Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
BMC Cancer ; 23(1): 966, 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37828434

ABSTRACT

BACKGROUND: In case of locally advanced and/or non-metastatic unresectable esophageal cancer, definitive chemoradiotherapy (CRT) delivering 50 Gy in 25 daily fractions in combination with platinum-based regimen remains the standard of care resulting in a 2-year disease-free survival of 25% which deserves to be associated with new systemic strategies. In recent years, several immune checkpoint inhibitors (anti-PD1/anti-PD-L1, anti-Program-Death 1/anti-Program-Death ligand 1) have been approved for the treatment of various solid malignancies including metastatic esophageal cancer. As such, we hypothesized that the addition of an anti-PD-L1 to CRT would provide clinical benefit for patients with locally advanced oesophageal cancer. To assess the efficacy of the anti-PD-L1 durvalumab in combination with CRT and then as maintenance therapy we designed the randomized phase II ARION (Association of Radiochemotherapy with Immunotherapy in unresectable Oesophageal carciNoma- UCGI 33/PRODIGE 67). METHODS: ARION is a multicenter, open-label, randomized, comparative phase II trial. Patients are randomly assigned in a 1:1 ratio in each arm with a stratification according to tumor stage, histology and centre. Experimental arm relies on CRT with 50 Gy in 25 daily fractions in combination with FOLFOX regimen administrated during and after radiotherapy every two weeks for a total of 6 cycles and durvalumab starting with CRT for a total of 12 infusions. Standard arm is CRT alone. Use of Intensity Modulated radiotherapy is mandatory. The primary endpoint is to increase progression-free survival at 12 months from 50 to 68% (HR = 0.55) (power 90%; one-sided alpha-risk, 10%). Progression will be defined with central external review of imaging. ANCILLARY STUDIES ARE PLANNED: PD-L1 Combined Positivity Score on carcinoma cells and stromal immune cells of diagnostic biopsy specimen will be correlated to disease free survival. The study of gut microbiota will aim to determine if baseline intestinal bacteria correlates with tumor response. Proteomic analysis on blood samples will compare long-term responder after CRT with durvalumab to non-responder to identify biomarkers. CONCLUSION: Results of the present study will be of great importance to evaluate the impact of immunotherapy in combination with CRT and decipher immune response in this unmet need clinical situation. TRIAL REGISTRATION: ClinicalTrials.gov, NCT: 03777813.Trial registration date: 5th December 2018.


Subject(s)
Carcinoma , Esophageal Neoplasms , Humans , Proteomics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy , Esophageal Neoplasms/therapy , Immunotherapy , Randomized Controlled Trials as Topic , Multicenter Studies as Topic , Clinical Trials, Phase II as Topic
2.
Cancers (Basel) ; 16(1)2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38201456

ABSTRACT

GSCs play an important role in GBM recurrence. Understanding the resistance mechanisms in these cells is therefore crucial for radiation therapy optimization. In this study, using patient-derived GSCs, we demonstrate that GDF15, a cytokine belonging to the TGF-ß superfamily, is regulated by irradiation (IR) and the transcription factor WWTR1/TAZ. Blocking WWTR1/TAZ using specific siRNAs significantly reduces GDF15 basal expression and reverses the upregulation of this cytokine induced by IR. Furthermore, we demonstrate that GDF15 plays an important role in GSC radioresistance. Targeting GDF15 expression by siRNA in GSCs expressing high levels of GDF15 sensitizes the cells to IR. In addition, we also found that GDF15 expression is critical for GSC spheroid formation, as GDF15 knockdown significantly reduces the number of GSC neurospheres. This study suggests that GDF15 targeting in combination with radiotherapy may be a feasible approach in patients with GBM.

3.
Cells ; 11(14)2022 07 06.
Article in English | MEDLINE | ID: mdl-35883571

ABSTRACT

Systematic recurrence of glioblastoma (GB) despite surgery and chemo-radiotherapy is due to GB stem cells (GBSC), which are particularly invasive and radioresistant. Therefore, there is a need to identify new factors that might be targeted to decrease GBSC invasive capabilities as well as radioresistance. Patient-derived GBSC were used in this study to demonstrate a higher expression of the glycoprotein M6a (GPM6A) in invasive GBSC compared to non-invasive cells. In 3D invasion assays performed on primary neurospheres of GBSC, we showed that blocking GPM6A expression by siRNA significantly reduced cell invasion. We also demonstrated a high correlation of GPM6A with the oncogenic protein tyrosine phosphatase, PTPRZ1, which regulates GPM6A expression and cell invasion. The results of our study also show that GPM6A and PTPRZ1 are crucial for GBSC sphere formation. Finally, we demonstrated that targeting GPM6A or PTPRZ1 in GBSC increases the radiosensitivity of GBSC. Our results suggest that blocking GPM6A or PTPRZ1 could represent an interesting approach in the treatment of glioblastoma since it would simultaneously target proliferation, invasion, and radioresistance.


Subject(s)
Glioblastoma , Membrane Glycoproteins , Nerve Tissue Proteins , Glioblastoma/genetics , Glioblastoma/radiotherapy , Humans , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/radiation effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/radiation effects , Radiation Tolerance , Receptor-Like Protein Tyrosine Phosphatases, Class 5
4.
Cancers (Basel) ; 13(1)2020 Dec 25.
Article in English | MEDLINE | ID: mdl-33375621

ABSTRACT

Recurrence of GBM is thought to be due to GBMSCs, which are particularly chemo-radioresistant and characterized by a high capacity to invade normal brain. Evidence is emerging that modulation of m6A RNA methylation plays an important role in tumor progression. However, the impact of this mRNA modification in GBM is poorly studied. We used patient-derived GBMSCs to demonstrate that high expression of the RNA demethylase, ALKBH5, increases radioresistance by regulating homologous recombination (HR). In cells downregulated for ALKBH5, we observed a decrease in GBMSC survival after irradiation likely due to a defect in DNA-damage repair. Indeed, we observed a decrease in the expression of several genes involved in the HR, including CHK1 and RAD51, as well as a persistence of γ-H2AX staining after IR. We also demonstrated in this study that ALKBH5 contributes to the aggressiveness of GBM by favoring the invasion of GBMSCs. Indeed, GBMSCs deficient for ALKBH5 exhibited a significant reduced invasion capability relative to control cells. Our data suggest that ALKBH5 is an attractive therapeutic target to overcome radioresistance and invasiveness of GBMSCs.

5.
NAR Cancer ; 2(3): zcaa020, 2020 Sep.
Article in English | MEDLINE | ID: mdl-34316689

ABSTRACT

Intrinsic resistance to current therapies, leading to dismal clinical outcomes, is a hallmark of glioblastoma multiforme (GBM), the most common and aggressive brain tumor. Understanding the underlying mechanisms of such malignancy is, therefore, an urgent medical need. Deregulation of the protein translation machinery has been shown to contribute to cancer initiation and progression, in part by driving selective translational control of specific mRNA transcripts involved in distinct cancer cell behaviors. Here, we focus on eIF3, a multimeric complex with a known role in the initiation of translation and that is frequently deregulated in cancer. Our results show that the deregulated expression of eIF3e, the e subunit of eIF3, in specific GBM regions could impinge on selective protein synthesis impacting the GBM outcome. In particular, eIF3e restricts the expression of proteins involved in the response to cellular stress and increases the expression of key functional regulators of cell stemness. Such a translation program can therefore serve as a double-edged sword promoting GBM tumor growth and resistance to radiation.

6.
Cancers (Basel) ; 11(3)2019 Mar 22.
Article in English | MEDLINE | ID: mdl-30909436

ABSTRACT

Glioblastoma (GBM) is the most lethal primary brain tumor in adults and is known to be particularly aggressive and resistant to anti-cancer therapies, mainly due to the presence of GBM stem cells (GBMSC). By in vitro approaches supported by analysis from patients' databases, we determined how α6-integrin and Fibroblast Growth Factor Receptor 1 (FGFR1) work in concert to regulate proliferation and stemness of GBMSC. We showed that α6-integrin regulates the expression of FGFR1 and its target gene Fokhead Box M1 (FOXM1) via the ZEB1/YAP1 transcription complex. These results were in accordance with the positive correlation observed in GBM between α6-integrin expression and its target genes ZEB1/YAP1, FGFR1, and FOXM1 in the databases, TCGA and Rembrandt. In addition, the clinical data demonstrate that GBM patients with high levels of the five genes signature, including α6-integrin, ZEB1/YAP1, FGFR1 and FOXM1, have a significantly shorter overall survival. In vitro, we observed a similar decrease in the expression of stemness-related factors, neurospheres forming capacity, as well as spheroids growth when α6-integrin or FGFR1 was blocked individually with specific siRNA, whereas the combination of both siRNA led to a significantly higher inhibition of spheres formation. These data suggest that co-administration of anti-FGFR1 and anti-α6-integrin could provide an improved therapeutic response in GBMSC.

7.
Cell Death Dis ; 9(9): 872, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30158599

ABSTRACT

Radiotherapy is the cornerstone of glioblastoma (GBM) standard treatment. However, radioresistance of cancer cells leads to an inevitable recurrence. In the present study, we showed that blocking α6-integrin in cells derived from GBM biopsy specimens cultured as neurospheres, sensitized cells to radiation. In cells downregulated for α6-integrin expression, we observed a decrease in cell survival after irradiation and an increase in radio-induced cell death. We also demonstrated that inhibition of α6-integrin expression affects DNA damage checkpoint and repair. Indeed, we observed a persistence of γ-H2AX staining after IR and the abrogation of the DNA damage-induced G2/M checkpoint, likely through the downregulation of the checkpoint kinase CHK1 and its downstream target Cdc25c. We also showed that α6-integrin contributes to GBM radioresistance by controlling the expression of the transcriptional network ZEB1/OLIG2/SOX2. Finally, the clinical data from TCGA and Rembrandt databases demonstrate that GBM patients with high levels of the five genes signature, including α6-integrin and its targets, CHK1, ZEB1, OLIG2 and SOX2, have a significantly shorter overall survival. Our study suggest that α6-integrin is an attractive therapeutic target to overcome radioresistance of GBM cancer cells.


Subject(s)
Brain Neoplasms/genetics , DNA Damage/genetics , Glioblastoma/genetics , Integrin alpha Chains/genetics , Radiation Tolerance/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Cell Division/genetics , Cell Line, Tumor , Cell Survival/genetics , Checkpoint Kinase 1/genetics , Down-Regulation/genetics , G2 Phase/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Oligodendrocyte Transcription Factor 2/genetics , SOXB1 Transcription Factors/genetics , Transcription, Genetic/genetics , cdc25 Phosphatases/genetics
8.
Exp Cell Res ; 362(2): 498-503, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29253536

ABSTRACT

Peritoneal metastasis is a major cause of recurrence of gastric cancer and integrins are key molecules involved in gastric cancer cells attachment to the peritoneum. The peptide hormone, gastrin, initially identified for its role in gastric acid secretion is also a growth factor for gastric mucosa. Gastrin has also been shown to contribute to gastric cancers progression. Here, we provide the first evidence that gastrin increases the adhesion of gastric cancer cells. Gastrin treatment induces the expression of α2 integrin subunit through a mechanism that involves the ERK pathway. We also observed in response to gastrin an increase in the amount of α2 integrin associated with ß1subunit. In addition, gastrin-stimulated cell adhesion was blocked with an anti-α2ß1 integrin neutralizing antibody. We also show that gastrin activates the integrin pathway via the phosphorylation of ß1 integrin by a Src family kinase. This mechanism may contribute to the enhancement of cell adhesion observed in response to gastrin since we found an inhibition of gastrin-mediated cell adhesion when cells were treated with a Src inhibitor. By regulating one of the key step of the metastatic process gastrin might contribute to increase the aggressive behaviour of human gastric tumours.


Subject(s)
Gastrins/pharmacology , Integrin alpha2beta1/genetics , Peritoneal Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Cell Adhesion/drug effects , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Neoplasm Metastasis , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/secondary , Peritoneum/pathology , Phosphorylation/drug effects , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
9.
Oncotarget ; 8(35): 58587-58600, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28938581

ABSTRACT

A high percentage of advanced rectal cancers are resistant to radiation. Therefore, increasing the efficacy of radiotherapy by targeting factors involved in radioresistance seems to be an attractive strategy. Here we demonstrated that the pro-hormone progastrin (PG), known to be over-expressed in CRC, and recognized as a pro-oncogenic factor, is a radioresistance factor that can be targeted to sensitize resistant rectal cancers to radiations. First, we observed an increase in PG mRNA expression under irradiation. Our results also demonstrated that down-regulating PG mRNA expression using a shRNA strategy, significantly increases the sensitivity to irradiation (IR) in a clonogenic assay of different colorectal cancer cell lines. We also showed that the combination of PG gene down-regulation and IR strongly inhibits tumours progression in vivo. Then, we demonstrated that targeting PG gene radiosensitizes cancer cells by increasing radio-induced apoptosis shown by an increase in annexin V positive cells, caspases activation and PARP cleavage. We also observed the up-regulation of the pro-apoptotic pathway, JNK and the induction of the expression of pro-apoptotic factors such as BIM. In addition, we demonstrated in this study that inhibition of PG gene expression enhances radiation-induced DNA damage. Our data also suggest that, in addition to increase radio-induced apoptosis, targeting PG gene also leads to the inhibition of the survival pathways, AKT and ERK induced by IR. Taken together, our results highlight the role of PG in radioresistance and provide a preclinical proof of concept that PG represents an attractive target for sensitizing resistant rectal tumours to irradiation. .

10.
BMC Cancer ; 13: 531, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24209454

ABSTRACT

BACKGROUND: In contrast to sessile serrated adenomas and traditional serrated adenomas which are associated with a significant cancer risk, the role of hyperplastic polyps (HP) in colorectal carcinogenesis as well as the molecular mechanisms underlying their development remain controversial and still need to be clarified. Several reports suggest that a subset of HP may represent precursor lesions of some colorectal cancers. However, biomarkers are needed to identify the subset of HP that may have a malignant potential. The hormone precursor, progastrin (PG) has been involved in colon carcinogenesis and is known to activate pro-oncogenic pathways such as the ERK or the STAT3 pathway. We therefore analyzed PG expression and the activation of these signaling factors in HP. METHODS: We retrospectively analyzed PG expression as well as the phosphorylation of ERK and STAT3 by immunohistochemistry in HP from 48 patients. RESULTS: Mean percentages of epithelial cells positive for PG or phospho-ERK were respectively, 31% and 33% in HP and were significantly higher in these lesions compared to normal colon (3%, p=0.0021 and 7%, p=0.0008, respectively). We found a significant correlation between PG and phospho-ERK expression in HP with ERK activation significantly stronger in lesions with high progastrin expression (p=0.015). In contrast, STAT3 was not significantly activated in HP compared to normal colon and we did not observe a significant correlation with PG expression. CONCLUSIONS: HP overexpressing PG that have the highest activation of the ERK pathway might reflect less latent lesions that might have a malignant potential.


Subject(s)
Colonic Polyps/metabolism , Oncogene Proteins/metabolism , Signal Transduction , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Adult , Aged , Aged, 80 and over , Colonic Polyps/genetics , Colonic Polyps/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gastrins/genetics , Gastrins/metabolism , Humans , Hyperplasia , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , Oncogene Proteins/genetics , Protein Precursors/genetics , Protein Precursors/metabolism , Retrospective Studies , Risk Factors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
11.
J Biol Chem ; 287(49): 41458-68, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23055519

ABSTRACT

F1 domain of F(1)F(o)-ATPase was initially believed to be strictly expressed in the mitochondrial membrane. Interestingly, recent reports have shown that the F1 complex can serve as a cell surface receptor for apparently unrelated ligands. Here we show for the first time the presence of the F(1)-ATPase at the cell surface of normal or cancerous colonic epithelial cells. Using surface plasmon resonance technology and mass spectrometry, we identified a peptide hormone product of the gastrin gene (glycine-extended gastrin (G-gly)) as a new ligand for the F(1)-ATPase. By molecular modeling, we identified the motif in the peptide sequence (E(E/D)XY), that directly interacts with the F(1)-ATPase and the amino acids in the F(1)-ATPase that bind this motif. Replacement of the Glu-9 residue by an alanine in the E(E/D)XY motif resulted in a strong decrease of G-gly binding to the F(1)-ATPase and the loss of its biological activity. In addition we demonstrated that F(1)-ATPase mediates the growth effects of the peptide. Indeed, blocking F(1)-ATPase activity decreases G-gly-induced cell growth. The mechanism likely involves ADP production by the membrane F(1)-ATPase, which is induced by G-gly. These results suggest an important contribution of cell surface F(1)-ATPase in the pro-proliferative action of this gastrointestinal peptide.


Subject(s)
Cell Membrane/enzymology , Colon/enzymology , Epithelial Cells/metabolism , Proton-Translocating ATPases/chemistry , Adenosine Diphosphate/chemistry , Amino Acid Sequence , Animals , Caco-2 Cells , Catalytic Domain , Cattle , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Endothelial Cells/cytology , Humans , Mitochondria/metabolism , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Sequence Homology, Amino Acid , Surface Plasmon Resonance
12.
Cancer Prev Res (Phila) ; 5(4): 675-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22366915

ABSTRACT

The most frequently occurring lesions in the colon are the hyperplastic polyps. Hyperplastic polyps have long been considered as lesions with no malignant potential and colonoscopy for these patients is not recommended. However, recent works suggest that hyperplastic polyps may represent precursor lesions of some sporadic colorectal cancers. Until now, no biomarker allows to identify the subset of hyperplastic polyps that may have a malignant potential. Because the hormone precursor progastrin has been involved in colon carcinogenesis, we investigated whether its expression in hyperplastic polyps predicts the occurrence of colonic neoplasm after resection of hyperplastic polyps. We retrospectively analyzed progastrin expression in hyperplastic polyps from 74 patients without history of colorectal pathology. In our study, 41% of patients presenting an initial hyperplastic polyp subsequently developed adenomatous polyps, recognized as precursor lesions for colorectal adenocarcinomas. Progastrin was overexpressed in the hyperplastic polyps in 40% of the patients. We showed a significant association between progastrin overexpression and shortened neoplasm-free survival (P = 0.001). Patients with high overexpression of progastrin had a 5-year neoplasm-free survival rate of 38% as compared with 100% for the patients with low progastrin expression. In addition, we established a predictive test on the basis of progastrin staining and patients' age that predicts occurrence of neoplasm after developing a first hyperplastic polyp with a sensitivity of 100% [95% confidence interval (CI), 79%-100%] and a specificity of 74% (51%-90%). We show that progastrin expression evaluation in hyperplastic polyps is an efficient prognostic tool to determine patients with higher risk of metachronous neoplasms who could benefit from an adapted follow-up.


Subject(s)
Biomarkers, Tumor/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Polyps/metabolism , Gastrins/biosynthesis , Protein Precursors/biosynthesis , Adult , Aged , Aged, 80 and over , Cohort Studies , Disease-Free Survival , Female , Humans , Immunohistochemistry , Male , Middle Aged , ROC Curve , Reproducibility of Results
13.
World J Gastroenterol ; 17(40): 4488-95, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22110279

ABSTRACT

AIM: To analyse αv integrin expression induced by gastrin in pancreatic cancer models. METHODS: αv integrin mRNA expression in human pancreatic cancer cells was analysed using a "cancer genes" array and confirmed by real-time reverse transcription-polymerase chain reaction (PCR). Western blotting and semi-quantitative immunohistochemistry were used to examine protein levels in human pancreatic cancer cell lines and pancreatic tissues, respectively. The role of αv integrin on gastrin-induced cell adhesion was examined using blocking anti-αv integrin monoclonal antibodies. Adherent cells were quantified by staining with crystal violet. RESULTS: Using a "cancer genes" array we identified αv integrin as a new gastrin target gene in human pancreatic cancer cells. A quantitative real-time PCR approach was used to confirm αv integrin gene expression. We also demonstrate that Src family kinases and the PI 3-kinase, two signalling pathways specifically activated by the CCK-2 receptor (CCK2R), are involved in gastrin-mediated αv integrin expression. In contrast, inhibition of the ERK pathway was without any effect on αv integrin expression induced by gastrin. Our results also show that gastrin modulates cell adhesion via αv integrins. Indeed, in vitro adhesion assays performed on fibronectin show that gastrin significantly increases adhesion of pancreatic cancer cells. The use of blocking anti-αv integrin monoclonal antibodies completely reversed the increase in cell-substrate adhesion induced by gastrin. In addition, we showed in vivo that the targeted CCK2R expression in the pancreas of Elas-CCK2 mice, leads to the overexpression of αv integrin. This process may contribute to pancreatic tumour development observed in these transgenic animals. CONCLUSION: αv integrin is a new gastrin target in pancreatic cancer models and contributes to gastrin effects on cell adhesion.


