Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Cancer Gene Ther ; 28(1-2): 18-26, 2021 02.
Article En | MEDLINE | ID: mdl-32595215

OPCML is a highly conserved glycosyl phosphatidylinositol (GPI)-anchored protein belonging to the IgLON family of cell adhesion molecules. OPCML functions as a tumor suppressor and is silenced in over 80% of ovarian cancers by loss of heterozygosity and by epigenetic mechanisms. OPCML inactivation is also observed in many other cancers suggesting a conservation of tumor suppressor function. Although epigenetic silencing and subsequent loss of OPCML expression correlate with poor progression-free and overall patient survival, its mechanism of action is only starting to be fully elucidated. Recent discoveries have demonstrated that OPCML exerts its tumor suppressor effect by inhibiting several cancer hallmark phenotypes in vitro and abrogating tumorigenesis in vivo, by downregulating/inactivating a specific spectrum of Receptor Tyrosine Kinases (RTKs), including EphA2, FGFR1, FGFR3, HER2, HER4, and AXL. This modulation of RTKs can also sensitize ovarian and breast cancers to lapatinib, erlotinib, and anti-AXL therapies. Furthermore, OPCML has also been shown to function in synergy with the tumor suppressor phosphatase PTPRG to inactivate pro-metastatic RTKs such as AXL. Recently, the identification of inactivating point mutations and the elucidation of the crystal structure of OPCML have provided valuable insights into its structure-function relationships, giving rise to its potential as an anti-cancer therapeutic.


Cell Adhesion Molecules/drug effects , Glycosylphosphatidylinositols/therapeutic use , Neoplasms/drug therapy , GPI-Linked Proteins/drug effects , Glycosylphosphatidylinositols/pharmacology , Humans
2.
Nat Commun ; 10(1): 3134, 2019 07 17.
Article En | MEDLINE | ID: mdl-31316070

OPCML, a tumor suppressor gene, is frequently silenced epigenetically in ovarian and other cancers. Here we report, by analysis of databases of tumor sequences, the observation of OPCML somatic missense mutations from various tumor types and the impact of these mutations on OPCML function, by solving the X-ray crystal structure of this glycoprotein to 2.65 Å resolution. OPCML consists of an extended arrangement of three immunoglobulin-like domains and homodimerizes via a network of contacts between membrane-distal domains. We report the generation of a panel of OPCML variants with representative clinical mutations and demonstrate clear phenotypic effects in vitro and in vivo including changes to anchorage-independent growth, interaction with activated cognate receptor tyrosine kinases, cellular migration, invasion in vitro and tumor growth in vivo. Our results suggest that clinically occurring somatic missense mutations in OPCML have the potential to contribute to tumorigenesis in a variety of cancers.


Cell Adhesion Molecules/genetics , Epigenesis, Genetic , Ovarian Neoplasms/genetics , Cell Adhesion Molecules/chemistry , Cell Transformation, Neoplastic , Crystallography, X-Ray , Female , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , Glycosylation , Humans , Mutation, Missense , Neoplasm Invasiveness , Protein Aggregation, Pathological/genetics , Protein Structure, Tertiary
3.
EMBO Rep ; 19(8)2018 08.
Article En | MEDLINE | ID: mdl-29907679

In ovarian cancer, the prometastatic RTK AXL promotes motility, invasion and poor prognosis. Here, we show that reduced survival caused by AXL overexpression can be mitigated by the expression of the GPI-anchored tumour suppressor OPCML Further, we demonstrate that AXL directly interacts with OPCML, preferentially so when AXL is activated by its ligand Gas6. As a consequence, AXL accumulates in cholesterol-rich lipid domains, where OPCML resides. Here, phospho-AXL is brought in proximity to the lipid domain-restricted phosphatase PTPRG, which de-phosphorylates the RTK/ligand complex. This prevents AXL-mediated transactivation of other RTKs (cMET and EGFR), thereby inhibiting sustained phospho-ERK signalling, induction of the EMT transcription factor Slug, cell migration and invasion. From a translational perspective, we show that OPCML enhances the effect of the phase II AXL inhibitor R428 in vitro and in vivo We therefore identify a novel mechanism by which two spatially restricted tumour suppressors, OPCML and PTPRG, coordinate to repress AXL-dependent oncogenic signalling.


Cell Adhesion Molecules/metabolism , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Benzocycloheptenes/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chickens , Cholesterol/metabolism , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fallopian Tubes/pathology , Female , GPI-Linked Proteins/metabolism , Gene Silencing/drug effects , Humans , Intercellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/drug effects , Membrane Microdomains/metabolism , Neoplasm Invasiveness , Ovarian Neoplasms/pathology , Phosphorylation/drug effects , Protein Binding/drug effects , Treatment Outcome , Triazoles/pharmacology , Axl Receptor Tyrosine Kinase
4.
Front Pharmacol ; 9: 1524, 2018.
Article En | MEDLINE | ID: mdl-30745873

Objective: Apolipoprotein A1 (ApoA1) is remarkably decreased in serum and ovarian tissues of ovarian cancer patients. ApoA1 and ApoA1 mimetic peptides can sequestrate pro-inflammatory phospholipids, some of which are known to activate a variety of oncogenic pathways. Besides, more intrinsic anti-tumorigenic properties, independent from interaction with lipids, have also been described for ApoA1. We aimed to disclose the effects of ApoA1 and a mimetic peptide on the malignant phenotype of ovarian cancer cells, particularly regarding cell viability, invasiveness and platinum sensitization. Methods: Cells viability was assessed by MTS assay. Extracellular matrix invasion was assessed by transwell and spheroid invasion assays. Western blotting was performed to evaluate the effect of test compounds on intracellular pathways. Sensitization assays were performed in vitro and in the biologically relevant in ovo chorioallantoic membrane model. Results: Both ApoA1 and the mimetic peptide, at a concentration of 100 µg/mL, were able to decrease the viability of SKOV3, CAOV3, and OVCAR3 cells (p < 0.05). The peptide at this concentration was not able to affect the viability of immortalized non-neoplastic ovarian cells (p > 0.05). ApoA1 decreased SKOV3 cells invasiveness at 300 µg/mL after 72 and 96 h of exposure (p < 0.05), while the ApoA1 mimetic peptide prevented cell invasion at 50 and 100 µg/mL (p < 0.01). Treatment with 100 µg/mL of ApoA1 mimetic peptide decreased Akt phosphorylation in SKOV3 cells (p < 0.01). Accordingly, treatment with increasing concentrations of the peptide sensitized SKOV3, OVCAR3 and CAOV3 cells to cisplatin. This synergistic effect was observed both in vitro and in ovo. Conclusions: These results support the role of ApoA1 and ApoA1 mimetics as suppressors of ovarian tumorigenesis and as chemo-sensitising agents.

5.
Mol Cancer Ther ; 16(10): 2246-2256, 2017 10.
Article En | MEDLINE | ID: mdl-28775148

Opioid-binding protein/cell adhesion molecule-like (OPCML) is a tumor-suppressor gene that is frequently inactivated in ovarian cancer and many other cancers by somatic methylation. We have previously shown that OPCML exerts its suppressor function by negatively regulating a spectrum of receptor tyrosine kinases (RTK), such as ErbB2/HER2, FGFR1, and EphA2, thus attenuating their related downstream signaling. The physical interaction of OPCML with this defined group of RTKs is a prerequisite for their downregulation. Overexpression/gene amplification of EGFR and HER2 is a frequent event in multiple cancers, including ovarian and breast cancers. Molecular therapeutics against EGFR/HER2 or EGFR only, such as lapatinib and erlotinib, respectively, were developed to target these receptors, but resistance often occurs in relapsing cancers. Here we show that, though OPCML interacts only with HER2 and not with EGFR, the interaction of OPCML with HER2 disrupts the formation of the HER2-EGFR heterodimer, and this translates into a better response to both lapatinib and erlotinib in HER2-expressing ovarian and breast cancer cell lines. Also, we show that high OPCML expression is associated with better response to lapatinib therapy in breast cancer patients and better survival in HER2-overexpressing ovarian cancer patients, suggesting that OPCML co-therapy could be a valuable sensitizing approach to RTK inhibitors. Mol Cancer Ther; 16(10); 2246-56. ©2017 AACR.


