Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Pharmacol Res ; 113(Pt A): 364-375, 2016 11.
Article in English | MEDLINE | ID: mdl-27620070

ABSTRACT

In this study, we have addressed the pharmacogenomic basis of the response of gastrointestinal tumors to six anticancer drugs using a panel of fifteen cell lines derived from pancreatic, stomach and biliary tract cancers. We determined the constitutive expression levels of 96 genes, whose encoded proteins contribute to drug action, and identified a major gene network that contains broad selectivity nucleoside transporter genes, as well as several genes known to be involved in cell proliferation and survival. All cell lines were exposed to 5'-DFUR, 5-FU, gemcitabine, cisplatin, doxorubicin and paclitaxel for 48h and cell response was measured using MTT assays. We correlated the cell response of the fifteen cell lines with the mRNA expression of the selected 96 genes and identified sets of 4-5 genes whose expression profiles correlated to responsiveness to each anticancer drug. These genes may be good candidates as response predictors to such therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Pharmacogenetics/methods , RNA, Messenger/metabolism
2.
BMC Cancer ; 15: 223, 2015 Apr 04.
Article in English | MEDLINE | ID: mdl-25886138

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma is a particularly challenging malignancy characterized by poor responsiveness to conventional chemotherapy. Although this tumor frequently overexpresses or possesses constitutively activated variants of IGF-IR and EGFR/Her-2, clinical trials using inhibitors of these receptors have failed. ErbB receptors have been proposed as one mechanism involved in the resistance to IGF-IR inhibitors. Therefore, combined treatment with inhibitors of both IGF-IR and ErbB receptors would appear to be a good strategy for overcoming the emergence of resistance. METHODS: Sensitivity of cells to NVP-AEW541 and lapatinib in single or combination treatment was assessed by MTT or WST-8 assays in a panel of human pancreatic cancer cell lines and cancer stem cells. Tumorspheres enriched in cancer stem cells were obtained from cultures growing in non-adherent cell plates. The effects on cell signalling pathways were analyzed by Western blot. RESULTS: We found that combined treatment with the IGF-IR and EGFR/Her-2 inhibitors NVP-AEW541 and lapatinib, respectively, synergistically inhibited pancreatic cancer cell growth. Analysis at molecular level argued in favor of cross-talk between IGF-IR and ErbBs pathways at IRS-1 level and indicated that the synergistic effect is associated with the total abolishment of Akt, Erk and IRS-1 phosphorylation. Moreover, these inhibitors acted synergistically in tumorsphere cultures to eliminate cancer stem cells, in contrast to their resistance to gemcitabine. CONCLUSIONS: Taken together, these data indicate that simultaneous blockade of IGF-IR and EGFR/Her-2 using NVP-AEW541 and lapatinib may overcome resistance in pancreatic cancer. Thus, the synergy observed with this combined treatment indicates that it may be possible to maximize patient benefit with the appropriate combination of currently known anticancer agents.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Humans , Lapatinib , Quinazolines , Signal Transduction/drug effects , Spheroids, Cellular , Tumor Cells, Cultured
3.
Stem Cell Res ; 12(1): 1-10, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24141108

ABSTRACT

Pancreatic tumors contain cancer stem cells highly resistant to chemotherapy. The identification of therapies that can eliminate this population of cells might provide with more effective treatments. In the current work we evaluated the potential of oncolytic adenoviruses to act against pancreatic cancer stem cells (PCSC). PCSC from two patient-derived xenograft models were isolated from orthotopic pancreatic tumors treated with saline, or with the chemotherapeutic agent gemcitabine. An enrichment in the number of PCSC expressing the cell surface marker CD133 and a marked enhancement on tumorsphere formation was observed in gemcitabine treated tumors. No significant increase in the CD44, CD24, and epithelial-specific antigen (ESA) positive cells was observed. Neoplastic sphere-forming cells were susceptible to adenoviral infection and exposure to oncolytic adenoviruses resulted in elevated cytotoxicity with both Adwt and the tumor specific AduPARE1A adenovirus. In vivo, intravenous administration of a single dose of AduPARE1A in human-derived pancreatic xenografts led to a remarkable anti-tumor effect. In contrast to gemcitabine AduPARE1A treatment did not result in PCSC enrichment. No enrichment on tumorspheres neither on the CD133(+) population was detected. Therefore our data provide evidences of the relevance of uPAR-controlled oncolytic adenoviruses for the elimination of pancreatic cancer stem cells.


Subject(s)
Adenoviridae , Neoplastic Stem Cells/cytology , Pancreatic Neoplasms/therapy , Receptors, Urokinase Plasminogen Activator/genetics , AC133 Antigen , Adenoviridae/genetics , Adenoviridae/metabolism , Adenovirus E1A Proteins/genetics , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , CD24 Antigen/metabolism , Cell Adhesion Molecules/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Epithelial Cell Adhesion Molecule , Glycoproteins/metabolism , Humans , Hyaluronan Receptors/metabolism , Male , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/virology , Oncolytic Virotherapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Peptides/metabolism , Promoter Regions, Genetic , Receptors, Urokinase Plasminogen Activator/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured , Gemcitabine
4.
Cell Oncol (Dordr) ; 34(6): 511-21, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21681527

ABSTRACT

BACKGROUND: Efforts to identify novel therapeutic options for human pancreatic ductal adenocarcinoma (PDAC) have failed to result in a clear improvement in patient survival to date. Pancreatic cancer requires efficient therapies that must be designed and assayed in preclinical models with improved predictor ability. Among the available preclinical models, the orthotopic approach fits with this expectation, but its use is still occasional. METHODS: An in vivo platform of 11 orthotopic tumor xenografts has been generated by direct implantation of fresh surgical material. In addition, a frozen tumorgraft bank has been created, ensuring future model recovery and tumor tissue availability. RESULTS: Tissue microarray studies allow showing a high degree of original histology preservation and maintenance of protein expression patterns through passages. The models display stable growth kinetics and characteristic metastatic behavior. Moreover, the molecular diversity may facilitate the identification of tumor subtypes and comparison of drug responses that complement or confirm information obtained with other preclinical models. CONCLUSIONS: This panel represents a useful preclinical tool for testing new agents and treatment protocols and for further exploration of the biological basis of drug responses.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Drug Evaluation, Preclinical/methods , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Blotting, Western , Carcinoma, Pancreatic Ductal/drug therapy , Female , Humans , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Nude , Middle Aged , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Tissue Array Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays , ras Proteins/genetics , ras Proteins/metabolism
5.
Mol Cancer Ther ; 10(3): 505-17, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21388975

ABSTRACT

Gemcitabine is a nucleoside analogue with anticancer activity. Inside the cell, it is sequentially phosphorylated to generate the active drug. Phosphorylated nucleoside analogues have been shown to traffic through gap junctions. We investigated the participation of gap junctional intercellular communication (GJIC) as a possible mechanism spreading gemcitabine cytotoxicity in pancreatic tumors. Immunohistochemical analysis of pancreatic cancer biopsies revealed increased connexin 26 (Cx26) content but loss of connexins 32 (Cx32) and 43 (Cx43) expression. Cx26 abundance in neoplastic areas was confirmed by Cx26 mRNA in situ hybridization. Heterogeneity on the expression levels and the localization of Cx26, Cx32, and Cx43 were identified in pancreatic cancer cells and found to be associated with the extent of GJIC, and correlated with gemcitabine bystander cytotoxic effect. The abundance of Cx26 at the contact points in tumoral regions prompted us to study the involvement of Cx26 in the GJIC of gemcitabine toxic metabolites and their influence on the antitumoral effects of gemcitabine. Knockdown of Cx26 led to decreased GJIC and reduced gemcitabine bystander killing whereas overexpression of Cx26 triggered increased GJIC and enhanced the gemcitabine cytotoxic bystander effect. Gemcitabine treatment of mice bearing tumors, with a high GJIC capacity, resulted in a significant delay in tumor progression. Interestingly, gemcitabine administration in mice bearing tumors that overexpress Cx26 triggered a dramatic tumor regression of 50% from the initial volume. This study shows that Cx26 participates in the gap junction-mediated bystander cytoxic effect of gemcitabine and provides evidence that upregulation of Cx26 improves gemcitabine anticancer efficacy.


Subject(s)
Bystander Effect/physiology , Connexins/metabolism , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/metabolism , Animals , Cell Communication/physiology , Cell Line, Tumor , Cell Proliferation , Connexin 26 , Connexin 43/genetics , Connexins/antagonists & inhibitors , Connexins/genetics , Deoxycytidine/pharmacology , Gap Junctions/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Phosphorylation , RNA, Messenger/genetics , Gemcitabine , Gap Junction beta-1 Protein
6.
Biochem Pharmacol ; 76(3): 322-9, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18589402