Subject(s)
Gastrins/metabolism , Integrin alphaV/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Gastrins/genetics , Gene Expression Regulation, Neoplastic , Humans , Integrin alphaV/genetics , Mice , Mice, Transgenic , Microarray Analysis/methods , Phosphatidylinositol 3-Kinases/metabolism , Real-Time Polymerase Chain Reaction/methods , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Signal Transduction/physiology , src-Family Kinases/metabolism
14.
Eur J Pain ; 15(2): 169.e1-169.e12, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20594879

ABSTRACT

The N-methyl-d-aspartate receptor (NMDAR) contributes to central sensitization in the spinal cord, a phenomenon which comprises various pathophysiological mechanisms responsible for neuropathic pain-like signs in animal models. NMDAR function is modulated by post-translational modifications including phosphorylation, and this is proposed to underlie its involvement in the production of pain hypersensitivity. As in diabetic patients, streptozotocin-induced diabetic rats exhibit or not somatic mechanical hyperalgesia; these rats were named DH and DNH respectively. At three weeks of diabetes, we present evidence that somatic mechanical hyperalgesia was correlated with an enhanced phosphorylation of the NMDAR NR1 subunit (pNR1) in the rat spinal cord. This increase was not found in normal and DNH rats, suggesting that this regulation was specific to hyperalgesia. Double immunofluorescence studies revealed that the numbers of pNR1-immunoreactive neurons and microglial cells were significantly increased in all laminae (I-II and III-VI) of the dorsal horn from DH animals. Western-blots analysis showed no change in NR1 protein levels, whatever the behavioural and glycemic status of the animals. Chronic intrathecal treatment (5µg/rat/day for 7days) by U0126 and MK801, which blocked MEK (an upstream kinase of extracellular signal-regulated protein kinase: ERK) and the NMDAR respectively, simultaneously suppressed somatic mechanical hyperalgesia developed by diabetic rats and decreased pNR1. These results indicate for the first time that increased expression of pNR1 is regulated by ERK and the NMDAR via a feedforward mechanism in spinal neurons and microglia and represents one mechanism involved in central sensitization and somatic mechanical hyperalgesia after diabetes.


Subject(s)
Diabetic Neuropathies/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Microglia/metabolism , Phosphorylation/physiology , Posterior Horn Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/metabolism , Analysis of Variance , Animals , Blotting, Western , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Enzyme Inhibitors/pharmacology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Immunohistochemistry , Male , Microglia/drug effects , Phosphorylation/drug effects , Posterior Horn Cells/drug effects , Posterior Horn Cells/physiopathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/drug effects , Spinal Cord/physiopathology
15.
Int J Cancer ; 126(12): 2847-57, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-19876923

ABSTRACT

One of the major angiogenic factor released by tumor cells is VEGF. Its high expression is correlated with poor prognosis in colorectal tumors. In colon cancer, gastrin gene expression is also upregulated. In these tumors, gastrin precursors are mainly produced and act as growth factors. Recently, a study has also shown that the gastrin precursor, G-gly induced in vitro tubules formation by vascular endothelial cells suggesting a potential proangiogenic role. Here, we demonstrate that stimulation of human colorectal cancer cell lines with G-gly increases the expression of the proangiogenic factor VEGF at the mRNA and protein levels. In addition, blocking the progastrin autocrine loop leads to a downregulation of VEGF. Although HIF-1 is a major transcriptional activator for VEGF our results suggest an alternative mechanism for VEGF regulation in normoxic conditions, independent of HIF-1 that involves the PI3K/AKT pathway. Indeed we show that G-gly does not lead to HIF-1 accumulation in colon cancer cells. Moreover, we found that G-gly activates the PI3K/AKT pathway and inhibition of this pathway reverses the effects of G-gly observed on VEGF mRNA and protein levels. In correlation with these results, we observed in vivo, on colon tissue sections from transgenic mice overexpressing G-gly, an increase in VEGF expression in absence of HIF-1 accumulation. In conclusion, our study demonstrates that gastrin precursors, known to promote colon epithelial cells proliferation and survival can also contribute to the angiogenesis process by stimulating the expression of the proangiogenic factor VEGF via the PI3K pathway and independently of hypoxia conditions.


Subject(s)
Colon/metabolism , Colonic Neoplasms/metabolism , Gastrins/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Blotting, Western , Colonic Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoenzyme Techniques , Mice , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Up-Regulation
16.
Endocrinology ; 148(3): 1039-49, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17122076

ABSTRACT

The role of cholecystokinin (CCK) as a satiety factor has been extensively documented. Although most work implies that CCK1 receptor mediates the control of food intake, a contributing role for CCK2 receptor (CCK2R) in the CCK-induced satiety cannot be totally excluded. The hypothesis that CCK2R invalidation disrupts regulatory pathways with impact on feeding behavior was examined in CCK2R(-/-) mice. CCK2R(-/-) mice developed obesity that was associated with hyperphagia. Obesity was related with increased fat deposition resulting from adipocyte hypertrophy. Expression of several adipokines was dysregulated consistently with obesity. Moreover, obesity was associated with disturbed glucose homeostasis as revealed by increased fasting glycemia and insulinemia, impaired glucose tolerance, and hepatic insulin resistance in CCK2R(-/-) mice. In vitro analysis of isolated adipocytes metabolism was consistent with increased storage but preserved insulin sensitivity. Suppression of feeding and concomitant increased expression of hypothalamic proopiomelanocortin after intracerebroventricular injection of gastrin into control mice demonstrates that hypothalamic CCK2 receptors mediate inhibition of food intake. Comparative analysis of hypothalamic mediator gene expression in fed knockout and control mice demonstrated overexpression of ghrelin receptors in CCK2R(-/-) mice, indicating up-regulation of orexigenic pathways. This effect was also observed after body weight normalization, indicating a causative role in the development of hyperphagia and obesity of CCK2R(-/-) mice. Our results give evidence that CCK2 receptor activity plays a contributing regulatory role in the control of food intake.


Subject(s)
Appetite Regulation/genetics , Hyperphagia/genetics , Obesity/genetics , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/physiology , Adiposity , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Body Weight , Energy Metabolism , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Motor Activity
17.
Biochim Biophys Acta ; 1763(10): 1098-107, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16963136

ABSTRACT

SHP-2 is a tyrosine phosphatase which functions as a positive regulator downstream of RTKs, activating growth-stimulatory signalling pathways. To date, very few G protein-coupled receptors (GPCRs) have been shown to be connected to SHP-2 and very little is known about the positive role of SHP-2 in GPCR signalling. The CCK2 receptor (CCK2R), a GPCR, is now recognized to mediate mitogenic effects of gastrin on gastrointestinal cells. In the present study, we demonstrate the role of SHP-2 in the activation of the AKT pathway by the CCK2R in COS-7 cells transfected with the CCK2R and in a pancreatic cancer cell line expressing the endogenous receptor. Using surface plasmon resonance analysis, we identified a highly conserved ITIM motif, containing the tyrosine residue 438, located in the C-terminal intracellular tail of the CCK2R which directly interacts with the SHP-2 SH2 domains. The interaction was confirmed by pull down assays and co-immunoprecipitation of the receptor with SHP-2. This interaction was transiently increased following gastrin stimulation of the CCK2R and correlated with the tyrosine phosphorylation of SHP-2. Mutational analysis of the key ITIM residue 438 confirmed that the CCK2R ITIM sequence is required for interaction with SHP-2 and the activation of the AKT pathway.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Pancreatic Neoplasms/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Cholecystokinin B/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Gastrins/pharmacology , Intracellular Signaling Peptides and Proteins/physiology , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/physiology , Rats , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/physiology , Signal Transduction , Tyrosine/metabolism
18.
World J Gastroenterol ; 12(28): 4498-503, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16874861

ABSTRACT

AIM: To investigate in vivo, whether CCK2 receptors (CCK2R) regulate proteins known to play a crucial role in cell proliferation and cancer development and analyse in vitro the molecular mechanisms that lead to Src activation; in particular, to identify the domains within the CCK2R sequence that are implicated in this activation. METHODS: The expression and activation of Src and ERK were studied in vivo using immuno-fluorescence and western-blot techniques. We used pancreatic tissues derived from wild type or Elas-CCK2 mice that expressed the CCK2R in pancreatic acini, displayed an increased pancreatic growth and developed preneoplastic lesions. The pancreatic tumor cell line AR4-2J expressing the endogenous CCK2R or COS-7 cells transiently transfected with wild type or mutant CCK2R were used as in vitro models to study the mechanism of Src activation. Src activation was measured by in vitro kinase assays, ERK activation by western blot using anti-phospho-ERK antibodies and the involvement of Src in gastrin-induced cell proliferation by MTT test. RESULTS: We showed in vivo that the targeted CCK2R expression in the pancreas of Elas-CCK2 mice, led to the activation of Src and the ERK pathway. Src was activated upstream of the ERK pathway by the CCK2R in pancreatic tumoral cells and contributed to the proliferative effects mediated by this receptor. In vitro results demonstrated that activation of the Src/ERK pathway by the CCK2R required the NPXXY motif, located within the CCK2R sequence at the end of the 7th transmembrane domain, and suggested the putative role of Gq in this mechanism. CONCLUSION: Deregulation of the Src/ERK pathway by the CCK2R might represent an early step that contributes to cell proliferation, formation of preneoplastic lesions and pancreatic tumor development.


Subject(s)
Pancreas/metabolism , Pancreatic Neoplasms/physiopathology , Precancerous Conditions/physiopathology , Receptor, Cholecystokinin B/physiology , src-Family Kinases/metabolism , Amino Acid Motifs/physiology , Animals , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Mice , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Protein Structure, Tertiary/physiology , Signal Transduction/physiology , src-Family Kinases/genetics
19.
Physiol Rev ; 86(3): 805-47, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16816139

ABSTRACT

Cholecystokinin and gastrin receptors (CCK1R and CCK2R) are G protein-coupled receptors that have been the subject of intensive research in the last 10 years with corresponding advances in the understanding of their functioning and physiology. In this review, we first describe general properties of the receptors, such as the different signaling pathways used to exert short- and long-term effects and the structural data that explain their binding properties, activation, and regulation. We then focus on peripheral cholecystokinin receptors by describing their tissue distribution and physiological actions. Finally, pathophysiological peripheral actions of cholecystokinin receptors and their relevance in clinical disorders are reviewed.


Subject(s)
Cholecystokinin/physiology , Receptor, Cholecystokinin B/physiology , Receptors, Cholecystokinin/physiology , Amino Acid Sequence , Animals , Binding Sites , Humans , Molecular Sequence Data , Receptor, Cholecystokinin B/chemistry , Receptors, Cholecystokinin/chemistry
20.
World J Gastroenterol ; 12(12): 1859-64, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-16609991

ABSTRACT

AIM: To investigate whether Src, JAK2 and phosphatidylinositol 3-kinase (PI3K) pathways are involved in the proliferation of human colonic tumour cells induced by glycine-extended gastrin (G-gly), the precursor of the mature amidated gastrin and to elucidate the molecular interaction between these three kinases in response to this peptide. METHODS: Using the human colonic tumour cell line HCT116 as a model, we first measured the activation of PI3K, p60-Src and JAK2 in response to G-gly by in vitro kinase assays. Then we investigated the involvement of these kinases in G-gly-induced cell proliferation by MTT test. RESULTS: G-gly stimulation induced p60-Src, JAK2 and PI3K activation in HCT116. The different pathways were involved in proliferation of human colon cancer cells induced by G-gly. Furthermore, we found that both Src and JAK2 were necessary to PI3K regulation by this peptide. However, we did not find any cross-talk between the two tyrosine kinases. CONCLUSION: Our results suggest that the p60-Src/PI3K and JAK2/PI3K pathways act independently to mediate G-gly proliferative effect on human colonic tumour cells.


Subject(s)
Colonic Neoplasms/enzymology , Gastrins/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , HCT116 Cells , Humans , Immunoprecipitation , Janus Kinase 2
SELECTION OF CITATIONS
SEARCH DETAIL
...