Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
Transl Oncol ; 16: 101315, 2022 Feb.
Article En | MEDLINE | ID: mdl-34906890

Pancreatic ductal adenocarcinoma (PDAC) patients are frequently treated by chemotherapy. Even if personalized therapy based on molecular analysis can be performed for some tumors, PDAC regimens selection is still mainly based on patients' performance status and expected efficacy. Therefore, the establishment of molecular predictors of chemotherapeutic efficacy could potentially improve prognosis by tailoring treatments. We have recently developed an RNA-based signature that predicts the efficacy of adjuvant gemcitabine using 38 PDAC primary cell cultures. While demonstrated its efficiency, a significant association with the classical/basal-like PDAC spectrum was observed. We hypothesized that this flaw was due to the basal-like biased phenotype of cellular models used in our strategy. To overcome this limitation, we generated a prospective cohort of 27 consecutive biopsied derived pancreatic organoids (BDPO) and include them in the signature identification strategy. As BDPO's do not have the same biased phenotype as primary cell cultures we expect they can compensate one with each other and cover a broader range of molecular phenotypes. We then obtained an improved signature predicting gemcitabine sensibility that was validated in a cohort of 300 resected PDAC patients that have or have not received adjuvant gemcitabine. We demonstrated a significant association between the improved signature and the overall and disease-free survival in patients predicted as sensitive and treated with adjuvant gemcitabine. We propose then that including BDPO along primary cell cultures represent a powerful strategy that helps to overcome primary cell cultures limitations producing unbiased RNA-based signatures predictive of adjuvant treatments in PDAC.

2.
Ann Oncol ; 32(2): 250-260, 2021 02.
Article En | MEDLINE | ID: mdl-33188873

BACKGROUND: Chemotherapy is the only systemic treatment approved for pancreatic ductal adenocarcinoma (PDAC), with a selection of regimens based on patients' performance status and expected efficacy. The establishment of a potent stratification associated with chemotherapeutic efficacy could potentially improve prognosis by tailoring treatments. PATIENTS AND METHODS: Concomitant chemosensitivity and genome-wide RNA profiles were carried out on preclinical models (primary cell cultures and patient-derived xenografts) derived from patients with PDAC included in the PaCaOmics program (NCT01692873). The RNA-based stratification was tested in a monocentric cohort and validated in a multicentric cohort, both retrospectively collected from resected PDAC samples (67 and 368 patients, respectively). Forty-three (65%) and 203 (55%) patients received adjuvant gemcitabine in the monocentric and the multicentric cohorts, respectively. The relationships between predicted gemcitabine sensitivity and patients' overall survival (OS) and disease-free survival were investigated. RESULTS: The GemPred RNA signature was derived from preclinical models, defining gemcitabine sensitive PDAC as GemPred+. Among the patients who received gemcitabine in the test and validation cohorts, the GemPred+ patients had a higher OS than GemPred- (P = 0.046 and P = 0.00216). In both cohorts, the GemPred stratification was not associated with OS among patients who did not receive gemcitabine. Among gemcitabine-treated patients, GemPred+ patients had significantly higher OS than the GemPred-: 91.3 months [95% confidence interval (CI): 61.2-not reached] versus 33 months (95% CI: 24-35.2); hazard ratio 0.403 (95% CI: 0.221-0.735, P = 0.00216). The interaction test for gemcitabine and GemPred+ stratification was significant (P = 0.0245). Multivariate analysis in the gemcitabine-treated population retained an independent predictive value. CONCLUSION: The RNA-based GemPred stratification predicts the benefit of adjuvant gemcitabine in PDAC patients.


Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Chemotherapy, Adjuvant , Deoxycytidine/analogs & derivatives , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Retrospective Studies , Transcriptome , Gemcitabine
3.
Cell Death Differ ; 21(7): 1107-18, 2014 Jul.
Article En | MEDLINE | ID: mdl-24608790

Tumor Protein p53-Induced Nuclear Protein 1 (TP53INP1) is a tumor suppressor that modulates the p53 response to stress. TP53INP1 is one of the key mediators of p53 antioxidant function by promoting the p53 transcriptional activity on its target genes. TP53INP1 expression is deregulated in many types of cancers including pancreatic ductal adenocarcinoma in which its decrease occurs early during the preneoplastic development. In this work, we report that redox-dependent induction of p53 transcriptional activity is enhanced by the oxidative stress-induced SUMOylation of TP53INP1 at lysine 113. This SUMOylation is mediated by PIAS3 and CBX4, two SUMO ligases especially related to the p53 activation upon DNA damage. Importantly, this modification is reversed by three SUMO1-specific proteases SENP1, 2 and 6. Moreover, TP53INP1 SUMOylation induces its binding to p53 in the nucleus under oxidative stress conditions. TP53INP1 mutation at lysine 113 prevents the pro-apoptotic, antiproliferative and antioxidant effects of TP53INP1 by impairing the p53 response on its target genes p21, Bax and PUMA. We conclude that TP53INP1 SUMOylation is essential for the regulation of p53 activity induced by oxidative stress.


Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , Oxidative Stress , Sumoylation , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Autophagy , Cell Proliferation , HEK293 Cells , Humans , Ligases , MCF-7 Cells , Molecular Chaperones/metabolism , Oxidation-Reduction , Polycomb-Group Proteins/metabolism , Protein Inhibitors of Activated STAT/metabolism , SUMO-1 Protein/metabolism , Serine Endopeptidases/metabolism , Transcriptional Activation , Ubiquitin-Protein Ligases/metabolism
4.
Cell Death Differ ; 19(9): 1525-35, 2012 Sep.
Article En | MEDLINE | ID: mdl-22421968

TP53INP1 (tumor protein 53-induced nuclear protein 1) is a tumor suppressor, whose expression is downregulated in cancers from different organs. It was described as a p53 target gene involved in cell death, cell-cycle arrest and cellular migration. In this work, we show that TP53INP1 is also able to interact with ATG8-family proteins and to induce autophagy-dependent cell death. In agreement with this finding, we observe that TP53INP1, which is mainly nuclear, relocalizes in autophagosomes during autophagy where it is eventually degraded. TP53INP1-LC3 interaction occurs via a functional LC3-interacting region (LIR). Inactivating mutations of this sequence abolish TP53INP1-LC3 interaction, relocalize TP53INP1 in autophagosomes and decrease TP53INP1 ability to trigger cell death. Interestingly, TP53INP1 binds to ATG8-family proteins with higher affinity than p62, suggesting that it could partially displace p62 from autophagosomes, modifying thereby their composition. Moreover, silencing the expression of autophagy related genes (ATG5 or Beclin-1) or inhibiting caspase activity significantly decreases cell death induced by TP53INP1. These data indicate that cell death observed after TP53INP1-LC3 interaction depends on both autophagy and caspase activity. We conclude that TP53INP1 could act as a tumor suppressor by inducing cell death by caspase-dependent autophagy.


Adaptor Proteins, Signal Transducing/metabolism , Autophagy/physiology , Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Autophagy-Related Protein 8 Family , Beclin-1 , Carrier Proteins/genetics , HEK293 Cells , HeLa Cells , Heat-Shock Proteins/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Mutation , Phagosomes/genetics , Phagosomes/metabolism , Protein Structure, Tertiary , Protein Transport/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Oncogene ; 30(27): 3049-61, 2011 Jul 07.
Article En | MEDLINE | ID: mdl-21339733

Tumor protein 53 induced nuclear protein 1 (TP53INP1) is a p53 target gene that induces cell growth arrest and apoptosis by modulating p53 transcriptional activity. TP53INP1 interacts physically with p53 and is a major player in the p53-driven oxidative stress response. Previously, we demonstrated that TP53INP1 is downregulated in an early stage of pancreatic cancerogenesis and when restored is able to suppress pancreatic tumor development. TP53INP1 downregulation in pancreas is associated with an oncogenic microRNA miR-155. In the present work, we studied the effects of TP53INP1 on cell migration. We found that TP53INP1 inactivation correlates with increased cell migration both in vivo and in vitro. The impact of TP53INP1 expression on cell migration was studied in different cellular contexts: mouse embryonic fibroblast and different pancreatic cancer cell lines. Its expression decreases cell migration by the transcriptional downregulation of secreted protein acidic and rich in cysteine (SPARC). SPARC is a matrix cellular protein, which governs diverse cellular functions and has a pivotal role in regulating cell-matrix interactions, cellular proliferation and migration. SPARC was also showed to be upregulated in normal pancreas and in pancreatic intraepithelial neoplasia lesions in a pancreatic adenocarcinoma mouse model only in the TP53INP1-deficient animals. This novel TP53INP1 activity on the regulation of SPARC expression could explain in part its tumor suppressor function in pancreatic adenocarcinoma by modulating cellular spreading during the metastatic process.


Carrier Proteins/physiology , Cell Movement/physiology , Heat-Shock Proteins/physiology , Osteonectin/metabolism , Pancreatic Neoplasms/pathology , Down-Regulation , Humans
6.
Oncogene ; 29(17): 2528-39, 2010 Apr 29.
Article En | MEDLINE | ID: mdl-20140013

Vav proteins are guanine nucleotide exchange factors for Rho GTPases that regulate cell adhesion, motility, spreading and proliferation in response to growth factor signalling. In this work, we show that Vav2 expression delayed epidermal growth factor receptor (EGFR) internalization and degradation, and enhanced EGFR, ERK and Akt phosphorylations. This effect of Vav2 on EGFR degradation is dependent on its guanine nucleotide exchange function. Knockdown of Vav2 in HeLa cells enhanced EGFR degradation and reduced cell proliferation. epidermal growth factor stimulation led to co-localization of Vav2 with EGFR and Rab5 in endosomes. We further show that the effect of Vav2 on EGFR stability is modulated by its interaction with two endosome-associated proteins and require RhoA function. Thus, in this work, we report for the first time that Vav2 can regulate growth factors receptor signalling by slowing receptor internalization and degradation through its interaction with endosome-associated proteins.