ABSTRACT

Nucleoside-derived anticancer agents must be transported across the plasma membrane as a preliminary step to their conversion into active drugs. Hence, modulation of a specific nucleoside transporter may affect bioavailability and contribute significantly to sensitizing tumor cells to these anticancer agents. We have generated and functionally characterized a new recombinant adenovirus (Ad-hENT1) that has allowed us to overexpress the equilibrative nucleoside transporter hENT1 and to analyze its effects in human pancreatic tumor cells. Overexpression of hENT1 is associated with changes in cell cycle profile, in a variable manner depending on the particular cell type, thus suggesting a metabolic link between hENT1-mediated transport processes and the enzymatic machinery responsible for intracellular nucleoside metabolism. When assayed in vivo in a human pancreatic adenocarcinoma xenograft, intratumoral Ad-hENT1 injection improved the therapeutic response to gemcitabine. In summary, hENT1 overexpression is associated with alterations in nucleoside enzymatic machinery and cell cycle progression in cultured cells and enhances gemcitabine action in vivo.


Subject(s)
Adenocarcinoma/drug therapy , Adenoviridae/genetics , Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Equilibrative Nucleoside Transporter 1/biosynthesis , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antimetabolites, Antineoplastic/therapeutic use , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Equilibrative Nucleoside Transporter 1/genetics , Female , Genetic Vectors , Humans , Injections, Intralesional , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Recombinant Fusion Proteins/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Uridine/metabolism , Xenograft Model Antitumor Assays , Gemcitabine
7.
Hum Gene Ther ; 17(12): 1187-200, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17069538

ABSTRACT

The CYP2B1/cyclophosphamide (CPA) suicide gene therapy approach has been shown to be highly promising in clinical trials for the treatment of pancreatic cancer. However, delivering the therapeutic gene to a sufficient number of tumor cells able to trigger a complete response remains a challenge. Target-specific delivery of adenovirus to fibroblast growth factor receptors (FGFRs) has been obtained in a variety of tumor models and has been shown to highly increase transduction efficiency. In the present paper we have tested the therapeutic outcome of retargeting the adenoviral vector, Ad-CYP2B1, to FGFRs, using an FGF2-Fab' conjugate, in pancreatic cancer models. First, we show a heterogeneous subcellular distribution of overexpressed FGFR-1 in pancreatic cancer cells. Higher transduction efficiency was observed in five of the six cell lines studied after FGF2-AdGFPLuc infection. Interestingly, an association between FGFR-1 membrane cell expression and viral entry was found. Moreover, tumors injected with FGF2-AdGFPLuc showed enhanced and persistent transgene expression. Importantly, we demonstrate the relevant enhanced cytotoxic effect of the FGF2-Ad-CYP2B]/CPA system in four of the six cell lines studied. Moreover, retargeting Ad-CYP2B1/CPA to FGFRs resulted in a potent antitumoral effect and in an increased survival rate, in two human pancreatic xenograft models. Thus, our results indicate that redirecting adenoviruses to FGFRs highly increases the potency of the suicide system CYP2B1/CPA. Consequently, it may constitute a promising approach to the treatment of patients with pancreatic tumors, in which a high proportion of FGF receptors precisely localize to the plasma membrane.


Subject(s)
Cyclophosphamide/pharmacology , Cytochrome P-450 CYP2B1/genetics , Fibroblast Growth Factor 2/genetics , Genetic Therapy/methods , Pancreatic Neoplasms/therapy , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adenoviridae/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Membrane/metabolism , Fibroblast Growth Factor 2/metabolism , Gene Expression , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transplantation, Heterologous
8.
Oncology ; 68(2-3): 179-89, 2005.
Article in English | MEDLINE | ID: mdl-16006755

ABSTRACT

Human pancreatic adenocarcinomas are highly resistant to conventional treatment modalities, specially to chemotherapy. Among the genes that modulate apoptosis in response to cytotoxic drugs, the role of p53 has been demonstrated to be of paramount importance. Moreover, p53 is mutated in close to 50% of pancreatic cancer, which renders attractive the reintroduction of this gene as a way to enhance the action of chemotherapeutics. In this paper, gemcitabine, the most effective drug for the treatment of pancreatic tumors, has been selected to develop a new combination approach in vivo based on an administration schedule previously optimized in vitro. In a human xenograft model, the sequential administration of gemcitabine and p53 resulted in potent tumor growth inhibition. Statistical differences were observed with respect to the growth of tumors receiving only gemcitabine or p53. Moreover, the chemosensitization observed in tumors treated with the combination gemcitabine-p53 correlated with differential histological features such as important increases in intratumoral fibrosis and apoptotic levels, when compared with unimodal treatments. Taken together, our data indicate that reintroduction of p53 function in human pancreatic tumors in vivo allows to restore molecular pathways improving the response to gemcitabine. It may constitute a useful step towards a better clinical treatment of patients harboring pancreatic cancer.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/enzymology , Animals , Colorimetry , Drug Administration Schedule , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Pancreatic Neoplasms/enzymology , Transplantation, Heterologous , Tumor Suppressor Protein p53/pharmacology , beta-Galactosidase/blood , Gemcitabine
9.
Clin Cancer Res ; 10(4): 1454-62, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14977849

ABSTRACT

PURPOSE: Gene transfer of a truncated variant of the retinoblastoma (RB) gene encoding a M(r) 94000 protein that lacks the NH(2)-terminal 112 amino acid residues, termed RB94, has been shown to inhibit proliferation of several human tumor cell types. We have assessed its therapeutic effectiveness on pancreatic cancer, one of the most aggressive and therapy-resistant types of cancer. For this purpose, preclinical studies aimed to evaluate the therapeutic potential of RB94 gene transfer in pancreatic cancer were carried out. EXPERIMENTAL DESIGN: We have compared the antiproliferative effects of adenovirus-mediated gene transfer of RBwt and RB94 at the in vitro and in vivo levels in three RB-positive human pancreatic tumor cell lines: (a). NP-9; (b). NP-18; and (c). NP-31. We have also examined their effects on cell cycle and their capacity to induce apoptosis. RESULTS: In vitro results indicate that RB94 gene transfer has stronger antiproliferative effects compared with RBwt. RB94 transduction correlated with accumulation at the S-G(2) phase of the cell cycle in the three cell lines tested and induction of apoptosis in two of them. In vivo studies show significant decreases in the growth rate of tumors treated with Ad-RB94 when compared with those treated with Ad-RBwt. Moreover, terminal deoxynucleotidyl transferase-mediated nick end labeling analyses of Ad-RB94-treated tumor sections revealed that only RB94 is able to significantly induce apoptosis. CONCLUSIONS: RB94 gene expression has antiproliferative effects also in human pancreatic tumor cells, being more effective than wild-type RB in preventing tumor growth.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Pancreatic Neoplasms/therapy , Retinoblastoma Protein/genetics , Animals , Annexin A5/pharmacology , Apoptosis , Blotting, Western , Cell Cycle , Cell Division , Cell Line, Tumor , Coloring Agents/pharmacology , Dose-Response Relationship, Drug , Humans , In Situ Nick-End Labeling , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Protein Structure, Tertiary , Time Factors
10.
Angiogenesis ; 7(4): 323-33, 2004.
Article in English | MEDLINE | ID: mdl-15886876

ABSTRACT

Recent evidence has established different functions for the tumor suppressor protein, p16(INK4A) aside from controlling the cell cycle. Here we report that cdk4/6 inhibition blocked both human umbilical vein endothelial cells (HUVEC) spreading on a vitronectin matrix and HUVEC migration on vitronectin. p16 can also act as an anti-angiogenic molecule in vitro since HUVEC and HMEC cells transfected with Ad-p16 or treated with Antennapedia p16 peptides are unable to differentiate on a Matrigel matrix. Both, p16, cyclin D1, cdk4 and cdk6 were immuno-colocalized with Ezrin, Rac, Vinculin, alphav-integrin, and FAK proteins in the ruffles and lamellipodia of migratory cells. Our results indicate that p16 is a key component of a new cytoplasmic pathway controlling angiogenesis of endothelial cells via the alphavbeta3-integrin-mediated migration.


Subject(s)
Cell Movement/physiology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Endothelium, Vascular/cytology , Neovascularization, Physiologic/physiology , Pseudopodia/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin alphaVbeta3/metabolism , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism , Vinculin/metabolism , rac GTP-Binding Proteins/metabolism
11.
Clin Cancer Res ; 9(13): 5000-8, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14581375

