Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Ann Oncol ; 35(9): 780-791, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38906254

ABSTRACT

BACKGROUND: After surgical resection of pancreatic ductal adenocarcinoma (PDAC), patients are predominantly treated with adjuvant chemotherapy, commonly consisting of gemcitabine (GEM)-based regimens or the modified FOLFIRINOX (mFFX) regimen. While mFFX regimen has been shown to be more effective than GEM-based regimens, it is also associated with higher toxicity. Current treatment decisions are based on patient performance status rather than on the molecular characteristics of the tumor. To address this gap, the goal of this study was to develop drug-specific transcriptomic signatures for personalized chemotherapy treatment. PATIENTS AND METHODS: We used PDAC datasets from preclinical models, encompassing chemotherapy response profiles for the mFFX regimen components. From them we identified specific gene transcripts associated with chemotherapy response. Three transcriptomic artificial intelligence signatures were obtained by combining independent component analysis and the least absolute shrinkage and selection operator-random forest approach. We integrated a previously developed GEM signature with three newly developed ones. The machine learning strategy employed to enhance these signatures incorporates transcriptomic features from the tumor microenvironment, leading to the development of the 'Pancreas-View' tool ultimately clinically validated in a cohort of 343 patients from the PRODIGE-24/CCTG PA6 trial. RESULTS: Patients who were predicted to be sensitive to the administered drugs (n = 164; 47.8%) had longer disease-free survival (DFS) than the other patients. The median DFS in the mFFX-sensitive group treated with mFFX was 50.0 months [stratified hazard ratio (HR) 0.31, 95% confidence interval (CI) 0.21-0.44, P < 0.001] and 33.7 months (stratified HR 0.40, 95% CI 0.17-0.59, P < 0.001) in the GEM-sensitive group when treated with GEM. Comparatively patients with signature predictions unmatched with the treatments (n = 86; 25.1%) or those resistant to all drugs (n = 93; 27.1%) had shorter DFS (10.6 and 10.8 months, respectively). CONCLUSIONS: This study presents a transcriptome-based tool that was developed using preclinical models and machine learning to accurately predict sensitivity to mFFX and GEM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Pancreatic Ductal , Irinotecan , Oxaliplatin , Pancreatic Neoplasms , Precision Medicine , Transcriptome , Humans , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Chemotherapy, Adjuvant/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Oxaliplatin/administration & dosage , Oxaliplatin/therapeutic use , Oxaliplatin/pharmacology , Male , Precision Medicine/methods , Irinotecan/administration & dosage , Irinotecan/therapeutic use , Irinotecan/pharmacology , Leucovorin/therapeutic use , Leucovorin/administration & dosage , Middle Aged , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Deoxycytidine/administration & dosage , Gemcitabine , Aged , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Artificial Intelligence , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects
2.
Ann Oncol ; 32(2): 250-260, 2021 02.
Article in English | MEDLINE | ID: mdl-33188873

ABSTRACT

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.


Subject(s)
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.
Front Pharmacol ; 15: 1432851, 2024.
Article in English | MEDLINE | ID: mdl-39114357

ABSTRACT

The xenobiotic transporter ABCC4/MRP4 is highly expressed in pancreatic ductal adenocarcinoma (PDAC) and correlates with a more aggressive phenotype and metastatic propensity. Here, we show that ABCC4 promotes epithelial-mesenchymal transition (EMT) in PDAC, a hallmark process involving the acquisition of mesenchymal traits by epithelial cells, enhanced cell motility, and chemoresistance. Modulation of ABCC4 levels in PANC-1 and BxPC-3 cell lines resulted in the dysregulation of genes present in the EMT signature. Bioinformatic analysis on several cohorts including tumor samples, primary patient-derived cultured cells, patient-derived xenografts, and cell lines, revealed a positive correlation between ABCC4 expression and EMT markers. We also characterized the ABCC4 cistrome and identified four candidate clusters in the distal promoter and intron one that showed differential binding of pro-epithelial FOXA1 and pro-mesenchymal GATA2 transcription factors in low ABCC4-expressing HPAF-II and high ABCC4-expressing PANC-1 xenografts. HPAF-II xenografts showed exclusive binding of FOXA1, and PANC-1 xenografts exclusive binding of GATA2, at ABCC4 clusters, consistent with their low and high EMT phenotype respectively. Our results underscore ABCC4/MRP4 as a valuable prognostic marker and a potential therapeutic target to treat PDAC subtypes with prominent EMT features, such as the basal-like/squamous subtype, characterized by worse prognosis and no effective therapies.

4.
Transl Oncol ; 16: 101315, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34906890

ABSTRACT

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.

5.
World J Surg ; 34(4): 830-5, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20145930

ABSTRACT

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.


Subject(s)
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
6.
Cell Death Dis ; 7: e2295, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27415425

ABSTRACT

Both clinical and experimental evidence have firmly established that chronic pancreatitis, in particular in the context of Kras oncogenic mutations, predisposes to pancreatic ductal adenocarcinoma (PDAC). However, the repertoire of molecular mediators of pancreatitis involved in Kras-mediated initiation of pancreatic carcinogenesis remains to be fully defined. In this study we demonstrate a novel role for vacuole membrane protein 1 (VMP1), a pancreatitis-associated protein critical for inducible autophagy, in the regulation of Kras-induced PDAC initiation. Using a newly developed genetically engineered model, we demonstrate that VMP1 increases the ability of Kras to give rise to preneoplastic lesions, pancreatic intraepithelial neoplasias (PanINs). This promoting effect of VMP1 on PanIN formation is due, at least in part, by an increase in cell proliferation combined with a decrease in apoptosis. Using chloroquine, an inhibitor of autophagy, we show that this drug antagonizes the effect of VMP1 on PanIN formation. Thus, we conclude that VMP1-mediated autophagy cooperate with Kras to promote PDAC initiation. These findings are of significant medical relevance, molecules targeting autophagy are currently being tested along chemotherapeutic agents to treat PDAC and other tumors in human trials.


Subject(s)
Carcinoma, Ductal/genetics , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatitis/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Autophagy/drug effects , Carcinoma, Ductal/etiology , Carcinoma, Ductal/metabolism , Carcinoma, Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chloroquine/pharmacology , Genes, Reporter , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis/complications , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis-Associated Proteins , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction
7.
Biochim Biophys Acta ; 1174(1): 99-102, 1993 Jul 18.
Article in English | MEDLINE | ID: mdl-7916640

ABSTRACT

The rat lithostathine gene was isolated from a genomic library using a rapid screening procedure involving PCR amplification. It was characterized over 2.7 kbp of gene sequence and 2.43 kbp of 5'-flanking sequence. The 5'-end of the coding sequence was determined by primer extension of lithostathine mRNA. The lithosathine sequence spanned over six exons. The promoter region of the gene contained the TATAAA and CCAAT consensus sequences 30 and 107 bp upstream of the cap site, respectively. Furthermore, a tract of (TG)22 repeat, with potential Z-DNA conformation, was found at position-1081.


Subject(s)
Calcium-Binding Proteins/genetics , Nerve Tissue Proteins , Animals , Base Sequence , Calcium-Binding Proteins/chemistry , Cloning, Molecular , Lithostathine , Molecular Conformation , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/analysis , Rats
8.
Biochim Biophys Acta ; 1261(2): 272-4, 1995 Apr 04.
Article in English | MEDLINE | ID: mdl-7711071

ABSTRACT

We have previously reported the coding sequence of the rat PAP II mRNA. We show in this paper the existence in rat pancreas of two forms of PAP II mRNA with identical coding sequence but a different 5'-untranslated region. We demonstrate that this is the result of a differential splicing.


Subject(s)
Antigens, Neoplasm , Biomarkers, Tumor , Lectins, C-Type , Pancreas/metabolism , Proteins/genetics , RNA, Messenger/analysis , Animals , Base Sequence , Molecular Sequence Data , Pancreatitis-Associated Proteins , Polymerase Chain Reaction , RNA Splicing , Rats
9.
Biochim Biophys Acta ; 1216(2): 329-31, 1993 Nov 16.
Article in English | MEDLINE | ID: mdl-8241280

ABSTRACT

A third member of the rat pancreatitis associated protein (PAP) gene family is described here. Its messenger RNA was cloned from an intestinal cDNA library and sequenced. The encoded protein, designated PAP III, shows 66% and 63% identity with the rat PAP I and II, respectively. The PAP III gene is constitutively expressed in the small intestine and in the pancreas with acute pancreatitis, but not in the healthy pancreas.


Subject(s)
Antigens, Neoplasm , Biomarkers, Tumor , Lectins, C-Type , Pancreatitis/genetics , Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/biosynthesis , DNA, Complementary/chemistry , Intestine, Small/metabolism , Molecular Sequence Data , Pancreas/metabolism , Pancreatitis-Associated Proteins , RNA, Messenger/analysis , Rats
10.
Cell Death Dis ; 6: e1592, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25590802

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a critical health issue in the field of cancer, with few therapeutic options. Evidence supports an implication of the intratumoral microenvironment (stroma) on PDA progression. However, its contribution to the role of neuroplastic changes within the pathophysiology and clinical course of PDA, through tumor recurrence and neuropathic pain, remains unknown, neglecting a putative, therapeutic window. Here, we report that the intratumoral microenvironment is a mediator of PDA-associated neural remodeling (PANR), and we highlight factors such as 'SLIT2' (an axon guidance molecule), which is expressed by cancer-associated fibroblasts (CAFs), that impact on neuroplastic changes in human PDA. We showed that 'CAF-secreted SLIT2' increases neurite outgrowth from dorsal root ganglia neurons as well as from Schwann cell migration/proliferation by modulating N-cadherin/ß-catenin signaling. Importantly, SLIT2/ROBO signaling inhibition disrupts this stromal/neural connection. Finally, we revealed that SLIT2 expression and CAFs are correlated with neural remodeling within human and mouse PDA. All together, our data demonstrate the implication of CAFs, through the secretion of axon guidance molecule, in PANR. Furthermore, it provides rationale to investigate the disruption of the stromal/neural compartment connection with SLIT2/ROBO inhibitors for the treatment of pancreatic cancer recurrence and pain.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Animals , Axons/drug effects , Axons/metabolism , Cadherins/metabolism , Cell Communication/drug effects , Cell Compartmentation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Culture Media/pharmacology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Nude , Models, Biological , Neurons/drug effects , Neurons/metabolism , Pancreatic Neoplasms/genetics , Schwann Cells/drug effects , Schwann Cells/metabolism , Schwann Cells/pathology , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Transcriptome/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , beta Catenin/metabolism , Pancreatic Neoplasms
11.
Cell Death Differ ; 22(1): 131-44, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25168244

ABSTRACT

Tribbles pseudokinase-3 (TRIB3) has been proposed to act as an inhibitor of AKT although the precise molecular basis of this activity and whether the loss of TRIB3 contributes to cancer initiation and progression remain to be clarified. In this study, by using a wide array of in vitro and in vivo approaches, including a Trib3 knockout mouse, we demonstrate that TRIB3 has a tumor-suppressing role. We also find that the mechanism by which TRIB3 loss enhances tumorigenesis relies on the dysregulation of the phosphorylation of AKT by the mTORC2 complex, which leads to an enhanced phosphorylation of AKT on Ser473 and the subsequent hyperphosphorylation and inactivation of the transcription factor FOXO3. These observations support the notion that loss of TRIB3 is associated with a more aggressive phenotype in various types of tumors by enhancing the activity of the mTORC2/AKT/FOXO axis.


Subject(s)
Cell Cycle Proteins/metabolism , Forkhead Transcription Factors/metabolism , Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Knockout , Mice, Nude , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Repressor Proteins/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics
12.
Eur J Cell Biol ; 80(2): 156-63, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11302520

ABSTRACT

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.


Subject(s)
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
13.
Pancreas ; 5(4): 421-6, 1990 Jul.
Article in English | MEDLINE | ID: mdl-1696380

ABSTRACT

Changes in pancreatic growth and in mRNA concentrations in rat pancreas were monitored by dot-blot hybridization with cloned cDNAs of rat amylase, chymotrypsinogen B, proinsulin I, and actin during the pre- and postnatal period in the rat. Wistar rats were killed at the 18th day of gestation and at the 1st, 10th, 20th, 35th, and 87th day of postnatal life. It was concluded from the ratio of pancreatic weight/body weight that pancreatic growth preceded body growth. Pancreatic protein and total RNA concentration increased 2.9 times during the period studies. All studied mRNAs increased in concentration during the postnatal development period. Messenger RNA for chymotrypsinogen B and proinsulin I exhibited a significant increase after birth, decreased by the 10th day of life, and increased thereafter. For amylase mRNA, no significant changes were observed around birth, a progressive increase occurring thereafter up to the 87th day of life. The mRNA for actin showed a progressive increase between the 18th day of gestation and the 20th postnatal day, after which it remained stable. We concluded that each mRNA showed a singular profile of increase during postnatal development.


Subject(s)
Actins/genetics , Amylases/genetics , Chymotrypsinogen/genetics , Gene Expression , Pancreas/growth & development , Proinsulin/genetics , Animals , Female , Gestational Age , Male , Nucleic Acid Hybridization , Pancreas/embryology , Pancreas/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Weight Gain
14.
Pancreas ; 8(5): 597-601, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8302796

ABSTRACT

Lithostathine is a pancreatic secretory protein which controls CaCO3 crystal growth in pancreatic juice. Trypsin hydrolysis of the molecule generates two fragments of 11 and 133 amino acids. The N-terminal undecapeptide bears the inhibitory activity for crystal growth. We demonstrate that the C-terminal part of the molecule, which is structurally related to Ca(2+)-dependent lectins, can induce bacterial aggregation. Ca(2+)- and pH-dependent aggregation was obtained for Escherichia coli strain KH 802 and 9 of 19 strains isolated from the predominant flora of human feces. Aggregation of E. coli could be reversed by dilution and bacteria could resume normal growth. Lithostathine is apparently the only component of normal pancreatic juice displaying such activity. Lithostathine is therefore a bifunctional protein which might be involved in the control of the bacterial ecosystem in the intestine.


Subject(s)
Calcium Carbonate/chemistry , Calcium-Binding Proteins/pharmacology , Escherichia coli/physiology , Nerve Tissue Proteins , Pancreatic Juice/chemistry , Animals , Bacterial Adhesion/drug effects , Calcium/pharmacology , Calcium-Binding Proteins/isolation & purification , Chromatography, High Pressure Liquid , Crystallization , Escherichia coli/drug effects , Escherichia coli/growth & development , Hemagglutination , Humans , Hydrogen-Ion Concentration , Lithostathine , Phosphoproteins/pharmacology , Rabbits , Rats , Trypsin/pharmacology
15.
Pancreas ; 6(2): 150-6, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1886883

ABSTRACT

Expression of the c-myc and H-ras oncogenes, and of several genes specifically expressed in adult rat pancreas was investigated by monitoring changes in corresponding mRNA concentrations, by dot-blot hybridization, during the early phase of regeneration following subtotal pancreatectomy. The oncogenes c-myc and H-ras were overexpressed after 12-24 and 48 h, respectively, then returned to basal levels. The concentrations of mRNAs encoding amylase, chymotrypsinogen B, and trypsinogen I decreased during the regeneration time. By contrast, proinsulin I mRNA concentration was increased at 12-48 h after surgical resection, and actin mRNA concentration was increased at 12-48 h after surgical resection, and actin mRNA concentration was increased at 12 h after subtotal pancreatectomy and remained elevated thereafter. We concluded that regeneration after subtotal pancreatectomy is accompanied by repression of certain genes that are expressed in differentiated pancreatic tissue, and that derepression of other genes may be necessary for starting and/or maintaining the process of pancreatic regeneration.


Subject(s)
Gene Expression Regulation/physiology , Genes, myc/genetics , Genes, ras/genetics , Pancreas/physiology , Regeneration/physiology , Animals , Collodion , DNA Probes , Immunoblotting , Male , Nucleic Acid Hybridization , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains
16.
Cell Death Differ ; 21(7): 1107-18, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24608790

ABSTRACT

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.


Subject(s)
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
17.
Cell Death Differ ; 19(9): 1525-35, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22421968

ABSTRACT

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.


Subject(s)
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
18.
Oncogene ; 30(27): 3049-61, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21339733

ABSTRACT

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.


Subject(s)
Carrier Proteins/physiology , Cell Movement/physiology , Heat-Shock Proteins/physiology , Osteonectin/metabolism , Pancreatic Neoplasms/pathology , Down-Regulation , Humans
19.
Oncogene ; 29(17): 2528-39, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20140013

ABSTRACT

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.


Subject(s)
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
20.
Cancer Lett ; 288(1): 116-23, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19631450

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL