Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 22(10): 1115-1127, 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37721536

ABSTRACT

Genome-wide gene expression analysis and animal modeling indicate that melanoma differentiation associated gene-9 (mda-9, Syntenin, Syndecan binding protein, referred to as MDA-9/Syntenin) positively regulates melanoma metastasis. The MDA-9/Syntenin protein contains two tandem PDZ domains serving as a nexus for interactions with multiple proteins that initiate transcription of metastasis-associated genes. Although targeting either PDZ domain abrogates signaling and prometastatic phenotypes, the integrity of both domains is critical for full biological function. Fragment-based drug discovery and NMR identified PDZ1i, an inhibitor of the PDZ1 domain that effectively blocks cancer invasion in vitro and in vivo in multiple experimental animal models. To maximize disruption of MDA-9/Syntenin signaling, an inhibitor has now been developed that simultaneously binds and blocks activity of both PDZ domains. PDZ1i was joined to the second PDZ binding peptide (TNYYFV) with a PEG linker, resulting in PDZ1i/2i (IVMT-Rx-3) that engages both PDZ domains of MDA-9/Syntenin. IVMT-Rx-3 blocks MDA-9/Syntenin interaction with Src, reduces NF-κB activation, and inhibits MMP-2/MMP-9 expression, culminating in repression of melanoma metastasis. The in vivo antimetastatic properties of IVMT-Rx-3 are enhanced when combined with an immune-checkpoint inhibitor. Collectively, our results support the feasibility of engineering MDA-9 dual-PDZ inhibitors with enhanced antimetastatic activities and applications of IVMT-Rx-3 for developing novel therapeutic strategies effectively targeting melanoma and in principle, a broad spectrum of human cancers that also overexpress MDA-9/Syntenin.


Subject(s)
Melanoma , Animals , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Syntenins/chemistry , Signal Transduction , Peptides/metabolism
2.
Eur J Med Chem ; 223: 113601, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34153575

ABSTRACT

Syntenin stimulates exosome production and its expression is upregulated in many cancers and implicated in the spread of metastatic tumor. These effects are supported by syntenin PDZ domains interacting with syndecans. We therefore aimed to develop, through a fragment-based drug design approach, novel inhibitors targeting syntenin-syndecan interactions. We describe here the optimization of a fragment, 'hit' C58, identified by in vitro screening of a PDZ-focused fragment library, which binds specifically to the syntenin-PDZ2 domain at the same binding site as the syndecan-2 peptide. X-ray crystallographic structures and computational docking were used to guide our optimization process and lead to compounds 45 and 57 (IC50 = 33 µM and 47 µM; respectively), two representatives of syntenin-syndecan interactions inhibitors, that selectively affect the syntenin-exosome release. These findings demonstrate that it is possible to identify small molecules inhibiting syntenin-syndecan interaction and exosome release that may be useful for cancer therapy.


Subject(s)
Amino Acids/pharmacology , Antineoplastic Agents/pharmacology , Benzene Derivatives/pharmacology , Exosomes/metabolism , Syntenins/metabolism , Amino Acids/chemical synthesis , Amino Acids/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Benzene Derivatives/chemical synthesis , Benzene Derivatives/metabolism , Drug Design , Humans , MCF-7 Cells , Molecular Docking Simulation , Molecular Structure , PDZ Domains , Protein Binding/drug effects , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Syndecans/metabolism , Syntenins/chemistry
3.
Int J Mol Sci ; 20(17)2019 Aug 26.
Article in English | MEDLINE | ID: mdl-31454940

ABSTRACT

Syntenin is an adaptor-like molecule that has two adjacent tandem postsynaptic density protein 95/Discs large protein/Zonula occludens 1 (PDZ) domains. The PDZ domains of syntenin recognize multiple peptide motifs with low to moderate affinity. Many reports have indicated interactions between syntenin and a plethora of proteins. Through interactions with various proteins, syntenin regulates the architecture of the cell membrane. As a result, increases in syntenin levels induce the metastasis of tumor cells, protrusion along the neurite in neuronal cells, and exosome biogenesis in various cell types. Here, we review the updated data that support various roles for syntenin in the regulation of neuronal synapses, tumor cell invasion, and exosome control.


Subject(s)
PDZ Domains , Syntenins/metabolism , Animals , Biomarkers , Cell Membrane/metabolism , Disease Susceptibility , Humans , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Structure-Activity Relationship , Synapses/metabolism , Syntenins/chemistry
4.
Mol Cancer Ther ; 18(11): 1997-2007, 2019 11.
Article in English | MEDLINE | ID: mdl-31345950

ABSTRACT

Metastasis is the primary determinant of death in patients with diverse solid tumors and MDA-9/Syntenin (SDCBP), a pro-metastatic and pro-angiogenic gene, contributes to this process. Recently, we documented that by physically interacting with IGF-1R, MDA-9/Syntenin activates STAT3 and regulates prostate cancer pathogenesis. These observations firmly established MDA-9/Syntenin as a potential molecular target in prostate cancer. MDA-9/Syntenin contains two highly homologous PDZ domains predicted to interact with a plethora of proteins, many of which are central to the cancerous process. An MDA-9/Syntenin PDZ1 domain-targeted small molecule (PDZ1i) was previously developed using fragment-based drug discovery (FBDD) guided by NMR spectroscopy and was found to be well-tolerated in vivo, had significant half-life (t 1/2 = 9 hours) and displayed substantial anti-prostate cancer preclinical in vivo activity. PDZ1i blocked tumor cell invasion and migration in vitro, and metastasis in vivo Hence, we demonstrate that PDZ1i an MDA-9/Syntenin PDZ1 target-specific small-molecule inhibitor displays therapeutic potential for prostate and potentially other cancers expressing elevated levels of MDA-9/Syntenin.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Prostatic Neoplasms/drug therapy , Small Molecule Libraries/administration & dosage , Syntenins/chemistry , Animals , Binding Sites/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Humans , Male , Mice , Protein Domains , Receptor, IGF Type 1/metabolism , Small Molecule Libraries/pharmacology , Syntenins/metabolism , Xenograft Model Antitumor Assays
5.
PLoS One ; 12(1): e0171169, 2017.
Article in English | MEDLINE | ID: mdl-28141839

ABSTRACT

Triple negative breast cancer (TNBC) progresses rapidly but lacks effective targeted therapies. Our previous study showed that downregulating syndecan-binding protein (SDCBP) in TNBC inhibits the proliferation of TNBC cells. Dasatinib is a new small-molecule inhibitor of c-src phosphorylation. The aim of this study was to investigate if SDCBP is a potential marker to indicate whether a TNBC is suitable for dasatinib therapy. This study applied co-immunoprecipitation to identify the interaction between SDCBP and c-src in TNBC cell lines. In addition, immunohistochemistry was used to investigate SDCBP and tyrosine-419 phosphorylated c-src (p-c-src-Y419) expression in TNBC tissues. SDCBP-overexpressing MDA-MB-231 cells were then constructed to evaluate the effects of dasatinib on SDCBP-induced TNBC progression in vitro and tumor formation in nude mice. We found wild-type SDCBP interacted with c-src and promoted the phosphorylation of c-src; this phosphorylation was completely blocked by dasatinib. SDCBP lacking the PDZ domain had no such effect. Among the 52 consecutive random TNBC cases examined, the expression of SDCBP was consistent with that of p-c-src-Y419, and positively correlated with histological grading or Ki-67 levels. SDCBP overexpression significantly accelerated the proliferation and cell cycle progression of the TNBC cell line MDA-MB-231; these effects were prevented by dasatinib treatment. However, the subsequent inhibition of p27 expression partially restored the proliferation and viability of the TNBC cells. The results of this study suggest that SDCBP interacts with c-src, regulates G1/S in TNBC cells, and enhances tumor cell proliferation by promoting the tyrosine phosphorylation of c-src at residue 419. Dasatinib inhibits such phosphorylation and blocks SDCBP-induced cell cycle progression. Therefore, SDCBP might be an important marker for identifying TNBC cases that are suitable for dasatinib therapy.


Subject(s)
Dasatinib/pharmacology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , src-Family Kinases/metabolism , Animals , CSK Tyrosine-Protein Kinase , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dasatinib/administration & dosage , Disease Progression , Female , Humans , Mice, Nude , PDZ Domains , Phosphorylation/drug effects , Protein Binding/drug effects , Syntenins/chemistry , Syntenins/metabolism
6.
Sci Rep ; 6: 36818, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27830760

ABSTRACT

The PDZ domain-containing scaffold protein, syntenin-1, binds to the transmembrane proteoglycan, syndecan-4, but the molecular mechanism/function of this interaction are unknown. Crystal structure analysis of syntenin-1/syndecan-4 cytoplasmic domains revealed that syntenin-1 forms a symmetrical pair of dimers anchored by a syndecan-4 dimer. The syndecan-4 cytoplasmic domain is a compact intertwined dimer with a symmetrical clamp shape and two antiparallel strands forming a cavity within the dimeric twist. The PDZ2 domain of syntenin-1 forms a direct antiparallel interaction with the syndecan-4 cytoplasmic domain, inhibiting the functions of syndecan-4 such as focal adhesion formation. Moreover, C-terminal region of syntenin-1 reveals an essential role for enhancing the molecular homodimerization. Mutation of key syntenin-1 residues involved in the syndecan-4 interaction or homodimer formation abolishes the inhibitory function of syntenin-1, as does deletion of the homodimerization-related syntenin-1 C-terminal domain. Syntenin-1, but not dimer-formation-incompetent mutants, rescued the syndecan-4-mediated inhibition of migration and pulmonary metastasis by B16F10 cells. Therefore, we conclude that syntenin-1 negatively regulates syndecan-4 function via oligomerization and/or syndecan-4 interaction, impacting cytoskeletal organization and cell migration.


Subject(s)
Syndecan-4/chemistry , Syntenins/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Movement , Crystallography, X-Ray , Humans , Lymphatic Metastasis , Melanoma, Experimental/metabolism , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , Rats , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Syndecan-4/physiology , Syntenins/physiology
7.
Nat Commun ; 7: 12101, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27386966

ABSTRACT

PDZ domain-containing proteins work as intracellular scaffolds to control spatio-temporal aspects of cell signalling. This function is supported by the ability of their PDZ domains to bind other proteins such as receptors, but also phosphoinositide lipids important for membrane trafficking. Here we report a crystal structure of the syntenin PDZ tandem in complex with the carboxy-terminal fragment of Frizzled 7 and phosphatidylinositol 4,5-bisphosphate (PIP2). The crystal structure reveals a tripartite interaction formed via the second PDZ domain of syntenin. Biophysical and biochemical experiments establish co-operative binding of the tripartite complex and identify residues crucial for membrane PIP2-specific recognition. Experiments with cells support the importance of the syntenin-PIP2 interaction for plasma membrane targeting of Frizzled 7 and c-jun phosphorylation. This study contributes to our understanding of the biology of PDZ proteins as key players in membrane compartmentalization and dynamics.


Subject(s)
Frizzled Receptors/chemistry , Phosphatidylinositol 4,5-Diphosphate/chemistry , Syntenins/chemistry , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , MCF-7 Cells , Models, Molecular , PDZ Domains , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Syntenins/genetics , Syntenins/metabolism
8.
FEBS Lett ; 590(1): 3-12, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26787460

ABSTRACT

Syntenin has crucial roles in cell adhesion, cell migration and synaptic transmission. Its closely linked postsynaptic density-95, discs large 1, zonula occludens-1 (PDZ) domains typically interact with C-terminal ligands. We profile syntenin PDZ1-2 through proteomic peptide phage display (ProP-PD) using a library that displays C-terminal regions of the human proteome. The protein recognizes a broad range of peptides, with a preference for hydrophobic motifs and has a tendency to recognize cryptic internal ligands. We validate the interaction with nectin-1 through orthogonal assays. The study demonstrates the power of ProP-PD as a complementary approach to uncover interactions of potential biological relevance.


Subject(s)
Models, Molecular , Syntenins/metabolism , Amino Acid Motifs , Animals , Binding Sites , COS Cells , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chlorocebus aethiops , Computational Biology , Humans , Hydrophobic and Hydrophilic Interactions , Immobilized Proteins/chemistry , Immobilized Proteins/genetics , Immobilized Proteins/metabolism , Kinetics , Ligands , MCF-7 Cells , Nectins , PDZ Domains , Peptide Fragments/chemistry , Peptide Fragments/classification , Peptide Fragments/metabolism , Peptide Library , Proteomics/methods , Recombinant Proteins/chemistry , Recombinant Proteins/classification , Recombinant Proteins/metabolism , Syntenins/chemistry , Syntenins/genetics , Two-Hybrid System Techniques
9.
J Biol Chem ; 290(6): 3333-48, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25505176

ABSTRACT

Melanoma differentiation associated gene-9 (MDA-9), also known as syntenin, is a novel gene that positively regulates cancer cell motility, invasion, and metastasis through distinct biochemical and signaling pathways, but how MDA-9/syntenin is regulated in response to signals with the extracellular environment and promotes tumor progression is unclear. We now demonstrate that MDA-9/syntenin is dramatically up-regulated by a combination of rFVIIa and factor F(X) in malignant melanoma. Induction of MDA-9/syntenin in melanoma was found to occur in a thrombin-independent signaling pathway and involves the PAR-1/c-Src/Rho GTPases Rac1 and Cdc42/c-Jun N-terminal kinase axis resulting in the activation of paxillin, NF-κB, and matrix metalloproteinase-2 (MMP-2). MDA-9/syntenin physically interacts with c-Src through its PDZ binding motif following stimulation of melanoma cells with rFVIIa and FX. We also document that induction of this signaling pathway is required for TF·FVIIa·Xa-induced cell migration, invasion, and metastasis by melanoma cells. The present finding uncovers a novel role of MDA-9/syntenin as an important TF·FVIIa·Xa/PAR-1-regulated gene that initiates a signaling circuit essential for cell motility and invasion of metastatic melanoma. In these contexts, targeting TF·FVIIa·Xa and its relevant downstream targets such as MDA-9/syntenin, may represent a novel therapeutic strategy to control the evolution of neoplastic cells.


Subject(s)
Cell Movement , Factor VIIa/metabolism , Melanoma/metabolism , Signal Transduction , Syntenins/metabolism , Animals , Cell Line, Tumor , Factor X/metabolism , Gene Expression Regulation, Neoplastic , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 2/metabolism , Melanoma/pathology , Mice , NF-kappa B/metabolism , NIH 3T3 Cells , Neoplasm Metastasis , PDZ Domains , Paxillin/metabolism , Protein Binding , Receptor, PAR-1/metabolism , Syntenins/chemistry , Syntenins/genetics , Up-Regulation , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
10.
PLoS Negl Trop Dis ; 8(8): e3107, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25144756

ABSTRACT

BACKGROUND: Schistosomiasis is a neglected tropical disease caused by several species of trematode of the genus Schistosoma. The disease affects more than 200 million people in the world and causes up to 280,000 deaths per year, besides having high morbidity due to chronic illness that damages internal organs. Current schistosomiasis control strategies are mainly based on chemotherapy, but many researchers believe that the best long-term strategy to control disease is a combination of drug treatment and immunization with an anti-schistosome vaccine. Among the most promising molecules as vaccine candidates are the proteins present in the tegument and digestive tract of the parasite. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we describe for the first time Schistosoma mansoni syntenin (SmSynt) and we evaluate its potential as a recombinant vaccine. We demonstrate by real-time PCR that syntenin is mainly expressed in intravascular life stages (schistosomula and adult worms) of the parasite life cycle and, by confocal microscopy, we localize it in digestive epithelia in adult worms and schistosomula. Administration of siRNAs targeting SmSynt leads to the knock-down of syntenin gene and protein levels, but this has no demonstrable impact on parasite morphology or viability, suggesting that high SmSynt gene expression is not essential for the parasites in vitro. Mice immunization with rSmSynt, formulated with Freund's adjuvant, induces a Th1-type response, as suggested by the production of IFN-γ and TNF-α by rSmSynt-stimulated cultured splenocytes. The protective effect conferred by vaccination with rSmSynt was demonstrated by 30-37% reduction of worm burden, 38-43% reduction in the number, and 35-37% reduction in the area, of liver granulomas. CONCLUSIONS/SIGNIFICANCE: Our report is the first characterization of syntenin in Schistosoma mansoni and our data suggest that this protein is a potential candidate for the development of a multi-antigen vaccine to control schistosomiasis.


Subject(s)
Antigens, Helminth , Schistosoma mansoni/immunology , Schistosomiasis mansoni , Syntenins , Vaccines/immunology , Amino Acid Sequence , Animals , Antigens, Helminth/chemistry , Antigens, Helminth/classification , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Female , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Schistosoma mansoni/genetics , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/prevention & control , Sequence Alignment , Syntenins/chemistry , Syntenins/classification , Syntenins/genetics , Syntenins/immunology
11.
Adv Exp Med Biol ; 991: 41-57, 2013.
Article in English | MEDLINE | ID: mdl-23775690

ABSTRACT

The discovery that PSD-95/Discs large/ZO-1 (PDZ) domains can function as lipid-binding modules, in particular interacting with phosphoinositides (PIs), was made more than 10 years ago (Mol Cell 9(6): 1215-1225, 2002). Confirmatory studies and a series of functional follow-ups established PDZ domains as dual specificity modules displaying both peptide and lipid binding, and prompted a rethinking of the mode of action of PDZ domains in the control of cell signaling. In this chapter, after introducing PDZ domains, PIs and methods for studying protein-lipid interactions, we focus on (i) the prevalence and the specificity of PDZ-PIs interactions, (ii) the molecular determinants of PDZ-PIs interactions, (iii) the integration of lipid and peptide binding by PDZ domains, (iv) the common features of PIs interacting PDZ domains and (v) the regulation and functional significance of PDZ-PIs interactions.


Subject(s)
PDZ Domains/physiology , Phosphatidylinositols/chemistry , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/physiology , Carrier Proteins/chemistry , Carrier Proteins/physiology , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/physiology , Humans , Membrane Proteins/chemistry , Membrane Proteins/physiology , Muscle Proteins/chemistry , Muscle Proteins/physiology , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Phosphatidylinositols/physiology , Syntenins/chemistry , Syntenins/physiology , Zonula Occludens-1 Protein/chemistry , Zonula Occludens-1 Protein/physiology
12.
PLoS One ; 8(2): e54581, 2013.
Article in English | MEDLINE | ID: mdl-23390500

ABSTRACT

BACKGROUND: PDZ domains are highly abundant protein-protein interaction modules involved in the wiring of protein networks. Emerging evidence indicates that some PDZ domains also interact with phosphoinositides (PtdInsPs), important regulators of cell polarization and signaling. Yet our knowledge on the prevalence, specificity, affinity, and molecular determinants of PDZ-PtdInsPs interactions and on their impact on PDZ-protein interactions is very limited. METHODOLOGY/PRINCIPAL FINDINGS: We screened the human proteome for PtdInsPs interacting PDZ domains by a combination of in vivo cell-localization studies and in vitro dot blot and Surface Plasmon Resonance (SPR) experiments using synthetic lipids and recombinant proteins. We found that PtdInsPs interactions contribute to the cellular distribution of some PDZ domains, intriguingly also in nuclear organelles, and that a significant subgroup of PDZ domains interacts with PtdInsPs with affinities in the low-to-mid micromolar range. In vitro specificity for the head group is low, but with a trend of higher affinities for more phosphorylated PtdInsPs species. Other membrane lipids can assist PtdInsPs-interactions. PtdInsPs-interacting PDZ domains have generally high pI values and contain characteristic clusters of basic residues, hallmarks that may be used to predict additional PtdInsPs interacting PDZ domains. In tripartite binding experiments we established that peptide binding can either compete or cooperate with PtdInsPs binding depending on the combination of ligands. CONCLUSIONS/SIGNIFICANCE: Our screen substantially expands the set of PtdInsPs interacting PDZ domains, and shows that a full understanding of the biology of PDZ proteins will require a comprehensive insight into the intricate relationships between PDZ domains and their peptide and lipid ligands.


Subject(s)
Membrane Proteins/metabolism , PDZ Domains , Peptides/metabolism , Phosphatidylinositols/metabolism , Syntenins/metabolism , Amino Acid Sequence , Bacterial Proteins , Binding Sites , Cell Line, Tumor , Genes, Reporter , High-Throughput Screening Assays , Humans , Immunoblotting , Kinetics , Ligands , Luminescent Proteins , Membrane Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Phosphatidylinositols/chemistry , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Surface Plasmon Resonance , Syntenins/chemistry
13.
FEBS Lett ; 586(10): 1445-51, 2012 May 21.
Article in English | MEDLINE | ID: mdl-22673509

ABSTRACT

Syntenin-1 is a PDZ protein involved in receptor recycling and clustering. Its two PDZ domains interact with various receptors and phosphoinositides, and are flanked by N- and C-terminal regions. Here, we report the identification of an autoinhibitory peptide stretch in the N-terminus that might be regulated by phosphorylation. We further establish that basic residues in the C-terminal region mediate electrostatic interactions with reconstituted liposomes and contribute to the plasma membrane targeting. Our study adds new components to the multi-dentate membrane targeting mechanism and highlights the role of N- and C-terminal PDZ extensions in the regulation of syntenin-1 plasma membrane localization.