ABSTRACT

PURPOSE: Concentrative nucleoside transporter (CNT) 1, CNT3, equilibrative nucleoside transporter (ENT) 1, and, to a lesser extent, ENT2, appear to be the transporters responsible for 2',2'-difluorodeoxycytidine (gemcitabine; Gemzar) uptake into cells. Gemcitabine is used currently in the treatment of pancreatic cancer, but the role of specific nucleoside carrier proteins in gemcitabine cytotoxicity has not been elucidated. Indeed, it is not known which nucleoside transporters are expressed in human pancreas. EXPERIMENTAL DESIGN: In this study we have used four cell lines [pancreatic neoplasia (NP)9, NP18, NP29, and NP31] derived from human pancreatic adenocarcinomas to monitor the pattern of nucleoside transporter expression, and we have heterologously expressed the high-affinity gemcitabine transporter human orthologue (h) CNT1 to monitor its role in drug responsiveness. RESULTS: All of the cell lines take up gemcitabine mostly via the hENT1 transporter, which is expressed at high levels. Reverse transcription-PCR analysis of the other four cloned plasma membrane transporter mRNAs revealed very different expression patterns among NP cell lines, with apparent selective loss or decrease of hCNT mRNAs. NP cells transiently express hCNT1-type Na(+)-dependent nucleoside transport activity at low/medium cell density but not in confluent cultures. Cells expressing hCNT1 in a more constitutive manner were cloned after stable transfection of hCNT1. Despite high constitutive hENT1 activity, this increased sensitivity to gemcitabine. CONCLUSION: In summary, human pancreatic adenocarcinoma cells overexpress hENT1, although they retain the ability to express a functional hCNT1 transporter, an isoform that confers sensitivity to gemcitabine.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Membrane Transport Proteins/physiology , Pancreatic Neoplasms/drug therapy , Biological Transport , Cell Line, Tumor , Cell Membrane/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Kinetics , Poly A , Polymerase Chain Reaction , Protein Isoforms , RNA/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Gemcitabine
12.
Cancer Res ; 63(17): 5544-50, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500393

ABSTRACT

Adenovirus synthesize proteins that interact with oncogene and tumor suppressor gene products to set the cell for virus replication. Mutant viruses defective in these functions replicate selectively in cancer cells and represent new tools to treat cancer. We report a selectivity strategy based on deletions of adenovirus Virus-Associated (VA) RNAs. In normal cells, these RNAs are necessary for virus replication because they inactivate the RNA-dependent protein kinase protein kinase R, a kinase that otherwise would block protein translation in response to infection. However, downstream effectors of Ras can also inactivate protein kinase R, and therefore, the need for VA RNA genes should be bypassed in cells with an active Ras pathway. We demonstrate here that a VAI RNA mutant presents a Ras-dependent replication and can be used for oncolytic virotherapy of pancreatic tumors.


Subject(s)
Adenoviruses, Human/physiology , RNA, Viral/genetics , ras Proteins/physiology , Adenoviruses, Human/genetics , Cytopathogenic Effect, Viral/genetics , Enzyme Activation , Humans , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/virology , Tumor Cells, Cultured , Virus Replication/genetics , eIF-2 Kinase/metabolism
13.
J Biol Chem ; 277(52): 50263-74, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12401786

ABSTRACT

Mitogenic stimulation leads to activation of G(1) cyclin-dependent kinases (CDKs), which phosphorylate pocket proteins and trigger progression through the G(0)/G(1) and G(1)/S transitions of the cell cycle. However, the individual role of G(1) cyclin-CDK complexes in the coordinated regulation of pocket proteins and their interaction with E2F family members is not fully understood. Here we report that individually or in concert cyclin D1-CDK and cyclin E-CDK complexes induce distinct and coordinated phosphorylation of endogenous pocket proteins, which also has distinct consequences in the regulation of pocket protein interactions with E2F4 and the expression of p107 and E2F1, both E2F-regulated genes. The up-regulation of these two proteins and the release of p130 and pRB from E2F4 complexes allows formation of E2F1 complexes not only with pRB but also with p130 and p107 as well as the formation of p107-E2F4 complexes. The formation of these complexes occurs in the presence of active cyclin D1-CDK and cyclin E-CDK complexes, indicating that whereas phosphorylation plays a role in the abrogation of certain pocket protein/E2F interactions, these same activities induce the formation of other complexes in the context of a cell expressing endogenous levels of pocket and E2F proteins. Of note, phosphorylated p130 "form 3," which does not interact with E2F4, readily interacts with E2F1. Our data also demonstrate that ectopic overexpression of either cyclin is sufficient to induce mitogen-independent growth in human T98G and Rat-1 cells, although the effects of cyclin D1 require downstream activation of cyclin E-CDK2 activity. Interestingly, in T98G cells, cyclin D1 induces cell cycle progression more potently than cyclin E. This suggests that cyclin D1 activates pathways independently of cyclin E that ensure timely progression through the cell cycle.


Subject(s)
Cell Cycle Proteins , Cell Cycle/physiology , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Transcription Factors/metabolism , Adenoviridae , Animals , Cell Line , Cells, Cultured , Cyclin G , Cyclin G1 , E2F Transcription Factors , E2F1 Transcription Factor , E2F4 Transcription Factor , Fibroblasts/cytology , Fibroblasts/physiology , G2 Phase , Mice , Mitosis , Phosphoproteins/metabolism , Phosphorylation , Rats , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL