Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Rev Mol Cell Biol ; 11(2): 103-12, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20094051

ABSTRACT

Networks of signal transducers determine the conversion of environmental cues into cellular actions. Among the main players in these networks are protein kinases, which can acutely and reversibly modify protein functions to influence cellular events. One group of kinases, the protein kinase C (PKC) family, have been increasingly implicated in the organization of signal propagation, particularly in the spatial distribution of signals. Examples of where and how various PKC isoforms direct this tier of signal organization are becoming more evident.


Subject(s)
Protein Kinase C/metabolism , Signal Transduction , Animals , Cell Communication , Cell Movement , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Kinase C/genetics , Protein Transport
2.
J Cell Sci ; 127(Pt 9): 1938-52, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24790222

ABSTRACT

Many carcinomas have acquired oncogenic mechanisms for activating c-Met, including c-Met overexpression and excessive autocrine or paracrine stimulation with hepatocyte growth factor (HGF). However, the biological outcome of c-Met activation through these distinct modes remains ambiguous. Here, we report that HGF-mediated c-Met stimulation triggers a mesenchymal-type collective cell invasion. By contrast, the overexpression of c-Met promotes cell rounding. Moreover, in a high-throughput siRNA screen that was performed using a library of siRNAs against putative regulators of integrin activity, we identified RhoA and the clathrin-adapter protein HIP1 as crucial c-Met effectors in these morphological changes. Transient RhoA activation was necessary for the HGF-induced invasion, whereas sustained RhoA activity regulated c-Met-induced cell rounding. In addition, c-Met-induced cell rounding correlated with the phosphorylation of filamin A and the downregulation of active cell-surface integrins. By contrast, a HIP1-mediated increase in ß1-integrin turnover was required for the invasion triggered by HGF. Taken together, our results indicate that c-Met induces distinct cell morphology alterations depending on the stimulus that activates c-Met.


Subject(s)
DNA-Binding Proteins/metabolism , Integrins/metabolism , Proto-Oncogene Proteins c-met/metabolism , rhoA GTP-Binding Protein/metabolism , Apoptosis/genetics , Apoptosis/physiology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Cell Line , DNA-Binding Proteins/genetics , Endocytosis/genetics , Endocytosis/physiology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Integrins/genetics , Proto-Oncogene Proteins c-met/genetics , rhoA GTP-Binding Protein/genetics
3.
Nature ; 465(7299): 813-7, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20535211

ABSTRACT

Down's syndrome (DS) is a genetic disorder caused by full or partial trisomy of human chromosome 21 and presents with many clinical phenotypes including a reduced incidence of solid tumours. Recent work with the Ts65Dn model of DS, which has orthologues of about 50% of the genes on chromosome 21 (Hsa21), has indicated that three copies of the ETS2 (ref. 3) or DS candidate region 1 (DSCR1) genes (a previously known suppressor of angiogenesis) is sufficient to inhibit tumour growth. Here we use the Tc1 transchromosomic mouse model of DS to dissect the contribution of extra copies of genes on Hsa21 to tumour angiogenesis. This mouse expresses roughly 81% of Hsa21 genes but not the human DSCR1 region. We transplanted B16F0 and Lewis lung carcinoma tumour cells into Tc1 mice and showed that growth of these tumours was substantially reduced compared with wild-type littermate controls. Furthermore, tumour angiogenesis was significantly repressed in Tc1 mice. In particular, in vitro and in vivo angiogenic responses to vascular endothelial growth factor (VEGF) were inhibited. Examination of the genes on the segment of Hsa21 in Tc1 mice identified putative anti-angiogenic genes (ADAMTS1and ERG) and novel endothelial cell-specific genes, never previously shown to be involved in angiogenesis (JAM-B and PTTG1IP), that, when overexpressed, are responsible for inhibiting angiogenic responses to VEGF. Three copies of these genes within the stromal compartment reduced tumour angiogenesis, explaining the reduced tumour growth in DS. Furthermore, we expect that, in addition to the candidate genes that we show to be involved in the repression of angiogenesis, the Tc1 mouse model of DS will permit the identification of other endothelium-specific anti-angiogenic targets relevant to a broad spectrum of cancer patients.