Breast Neoplasms/drug therapy , Cell Adhesion Molecules/genetics , ErbB Receptors/genetics , Ovarian Neoplasms/drug therapy , Receptor, ErbB-2/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/administration & dosage , Female , GPI-Linked Proteins/genetics , Gene Amplification/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lapatinib , Molecular Targeted Therapy , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Quinazolines/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/administration & dosage
6.
Curr Top Med Chem ; 17(1): 16-29, 2017.
Article En | MEDLINE | ID: mdl-27530972

The Ras superfamily of small monomeric GTPases includes some of the most prominent cancer targets for which no selective therapeutic agent has yet been successfully developed. The turn of the millennium saw a resurgence of efforts to target these enzymes using new and improved biophysical techniques to overcome the perceived difficulties of insurmountably high affinity for guanosine nucleotides and flat, flexible topology lacking suitable pockets for small molecule inhibitors. Further, recent investigations have begun to probe the dynamic conformational status of GTP-bound Ras, opening up new mechanisms of inhibition. While much of the literature has focused on the oncogenic Ras proteins, particularly K-Ras, these represent only a small minority of therapeutically interesting targets within the superfamily; for example, the Rab GTPases are the largest subfamily of about 70 members, and present an as yet untapped class of potential targets. The present review documents the key methodologies employed to date in structure-guided attempts to drug the Ras GTPases, and forecasts their transferability to other similarly challenging proteins in the superfamily.


Drug Delivery Systems , Drug Design , ras Proteins/drug effects , Molecular Structure , ras Proteins/chemistry
7.
Sci Signal ; 9(448): ra97, 2016 10 04.
Article En | MEDLINE | ID: mdl-27703030

Ovarian cancer is a complex disease with heterogeneity among the gene expression molecular subtypes (GEMS) between patients. Patients with tumors of a mesenchymal ("Mes") subtype have a poorer prognosis than patients with tumors of an epithelial ("Epi") subtype. We evaluated GEMS of ovarian cancer patients for molecular signaling profiles and assessed how the differences in these profiles could be leveraged to improve patient clinical outcome. Kinome enrichment analysis identified AXL as a particularly abundant kinase in Mes-subtype tumor tissue and cell lines. In Mes cells, upon activation by its ligand GAS6, AXL coclustered with and transactivated the receptor tyrosine kinases (RTKs) cMET, EGFR, and HER2, producing sustained extracellular signal-regulated kinase (ERK) activation. In Epi-A cells, AXL was less abundant and induced a transient activation of ERK without evidence of RTK transactivation. AXL-RTK crosstalk also stimulated sustained activation of the transcription factor FRA1, which correlated with the induction of the epithelial-mesenchymal transition (EMT)-associated transcription factor SLUG and stimulation of motility exclusively in Mes-subtype cells. The AXL inhibitor R428 attenuated RTK and ERK activation and reduced cell motility in Mes cells in culture and reduced tumor growth in a chick chorioallantoic membrane model. A higher concentration of R428 was needed to inhibit ERK activation and cell motility in Epi-A cells. Silencing AXL in Mes-subtype cells reversed the mesenchymal phenotype in culture and abolished tumor formation in an orthotopic xenograft mouse model. Thus, AXL-targeted therapy may improve clinical outcome for patients with Mes-subtype ovarian cancer.


Benzocycloheptenes/pharmacology , Cell Movement/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/drug effects , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Triazoles/pharmacology , Animals , Cell Line, Tumor , Cell Movement/genetics , Chick Embryo , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/genetics , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
9.
Breast Cancer Res Treat ; 148(2): 455-62, 2014 Nov.
Article En | MEDLINE | ID: mdl-25248409

The goal of targeted cancer therapies is to specifically block oncogenic signalling, thus maximising efficacy, while reducing side-effects to patients. The gamma-secretase (GS) complex is an attractive therapeutic target in haematological malignancies and solid tumours with major pharmaceutical activity to identify optimal inhibitors. Within GS, nicastrin (NCSTN) offers an opportunity for therapeutic intervention using blocking monoclonal antibodies (mAbs). Here we explore the role of anti-nicastrin monoclonal antibodies, which we have developed as specific, multi-faceted inhibitors of proliferation and invasive traits of triple-negative breast cancer cells. We use 3D in vitro proliferation and invasion assays as well as an orthotopic and tail vail injection triple-negative breast cancer in vivo xenograft model systems. RNAScope assessed nicastrin in patient samples. Anti-NCSTN mAb clone-2H6 demonstrated a superior anti-tumour efficacy than clone-10C11 and the RO4929097 small molecule GS inhibitor, acting by inhibiting GS enzymatic activity and Notch signalling in vitro and in vivo. Confirming clinical relevance of nicastrin as a target, we report evidence of increased NCSTN mRNA levels by RNA in situ hybridization (RNAScope) in a large cohort of oestrogen receptor negative breast cancers, conferring independent prognostic significance for disease-free survival, in multivariate analysis. We demonstrate here that targeting NCSTN using specific mAbs may represent a novel mode of treatment for invasive triple-negative breast cancer, for which there are few targeted therapeutic options. Furthermore, we propose that measuring NCSTN in patient samples using RNAScope technology may serve as companion diagnostic for anti-NCSTN therapy in the clinic.


Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Membrane Glycoproteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Flow Cytometry , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
PLoS One ; 9(7): e102851, 2014.
Article En | MEDLINE | ID: mdl-25051489

The mechanism(s) by which Rab GTPases are specifically recruited to distinct intracellular membranes remains elusive. Here we used Rab27a localisation onto melanosomes as a model to investigate Rab targeting. We identified the α1 subunit of Na+,K+-ATPase (ATP1a1) as a novel Rab27a interacting protein in melanocytes and showed that this interaction is direct with the intracellular M4M5 loop of ATP1a1 and independent of nucleotide bound status of the Rab. Knockdown studies in melanocytes revealed that ATP1a1 plays an essential role in Rab27a-dependent melanosome transport. Specifically, expression of ATP1a1, like the Rab27a GDP/GTP exchange factor (Rab3GEP), is essential for targeting and activation of Rab27a to melanosomes. Finally, we showed that the ability of Rab27a mutants to target to melanosomes correlates with the efficiency of their interaction with ATP1a1. Altogether these studies point to a new role for ATP1a1 as a regulator of Rab27a targeting and activation.


Melanocytes/metabolism , Melanosomes/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Binding Sites , Cells, Cultured , Humans , Immunoblotting , Mice , Microscopy, Confocal , Mutation , Protein Binding , RNA Interference , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins
11.
PLoS One ; 9(6): e101087, 2014.
Article En | MEDLINE | ID: mdl-24972036

BACKGROUND: Infection-related exacerbations of respiratory diseases are a major health concern; thus understanding the mechanisms driving them is of paramount importance. Despite distinct inflammatory profiles and pathological differences, asthma and COPD share a common clinical facet: raised airway ATP levels. Furthermore, evidence is growing to suggest that infective agents can cause the release of extracellular vesicle (EVs) in vitro and in bodily fluids. ATP can evoke the P2X7/caspase 1 dependent release of IL-1ß/IL-18 from EVs; these cytokines are associated with neutrophilia and are increased during exacerbations. Thus we hypothesized that respiratory infections causes the release of EVs in the airway and that the raised ATP levels, present in respiratory disease, triggers the release of IL-1ß/IL-18, neutrophilia and subsequent disease exacerbations. METHODS: To begin to test this hypothesis we utilised human cell-based assays, ex vivo murine BALF, in vivo pre-clinical models and human samples to test this hypothesis. RESULTS: Data showed that in a murine model of COPD, known to have increased airway ATP levels, infective challenge causes exacerbated inflammation. Using cell-based systems, murine models and samples collected from challenged healthy subjects, we showed that infection can trigger the release of EVs. When exposed to ATP the EVs release IL-1ß/IL-18 via a P2X7/caspase-dependent mechanism. Furthermore ATP challenge can cause a P2X7 dependent increase in LPS-driven neutrophilia. CONCLUSIONS: This preliminary data suggests a possible mechanism for how infections could exacerbate respiratory diseases and may highlight a possible signalling pathway for drug discovery efforts in this area.


Asthma/metabolism , Cell-Derived Microparticles/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Respiratory Tract Infections/metabolism , Adenosine Triphosphate/pharmacology , Animals , Asthma/complications , Caspases/metabolism , Cell Line, Tumor , Cell-Derived Microparticles/drug effects , Cell-Derived Microparticles/ultrastructure , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive/complications , Receptors, Purinergic P2X7/metabolism , Respiratory Tract Infections/complications
12.
FEBS J ; 280(3): 892-903, 2013 Feb.
Article En | MEDLINE | ID: mdl-23281710

Mediator release from mast cells is a critical step in allergic and inflammatory disease. However, the processes regulating the latter stages of granule release are yet to be fully understood. Rab27 small GTPases regulate release of secretory lysosomes in a variety of cells, including mast cell granules. In the present study, using murine bone marrow-derived mast cells (BMMC) from Rab27-deficient mutant mice, we found that, in contrast to Rab27b, Rab27a primarily plays an inhibitory role in regulating degranulation. Immunofluorescence analysis revealed that resting Rab27a-deficient (ashen) BMMCs display abnormal cortical F-actin distribution. Actin disassembly prior to IgE cross-linking increased wild-type BMMC secretion to ashen levels, suggesting that changes in the integrity of cortical F-actin underlie the ashen phenotype. Comparison of the secretory impairment of Rab27b knockout and Rab27a/b double knockout BMMCs highlighted a secondary positive role for Rab27a in enhancing degranulation. Rab27 is known to interact with actin via its effectors melanophilin (Mlph) and myosin Va (MyoVa) in other cell types. To better understand the differing roles of Rab27 proteins, we analysed the secretory phenotype of BMMCs derived from mice lacking Rab27 effector proteins. These experiments revealed that the phenotype of BMMCs deficient in Mlph (leaden) and BMMCs deficient in MyoVa (dilute) resembles the hyper-secretion of ashen BMMCs, while Munc13-4-deficient (jinx) BMMCs phenocopy the Rab27b knockout and double Rab27a/b knockout secretory impairment. We conclude that Rab27a and Rab27b regulate distinct steps in the BMMC degranulation pathway, with Rab27a/Mlph/MyoVa regulating cortical actin stability upstream of Rab27a/b/Munc13-4-dependent granule exocytosis.


Adaptor Proteins, Signal Transducing/metabolism , Mast Cells/metabolism , Membrane Proteins/metabolism , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , rab GTP-Binding Proteins/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Degranulation/genetics , Cell Degranulation/physiology , Cells, Cultured , Cytoskeleton/metabolism , Exocytosis/genetics , Exocytosis/physiology , Female , Immunoblotting , Male , Mast Cells/physiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Receptors, IgE/metabolism , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins
13.
Biochem Soc Trans ; 40(6): 1398-403, 2012 Dec 01.
Article En | MEDLINE | ID: mdl-23176488

Rab GTPases are master regulators of intracellular trafficking and, in recent years, their role in the control of different aspects of tumour progression has emerged. In the present review, we show that Rab GTPases are disregulated in many cancers and have central roles in tumour cell migration, invasion, proliferation, communication with stromal cells and the development of drug resistance. As a consequence, Rab proteins may be novel potential candidates for the development of anticancer drugs and, in this context, the preliminary results obtained with an inhibitor of Rab function are also discussed.


Neoplasms/enzymology , rab GTP-Binding Proteins/physiology , Animals , Cell Cycle , Cell Movement , Disease Progression , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Protein Transport , Signal Transduction
14.
Cancer Res ; 72(19): 4920-30, 2012 Oct 01.
Article En | MEDLINE | ID: mdl-22865453

During progression from single cancer cells to a tumor mass and metastases, tumor cells send signals that can subvert their tissue microenvironment. These signals involve soluble molecules and various extracellular vesicles, including a particular type termed exosomes. The specific roles of exosomes secreted in the tumor microenvironment, however, is unclear. The small GTPases RAB27A and RAB27B regulate exocytosis of multivesicular endosomes, which lead to exosome secretion, in human HeLa cells. Here, we used mouse models to show that Rab27a blockade in mammary carcinoma cells decreased secretion of exosomes characterized by endocytic markers, but also of matrix metalloproteinase 9, which is not associated with exosomes. Rab27a blockade resulted in decreased primary tumor growth and lung dissemination of a metastatic carcinoma (4T1), but not of a nonmetastatic carcinoma (TS/A). Local growth of 4T1 tumors required mobilization of a population of neutrophil immune cells induced by Rab27a-dependent secretion of exosomes together with a specific combination of cytokines and/or metalloproteinases. Our findings offer in vivo validation of the concept that exosome secretion can exert key pathophysiologic roles during tumor formation and progression, but they also highlight the idiosyncratic character of the tumor context.


Exosomes/metabolism , Mammary Neoplasms, Experimental/metabolism , Tumor Microenvironment , rab GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Chemokines/metabolism , Cytokines/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinases, Secreted/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Neutrophils/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/genetics , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins
15.
J Cell Sci ; 125(Pt 7): 1652-6, 2012 Apr 01.
Article En | MEDLINE | ID: mdl-22375060

Neutrophil migration is vital for immunity and precedes effector functions such as pathogen killing. Here, we report that this process is regulated by the Rab27a GTPase, a protein known to control granule exocytosis. Rab27a-deficient (Rab27a KO) neutrophils exhibit migration defects in vitro and in vivo, and live-cell microscopy suggests that delayed uropod detachment causes the migratory defect. Surface expression of CD11b, a key adhesion molecule, is increased in chemokine-stimulated Rab27a KO neutrophils compared with the control, suggesting a turnover delay caused by a defect in elastase secretion from azurophilic granules at the rear of bone marrow polymorphonuclear leukocytes (BM-PMNs). We suggest that Rab27a-dependent protease secretion regulates neutrophil migration through proteolysis-dependent de-adhesion of uropods, a mechanism that could be conserved in cell migration and invasion.


Neutrophils/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cell Movement , Mice , Mice, Knockout , Mice, Transgenic , Neutrophils/cytology , rab GTP-Binding Proteins/deficiency , rab27 GTP-Binding Proteins
16.
J Cell Biol ; 181(6): 985-98, 2008 Jun 16.
Article En | MEDLINE | ID: mdl-18541705

Invadopodia are actin-based membrane protrusions formed at contact sites between invasive tumor cells and the extracellular matrix with matrix proteolytic activity. Actin regulatory proteins participate in invadopodia formation, whereas matrix degradation requires metalloproteinases (MMPs) targeted to invadopodia. In this study, we show that the vesicle-tethering exocyst complex is required for matrix proteolysis and invasion of breast carcinoma cells. We demonstrate that the exocyst subunits Sec3 and Sec8 interact with the polarity protein IQGAP1 and that this interaction is triggered by active Cdc42 and RhoA, which are essential for matrix degradation. Interaction between IQGAP1 and the exocyst is necessary for invadopodia activity because enhancement of matrix degradation induced by the expression of IQGAP1 is lost upon deletion of the exocyst-binding site. We further show that the exocyst and IQGAP1 are required for the accumulation of cell surface membrane type 1 MMP at invadopodia. Based on these results, we propose that invadopodia function in tumor cells relies on the coordination of cytoskeletal assembly and exocytosis downstream of Rho guanosine triphosphatases.


Breast Neoplasms/metabolism , Vesicular Transport Proteins/metabolism , cdc42 GTP-Binding Protein/metabolism , ras GTPase-Activating Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Cell Line , Cell Line, Tumor , Cross-Linking Reagents/pharmacology , Female , Humans , Matrix Metalloproteinase 14/metabolism , Models, Biological , Mutant Proteins/metabolism , Neoplasm Invasiveness , Protein Binding/drug effects , Protein Processing, Post-Translational/drug effects , Protein Structure, Tertiary , Protein Subunits/metabolism , Protein Transport/drug effects , Pseudopodia/drug effects , Pseudopodia/enzymology , ras GTPase-Activating Proteins/chemistry
17.
Curr Biol ; 18(12): 926-31, 2008 Jun 24.
Article En | MEDLINE | ID: mdl-18571410

Proteolytic degradation of the extracellular matrix (ECM) is one intrinsic property of metastatic tumor cells to breach tissue barriers and to disseminate into different tissues. This process is initiated by the formation of invadopodia, which are actin-driven, finger-like membrane protrusions. Yet, little is known on how invadopodia are endowed with the functional machinery of proteolytic enzymes [1, 2]. The key protease MT1-MMP (membrane type 1-matrix metalloproteinase) confers proteolytic activity to invadopodia and thus invasion capacity of cancer cells [3-6]. Here, we report that MT1-MMP-dependent matrix degradation at invadopodia is regulated by the v-SNARE TI-VAMP/VAMP7, hence providing the molecular inventory mediating focal degradative activity of cancer cells. As observed by TIRF microscopy, MT1-MMP-mCherry and GFP-VAMP7 were simultaneously detected at proteolytic sites. Functional ablation of VAMP7 decreased the ability of breast cancer cells to degrade and invade in a MT1-MMP-dependent fashion. Moreover, the number of invadopodia was dramatically decreased in VAMP7- and MT1-MMP-depleted cells, indicative of a positive-feedback loop in which the protease as a cargo of VAMP7-targeted transport vesicles regulates maturation of invadopodia. Collectively, these data point to a specific role of VAMP7 in delivering MT1-MMP to sites of degradation, maintaining the functional machinery required for invasion.


Breast Neoplasms/pathology , Cell Surface Extensions/enzymology , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 14/metabolism , R-SNARE Proteins/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Humans , Matrix Metalloproteinase 14/genetics
19.
J Biol Chem ; 278(36): 33763-73, 2003 Sep 05.
Article En | MEDLINE | ID: mdl-12826660

Rv1395 is annotated as a potential transcriptional regulator of the AraC family. The Rv1395 insertional mutant was identified in a signature tag mutagenesis study in Mycobacterium tuberculosis and was shown to be attenuated in the lungs of mice. Here, we used comparative genomics and biochemical methods to show that Rv1395 is unique to the M. tuberculosis complex and that it encodes a protein that binds the region between two divergent genes, a member of the cytochrome P450 family (Rv1394c or cyp132) and Rv1395 itself. Rv1395 binds to this DNA region by its helix-turn-helix-containing C-terminal domain, and it recognizes two sites with different affinity. We identified the transcriptional start points (TSP) of Rv1394c and Rv1395: both genes have two TSPs, three of which are located in the intergenic region. We constructed and compared various transcriptional fusions consisting of the promoter regions and a reporter gene in Mycobacterium smegmatis: this showed that Rv1395 induces the expression of the cytochrome P450 gene (Rv1394c) and represses its own transcription. This was confirmed in M. tuberculosis when the wild type and a Rv1395-overexpressing strain were used as hosts for the fusions. Site-directed mutagenesis showed that Rv1395 binds to the two sites in a co-operative manner and that binding to both sites is required for Rv1395 optimal activity. A model describing the potential mode of action of Rv1395 is discussed.


Bacterial Proteins , Cytochrome P-450 Enzyme System/metabolism , Mycobacterium tuberculosis/metabolism , Repressor Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/physiology , Transcription, Genetic , Amino Acid Motifs , AraC Transcription Factor , Base Sequence , Binding Sites , Deoxyribonuclease I/metabolism , Dose-Response Relationship, Drug , Models, Genetic , Molecular Sequence Data , Mutagenesis, Site-Directed , Mycobacterium smegmatis/metabolism , Open Reading Frames , Protein Binding , Protein Structure, Tertiary , beta-Galactosidase/metabolism
20.
Cell Microbiol ; 5(1): 65-73, 2003 Jan.
Article En | MEDLINE | ID: mdl-12542471

Erp (exported repetitive protein), also known as P36, Pirg and Rv3810, is a member of a mycobacteria-specific family of extracellular proteins. These proteins consist of three domains, the N- and C-terminal domains are similar in all mycobacterial species, however, the central domain contains a repeated PGLTS module and differs considerably between species. The erp knockout mutant of Mycobacterium tuberculosis displays very low levels of multiplication both in macrophage cell lines and in vivo in a mouse model of infection. The high interspecies variability of the central repeated region of the Erp protein led us to investigate whether these orthologous proteins were functionally equivalent in a mouse model of tuberculosis. We expressed a gene fusion with the erp gene of Mycobacterium smegmatis, Mycobacterium leprae or M. tuberculosis in trans in an erp-M. tuberculosis mutant and found that these three alleles restored multiplication to similar levels in the spleen of infected mice. However, these alleles gave different levels of colonization in the lung, for the early time-points. Quantitative histological analyses of the lungs of infected animals showed that the nature of the erp allele strongly affected the number and the size of lung lesions, demonstrating the importance of surface determinants for virulence and tissue damage.


Alleles , Bacterial Proteins/genetics , Membrane Proteins/genetics , Mycobacterium tuberculosis/pathogenicity , Tuberculosis, Pulmonary/microbiology , Animals , Bacterial Proteins/metabolism , Cells, Cultured , Female , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Tuberculosis, Pulmonary/pathology
...