Endocytosis , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-vav/physiology , Cell Line, Tumor , Endosomes/metabolism , ErbB Receptors/analysis , Humans , Proto-Oncogene Proteins c-vav/analysis , rab5 GTP-Binding Proteins/analysis
7.
World J Surg ; 34(4): 830-5, 2010 Apr.
Article En | MEDLINE | ID: mdl-20145930

BACKGROUND: Medullary thyroid cancer (MTC) is characterized by early regional lymph node metastasis, the presence of which represents a critical obstacle to cure. At present no molecular markers have been successfully integrated into the clinical care of sporadic MTC. The present study was designed to evaluate TP53INP1 expression in MTC and to assess its ability to guide the surgeon to the optimal extent of surgery performed with curative intent. METHODS: Thirty-eight patients with sporadic MTC were evaluated. TP53INP1 immunoexpression was studied on embedded paraffin material and on cytological smears. RESULTS: TP53INP1 was expressed in normal C cells, in C-cell hyperplasia, and in 57.9% of MTC. It was possible to identify two groups of MTC according to the proportion of TP53INP1 expressing tumor cells: group 1 from 0% to <50% and group 2 from 50% to 100% of positive cells. Patients with a decreased expression of TP53INP1 (group 1) had a lower rate of nodal metastasis (18.8% versus 63.4% in group 2; P = 0.009), with only minimal lymph node involvement per N1 patient (2.7% of positive lymph nodes versus 22.9%; P < 0.001) and better outcomes (100% of biochemical cure versus 55.5%; P < 0.001). Patients with distant metastases were only observed in group 2. Cytological samples exhibit similar results to their embedded counterparts. CONCLUSIONS: TP53INP1 immunoexpression appears to be a clinical predictor of lymph node metastasis in MTC. The evaluation of TP53INP1 expression may guide the extent of lymph node dissection in the clinically node-negative neck. These findings require prospective validation.


Carcinoma, Medullary/metabolism , Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , Thyroid Neoplasms/metabolism , Adult , Aged , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Medullary/pathology , Carcinoma, Medullary/surgery , Chi-Square Distribution , Female , Humans , Immunohistochemistry , Logistic Models , Lymph Node Excision , Lymphatic Metastasis , Male , Middle Aged , Statistics, Nonparametric , Thyroid Neoplasms/pathology , Thyroid Neoplasms/surgery
8.
Cancer Lett ; 288(1): 116-23, 2010 Feb 01.
Article En | MEDLINE | ID: mdl-19631450

ArgBP2 is a multi-adapter protein involved in signal transduction associated to the cytoskeleton and was shown to regulate the migration and adhesion of pancreatic cancer cells thereby modulating their tumorigenicity. Here we describe the interaction of ArgBP2 with CIP4, a new associated protein identified by yeast two-hybrid. We found that both proteins modulated their reciprocal tyrosine phosphorylation catalyzed by the non-receptor tyrosine kinase c-Abl. We observed that, like ArgBP2, CIP4 directly interacted with WAVE1 and could enhance its phosphorylation by c-Abl. ArgBP2 and CIP4 acted synergistically to increase WAVE1 tyrosine phosphorylation. Finally, we could show that CIP4 was dispensable for the ArgBP2 induced blockade of cell migration whereas its overexpression was deleterious for this important function of ArgBP2.


Cell Movement , Homeodomain Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Pancreatic Neoplasms/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , Adaptor Proteins, Signal Transducing , Binding Sites , Cell Line, Tumor , Homeodomain Proteins/genetics , Humans , Microtubule-Associated Proteins/genetics , Minor Histocompatibility Antigens , Neoplasm Invasiveness , Pancreatic Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-abl/metabolism , RNA Interference , RNA-Binding Proteins , Signal Transduction , Transfection , Two-Hybrid System Techniques , src Homology Domains
9.
J Biol Chem ; 276(47): 44185-92, 2001 Nov 23.
Article En | MEDLINE | ID: mdl-11557757

We have used a quantitative fluorescent cDNA microarray hybridization approach to identify pancreatic genes induced by the cellular stress promoted by acute pancreatitis in the mouse. We report the cloning and characterization of one of them that encodes the stress-induced proteins (SIP). The mouse SIP gene is organized into five exons and expands over approximately 20 kilobase pairs. Exon 4 (38 base pairs) is alternatively spliced to generate two transcripts. Northern blot and in situ hybridization showed that both SIP mRNAs are rapidly and strongly induced in acinar cells of the pancreas with acute pancreatitis. They are also constitutively expressed in several other tissues, although with different ratios. They encode proteins of 18 and 27 kDa (SIP(18) and SIP(27)). SIP(27) is identical to the thymus-expressed acidic protein (TEAP) protein, formerly described as a thymus-specific protein. Expression of the SIP(18) and SIP(27)/EGFP or V5 fusion proteins showed that both are nuclear factors. We monitored SIP expression in NIH3T3 cells submitted to various stress agents. UV stress, base damaging, mutagenic stress, ethanol, heat shock, and oxidative stress induced the concomitant expression of SIP(18) and SIP(27) mRNAs. Finally, transient transfection of SIP(18) and SIP(27) expression plasmids induced death by apoptosis in COS7 cells as measured by terminal deoxynucleotidyltransferase-mediated dUTP nick end-labeling staining. In conclusion, the SIP gene is an important element of cellular stress response. It is expressed in many tissues and induced by a variety of stress agents affecting many cellular pathways. SIP generates, by alternative splicing, two nuclear proteins that can promote cell death by apoptosis.


Alternative Splicing , Carrier Proteins/physiology , Heat-Shock Proteins , 3T3 Cells , Amino Acid Sequence , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Base Sequence , Carrier Proteins/genetics , Cloning, Molecular , In Situ Hybridization, Fluorescence/methods , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
10.
Eur J Cell Biol ; 80(2): 156-63, 2001 Feb.
Article En | MEDLINE | ID: mdl-11302520

Expression of the Cdx1 homeobox gene in epithelial intestinal cells promotes cellular growth and differentiation. Cdx1and the Pancreatitis Associated Protein I (PAP I) are concomitantly expressed in the epithelial cells of the lower part of the intestinal crypts. Because Cdx1 is a transcription factor and PAP I, in other tissues, is a proliferative factor, we looked for a relationship between these two proteins in the intestinal-derived IEC-6 cells. After stable transfection with a Cdx1 expression vector, they produce high levels of the PAP I transcript and protein indicating a functional link between the two genes. Demonstration of Cdx1 binding to the PAP I promoter region and suppression of PAP I induction after deletion of the corresponding sequence indicated that Cdx1 is a transcription factor controlling PAP I gene expression in intestinal cells. By infecting IEC-6 cells with adenoviruses expressing PAP I, we demonstrated that PAP I induces mitosis in these cells. On the other hand, inhibition of the PAP I expression in the IEC-6 Cdxl-expressing cells using an antisense strategy confirmed the requirement of this protein for the effect of Cdx1 on cell growth. Finally, addition of the immunopurified PAP I to the culture medium promotes cell growth of the IEC-6 cells in a dose-dependent manner. Maximal effect was obtained at 1 ng/ml. Taken together these results demonstrate that PAP I is a target of the Cdx1 homeobox gene in intestinal cells which participates in the regulation of intestinal cell growth via an autocrine and/or paracrine mechanism.


Acute-Phase Proteins/metabolism , Antigens, Neoplasm , Biomarkers, Tumor , Homeodomain Proteins/metabolism , Intestinal Mucosa/cytology , Lectins, C-Type , Transcription Factors/metabolism , Acute-Phase Proteins/genetics , Animals , Cell Division , Cell Line , Epithelial Cells/cytology , Gene Expression Regulation , Homeodomain Proteins/genetics , Humans , Mitosis , Pancreatitis-Associated Proteins , Promoter Regions, Genetic , Rats , Response Elements , Transcription Factors/genetics , Transcriptional Activation
11.
Biochem Biophys Res Commun ; 277(3): 660-7, 2000 Nov 02.
Article En | MEDLINE | ID: mdl-11062010

Most attacks of acute pancreatitis are self-limiting, suggesting that the pancreatic cells adapt their phenotype to prevent progression of the disease. Such phenotypic change must involve a coordinated modification in the expression of numerous genes. To identify differentially expressed genes, high-density mouse cDNA microarrays were hybridized with cDNA probes from both healthy pancreas and pancreas affected by acute pancreatitis. From the 7981 mouse genes analyzed, 239 showed significant changes in their expression during the acute phase of pancreatitis. Among them, 107 genes were up-regulated whereas 132 were down-regulated. They include genes whose function was not previously related to pancreatitis, suggesting that they are involved in some way into the acute pancreatic response. Finally, 40% of differentially expressed genes corresponded to ESTs. Demonstration that a large quantity of unexpected or yet uncharacterized genes showed altered expression during acute pancreatitis underscores the interest of a genome-based investigation. Some of these genes are certainly involved in the cellular defense against pancreatitis and, as such, deserve being studied further.


Gene Expression Profiling , Gene Expression Regulation , Oligonucleotide Array Sequence Analysis/methods , Pancreatitis/genetics , Acute-Phase Reaction , Animals , Down-Regulation , Mice , Mice, Inbred BALB C , Up-Regulation
12.
Gastroenterology ; 119(3): 816-28, 2000 Sep.
Article En | MEDLINE | ID: mdl-10982776

BACKGROUND & AIMS: Tumor necrosis factor (TNF)-alpha contributes to the development of acute pancreatitis. Because TNF-alpha is involved in the control of apoptosis, we studied its interaction with the pancreatic apoptotic pathway. METHODS: Pancreatic acinar AR4-2J cells were used. Apoptosis was monitored by morphologic and biochemical criteria. RESULTS: TNF-alpha induced apoptosis in AR4-2J cells. Induction was strongly enhanced in cells treated with actinomycin D, suggesting that TNF-alpha activated concomitantly an antiapoptotic mechanism through newly synthesized proteins. This mechanism involved activation of nuclear factor-kappaB (NF-kappaB) and mitogen-activated protein (MAP) kinases because their inhibition worsened TNF-alpha-induced apoptosis. The antiapoptotic pancreatitis-associated protein (PAP) I is a candidate for mediating TNF-alpha activity. Its expression is induced by TNF-alpha, and cells overexpressing PAP I show significantly less apoptosis on exposure to TNF-alpha. We examined whether TNF-alpha induction of PAP I expression was mediated by NF-kappaB or MAP kinases by using specific inhibitors of both pathways. Inhibition of NF-kappaB had no effect. However, inhibitors of MEK1 eliminated PAP I induction. CONCLUSIONS: TNF-alpha induces concomitantly proapoptotic and antiapoptotic mechanisms in pancreatic AR4-2J cells. Antiapoptotic mechanisms are mediated by NF-kappaB and MAP kinases, and PAP I is one of the effectors of apoptosis inhibition.


Acute-Phase Proteins/physiology , Antigens, Neoplasm , Apoptosis/drug effects , Biomarkers, Tumor , Lectins, C-Type , NF-kappa B/antagonists & inhibitors , Pancreas/drug effects , Pancreas/physiopathology , Tumor Necrosis Factor-alpha/pharmacology , Acute-Phase Proteins/metabolism , Animals , Apoptosis/physiology , Cell Line , Dactinomycin/pharmacology , Drug Synergism , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/physiology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pancreas/pathology , Pancreatitis-Associated Proteins , Protein Serine-Threonine Kinases/physiology , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
13.
Biochem Biophys Res Commun ; 251(3): 710-3, 1998 Oct 29.
Article En | MEDLINE | ID: mdl-9790974

We report in this paper that cycloheximide induces PAP mRNA expression in the pancreatic acinar cell line AR4-2J in a dose- and time-dependent manner. We analyzed whether stabilization of the PAP mRNA or the direct induction of its transcription contributed to the induction of PAP mRNA expression by the drug. We first infected the cells, which do not express PAP mRNA constitutively, with a recombinant adenovirus in which the PAP cDNA was subcloned downstream of the CMV promotor, to obtain high levels of transcript. Then, transcription was pharmacologically blocked, the cells were treated with cycloheximide, and the PAP mRNA concentration was monitored over 8 h by Northern blot. PAP mRNA concentration remained unchanged for 4 h and then decreased in both cycloheximide-treated and control cells, ruling out a significant contribution of posttranscriptional regulation in cycloheximide induction. Direct regulation of gene transcription is therefore likely and we investigated whether it could involve ADP-ribosylation. Cycloheximide-induced cells were treated with two chemical inhibitors of poly(ADP-ribose) polymerase. 3-Aminobenzamide inhibited 75% of PAP gene induction and 4-hydroxyquinazolone, the highly specific inhibitor of the enzyme, blocked almost completely PAP expression, suggesting that ADP-ribosylation was indeed required for the upregulation of PAP gene expression by cycloheximide.


Acute-Phase Proteins/biosynthesis , Adenosine Diphosphate Ribose/metabolism , Antigens, Neoplasm , Biomarkers, Tumor , Cycloheximide/pharmacology , Lectins, C-Type , Pancreas/metabolism , Protein Processing, Post-Translational , Acute-Phase Proteins/genetics , Benzamides/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation , Half-Life , Models, Genetic , Pancreas/cytology , Pancreas/drug effects , Pancreatitis-Associated Proteins , Poly(ADP-ribose) Polymerase Inhibitors , Transcription, Genetic , Transcriptional Activation
14.
Gastroenterology ; 114(4): 808-16, 1998 Apr.
Article En | MEDLINE | ID: mdl-9516402

BACKGROUND & AIMS: Free radicals are involved in the pathogenesis of acute pancreatitis, during which pancreatitis-associated protein (PAP)-I is overexpressed. We explored whether PAP-I expression could be induced by oxidative stress and whether it could affect apoptosis. METHODS: AR4-2J cells were exposed to H2O2 or menadione, and PAP-I messenger RNA (mRNA) expression was analyzed by Northern blotting. RESULTS: Maximal expression was observed with 0.1 mmol/L H2O2 or with 0.05 mmol/L menadione. Induction was detectable after 12 hours, reached a climax at 18 hours, and then decreased. Pretreatment of the cells with pyrrolidine dithiocarbamate completely abolished PAP-I mRNA induction, suggesting involvement of NFkappaB in the signaling pathway. These findings were confirmed in transient transfection assays using a plasmid containing the PAP-I promoter linked to the chloramphenicol acetyltransferase reporter gene. Then the relationship between PAP-I induction and protection against cell damage during oxidative stress was considered. Constitutive PAP-I expression in AR4-2J cells after transfection with PAP-I complementary DNA conferred significant resistance to apoptosis induced by low doses of H2O2 but not to necrosis induced by high doses of H2O2. CONCLUSIONS: These results suggest that during oxidative stress, PAP-I might be part of a mechanism of pancreatic cell protection against apoptosis.


Acute-Phase Proteins/biosynthesis , Antigens, Neoplasm , Apoptosis , Biomarkers, Tumor , Lectins, C-Type , Pancreas/metabolism , Acute-Phase Proteins/genetics , Antioxidants/pharmacology , Cell Line , Free Radicals , Hydrogen Peroxide/toxicity , Oxidative Stress , Pancreas/drug effects , Pancreas/pathology , Pancreatitis-Associated Proteins , Promoter Regions, Genetic , RNA, Messenger/analysis
15.
Arch Biochem Biophys ; 340(1): 111-6, 1997 Apr 01.
Article En | MEDLINE | ID: mdl-9126283

Previous analysis of the rat PAP I promoter indicated that the region between nt -180 and -81 possessed silencer activity in cells that did not express PAP I. Based on this finding, we performed a series of experiments to characterize functionally that region and analyze the nuclear proteins interacting with it. Transient transfection assays were conducted in the fibroblast Rat2 cell line, in which PAP I is not expressed, and in the pancreatic cell line AR-42J, expressing PAP I, using the CAT gene as reporter. Experiments in Rat2 cells revealed that the sequence with silencer activity was located within the rep27 region (position -180/-153). Suppressor activity was observed when rep27 was inserted upstream from the core PAP I promoter, in both orientations. By contrast, inserting the rep27 region in front of the promoters of SV40 or thymidine kinase did not affect or weakly enhanced CAT activity. Suppressor activity is therefore position-independent and promoter-dependent. In pancreatic AR-42J cells, rep27 act as a positive element but did not alter CAT expression when inserted in front of the core PAP I promoter or heterologous promoters. Electrophoretic mobility shift assays allowed identification of specific DNA-protein complexes. The shifted complex migrated at the same position with both Rat2 and AR-42J nuclear extracts. Moreover, similar band shifts were obtained with rat nuclear extracts from healthy pancreas, pancreas with acute pancreatitis, liver, kidney, spleen, and small intestine. Results suggest that the rep27 cis-acting element contributes to the tissue specific expression of the PAP I gene. That activity could be mediated by the synergistic action of several transcription factors, one of which being present in all cells.


Acute-Phase Proteins/genetics , Antigens, Neoplasm , Biomarkers, Tumor , Lectins, C-Type , Regulatory Sequences, Nucleic Acid , Animals , Cell Line , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Multigene Family , Nuclear Proteins/metabolism , Pancreatitis/metabolism , Pancreatitis-Associated Proteins , Promoter Regions, Genetic , Rats , Tissue Distribution , Transcription Factors/metabolism
16.
J Biol Chem ; 272(9): 5800-4, 1997 Feb 28.
Article En | MEDLINE | ID: mdl-9038194

The pancreatitis-associated protein I (PAP I) is a pancreatic secretory protein expressed in pancreas during acute pancreatitis but not in the healthy pancreas. The promoter of the PAP I gene thus represents a potential candidate to drive expression of therapeutic molecules to the diseased pancreas. In this work, we have constructed recombinant adenoviruses harboring the chloramphenicol acetyltransferase (CAT) gene driven by several fragments of the PAP I promoter and have characterized their properties in vitro and in vivo. In vitro studies showed that the transduction of the pancreatic cell line AR-42J with these adenoviruses led to low levels of CAT activity in basal conditions. After stimulation with a combination of interleukin-6 and dexamethasone or after induction of oxidative stress, CAT activity was strongly induced, a characteristic of the endogenous PAP I gene. Stimulation was maximal when constructs comprised 1253 base pairs of the PAP I promoter, upstream from initiation of transcription, and decreased with shorter fragments of 317, 180, 118 or 61 base pairs. The recombinant adenovirus containing the CAT gene under the control of the PAP I promoter fragment (-1253/+10) was also tested in vivo. Following administration by intravenous injection into mice, CAT activity was measured in several tissues 96 h later. In healthy animals, low but significant CAT activity was detected in pancreas, compared with near background values observed in the other tissues. When experimental acute pancreatitis was induced, CAT expression was strongly enhanced only in pancreas. In control experiments with adenoviruses in which the CAT gene was driven by the cytomegalovirus promoter, higher levels of expression were observed in all tissues. Expression was not modified after induction of acute pancreatitis. In conclusion, this study shows that (i) a recombinant adenovirus containing a fragment of the PAP I promoter allows specific targeting of a reporter gene to the mouse pancreas and (ii) expression of the reporter gene in pancreas is induced during acute pancreatitis. Adenovirus-mediated gene therapy of acute pancreatitis is therefore conceivable.


