Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Anticancer Agents Med Chem ; 21(14): 1901-1910, 2021.
Article in English | MEDLINE | ID: mdl-33292143

ABSTRACT

BACKGROUND: Triple-negative BC is the most aggressive type of breast cancer and its lack of responsiveness to conventional therapies requires screening of new chemical entities. Anti-migratory compounds are promising to treat metastatic cancer since they inhibit one of the main steps of the metastatic cascade. Spirocyclic compounds are non-conventional structures used as building blocks for the synthesis of biologically active molecules and considered interesting structures in the search for new targets in cancer research. OBJECTIVE: Here, we evaluated the potential of eight synthetic spirocyclohexadienones as cell migration inhibitors. METHODS: The anti-migratory ability of compounds was tested by wound healing and Boyden chamber approaches. Experiments in tubulin were performed by fluorescence and tubulin polymerization techniques. Finally, compounds were submitted to cell proliferation inhibition and flow cytometry assays to explore the mechanism by which they inhibit cell migration. RESULTS: Four compounds inhibited cell migration significantly. Analogs containing the 3,4,5-trimethoxyphenil ring at R1 position were the most potent and, thus, selected for additional experiments. Tubulin polymerization and fluorescence assays highlighted a possible binding of spirocyclohexadienones in the colchicine binding site; however, these compounds did not affect the cell cycle to the same extent as colchicine. Cell proliferation was affected and, notably, the most potent analogs induced apoptosis of tumor cells, suggesting a different mechanism by which they inhibit cell migration. CONCLUSION: We presented, for the first time, a series of eight synthetic spirocyclohexadienones with the ability to inhibit TNBC cell migration. These compounds represent a new category to be explored as anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclohexenes/pharmacology , Spiro Compounds/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclohexenes/chemical synthesis , Cyclohexenes/chemistry , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Polymerization/drug effects , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Triple Negative Breast Neoplasms/pathology , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
2.
Cell Commun Signal ; 18(1): 158, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32988382

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs) are lipid-bound particles that are naturally released from cells and mediate cell-cell communication. Integrin adhesion receptors are enriched in small EVs (SEVs) and SEV-carried integrins have been shown to promote cancer cell migration and to mediate organ-specific metastasis; however, how integrins mediate these effects is not entirely clear and could represent a combination of EV binding to extracellular matrix and cells. METHODS: To probe integrin role in EVs binding and uptake, we employed a disintegrin inhibitor (DisBa-01) of integrin binding with specificity for αvß3 integrin. EVs were purified from MDA-MB-231 cells conditioned media by serial centrifugation method. Isolated EVs were characterized by different techniques and further employed in adhesion, uptake and co-culture experiments. RESULTS: We find that SEVs secreted from MDA-MB-231 breast cancer cells carry αvß3 integrin and bind directly to fibronectin-coated plates, which is inhibited by DisBa-01. SEV coating on tissue culture plates also induces adhesion of MDA-MB-231 cells, which is inhibited by DisBa-01 treatment. Analysis of EV uptake and interchange between cells reveals that the amount of CD63-positive EVs delivered from malignant MDA-MB-231 breast cells to non-malignant MCF10A breast epithelial cells is reduced by DisBa-01 treatment. Inhibition of αvß3 integrin decreases CD63 expression in cancer cells suggesting an effect on SEV content. CONCLUSION: In summary, our findings demonstrate for the first time a key role of αvß3 integrin in cell-cell communication through SEVs. Video Abstract.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Extracellular Vesicles/metabolism , Integrin alphaVbeta3/metabolism , Breast/pathology , Cell Adhesion , Cell Line, Tumor , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Vesicles/ultrastructure , Female , Green Fluorescent Proteins/metabolism , Humans , Models, Biological , Protein Binding
3.
Biochimie ; 174: 144-158, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32360415

ABSTRACT

Angiogenesis, a crucial process in tumor progression, is mainly regulated by vascular endothelial growth factor (VEGF) and its receptor, VEGFR2. Studies have shown the interaction between α2ß1 integrin, a collagen receptor, and VEGFR2 in VEGF-driven angiogenesis in vitro and in vivo. Alternagin-C (ALT-C), an ECD-disintegrin-like protein from Bothrops alternatus snake venom, has high affinity for α2ß1 integrin and shows antiangiogenic activity in concentrations higher than 100 nM. Despite previous results, its mechanism of action on angiogenic signaling pathways has not been addressed. Here we evaluate the antiangiogenic activity of ALT-C in human umbilical vein endothelial cells (HUVECs) associated or not with VEGF, as well as its interference in the α2ß1/VEGFR2 crosstalk. ALT-C (1000 nM) affected actin cytoskeleton, decreased the number of cell filopodia, and strongly inhibited HUVEC tube formation, adhesion to type I collagen and cell migration. Down-regulation of α2ß1/VEGFR2 crosstalk by ALT-C decreased the protein content and phosphorylation of VEGFR2 and ß1 integrin subunit, inhibited ERK 1/2 and PI3K signaling and regulated FAK/Src and paxillin pathways. Furthermore, ALT-C increased the content of the autophagic markers LC3B and Beclin-1 in the presence of VEGF, which is associated with decreased angiogenesis. In conclusion, we suggest that ALT-C, after binding to α2ß1 integrin, inhibits VEGF/VEGFR2 signaling, which results in impaired angiogenesis. These results demonstrate that ALT-C may be a potential candidate for the development of antiangiogenic therapies for tumor and metastasis treatment and help to understand the complexity and fundamental role of integrin inhibition in the tumor microenvironment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Disintegrins/pharmacology , Integrin alpha2beta1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Bothrops , Cell Adhesion/drug effects , Cell Movement/drug effects , Crotalid Venoms/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Signal Transduction/drug effects
4.
Cell Commun Signal ; 17(1): 27, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894182

ABSTRACT

BACKGROUND: Integrins mediate cell adhesion, migration, and survival by connecting the intracellular machinery with the surrounding extracellular matrix. Previous studies demonstrated the interaction between αvß3 integrin and VEGF type 2 receptor (VEGFR2) in VEGF-induced angiogenesis. DisBa-01, a recombinant His-tag fusion, RGD-disintegrin from Bothrops alternatus snake venom, binds to αvß3 integrin with nanomolar affinity blocking cell adhesion to the extracellular matrix. Here we present in vitro evidence of a direct interference of DisBa-01 with αvß3/VEGFR2 cross-talk and its downstream pathways. METHODS: Human umbilical vein (HUVECs) were cultured in plates coated with fibronectin (FN) or vitronectin (VN) and tested for migration, invasion and proliferation assays in the presence of VEGF, DisBa-01 (1000 nM) or VEGF and DisBa-01 simultaneously. Phosphorylation of αvß3/VEGFR2 receptors and the activation of intracellular signaling pathways were analyzed by western blotting. Morphological alterations were observed and quantified by fluorescence confocal microscopy. RESULTS: DisBa-01 treatment of endothelial cells inhibited critical steps of VEGF-mediated angiogenesis such as migration, invasion and tubulogenesis. The blockage of αvß3/VEGFR2 cross-talk by this disintegrin decreases protein expression and phosphorylation of VEGFR2 and ß3 integrin subunit, regulates FAK/SrC/Paxillin downstream signals, and inhibits ERK1/2 and PI3K pathways. These events result in actin re-organization and inhibition of HUVEC migration and adhesion. Labelled-DisBa-01 colocalizes with αvß3 integrin and VEGFR2 in treated cells. CONCLUSIONS: Disintegrin inhibition of αvß3 integrin blocks VEGFR2 signalling, even in the presence of VEGF, which impairs the angiogenic mechanism. These results improve our understanding concerning the mechanisms of pharmacological inhibition of angiogenesis.


Subject(s)
Cell Movement/drug effects , Crotalid Venoms/pharmacology , Disintegrins/pharmacology , Human Umbilical Vein Endothelial Cells , Integrin alphaVbeta3/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cell Adhesion , Cells, Cultured , Focal Adhesion Kinase 1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Paxillin/metabolism , Phosphatidylinositol 3-Kinase/metabolism , src-Family Kinases/metabolism
5.
PLoS One ; 12(4): e0176226, 2017.
Article in English | MEDLINE | ID: mdl-28437464

ABSTRACT

The connective tissue formed by extracellular matrix (ECM) rich in fibronectin and collagen consists a barrier that cancer cells have to overpass to reach blood vessels and then a metastatic site. Cell adhesion to fibronectin is mediated by αvß3 and α5ß1 integrins through an RGD motif present in this ECM protein, thus making these receptors key targets for cell migration studies. Here we investigated the effect of an RGD disintegrin, DisBa-01, on the migration of human fibroblasts (BJ) and oral squamous cancer cells (OSCC, SCC25) on a fibronectin-rich environment. Time-lapse images were acquired on fibronectin-coated glass-bottomed dishes. Migration speed and directionality analysis indicated that OSCC cells, but not fibroblasts, showed significant decrease in both parameters in the presence of DisBa-01 (1µM and 2µM). Integrin expression levels of the α5, αv and ß3 subunits were similar in both cell lines, while ß1 subunit is present in lower levels on the cancer cells. Next, we examined whether the effects of DisBa-01 were related to changes in adhesion properties by using paxillin immunostaining and total internal reflection fluorescence TIRF microscopy. OSCCs in the presence of DisBa-01 showed increased adhesion sizes and number of maturing adhesion. The same parameters were analyzed usingß3-GFP overexpressing cells and showed that ß3 overexpression restored cell migration velocity and the number of maturing adhesion that were altered by DisBa-01. Surface plasmon resonance analysis showed that DisBa-01 has 100x higher affinity for αvß3 integrin than forα5ß1 integrin. In conclusion, our results suggest that the αvß3 integrin is the main receptor involved in cell directionality and its blockage may be an interesting alternative against metastasis.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Integrin alphaVbeta3/metabolism , Mouth Neoplasms/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Crotalid Venoms/pharmacology , Disintegrins/pharmacology , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Humans , Mouth Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...