Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Bioinform Adv ; 3(1): vbad156, 2023.
Article in English | MEDLINE | ID: mdl-37928344

ABSTRACT

Motivation: Cells adhering to the extracellular matrix can sense and respond to a wide variety of chemical and physical features of the adhesive surface. Traction force microscopy (TFM) allows determining the tensile forces exerted by the cells on their substrate with high resolution. Results: To allow broad access of this techniques to cell biology laboratories we developed JeasyTFM, an open-source ImageJ package able to process multi-color and multi-position time-lapse pictures thus suitable for the automatic analysis of large TFM data. Availability and implementation: JEasyTFM is implemented as an ImageJ plugin and available at: http://questpharma.u-strasbg.fr/JEasyTFM.html.

2.
Cell Death Dis ; 14(3): 190, 2023 03 11.
Article in English | MEDLINE | ID: mdl-36899008

ABSTRACT

Cell invasion is a highly complex process that requires the coordination of cell migration and degradation of the extracellular matrix. In melanoma cells, as in many highly invasive cancer cell types these processes are driven by the regulated formation of adhesives structures such as focal adhesions and invasive structures like invadopodia. Structurally, focal adhesion and invadopodia are quite distinct, yet they share many protein constituents. However, quantitative understanding of the interaction of invadopodia with focal adhesion is lacking, and how invadopodia turn-over is associated with invasion-migration transition cycles remains unknown. In this study, we investigated the role of Pyk2, cortactin and Tks5 in invadopodia turnover and their relation with focal adhesions. We found that active Pyk2 and cortactin are localised at both focal adhesions and invadopodia. At invadopodia, localisation of active Pyk2 is correlated with ECM degradation. During invadopodia disassembly, Pyk2 and cortactin but not Tks5 are often relocated at nearby nascent adhesions. We also show that during ECM degradation, cell migration is reduced which is likely related to the sharing of common molecules within the two structures. Finally, we found that the dual FAK/Pyk2 inhibitor PF-431396 inhibits both focal adhesion and invadopodia activities thereby reducing both migration and ECM degradation.


Subject(s)
Melanoma , Podosomes , Humans , Cortactin/metabolism , Podosomes/metabolism , Focal Adhesion Kinase 2/metabolism , Neoplasm Invasiveness , Cell Line, Tumor , Extracellular Matrix/metabolism , Melanoma/metabolism
3.
Hepatology ; 76(4): 1164-1179, 2022 10.
Article in English | MEDLINE | ID: mdl-35388524

ABSTRACT

BACKGROUND AND AIMS: Numerous HCV entry factors have been identified, and yet information regarding their spatiotemporal dynamics is still limited. Specifically, one of the main entry factors of HCV is occludin (OCLN), a protein clustered at tight junctions (TJs), away from the HCV landing site. Thus, whether HCV particles slide toward TJs or, conversely, OCLN is recruited away from TJs remain debated. APPROACH AND RESULTS: Here, we generated CRISPR/CRISPR-associated protein 9 edited Huh7.5.1 cells expressing endogenous levels of enhanced green fluorescent protein/OCLN and showed that incoming HCV particles recruit OCLN outside TJs, independently of claudin 1 (CLDN1) expression, another important HCV entry factor located at TJs. Using ex vivo organotypic culture of hepatic slices obtained from human liver explants, a physiologically relevant model that preserves the overall tissue architecture, we confirmed that HCV associates with OCLN away from TJs. Furthermore, we showed, by live cell imaging, that increased OCLN recruitment beneath HCV particles correlated with lower HCV motility. To decipher the mechanism underlying virus slow-down upon OCLN recruitment, we performed CRISPR knockout (KO) of CLDN1, an HCV entry factor proposed to act upstream of OCLN. Although CLDN1 KO potently inhibits HCV infection, OCLN kept accumulating underneath the particle, indicating that OCLN recruitment is CLDN1 independent. Moreover, inhibition of the phosphorylation of Ezrin, a protein involved in HCV entry that links receptors to the actin cytoskeleton, increased OCLN accumulation and correlated with more efficient HCV internalization. CONCLUSIONS: Together, our data provide robust evidence that HCV particles interact with OCLN away from TJs and shed mechanistic insights regarding the manipulation of transmembrane receptor localization by extracellular virus particles.


Subject(s)
Hepatitis C , Tight Junctions , CRISPR-Associated Protein 9/metabolism , Claudin-1/genetics , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatocytes/metabolism , Humans , Occludin , Virion , Virus Internalization
4.
Cells ; 10(11)2021 11 21.
Article in English | MEDLINE | ID: mdl-34831480

ABSTRACT

EGFR (epidermal growth factor receptor), a member of the ErbB tyrosine kinase receptor family, is a clinical therapeutic target in numerous solid tumours. EGFR overexpression in glioblastoma (GBM) drives cell invasion and tumour progression. However, clinical trials were disappointing, and a molecular basis to explain these poor results is still missing. EGFR endocytosis and membrane trafficking, which tightly regulate EGFR oncosignaling, are often dysregulated in glioma. In a previous work, we showed that EGFR tyrosine kinase inhibitors, such as gefitinib, lead to enhanced EGFR endocytosis into fused early endosomes. Here, using pharmacological inhibitors, siRNA-mediated silencing, or expression of mutant proteins, we showed that dynamin 2 (DNM2), the small GTPase Rab5 and the endocytosis receptor LDL receptor-related protein 1 (LRP-1), contribute significantly to gefitinib-mediated EGFR endocytosis in glioma cells. Importantly, we showed that inhibition of DNM2 or LRP-1 also decreased glioma cell responsiveness to gefitinib during cell evasion from tumour spheroids. By highlighting the contribution of endocytosis proteins in the activity of gefitinib on glioma cells, this study suggests that endocytosis and membrane trafficking might be an attractive therapeutic target to improve GBM treatment.


Subject(s)
Endocytosis , ErbB Receptors/metabolism , Gefitinib/pharmacology , Cell Line, Tumor , Dynamin II/metabolism , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , Epidermal Growth Factor/metabolism , Gene Silencing , Humans , Low Density Lipoprotein Receptor-Related Protein-1/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , rab5 GTP-Binding Proteins/metabolism
5.
Nanoscale ; 13(20): 9236-9251, 2021 May 27.
Article in English | MEDLINE | ID: mdl-33977943

ABSTRACT

Glioblastoma are characterized by an invasive phenotype, which is thought to be responsible for recurrences and the short overall survival of patients. In the last decade, the promising potential of ultrasmall gadolinium chelate-coated gold nanoparticles (namely Au@DTDTPA(Gd)) was evidenced for image-guided radiotherapy in brain tumors. Considering the threat posed by invasiveness properties of glioma cells, we were interested in further investigating the biological effects of Au@DTDTPA(Gd) by examining their impact on GBM cell migration and invasion. In our work, exposure of U251 glioma cells to Au@DTDTPA(Gd) led to high accumulation of gold nanoparticles, that were mainly diffusely distributed in the cytoplasm of the tumor cells. Experiments pointed out a significant decrease in glioma cell invasiveness when exposed to nanoparticles. As the proteolysis activities were not directly affected by the intracytoplasmic accumulation of Au@DTDTPA(Gd), the anti-invasive effect cannot be attributed to matrix remodeling impairment. Rather, Au@DTDTPA(Gd) nanoparticles affected the intrinsic biomechanical properties of U251 glioma cells, such as cell stiffness, adhesion and generated traction forces, and significantly reduced the formation of protrusions, thus exerting an inhibitory effect on their migration capacities. Consistently, analysis of talin-1 expression and membrane expression of beta 1 integrin evoke the stabilization of focal adhesion plaques in the presence of nanoparticles. Taken together, our results highlight the interest in Au@DTDTPA(Gd) nanoparticles for the therapeutic management of astrocytic tumors, not only as a radio-enhancing agent but also by reducing the invasive potential of glioma cells.


Subject(s)
Glioma , Metal Nanoparticles , Cell Line, Tumor , Gadolinium , Glioma/drug therapy , Gold , Humans , Metal Nanoparticles/toxicity , Neoplasm Invasiveness
6.
Cancers (Basel) ; 13(8)2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33919725

ABSTRACT

The nonreceptor tyrosine kinase FAK is a promising target for solid tumor treatment because it promotes invasion, tumor progression, and drug resistance when overexpressed. Investigating the role of FAK in human melanoma cells, we found that both in situ and metastatic melanoma cells strongly express FAK, where it controls tumor cells' invasiveness by regulating focal adhesion-mediated cell motility. Inhibiting FAK in human metastatic melanoma cells with either siRNA or a small inhibitor targeting the kinase domain impaired migration but led to increased invadopodia formation and extracellular matrix degradation. Using FAK mutated at Y397, we found that this unexpected increase in invadopodia activity is due to the lack of phosphorylation at this residue. To preserve FAK-Src interaction while inhibiting pro-migratory functions of FAK, we found that altering FAK-paxillin interaction, with either FAK mutation in the focal adhesion targeting (FAT) domain or a competitive inhibitor peptide mimicking paxillin LD domains drastically reduces cell migration and matrix degradation by preserving FAK activity in the cytoplasm. In conclusion, our data show that targeting FAK-paxillin interactions could be a potential therapeutic strategy to prevent metastasis formation, and molecules targeting this interface could be alternative to inhibitors of FAK kinase activity which display unexpected effects.

7.
Biol Cell ; 113(6): 272-280, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33554340

ABSTRACT

Cancer is a multi-step disease where an initial tumour progresses through critical steps shaping, in most cases, life-threatening secondary foci called metastases. The oncogenic cascade involves genetic, epigenetic, signalling pathways, intracellular trafficking and/or metabolic alterations within cancer cells. In addition, pre-malignant and malignant cells orchestrate complex and dynamic interactions with non-malignant cells and acellular matricial components or secreted factors within the tumour microenvironment that is instrumental in the progression of the disease. As our aptitude to effectively treat cancer mostly depends on our ability to decipher, properly diagnose and impede cancer progression and metastasis formation, full characterisation of molecular complexes and cellular processes at play along the metastasis cascade is crucial. For many years, the scientific community lacked adapted imaging and molecular technologies to accurately dissect, at the highest resolution possible, tumour and stromal cells behaviour within their natural microenvironment. In that context, the NANOTUMOR consortium is a French national multi-disciplinary workforce which aims at a providing a multi-scale characterisation of the oncogenic cascade, from the atomic level to the dynamic organisation of the cell in response to genetic mutations, environmental changes or epigenetic modifications. Ultimately, this program aims at identifying new therapeutic targets using innovative drug design.


Subject(s)
Databases as Topic , Neoplasms/pathology , Humans
8.
J Biol Chem ; 295(39): 13474-13487, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32690605

ABSTRACT

Yes-associated protein (YAP) signaling has emerged as a crucial pathway in several normal and pathological processes. Although the main upstream effectors that regulate its activity have been extensively studied, the role of the endosomal system has been far less characterized. Here, we identified the late endosomal/lysosomal adaptor MAPK and mTOR activator (LAMTOR) complex as an important regulator of YAP signaling in a preosteoblast cell line. We found that p18/LAMTOR1-mediated peripheral positioning of late endosomes allows delivery of SRC proto-oncogene, nonreceptor tyrosine kinase (SRC) to the plasma membrane and promotes activation of an SRC-dependent signaling cascade that controls YAP nuclear shuttling. Moreover, ß1 integrin engagement and mechano-sensitive cues, such as external stiffness and related cell contractility, controlled LAMTOR targeting to the cell periphery and thereby late endosome recycling and had a major impact on YAP signaling. Our findings identify the late endosome recycling pathway as a key mechanism that controls YAP activity and explains YAP mechano-sensitivity.


Subject(s)
Cell Cycle Proteins/metabolism , Endosomes/metabolism , Integrin beta1/metabolism , Transcription Factors/metabolism , src-Family Kinases/metabolism , Animals , Cell Cycle Proteins/deficiency , Cell Line , HEK293 Cells , Humans , Mice , Mice, Knockout , Proto-Oncogene Mas , Signal Transduction , Transcription Factors/deficiency , src-Family Kinases/deficiency
9.
Biol Cell ; 112(5): 140-151, 2020 May.
Article in English | MEDLINE | ID: mdl-32034780

ABSTRACT

BACKGROUND INFORMATION: Claudin-1 (CLDN1) is a four-span transmembrane protein localised at cell-cell tight junctions (TJs), playing an important role in epithelial impermeability and tissue homoeostasis under physiological conditions. Moreover, CLDN1 expression is up-regulated in several cancers, and the level of CLDN1 expression has been proposed as a prognostic marker of patient survival. RESULTS: Here, we generated and characterised a novel reporter cell line expressing endogenous fluorescent levels of CLDN-1, allowing dynamic monitoring of CLDN-1 expression levels. Specifically, a hepatocellular carcinoma Huh7.5.1 monoclonal cell line was bioengineered using CRISPR/Cas9 to endogenously express a fluorescent TagRFP-T protein fused at the N-terminus of the CLDN1 protein. These cells were proved useful to measure CLDN1 expression and distribution in live cells. However, the cells were resistant to hepatitis C virus (HCV) infection, of which CLDN1 is a viral receptor, while retaining permissiveness to VSV-G-decorated pseudoparticles. Nonetheless, the TagRFP-CLDN1+/+ cell line showed expected CLDN1 protein localisation at TJs and the cell monolayer had similar impermeability and polarisation features as its wild-type counterpart. Finally, using fluorescence recovery after photobleaching (FRAP) approaches, we measured that the majority of endogenous and overexpressed TagRFP-CLDN1 diffuses rapidly within the TJ, whereas half of the overexpressed EGFP-CLDN1 proteins were stalled at TJs. CONCLUSIONS: The Huh7.5.1 TagRFP-CLDN1+/+ edited cell line showed physiological features comparable to that of non-edited cells, but became resistant to HCV infection. Our data also highlight the important impact of the fluorescent protein chosen for endogenous tagging. SIGNIFICANCE: Although HCV-related studies may not be achieved with these cells, our work provides a novel tool to study the cell biology of TJ-associated proteins and a potential screening strategy measuring CLDN1 expression levels.


Subject(s)
Claudin-1/metabolism , Gene Knock-In Techniques , Hepacivirus/physiology , Hepatocytes/metabolism , Virus Internalization , CRISPR-Cas Systems , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Cell Movement , Hepatocytes/virology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/virology
10.
Biol Cell ; 112(2): 53-72, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31859373

ABSTRACT

BACKGROUND: Fibroblasts executing directional migration position their centrosome, and their Golgi apparatus, in front of the nucleus towards the cell leading edge. Centrosome positioning relative to the nucleus has been associated to mechanical forces exerted on the centrosome by the microtubule-dependent molecular motor cytoplasmic dynein 1, and to nuclear movements such as rearward displacement and rotation events. Dynein has been proposed to regulate the position of the centrosome by exerting pulling forces on microtubules from the cell leading edge, where the motor is enriched during migration. However, the mechanism explaining how dynein acts at the front of the cells has not been elucidated. RESULTS: We present here results showing that the protein Focal Adhesion Kinase (FAK) interacts with dynein and regulates the enrichment of the dynein/dynactin complex at focal adhesions at the cell the leading edge of migrating fibroblasts. This suggests that focal adhesions provide anchoring sites for dynein during the polarisation process. In support of this, we present evidence indicating that the interaction between FAK and dynein, which is regulated by the phosphorylation of FAK on its Ser732 residue, is required for proper centrosome positioning. Our results further show that the polarisation of the centrosome can occur independently of nuclear movements. Although FAK regulates both nuclear and centrosome motilities, downregulating the interaction between FAK and dynein affects only the nuclear independent polarisation of the centrosome. CONCLUSIONS: Our work highlights the role of FAK as a key player in the regulation of several aspects of cell polarity. We thus propose a model in which the transient localisation of dynein with focal adhesions provides a tuneable mechanism to bias dynein traction forces on microtubules allowing proper centrosome positioning in front of the nucleus. SIGNIFICANCE: We unravel here a new role for the cancer therapeutic target FAK in the regulation of cell morphogenesis.


Subject(s)
Cell Movement , Cell Polarity , Dyneins/metabolism , Focal Adhesion Kinase 1/metabolism , Animals , Dyneins/genetics , Focal Adhesion Kinase 1/genetics , Mice , NIH 3T3 Cells , Protein Transport
11.
Cancers (Basel) ; 10(9)2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30134553

ABSTRACT

Focal adhesion kinase (FAK) is a cytoplasmic non-receptor protein tyrosine kinase that is overexpressed and activated in many human cancers. FAK transmits signals to a wide range of targets through both kinase-dependant and independent mechanism thereby playing essential roles in cell survival, proliferation, migration and invasion. In the past years, small molecules that inhibit FAK kinase function have been developed and show reduced cancer progression and metastasis in several preclinical models. Clinical trials have been conducted and these molecules display limited adverse effect in patients. FAK contain multiple functional domains and thus exhibit both important scaffolding functions. In this review, we describe the major FAK interactions relevant in cancer signalling and discuss how such knowledge provide rational for the development of Protein-Protein Interactions (PPI) inhibitors.

12.
Cancer Lett ; 376(2): 328-38, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27063097

ABSTRACT

Glioblastoma multiform (GBM) is the most common and most aggressive primary brain tumor. The fibronectin receptor, α5 integrin is a pertinent novel therapeutic target. Despite numerous data showing that α5 integrin support tumor cell migration and invasion, it has been reported that α5 integrin can also limit cell dispersion by increasing cell-cell interaction. In this study, we showed that α5 integrin was involved in cell-cell interaction and gliomasphere formation. α5-mediated cell-cell cohesion limited cell dispersion from spheroids in fibronectin-poor microenvironment. However, in fibronectin-rich microenvironment, α5 integrin promoted cell dispersion. Ligand-occupied α5 integrin and fibronectin were distributed in fibril-like pattern at cell-cell junction of evading cells, forming cell-cell fibrillar adhesions. Activated focal adhesion kinase was not present in these adhesions but was progressively relocalized with α5 integrin as cell migrates away from the spheroids. α5 integrin function in GBM appears to be more complex than previously suspected. As GBM overexpressed fibronectin, it is most likely that in vivo, α5-mediated dissemination from the tumor mass overrides α5-mediated tumor cell cohesion. In this respect, α5-integrin antagonists may be useful to limit GBM invasion in brain parenchyma.


Subject(s)
Brain Neoplasms/metabolism , Cell Adhesion , Cell Communication , Cell Movement , Cell-Matrix Junctions/metabolism , Extracellular Matrix/metabolism , Integrin alphaV/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Fibronectins/metabolism , Focal Adhesion Kinase 1/metabolism , Humans , Integrin alphaV/genetics , Neoplasm Invasiveness , RNA Interference , Signal Transduction , Spheroids, Cellular , Time Factors , Transfection
13.
J Cell Biochem ; 117(5): 1167-75, 2016 May.
Article in English | MEDLINE | ID: mdl-26477879

ABSTRACT

A large number of plants used in traditional medicines have been shown to possess antitumor activities. The aims of this study were to evaluate any anticancer effect of the essential oil (EO) extracted from P. tortuosus against B16F10 melanoma cancer cells in vitro as well as in vivo. In vitro, EO was shown to induce apoptosis and to inhibit migration and invasion processes. Further investigation revealed that EO decreased focal adhesion and invadopodia formation which was accompanied by a drastic downregulation of FAK, Src, ERK, p130Cas and paxillin. Moreover, EO treatment decreased the expression level of p190RhoGAP, and Grb2, which impair cell migration and actin assembly. Mice bearing the melanoma cells were used to confirm any in vivo effectiveness of the EO as an anti-tumor promoting agent. In mice dosed with 100 mg EO/kg/d (for 27 days), tumor weight was inhibited by 98% compared to that in mice that did not receive the product. In conclusion, these data suggested to us that an EO of P. tortuosus could evolve to be a potential medicinal resource for use in the treatment of cancers.


Subject(s)
Apiaceae/chemistry , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Melanoma, Experimental/drug therapy , Oils, Volatile/pharmacology , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/drug effects , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred BALB C , Neoplasm Invasiveness , Phytotherapy , Proto-Oncogene Proteins pp60(c-src)/metabolism , Tumor Burden/drug effects
14.
PLoS One ; 10(2): e0116921, 2015.
Article in English | MEDLINE | ID: mdl-25723396

ABSTRACT

The nucleocapsid protein (NCp7) of the Human immunodeficiency virus type 1 (HIV-1) is a small basic protein containing two zinc fingers. About 2000 NCp7 molecules coat the genomic RNA in the HIV-1 virion. After infection of a target cell, the viral core enters into the cytoplasm, where NCp7 chaperones the reverse transcription of the genomic RNA into the proviral DNA. As a consequence of their much lower affinity for double-stranded DNA as compared to single-stranded RNAs, NCp7 molecules are thought to be released in the cytoplasm and the nucleus of infected cells in the late steps of reverse transcription. Yet, little is known on the cellular distribution of the released NCp7 molecules and on their possible interactions with cell components. Hence, the aim of this study was to identify potential cellular partners of NCp7 and to monitor its intracellular distribution and dynamics by means of confocal fluorescence microscopy, fluorescence lifetime imaging microscopy, fluorescence recovery after photobleaching, fluorescence correlation and cross-correlation spectroscopy, and raster imaging correlation spectroscopy. HeLa cells transfected with eGFP-labeled NCp7 were used as a model system. We found that NCp7-eGFP localizes mainly in the cytoplasm and the nucleoli, where it binds to cellular RNAs, and notably to ribosomal RNAs which are the most abundant. The binding of NCp7 to ribosomes was further substantiated by the intracellular co-diffusion of NCp7 with the ribosomal protein 26, a component of the large ribosomal subunit. Finally, gradient centrifugation experiments demonstrate a direct association of NCp7 with purified 80S ribosomes. Thus, our data suggest that NCp7 molecules released in newly infected cells may primarily bind to ribosomes, where they may exert a new potential role in HIV-1 infection.


Subject(s)
HIV Infections/metabolism , HIV Infections/virology , HIV-1/metabolism , Nucleocapsid Proteins/metabolism , Carrier Proteins/metabolism , Cell Line , Gene Expression , Genes, Reporter , HIV-1/genetics , Humans , Intracellular Space/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Nucleocapsid Proteins/genetics , Protein Binding , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
15.
Cancer Cell Int ; 14: 42, 2014.
Article in English | MEDLINE | ID: mdl-24860258

ABSTRACT

BACKGROUND: Astrocytoma are known to have altered glutamate machinery that results in the release of large amounts of glutamate into the extracellular space but the precise role of glutamate in favoring cancer processes has not yet been fully established. Several studies suggested that glutamate might provoke active killing of neurons thereby producing space for cancer cells to proliferate and migrate. Previously, we observed that calcium promotes disassembly of integrin-containing focal adhesions in astrocytoma, thus providing a link between calcium signaling and cell migration. The aim of this study was to determine how calcium signaling and glutamate transmission cooperate to promote enhanced astrocytoma migration. METHODS: The wound-healing model was used to assay migration of human U87MG astrocytoma cells and allowed to monitor calcium signaling during the migration process. The effect of glutamate on calcium signaling was evaluated together with the amount of glutamate released by astrocytoma during cell migration. RESULTS: We observed that glutamate stimulates motility in serum-starved cells, whereas in the presence of serum, inhibitors of glutamate receptors reduce migration. Migration speed was also reduced in presence of an intracellular calcium chelator. During migration, cells displayed spontaneous Ca(2+) transients. L-THA, an inhibitor of glutamate re-uptake increased the frequency of Ca(2+) oscillations in oscillating cells and induced Ca(2+) oscillations in quiescent cells. The frequency of migration-associated Ca(2+) oscillations was reduced by prior incubation with glutamate receptor antagonists or with an anti-ß1 integrin antibody. Application of glutamate induced increases in internal free Ca(2+) concentration ([Ca(2+)]i). Finally we found that compounds known to increase [Ca(2+)]i in astrocytomas such as thapsigagin, ionomycin or the metabotropic glutamate receptor agonist t-ACPD, are able to induce glutamate release. CONCLUSION: Our data demonstrate that glutamate increases migration speed in astrocytoma cells via enhancement of migration-associated Ca(2+) oscillations that in turn induce glutamate secretion via an autocrine mechanism. Thus, glutamate receptors are further validated as potential targets for astrocytoma cancer therapy.

16.
Nucleic Acids Res ; 42(9): 5616-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24598253

ABSTRACT

The repair of toxic double-strand breaks (DSB) is critical for the maintenance of genome integrity. The major mechanisms that cope with DSB are: homologous recombination (HR) and classical or alternative nonhomologous end joining (C-NHEJ versus A-EJ). Because these pathways compete for the repair of DSB, the choice of the appropriate repair pathway is pivotal. Among the mechanisms that influence this choice, deoxyribonucleic acid (DNA) end resection plays a critical role by driving cells to HR, while accurate C-NHEJ is suppressed. Furthermore, end resection promotes error-prone A-EJ. Increasing evidence define Poly(ADP-ribose) polymerase 3 (PARP3, also known as ARTD3) as an important player in cellular response to DSB. In this work, we reveal a specific feature of PARP3 that together with Ku80 limits DNA end resection and thereby helps in making the choice between HR and NHEJ pathways. PARP3 interacts with and PARylates Ku70/Ku80. The depletion of PARP3 impairs the recruitment of YFP-Ku80 to laser-induced DNA damage sites and induces an imbalance between BRCA1 and 53BP1. Both events result in compromised accurate C-NHEJ and a concomitant increase in DNA end resection. Nevertheless, HR is significantly reduced upon PARP3 silencing while the enhanced end resection causes mutagenic deletions during A-EJ. As a result, the absence of PARP3 confers hypersensitivity to anti-tumoral drugs generating DSB.


Subject(s)
Cell Cycle Proteins/physiology , DNA End-Joining Repair , Poly(ADP-ribose) Polymerases/physiology , Recombinational DNA Repair , Antigens, Nuclear/metabolism , Antineoplastic Agents/pharmacology , BRCA1 Protein/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Drug Screening Assays, Antitumor , Etoposide/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ku Autoantigen , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Protein Transport , Replication Protein A/metabolism , Tumor Suppressor p53-Binding Protein 1
17.
PLoS One ; 9(3): e92059, 2014.
Article in English | MEDLINE | ID: mdl-24642576

ABSTRACT

Focal adhesion kinase (FAK) plays an important role in signal transduction pathways initiated at sites of integrin-mediated cell adhesion to the extracellular matrix. Thus, FAK is involved in many aspects of the metastatic process including adhesion, migration and invasion. Recently, several small molecule inhibitors which target FAK catalytic activity have been developed by pharmaceutical companies. The current study was aimed at addressing whether inhibiting FAK targeting to focal adhesions (FA) represents an efficient alternative strategy to inhibit FAK downstream pathways. Using a mutagenesis approach to alter the targeting domain of FAK, we constructed a FAK mutant that fails to bind paxillin. Inhibiting FAK-paxillin interactions led to a complete loss of FAK localization at FAs together with reduced phosphorylation of FAK and FAK targets such as paxillin and p130Cas. This in turn resulted in altered FA dynamics and inhibition of cell adhesion, migration and invasion. Moreover, the migration properties of cells expressing the FAK mutant were reduced as compared to FAK-/- cells. This was correlated with a decrease in both phospho-Src and phospho-p130Cas levels at FAs. We conclude that targeting FAK-paxillin interactions is an efficient strategy to reduce FAK signalling and thus may represent a target for the development of new FAK inhibitors.


Subject(s)
Crk-Associated Substrate Protein/genetics , Fibroblasts/drug effects , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation , Paxillin/genetics , src-Family Kinases/genetics , Animals , Binding Sites , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Crk-Associated Substrate Protein/metabolism , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Humans , Mice , Mutagenesis, Site-Directed , Paxillin/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Recombinant Fusion Proteins , Signal Transduction , src-Family Kinases/metabolism
18.
Invest New Drugs ; 30(6): 2121-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22170088

ABSTRACT

Glioblastoma represent the most frequent primary tumors of the central nervous system and remain among the most aggressive human cancers as available therapeutic approaches still fail to contain their invasiveness. Many studies have reported elevated expression of the Focal Adhesion Kinase (FAK) protein in glioblastoma, associated with an increase in the rates of both migration and invasion. This designates FAK as a promising target to limit invasiveness in glioblastoma. Thymoquinone (TQ), the main phytoactive compound of Nigella sativa has shown remarkable anti-neoplasic activities on a variety of cancer cells. Here, we studied the anti-invasive and anti-migratory effects of TQ on human glioblastoma cells. The results obtained indicated that TQ treatment reduced migration, adhesion and invasion of both U-87 and CCF-STTG1 cells. This was accompanied by a drastic down-regulation of FAK, associated with a reduction of ERK phosphorylation as well as MMP-2 and MMP-9 secretion. This study provides new data on FAK regulation by a natural product (TQ) which could be of a great value for the development of novel therapies in glioblastoma.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzoquinones/pharmacology , Focal Adhesion Kinase 1/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation , Fibroblasts/drug effects , Fibroblasts/physiology , Glioblastoma , Humans , Neoplasm Invasiveness , Nigella sativa
19.
Mol Biol Cell ; 22(7): 964-75, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21289086

ABSTRACT

Cell migration is a highly complex process that requires the coordinated formation of membrane protrusion and focal adhesions (FAs). Focal adhesion kinase (FAK), a major signaling component of FAs, is involved in the disassembly process of FAs through phosphorylation and dephosphorylation of its tyrosine residues, but the role of such phosphorylations in nascent FA formation and turnover near the cell front and in cell protrusion is less well understood. In the present study, we demonstrate that, depending on the phosphorylation status of Tyr-925 residue, FAK modulates cell migration via two specific mechanisms. FAK⁻/⁻ mouse embryonic fibroblasts (MEFs) expressing nonphosphorylatable Y925F-FAK show increased interactions between FAK and unphosphorylated paxillin, which lead to FA stabilization and thus decreased FA turnover and reduced cell migration. Conversely, MEFs expressing phosphomimetic Y925E-FAK display unchanged FA disassembly rates, show increase in phosphorylated paxillin in FAs, and exhibit increased formation of nascent FAs at the cell leading edges. Moreover, Y925E-FAK cells present enhanced cell protrusion together with activation of the p130(CAS)/Dock180/Rac1 signaling pathway. Together, our results demonstrate that phosphorylation of FAK at Tyr-925 is required for FAK-mediated cell migration and cell protrusion.


Subject(s)
Cell Surface Extensions/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Animals , Cells, Cultured , Crk-Associated Substrate Protein/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , Mice , Mice, Knockout , Paxillin/metabolism , Phosphorylation , rac1 GTP-Binding Protein/metabolism
20.
Cell Signal ; 23(1): 228-38, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20837140

ABSTRACT

The human NPY Y(1) receptor undergoes fast agonist-induced internalization via clathrin-coated pits then recycles back to the cell membrane. In an attempt to identify the molecular determinants involved in this process, we studied several C-terminal truncation mutants tagged with EFGP. In the absence of agonist, Y(1) receptors lacking the last 32 C-terminal amino acids (Y(1)Δ32) are constitutively internalized, unlike full-length Y(1) receptors. At steady state, internalized Y(1)Δ32 receptors co-localize with transferrin, a marker of early and recycling endosomes. Inhibition of constitutive internalization of Y(1)Δ32 receptors by hypertonic sucrose or by co-expression of Rab5aS34N, a dominant negative form of the small GTPase Rab5a or depletion of all three isoforms of Rab5 indicates the involvement of clathrin-coated pits. In contrast, a truncated receptor lacking the last 42 C-terminal amino acids (Y(1)Δ42) does not constitutively internalize, consistent with the possibility that there is a molecular determinant responsible for constitutive internalization located in the last 10 amino acids of Y(1)Δ32 receptors. We show that the agonist-independent internalization of Y(1)Δ32 receptors involves a tyrosine-based motif YXXΦ. The potential role of this motif in the behaviour of full-length Y(1) receptors has also been explored. Our results indicate that a C-terminal tyrosine-based motif is critical for the constitutive internalization of truncated Y(1)Δ32 receptors. We suggest that this motif is masked in full-length Y(1) receptors which do not constitutively internalize in the absence of agonist.


Subject(s)
Receptors, Neuropeptide Y/metabolism , Tyrosine/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Clathrin/chemistry , Clathrin/metabolism , HEK293 Cells , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Neuropeptide Y/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/genetics , Signal Transduction , Transferrin/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...