Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Cell Biol ; 223(5)2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38358349

RESUMEN

Different membrane microdomain compositions provide unique environments that can regulate signaling receptor function. We identify microdomains on the endosome membrane of Drosophila endosomes, enriched in lipid-raft or clathrin/ESCRT-0, which are associated with Notch activation by distinct, ligand-independent mechanisms. Transfer of Notch between microdomains is regulated by Deltex and Suppressor of deltex ubiquitin ligases and is limited by a gate-keeper role for ESCRT complexes. Ubiquitination of Notch by Deltex recruits it to the clathrin/ESCRT-0 microdomain and enhances Notch activation by an ADAM10-independent/TRPML-dependent mechanism. This requirement for Deltex is bypassed by the downregulation of ESCRT-III. In contrast, while ESCRT-I depletion also activates Notch, it does so by an ADAM10-dependent/TRPML-independent mechanism and Notch is retained in the lipid raft-like microdomain. In the absence of such endosomal perturbation, different activating Notch mutations also localize to different microdomains and are activated by different mechanisms. Our findings demonstrate the interplay between Notch regulators, endosomal trafficking components, and Notch genetics, which defines membrane locations and activation mechanisms.


Asunto(s)
Proteínas de Drosophila , Drosophila , Proteínas de la Membrana , Receptores Notch , Canales de Potencial de Receptor Transitorio , Animales , Proteína ADAM10/metabolismo , Clatrina/metabolismo , Regulación hacia Abajo , Proteínas de Drosophila/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/metabolismo , Receptores Notch/metabolismo , Ubiquitinación , Proteínas de la Membrana/metabolismo , Microdominios de Membrana/metabolismo
2.
Pharmacol Ther ; 238: 108179, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35358569

RESUMEN

Gastrointestinal cancers are responsible for more cancer deaths than any other system of the body. This review summarises how Wnt pathway dysregulation contributes to the development of the most common gastrointestinal cancers, with a particular focus on the nature and frequency of upstream pathway aberrations. Tumors with upstream aberrations maintain a dependency on the presence of functional Wnt ligand, and are predicted to be tractable to inhibitors of Porcupine, an enzyme that plays a key role in Wnt secretion. We summarise available pre-clinical efficacy data from Porcupine inhibitors in vitro and in vivo, as well as potential toxicities and the data from early phase clinical trials. We appraise the rationale for biomarker-defined targeted approaches, as well as outlining future opportunities for combination with other therapeutics.


Asunto(s)
Neoplasias Gastrointestinales , Vía de Señalización Wnt , Aciltransferasas/metabolismo , Neoplasias Gastrointestinales/tratamiento farmacológico , Humanos , Ligandos , Proteínas de la Membrana/metabolismo
3.
Cancer Res Commun ; 2(9): 914-928, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36922934

RESUMEN

Wnt signaling is implicated in the etiology of gastrointestinal tract cancers. Targeting Wnt signaling is challenging due to on-target toxicity concerns and lack of druggable pathway components. We describe the discovery and characterization of RXC004, a potent and selective inhibitor of the membrane-bound o-acyl transferase Porcupine, essential for Wnt ligand secretion. Absorption, distribution, metabolism, and excretion and safety pharmacology studies were conducted with RXC004 in vitro, and pharmacokinetic exposure assessed in vivo. RXC004 effects on proliferation and tumor metabolism were explored in genetically defined colorectal and pancreatic cancer models in vitro and in vivo. RXC004 effects on immune evasion were assessed in B16F10 immune "cold" and CT26 immune "hot" murine syngeneic models, and in human cell cocultures. RXC004 showed a promising pharmacokinetic profile, inhibited Wnt ligand palmitoylation, secretion, and pathway activation, and demonstrated potent antiproliferative effects in Wnt ligand-dependent (RNF43-mutant or RSPO3-fusion) colorectal and pancreatic cell lines. Reduced tumor growth and increased cancer cell differentiation were observed in SNU-1411 (RSPO3-fusion), AsPC1 and HPAF-II (both RNF43-mutant) xenograft models, with a therapeutic window versus Wnt homeostatic functions. Additional effects of RXC004 on tumor cell metabolism were confirmed in vitro and in vivo by glucose uptake and 18fluorodeoxyglucose-PET, respectively. RXC004 stimulated host tumor immunity; reducing resident myeloid-derived suppressor cells within B16F10 tumors and synergizing with anti-programmed cell death protein-1 (PD-1) to increase CD8+/regulatory T cell ratios within CT26 tumors. Moreover, RXC004 reversed the immunosuppressive effects of HPAF-II cells cocultured with human peripheral blood mononuclear cells, confirming the multiple anticancer mechanisms of this compound, which has progressed into phase II clinical trials. Significance: Wnt pathway dysregulation drives many gastrointestinal cancers; however, there are no approved therapies that target the pathway. RXC004 has demonstrated the potential to block both tumor growth and tumor immune evasion in a genetically defined, clinically actionable subpopulation of Wnt ligand-dependent gastrointestinal cancers. The clinical utility of RXC004, and other Porcupine inhibitors, in such Wnt ligand-dependent cancers is currently being assessed in patient trials.


Asunto(s)
Neoplasias Colorrectales , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Vía de Señalización Wnt , Ligandos , Evasión Inmune , Leucocitos Mononucleares/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico
4.
Sci Rep ; 11(1): 9096, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33907274

RESUMEN

Notch and Wnt are two essential signalling pathways that help to shape animals during development and to sustain adult tissue homeostasis. Although they are often active at the same time within a tissue, they typically have opposing effects on cell fate decisions. In fact, crosstalk between the two pathways is important in generating the great diversity of cell types that we find in metazoans. Several different mechanisms have been proposed that allow Notch to limit Wnt signalling, driving a Notch-ON/Wnt-OFF state. Here we explore these different mechanisms in human cells and demonstrate two distinct mechanisms by which Notch itself, can limit the transcriptional activity of ß-catenin. At the membrane, independently of DSL ligands, Notch1 can antagonise ß-catenin activity through an endocytic mechanism that requires its interaction with Deltex and sequesters ß-catenin into the membrane fraction. Within the nucleus, the intracellular domain of Notch1 can also limit ß-catenin induced transcription through the formation of a complex that requires its interaction with RBPjκ. We believe these mechanisms contribute to the robustness of cell-fate decisions by sharpening the distinction between opposing Notch/Wnt responses.


Asunto(s)
Receptores Notch/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Núcleo Celular/metabolismo , Células HCT116 , Células HEK293 , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores Notch/genética , Transcripción Genética , beta Catenina/genética , Proteínas de Unión al GTP rab5/metabolismo
5.
J Med Chem ; 64(6): 3165-3184, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33683117

RESUMEN

Mer is a member of the TAM (Tyro3, Axl, Mer) kinase family that has been associated with cancer progression, metastasis, and drug resistance. Their essential function in immune homeostasis has prompted an interest in their role as modulators of antitumor immune response in the tumor microenvironment. Here we illustrate the outcomes of an extensive lead-generation campaign for identification of Mer inhibitors, focusing on the results from concurrent, orthogonal high-throughput screening approaches. Data mining, HT (high-throughput), and DECL (DNA-encoded chemical library) screens offered means to evaluate large numbers of compounds. We discuss campaign strategy and screening outcomes, and exemplify series resulting from prioritization of hits that were identified. Concurrent execution of HT and DECL screening successfully yielded a large number of potent, selective, and novel starting points, covering a range of selectivity profiles across the TAM family members and modes of kinase binding, and offered excellent start points for lead development.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Tirosina Quinasa c-Mer/antagonistas & inhibidores , Animales , Cristalografía por Rayos X , Minería de Datos , Descubrimiento de Drogas , Humanos , Modelos Moleculares , Tirosina Quinasa c-Mer/química , Tirosina Quinasa c-Mer/metabolismo
6.
Open Biol ; 7(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28424321

RESUMEN

The developmental signalling protein Notch can be proteolytically activated following ligand-interaction at the cell surface, or can be activated independently of its ligands, following Deltex (Dx)-induced Notch endocytosis and trafficking to the lysosomal membrane. The means by which different pools of Notch are directed towards these alternative outcomes remains poorly understood. We found that the Drosophila ZO-1 protein Polychaetoid (Pyd) suppresses specifically the Dx-induced form of Notch activation both in vivo and in cell culture assays. In vivo we confirmed the physiological relevance and direction of the Pyd/Dx interaction by showing that the expanded ovary stem cell niche phenotypes of pyd mutants require the presence of functional Dx and other components that are specific to the Dx-induced Notch activation mechanism. In S2 cells we found that Pyd can form a complex with Dx and Notch at the cell surface and reduce Dx-induced Notch endocytosis. Similar to other known activities of ZO-1 family proteins, the action of Pyd on Dx-induced endocytosis and signalling was found to be cell density dependent. Thus, together, our results suggest an alternative means by which external cues can tune Notch signalling through Pyd regulation of Dx-induced Notch trafficking.


Asunto(s)
Proteínas de Drosophila/metabolismo , Células Germinativas/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Nicho de Células Madre , Proteínas de Uniones Estrechas/metabolismo , Proteínas de Drosophila/genética , Expresión Génica , Proteínas de la Membrana/genética , Mutación , Transporte de Proteínas , Transducción de Señal
7.
Health Econ Rev ; 7(1): 3, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28105591

RESUMEN

In health economic evaluation studies, to value productivity loss due to absenteeism, existing methods use wages as a proxy value for marginal productivity. This study is the first to test the equality between wage and marginal productivity losses due to absenteeism separately for team workers and non-team workers. Our estimates are based on linked employer-employee data from Canada. Results indicate that team workers are more productive and earn higher wages than non-team workers. However, the productivity gap between these two groups is considerably larger than the wage gap. In small firms, employee absenteeism results in lower productivity and wages, and the marginal productivity loss due to team worker absenteeism is significantly higher than the wage loss. No similar wage-productivity gap exists for large firms. Our findings suggest that productivity loss or gain is most likely to be underestimated when valued according to wages for team workers. The findings help to value the burden of illness-related absenteeism. This is important for economic evaluations that seek to measure the productivity gain or loss of a health care technology or intervention, which in turn can impact policy makers' funding decisions.

8.
Soc Sci Med ; 147: 62-71, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26547046

RESUMEN

One source of productivity loss due to illness is the reduced "quantity" or "quality" of labor input while working, often referred to as presenteeism. Illness-related presenteeism has been found to be potentially more costly than absenteeism. To value presenteeism, existing methods use wages as a proxy for marginal productivity at the firm level. However, wage may not equal marginal productivity in some scenarios. One instance is when a job involves team production and perfect substitutes for workers are not readily available. Using a Canadian linked employer-employee survey (2001-2005), we test whether relative wage equals relative marginal productivity among team workers and non-team workers with different frequencies of presenteeism (reduction at work due to illness). For the pooled cross-sectional estimates (2001, 2003, 2005) we obtain 13,755 observations with 6842 unique workplaces. There are 6490 observations for the first differences estimates from the odd years and 5263 observations for the first differences estimates from 2001 to 2002 and 2003 to 2004. We find that in both small and large firms, team workers with frequent reductions at work are less productive but earn similarly compared with non-team workers without reductions. We also find that in small firms, workers with occasional work reductions are more productive than workers without reductions, but the reverse is true in large firms. The study findings partially support the literature stating that productivity loss resulting from employee presenteeism could exceed wages if team work is involved.


Asunto(s)
Costo de Enfermedad , Eficiencia , Presentismo , Salarios y Beneficios/economía , Absentismo , Canadá , Estudios Transversales , Humanos , Modelos Teóricos , Encuestas y Cuestionarios , Trabajo/economía , Lugar de Trabajo/estadística & datos numéricos
9.
J Biomol Screen ; 20(8): 1040-51, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26048892

RESUMEN

For more than a decade, RNA interference (RNAi) has brought about an entirely new approach to functional genomics screening. Enabling high-throughput loss-of-function (LOF) screens against the human genome, identifying new drug targets, and significantly advancing experimental biology, RNAi is a fast, flexible technology that is compatible with existing high-throughput systems and processes; however, the recent advent of clustered regularly interspaced palindromic repeats (CRISPR)-Cas, a powerful new precise genome-editing (PGE) technology, has opened up vast possibilities for functional genomics. CRISPR-Cas is novel in its simplicity: one piece of easily engineered guide RNA (gRNA) is used to target a gene sequence, and Cas9 expression is required in the cells. The targeted double-strand break introduced by the gRNA-Cas9 complex is highly effective at removing gene expression compared to RNAi. Together with the reduced cost and complexity of CRISPR-Cas, there is the realistic opportunity to use PGE to screen for phenotypic effects in a total gene knockout background. This review summarizes the exciting development of CRISPR-Cas as a high-throughput screening tool, comparing its future potential to that of well-established RNAi screening techniques, and highlighting future challenges and opportunities within these disciplines. We conclude that the two technologies actually complement rather than compete with each other, enabling greater understanding of the genome in relation to drug discovery.


Asunto(s)
Sistemas CRISPR-Cas , Genómica/métodos , Ensayos Analíticos de Alto Rendimiento , Interferencia de ARN , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Humanos , Edición de ARN
10.
Cell ; 157(5): 1160-74, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24855951

RESUMEN

Developmental signaling is remarkably robust to environmental variation, including temperature. For example, in ectothermic animals such as Drosophila, Notch signaling is maintained within functional limits across a wide temperature range. We combine experimental and computational approaches to show that temperature compensation of Notch signaling is achieved by an unexpected variety of endocytic-dependent routes to Notch activation which, when superimposed on ligand-induced activation, act as a robustness module. Thermal compensation arises through an altered balance of fluxes within competing trafficking routes, coupled with temperature-dependent ubiquitination of Notch. This flexible ensemble of trafficking routes supports Notch signaling at low temperature but can be switched to restrain Notch signaling at high temperature and thus compensates for the inherent temperature sensitivity of ligand-induced activation. The outcome is to extend the physiological range over which normal development can occur. Similar mechanisms may provide thermal robustness for other developmental signals.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Endocitosis , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Regulación hacia Abajo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Transducción de Señal , Temperatura
11.
J Cell Sci ; 127(Pt 13): 2849-61, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24816559

RESUMEN

The GTPase Ras can either promote or inhibit cell survival. Inactivating mutations in Drosophila RasGAP (encoded by vap), a Ras GTPase-activating protein, lead to age-related brain degeneration. Genetic interactions implicate the epidermal growth factor receptor (EGFR)-Ras pathway in promoting neurodegeneration but the mechanism is not known. Here, we show that the Src homology 2 (SH2) domains of RasGAP are essential for its neuroprotective function. By using affinity purification and mass spectrometry, we identify a complex containing RasGAP together with Sprint, which is a Ras effector and putative activator of the endocytic GTPase Rab5. Formation of the RasGAP-Sprint complex requires the SH2 domains of RasGAP and tyrosine phosphorylation of Sprint. RasGAP and Sprint colocalize with Rab5-positive early endosomes but not with Rab7-positive late endosomes. We demonstrate a key role for this interaction in neurodegeneration: mutation of Sprint (or Rab5) suppresses neuronal cell death caused by the loss of RasGAP. These results indicate that the long-term survival of adult neurons in Drosophila is crucially dependent on the activities of two GTPases, Ras and Rab5, regulated by the interplay of RasGAP and Sprint.


Asunto(s)
Drosophila/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Supervivencia Celular/fisiología , Drosophila/genética , Endocitosis , Femenino , Masculino , Fosforilación , Transducción de Señal
12.
EMBO Rep ; 11(4): 292-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20224579

RESUMEN

Focal adhesion (FA) disassembly required for optimal cell migration is mediated by microtubules (MTs); targeting of FAs by MTs coincides with their disassembly. Regrowth of MTs, induced by removal of the MT destabilizer nocodazole, activates the Rho-like GTPase Rac, concomitant with FA disassembly. Here, we show that the Rac guanine nucleotide exchange factor (GEF) Sif and Tiam1-like exchange factor (STEF) is responsible for Rac activation during MT regrowth. Importantly, STEF is required for multiple targeting of FAs by MTs. As a result, FAs in STEF-knockdown cells have a reduced disassembly rate and are consequently enlarged. This leads to reduced speed of migration. Together, these findings suggest a new role for STEF in FA disassembly and cell migration through MT-mediated mechanisms.


Asunto(s)
Adhesiones Focales/metabolismo , Factores de Intercambio de Guanina Nucleótido/fisiología , Microtúbulos/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/genética , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Microscopía Fluorescente , Microtúbulos/efectos de los fármacos , Nocodazol/farmacología , Interferencia de ARN/fisiología
13.
Curr Biol ; 20(7): 669-75, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20346677

RESUMEN

Centrosome separation, critical for bipolar spindle formation and subsequent chromosome segregation during mitosis, occurs via distinct prophase and prometaphase pathways. Kinesin-5 (Eg5), a microtubule (MT) motor, pushes centrosomes apart during bipolar spindle assembly; its suppression results in monopolar spindles and mitotic arrest. Forces that antagonize Eg5 in prophase are unknown. Here we identify a new force generating mechanism mediated by the guanine nucleotide exchange factor (GEF) Tiam1, dependent on its ability to activate the GTPase Rac. We reveal that Tiam1 and Rac localize to centrosomes during prophase and prometaphase, and Tiam1, acting through Rac, ordinarily retards centrosome separation. Importantly, both Tiam1-depleted cells in culture and Rac1-deficient epithelial cells in vivo escape the mitotic arrest induced by Eg5 suppression. Moreover, Tiam1-depleted cells transit more slowly through prometaphase and display increased chromosome congression errors. Significantly, Eg5 suppression in Tiam1-depleted cells rectifies not only their increased centrosome separation but also their chromosome congression errors and mitotic delay. These findings identify Tiam1-Rac signaling as the first antagonist of centrosome separation during prophase, demonstrate its requirement in balancing Eg5-induced forces during bipolar spindle assembly in vitro and in vivo, and show that proper centrosome separation in prophase facilitates subsequent chromosome congression.


Asunto(s)
Segregación Cromosómica/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Cinesinas/metabolismo , Huso Acromático/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Secuencia de Bases , Línea Celular , Centrosoma/metabolismo , Perros , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitosis , Modelos Biológicos , Neuropéptidos/genética , Neuropéptidos/metabolismo , Interferencia de ARN , Transducción de Señal , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1
14.
J Proteome Res ; 8(12): 5629-41, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19899799

RESUMEN

The Rac-specific GEF (guanine-nucleotide exchange factor) Tiam1 has important functions in multiple cellular processes including proliferation, apoptosis and adherens junction maintenance. Here we describe a modified tandem affinity purification (TAP) technique that we have applied to specifically enrich Tiam1-containing protein complexes from mammalian cells. Using this technique in conjunction with LC-MS/MS mass spectrometry, we have identified additional Tiam1-interacting proteins not seen with the standard technique, and have identified multiple 14-3-3 family members as Tiam1 interactors. We confirm the Tiam1/14-3-3 protein interaction by GST-pulldown and coimmunoprecipitation experiments, show that it is phosphorylation-dependent, and that they colocalize in cells. The interaction is largely dependent on the N-terminal region of Tiam1; within this region, there are four putative phospho-serine-containing 14-3-3 binding motifs, and we confirm that two of them (Ser172 and Ser231) are phosphorylated in cells using mass spectrometry. Moreover, we show that phosphorylation at three of these motifs (containing Ser60, Ser172 and Ser231) is required for the binding of 14-3-3 proteins to this region of Tiam1. We show that phosphorylation of these sites does not affect Tiam1 activity; significantly however, we demonstrate that phosphorylation of the Ser60-containing motif is required for the degradation of Tiam1. Thus, we have established and proven methodology that allows the identification of additional protein-protein interactions in mammalian cells, resulting in the discovery of a novel mechanism of regulating Tiam1 stability.


Asunto(s)
Proteínas 14-3-3/química , Cromatografía de Afinidad/métodos , Factores de Intercambio de Guanina Nucleótido/química , Proteínas 14-3-3/metabolismo , Animales , Sitios de Unión , Línea Celular , Cromatografía de Afinidad/instrumentación , Factores de Intercambio de Guanina Nucleótido/aislamiento & purificación , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Ratones , Complejos Multiproteicos/química , Complejos Multiproteicos/aislamiento & purificación , Fosforilación , Unión Proteica , Estabilidad Proteica , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Espectrometría de Masas en Tándem
15.
Mol Cell ; 33(5): 639-53, 2009 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-19285946

RESUMEN

The Rac activator Tiam1 is required for adherens junction (AJ) maintenance, and its depletion results in AJ disassembly. Conversely, the oncoprotein Src potently induces AJ disassembly and epithelial-mesenchymal transition (EMT). Here, we show that Tiam1 is phosphorylated on Y384 by Src. This occurs predominantly at AJs, is required for Src-induced AJ disassembly and cell migration, and creates a docking site on Tiam1 for Grb2. We find that Tiam1 is associated with ERK. Following recruitment of the Grb2-Sos1 complex, ERK becomes activated and triggers the localized degradation of Tiam1 at AJs, likely involving calpain proteases. Furthermore, we demonstrate that, in human tumors, Y384 phosphorylation positively correlates with Src activity, and total Tiam1 levels are inversely correlated. Thus, our data implicate Tiam1 phosphorylation and consequent degradation in Src-mediated EMT and resultant cell motility and establish a paradigm for regulating local concentrations of Rho-GEFs.


Asunto(s)
Uniones Adherentes/enzimología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Procesamiento Proteico-Postraduccional , Familia-src Quinasas/metabolismo , Animales , Calpaína/deficiencia , Calpaína/genética , Calpaína/metabolismo , Línea Celular , Movimiento Celular , Clonación Molecular , Perros , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Adaptadora GRB2/metabolismo , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Mutación , Invasividad Neoplásica , Neoplasias/enzimología , Neoplasias/patología , Proteína Oncogénica pp60(v-src)/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-yes/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteína SOS1/metabolismo , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Factores de Tiempo , Transfección , Tirosina , Dominios Homologos src , Familia-src Quinasas/genética
16.
Biochem J ; 380(Pt 3): 767-74, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15030317

RESUMEN

Btl (breathless) and Htl (heartless), the two FGFRs (fibroblast growth factor receptors) in Drosophila melanogaster, control cell migration and differentiation in the developing embryo. These receptors signal through the conserved Ras/mitogen-activated protein kinase pathway, but how they regulate Ras activity is not known. The present study shows that there is a direct interaction between p120 RasGAP (Ras GTPase-activating protein), a negative regulator of Ras, and activated FGFRs in Drosophila. The interaction is dependent on the SH2 (Src homology 2) domains of RasGAP, which have been shown to interact with a phosphotyrosine residue within the consensus sequence (phospho)YXXPXD. A potential binding site that matches this consensus is found in both Btl and Htl, located between the transmembrane and kinase domains of each receptor. A peptide corresponding to this region was capable of binding RasGAP only when the tyrosine residue was phosphorylated. This tyrosine residue appears to be conserved in human FGFR-1 and mediates the association with the adapter protein CrkII, but no association between dCrk (Drosophila homologue of CrkII) and the activated FGFRs was detected. RasGAP was a substrate of the activated FGFR kinase domain, and mutation of the tyrosine residue within the potential binding site on the receptor prevented tyrosine phosphorylation of RasGAP. RasGAP attenuated FGFR signalling in vivo and this ability was dependent on both its SH2 domains and its GAP activity. On the basis of these results, we propose that RasGAP is directly recruited into activated FGFRs in Drosophila and plays a role in regulating the strength of signalling through Ras and the mitogen-activated protein kinase pathway.


Asunto(s)
Drosophila melanogaster/enzimología , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteína Activadora de GTPasa p120/metabolismo , Animales , Sitios de Unión/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Fosforilación , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-crk , Receptores de Factores de Crecimiento de Fibroblastos/química , Receptores de Factores de Crecimiento de Fibroblastos/genética , Transducción de Señal/fisiología , Tirosina/metabolismo , Proteína Activadora de GTPasa p120/química , Proteína Activadora de GTPasa p120/fisiología , Dominios Homologos src/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...