Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
J Visc Surg ; 154(5): 329-333, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28844705

ABSTRACT

PURPOSE: To assess the K-ras gene mutation in the histologically negative venous margin of a pancreaticoduodenectomy (PD) specimen and its impact on survival. METHOD: From 2007 to 2010, 22 patients underwent R0 PD for resecable pancreatic adenocarcinoma. All specimens were stained and the portal vein (PV) bed was identified by blue ink; a 2mm3 sample (including the blue ink) was cut from a microscopic free-tumor block. DNA was extracted and assessed by quantitative real time polymerase chain reaction to detect the K-ras gene mutation. Twelve specimens (55%) (kras+ group) were identified with a K-ras mutation in the venous margin resection, and 10 specimens (kras- group) did not have K-ras mutation detected in the venous margin resection. RESULTS: The two groups were comparable. Overall 3years survival of patients of kras+ group versus patients of kras- group was 0 and 17% (P=0.03), respectively. Median survival time of patients of kras+ group versus patients of kras- group was 16months vs 25months (P=0.04; 95% confidence interval [1,11-1,88]), respectively. CONCLUSION: Genetic evaluation of venous resection margin affirmed unrecognized disease with strong impact on survival in more than 50% of patients with histologically R0 resection.


Subject(s)
Adenocarcinoma/surgery , Gene Expression Regulation , Margins of Excision , Pancreatic Neoplasms/surgery , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Cohort Studies , Disease-Free Survival , Female , Humans , Male , Mesenteric Veins/surgery , Middle Aged , Mutation/genetics , Neoplasm Grading , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreaticoduodenectomy/methods , Pancreaticoduodenectomy/mortality , Portal Vein/surgery , Prognosis , Retrospective Studies , Risk Assessment , Survival Analysis , Treatment Outcome
2.
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
3.
Cell Death Dis ; 7(6): e2269, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27336713

ABSTRACT

Sorafenib, an oral multikinase inhibitor, is the only approved agent for the treatment of advanced hepatocellular carcinoma (HCC). However, its benefits are modest, and as its mechanisms of action remain elusive, a better understanding of its anticancer effects is needed. Based on our previous study results, we investigated here the implication of the nuclear protein 1 (NUPR1) in HCC and its role in sorafenib treatment. NUPR1 is a stress-inducible protein that is overexpressed in various malignancies, but its role in HCC is not yet fully understood. We found that NUPR1 expression was significantly higher in primary human HCC samples than in the normal liver. Knockdown of NUPR1 significantly increased cell sensitivity to sorafenib and inhibited the cell growth, migration and invasion of HCC cells, both in vitro and in vivo. Moreover, NUPR1 silencing influenced the expression of RELB and IER3 genes. Unsurprisingly, RELB and IER3 knockdown also inhibited HCC cell viability, growth and migration. Using gene expression profiling of HCC cells following stable NUPR1 knockdown, we found that genes functionally involved in cell death and survival, cellular response to therapies, lipid metabolism, cell growth and proliferation, molecular transport and cellular movement were mostly suppressed. Network analysis of dynamic gene expression identified NF-κB and ERK as downregulated gene nodes, and several HCC-related oncogenes were also suppressed. We identified Runt-related transcription factor 2 (RUNX2) gene as a NUPR1-regulated gene and demonstrated that RUNX2 gene silencing inhibits HCC cell viability, growth, migration and increased cell sensitivity to sorafenib. We propose that the NUPR1/RELB/IER3/RUNX2 pathway has a pivotal role in hepatocarcinogenesis. The identification of the NUPR1/RELB/IER3/RUNX2 pathway as a potential therapeutic target may contribute to the development of new treatment strategies for HCC management.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement/drug effects , Drug Resistance, Neoplasm/drug effects , Liver Neoplasms/pathology , Molecular Targeted Therapy , Neoplasm Proteins/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Computational Biology , Core Binding Factor Alpha 1 Subunit/metabolism , Down-Regulation/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing/drug effects , Humans , Liver Neoplasms/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Niacinamide/pharmacology , RNA, Small Interfering/metabolism , Sorafenib , Transcription Factor RelB/genetics , Transcription Factor RelB/metabolism , Transcriptome/genetics , Young Adult
4.
Cell Death Differ ; 23(8): 1358-70, 2016 08.
Article in English | MEDLINE | ID: mdl-26943320

ABSTRACT

Advances made in pancreatic cancer therapy have been far from sufficient and have allowed only a slight improvement in global survival of patients with pancreatic ductal adenocarcinoma (PDA). Recent progresses in chemotherapy have offered some hope for an otherwise gloomy outlook, however, only a limited number of patients are eligible because of important cytotoxicity. In this context, enhancing our knowledge on PDA initiation and evolution is crucial to highlight certain weaknesses on which to specifically target therapy. We found that loss of transcriptionally active p73 (TAp73), a p53 family member, impacted PDA development. In two relevant and specific engineered pancreatic cancer mouse models, we observed that TAp73 deficiency reduced survival and enhanced epithelial-to-mesenchymal transition (EMT). Through proteomic analysis of conditioned media from TAp73 wild-type (WT) and deficient pancreatic tumor cells, we identified a secreted protein, biglycan (BGN), which is necessary and sufficient to mediate this pro-EMT effect. Interestingly, BGN is modulated by and modulates the transforming growth factor-ß (TGF-ß) pathway, a key regulator of the EMT process. We further examined this link and revealed that TAp73 impacts the TGF-ß pathway by direct regulation of BGN expression and Sma and Mad-related proteins (SMADs) expression/activity. Absence of TAp73 leads to activation of TGF-ß signaling through a SMAD-independent pathway, favoring oncogenic TGF-ß effects and EMT. Altogether, our data highlight the implication of TAp73 in the aggressiveness of pancreatic carcinogenesis through modulation of the TGF-ß signaling. By suggesting TAp73 as a predictive marker for response to TGF-ß inhibitors, our study could improve the classification of PDA patients with a view to offering combined therapy involving TGF-ß inhibitors.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Nuclear Proteins/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Biglycan/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition , Humans , Male , Mice , Mice, Knockout , Mice, Nude , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , RNA Interference , Signal Transduction/physiology , Survival Rate , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured
5.
Biochim Biophys Acta ; 1852(5): 962-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25638293

ABSTRACT

We recently reported that deletion of the stress-regulated nuclear protein 1 (Nupr1) protected against obesity-associated metabolic alterations due to increased beta cell mass, but complete Nupr1 ablation was not advantageous since it led to insulin resistance on a normal diet. The current study used Nupr1 haplodeficient mice to investigate whether a partial reduction in Nupr1 expression conferred beneficial effects on glucose homeostasis. Islet number, morphology and area, assessed by immunofluorescence and morphometric analyses, were not altered in Nupr1 haplodeficient mice under normal diet conditions and nor was beta cell BrdU incorporation. Glucose and insulin tolerance tests indicated that there were no significant changes in in vivo insulin secretion and glucose clearance in Nupr1 haplodeficient mice, and beta cell function in vitro was normal. However, reduced Nupr1 expression decreased visceral fat deposition and significantly increased insulin sensitivity in vivo. In contrast to wild type animals, high fat diet-fed Nupr1 haplodeficient mice were not hyperinsulinaemic or glucose intolerant, and their sustained insulin sensitivity was demonstrated by appropriate insulin-induced Akt phosphorylation, as determined by Western blotting. At the molecular level, measurements of gene expression levels and promoter activities identified Nupr1-dependent inhibition of heat shock factor-1-induced heat shock protein 70 (Hsp70) expression as a mechanism through which Nupr1 regulates insulin sensitivity. We have shown for the first time that Nupr1 plays a central role in inhibiting Hsp70 expression in tissues regulating glucose homeostasis, and reductions in Nupr1 expression could be used to protect against the metabolic defects associated with obesity-induced insulin resistance.


Subject(s)
DNA-Binding Proteins/genetics , Glucose Intolerance/genetics , HSP70 Heat-Shock Proteins/genetics , Insulin Resistance/genetics , Neoplasm Proteins/genetics , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/genetics , DNA-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Gene Expression , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , HSP70 Heat-Shock Proteins/metabolism , Immunohistochemistry , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/metabolism , Phosphorylation , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Up-Regulation
6.
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
7.
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
8.
Cell Death Differ ; 21(10): 1633-41, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24902898

ABSTRACT

Nuclear protein 1 (Nupr1), a small chromatin protein, has a critical role in cancer development, progression and resistance to therapy. Previously, we had demonstrated that Nupr1 cooperates with Kras(G12D) to induce pancreas intraepithelial neoplasias (PanIN) formation and pancreatic ductal adenocarcinoma development in mice. However, the molecular mechanisms by which Nupr1 influences Kras-mediated preneoplastic growth remain to be fully characterized. In the current study, we report evidence supporting a role for Nupr1 as a gene modifier of Kras(G12D)-induced senescence, which must be overcome to promote PanIN formation. We found that genetic inactivation of Nupr1 in mice impairs Kras-induced PanIN, leading to an increase in ß-galactosidase-positive cells and an upregulation of surrogate marker genes for senescence. More importantly, both of these cellular and molecular changes are recapitulated by the results of mechanistic experiments using RNAi-based inactivation of Nupr1 in human pancreatic cancer cell models. In addition, the senescent phenotype, which results from Nupr1 inactivation, is accompanied by activation of the FoxO3a-Skp2-p27(Kip1)-pRb-E2F pathway in vivo and in vitro. Thus, combined, these results show, for the first time, that Nupr1 aids oncogenic Kras to bypass senescence in a manner that cooperatively promotes PanIN formation. Besides its mechanistic importance, this new knowledge bears medical relevance as it delineates early pathobiological events that may be targeted in the future as a means to interfere with the formation of preneoplastic lesions early during pancreatic carcinogenesis.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cellular Senescence/genetics , DNA-Binding Proteins/genetics , Neoplasm Proteins/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Knockout , Proto-Oncogene Proteins p21(ras)/biosynthesis , RNA Interference , RNA, Small Interfering , S-Phase Kinase-Associated Proteins/biosynthesis , Up-Regulation , beta-Galactosidase/biosynthesis
9.
Cell Death Differ ; 20(2): 293-301, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22976836

ABSTRACT

The multiple isoforms of p73, a member of the p53 family, share the ability to modulate p53 activities but also have unique properties, leading to a complex and poorly understood functional network. In vivo, p73 isoforms have been implicated in tumor suppression (TAp73(-/-) mice), DNA damage (ΔNp73(-/-) mice) and development (p73(-/-) mice). In this study, we investigated whether TAp73 contributes to innate immunity and septic shock. In response to a lethal lipopolysaccharide (LPS) challenge, TAp73(-/-) mice showed higher blood levels of proinflammatory cytokines and greater mortality than their wild-type littermates. In vitro, TAp73(-/-) macrophages exhibited elevated production of tumor necrosis factor alpha , interleukin-6 and macrophage inflammatory protein-2 as well as prolonged survival, decreased phagocytosis and increased major histocompatibility complex class II expression. Mice depleted of endogenous macrophages and reconstituted with TAp73(-/-) macrophages showed increased sensitivity to LPS challenge. These results suggest that macrophage polarization is altered in the absence of TAp73 such that maintenance of the M1 effector phenotype is prolonged at the expense of the M2 phenotype, thus impairing resolution of the inflammatory response. Our data indicate that TAp73 has a role in macrophage polarization and innate immunity, enhancing the action field of this important regulatory molecule.


Subject(s)
DNA-Binding Proteins/metabolism , Immunity, Innate , Macrophages/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cells, Cultured , Chemokine CXCL2/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Interferon-beta/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phenotype , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Protein p73 , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
10.
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
11.
Oncogene ; 31(5): 664-70, 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-21725356

ABSTRACT

p53 is a tumor suppressor that responds to various stress signals by initiating cell-cycle arrest, senescence and apoptosis. Mutations of the p53 gene are found in over 50% of human tumors, highlighting the importance of p53 in tumor suppression. Numerous studies have reported on the interactions between p53, IGF-1-AKT and mTOR pathways as potentially explaining some of the tumor suppressive activities of p53. To further understand the basis of these interactions, we analyzed the involvement of DJ-1, an oncogene known to drive AKT-mediated cell survival, in the p53-AKT axis. In this study, we show that DJ-1 and p53 are tightly 'linked': p53 prevents the accumulation of DJ-1 protein, whereas loss of p53 leads to stabilization and enhancement of DJ-1 expression. Interestingly, this increase in DJ-1 level is only observed when p53 loss is accompanied by transformation of cells. Moreover, DJ-1 seems to be required for the enhanced activation of AKT observed in p53-deficient cells. Such observation confers a new property to DJ-1 associated to transforming-process to its oncogenic ability to drive AKT activation. We also show that DJ-1 is necessary for p53 activation following oxidative stress, suggesting the existence of a finely regulated loop between these two proteins in transformed cells. Finally, we demonstrate that in the absence of p53, DJ-1 is stabilized by ROS accumulation, and surprisingly seems to be required for this high intracellular ROS production. These data offer new insights into the regulation of DJ-1 and suggest that DJ-1 is a target of p53. Importantly, our study highlights that during transformation, DJ-1 is having a key role in the p53-regulated AKT pathway and p53-driven oxidative-stress response.


Subject(s)
Cell Transformation, Neoplastic , Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/deficiency , Up-Regulation , Animals , Blotting, Western , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins/genetics , Peroxiredoxins , Protein Deglycase DJ-1 , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics
12.
Ann Oncol ; 23(3): 570-576, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21810728

ABSTRACT

Although the treatment of pancreatic ductal adenocarcinoma (PDAC) remains a huge challenge, it is entering a new era with the development of new strategies and trial designs. Because there is an increasing number of novel therapeutic agents and potential combinations available to test in patients with PDAC, the identification of robust prognostic and predictive markers and of new targets and relevant pathways is a top priority as well as the design of adequate trials incorporating molecular-driven hypothesis. We presently report a consensus strategy for research in pancreatic cancer that was developed by a multidisciplinary panel of experts from different European institutions and collaborative groups involved in pancreatic cancer. The expert panel embraces the concept of exploratory early proof of concept studies, based on the prediction of response to novel agents and combinations, and randomised phase II studies permitting the selection of the best therapeutic approach to go forward into phase III, where the recommended primary end point remains overall survival. Trials should contain as many translational components as possible, relying on standardised tissue and blood processing and robust biobanking, and including dynamic imaging. Attention should not only be paid to the pancreatic cancer cells but also to microenvironmental factors and stem/stellate cells.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Research Design , Antineoplastic Agents/pharmacology , Europe , Humans , Randomized Controlled Trials as Topic , Research Design/standards , Research Design/trends
13.
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
14.
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
15.
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
16.
Oncogene ; 29(13): 1883-96, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20101233

ABSTRACT

One strategy to improve therapies in advanced prostate cancer (PC) involves targeting genes that are activated by androgen withdrawal to delay the emergence of the androgen-independent (AI) phenotype. Heat shock protein 27 (Hsp27) expression becomes highly upregulated in PC cells after androgen withdrawal or chemotherapy, in which it functions as a cytoprotective chaperone to confer broad-spectrum treatment resistance. The purpose of this study is to elucidate anti-apoptotic pathways regulated by Hsp27 that are activated during PC progression. Using two-hybrid experiment, we found that Hsp27 was having a major role in the protein translational initiation process. Furthermore, using complementary DNA (cDNA) microarray analysis, 4E binding protein 1 was identified as being proportionately and highly regulated by Hsp27. These data led us to analyze the protein synthesis initiation pathway, which is a prerequisite for cell growth and proliferation. Using northern and western blot analysis, we found that Hsp27 downregulation decreased eukaryotic translation initiation factor 4E (eIF4E) expression at the protein, but not mRNA, level. The cytoprotection afforded by Hsp27 overexpression was attenuated by eIF4E knockdown using specific eIF4E short interfering RNA (siRNA). Co-immunoprecipitation and co-immunofluorescence confirmed that Hsp27 colocalizes and interacts directly with eIF4E. Hsp27-eIF4E interaction decreases eIF4E ubiquitination and proteasomal degradation. By chaperoning eIF4E, Hsp27 seems to protect the protein synthesis initiation process to enhance cell survival during cell stress induced by castration or chemotherapy. Forced overexpression of eIF4E induces resistance to androgen-withdrawal and paclitaxel treatment in the prostate LNCaP cells in vitro. These findings identify Hsp27 as a modulator of eIF4E and establish a potential mechanism for the eIF4E-regulated apoptosis after androgen ablation and chemotherapy. Targeting Hsp27-eIF4E interaction may serve as a therapeutic target in advanced PC.


Subject(s)
Androgens/administration & dosage , HSP27 Heat-Shock Proteins/metabolism , Prostatic Neoplasms/metabolism , Androgen Antagonists/pharmacology , Androgens/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Disease Progression , Eukaryotic Initiation Factor-4E , HeLa Cells , Heat-Shock Proteins , Humans , Male , Molecular Chaperones/pharmacology , Prostatic Neoplasms/enzymology , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
17.
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
18.
Cell Mol Life Sci ; 66(13): 2195-204, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19434369

ABSTRACT

Pancreatitis-associated protein 1 (PAP1) belongs to the Reg family of secretory proteins. Several important biological roles have been attributed to PAP1 but the signaling pathways activated by this protein remain only partially understood. Here, we describe the intracellular pathways triggered by PAP1 in a pancreatic acinar cell line. Taking advantage of the fact that PAP1 induces its own transcription, we performed ChIP assays to analyze the recruitment of transcriptional factors on its promoter. Our results show that PAP1 increased the transactivation activity of pap1 and the binding on its promoter of the nuclear factors C/EBPbeta, P-CREB, P-ELK1, EGR1, STAT3, and ETS2, which are downstream targets of MAPK signaling. p44/42, p38, and JNK MAPKs activity increased after PAP1 treatment. In addition, pharmacological inhibition of these kinases markedly inhibited the induction of pap1 mRNA. Taken together, these results indicated that the mechanism of PAP1 action involves the activation of the MAPK superfamily.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Lectins, C-Type/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Animals , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Cell Line , Gene Expression Regulation, Enzymologic , Humans , Lectins, C-Type/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Pancreas/cytology , Pancreatitis-Associated Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Transcriptional Activation
19.
Cancer Gene Ther ; 16(9): 731-40, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19363471

ABSTRACT

A panel of kinases whose inhibition increased apoptosis of pancreatic adenocarcinoma cells in vitro was recently established. The aim of this work was to observe in a mouse xenograft model whether inhibition of these kinases would alter pancreatic tumor growth. Rate of apoptosis, caspase-3 activity and cell viability were assessed in two pancreatic cancer cell lines, MiaPaCa2 and BxPC3, after inhibiting selected kinases by transfection of specific siRNAs. For in vivo experiments, MiaPaCa2 cells were injected into the pancreas of nude mice, where they formed tumors. Inhibition of kinases was obtained by repeated intraperitoneal (i.p.) injections of modified O-Methyl (OMe) siRNAs specific for the selected kinases. Tumor volumes were assessed after 21 days. Among selected kinases, PAK7, MAP3K7 and CK2alpha were those whose inhibition increased apoptosis the most in vitro. Simultaneous inhibition of two of them increased apoptosis up to five times. Moreover, inhibiting these kinases had little effect on 10 non-pancreatic cell lines, suggesting pancreatic specificity. In vivo, OMe-siRNAs induced significant but incomplete inhibition of kinase expression (45-75%). Nevertheless, such inhibition resulted in a twofold increase in caspase-3 activity in tumors and a strong reduction in tumor volume (about 75%). In vivo inhibition by OMe-siRNAs of three survival kinases apparently specific for pancreatic cancer cells, PAK7, MAP3K7 and CK2alpha, decreases significantly the growth of xenografted MiaPaCa2 cells. This strategy is therefore of potential clinical interest.


Subject(s)
Casein Kinase II/genetics , Genetic Therapy , MAP Kinase Kinase Kinases/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , RNA, Small Interfering/genetics , p21-Activated Kinases/genetics , Animals , Apoptosis , Casein Kinase II/metabolism , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Nude , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , Xenograft Model Antitumor Assays , p21-Activated Kinases/metabolism
20.
Gut ; 54(9): 1244-53, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15870231

ABSTRACT

BACKGROUND AND AIMS: Increased pancreatitis associated protein (PAP) mRNA has been reported in active inflammatory bowel disease (IBD). The aims of the current study were to characterise PAP production in IBD and the effects of PAP on inflammation. PATIENTS AND METHODS: Serum PAP levels were determined in healthy controls (n = 29), inflammatory controls (n = 14), and IBD patients (n = 171). Ex vivo PAP secretion in intestinal tissue was measured in 56 IBD patients and 13 healthy controls. Cellular origin of PAP was determined by immunohistochemistry. The effects of exogenous PAP on nuclear factor kappaB (NFkappaB) activation, proinflammatory cytokine production, and endothelial adhesion molecule expression were also analysed ex vivo. RESULTS: Patients with active IBD had increased serum PAP levels compared with controls, and these levels correlated with clinical and endoscopic disease severity. Ex vivo intestinal PAP synthesis was increased in active IBD and correlated with endoscopic and histological severity of inflammatory lesions. PAP localised to colonic Paneth cells. Incubation of mucosa from active Crohn's disease with PAP dose dependently reduced proinflammatory cytokines secretion. PAP prevented TNF-alpha induced NFkappaB activation in monocytic, epithelial, and endothelial cells and reduced proinflammatory cytokine mRNA levels and adhesion molecule expression. CONCLUSIONS: PAP is synthesised by Paneth cells and is overexpressed in colonic tissue of active IBD. PAP inhibits NFkappaB activation and downregulates cytokine production and adhesion molecule expression in inflamed tissue. It may represent an anti-inflammatory mechanism and new therapeutic strategy in IBD.


Subject(s)
Antigens, Neoplasm/blood , Biomarkers, Tumor/blood , Inflammatory Bowel Diseases/blood , Lectins, C-Type/blood , Analysis of Variance , Antigens, Neoplasm/pharmacology , Biological Transport , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/pharmacology , Case-Control Studies , Cell Adhesion Molecules/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Colitis/blood , Colitis/immunology , Colitis/pathology , Colitis, Ulcerative/blood , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Crohn Disease/blood , Crohn Disease/immunology , Crohn Disease/pathology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukins/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , NF-kappa B/metabolism , Pancreatitis-Associated Proteins , Paneth Cells/metabolism , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL