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










Publication year range
1.
J Cell Biol ; 222(7)2023 07 03.
Article in English | MEDLINE | ID: mdl-37204781

ABSTRACT

The mechanisms that regulate the spatial sorting of nonmuscle myosins-2 (NM2) isoforms and couple them mechanically to the plasma membrane are unclear. Here we show that the cytoplasmic junctional proteins cingulin (CGN) and paracingulin (CGNL1) interact directly with NM2s through their C-terminal coiled-coil sequences. CGN binds strongly to NM2B, and CGNL1 to NM2A and NM2B. Knockout (KO), exogenous expression, and rescue experiments with WT and mutant proteins show that the NM2-binding region of CGN is required for the junctional accumulation of NM2B, ZO-1, ZO-3, and phalloidin-labeled actin filaments, and for the maintenance of tight junction membrane tortuosity and apical membrane stiffness. CGNL1 expression promotes the junctional accumulation of both NM2A and NM2B and its KO results in myosin-dependent fragmentation of adherens junction complexes. These results reveal a mechanism for the junctional localization of NM2A and NM2B and indicate that, by binding to NM2s, CGN and CGNL1 mechanically couple the actomyosin cytoskeleton to junctional protein complexes to mechanoregulate the plasma membrane.


Subject(s)
Cell Membrane , Cytoskeletal Proteins , Cytoskeleton , Myosins , Adherens Junctions/metabolism , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Myosins/metabolism , Tight Junctions/metabolism
2.
J Biol Chem ; 298(8): 102138, 2022 08.
Article in English | MEDLINE | ID: mdl-35714771

ABSTRACT

The plasma membrane calcium ATPase (PMCA) extrudes calcium from the cytosol to the extracellular space to terminate calcium-dependent signaling. Although the distribution of PMCA is crucial for its function, the molecular mechanisms that regulate the localization of PMCA isoforms are not well understood. PLEKHA7 is implicated by genetic studies in hypertension and the regulation of calcium handling. PLEKHA7 recruits the small adapter protein PDZD11 to adherens junctions, and together they control the trafficking and localization of plasma membrane associated proteins, including the Menkes copper ATPase. Since PDZD11 binds to the C-terminal domain of b-isoforms of PMCA, PDZD11 and its interactor PLEKHA7 could control the localization and activity of PMCA. Here, we test this hypothesis using cultured cell model systems. We show using immunofluorescence microscopy and a surface biotinylation assay that KO of either PLEKHA7 or PDZD11 in mouse kidney collecting duct epithelial cells results in increased accumulation of endogenous PMCA at lateral cell-cell contacts and PDZ-dependent ectopic apical localization of exogenous PMCA4x/b isoform. In HeLa cells, coexpression of PDZD11 reduces membrane accumulation of overexpressed PMCA4x/b, and analysis of cytosolic calcium transients shows that PDZD11 counteracts calcium extrusion activity of overexpressed PMCA4x/b, but not PMCA4x/a, which lacks the PDZ-binding motif. Moreover, KO of PDZD11 in either endothelial (bEnd.3) or epithelial (mouse kidney collecting duct) cells increases the rate of calcium extrusion. Collectively, these results suggest that the PLEKHA7-PDZD11 complex modulates calcium homeostasis by regulating the localization of PMCA.


Subject(s)
Calcium , Carrier Proteins , Plasma Membrane Calcium-Transporting ATPases , Adherens Junctions/metabolism , Animals , Calcium/metabolism , Carrier Proteins/metabolism , Cells, Cultured , HeLa Cells , Humans , Mice , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasma Membrane Calcium-Transporting ATPases/metabolism , Protein Isoforms/metabolism
3.
J Biol Chem ; 298(4): 101797, 2022 04.
Article in English | MEDLINE | ID: mdl-35259394

ABSTRACT

Zonula occludens-1 (ZO-1), the major scaffolding protein of tight junctions (TJs), recruits the cytoskeleton-associated proteins cingulin (CGN) and paracingulin (CGNL1) to TJs by binding to their N-terminal ZO-1 interaction motif. The conformation of ZO-1 can be either folded or extended, depending on cytoskeletal tension and intramolecular and intermolecular interactions, and only ZO-1 in the extended conformation recruits the transcription factor DbpA to TJs. However, the sequences of ZO-1 that interact with CGN and CGNL1 and the role of TJ proteins in ZO-1 TJ assembly are not known. Here, we used glutathione-S-transferase pulldowns and immunofluorescence microscopy to show that CGN and CGNL1 bind to the C-terminal ZU5 domain of ZO-1 and that this domain is required for CGN and CGNL1 recruitment to TJs and to phase-separated ZO-1 condensates in cells. We show that KO of CGN, but not CGNL1, results in decreased accumulation of ZO-1 at TJs. Furthermore, ZO-1 lacking the ZU5 domain showed decreased accumulation at TJs, was detectable along lateral contacts, had a higher mobile fraction than full-length ZO-1 by fluorescence recovery after photobleaching analysis, and had a folded conformation, as determined by structured illumination microscopy of its N-terminal and C-terminal ends. The CGN-ZU5 interaction promotes the extended conformation of ZO-1, since binding of the CGN-ZO-1 interaction motif region to ZO-1 resulted in its interaction with DbpA in cells and in vitro. Together, these results show that binding of CGN to the ZU5 domain of ZO-1 promotes ZO-1 stabilization and accumulation at TJs by promoting its extended conformation.


Subject(s)
Cytoskeletal Proteins , Tight Junctions , Zonula Occludens-1 Protein , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Gene Knockdown Techniques , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Domains , Protein Folding , Protein Stability , Protein Structure, Quaternary , Tight Junctions/metabolism , Zonula Occludens-1 Protein/chemistry , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
4.
Cells ; 11(4)2022 02 11.
Article in English | MEDLINE | ID: mdl-35203278

ABSTRACT

Transmembrane proteins of adherens and tight junctions are known targets for viruses and bacterial toxins. The coronavirus receptor ACE2 has been localized at the apical surface of epithelial cells, but it is not clear whether ACE2 is localized at apical Cell-Cell junctions and whether it associates with junctional proteins. Here we explored the expression and localization of ACE2 and its association with transmembrane and tight junction proteins in epithelial tissues and cultured cells by data mining, immunoblotting, immunofluorescence microscopy, and co-immunoprecipitation experiments. ACE2 mRNA is abundant in epithelial tissues, where its expression correlates with the expression of the tight junction proteins cingulin and occludin. In cultured epithelial cells ACE2 mRNA is upregulated upon differentiation and ACE2 protein is widely expressed and co-immunoprecipitates with the transmembrane proteins ADAM17 and CD9. We show by immunofluorescence microscopy that ACE2 colocalizes with ADAM17 and CD9 and the tight junction protein cingulin at apical junctions of intestinal (Caco-2), mammary (Eph4) and kidney (mCCD) epithelial cells. These observations identify ACE2, ADAM17 and CD9 as new epithelial junctional transmembrane proteins and suggest that the cytokine-enhanced endocytic internalization of junction-associated protein complexes comprising ACE2 may promote coronavirus entry.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Intercellular Junctions/metabolism , Intercellular Junctions/virology , ADAM17 Protein/metabolism , Adherens Junctions/metabolism , Angiotensin-Converting Enzyme 2/genetics , Cadherins/metabolism , Carrier Proteins/metabolism , Cell Line , Cell Membrane Permeability , Coronavirus/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression/genetics , Tetraspanin 29/metabolism , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Transcriptome/genetics
5.
Biochim Biophys Acta Biomembr ; 1862(10): 183399, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32553946

ABSTRACT

Tight and adherens junctions are specialized sites of cell-cell interaction in epithelia and endothelia, and are involved in barrier, adhesion, and signaling functions. These functions are orchestrated by a highly organized meshwork of macromolecules in the membrane and cytoplasmic compartments. In this review, we discuss the structural organization and functions of the major cytoplasmic scaffolding and adaptor proteins of vertebrate apical junctions (ZO proteins, afadin, PLEKHA7, cingulin, paracingulin, polarity complex proteins, and a few others), focusing on their interactions with cytoskeletal and signaling proteins. Furthermore, we discuss recent results highlighting how mechanical tension, protein-protein interactions and post-translational modifications regulate the conformation and function of scaffolding proteins, and how spontaneous phase separation into biomolecular condensates contributes to apical junction assembly. Using a sequence-based algorithm, a large fraction of cytoplasmic proteins of apical junctions are predicted to be phase separating proteins (PSPs), suggesting that formation of biomolecular condensates is a general mechanism to organize cell-cell contacts by clustering proteins.


Subject(s)
Intercellular Junctions/metabolism , Vertebrates/metabolism , Animals , Biophysical Phenomena , Cytoplasm/metabolism , Ligands
6.
J Biol Chem ; 295(28): 9299-9312, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32371390

ABSTRACT

Pleckstrin homology domain-containing A7 (PLEKHA7) is a cytoplasmic protein at adherens junctions that has been implicated in hypertension, glaucoma, and responses to Staphylococcus aureus α-toxin. Complex formation between PLEKHA7, PDZ domain-containing 11 (PDZD11), tetraspanin 33, and the α-toxin receptor ADAM metallopeptidase domain 10 (ADAM10) promotes junctional clustering of ADAM10 and α-toxin-mediated pore formation. However, how the N-terminal region of PDZD11 interacts with the N-terminal tandem WW domains of PLEKHA7 and how this interaction promotes tetraspanin 33 binding to the WW1 domain is unclear. Here, we used site-directed mutagenesis, glutathione S-transferase pulldown experiments, immunofluorescence, molecular modeling, and docking experiments to characterize the mechanisms driving these interactions. We found that Asp-30 of WW1 and His-75 of WW2 interact through a hydrogen bond and, together with Thr-35 of WW1, form a binding pocket that accommodates a polyproline stretch within the N-terminal PDZD11 region. By strengthening the interactions of the ternary complex, the WW2 domain stabilized the WW1 domain and cooperatively promoted the interaction with PDZD11. Modeling results indicated that, in turn, PDZD11 binding induces a conformational rearrangement, which strengthens the ternary complex, and contributes to enlarging a "hydrophobic hot spot" region on the WW1 domain. The last two lipophilic residues of tetraspanin 33, Trp-283 and Tyr-282, were required for its interaction with PLEKHA7. Docking of the tetraspanin 33 C terminus revealed that it fits into the hydrophobic hot spot region of the accessible surface of WW1. We conclude that communication between the two tandem WW domains of PLEKHA7 and the PLEKHA7-PDZD11 interaction modulate the ligand-binding properties of PLEKHA7.


Subject(s)
Carrier Proteins/chemistry , Tetraspanins/chemistry , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Humans , Hydrogen Bonding , Mice , Mice, Knockout , Molecular Docking Simulation , Protein Domains , Protein Structure, Quaternary , Tetraspanins/genetics , Tetraspanins/metabolism
7.
Tissue Barriers ; 7(3): e1653748, 2019.
Article in English | MEDLINE | ID: mdl-31438766

ABSTRACT

The barrier function of epithelia and endothelia depends on tight junctions, which are formed by the polymerization of claudins on a scaffold of ZO proteins. Two differentially spliced isoforms of ZO-1 have been described, depending on the presence of the α domain, but the function of this domain is unclear. ZO-1 also contains a C-terminal ZU5 domain, which is involved in a mechano-sensitive intramolecular interaction with the central (ZPSG) region of ZO-1. Here we use immunoblotting and immunofluorescence to map the binding sites for commercially available monoclonal and polyclonal antibodies against ZO-1, and for a new polyclonal antibody (R3) that we developed against the ZO-1 C-terminus. We demonstrate that antibody R40.76 binds to the α domain, and the R3 antibody binds to the ZU5 domain. The (α+) isoform of ZO-1 shows higher expression in epithelial versus endothelial cells, and in differentiated versus undifferentiated primary keratinocytes, suggesting a link to epithelial differentiation and a potential molecular adaptation to junctions subjected to stronger mechanical forces. These results provide new tools and hypotheses to investigate the role of the α and ZU5 domains in ZO-1 mechano-sensing and dynamic interactions with the cytoskeleton and junctional ligands.


Subject(s)
Epithelium/metabolism , Keratinocytes/metabolism , Tight Junctions/metabolism , Zonula Occludens-1 Protein/physiology , Animals , Cell Differentiation , Humans
8.
Cell Rep ; 25(8): 2132-2147.e7, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30463011

ABSTRACT

We previously identified PLEKHA7 and other junctional proteins as host factors mediating death by S. aureus α-toxin, but the mechanism through which junctions promote toxicity was unclear. Using cell biological and biochemical methods, we now show that ADAM10 is docked to junctions by its transmembrane partner Tspan33, whose cytoplasmic C terminus binds to the WW domain of PLEKHA7 in the presence of PDZD11. ADAM10 is locked at junctions through binding of its cytoplasmic C terminus to afadin. Junctionally clustered ADAM10 supports the efficient formation of stable toxin pores. Instead, disruption of the PLEKHA7-PDZD11 complex inhibits ADAM10 and toxin junctional clustering. This promotes toxin pore removal from the cell surface through an actin- and macropinocytosis-dependent process, resulting in cell recovery from initial injury and survival. These results uncover a dock-and-lock molecular mechanism to target ADAM10 to junctions and provide a paradigm for how junctions regulate transmembrane receptors through their clustering.


Subject(s)
ADAM10 Protein/metabolism , Bacterial Toxins/toxicity , Hemolysin Proteins/toxicity , Intercellular Junctions/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Death/drug effects , Cell Line , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Humans , Intercellular Junctions/drug effects , Microfilament Proteins/metabolism , Pinocytosis/drug effects , Protein Binding , Protein Domains , Protein Structure, Secondary , Tetraspanins/metabolism
9.
Cell Death Dis ; 9(5): 527, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29743521

ABSTRACT

Melanoma is one of the most lethal cancers when it reaches a metastatic stage. Despite advancements in targeted therapies (BRAF inhibitors) or immunotherapies (anti-CTLA-4 or anti-PD1), most patients with melanoma will need additional treatment. Thus, there is an urgent need to develop new therapeutical approaches to bypass resistance and achieve more prolonged responses. In this context, we were interested in E2F1, a transcription factor that plays a major role in the control of cell cycle under physiological and pathological conditions. Here we confirmed that E2F1 is highly expressed in melanoma cells. Inhibition of E2F1 activity further increased melanoma cell death and senescence, both in vitro and in vivo. Moreover, blocking E2F1 also induced death of melanoma cells resistant to BRAF inhibitors. In conclusion, our studies suggest that targeting the E2F1 signaling pathway may be therapeutically relevant for melanoma.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Cell Death/drug effects , E2F1 Transcription Factor , Melanoma, Experimental , Signal Transduction/drug effects , Animals , Cell Line, Tumor , E2F1 Transcription Factor/antagonists & inhibitors , E2F1 Transcription Factor/metabolism , Female , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Nude , Neoplasm Metastasis , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism
11.
Cell Death Differ ; 25(11): 2010-2022, 2018 11.
Article in English | MEDLINE | ID: mdl-29515254

ABSTRACT

HACE1 is an E3 ubiquitin ligase described as a tumour suppressor because HACE1-knockout mice develop multi-organ, late-onset cancers and because HACE1 expression is lost in several neoplasms, such as Wilms' tumours and colorectal cancer. However, a search of public databases indicated that HACE1 expression is maintained in melanomas. We demonstrated that HACE1 promoted melanoma cell migration and adhesion in vitro and was required for mouse lung colonisation by melanoma cells in vivo. Transcriptomic analysis of HACE1-depleted melanoma cells revealed an inhibition of ITGAV and ITGB1 as well changes in other genes involved in cell migration. We revealed that HACE1 promoted the K27 ubiquitination of fibronectin and regulated its secretion. Secreted fibronectin regulated ITGAV and ITGB1 expression, as well as melanoma cell adhesion and migration. Our findings disclose a novel molecular cascade involved in the regulation of fibronectin secretion, integrin expression and melanoma cell adhesion. By controlling this cascade, HACE1 displays pro-tumoural properties and is an important regulator of melanoma cell invasive properties.


Subject(s)
Ubiquitin-Protein Ligases/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Fibronectins/metabolism , Humans , Integrins/genetics , Integrins/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , RNA Interference , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
12.
Biol Cell ; 109(4): 139-161, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28220498

ABSTRACT

Tissues of multicellular organisms are characterised by several types of specialised cell-cell junctions. In vertebrate epithelia and endothelia, tight and adherens junctions (AJ) play critical roles in barrier and adhesion functions, and are connected to the actin and microtubule cytoskeletons. The interaction between junctions and the cytoskeleton is crucial for tissue development and physiology, and is involved in the molecular mechanisms governing cell shape, motility, growth and signalling. The machineries which functionally connect tight and AJ to the cytoskeleton comprise proteins which either bind directly to cytoskeletal filaments, or function as adaptors for regulators of the assembly and function of the cytoskeleton. In the last two decades, specific cytoskeleton-associated junctional molecules have been implicated in mechanotransduction, revealing the existence of multimolecular complexes that can sense mechanical cues and translate them into adaptation to tensile forces and biochemical signals. Here, we summarise the current knowledge about the machineries that link tight and AJ to actin filaments and microtubules, and the molecular basis for mechanotransduction at epithelial and endothelial AJ.


Subject(s)
Cytoskeleton/metabolism , Mechanotransduction, Cellular/physiology , Animals , Cadherins/metabolism , Cell Polarity , Humans , Intercellular Junctions/metabolism
13.
Nucleic Acids Res ; 45(7): 4131-4141, 2017 04 20.
Article in English | MEDLINE | ID: mdl-27994032

ABSTRACT

Extracellular vesicles (EVs) have been shown to play an important role in intercellular communication as carriers of DNA, RNA and proteins. While the intercellular transfer of miRNA through EVs has been extensively studied, the stability of extracellular miRNA (ex-miRNA) once engulfed by a recipient cell remains to be determined. Here, we identify the ex-miRNA-directed phenotype to be transient due to the rapid decay of ex-miRNA. We demonstrate that the ex-miR-223-3p transferred from polymorphonuclear leukocytes to cancer cells were functional, as demonstrated by the decreased expression of its target FOXO1 and the occurrence of epithelial-mesenchymal transition reprogramming. We showed that the engulfed ex-miRNA, unlike endogenous miRNA, was unstable, enabling dynamic regulation and a return to a non-invasive phenotype within 8 h. This transient phenotype could be modulated by targeting XRN1/PACMAN exonuclease. Indeed, its silencing was associated with slower decay of ex-miR-223-3p and subsequently prolonged the invasive properties. In conclusion, we showed that the 'steady step' level of engulfed miRNA and its subsequent activity was dependent on the presence of a donor cell in the surroundings to constantly fuel the recipient cell with ex-miRNAs and of XRN1 exonuclease, which is involved in the decay of these imported miRNA.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Exoribonucleases/metabolism , MicroRNAs/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasms/genetics , RNA Stability , Cell Line, Tumor , Exosomes/metabolism , Humans , Neoplasm Invasiveness , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/pathology , Neutrophils/metabolism
14.
Oncotarget ; 7(50): 82804-82819, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27756874

ABSTRACT

Melanoma is one of the most lethal cancers when it reaches a metastatic stage. Despite the spectacular achievements of targeted therapies (BRAF inhibitors) or immuno-therapies (anti-CTLA4 or anti-PD1), most patients with melanoma will need additional treatments. Here we used a photoactive NADPH analogue called NS1 to induce cell death by inhibition of NADPH oxidases NOX in melanoma cells, including melanoma cells isolated from patients. In contrast, healthy melanocytes growth was unaffected by NS1 treatment.NS1 established an early Endoplasmic Reticulum stress by the early release of calcium mediated by (a) calcium-dependent redox-sensitive ion channel(s). These events initiated autophagy and apoptosis in all tested melanoma cells independently of their mutational status. The autophagy promoted by NS1 was incomplete. The autophagic flux was blocked at late stage events, consistent with the accumulation of p62, and a close localization of LC3 with NS1 associated with NS1 inhibition of NOX1 in autophagosomes. This hypothesis of a specific incomplete autophagy and apoptosis driven by NS1 was comforted by the use of siRNAs and pharmacological inhibitors blocking different processes. This study highlights the potential therapeutic interest of NS1 inducing cell death by triggering a selective ER stress and incomplete autophagy in melanoma cells harbouring wt and BRAF mutation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Melanoma/drug therapy , NADPH Oxidases/antagonists & inhibitors , NADP/pharmacology , Skin Neoplasms/drug therapy , Animals , Autophagy/drug effects , Calcium/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Microtubule-Associated Proteins/metabolism , Mutation , NADP/analogs & derivatives , NADPH Oxidases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , RAW 264.7 Cells , RNA Interference , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism , Signal Transduction/drug effects , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Time Factors , Transfection
16.
Cancer Cell ; 29(6): 805-819, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27238082

ABSTRACT

We have discovered and developed a series of molecules (thiazole benzenesulfonamides). HA15, the lead compound of this series, displayed anti-cancerous activity on all melanoma cells tested, including cells isolated from patients and cells that developed resistance to BRAF inhibitors. Our molecule displayed activity against other liquid and solid tumors. HA15 also exhibited strong efficacy in xenograft mouse models with melanoma cells either sensitive or resistant to BRAF inhibitors. Transcriptomic, proteomic, and biochemical studies identified the chaperone BiP/GRP78/HSPA5 as the specific target of HA15 and demonstrated that the interaction increases ER stress, leading to melanoma cell death by concomitant induction of autophagic and apoptotic mechanisms.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum Stress/drug effects , Melanoma/drug therapy , Sulfonamides/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Melanoma/metabolism , Mice , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
17.
Cell Discov ; 1: 15030, 2015.
Article in English | MEDLINE | ID: mdl-27462428

ABSTRACT

Specific BRAFV600E inhibitors (BRAFi) are highly effective in the treatment of melanoma. However, acquired drug resistances invariably develop after the initial response. Therefore, the identification of new mechanisms of acquired resistance gives important clues towards the development of therapies that could elicit long lasting responses. Here we report that CD271 confers resistance to BRAFi in melanoma cells. The expression of CD271 is increased by BRAFi through a stimulation of tumor necrosis factor-alpha (TNFα) secretion that leads to NF-κB signaling pathway activation. CD271 is upregulated in a subset of BRAFi-resistant melanoma cells. The inhibition of TNFα/NF-κB pathway and CD271 silencing restore the BRAFi sensitivity of resistant melanoma cells. Finally, increase of CD271 expression is validated in BRAFi-resistant xenografts tumors and also in tumors from the patients who relapsed under BRAFi. In summary, these results reveal a novel TNFα/NF-κB/CD271 axis whose activation contributes to the acquisition of resistance to BRAFi and therefore may represent a novel therapeutic target to improve the efficacy of therapy in melanoma.

18.
Oncotarget ; 5(21): 10650-64, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25296975

ABSTRACT

Nitric Oxide (NO) and Reactive oxygen species (ROS) are endogenous regulators of angiogenesis-related events as endothelial cell proliferation and survival, but NO/ROS defect or unbalance contribute to cancers. We recently designed a novel photoactive inhibitor of NO-Synthases (NOS) called NS1, which binds their NADPH site in vitro. Here, we show that NS1 inhibited NO formed in aortic rings. NS1-induced NO decrease led to an inhibition of angiogenesis in a model of VEGF-induced endothelial tubes formation. Beside this effect, NS1 reduced ROS levels in endothelial and melanoma A375 cells and in aorta. In metastatic melanoma cells, NS1 first induced a strong decrease of VEGF and blocked melanoma cell cycle at G2/M. NS1 decreased NOX(4) and ROS levels that could lead to a specific proliferation arrest and cell death. In contrast, NS1 did not perturb melanocytes growth. Altogether, NS1 revealed a possible cross-talk between eNOS- and NOX(4) -associated pathways in melanoma cells via VEGF, Erk and Akt modulation by NS1 that could be targeted to stop proliferation. NS1 thus constitutes a promising tool that modulates NO and redox stresses by targeting and directly inhibiting eNOS and, at least indirectly, NADPH oxidase(s), with great potential to control angiogenesis.


Subject(s)
Enzyme Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Light , Melanoma/metabolism , NADP/pharmacology , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Apoptosis , Blotting, Western , Cell Cycle , Cell Proliferation , Electron Spin Resonance Spectroscopy , Flow Cytometry , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Male , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, Inbred C57BL , NADP/analogs & derivatives , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neovascularization, Pathologic , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
19.
J Invest Dermatol ; 134(10): 2589-2597, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24756109

ABSTRACT

Several reports have demonstrated the inhibitory effect of metformin, a widely used drug in the treatment of type 2 diabetes, on the proliferation of many cancers including melanoma. Recently, it has been shown that metformin is able to modulate the cAMP level in the liver. As cAMP has a crucial role in melanin synthesis and skin pigmentation, we investigated the effect of metformin on melanogenesis both in vitro and in vivo. We showed that metformin led to reduced melanin content in melanoma cells and in normal human melanocytes by decreasing cAMP accumulation and cAMP-responsive element-binding protein phosphorylation. This inhibitory effect is correlated with decreased expression of master genes of melanogenesis, microphthalmia-associated transcription factor, tyrosinase, dopachrome tautomerase, and tyrosinase-related protein 1. Furthermore, we demonstrated that the antimelanogenic effect of metformin is independent of the AMPK pathway. Interestingly, topical application of metformin induced tail whitening in mice. Finally, we confirmed the antimelanogenic effect of metformin on reconstituted human epidermis and on human skin biopsies. These data emphasize the depigmenting effect of metformin and suggest a clinical strategy for using metformin in the topical treatment of hyperpigmentation disorders.


Subject(s)
Hypoglycemic Agents/pharmacology , Melanins/metabolism , Melanocytes/drug effects , Melanocytes/metabolism , Metformin/pharmacology , Skin/drug effects , Skin/metabolism , Animals , Biopsy , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Melanocytes/pathology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Microphthalmia-Associated Transcription Factor/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Pigmentation/drug effects , Time Factors
20.
Mol Cancer Ther ; 12(8): 1605-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23741061

ABSTRACT

Metformin was reported to inhibit the proliferation of many cancer cells, including melanoma cells. In this report, we investigated the effect of metformin on melanoma invasion and metastasis development. Using different in vitro approaches, we found that metformin inhibits cell invasion without affecting cell migration and independently of antiproliferation action. This inhibition is correlated with modulation of expression of proteins involved in epithelial-mesenchymal transition such as Slug, Snail, SPARC, fibronectin, and N-cadherin and with inhibition of MMP-2 and MMP-9 activation. Furthermore, our data indicate that this process is dependent on activation of AMPK and tumor suppressor protein p53. Finally, we showed that metformin inhibits melanoma metastasis development in mice using extravasation and metastasis models. The presented data reinforce the fact that metformin might be a good candidate for clinical trial in melanoma treatment.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Melanoma/metabolism , Melanoma/pathology , Metformin/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Melanoma/genetics , Metalloendopeptidases/metabolism , Mice , Neoplasm Invasiveness , Neoplasm Metastasis
SELECTION OF CITATIONS
SEARCH DETAIL
...