Subject(s)
Carcinoma, Lewis Lung/blood supply , Disease Models, Animal , Down Syndrome/genetics , Gene Dosage/genetics , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/genetics , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS1 Protein , Animals , Carcinoma, Lewis Lung/complications , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chromosomes, Mammalian/genetics , Down Syndrome/complications , Down Syndrome/physiopathology , Female , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Intracellular Signaling Peptides and Proteins , Male , Melanoma, Experimental/complications , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Transcription Factors , Transcriptional Regulator ERG , Trisomy/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
Breast Cancer Res ; 17: 52, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25887320

ABSTRACT

c-Met is a receptor tyrosine kinase that upon binding of its ligand, hepatocyte growth factor (HGF), activates downstream pathways with diverse cellular functions that are important in organ development and cancer progression. Anomalous c-Met signalling has been described in a variety of cancer types, and the receptor is regarded as a novel therapeutic target. In breast cancer there is a need to develop new treatments, particularly for the aggressive subtypes such as triple-negative and basal-like cancer, which currently lack targeted therapy. Over the last two decades, much has been learnt about the functional role of c-Met signalling in different models of breast development and cancer. This work has been complemented by clinical studies, establishing the prognostic significance of c-Met in tissue samples of breast cancer. While the clinical trials of anti-c-Met therapy in advanced breast cancer progress, there is a need to review the existing evidence so that the potential of these treatments can be better appreciated. The aim of this article is to examine the role of HGF/c-Met signalling in in vitro and in vivo models of breast cancer, to describe the mechanisms of aberrant c-Met signalling in human tissues, and to give a brief overview of the anti-c-Met therapies currently being evaluated in breast cancer patients. We will show that the HGF/c-Met pathway is associated with breast cancer progression and suggest that there is a firm basis for continued development of anti-c-Met treatment, particularly for patients with basal-like and triple-negative breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Hepatocyte Growth Factor/metabolism , Humans , Molecular Targeted Therapy , Prognosis , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
5.
J Cell Sci ; 126(Pt 11): 2381-91, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23549785

ABSTRACT

Aberrant signalling of receptor tyrosine kinases (RTKs), such as c-Met, the receptor for hepatocyte growth factor (HGF), has been implicated in the oncogenesis of various tumours including non-small cell lung carcinoma (NSCLC). Through its pro-migratory properties, c-Met has been implicated specifically in the process of tumour metastasis, demanding a better understanding of the underlying signalling pathways. Various players downstream of c-Met have been well characterised, including the extracellular-signal-regulated kinases (ERKs) 1 and 2. In a small interfering RNA (siRNA)-based high-throughput wound healing screen performed in A549 lung carcinoma cells, we identified ERK2 but not ERK1 as a strong mediator of HGF-induced motility. This finding was confirmed in several NSCLC cell lines as well as in HeLa cells. One known substrate for ERK kinases in cell migration, the focal adhesion protein paxillin, was also one of the hits identified in the screen. We demonstrate that HGF stimulation results in a time-dependent phosphorylation of paxillin on serine 126, a process that can be blocked by inhibition of the ERK1/2 upstream kinase mitogen-activated protein kinase/ERK kinase 1 (MEK1) or inhibition of glycogen synthase kinase 3 (GSK3). Further, we show that paxillin turnover at focal adhesions is increased upon stimulation by HGF, an effect that is dependent on serine residues 126 (GSK3 site) and 130 (ERK site) within paxillin. In line with the isoform-specific requirement of ERK2 for HGF-mediated migration in lung tumour cell models, ERK2 but not ERK1 is shown to be responsible for paxillin serine 126 phosphorylation and its increased turnover at focal adhesions.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Hepatocyte Growth Factor/metabolism , Lung Neoplasms/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Focal Adhesions/genetics , Focal Adhesions/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , HeLa Cells , Hepatocyte Growth Factor/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Paxillin/genetics , Paxillin/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism
6.
Cancer ; 120(2): 163-71, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24150964

ABSTRACT

BACKGROUND: Basal-like (BL) breast cancer is an aggressive form of breast cancer with limited treatment options. Recent work has identified BL breast cancer as a biologically distinct form of triple-negative breast cancer, with a worse outlook. The receptor tyrosine kinase c-Met is a novel therapeutic target associated with reduced survival in breast cancer. Few studies have specifically addressed the association between c-Met and molecular subtype of breast cancer, yet this is a key consideration when selecting patients for clinical trials. The aim of this study is to evaluate c-Met expression in a large cohort of invasive breast cancers and in particular, its correlation with molecular subtype. METHODS: Immunohistochemistry for c-Met was performed and evaluated on 1274 invasive breast cancers using tissue microarray technology. The c-Met scores were correlated with molecular subtype, survival, and other standard clinicopathological prognostic factors. RESULTS: Multivariate logistic regression showed c-Met was independently associated with BL status (odds ratio=6.44, 95% confidence interval=1.74-23.78, P=.005). There was a positive correlation between c-Met and Her2 (P=.005) and an inverse correlation with tumor size (P<.001). C-Met was an independent poor prognostic factor at Cox regression analysis in all subtypes (hazard ratio=1.85, 95% confidence interval=1.07-3.19, P=.027) and there was a trend toward reduced survival in BL tumors overexpressing c-Met, but this was not significant. CONCLUSIONS: C-Met is independently associated with BL breast cancer. In the future, patients with BL tumors should be included in clinical trials of anti-c-Met therapy.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , Adult , Aged , Breast Neoplasms/mortality , Cohort Studies , Female , Humans , Logistic Models , Middle Aged , Odds Ratio , Prognosis , Proportional Hazards Models , Receptor, ErbB-2/metabolism
7.
Mol Oncol ; 17(11): 2257-2274, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36799689

ABSTRACT

Exon skipping mutations of the MET receptor tyrosine kinase (METex14), increasingly reported in cancers, occur in 3-4% of non-small-cell lung cancer (NSCLC). Only 50% of patients have a beneficial response to treatment with MET-tyrosine kinase inhibitors (TKIs), underlying the need to understand the mechanism of METex14 oncogenicity and sensitivity to TKIs. Whether METex14 is a driver mutation and whether it requires hepatocyte growth factor (HGF) for its oncogenicity in a range of in vitro functions and in vivo has not been fully elucidated from previous preclinical models. Using CRISPR/Cas9, we developed a METex14/WT isogenic model in nontransformed human lung cells and report that the METex14 single alteration was sufficient to drive MET-dependent in vitro anchorage-independent survival and motility and in vivo tumorigenesis, sensitising tumours to MET-TKIs. However, we also show that human HGF (hHGF) is required, as demonstrated in vivo using a humanised HGF knock-in strain of mice and further detected in tumour cells of METex14 NSCLC patient samples. Our results also suggest that METex14 oncogenicity is not a consequence of an escape from degradation in our cell model. Thus, we developed a valuable model for preclinical studies and present results that have potential clinical implication.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Exons , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Lung Neoplasms/pathology , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Animals , Mice
8.
Int J Cancer ; 130(5): 1060-70, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21400516

ABSTRACT

c-Met [the hepatocyte growth factor (HGF) receptor] is a receptor tyrosine kinase playing a role in various biological events. Overexpression of the receptor has been observed in a number of cancers, correlating with increased metastatic tendency and poor prognosis. Additionally, activating mutations in c-Met kinase domain have been reported in a subset of familial cancers causing resistance to treatment. Receptor trafficking, relying on the integrity of the microtubule network, plays an important role in activation of downstream targets and initiation of signalling events. Aurintricarboxylic acid (ATA) is a triphenylmethane derivative that has been reported to inhibit microtubule motor proteins kinesins. Additional reported properties of this inhibitor include inhibition of protein tyrosine phosphatases, nucleases and members of the Jak family. Here we demonstrate that ATA prevents HGF-induced c-Met phosphorylation, internalisation, subsequent receptor trafficking and degradation. In addition, ATA prevented HGF-induced downstream signalling which also affected cellular function, as assayed by collective cell migration of A549 cells. Surprisingly, the inhibitory effect of ATA on HGF-induced phosphorylation and signalling in vivo was associated with an increase in basal c-Met kinase activity in vitro. It is concluded that the inhibitory effects of ATA on c-Met in vivo is an allosteric effect mediated through the kinase domain of the receptor. As the currently tested adenosine triphosphate competitive tyrosine kinase inhibitors (TKIs) may lead to tumor resistance (McDermott U, et al., Cancer Res 2010;70:1625-34), our findings suggest that novel anti-c-Met therapies could be developed in the future for cancer treatment.


Subject(s)
Aurintricarboxylic Acid/pharmacology , Kinesins/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Allosteric Regulation , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation , HeLa Cells , Humans , Lung Neoplasms , Phosphorylation/drug effects , Signal Transduction/drug effects
9.
Front Cell Dev Biol ; 10: 994528, 2022.
Article in English | MEDLINE | ID: mdl-36330337

ABSTRACT

c-Met is a receptor tyrosine kinase which upon activation by its ligand, the hepatocyte growth factor, mediates many important signalling pathways that regulate cellular functions such as survival, proliferation, and migration. Its oncogenic and tumorigenic signalling mechanisms, greatly contributing to cancer development and progression, are well documented. Integrins, heterogeneous adhesion receptors which facilitate cell-extracellular matrix interactions, are important in biomechanically sensitive cell adhesion and motility but also modulate diverse cell behaviour. Here we review the studies which reported cooperation between c-Met and several integrins, particularly ß1 and ß4, in various cell models including many tumour cell types. From the various experimental models and results analysed, we propose that c-Met-integrin cooperation occurs via inside-out or outside-in signalling. Thus, either c-Met activation triggers integrin activation and cell adhesion or integrin adhesion to its extracellular ligand triggers c-Met activation. These two modes of cooperation require the adhesive function of integrins and mostly lead to cell migration and invasion. In a third, less conventional, mode of cooperation, the integrin plays the role of a signalling adaptor for c-Met, independently from its adhesive property, leading to anchorage independent survival. Recent studies have revealed the influence of endocytic trafficking in c-Met-integrin cooperation including the adaptor function of integrin occurring on endomembranes, triggering an inside-in signalling, believed to promote survival of metastatic cells. We present the evidence of the cooperation in vivo and in human tissues and highlight its therapeutic relevance. A better understanding of the mechanisms regulating c-Met-integrin cooperation in cancer progression could lead to the design of new therapies targeting this cooperation, providing more effective therapeutic approaches than c-Met or integrin inhibitors as monotherapies used in the clinic.

10.
Oncogene ; 40(11): 1927-1941, 2021 03.
Article in English | MEDLINE | ID: mdl-33526881

ABSTRACT

Chemotherapy remains the mainstay of treatment in the majority of solid and haematological malignancies. Resistance to cytotoxic chemotherapy is a major clinical problem and substantial research is ongoing into potential methods of overcoming this resistance. One major target, the receptor tyrosine kinase MET, has generated increasing interest with multiple clinical trials in progress. Overexpression of MET is frequently observed in a range of different cancers and is associated with poor prognosis. Studies have shown that MET promotes resistance to targeted therapies, including those targeting EGFR, BRAF and MEK. More recently, several reports suggest that MET also contributes to cytotoxic chemotherapy resistance. Here we review the preclinical evidence of MET's role in chemotherapy resistance, the mechanisms by which this resistance is mediated and the translational relevance of MET inhibitor therapy for patients with chemotherapy resistant disease.


Subject(s)
Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , Proteomics , Proto-Oncogene Proteins c-met/genetics , Humans , Neoplasms/genetics
11.
Mol Cell Oncol ; 7(6): 1803029, 2020 Aug 18.
Article in English | MEDLINE | ID: mdl-33235904

ABSTRACT

We reported that RAC1 is a master regulator of cell migration and anchorage-independent growth, downstream of the oncogenic Receptor Tyrosine Kinase (RTK) MET. RAC1 growth-promoting role is guanosine triphosphatase (GTPase)- and phosphatidylinositol 3-kinase (PI3K)-independent but promotes mammalian target of rapamycin (mTOR) signaling through triggering its plasma membrane localization.

12.
Sci Signal ; 13(637)2020 06 23.
Article in English | MEDLINE | ID: mdl-32576681

ABSTRACT

Receptor tyrosine kinases (RTKs) are often overexpressed or mutated in cancers and drive tumor growth and metastasis. In the current model of RTK signaling, including that of MET, downstream phosphatidylinositol 3-kinase (PI3K) mediates both cell proliferation and cell migration, whereas the small guanosine triphosphatase (GTPase) Rac1 mediates cell migration. However, in cultured NIH3T3 and glioblastoma cells, we found that class I PI3K mediated oncogenic MET-induced cell migration but not anchorage-independent growth. In contrast, Rac1 regulated both processes in distinct ways. Downstream of PI3K, Rac1 mediated cell migration through its GTPase activity, whereas independently of PI3K, Rac1 mediated anchorage-independent growth in a GTPase-independent manner through an adaptor function. Through its RKR motif, Rac1 formed a complex with the kinase mTOR to promote its translocation to the plasma membrane, where its activity promoted anchorage-independent growth of the cell cultures. Inhibiting mTOR with rapamycin suppressed the growth of subcutaneous MET-mutant cell grafts in mice, including that of MET inhibitor-resistant cells. These findings reveal a GTPase-independent role for Rac1 in mediating a PI3K-independent MET-to-mTOR pathway and suggest alternative or combined strategies that might overcome resistance to RTK inhibitors in patients with cancer.


Subject(s)
Neuropeptides/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Movement , Mice , NIH 3T3 Cells , Neuropeptides/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-met/genetics , TOR Serine-Threonine Kinases/genetics , rac1 GTP-Binding Protein/genetics
14.
Front Mol Biosci ; 5: 86, 2018.
Article in English | MEDLINE | ID: mdl-30406111

ABSTRACT

The Receptor Tyrosine Kinase (RTK) Met, overexpressed or mutated in cancer, plays a major role in cancer progression and represents an attractive target for cancer therapy. However RTK inhibitors can lead to drug resistance, explaining the necessity to develop therapies that target downstream signaling. Phosphatidylinositide 3-kinase (PI3K) is one of the most deregulated pathways in cancer and implicated in various types of cancer. PI3K signaling is also a major signaling pathway downstream of RTK, including Met. PI3K major effectors include Akt and "mechanistic Target of Rapamycin" (mTOR), which each play key roles in numerous and various cell functions. Advancements made due to the development of molecular and pharmaceutical tools now allow us to delve into the roles of each independently. In this review, we summarize the current understanding we possess of the activation and role of PI3K/Akt/mTOR, downstream of Met, in cancer.

15.
Cell Adh Migr ; 11(3): 211-232, 2017 05 04.
Article in English | MEDLINE | ID: mdl-27254775

ABSTRACT

E-cadherin mediated cell-cell adhesion plays a critical role in epithelial cell polarization and morphogenesis. Our recent studies suggest that the desmosomal cadherin, desmoglein 3 (Dsg3) cross talks with E-cadherin and regulates its adhesive function in differentiating keratinocytes. However, the underlying mechanism remains not fully elucidated. Since E-cadherin trafficking has been recognized to be a central determinant in cell-cell adhesion and homeostasis we hypothesize that Dsg3 may play a role in regulating E-cadherin trafficking and hence the cell-cell adhesion. Here we investigated this hypothesis in cells with loss of Dsg3 function through RNAi mediated Dsg3 knockdown or the stable expression of the truncated mutant Dsg3ΔC. Our results showed that loss of Dsg3 resulted in compromised cell-cell adhesion and reduction of adherens junction and desmosome protein expression as well as the cortical F-actin formation. As a consequence, cells failed to polarize but instead displayed aberrant cell flattening. Furthermore, retardation of E-cadherin internalization and recycling was consistently observed in these cells during the process of calcium induced junction assembling. In contrast, enhanced cadherin endocytosis was detected in cells with overexpression of Dsg3 compared to control cells. Importantly, this altered cadherin trafficking was found to be coincided with the reduced expression and activity of Rab proteins, including Rab5, Rab7 and Rab11 which are known to be involved in E-cadherin trafficking. Taken together, our findings suggest that Dsg3 functions as a key in cell-cell adhesion through at least a mechanism of regulating E-cadherin membrane trafficking.


Subject(s)
Cadherins/genetics , Cell Adhesion/genetics , Desmoglein 3/genetics , Animals , Cadherins/metabolism , Cell Line , Desmoglein 3/metabolism , Epithelial Cells/metabolism , Humans , Mice , Protein Transport/genetics
16.
Nat Commun ; 7: 11942, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27336951

ABSTRACT

Receptor tyrosine kinases (RTKs) and integrins cooperate to stimulate cell migration and tumour metastasis. Here we report that an integrin influences signalling of an RTK, c-Met, from inside the cell, to promote anchorage-independent cell survival. Thus, c-Met and ß1-integrin co-internalize and become progressively recruited on LC3B-positive 'autophagy-related endomembranes' (ARE). In cells growing in suspension, ß1-integrin promotes sustained c-Met-dependent ERK1/2 phosphorylation on ARE. This signalling is dependent on ATG5 and Beclin1 but not on ATG13, suggesting ARE belong to a non-canonical autophagy pathway. This ß1-integrin-dependent c-Met-sustained signalling on ARE supports anchorage-independent cell survival and growth, tumorigenesis, invasion and lung colonization in vivo. RTK-integrin cooperation has been assumed to occur at the plasma membrane requiring integrin 'inside-out' or 'outside-in' signalling. Our results report a novel mode of integrin-RTK cooperation, which we term 'inside-in signalling'. Targeting integrin signalling in addition to adhesion may have relevance for cancer therapy.


Subject(s)
Integrin beta1/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Autophagy , Carcinogenesis , Cell Adhesion , Cell Line , Cell Movement , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hepatocyte Growth Factor/pharmacology , Humans , Integrin beta1/genetics , Mice , Proto-Oncogene Proteins c-met/genetics , Signal Transduction
17.
J Clin Endocrinol Metab ; 90(11): 6177-84, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16144948

ABSTRACT

CONTEXT: Leptin, partially produced by the stomach, is a hormone involved in energy balance and regulation of food intake. It also regulates some digestive functions through its functional receptor Ob-Rb expressed by gastrointestinal epithelial cells. OBJECTIVE: The objective of the study was to investigate the temporal and spatial appearance of Ob-Rb in the human digestive tract and leptin in the stomach. DESIGN: The esophagus, stomach, and intestine samples of 7- to 24-wk-old human fetuses and adult mucosae were studied by RT-PCR, immunohistochemistry, and Western blot. Leptin was measured by RIA in amniotic fluids at 16-33 wk gestation. RESULTS: All mucosae expressed Ob-Rb (mRNA and/or protein) between 7 and 9 wk gestation. Leptin protein appeared by 8 wk in the gastric mucosa, whereas leptin mRNA was detected around 11 wk. Leptin levels in amniotic fluids were significantly higher during the second than during the third trimester. Overall, Ob-Rb immunoreactivity was higher in young fetuses, during the period corresponding to the formation of gastric buds and primitive intestinal crypts and the beginning of differentiation of epithelial cell types, than in the oldest. Leptin added to culture medium of gastrointestinal explants from 10- to 12-wk-old fetuses appeared to affect DNA synthesis as compared with controls, indicating that leptin receptor functionality was developing. CONCLUSIONS: The strong expression of leptin, in amniotic fluid when fetuses begin swallowing then in the gastric mucosa, and the early presence of Ob-Rb in mucosae suggest a possible role for leptin, exerted endoluminally and in a paracrine pathway, in the developmental process (growth and/or maturation) of the human digestive tract.


Subject(s)
Fetus/chemistry , Leptin/analysis , Receptors, Cell Surface/analysis , Amniotic Fluid/chemistry , DNA/biosynthesis , Esophagus/chemistry , Fetal Development , Humans , Immunohistochemistry , Intestines/chemistry , Leptin/physiology , Protein Isoforms , Receptors, Cell Surface/physiology , Receptors, Leptin , Reverse Transcriptase Polymerase Chain Reaction , Stomach/chemistry
18.
Clin Cancer Res ; 9(1): 273-84, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12538480

ABSTRACT

PURPOSE: High prevalence of squamous cervical intraepithelial neoplasia (CIN) linked to oncogenic human papillomavirus (HPV) exits in HIV-infected women. Hepatocyte growth factor (HGF) and its receptor, c-Met, promote cell proliferation and are involved in tumor progression. Nothing is yet known about their expression in low- and high-grade CIN. Therefore, the expression, localization, and behavior of HGF and c-Met in normal and dysplastic cervical epithelium were investigated. EXPERIMENTAL DESIGN: We studied normal cervical mucosa from 10 healthy women, and low- and high-grade cervical lesions, uninfected (condyloma acuminata) or infected with oncogenic HPVs, from 40 HIV-negative and 48 HIV-positive women, using in situ molecular techniques, immunocytochemistry and morphoquantitative methods. RESULTS: In 154 oncogenic HPV-infected CIN encountered in biopsy samples, the total number of epithelial cell layers increased significantly during lesion progression. This number was significantly higher in HIV-positive than in HIV-negative women for CIN1 and CIN2 (P < 0.025 to P < 0.01). In HIV-negative women, the number and percentage of HGF and c-Met immunostained cell layers, and the intensity of immunostaining were enhanced in oncogenic HPV-infected lesions as compared with normal mucosa and condyloma acuminata. The latter parameters were significantly higher in tissues of HIV-positive women (oncogenic HPV-infected CIN1 and CIN2, normal-appearing mucosa contiguous to CIN, condyloma acuminata) than in the corresponding tissues of HIV-negative women (P < 0.025 to P < 0.0001). CONCLUSIONS: Overexpression of HGF/c-Met complex strongly correlates with oncogenic HPV and HIV infection. This overexpressed complex may stimulate cell proliferation in condyloma acuminata and participate in tumor progression in oncogenic HPV-infected lesions.


Subject(s)
HIV/metabolism , Hepatocyte Growth Factor/biosynthesis , Papillomaviridae/metabolism , Proto-Oncogene Proteins c-met/biosynthesis , Uterine Cervical Dysplasia/metabolism , Adolescent , Adult , Aged , Cell Division , Disease Progression , Epithelium/metabolism , Female , HIV Seropositivity , Humans , Immunohistochemistry , Middle Aged , Mucous Membrane/pathology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
Int J Biochem Cell Biol ; 49: 69-74, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440758

ABSTRACT

Deregulated signalling of the Receptor Tyrosine Kinase (RTK), Met, and/or its ligand HGF have been associated with cancer formation and progression to metastasis, with Met/HGF often overexpressed or mutated. Thus, Met has become a major target for cancer therapy and its inhibition is currently being tested in the clinic. It has recently become evident that, instead of signalling at the plasma membrane only, Met signals post-internalisation from endosomal compartments. Thus, Met endocytic trafficking is required for the full activation of signals such as Gab1, ERK 1/2, STAT3 and Rac1, all implicated in cell survival, invasion and metastasis. Modifications in the balance between degradation and recycling of Met may also impinge on Met signalling. Moreover, oncogenic Met mutations in the kinase domain trigger constitutive Met internalisation/recycling, leading to "endosomal signalling" and consequent cell transformation. Using Met as an example, this review outlines the evidence that the molecular mechanisms regulating trafficking and endosomal signalling may be exploited to design future cancer therapies.


Subject(s)
Endocytosis/physiology , Endosomes/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Endocytosis/genetics , Humans , Models, Biological , Mutation , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/genetics
20.
Methods Enzymol ; 535: 121-40, 2014.
Article in English | MEDLINE | ID: mdl-24377921

ABSTRACT

Met is a receptor tyrosine kinase, often overexpressed or mutated in human cancer. Upon activation by its ligand, the hepatocyte growth factor, Met controls several cell functions such as proliferation, migration, and survival through the activation of multiple pathways. Upon ligand binding, Met rapidly internalizes and continues to signal from endosomal compartments prior to its degradation. Importantly, this "endosomal signaling" has recently been shown to be involved in tumorigenesis and experimental metastasis. Consequently, interfering with Met endosomal signaling may provide a novel therapeutic approach in cancer treatment. However, there is a need for additional studies in various experimental models to confirm this and find the most specific ways of achieving it. Thus, outlined in this review are the techniques and tools we have been using to study Met endocytosis and Met endosomal signaling.


Subject(s)
Carcinogenesis/metabolism , Endosomes/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Animals , Binding Sites , Biotinylation , Cell Line, Tumor , Flow Cytometry , Fluorescent Antibody Technique , Hepatocyte Growth Factor/metabolism , Humans , Image Processing, Computer-Assisted , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasm Transplantation , Protein Transport , Proteolysis , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL