Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nucleic Acids Res ; 52(9): 5002-5015, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38477356

RESUMEN

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.


Asunto(s)
Proteínas Argonautas , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , MicroARNs , Animales , MicroARNs/genética , MicroARNs/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas Argonautas/metabolismo , Proteínas Argonautas/genética , Mutación , Proteínas de Unión al ARN
2.
PLoS Genet ; 17(4): e1009511, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33826611

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Biosíntesis de Proteínas , Proteínas de Unión al ARN/genética , Proteínas de Unión al GTP rab/genética , Regiones no Traducidas 3'/genética , Animales , Caenorhabditis elegans/genética , Regulación del Desarrollo de la Expresión Génica/genética , Silenciador del Gen , MicroARNs/genética , ARN Mensajero/genética
3.
J Cell Sci ; 129(21): 4105-4117, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27637266

RESUMEN

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.


Asunto(s)
Biosíntesis de Proteínas , Migración Transendotelial y Transepitelial , Adhesión Celular , Línea Celular Tumoral , Forma de la Célula , Activación Enzimática , Transición Epitelial-Mesenquimal , Adhesiones Focales/metabolismo , GTP Fosfohidrolasas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mesodermo/metabolismo , Modelos Biológicos , Metástasis de la Neoplasia , Poliadenilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo
4.
bioRxiv ; 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36711744

RESUMEN

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.

5.
J Biol Chem ; 287(36): 30541-51, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22773844

RESUMEN

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.


Asunto(s)
Anexina A1/metabolismo , Movimiento Celular/fisiología , Regulación de la Expresión Génica/fisiología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , MicroARNs/biosíntesis , Factor A de Crecimiento Endotelial Vascular/metabolismo , Regiones no Traducidas 3'/fisiología , Anexina A1/genética , Movimiento Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , MicroARNs/genética , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Seudópodos/genética , Seudópodos/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
6.
Breast Cancer Res ; 14(3): R92, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22691413

RESUMEN

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.


Asunto(s)
Movimiento Celular , Estradiol Deshidrogenasas/metabolismo , Nucleósido Difosfato Quinasas NM23/metabolismo , ARN Mensajero/metabolismo , Proteína BRCA2/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Estradiol Deshidrogenasas/genética , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Humanos , Células MCF-7 , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Análisis por Matrices de Proteínas , Proteoma/análisis , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño , Receptores Androgénicos/genética
7.
Angiogenesis ; 15(4): 593-608, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22696064

RESUMEN

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.


Asunto(s)
Movimiento Celular/fisiología , Endotelio Vascular/citología , MAP Quinasa Quinasa 3/metabolismo , MicroARNs/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Activación Enzimática , Humanos , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
J Immunol ; 184(8): 4531-7, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20228195

RESUMEN

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.


Asunto(s)
Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Mediadores de Inflamación/fisiología , Interleucina-17/fisiología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Adulto , Animales , Asma/inmunología , Asma/metabolismo , Asma/patología , Células Cultivadas , Endotelio Vascular/enzimología , Endotelio Vascular/patología , Células HT29 , Humanos , Inflamación/enzimología , Inflamación/inmunología , Inflamación/patología , Interleucina-17/biosíntesis , Interleucina-8/biosíntesis , Células Jurkat , Sistema de Señalización de MAP Quinasas/inmunología , Masculino , Ratones , Neutrófilos/enzimología , Neutrófilos/patología
9.
Cell Rep ; 41(11): 111822, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36516777

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans , MicroARNs , Animales , Caenorhabditis elegans/metabolismo , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fosforilación , Proteínas de Unión al ARN/metabolismo , Serina/metabolismo
10.
J Biol Chem ; 285(11): 8013-21, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20061392

RESUMEN

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.


Asunto(s)
Anexina A1/metabolismo , Movimiento Celular/fisiología , Células Endoteliales/enzimología , Quinasas Lim/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Anexina A1/genética , Células Cultivadas , Colágeno , Combinación de Medicamentos , Células Endoteliales/citología , Humanos , Laminina , Quinasas Lim/genética , Espectrometría de Masas , Neovascularización Fisiológica/fisiología , Fosforilación/fisiología , Proteoglicanos , ARN Interferente Pequeño , Venas Umbilicales/citología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
BMC Cancer ; 11: 285, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21722370

RESUMEN

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.


Asunto(s)
Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Selectina E/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Secuencia de Aminoácidos , Apoptosis/fisiología , Adhesión Celular , Supervivencia Celular/fisiología , Cromonas/farmacología , Células HT29 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Jurkat , Sistema de Señalización de MAP Quinasas , Microscopía Fluorescente , Datos de Secuencia Molecular , Morfolinas/farmacología , Metástasis de la Neoplasia , Fosforilación , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/química , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Familia-src Quinasas/metabolismo
12.
Oncogene ; 40(18): 3231-3244, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33833398

RESUMEN

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.


Asunto(s)
Proteínas Argonautas , MicroARNs , Células HEK293 , Humanos , Melanoma/genética , ARN Mensajero/genética , Proteínas de Unión al ARN
13.
Life Sci Alliance ; 3(4)2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32161113

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , MicroARNs/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Proteínas Argonautas/genética , Drosophila melanogaster/genética , Células Madre Embrionarias , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Silenciador del Gen/fisiología , Células HEK293 , Células HeLa , Humanos , Ratones , MicroARNs/genética , Células 3T3 NIH , Estabilidad del ARN/genética , Estabilidad del ARN/fisiología , ARN Mensajero/genética
14.
Mol Biol Cell ; 17(8): 3508-20, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16760434

RESUMEN

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.


Asunto(s)
Quinasa 2 de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Fosfoserina/metabolismo , Fosfotirosina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Benzoquinonas/farmacología , Bovinos , Movimiento Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/química , Adhesiones Focales/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lactamas Macrocíclicas/farmacología , Ratones , Mutación/genética , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Porcinos , Vinculina/metabolismo , Quinasas Asociadas a rho
15.
Cancer Res ; 66(18): 9117-24, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16982754

RESUMEN

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.


Asunto(s)
Movimiento Celular/fisiología , Neoplasias del Colon/patología , Selectina E/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Apoptosis/fisiología , Supervivencia Celular/fisiología , Neoplasias del Colon/enzimología , Neoplasias del Colon/metabolismo , Células Endoteliales/citología , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Activación Enzimática , Células HT29 , Humanos
16.
Mol Biol Cell ; 14(4): 1418-32, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12686598

RESUMEN

Oxidative stress induces in endothelial cells a quick and transient coactivation of both stress-activated protein kinase-2/p38 and extracellular signal-regulated kinase (ERK) mitogen-activated protein kinases. We found that inhibiting the ERK pathway resulted, within 5 min of oxidative stress, in a misassembly of focal adhesions characterized by mislocalization of key proteins such as paxillin. The focal adhesion misassembly that followed ERK inhibition with the mitogen-activated protein kinase kinase (MEK) inhibitor PD098059 (2'-amino-3'-methoxyflavone) or with a kinase negative mutant of ERK in the presence of H(2)O(2) resulted in a quick and intense membrane blebbing that was associated with important damage to the endothelium. We isolated by two-dimensional gel electrophoresis a PD098059-sensitive phosphoprotein of 38 kDa that we identified, by mass spectrometry, as tropomyosin-1. In fact, H(2)O(2) induced a time-dependent phosphorylation of tropomyosin that was sensitive to inhibition by PD098059 and UO126 (1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio]butanediane). Tropomyosin phosphorylation was also induced by expression of a constitutively activated form of MEK1 (MEK(CA)), which confirms that its phosphorylation resulted from the activation of ERK. In unstimulated cells, tropomyosin-1 was found diffuse in the cells, whereas it quickly colocalized with actin and stress fibers upon stimulation of ERK by H(2)O(2) or by expression of MEK(CA). We propose that phosphorylation of tropomyosin-1 downstream of ERK by contributing to formation of actin filaments increases cellular contractility and promotes the formation of focal adhesions. Incidentally, ML-7 (1-[5iodonaphthalene-1-sulfonyl]homopiperazine, HCl), an inhibitor of cell contractility, inhibited phosphorylation of tropomyosin and blocked the formation of stress fibers and focal adhesions, which also led to membrane blebbing in the presence of oxidative stress. Our finding that tropomyosin-1 is phosphorylated downstream of ERK, an event that modulates its interaction with actin, may lead to further understanding of the role of this protein in regulating cellular functions associated with cytoskeletal remodeling.


Asunto(s)
Citoesqueleto/metabolismo , Proteínas de Drosophila , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Tropomiosina/metabolismo , Secuencia de Aminoácidos , Membrana Celular/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Adhesiones Focales , Humanos , Peróxido de Hidrógeno/farmacología , Sistema de Señalización de MAP Quinasas , Estrés Oxidativo , Fosforilación , Tropomiosina/química , Tropomiosina/genética
17.
Oncogene ; 23(2): 434-45, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-14724572

RESUMEN

Activation of the tyrosine kinase receptor vascular endothelial growth factor receptor 2 (VEGFR2) by VEGF leads to the activation of stress-activated protein kinase (SAPK)2/p38 and then to actin polymerization and reorganization into stress fibers in endothelial cells. In turn, this triggers endothelial cell migration. Yet, nothing is known about the molecular mechanisms that couple VEGFR2 to SAPK2/p38. Here, we found that VEGF increased by twofold the activity of the small GTPase Cdc42 and that the expression of two different constitutively active forms of Cdc42 (Cdc42 V12 and Cdc42 L61) led to a marked increase in the formation of stress fibers that was sensitive to SAPK2/p38 inhibition by SB203580. Moreover, the expression of a dominant-negative form of Cdc42 (Cdc42 N17) inhibited the activation of SAPK2/p38 and of its direct target MAP kinase-activated protein kinase 2. These results indicate that Cdc42 is upstream of SAPK2/p38 in response to the activation of VEGFR2 by VEGF. In contrast, we found that neither RhoA nor Rac was involved in the SAPK2/p38-mediated actin reorganization induced by VEGF. Using a site-specific mutant of the major autophosphorylation site Y1214 on VEGFR2, we found that the mutant Y1214F inhibited the activation of both Cdc42 and SAPK2/p38 in response to VEGF. We conclude that phosphorylation of Y1214 on VEGFR2 is required to trigger the sequential activation of Cdc42 and SAPK2/p38 and to drive the SAPK2/p38-mediated actin remodeling in stress fibers in endothelial cells exposed to VEGF.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfotirosina/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Actinas/metabolismo , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Humanos , Modelos Biológicos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Fibras de Estrés/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
18.
Clin Exp Metastasis ; 21(3): 257-64, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15387376

RESUMEN

HT-29 colon carcinoma cells attach to TNFalpha-activated human umbilical vein endothelial cells (HUVECs) by their specific binding to E-selectin. This interaction activates, in the cancer cells, the MAPK SAPK2/p38, which leads to their transendothelial migration (Laferrière et al., J Biol Chem 2001; 276: 33762). In this study, we investigated the role of E-selectin in activating integrins to modulate adhesion and regulate integrin-mediated events. Blocking the integrins from HT-29 cells (alpha2, alpha3, alpha6, alphav/beta5, beta1 and beta4) with specific antibodies revealed a role for beta4 integrin in their adhesion to TNFalpha-treated HUVEC. The beta4 integrin-dependent adhesion was maximal after 30 min, whereas the-E-selectin-dependent adhesion was maximal after 15 min. Integrin beta4 became quickly phosphorylated upon addition of HT-29 cells to endothelial cells and the effect was independent of the expression of E-selectin. Moreover, a recombinant E-selectin/Fc chimera did not induce the phosphorylation of beta4. The phosphorylation of beta4 is not required for adhesion since adhesion was not affected in HT-29 cells that express a truncated form of beta4 that is deleted from its cytoplasmic phosphorylatable domain. However, the expression of the non-phosphorylatable deletant of beta4 was associated with decreased transendothelial cell migration underscoring the key role for the cytoplasmic domain of beta4 in cell migration. We suggest: 1) that the adhesion of HT-29 cells to activated endothelial cells follows at least two essential sequential steps involving the binding of E-selectin to its receptor on carcinoma cells and then the binding of beta4 to its own receptor on endothelial cells; 2) that the phosphorylation of integrin beta4 contributes to enhance the motile potential of cancer cells and increase their trans-endothelial migration. Overall, our results indicate that the interaction of metastatic cancer cells with endothelial cells implies a specific sequence of signaling events that ultimately leads to an increase in their efficient transendothelial migration.


Asunto(s)
Adhesión Celular/fisiología , Neoplasias del Colon/patología , Selectina E/fisiología , Endotelio Vascular/patología , Integrina beta4/fisiología , Línea Celular Tumoral , Movimiento Celular , Células Cultivadas , Humanos , Fosforilación , Transducción de Señal
19.
Cell Stress Chaperones ; 8(1): 37-52, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12820653

RESUMEN

Endothelial cell migration, a key process in angiogenesis, requires the coordinated integration of motogenic signals elicited by the adhesion of endothelial cells to extracellular matrices and by angiogenic cytokines such as the vascular endothelial growth factor (VEGF). In this study, we found that addition of VEGF to human umbilical vein endothelial cells cultivated on vitronectin triggers a synergistic interaction between the VEGF receptor VEGFR2 and the clustered integrin receptor alphavbeta3. The interaction between VEGFR2 and alphavbeta3 is required for full phosphorylation of VEGFR2 and to drive the activation of motogenic pathways involving focal adhesion kinase (FAK) and stress-activated protein kinase-2/p38 (SAPK2/p38). The signal emanating from the VEGFR2 and alphavbeta3 interaction and leading to SAPK2/p38 activation proceeds directly from VEGFR2. The chaperone Hsp90 is found in a complex that coprecipitates with inactivated VEGFR2, and the association is increased by VEGF and decreased by geldanamycin, a specific inhibitor of Hsp90-mediated events. Geldanamycin also impairs the phosphorylation of FAK that results from the interaction between VEGFR2 and alphavbeta3, and this is accompanied by an inhibition of the recruitment of vinculin to VEGFR2. We conclude that a necessary cross talk should occur between VEGFR2 and the integrin alphavbeta3, to transduce the VEGF signals to SAPK2/p38 and FAK and that Hsp90 is instrumental in the building up of focal adhesions by allowing the phosphorylation of FAK and the recruitment of vinculin to VEGFR2.


Asunto(s)
Endotelio Vascular/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Integrina alfaVbeta3/agonistas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/agonistas , Anticuerpos Monoclonales , Benzoquinonas , Movimiento Celular/fisiología , Células Cultivadas , Endotelio Vascular/citología , Activación Enzimática , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Técnicas de Transferencia de Gen , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Lactamas Macrocíclicas , Proteínas Quinasas Activadas por Mitógenos/genética , Fosforilación/efectos de los fármacos , Quinonas/farmacología , Venas Umbilicales/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
20.
Ann N Y Acad Sci ; 973: 562-72, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12485930

RESUMEN

The formation of metastasis is a dreadful complication of cancer that is associated with a poor prognosis. Several clinical observations and experimental findings indicate that the metastatic process is nonrandom and involves a sequence of multistep events that may all be targeted for therapy. This includes angiogenesis of the primary neoplasm, release of malignant cells from this neoplasm, entry of cancer cells into the blood circulation, interaction of cancer cells with vascular endothelial cells in distant organs, and growth of blood-borne cancer cells locally in the vessels or distally following extravasation. Our working hypothesis is that metastatic cancer cells exploit the mechanisms of the inflammation process to successfully migrate into distant organs. This implies a pivotal role for specific adhesive interactions between cancer cells and vascular endothelial cells and activation of migratory pathways in the cancer cells. We review here the roles played by the endothelial adhesive molecule E-selectin and by the motogenic stress-activated protein kinase-2 (SAPK2/p38) pathway of cancer cells in modulating transendothelial migration of cancer cells.


Asunto(s)
Selectina E/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Metástasis de la Neoplasia/fisiopatología , Adhesión Celular/fisiología , Endotelio Vascular/fisiopatología , Humanos , Modelos Biológicos , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA