Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 52(9): 5002-5015, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38477356

ABSTRACT

microRNAs regulate gene expression through interaction with an Argonaute protein. While some members of this protein family retain an enzymatic activity capable of cleaving RNA molecules complementary to Argonaute-bound small RNAs, the role of the slicer residues in the canonical microRNA pathway is still unclear in animals. To address this, we created Caenorhabditis elegans strains with mutated slicer residues in the endogenous ALG-1 and ALG-2, the only two slicing Argonautes essential for the miRNA pathway in this animal model. We observe that the mutation in ALG-1 and ALG-2 catalytic residues affects overall animal fitness and causes phenotypes reminiscent of miRNA defects only when grown and maintained at restrictive temperature. Furthermore, the analysis of global miRNA expression shows that the slicer residues of ALG-1 and ALG-2 contribute differentially to regulate the level of specific subsets of miRNAs in young adults. We also demonstrate that altering the catalytic tetrad of those miRNA-specific Argonautes does not result in any defect in the production of canonical miRNAs. Together, these data support that the slicer residues of miRNA-specific Argonautes contribute to maintaining levels of a set of miRNAs for optimal viability and fitness in animals particularly exposed to specific growing conditions.


Subject(s)
Argonaute Proteins , Caenorhabditis elegans Proteins , Caenorhabditis elegans , MicroRNAs , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Mutation , RNA-Binding Proteins
2.
bioRxiv ; 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-36711744

ABSTRACT

microRNAs regulate gene expression through interaction with an Argonaute protein family member. While some members of this protein family retain an enzymatic activity capable of cleaving RNA molecules complementary to Argonaute-bound small RNAs, the role of the slicing activity in the canonical microRNA pathway is still unclear in animals. To address the importance of slicing Argonautes in animals, we created Caenorhabditis elegans strains, carrying catalytically dead endogenous ALG-1 and ALG-2, the only two slicing Argonautes essential for the miRNA pathway in this animal model. We observe that the loss of ALG-1 and ALG-2 slicing activity affects overall animal fitness and causes phenotypes, reminiscent of miRNA defects, only when grown and maintained at restrictive temperature. Furthermore, the analysis of global miRNA expression shows that the catalytic activity of ALG-1 and ALG-2 differentially regulate the level of specific subsets of miRNAs in young adults. We also demonstrate that altering the slicing activity of those miRNA-specific Argonautes does not result in any defect in the production of canonical miRNAs. Together, these data support that the slicing activity of miRNA-specific Argonautes function to maintain the levels of a set of miRNAs for optimal viability and fitness in animals particularly exposed to specific growing conditions.

3.
Cell Rep ; 41(11): 111822, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36516777

ABSTRACT

Argonaute proteins are at the core of the microRNA-mediated gene silencing pathway essential for animals. In C. elegans, the microRNA-specific Argonautes ALG-1 and ALG-2 regulate multiple processes required for proper animal developmental timing and viability. Here we identified a phosphorylation site on ALG-1 that modulates microRNA association. Mutating ALG-1 serine 642 into a phospho-mimicking residue impairs microRNA binding and causes embryonic lethality and post-embryonic phenotypes that are consistent with alteration of microRNA functions. Monitoring microRNA levels in alg-1 phosphorylation mutant animals shows that microRNA passenger strands increase in abundance but are not preferentially loaded into ALG-1, indicating that the miRNA binding defects could lead to microRNA duplex accumulation. Our genetic and biochemical experiments support protein kinase A (PKA) KIN-1 as the putative kinase that phosphorylates ALG-1 serine 642. Our data indicate that PKA triggers ALG-1 phosphorylation to regulate its microRNA association during C. elegans development.


Subject(s)
Caenorhabditis elegans Proteins , MicroRNAs , Animals , Caenorhabditis elegans/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Caenorhabditis elegans Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphorylation , RNA-Binding Proteins/metabolism , Serine/metabolism
4.
PLoS Genet ; 17(4): e1009511, 2021 04.
Article in English | MEDLINE | ID: mdl-33826611

ABSTRACT

Once loaded onto Argonaute proteins, microRNAs form a silencing complex called miRISC that targets mostly the 3'UTR of mRNAs to silence their translation. How microRNAs are transported to and from their target mRNA remains poorly characterized. While some reports linked intracellular trafficking to microRNA activity, it is still unclear how these pathways coordinate for proper microRNA-mediated gene silencing and turnover. Through a forward genetic screen using Caenorhabditis elegans, we identified the RabGAP tbc-11 as an important factor for the microRNA pathway. We show that TBC-11 acts mainly through the small GTPase RAB-6 and that its regulation is required for microRNA function. The absence of functional TBC-11 increases the pool of microRNA-unloaded Argonaute ALG-1 that is likely associated to endomembranes. Furthermore, in this condition, this pool of Argonaute accumulates in a perinuclear region and forms a high molecular weight complex. Altogether, our data suggest that the alteration of TBC-11 generates a fraction of ALG-1 that cannot bind to target mRNAs, leading to defective gene repression. Our results establish the importance of intracellular trafficking for microRNA function and demonstrate the involvement of a small GTPase and its GAP in proper Argonaute localization in vivo.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Protein Biosynthesis , RNA-Binding Proteins/genetics , rab GTP-Binding Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental/genetics , Gene Silencing , MicroRNAs/genetics , RNA, Messenger/genetics
5.
Oncogene ; 40(18): 3231-3244, 2021 05.
Article in English | MEDLINE | ID: mdl-33833398

ABSTRACT

MicroRNAs and RNA-binding proteins (RBPs) primarily target the 3' UTR of mRNAs to control their translation and stability. However, their co-regulatory effects on specific mRNAs in physiology and disease are yet to be fully explored. CSDE1 is an RBP that promotes metastasis in melanoma and mechanisms underlying its oncogenic activities need to be completely defined. Here we report that CSDE1 interacts with specific miRNA-induced silencing complexes (miRISC) in melanoma. We find an association of CSDE1 with AGO2, the essential component of miRISC, which is facilitated by target mRNAs and depends on the first cold shock domain of CSDE1. Both CSDE1 and AGO2 bind to 3' UTR of PMEPA1. CSDE1 counters AGO2 binding, leading to an increase of PMEPA1 expression. We also identify a miRNA, miR-129-5p, that represses PMEPA1 expression in melanoma. Collectively, our results show that PMEPA1 promotes tumorigenic traits and that CSDE1 along with miR-129-5p/AGO2 miRISC act antagonistically to fine-tune PMEPA1 expression toward the progression of melanoma.


Subject(s)
Argonaute Proteins , MicroRNAs , HEK293 Cells , Humans , Melanoma/genetics , RNA, Messenger/genetics , RNA-Binding Proteins
6.
Life Sci Alliance ; 3(4)2020 04.
Article in English | MEDLINE | ID: mdl-32161113

ABSTRACT

In animals, miRNAs are the most prevalent small non-coding RNA molecules controlling posttranscriptional gene regulation. The Argonaute proteins (AGO) mediate miRNA-guided gene silencing by recruiting multiple factors involved in translational repression, deadenylation, and decapping. Here, we report that CSDE1, an RNA-binding protein linked to stem cell maintenance and metastasis in cancer, interacts with AGO2 within miRNA-induced silencing complex and mediates gene silencing through its N-terminal domains. We show that CSDE1 interacts with LSM14A, a constituent of P-body assembly and further associates to the DCP1-DCP2 decapping complex, suggesting that CSDE1 could promote the decay of miRNA-induced silencing complex-targeted mRNAs. Together, our findings uncover a hitherto unknown mechanism used by CSDE1 in the control of gene expression mediated by the miRNA pathway.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , MicroRNAs/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Animals , Argonaute Proteins/genetics , Drosophila melanogaster/genetics , Embryonic Stem Cells , Gene Expression/genetics , Gene Expression Regulation/genetics , Gene Silencing/physiology , HEK293 Cells , HeLa Cells , Humans , Mice , MicroRNAs/genetics , NIH 3T3 Cells , RNA Stability/genetics , RNA Stability/physiology , RNA, Messenger/genetics
7.
J Cell Sci ; 129(21): 4105-4117, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27637266

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a process by which cancer cells gain the ability to leave the primary tumor site and invade surrounding tissues. These metastatic cancer cells can further increase their plasticity by adopting an amoeboid-like morphology, by undergoing mesenchymal-to-amoeboid transition (MAT). We found that adhering cells produce spreading initiation centers (SICs), transient structures that are localized above nascent adhesion complexes, and share common biological and morphological characteristics associated with amoeboid cells. Meanwhile, spreading cells seem to return to a mesenchymal-like morphology. Thus, our results indicate that SIC-induced adhesion recapitulates events  that are associated with amoeboid-to-mesenchymal transition (AMT). We found that polyadenylated RNAs are enriched within SICs, blocking their translation decreased adhesion potential of metastatic cells that progressed through EMT. These results point to a so-far-unknown checkpoint that regulates cell adhesion and allows metastatic cells to alter adhesion strength to modulate their dissemination.


Subject(s)
Protein Biosynthesis , Transendothelial and Transepithelial Migration , Cell Adhesion , Cell Line, Tumor , Cell Shape , Enzyme Activation , Epithelial-Mesenchymal Transition , Focal Adhesions/metabolism , GTP Phosphohydrolases/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mesoderm/metabolism , Models, Biological , Neoplasm Metastasis , Polyadenylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
8.
Vasc Cell ; 4(1): 18, 2012 Nov 17.
Article in English | MEDLINE | ID: mdl-23157718

ABSTRACT

BACKGROUND: Loss of endothelial cell integrity and selective permeability barrier is an early event in the sequence of oxidant-mediated injury and may result in atherosclerosis, hypertension and facilitation of transendothelial migration of cancer cells during metastasis. We already reported that endothelial cell integrity is tightly regulated by the balanced co-activation of p38 and ERK pathways. In particular, we showed that phosphorylation of tropomyosin-1 (tropomyosin alpha-1 chain = Tm1) at Ser283 by DAP kinase, downstream of the ERK pathway might be a key event required to maintain the integrity and normal functions of the endothelium in response to oxidative stress. METHODS: Endothelial permeability was assayed by monitoring the passage of Dextran-FITC through a tight monolayer of HUVECs grown to confluence in Boyden chambers. Actin and Tm1 dynamics and distribution were evaluated by immunofluorescence. We modulated the expression of Tm1 by siRNA and lentiviral-mediated expression of wild type and mutated forms of Tm1 insensitive to the siRNA. Transendothelial migration of HT-29 colon cancer cells was monitored in Boyden chambers similarly as for permeability. RESULTS: We provide evidence indicating that Tm1 phosphorylation at Ser283 is essential to regulate endothelial permeability under oxidative stress by modulating actin dynamics. Moreover, the transendothelial migration of colon cancer cells is also regulated by the phosphorylation of Tm1 at Ser283. CONCLUSION: Our finding strongly support the role for the phosphorylation of endothelial Tm1 at Ser283 to prevent endothelial barrier dysfunction associated with oxidative stress injury.

9.
J Biol Chem ; 287(36): 30541-51, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22773844

ABSTRACT

Endothelial cell migration induced in response to vascular endothelial growth factor (VEGF) is an essential step of angiogenesis. It depends in part on the activation of the p38/MAPKAP kinase-2/LIMK1/annexin-A1 (ANXA1) signaling axis. In the present study, we obtained evidence indicating that miR-196a specifically binds to the 3'-UTR region of ANXA1 mRNA to repress its expression. In accordance with the role of ANXA1 in cell migration and angiogenesis, the ectopic expression of miR-196a is associated with decreased cell migration in wound closure assays, and the inhibitory effect of miR-196a is rescued by overexpressing ANXA1. This finding highlights the fact that ANXA1 is a required mediator of VEGF-induced cell migration. miR-196a also reduces the formation of lamellipodia in response to VEGF suggesting that ANXA1 regulates cell migration by securing the formation of lamellipodia at the leading edge of the cell. Additionally, in line with the fact that cell migration is an essential step of angiogenesis, the ectopic expression of miR-196a impairs the formation of capillary-like structures in a tissue-engineered model of angiogenesis. Here again, the effect of miR-196a is rescued by overexpressing ANXA1. Moreover, the presence of miR-196a impairs the VEGF-induced in vivo neo-vascularization in the Matrigel Plug assay. Interestingly, VEGF reduces the expression of miR-196a, which is associated with an increased level of ANXA1. Similarly, the inhibition of miR-196a with an antagomir results in an increased level of ANXA1. We conclude that the VEGF-induced decrease of miR-196a expression may participate to the angiogenic switch by maintaining the expression of ANXA1 to levels required to enable p38-ANXA1-dependent endothelial cell migration and angiogenesis in response to VEGF.


Subject(s)
Annexin A1/metabolism , Cell Movement/physiology , Gene Expression Regulation/physiology , Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/biosynthesis , Vascular Endothelial Growth Factor A/metabolism , 3' Untranslated Regions/physiology , Annexin A1/genetics , Cell Movement/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , Human Umbilical Vein Endothelial Cells/cytology , Humans , MicroRNAs/genetics , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Pseudopodia/genetics , Pseudopodia/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology , Wound Healing/drug effects , Wound Healing/physiology
10.
Breast Cancer Res ; 14(3): R92, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22691413

ABSTRACT

INTRODUCTION: Human 17beta-hydroxysteroid dehydrogenase type 1 (17ß-HSD1) is a steroid-converting enzyme that has long been known to play critical roles in estradiol synthesis and more recently in dihydrotestosterone (DHT) inactivation, showing a dual function that promotes breast cancer cell proliferation. Previously, we reported the first observation of the influence of the enzyme on endogenous estrogen-responsive gene expression. Here, we demonstrate the impact of 17ß-HSD1 expression on the breast cancer cell proteome and investigate its role in cell migration. METHODS: 17ß-HSD1 was stably transfected in MCF7 cells and the proteome of the generated cells overexpressing 17ß-HSD1 (MCF7-17ßHSD1 cells) was compared to that of the wild type MCF7 cells. Proteomics study was performed using two-dimensional gel electrophoresis followed by mass spectrometry analysis of differentially expressed protein spots. Reverse transcription quantitative real-time PCR (RT-qPCR) was used to investigate the transcription of individual gene. The effect of 17ß-HSD1 on MCF7 cell migration was verified by a wound-healing assay. RESULTS: Proteomic data demonstrate that the expression of more than 59 proteins is modulated following 17ß-HSD1 overexpression. 17ß-HSD1 regulates the expression of important genes and proteins that are relevant to cell growth control, such as BRCA2 and CDKN1A interacting protein (BCCIP) and proliferating cell nuclear antigen (PCNA) which are down- and upregulated in MCF7-17ßHSD1 cells, respectively. RT-qPCR data reveal that 17ß-HSD1 increases the mRNA levels of estrogen receptors (ER) alpha and beta by 171 and 120%, respectively, while decreasing that of the androgen receptor by 64%. Interestingly, 17ß-HSD1 increases the mRNA transcript (by 3.6 times) and the protein expression of the metastasis suppressor gene nm23-H1 and the expression of the two enzymes are closely correlated. We have further shown that 17ß-HSD1 expression is associated with an increase of MCF7 cell migration. CONCLUSIONS: In addition to the regulation of important genes, we have demonstrated for the first time that 17ß-HSD1 increases breast cancer cell migration, in spite of its positive regulation of the antimetastatic gene NM23. This is also correlated to its stimulation of breast cancer cell growth, further confirming its targeting in ER positive breast cancer. The novel findings in this study suggest several directions for future research on the contribution of 17ß-HSD1 to breast cancer progression and related treatment.


Subject(s)
Cell Movement , Estradiol Dehydrogenases/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , RNA, Messenger/metabolism , BRCA2 Protein/metabolism , Breast Neoplasms/metabolism , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Estradiol Dehydrogenases/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Humans , MCF-7 Cells , Nuclear Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Array Analysis , Proteome/analysis , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering , Receptors, Androgen/genetics
11.
Angiogenesis ; 15(4): 593-608, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22696064

ABSTRACT

Endothelial cell migration induced in response to vascular endothelial growth factor (VEGF) is a crucial step of angiogenesis and it depends on the activation of the p38 MAP-kinase pathway downstream of VEGFR2. In this study, we investigated the role of microRNAs (miRNAs) in regulating these processes. We found that the VEGF-induced p38 activation and cell migration are modulated by overexpression of Argonaute 2, a key protein in the functioning of miRNAs. Thereafter, we found that miR-20a expression is increased by VEGF and that its ectopic expression inhibits VEGF-induced actin remodeling and cell migration. Moreover, the expression of miR-20a impairs the formation of branched capillaries in a tissue-engineered model of angiogenesis. In addition, the lentivirus-mediated expression of miR-20a precursor (pmiR-20a) is associated with a decrease in the VEGF-induced activation of p38. In contrast, these processes are increased by inhibiting miR-20a with a specific antagomir. Interestingly, miR-20a does not modulate VEGFR2 or p38 protein expression level. miR-20a does not affect either the expression of other known actors of the p38 MAP kinase pathway except MKK3. Indeed, by using quantitative PCR and Western Blot analysis, we found that pmiR-20a decreases the expression of MKK3 and we obtained evidence indicating that miR-20a specifically binds to the 3'UTR region of MKK3 mRNA. In accordance, the VEGF-induced activation of p38 and cell migration are impaired when the MKK3 expression is knocked down by siRNA. We conclude that miR-20a acts in a feedback loop to repress the expression of MKK3 and to negatively regulate the p38 pathway-mediated VEGF-induced endothelial cell migration and angiogenesis.


Subject(s)
Cell Movement/physiology , Endothelium, Vascular/cytology , MAP Kinase Kinase 3/metabolism , MicroRNAs/physiology , Vascular Endothelial Growth Factor A/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Enzyme Activation , Humans , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction
12.
BMC Cancer ; 11: 285, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21722370

ABSTRACT

BACKGROUND: Extravasation of circulating cancer cells is a key event of metastatic dissemination that is initiated by the adhesion of cancer cells to endothelial cells. It requires interactions between adhesion receptors on endothelial cells and their counter-receptors on cancer cells. Notably, E-selectin, a major endothelial adhesion receptor, interacts with Death receptor-3 present on metastatic colon carcinoma cells. This interaction confers metastatic properties to colon cancer cells by promoting the adhesion of cancer cells to endothelial cells and triggering the activation of the pro-migratory p38 and pro-survival ERK pathways in the cancer cells. In the present study, we investigated further the mechanisms by which the E-selectin-activated pathways downstream of DR3 confer a survival advantage to colon cancer cells. METHODS: Cell survival has been ascertained by using the WST-1 assay and by evaluating the activation of the PI3 kinase/NFκB survival axis. Apoptosis has been assayed by determining DNA fragmentation by Hoechst staining and by measuring cleavage of caspases-8 and -3. DR3 isoforms have been identified by PCR. For more precise quantification, targeted PCR reactions were carried out, and the amplified products were analyzed by automated chip-based microcapillary electrophoresis on an Agilent 2100 Bioanalyzer instrument. RESULTS: Interaction between DR3-expressing HT29 colon carcinoma cells and E-selectin induces the activation of the PI3K/Akt pathway. Moreover, p65/RelA, the anti-apoptotic subunit of NFκB, is rapidly translocated to the nucleus in response to E-selectin. This translocation is impaired by the PI3K inhibitor LY294002. Furthermore, inhibition of the PI3K/Akt pathway increases the cleavage of caspase 8 in colon cancer cells treated with E-selectin and this effect is still further increased when both ERK and PI3K pathways are concomitantly inhibited. Intriguingly, metastatic colon cancer cell lines such as HT29 and SW620 express higher levels of a splice variant of DR3 that has no trans-membrane domain and no death domain. CONCLUSION: Colon cancer cells acquire an increased capacity to survive via the activation of the PI3K/NFκB pathway following the stimulation of DR3 by E-selectin. Generation of a DR3 splice variant devoid of death domain can further contribute to protect against apoptosis.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , E-Selectin/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Amino Acid Sequence , Apoptosis/physiology , Cell Adhesion , Cell Survival/physiology , Chromones/pharmacology , HT29 Cells , Human Umbilical Vein Endothelial Cells , Humans , Jurkat Cells , MAP Kinase Signaling System , Microscopy, Fluorescence , Molecular Sequence Data , Morpholines/pharmacology , Neoplasm Metastasis , Phosphorylation , Protein Isoforms , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Tumor Necrosis Factor, Member 25/chemistry , Receptors, Tumor Necrosis Factor, Member 25/genetics , src-Family Kinases/metabolism
14.
J Immunol ; 184(8): 4531-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20228195

ABSTRACT

Neutrophilic inflammation plays an important role in lung tissue destruction occurring in many chronic pulmonary diseases. Neutrophils can be recruited to sites of inflammation via the action of the cytokine IL-17. In this study, we report that IL-17RA and IL-17RC mRNA expression is significantly increased in asthmatic bronchoscopic biopsies and that these receptors are not only expressed on epithelial and inflammatory cells but also on endothelial cells. IL-17 potently stimulates lung microvascular endothelial cells to produce chemoattractants (CXCL8 and derivatives of the 5-lipoxygenase pathway) that selectively drive neutrophil but not lymphocyte chemotaxis. Moreover, IL-17 promotes endothelial activation by inducing the expression of endothelial adhesion markers (E-selectin, VCAM-1, and ICAM-1) in a p38 MAPK-dependent manner. This increased expression of adhesion molecules stimulates the trans-endothelial migration of neutrophils, as well as the transmigration of HT-29 colon carcinoma cells, suggesting a further role in promoting lung metastasis. Finally, IL-17 increased neutrophil adhesion to the endothelium in vivo as determined by intravital microscopy of mice cremaster muscle. Overall, our results demonstrate that IL-17 is a potent activator of the endothelium in vivo leading to neutrophil infiltration. Therefore, preventing neutrophil recruitment by blocking the action of IL-17 on endothelial cells may prove to be highly beneficial in diseases in which neutrophilic inflammation plays a key role.


Subject(s)
Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Inflammation Mediators/physiology , Interleukin-17/physiology , Neutrophil Infiltration/immunology , Neutrophils/immunology , p38 Mitogen-Activated Protein Kinases/physiology , Adult , Animals , Asthma/immunology , Asthma/metabolism , Asthma/pathology , Cells, Cultured , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , HT29 Cells , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Interleukin-17/biosynthesis , Interleukin-8/biosynthesis , Jurkat Cells , MAP Kinase Signaling System/immunology , Male , Mice , Neutrophils/enzymology , Neutrophils/pathology
15.
J Biol Chem ; 285(11): 8013-21, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20061392

ABSTRACT

In this study, we obtained evidence indicating that annexin 1 is a new target of the p38/MAPKAP kinase-2 pathway and that it regulates endothelial cell migration in response to vascular endothelial growth factor (VEGF). These conclusions are supported by a series of substantiating experiments. First, by two-dimensional gel electrophoresis and mass spectrometry, we identified annexin 1 as a protein whose phosphorylation is induced by VEGF and is impaired by inhibiting p38. Second, using in vitro kinase assays and in vivo phosphorylation assays, we found that VEGF-mediated activation of LIM kinase 1 downstream of the p38 pathway triggers the phosphorylation of annexin 1. Third, VEGF-induced cell migration and tube formation in Matrigel are inhibited following small interfering RNA-mediated knockdown of annexin 1. Fourth, both processes are rescued in cells expressing an annexin 1 construct insensitive to the small interfering RNA knockdown. Finally, the VEGF/annexin 1-mediated cell migration is impaired by inhibiting p38. We therefore conclude that phosphorylation of annexin 1 regulates the angiogenic effect that is associated with the activation of the p38/LIM kinase 1 axis by VEGF.


Subject(s)
Annexin A1/metabolism , Cell Movement/physiology , Endothelial Cells/enzymology , Lim Kinases/metabolism , MAP Kinase Signaling System/physiology , Vascular Endothelial Growth Factor A/metabolism , Annexin A1/genetics , Cells, Cultured , Collagen , Drug Combinations , Endothelial Cells/cytology , Humans , Laminin , Lim Kinases/genetics , Mass Spectrometry , Neovascularization, Physiologic/physiology , Phosphorylation/physiology , Proteoglycans , RNA, Small Interfering , Umbilical Veins/cytology , p38 Mitogen-Activated Protein Kinases/metabolism
16.
J Cell Sci ; 120(Pt 20): 3666-77, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17895359

ABSTRACT

Endothelial cells are actively involved in regulating the exchanges between blood and tissues. This function is tightly dependent on actin cytoskeleton dynamics and is challenged by a wide variety of stimuli, including oxidative stress. In endothelial cells, oxidative stress quickly activates the extracellular-signal-regulated kinase (ERK) MAP kinase, which results in the phosphorylation of tropomyosin. Here, we investigated further the mechanisms of tropomyosin phosphorylation and its function in actin remodeling. We identified, for the first time, death-associated protein kinase 1 (DAP kinase 1) as the kinase that phosphorylates tropomyosin-1 in response to ERK activation by hydrogen peroxide (H(2)O(2)). We also report that the phosphorylation of tropomyosin-1 mediated by DAP kinase occurs on Ser283. Moreover, the expression of the pseudophosphorylated tropomyosin mutant Ser283Glu triggers by itself the formation of stress fibers in untreated cells, and the effect is maintained in H(2)O(2)-treated cells in which DAP kinase expression is knocked-down by siRNA. By contrast, the expression of the nonphosphorylatable tropomyosin mutant Ser283Ala is not associated with stress fibers and leads to membrane blebbing in response to H(2)O(2). Our finding that tropomyosin-1 is phosphorylated downstream of ERK and DAP kinase and that it helps regulate the formation of stress fibers will aid understanding the role of this protein in regulating the endothelial functions associated with cytoskeletal remodeling.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Endothelial Cells/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Oxidative Stress , Stress Fibers/physiology , Tropomyosin/metabolism , Cell Line , Cells, Cultured , Death-Associated Protein Kinases , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System , Phosphorylation , Umbilical Veins/cytology
17.
Cancer Res ; 66(18): 9117-24, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16982754

ABSTRACT

E-selectin-mediated adhesion of colon cancer cells to endothelial cells is a key event in metastasis. However, the signaling mechanisms that confer metastatic advantages to cancer cells adhering to E-selectin are ill defined. By using affinity column chromatography and pull-down assays on purified membrane extracts of HT29 and LoVo cells coupled to mass spectrometry analysis, we obtained the first evidence indicating that E-selectin binds to death receptor-3 (DR3) expressed by the cancer cells. Thereafter, we accumulated several results, suggesting that DR3 is an E-selectin receptor on colon cancer cells and that its activation by E-selectin triggers the activation of p38 and extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) and confers migration and survival advantages. First, by Western blotting, we found that the E-selectin-binding protein, identified as DR3, is recognized by two anti-DR3 antibodies. Second, the neutralization of DR3 with an antibody and its knockdown by small interfering RNA decrease the adhesion of colon cancer cells to E-selectin and E-selectin-expressing human umbilical vein endothelial cells. Third, inhibiting DR3 and knocking down its expression impair transendothelial migration of HT29 cells and block the activation of p38 and ERK by E-selectin. Fourth, high molecular weight isoforms of DR3 are expressed in samples of primary human colon carcinoma but not in samples from normal colon tissue. Intriguingly, DR3 is a death receptor but its activation by E-selectin does not induce apoptosis in colon cancer cells, except when ERK is inhibited. Our findings identify novel signaling and functional roles of DR3 activated in response to E-selectin and highlight the potential link between DR3 and metastasis.


Subject(s)
Cell Movement/physiology , Colonic Neoplasms/pathology , E-Selectin/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis/physiology , Cell Survival/physiology , Colonic Neoplasms/enzymology , Colonic Neoplasms/metabolism , Endothelial Cells/cytology , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Enzyme Activation , HT29 Cells , Humans
18.
J Biol Chem ; 281(45): 34009-20, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16966330

ABSTRACT

VEGFR-2 is the major receptor that regulates the different functions of VEGF in adults. We have previously reported that following VEGF treatment of endothelial cells, VEGFR-2 is phosphorylated on Tyr1214 upstream of the Cdc42-SAPK2/p38-MAPKAP K2 pathway. However, little is known of the earliest molecular events that compose the SAPK2/p38 pathway following VEGFR-2 activation. In this study, we address this question using HA-tagged constructs of either wild-type VEGFR-2 or Y1214F VEGFR-2 mutant in immunoprecipitation assays. We show that the Src family kinase member Fyn, but not c-Src itself, is recruited to VEGFR-2 and is activated in a p-Tyr1214-dependent manner. We also report that the SH2 domain-containing adapter molecule Nck, but not Grb2, is recruited to VEGFR-2 in a p-Tyr1214-dependent manner and that it associates with Fyn. Moreover, PAK-2 is phosphorylated in a Fyn-dependent manner. Using chemical and genetic inhibitors, we show that Fyn activity is required for SAPK2/p38 but not for FAK activation in response to VEGF. In contrast, c-Src permits activation of FAK, but not that of SAPK2/p38. In addition, Fyn is required for stress fiber formation and endothelial cell migration. We propose a model in which Fyn forms a molecular complex with Nck and PAK-2 and suggest that this complex assembles in a p-Tyr1214-dependent manner within VEGFR-2 following VEGF treatment. In turn, this triggers the activation of the SAPK2/p38 MAP kinase module, and promotes stress fiber formation and endothelial cell migration.


Subject(s)
Cell Movement , Mitogen-Activated Protein Kinase 11/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Activation , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Mice , NIH 3T3 Cells , Phosphorylation , Umbilical Veins , src-Family Kinases
19.
Mol Biol Cell ; 17(8): 3508-20, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16760434

ABSTRACT

Focal adhesion kinase (FAK) is phosphorylated on tyrosine and serine residues after cell activation. In the present work, we investigated the relationship between tyrosine and serine phosphorylation of FAK in promoting endothelial cell migration in response to vascular endothelial growth factor (VEGF). We found that VEGF induces the activation of the Rho-dependent kinase (ROCK) downstream from vascular endothelial growth factor receptor (VEGFR) 2. In turn, activated ROCK directly phosphorylates FAK on Ser732. Proline-rich tyrosine kinase-2 (Pyk2) is also activated in response to VEGF. Its activation requires the clustering of integrin alphavbeta3 and triggers directly the phosphorylation of Tyr407 within FAK, an event necessary for cell migration. Interestingly, ROCK-mediated phosphorylation of Ser732 is essential for Pyk2-dependent phosphorylation of Tyr407, because the latter is abrogated in cells expressing a FAK mutant that is nonphosphorylatable on Ser732. We suggest that VEGF elicits the activation of the VEGFR2-ROCK pathway, leading to phosphorylation of Ser732 within FAK. In turn, phosphorylation of Ser732 would change the conformation of FAK, making it accessible to Pyk2 activated in response to its association with integrin beta3. Then, activated Pyk2 triggers the phosphorylation of FAK on Tyr407, promoting cell migration.


Subject(s)
Focal Adhesion Kinase 2/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Phosphoserine/metabolism , Phosphotyrosine/metabolism , Protein Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , Benzoquinones/pharmacology , Cattle , Cell Movement/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Focal Adhesions/drug effects , HSP90 Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Lactams, Macrocyclic/pharmacology , Mice , Mutation/genetics , Phosphorylation/drug effects , Protein Transport/drug effects , Swine , Vinculin/metabolism , rho-Associated Kinases
20.
Mol Carcinog ; 45(6): 362-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16637066

ABSTRACT

The traffic of molecules and cells across the vessel wall is gated by vascular endothelial cells. In accordance, these cells play an active role in regulating cardiovascular and systemic homeostasis and in modulating physiopathological processes such as inflammation. Dysfunction of the regulatory systems of the endothelium and its incapacity to efficiently deal with its physicochemical surrounding leads to disruption of endothelial integrity. For example, alterations of the selective endothelial cell permeability barrier are early events in the sequence of oxidative stress-mediated injury that may contribute to extravasation of circulating cancer cells. Several lines of evidence indicate that the regulation of the endothelial barrier is tightly regulated by activation of signaling pathways that converge on the regulation of actin cytoskeletal dynamics. In particular, the integrity of the endothelial layer in response to oxidative stress is tightly regulated by the balanced activation of the extracellular-signal regulated kinase (ERK) and the stress-activated protein kinase-2/p38 (SAPK2/p38) pathways. Activation of the SAPK2/p38 pathway is required to trigger actin polymerization, whereas activation of the ERK pathway by contributing to phosphorylate tropomyosin-1 triggers the formation of focal adhesions allowing the anchorage of actin filaments generated by SAPK2/p38 to bundle into stress fibers. Dysregulation of this equilibrium by inhibiting ERK leads to membrane blebbing, an early manifestation of oxidative toxicity that is associated with disruption of the endothelial layer integrity.


Subject(s)
Endothelium, Vascular/pathology , Neoplasms/pathology , Oxidative Stress , Actins/metabolism , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/enzymology , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...