Subject(s)
Guanylate Kinases/metabolism , Lipid Metabolism , Membrane Proteins/metabolism , PDZ Domains , Phosphoproteins/metabolism , Syntenins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line, Tumor , Cell Membrane/metabolism , Disks Large Homolog 4 Protein , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid , Static Electricity , Surface Plasmon Resonance , Syntenins/chemistry , Zonula Occludens-1 Protein
14.
Front Biosci (Landmark Ed) ; 17(1): 1-15, 2012 01 01.
Article in English | MEDLINE | ID: mdl-22201728

ABSTRACT

Melanoma differentiation associated gene-9 (MDA-9), synonymous with syntenin, is an adapter protein that provides a central role in regulating cell-cell and cell-matrix adhesion. MDA-9/syntenin transduces signals from the cell-surface to the interior through its interaction with a plethora of additional proteins and actively participates in intracellular trafficking and cell-surface targeting, synaptic transmission, and axonal outgrowth. Recent studies demarcate a seminal role of MDA-9/syntenin in cancer metastasis. In the context of melanoma, MDA-9/syntenin functions as a positive regulator of melanoma progression and metastasis through interactions with c-Src and promotes the formation of an active FAK/c-Src signaling complex leading to NF-k B and matrix metalloproteinase (MMP) activation. The present review provides a current perspective of our understanding of the important features of MDA-9/syntenin and its significant role in tumor cell metastasis with special focus on molecular mechanism of action.


Subject(s)
Melanoma/secondary , Syntenins/physiology , Enzyme Precursors/metabolism , Focal Adhesion Kinase 1/chemistry , Focal Adhesion Kinase 1/metabolism , Gelatinases/metabolism , Humans , Melanoma/pathology , Melanoma/physiopathology , Models, Biological , Multiprotein Complexes/chemistry , Nervous System/physiopathology , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins pp60(c-src)/chemistry , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction , Syndecans/metabolism , Syntenins/chemistry , Syntenins/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
15.
Mol Cell Biochem ; 352(1-2): 163-72, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21359963

ABSTRACT

Ubiquitination appears to be involved in proteasome-dependent proteolysis and in the membrane trafficking system including endocytosis and exocytosis. In this study, we identified MDA-9/syntenin as a novel ubiquitin-binding protein by a yeast two-hybrid system using modified ubiquitin in which lysine 48 is substituted by arginine. It has been reported that MDA-9/syntenin is a membrane-associated protein and regulates a cellular process involving endocytosis and intracellular transport. We found that MDA-9/syntenin binds to ubiquitin by a non-covalent bond and is ubiquitinated covalently. MDA-9/syntenin has no ubiquitin-binding motifs that have so far been reported, suggesting that MDA-9/syntenin physically interacts with ubiquitin via a novel binding motif. MDA-9/syntenin is stable in the cell, suggesting that ubiquitin binding of MDA-9/syntenin or ubiquitination of MDA-9/syntenin is not related to proteolysis. Furthermore, we showed that overexpression of wild-type MDA-9/syntenin enhances formation of filopodia, whereas MDA-9/syntenin lacking the PDZ domain inhibits the formation of filopodia, suggesting that MDA-9/syntenin plays an important role via interaction with ubiquitin in the regulation of cancer metastasis and invasion.


Subject(s)
Syntenins/metabolism , Ubiquitin/metabolism , Animals , Binding Sites , Cell Line , Cloning, Molecular , Humans , Protein Binding , Syntenins/chemistry , Syntenins/genetics , Two-Hybrid System Techniques , Ubiquitin/chemistry
16.
J Biol Chem ; 285(46): 35320-9, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20826818

ABSTRACT

CAPS (aka CADPS) is required for optimal vesicle exocytosis in neurons and endocrine cells where it functions to prime the exocytic machinery for Ca(2+)-triggered fusion. Fusion is mediated by trans complexes of the SNARE proteins VAMP-2, syntaxin-1, and SNAP-25 that bridge vesicle and plasma membrane. CAPS promotes SNARE complex formation on liposomes, but the SNARE binding properties of CAPS are unknown. The current work revealed that CAPS exhibits high affinity binding to syntaxin-1 and SNAP-25 and moderate affinity binding to VAMP-2. CAPS binding is specific for a subset of exocytic SNARE protein isoforms and requires membrane integration of the SNARE proteins. SNARE protein binding by CAPS is novel and mediated by interactions with the SNARE motifs in the three proteins. The C-terminal site for CAPS binding on syntaxin-1 does not overlap the Munc18-1 binding site and both proteins can co-reside on membrane-integrated syntaxin-1. As expected for a C-terminal binding site on syntaxin-1, CAPS stimulates SNARE-dependent liposome fusion with N-terminal truncated syntaxin-1 but exhibits impaired activity with C-terminal syntaxin-1 mutants. Overall the results suggest that SNARE complex formation promoted by CAPS may be mediated by direct interactions of CAPS with each of the three SNARE proteins required for vesicle exocytosis.


Subject(s)
Calcium-Binding Proteins/metabolism , Membrane Fusion , Proteolipids/metabolism , SNARE Proteins/metabolism , Animals , Binding, Competitive , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Cell Line , HEK293 Cells , Humans , Immunoblotting , Kinetics , Liposomes/chemistry , Liposomes/metabolism , Mice , Neurons/metabolism , Phosphatidylcholines/chemistry , Phosphatidylserines/chemistry , Protein Binding , Protein Multimerization , Proteolipids/chemistry , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , SNARE Proteins/chemistry , SNARE Proteins/genetics , Spodoptera , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Syntenins/chemistry , Syntenins/genetics , Syntenins/metabolism , Vesicle-Associated Membrane Protein 2/chemistry , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 2/metabolism
17.
Comp Biochem Physiol C Toxicol Pharmacol ; 152(2): 195-201, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20382261

ABSTRACT

Syntenin is a scaffolding PDZ domain-containing protein with diverse biological activities, including organization of protein complexes in the plasma membrane, regulation of B-cell development, intracellular trafficking, synaptic transmission, and cancer metastasis. In the present study, we isolated and characterized the cDNA of the olive flounder Paralichthys olivaceus syntenin, designated PoSyntenin. The full-length CDS of PoSyntenin with 5'- and 3'-UTR sequences is 2618bp long and consists of a 909bp open reading frame preceded by a 161bp 5'-UTR and followed by a 1551bp 3'-UTR. The PoSyntenin cDNA encodes a polypeptide of 302 amino acids containing two PDZ domains, which shares 61-80% homology with those of other species, including humans. Expression of the PoSyntenin mRNA was detectable from 1day post-hatching and constitutively in the brain, spleen, intestine, stomach, eye, liver, kidney, and gill of normal conditioned fish. Expression of the PoSyntenin mRNA was upregulated in the eye, liver, kidney, spleen, brain, gill, and intestine of flounder under hypoxia and was increased by treatment with the hypoxia-mimic CoCl(2) (a HIF-1 inducer) in HINAE cells. Taken together, these results suggest that PoSyntenin is a hypoxia target gene that has a potential role in the hypoxia response mechanism of fish.


Subject(s)
Flatfishes/metabolism , Hypoxia/veterinary , Syntenins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cobalt/toxicity , Hypoxia/metabolism , Molecular Sequence Data , PDZ Domains , Syntenins/chemistry , Syntenins/genetics
18.
Cancer Res ; 70(4): 1645-55, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20145126

ABSTRACT

Aberrant adhesion signaling pathways in cancer cells underlie their deadly invasive capabilities. The adhesion-related PDZ adapter protein mda-9/syntenin is a positive regulator of cancer cell progression in breast cancer, melanoma, and other human cancers. In this study, we report that mda-9/syntenin mediates adhesion-mediated activation of protein kinase Calpha (PKCalpha) and focal adhesion kinase (FAK) by fibronectin (FN) in human breast cancer and melanoma cells. FN rapidly stimulated the expression of mda-9/syntenin and the activation of PKCalpha prior to activation of FAK. Inhibiting PKCalpha suppressed basal or FN-induced expression of mda-9/syntenin, as well as cell migration and invasion toward FN stimulated by mda-9/syntenin. Several lines of evidence suggested that activation of PKCalpha and expression of mda-9/syntenin were interdependent. First, mda-9/syntenin inhibition suppressed basal or FN-induced phosphorylation of PKCalpha at Thr(638/641), whereas PKCalpha inhibition suppressed basal or FN-induced expression of mda-9/syntenin. Second, inhibiting either mda-9/syntenin or PKCalpha suppressed FN-induced formation of integrin-beta(1)/FAK/c-Src signaling complexes. Third, inhibiting either mda-9/syntenin or PKCalpha suppressed FN-induced phosphorylation of FAK Tyr(397) and c-Src Tyr(416) and the induction of downstream effector signals to p38 and mitogen-activated protein kinase, Cdc42, and NF-kappaB. In summary, our findings offer evidence that mda-9/syntenin acts as a molecular adaptor linking PKCalpha and FAK activation in a pathway of FN adhesion by human breast cancer and melanoma cells.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Neoplasms/metabolism , Protein Kinase C-alpha/metabolism , Syntenins/metabolism , CSK Tyrosine-Protein Kinase , Cell Adhesion/drug effects , Cell Adhesion/genetics , Enzyme Activation/drug effects , Fibronectins/genetics , Fibronectins/physiology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrin beta1/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , PDZ Domains/physiology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase C-alpha/physiology , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/pharmacology , Receptor Cross-Talk/physiology , Syntenins/antagonists & inhibitors , Syntenins/chemistry , Syntenins/genetics , Tumor Cells, Cultured , src-Family Kinases
19.
Biophys J ; 96(2): 464-75, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19167297

ABSTRACT

Normal mode analysis using elastic network models has grown popular for probing the low-frequency collective dynamics of proteins and other biomolecular assemblies. In most previous studies, these models were validated by comparing calculated atomic fluctuations for isolated proteins with experimental temperature factors determined in the crystalline state, although there were also hints that including crystal contacts in the calculations has an impact on the comparison. In this study, a set of 83 ultra-high resolution crystal structures with experimentally determined anisotropic displacement parameters is used to evaluate several C(alpha)-based elastic network models that either ignore or treat the crystal environment in different ways; the latter include using periodic boundary conditions defined with respect to the asymmetric unit or the primitive unit cell as well as using the Born-von Kármán boundary condition that accounts for lattice vibrations. For all elastic network models, treating the crystal environment leads to better agreement with experimental anisotropic displacement parameters with the Born-von Kármán boundary condition giving the best agreement. Atomic correlations over the entire protein are clearly affected by the presence of the crystal contacts and fairly sensitive to the way that the crystal environment is treated. These observations highlight the importance of properly treating the protein system in an environment consistent with experiment when either evaluating approximate protein models or using approximate dynamic models in structural refinement application types. Finally, investigation of the scaling behaviors of the cumulative density of states and the heat capacity indicates that there are still gaps between simplified elastic models and all-atom models for proteins.


Subject(s)
Models, Molecular , Proteins/chemistry , Anisotropy , Computer Simulation , Crystallization , Environment , Hot Temperature , Models, Statistical , Neural Networks, Computer , Syntenins/chemistry , Temperature , X-Ray Diffraction/methods
20.
J Cell Sci ; 121(Pt 9): 1349-55, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18434645

ABSTRACT

One of the most challenging issues currently facing cell biologists is how signal specificity and compartmentalization is achieved, allowing extracellular stimulation to result in a unique and pre-defined intracellular outcome. For this to occur, intracellular components must be correctly positioned in both space and time. Adaptor molecules, which contain protein-interaction domains, are often involved in the assembly of multimeric complexes that organize intracellular signal-transduction pathways. One such protein is syntenin, a PDZ-domain-containing molecule that has a surprising variety and diversity of interaction partners. Here we assimilate and discuss current data that support a role for syntenin in regulating transmembrane-receptor trafficking, tumour-cell metastasis and neuronal-synapse function.


Subject(s)
Intracellular Space/metabolism , Syntenins/metabolism , Amino Acid Sequence , Humans , Ligands , Molecular Sequence Data , Neoplasm Metastasis , Neurons/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Cell Surface/metabolism , Synapses/metabolism , Syntenins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...