Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Biophys Res Commun ; 701: 149552, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38335918

ABSTRACT

The Interleukin-17 (IL17) family is a group of cytokines implicated in the etiology of several inflammatory diseases. Interleukin-17 receptor D (IL17RD), also known as Sef (similar expression to fibroblast growth factor) belonging to the family of IL17 receptors, has been shown to modulate IL17A-associated inflammatory phenotypes. The objective of this study was to test the hypothesis that IL17RD promotes endothelial cell activation and consequent leukocyte adhesion. We utilized primary human aortic endothelial cells and demonstrated that RNAi targeting of IL17RD suppressed transcript levels by 83 % compared to non-targeted controls. Further, RNAi knockdown of IL17RD decreased the adhesion of THP-1 monocytic cells onto a monolayer of aortic endothelial cells in response to IL17A. Additionally, we determined that IL17A did not significantly enhance the activation of canonical MAPK and NFκB pathways in endothelial cells, and further did not significantly affect the expression of VCAM-1 and ICAM-1 in aortic endothelial cells, which is contrary to previous findings. We also determined the functional relevance of our findings in vivo by comparing the expression of endothelial VCAM-1 and ICAM-1 and leukocyte infiltration in the aorta in Western diet-fed Il17rd null versus wild-type mice. Our results showed that although Il17rd null mice do not have significant alteration in aortic expression of VCAM-1 and ICAM-1 in endothelial cells, they exhibit decreased accumulation of proinflammatory monocytes and neutrophils, suggesting that endothelial IL17RD induced in vivo myeloid cell accumulation is not dependent on upregulation of VCAM-1 and ICAM-1 expression. We further performed proteomics analysis to identify potential molecular mediators of the IL17A/IL17RD signaling axis. Collectively, our results underscore a critical role for Il17rd in the regulation of aortic myeloid cell infiltration in the context of Western diet feeding.


Subject(s)
Endothelial Cells , Intercellular Adhesion Molecule-1 , Humans , Animals , Mice , Intercellular Adhesion Molecule-1/metabolism , Endothelial Cells/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Diet, Western , Aorta/metabolism , Myeloid Cells/metabolism , Monocytes/metabolism , Cell Adhesion , Receptors, Interleukin/metabolism
3.
Sci Rep ; 6: 23216, 2016 03 15.
Article in English | MEDLINE | ID: mdl-26976794

ABSTRACT

Sprouty (Spry) proteins have been implicated in cancer progression, but their role in triple-negative breast cancer (TNBC), a subtype of lethal and aggressive breast cancer, is unknown. Here, we reported that Spry1 is significantly expressed in TNBC specimen and MDA-MB-231 cells. To understand Spry1 regulation of signaling events controlling breast cancer phenotype, we used lentiviral delivery of human Spry1 shRNAs to suppress Spry1 expression in MDA-MB-231, an established TNBC cell line. Spry1 knockdown MDA-MB-231 cells displayed an epithelial phenotype with increased membrane E-cadherin expression. Knockdown of Spry1 impaired MDA-MB-231 cell migration, Matrigel invasion, and anchorage-dependent and -independent growth. Tumor xenografts originating from Spry1 knockdown MDA-MB-231 cells grew slower, had increased E-cadherin expression, and yielded fewer lung metastases compared to control. Furthermore, suppressing Spry1 in MDA-MB-231 cells impaired the induction of Snail and Slug expression by EGF, and this effect was associated with increased EGFR degradation and decreased EGFR/Grb2/Shp2/Gab1 signaling complex formation. The same phenotype was also observed in the TNBC cell line MDA-MB-157. Together, our results show that unlike in some tumors, where Spry may mediate tumor suppression, Spry1 plays a selective role in at least a subset of TNBC to promote the malignant phenotype via enhancing EGF-mediated mesenchymal phenotype.


Subject(s)
Epidermal Growth Factor/metabolism , Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Collagen , Drug Combinations , Female , Gene Knockdown Techniques , Humans , Laminin , Membrane Proteins/genetics , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness/pathology , Phosphoproteins/genetics , Proteoglycans , Triple Negative Breast Neoplasms/pathology
4.
J Cell Biochem ; 117(10): 2346-56, 2016 10.
Article in English | MEDLINE | ID: mdl-26950413

ABSTRACT

Sef (similar expression to fgf), also know as IL17RD, is a transmembrane protein shown to inhibit fibroblast growth factor signaling in developmental and cancer contexts; however, its role as a tumor suppressor remains to be fully elucidated. Here, we show that Sef regulates epithelial-mesenchymal transition (EMT) in breast cancer cell lines. Sef expression was highest in the normal breast epithelial cell line MCF10A, intermediate expression in MCF-7 cells and lowest in MDA-MB-231 cells. Knockdown of Sef increased the expression of genes associated with EMT, and promoted cell migration, invasion, and a fibroblastic morphology of MCF-7 cells. Overexpression of Sef inhibited the expression of EMT marker genes and inhibited cell migration and invasion in MCF-7 cells. Induction of EMT in MCF10A cells by TGF-ß and TNF-α resulted in downregulation of Sef expression concomitant with upregulation of EMT gene expression and loss of epithelial morphology. Overexpression of Sef in MCF10A cells partially blocked cytokine-induced EMT. Sef was shown to block ß-catenin mediated luciferase reporter activity and to cause a decrease in the nuclear localization of active ß-catenin. Furthermore, Sef was shown to co-immunoprecipitate with ß-catenin. In a mouse orthotopic xenograft model, Sef overexpression in MDA-MB-231 cells slowed tumor growth and reduced expression of EMT marker genes. Together, these data indicate that Sef plays a role in the negative regulation of EMT in a ß-catenin dependent manner and that reduced expression of Sef in breast tumor cells may be permissive for EMT and the acquisition of a more metastatic phenotype. J. Cell. Biochem. 117: 2346-2356, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Receptors, Interleukin/metabolism , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured
5.
Angiogenesis ; 16(4): 861-75, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23955631

ABSTRACT

Angiogenesis is mediated by signaling through receptor tyrosine kinases (RTKs), Src family kinases and adhesion receptors such as integrins, yet the mechanism how these signaling pathways regulate one another remains incompletely understood. The RTK modulator, Sprouty4 (Spry4) inhibits endothelial cell functions and angiogenesis, but the mechanisms remain to be fully elucidated. In this study, we demonstrate that Spry4 regulates angiogenesis in part by regulating endothelial cell migration. Overexpression of Spry4 in human endothelial cells inhibited migration and adhesion on vitronectin (VTN), whereas knockdown of Spry4 enhanced these behaviors. These activities were shown to be c-Src-dependent and Ras-independent. Spry4 disrupted the crosstalk between vascular endothelial growth factor-2 and integrin αVß3, the receptor for VTN. Spry4 overexpression resulted in decreased integrin ß3 protein levels in a post-transcriptional manner in part by modulating its tyrosine phosphorylation by c-Src. Conversely, knockdown of Spry4 resulted in increased integrin ß3 protein levels and tyrosine phosphorylation. Moreover, in vivo analysis revealed that Spry4 regulated integrin ß3 levels in murine embryos and yolk sacs. Our findings identify an unanticipated role for Spry4 in regulating c-Src activity and integrin ß3 protein levels, which contributes to the regulation of migration and adhesion of endothelial cells. Thus, targeting Spry4 may be exploited as a target in anti-angiogenesis therapies.


Subject(s)
Endothelial Cells/cytology , Integrin beta3/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Neovascularization, Physiologic/physiology , Nerve Tissue Proteins/physiology , src-Family Kinases/metabolism , Animals , Aorta/cytology , CSK Tyrosine-Protein Kinase , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Embryo, Mammalian/cytology , Endothelial Cells/metabolism , Enzyme Activation , Female , Human Umbilical Vein Endothelial Cells , Humans , Integrin alphaVbeta3/physiology , Integrin beta3/physiology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Phosphorylation , Phosphotyrosine/metabolism , Protein Processing, Post-Translational , Protein Stability , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Vessels/growth & development , Up-Regulation , Vascular Endothelial Growth Factor Receptor-2/physiology , Vitronectin/metabolism , Yolk Sac/cytology
6.
PLoS One ; 8(3): e58746, 2013.
Article in English | MEDLINE | ID: mdl-23554919

ABSTRACT

BACKGROUND: Changes in the vascular smooth muscle cell (VSMC) contractile phenotype occur in pathological states such as restenosis and atherosclerosis. Multiple cytokines, signaling through receptor tyrosine kinases (RTK) and PI3K/Akt and MAPK/ERK pathways, regulate these phenotypic transitions. The Spry proteins are feedback modulators of RTK signaling, but their specific roles in VSMC have not been established. METHODOLOGY/PRINCIPAL FINDINGS: Here, we report for the first time that Spry1, but not Spry4, is required for maintaining the differentiated state of human VSMC in vitro. While Spry1 is a known MAPK/ERK inhibitor in many cell types, we found that Spry1 has little effect on MAPK/ERK signaling but increases and maintains Akt activation in VSMC. Sustained Akt signaling is required for VSMC marker expression in vitro, while ERK signaling negatively modulates Akt activation and VSMC marker gene expression. Spry4, which antagonizes both MAPK/ERK and Akt signaling, suppresses VSMC differentiation marker gene expression. We show using siRNA knockdown and ChIP assays that FoxO3a, a downstream target of PI3K/Akt signaling, represses myocardin promoter activity, and that Spry1 increases, while Spry4 decreases myocardin mRNA levels. CONCLUSIONS: Together, these data indicate that Spry1 and Spry4 have opposing roles in VSMC phenotypic modulation, and Spry1 maintains the VSMC differentiation phenotype in vitro in part through an Akt/FoxO/myocardin pathway.


Subject(s)
Aorta/metabolism , Forkhead Transcription Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Myocytes, Smooth Muscle/cytology , Nerve Tissue Proteins/genetics , Phenotype , Phosphoproteins/genetics , Signal Transduction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...