Acute-Phase Proteins/genetics , Antigens, Neoplasm , Biomarkers, Tumor , Lectins, C-Type , Lectins/genetics , Pancreatitis/genetics , Promoter Regions, Genetic , Proteins , Up-Regulation , Animals , Chloramphenicol O-Acetyltransferase/genetics , Genes, Reporter , Mice , Pancreas/chemistry , Pancreatitis/pathology , Pancreatitis-Associated Proteins , Sequence Deletion
17.
Int J Cancer ; 74(1): 35-44, 1997 Feb 20.
Article En | MEDLINE | ID: mdl-9036867

Defining the molecular mechanisms involved in cancer formation and progression is still a major challenge in colorectal-cancer research. Our strategy was to characterize genes whose expression is altered during colorectal carcinogenesis. To this end, the phenotype of a colorectal tumour was previously established by partial sequencing of a large number of its transcripts and the genes of interest were selected by differential screening on high-density filters with mRNA of colorectal cancer and normal adjacent mucosa. Fifty-one clones were found over-expressed and 23 were underexpressed in the colorectal-cancer tissues of the 5 analyzed patients. Among the latter, clones 6G2 and 32D6 were found of particular interest, since they had significant homology with several homeodomain-containing genes. The highest degree of similarity was with the murine Cdx1 for 6G2, and with the murine Cdx2 and hamster Cdx3 for 32D6. Using a RT-PCR approach, complete sequence of both types of homeobox-containing cDNA was obtained. The amino-acid sequence of the human Cdx1 is 85% identical to the mouse protein, and human Cdx2 has 94% identity with the mouse Cdx2 and hamster Cdx3. Tissue-distribution analysis of Cdx1 and Cdx2 mRNA showed that both transcripts were specifically expressed in small intestine, in colon and rectum. Twelve tissue samples from colorectal adenocarcinomas and the corresponding normal mucosa were analyzed by Northern blot. Expression of the 2 types of mRNA was either reduced or absent in 10 of them. Several colon-cancer cell lines were also analyzed. Cdx2 mRNA was absent from LS174T cells and Cdx1 mRNA was absent in PF11, TC7 and SW480 cells; none was detected in HT29 cells. It was concluded that decrease in human Cdx1 and/or Cdx2 expression is associated with colorectal tumorigenesis.


Avian Proteins , Colorectal Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/biosynthesis , Intestinal Mucosa/physiopathology , Adenocarcinoma , Adult , Amino Acid Sequence , Animals , Base Sequence , CDX2 Transcription Factor , Cell Line , Cloning, Molecular , Colonic Neoplasms , Colorectal Neoplasms/pathology , Cricetinae , DNA Primers , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Intestinal Mucosa/pathology , Male , Mice , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , Sequence Homology, Amino Acid , Trans-Activators , Transcription, Genetic , Tumor Cells, Cultured
18.
Br J Cancer ; 74(11): 1767-75, 1996 Dec.
Article En | MEDLINE | ID: mdl-8956791

Pancreatitis-associated protein I (PAP I) is a secretory protein first described as an acute phase reactant during acute pancreatitis. Recently, induction of the PAP I gene was also described in liver during hepatocarcinogenesis. To investigate the molecular mechanisms of this induction, we used constructs carrying progressive deletions of the PAP I promoter fused to the CAT gene. We showed that the silencer conferring tissue specificity on the PAP I gene was inactive in hepatoma cells. Then, in an vitro transcription system, we compared the transcription capacity of nuclear extracts from normal liver and HepG2 cells on constructs containing the silencer. The results confirmed that a trans-acting factor interacting with the PAP I silencer was present in liver cells and absent from hepatoma cells. On the other hand, immunohistochemistry showed that PAP I was expressed in a limited number of transformed hepatocytes. It was concluded that expression of PAP I in hepatocarcinoma occurred through inactivation of its silencer element and was not concomitant in all malignant cells. On that basis, we assayed PAP I in serum from patients with chronic hepatitis, liver cirrhosis or hepatocarcinoma. PAP I levels were normal in chronic active or persistent hepatitis, significantly higher in cirrhosis and strongly elevated in hepatocarcinoma. Because those clinical entities often develop in that sequence, serum PAP I appeared as a potential marker of hepatocarcinoma development.


Acute-Phase Proteins/metabolism , Antigens, Neoplasm , Biomarkers, Tumor , Carcinoma, Hepatocellular/blood , Hepatitis, Chronic/blood , Lectins, C-Type , Liver Cirrhosis/blood , Liver Neoplasms/blood , Liver/metabolism , Promoter Regions, Genetic/physiology , Acute-Phase Proteins/genetics , Adult , Animals , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA Footprinting , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Male , Middle Aged , Pancreatitis-Associated Proteins , Promoter Regions, Genetic/genetics , Rats , Sequence Deletion , Transcription, Genetic , Transcriptional Activation , Tumor Cells, Cultured
19.
Arch Biochem Biophys ; 330(1): 129-32, 1996 Jun 01.
Article En | MEDLINE | ID: mdl-8651687

During the acute phase of pancreatitis, expression of most pancreatic enzymes decreases, whereas mRNAs of pancreatitis associated protein and lithostathine/reg increase dramatically. In the present study we have investigated the effect of serum from rats with acute pancreatitis (SAP) and cytokines on the lithostathine/reg mRNA expression in AR-42J cells. Lithostathine/reg mRNA was strongly induced by SAP in a dose-dependent manner. Induction was abolished by preheating the SAP or by treating the cells with cycloheximide. Treatment with interleukins (IL) IL-1 or IL-6 or dexamethasone alone was ineffective. Combination of IL-1 with IL-6 was also ineffective. Combination of IL-6 with dexamethasone resulted in strong induction of the lithostathine/reg gene, but the further addition of IL-1 to the mixture reduced induction. Treatment with tumor necrosis factor-alpha (TNFalpha) or interferon-gamma (IFNgamma) induced lithostathine/reg mRNA expression. Combination of dexamethasone with TNFalpha or IFNgamma showed an inhibitory effect on lithostathine/reg mRNA expression. These findings suggest that expression of the lithostathine/reg mRNA during acute pancreatitis could be mediated by specific combinations of cytokines and/or glucocorticoids.


Calcium-Binding Proteins/biosynthesis , Cytokines/pharmacology , Nerve Tissue Proteins , Pancreas/metabolism , Pancreatitis/blood , Transcription, Genetic , Acute Disease , Animals , Cattle , Cell Line , Culture Media , Cycloheximide/pharmacology , Dexamethasone/pharmacology , Interleukin-1/pharmacology , Interleukin-6/pharmacology , Lithostathine , Pancreas/drug effects , Pancreatitis/chemically induced , Pancreatitis-Associated Proteins , Phosphoproteins/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Taurocholic Acid , Transcription, Genetic/drug effects
20.
Biochem J ; 311 ( Pt 2): 643-7, 1995 Oct 15.
Article En | MEDLINE | ID: mdl-7487908

We have previously characterized the rat pancreatitis-associated protein I (PAP I) gene by nucleotide sequencing. We describe in this paper its promoter region by analysing the regulatory functions associated with the DNA sequence comprising nt -1253 to + 10 of the gene. That sequence strongly promoted the transcription of the promotorless chloramphenicol acetyltranferase (CAT) gene in cells of pancreatic origin (AR-42J) but not in cells of non-pancreatic origin (Rat 2 and IEC 6). The influence on CAT expression of stepwise 5' deletions in the promoter sequence was monitored in the three cell lines. In pancreatic AR-42J cells, deletion down to position -926 did not affect significantly the expression of the reporter gene. Deletion to nt -685 caused about a 30% decrease in expression. Extending the deletion to nt -444 did not have any additional effect, but a further deletion to nt -180, resulted in a reduction to about 25%. Moreover, deletion from nt -180 to -118 resulted in a further reduction to about one-third of that. Finally, deletion down to nt -61 further reduced activity by a factor of 3, although it remained above background. These results suggest the presence of several positive cis-acting elements in the PAP I promoter. In non-pancreatic cells, CAT expression remained very low when the promoter was deleted down to nt -180. Yet, deletion from -180 to -118 significantly increased CAT expression, suggesting suppression of a negative cis-acting element. Further deletion down to nt -61 decreased CAT activity by a factor of 5. The region between nt -180 and -61 was subjected to footprint analysis. A similar pattern of DNase protection was obtained with AR-42J and Rat 2 nuclear extracts, the only protected region extending from nt -125 to -95. That region was further analysed by inserting the nt -180 to -81 fragment, in both orientations, upstream of thymidine kinase (TK) or simian virus 40 (SV40) promoter-CAT constructs. In all cases CAT expression was increased in pancreatic cells but reduced in Rat 2 cells. These results indicated the presence of cell-specific positive and negative elements within that region.


Acute-Phase Proteins/genetics , Antigens, Neoplasm , Biomarkers, Tumor , Gene Expression Regulation , Genes, Regulator/genetics , Lectins, C-Type , Transcription, Genetic/genetics , Acute-Phase Proteins/biosynthesis , Animals , Base Sequence , Cell Line , Cells, Cultured , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/genetics , DNA Primers/chemistry , Lectins/genetics , Molecular Sequence Data , Organ Specificity , Pancreas/cytology , Pancreas/metabolism , Pancreatitis-Associated Proteins , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , Rats , Regulatory Sequences, Nucleic Acid
...