Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38979380

RESUMEN

Integrin α5ß1 is crucial for cell attachment and migration in development and tissue regeneration, and α5ß1 binding proteins could have considerable utility in regenerative medicine and next-generation therapeutics. We use computational protein design to create de novo α5ß1-specific modulating miniprotein binders, called NeoNectins, that bind to and stabilize the open state of α5ß1. When immobilized onto titanium surfaces and throughout 3D hydrogels, the NeoNectins outperform native fibronectin and RGD peptide in enhancing cell attachment and spreading, and NeoNectin-grafted titanium implants outperformed fibronectin and RGD-grafted implants in animal models in promoting tissue integration and bone growth. NeoNectins should be broadly applicable for tissue engineering and biomedicine.

2.
Sci Transl Med ; 16(747): eadj7685, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38748774

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) is an aggressive bile duct malignancy that frequently exhibits isocitrate dehydrogenase (IDH1/IDH2) mutations. Mutant IDH (IDHm) ICC is dependent on SRC kinase for growth and survival and is hypersensitive to inhibition by dasatinib, but the molecular mechanism underlying this sensitivity is unclear. We found that dasatinib reduced p70 S6 kinase (S6K) and ribosomal protein S6 (S6), leading to substantial reductions in cell size and de novo protein synthesis. Using an unbiased phosphoproteomic screen, we identified membrane-associated guanylate kinase, WW, and PDZ domain containing 1 (MAGI1) as an SRC substrate in IDHm ICC. Biochemical and functional assays further showed that SRC inhibits a latent tumor-suppressing function of the MAGI1-protein phosphatase 2A (PP2A) complex to activate S6K/S6 signaling in IDHm ICC. Inhibiting SRC led to activation and increased access of PP2A to dephosphorylate S6K, resulting in cell death. Evidence from patient tissue and cell line models revealed that both intrinsic and extrinsic resistance to dasatinib is due to increased phospho-S6 (pS6). To block pS6, we paired dasatinib with the S6K/AKT inhibitor M2698, which led to a marked reduction in pS6 in IDHm ICC cell lines and patient-derived organoids in vitro and substantial growth inhibition in ICC patient-derived xenografts in vivo. Together, these results elucidated the mechanism of action of dasatinib in IDHm ICC, revealed a signaling complex regulating S6K phosphorylation independent of mTOR, suggested markers for dasatinib sensitivity, and described a combination therapy for IDHm ICC that may be actionable in the clinic.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Colangiocarcinoma , Dasatinib , Isocitrato Deshidrogenasa , Mutación , Familia-src Quinasas , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de los Conductos Biliares/patología , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/patología , Colangiocarcinoma/metabolismo , Colangiocarcinoma/genética , Dasatinib/farmacología , Isocitrato Deshidrogenasa/metabolismo , Isocitrato Deshidrogenasa/genética , Mutación/genética , Fosforilación/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo
3.
J Cell Biol ; 223(6)2024 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-38477830

RESUMEN

Pediatric high-grade gliomas are highly invasive and essentially incurable. Glioma cells migrate between neurons and glia, along axon tracts, and through extracellular matrix surrounding blood vessels and underlying the pia. Mechanisms that allow adaptation to such complex environments are poorly understood. N-cadherin is highly expressed in pediatric gliomas and associated with shorter survival. We found that intercellular homotypic N-cadherin interactions differentially regulate glioma migration according to the microenvironment, stimulating migration on cultured neurons or astrocytes but inhibiting invasion into reconstituted or astrocyte-deposited extracellular matrix. N-cadherin localizes to filamentous connections between migrating leader cells but to epithelial-like junctions between followers. Leader cells have more surface and recycling N-cadherin, increased YAP1/TAZ signaling, and increased proliferation relative to followers. YAP1/TAZ signaling is dynamically regulated as leaders and followers change position, leading to altered N-cadherin levels and organization. Together, the results suggest that pediatric glioma cells adapt to different microenvironments by regulating N-cadherin dynamics and cell-cell contacts.


Asunto(s)
Cadherinas , Glioma , Niño , Humanos , Astrocitos , Axones , Cadherinas/metabolismo , Movimiento Celular , Glioma/metabolismo , Glioma/patología , Microambiente Tumoral
4.
bioRxiv ; 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38260559

RESUMEN

Pediatric high-grade gliomas are highly invasive and essentially incurable. Glioma cells migrate between neurons and glia, along axon tracts, and through extracellular matrix surrounding blood vessels and underlying the pia. Mechanisms that allow adaptation to such complex environments are poorly understood. N-cadherin is highly expressed in pediatric gliomas and associated with shorter survival. We found that inter-cellular homotypic N-cadherin interactions differentially regulate glioma migration according to the microenvironment, stimulating migration on cultured neurons or astrocytes but inhibiting invasion into reconstituted or astrocyte-deposited extracellular matrix. N-cadherin localizes to filamentous connections between migrating leader cells but to epithelial-like junctions between followers. Leader cells have more surface and recycling N-cadherin, increased YAP1/TAZ signaling, and increased proliferation relative to followers. YAP1/TAZ signaling is dynamically regulated as leaders and followers change position, leading to altered N-cadherin levels and organization. Together, the results suggest that pediatric glioma cells adapt to different microenvironments by regulating N-cadherin dynamics and cell-cell contacts.

5.
Elife ; 122023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37489578

RESUMEN

Integrin-mediated cell attachment rapidly induces tyrosine kinase signaling. Despite years of research, the role of this signaling in integrin activation and focal adhesion assembly is unclear. We provide evidence that the Src-family kinase (SFK) substrate Cas (Crk-associated substrate, p130Cas, BCAR1) is phosphorylated and associated with its Crk/CrkL effectors in clusters that are precursors of focal adhesions. The initial phospho-Cas clusters contain integrin ß1 in its inactive, bent closed, conformation. Later, phospho-Cas and total Cas levels decrease as integrin ß1 is activated and core focal adhesion proteins including vinculin, talin, kindlin, and paxillin are recruited. Cas is required for cell spreading and focal adhesion assembly in epithelial and fibroblast cells on collagen and fibronectin. Cas cluster formation requires Cas, Crk/CrkL, SFKs, and Rac1 but not vinculin. Rac1 provides positive feedback onto Cas through reactive oxygen, opposed by negative feedback from the ubiquitin proteasome system. The results suggest a two-step model for focal adhesion assembly in which clusters of phospho-Cas, effectors and inactive integrin ß1 grow through positive feedback prior to integrin activation and recruitment of core focal adhesion proteins.


Asunto(s)
Adhesiones Focales , Fosfoproteínas , Fosforilación , Adhesiones Focales/metabolismo , Fosfoproteínas/metabolismo , Integrina beta1/metabolismo , Proteína Sustrato Asociada a CrK/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Integrinas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo
6.
Elife ; 102021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34169835

RESUMEN

Integrin adhesion complexes regulate cytoskeletal dynamics during cell migration. Adhesion activates phosphorylation of integrin-associated signaling proteins, including Cas (p130Cas, BCAR1), by Src-family kinases. Cas regulates leading-edge protrusion and migration in cooperation with its binding partner, BCAR3. However, it has been unclear how Cas and BCAR3 cooperate. Here, using normal epithelial cells, we find that BCAR3 localization to integrin adhesions requires Cas. In return, Cas phosphorylation, as well as lamellipodia dynamics and cell migration, requires BCAR3. These functions require the BCAR3 SH2 domain and a specific phosphorylation site, Tyr 117, that is also required for BCAR3 downregulation by the ubiquitin-proteasome system. These findings place BCAR3 in a co-regulatory positive-feedback circuit with Cas, with BCAR3 requiring Cas for localization and Cas requiring BCAR3 for activation and downstream signaling. The use of a single phosphorylation site in BCAR3 for activation and degradation ensures reliable negative feedback by the ubiquitin-proteasome system.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteína Sustrato Asociada a CrK/genética , Factores de Intercambio de Guanina Nucleótido/genética , Seudópodos/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adhesión Celular , Línea Celular , Proteína Sustrato Asociada a CrK/metabolismo , Células Epiteliales , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Integrinas/metabolismo , Fosforilación , Dominios Homologos src
7.
Mol Cell Biol ; 40(14)2020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32341084

RESUMEN

Several events during the normal development of the mammalian neocortex depend on N-cadherin, including the radial migration of immature projection neurons into the cortical plate. Remarkably, radial migration requires the N-cadherin extracellular domain but not N-cadherin-dependent homophilic cell-cell adhesion, suggesting that other N-cadherin-binding proteins may be involved. We used proximity ligation and affinity purification proteomics to identify N-cadherin-binding proteins. Both screens detected MycBP2 and SPRY domain protein Fbxo45, two components of an intracellular E3 ubiquitin ligase. Fbxo45 appears to be secreted by a nonclassical mechanism, not involving a signal peptide and not requiring transport from the endoplasmic reticulum to the Golgi apparatus. Fbxo45 binding requires N-cadherin SPRY motifs that are not involved in cell-cell adhesion. SPRY mutant N-cadherin does not support radial migration in vivo Radial migration was similarly inhibited when Fbxo45 expression was suppressed. The results suggest that projection neuron migration requires both Fbxo45 and the binding of Fbxo45 or another protein to SPRY motifs in the extracellular domain of N-cadherin.


Asunto(s)
Encéfalo/embriología , Cadherinas/metabolismo , Proteínas F-Box/metabolismo , Neuronas/citología , Animales , Dominio B30.2-SPRY , Encéfalo/citología , Encéfalo/metabolismo , Cadherinas/análisis , Movimiento Celular , Proteínas F-Box/análisis , Femenino , Células HEK293 , Células HeLa , Humanos , Ratones , Neuronas/metabolismo , Unión Proteica
8.
Elife ; 82019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31577229

RESUMEN

The functions of FGF receptors (FGFRs) in early development of the cerebral cortex are well established. Their functions in the migration of neocortical projection neurons, however, are unclear. We have found that FGFRs regulate multipolar neuron orientation and the morphological change into bipolar cells necessary to enter the cortical plate. Mechanistically, our results suggest that FGFRs are activated by N-Cadherin. N-Cadherin cell-autonomously binds FGFRs and inhibits FGFR K27- and K29-linked polyubiquitination and lysosomal degradation. Accordingly, FGFRs accumulate and stimulate prolonged Erk1/2 phosphorylation. Neurons inhibited for Erk1/2 are stalled in the multipolar zone. Moreover, Reelin, a secreted protein regulating neuronal positioning, prevents FGFR degradation through N-Cadherin, causing Erk1/2 phosphorylation. These findings reveal novel functions for FGFRs in cortical projection neuron migration, suggest a physiological role for FGFR and N-Cadherin interaction in vivo and identify Reelin as an extracellular upstream regulator and Erk1/2 as downstream effectors of FGFRs during neuron migration.


Asunto(s)
Cadherinas/metabolismo , Neocórtex/embriología , Neurogénesis , Neuronas/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Ubiquitinación , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Proteína Reelina , Serina Endopeptidasas/metabolismo
9.
Dev Dyn ; 247(11): 1227-1236, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30269386

RESUMEN

BACKGROUND: The E3 Cullin 5-RING ubiquitin ligase (CRL5) is a multiprotein complex that has recently been highlighted as a major regulator of central nervous system development. Cullin 5 (Cul5) and the RING finger protein Rbx2 are two CRL5 core components required for CRL5 function in the brain, but their full expression patterns and developmental functions have not been described in detail. RESULTS: Using a gene-trap mouse model for Cul5 and a knock-in-knockout mouse model for Rbx2, we show that lack of Cul5, but not Rbx2, disrupts blastocyst formation. However, Rbx2 is required for embryo survival at later embryonic stages. We also show that cul5 is expressed in the embryo proper as early as E7.5 and its expression is mostly restricted to the central nervous system and limbs at later time points. Finally, we show that rbx2 and cul5 are co-expressed in most areas of the brain during development and in the adult. CONCLUSIONS: Our results show that Cul5, but not Rbx2, is required during early embryogenesis and suggests that Cul5 has Rbx2-independent functions in early development. In the brain, Cul5 and Rbx2 are expressed in a similar fashion, allowing the nucleation of an active CRL5 complex. Developmental Dynamics 247:1227-1236, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Química Encefálica , Proteínas Cullin/biosíntesis , Desarrollo Embrionario , Ubiquitina-Proteína Ligasas/análisis , Animales , Embrión de Mamíferos/metabolismo , Ratones , Factores de Tiempo , Ubiquitina-Proteína Ligasas/biosíntesis
10.
Am J Hum Genet ; 102(6): 1143-1157, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29805042

RESUMEN

Non-syndromic cleft lip with or without cleft palate (NS-CL/P) is one of the most common human birth defects and is generally considered a complex trait. Despite numerous loci identified by genome-wide association studies, the effect sizes of common variants are relatively small, with much of the presumed genetic contribution remaining elusive. We report exome-sequencing results in 209 people from 72 multi-affected families with pedigree structures consistent with autosomal-dominant inheritance and variable penetrance. Herein, pathogenic variants are described in four genes encoding components of the p120-catenin complex (CTNND1, PLEKHA7, PLEKHA5) and an epithelial splicing regulator (ESRP2), in addition to the known CL/P-associated gene, CDH1, which encodes E-cadherin. The findings were also validated in a second cohort of 497 people with NS-CL/P, comprising small families and singletons with pathogenic variants in these genes identified in 14% of multi-affected families and 2% of the replication cohort of smaller families. Enriched expression of each gene/protein in human and mouse embryonic oro-palatal epithelia, demonstration of functional impact of CTNND1 and ESRP2 variants, and recapitulation of the CL/P spectrum in Ctnnd1 knockout mice support a causative role in CL/P pathogenesis. These data show that primary defects in regulators of epithelial cell adhesion are the most significant contributors to NS-CL/P identified to date and that inherited and de novo single gene variants explain a substantial proportion of NS-CL/P.


Asunto(s)
Cadherinas/genética , Cateninas/genética , Labio Leporino/genética , Fisura del Paladar/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Alelos , Secuencia de Aminoácidos , Animales , Biotinilación , Epitelio/metabolismo , Epitelio/patología , Femenino , Eliminación de Gen , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Hueso Paladar/patología , Linaje , Síndrome , Secuenciación del Exoma , Catenina delta
11.
Neoplasia ; 20(3): 233-243, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29444487

RESUMEN

Cross talk of stromal-epithelial cells plays an essential role in both normal development and tumor initiation and progression. Fibroblast growth factor (FGF)-FGF receptor (FGFR)-Src kinase axis is one of the major signal transduction pathways to mediate this cross talk. Numerous genomic studies have demonstrated that expression levels of FGFR/Src are deregulated in a variety of cancers including prostate cancer; however, the role that paracrine FGF (from stromal cells) plays in dysregulated expression of epithelial FGFRs/Src and tumor progression in vivo is not well evaluated. In this study, we demonstrate that ectopic expression of wild-type FGFR1/2 or Src kinase in epithelial cells was not sufficient to initiate prostate tumorigenesis under a normal stromal microenvironment in vivo. However, paracrine FGF10 synergized with ectopic expression of epithelial FGFR1 or FGFR2 to induce epithelial-mesenchymal transition. Additionally, paracrine FGF10 sensitized FGFR2-transformed epithelial cells to initiate prostate tumorigenesis. Next, paracrine FGF10 also synergized with overexpression of epithelial Src kinase to high-grade tumors. But loss of the myristoylation site in Src kinase inhibited paracrine FGF10-induced prostate tumorigenesis. Loss of myristoylation alters Src levels in the cell membrane and inhibited FGF-mediated signaling including inhibition of the phosphotyrosine pattern and FAK phosphorylation. Our study demonstrates the potential tumor progression by simultaneous deregulation of proteins in the FGF/FGFRs/Src signal axis and provides a therapeutic strategy of targeting myristoylation of Src kinase to interfere with the tumorigenic process.


Asunto(s)
Transformación Celular Neoplásica/patología , Factores de Crecimiento de Fibroblastos/genética , Genes src/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Oncogenes/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular , Transformación Celular Neoplásica/genética , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/genética , Células del Estroma/metabolismo , Células del Estroma/patología
12.
Sci Rep ; 7(1): 10838, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28883622

RESUMEN

Suppressors of cytokine signaling (SOCS) proteins inhibit signaling by serving as substrate receptors for the Cullin5-RING E3 ubiquitin ligase (CRL5) and through a variety of CRL5-independent mechanisms. CRL5, SOCS2 and SOCS6 are implicated in suppressing transformation of epithelial cells. We identified cell proteins that interact with SOCS2 and SOCS6 using two parallel proteomics techniques: BioID and Flag affinity purification mass spectrometry. The receptor tyrosine kinase ephrin type-A receptor 2 (EphA2) was identified as a SOCS2-interacting protein. SOCS2-EphA2 binding requires the SOCS2 SH2 domain and EphA2 activation loop autophosphorylation, which is stimulated by Ephrin A1 (EfnA1) or by phosphotyrosine phosphatase inhibition. Surprisingly, EfnA1-stimulated EphA2-SOCS2 binding is delayed until EphA2 has been internalized into endosomes. This suggests that SOCS2 binds to EphA2 in the context of endosomal membranes. We also found that SOCS2 overexpression decreases steady state levels of EphA2, consistent with increased EphA2 degradation. This effect is indirect: SOCS2 induces EfnA1 expression, and EfnA1 induces EphA2 down-regulation. Other RTKs have been reported to bind, and be regulated by, over-expressed SOCS proteins. Our data suggest that SOCS protein over-expression may regulate receptor tyrosine kinases through indirect and direct mechanisms.


Asunto(s)
Efrina-A2/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Biomarcadores , Línea Celular , Efrina-A2/genética , Expresión Génica , Humanos , Ratones , Mutación , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteoma , Proteómica/métodos , Receptor EphA2 , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética
13.
Dev Cell ; 41(5): 481-495.e5, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28552558

RESUMEN

Malformations of the cerebral cortex (MCCs) are devastating developmental disorders. We report here that mice with embryonic neural stem-cell-specific deletion of Llgl1 (Nestin-Cre/Llgl1fl/fl), a mammalian ortholog of the Drosophila cell polarity gene lgl, exhibit MCCs resembling severe periventricular heterotopia (PH). Immunohistochemical analyses and live cortical imaging of PH formation revealed that disruption of apical junctional complexes (AJCs) was responsible for PH in Nestin-Cre/Llgl1fl/fl brains. While it is well known that cell polarity proteins govern the formation of AJCs, the exact mechanisms remain unclear. We show that LLGL1 directly binds to and promotes internalization of N-cadherin, and N-cadherin/LLGL1 interaction is inhibited by atypical protein kinase C-mediated phosphorylation of LLGL1, restricting the accumulation of AJCs to the basolateral-apical boundary. Disruption of the N-cadherin-LLGL1 interaction during cortical development in vivo is sufficient for PH. These findings reveal a mechanism responsible for the physical and functional connection between cell polarity and cell-cell adhesion machineries in mammalian cells.


Asunto(s)
Encéfalo/anomalías , Adhesión Celular/fisiología , Polaridad Celular/fisiología , Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/fisiología , Células-Madre Neurales/fisiología , Heterotopia Nodular Periventricular/patología , Proteínas Supresoras de Tumor/fisiología , Animales , Apoptosis , Encéfalo/metabolismo , Encéfalo/patología , Cadherinas/genética , Cadherinas/metabolismo , Proliferación Celular , Células Cultivadas , Proteínas del Citoesqueleto , Células Madre Embrionarias/citología , Femenino , Ratones , Ratones Transgénicos , Nestina/genética , Nestina/metabolismo , Células-Madre Neurales/citología , Heterotopia Nodular Periventricular/metabolismo , Fosforilación
14.
Sci Rep ; 7: 43760, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28272509

RESUMEN

The Reelin-Dab1 signaling pathway regulates development of the mammalian brain, including neuron migrations in various brain regions, as well as learning and memory in adults. Extracellular Reelin binds to cell surface receptors and activates phosphorylation of the intracellular Dab1 protein. Dab1 is required for most effects of Reelin, but Dab1-independent pathways may contribute. Here we developed a single-component, photoactivatable Dab1 (opto-Dab1) by using the blue light-sensitive dimerization/oligomerization property of A. thaliana Cryptochrome 2 (Cry2). Opto-Dab1 can activate downstream signals rapidly, locally, and reversibly upon blue light illumination. The high spatiotemporal resolution of the opto-Dab1 probe also allows us to control membrane protrusion, retraction and ruffling by local illumination in both COS7 cells and in primary neurons. This shows that Dab1 activation is sufficient to orient cell movement in the absence of other signals. Opto-Dab1 may be useful to study the biological functions of the Reelin-Dab1 signaling pathway both in vitro and in vivo.


Asunto(s)
Luz , Proteínas del Tejido Nervioso/metabolismo , Optogenética/métodos , Transducción de Señal/efectos de la radiación , Animales , Células COS , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Chlorocebus aethiops , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Células HEK293 , Humanos , Ratones , Microscopía Confocal , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neuronas/efectos de la radiación , Fosforilación/efectos de la radiación , Proteína Reelina , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo
15.
Elife ; 52016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27656905

RESUMEN

Cell migration requires the cyclical assembly and disassembly of focal adhesions. Adhesion induces phosphorylation of focal adhesion proteins, including Cas (Crk-associated substrate/p130Cas/BCAR1). However, Cas phosphorylation stimulates adhesion turnover. This raises the question of how adhesion assembly occurs against opposition from phospho-Cas. Here we show that suppressor of cytokine signaling 6 (SOCS6) and Cullin 5, two components of the CRL5SOCS6 ubiquitin ligase, inhibit Cas-dependent focal adhesion turnover at the front but not rear of migrating epithelial cells. The front focal adhesions contain phospho-Cas which recruits SOCS6. If SOCS6 cannot access focal adhesions, or if cullins or the proteasome are inhibited, adhesion disassembly is stimulated. This suggests that the localized targeting of phospho-Cas within adhesions by CRL5SOCS6 and concurrent cullin and proteasome activity provide a negative feedback loop, ensuring that adhesion assembly predominates over disassembly at the leading edge. By this mechanism, ubiquitination provides a new level of spatio-temporal control over cell migration.


Asunto(s)
Movimiento Celular , Células Epiteliales/fisiología , Adhesiones Focales/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Línea Celular , Proteína Sustrato Asociada a CrK/metabolismo , Proteínas Cullin/metabolismo , Humanos , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
16.
J Clin Invest ; 126(4): 1311-22, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26927671

RESUMEN

Acute and chronic tissue injury results in the generation of a myriad of environmental cues that macrophages respond to by changing their phenotype and function. This phenotypic regulation is critical for controlling tissue inflammation and resolution. Here, we have identified the adaptor protein disabled homolog 2 (DAB2) as a regulator of phenotypic switching in macrophages. Dab2 expression was upregulated in M2 macrophages and suppressed in M1 macrophages isolated from both mice and humans, and genetic deletion of Dab2 predisposed macrophages to adopt a proinflammatory M1 phenotype. In mice with myeloid cell-specific deletion of Dab2 (Dab2fl/fl Lysm-Cre), treatment with sublethal doses of LPS resulted in increased proinflammatory gene expression and macrophage activation. Moreover, chronic high-fat feeding exacerbated adipose tissue inflammation, M1 polarization of adipose tissue macrophages, and the development of insulin resistance in DAB2-deficient animals compared with controls. Mutational analyses revealed that DAB2 interacts with TNF receptor-associated factor 6 (TRAF6) and attenuates IκB kinase ß-dependent (IKKß-dependent) phosphorylation of Ser536 in the transactivation domain of NF-κB p65. Together, these findings reveal that DAB2 is critical for controlling inflammatory signaling during phenotypic polarization of macrophages and suggest that manipulation of DAB2 expression and function may hold therapeutic potential for the treatment of acute and chronic inflammatory disorders.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras del Transporte Vesicular/biosíntesis , Tejido Adiposo/metabolismo , Regulación de la Expresión Génica , Macrófagos/metabolismo , Paniculitis/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Tejido Adiposo/patología , Animales , Proteínas Reguladoras de la Apoptosis , Línea Celular , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Paniculitis/genética , Paniculitis/patología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Proteínas Supresoras de Tumor/genética
17.
Mol Cell Biol ; 35(11): 1886-97, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25776560

RESUMEN

Three classes of E3 ubiquitin ligases, members of the Cbl, Hakai, and SOCS-Cul5-RING ligase families, stimulate the ubiquitination of phosphotyrosine-containing proteins, including receptor and nonreceptor tyrosine kinases and their phosphorylated substrates. Because ubiquitination frequently routes proteins for degradation by the lysosome or proteasome, these E3 ligases are able to potently inhibit tyrosine kinase signaling. Their loss or mutational inactivation can contribute to cancer, autoimmunity, or endocrine disorders, such as diabetes. However, these ligases also have biological functions that are independent of their ubiquitination activity. Here we review relevant literature and then focus on more-recent developments in understanding the structures, substrates, and pathways through which the phosphotyrosine-specific ubiquitin ligases regulate diverse aspects of cell biology.


Asunto(s)
Fosfotirosina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Humanos , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/fisiología , Ubiquitinación/fisiología
18.
Front Cell Neurosci ; 8: 386, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25452716

RESUMEN

Most neurons migrate with an elongated, "bipolar" morphology, extending a long leading process that explores the environment. However, when immature projection neurons enter the intermediate zone (IZ) of the neocortex they become "multipolar". Multipolar cells extend and retract cytoplasmic processes in different directions and move erratically-sideways, up and down. Multipolar cells extend axons while they are in the lower half of the IZ. Remarkably, the cells then resume radial migration: they reorient their centrosome and Golgi apparatus towards the pia, transform back to bipolar morphology, and commence locomotion along radial glia (RG) fibers. This reorientation implies the existence of directional signals in the IZ that are ignored during the multipolar stage but sensed after axonogenesis. In vivo genetic manipulation has implicated a variety of candidate directional signals, cell surface receptors, and signaling pathways, that may be involved in polarizing multipolar cells and stabilizing a pia-directed leading process for radial migration. Other signals are implicated in starting multipolar migration and triggering axon outgrowth. Here we review the molecules and mechanisms that regulate multipolar migration, and also discuss how multipolar migration affects the orderly arrangement of neurons in layers and columns in the developing neocortex.

19.
Arterioscler Thromb Vasc Biol ; 34(11): 2404-12, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25212232

RESUMEN

OBJECTIVE: The essential role of platelet activation in hemostasis and thrombotic diseases focuses attention on unveiling the underlying intracellular signals of platelet activation. Disabled-2 (Dab2) has been implicated in platelet aggregation and in the control of clotting responses. However, there is not yet any in vivo study to provide direct evidence for the role of Dab2 in hemostasis and platelet activation. APPROACH AND RESULTS: Megakaryocyte lineage-restricted Dab2 knockout (Dab2(-/-)) mice were generated to delineate in vivo functions of Dab2 in platelets. Dab2(-/-) mice appeared normal in size with prolonged bleeding time and impaired thrombus formation. Although normal in platelet production and granule biogenesis, Dab2(-/-) platelets elicited a selective defect in platelet aggregation and spreading on fibrinogen in response to low concentrations of thrombin, but not other soluble agonists. Investigation of the role of Dab2 in thrombin signaling revealed that Dab2 has no effect on the expression of thrombin receptors and the outside-in signaling. Dab2(-/-) platelets stimulated by low concentrations of thrombin were normal in Gαq-mediated calcium mobilization and protein kinase C activation, but were defective in Gα12/13-mediated RhoA-ROCKII activation. The attenuated Gα12/13 signaling led to impaired ADP release, Akt-mammalian target of rapamycin and integrin αIIbß3 activation, fibrinogen binding, and clot retraction. The defective responses of Dab2(-/-) platelets to low concentrations of thrombin stimulation may contribute to the impaired hemostasis and thrombosis of Dab2(-/-) mice. CONCLUSIONS: This study sheds new insight in platelet biology and represents the first report demonstrating that Dab2 is a key regulator of hemostasis and thrombosis by functional interplay with Gα12/13-mediated thrombin signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de Unión al ADN/fisiología , Hemostasis/fisiología , Activación Plaquetaria/fisiología , Transducción de Señal/fisiología , Trombina/fisiología , Trombosis/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Adenosina Difosfato/fisiología , Animales , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología , Proteína Quinasa C/fisiología , Serina-Treonina Quinasas TOR/fisiología , Quinasas Asociadas a rho/fisiología , Proteína de Unión al GTP rhoA/fisiología
20.
J Cell Sci ; 127(Pt 3): 509-20, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24284072

RESUMEN

Phosphorylation-dependent protein ubiquitylation and degradation provides an irreversible mechanism to terminate protein kinase signaling. Here, we report that mammary epithelial cells require cullin-5-RING-E3-ubiquitin-ligase complexes (Cul5-CRLs) to prevent transformation by a Src-Cas signaling pathway. Removal of Cul5 stimulates growth-factor-independent growth and migration, membrane dynamics and colony dysmorphogenesis, which are all dependent on the endogenous tyrosine kinase Src. Src is activated in Cul5-deficient cells, but Src activation alone is not sufficient to cause transformation. We found that Cul5 and Src together stimulate degradation of the Src substrate p130Cas (Crk-associated substrate). Phosphorylation stimulates Cas binding to the Cul5-CRL adaptor protein SOCS6 and consequent proteasome-dependent degradation. Cas is necessary for the transformation of Cul5-deficient cells. Either knockdown of SOCS6 or use of a degradation-resistant Cas mutant stimulates membrane ruffling, but not other aspects of transformation. Our results show that endogenous Cul5 suppresses epithelial cell transformation by several pathways, including inhibition of Src-Cas-induced ruffling through SOCS6.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteína Sustrato Asociada a CrK/metabolismo , Proteínas Cullin/genética , Familia-src Quinasas/metabolismo , Animales , Movimiento Celular/genética , Proliferación Celular , Proteínas Cullin/metabolismo , Células Epiteliales/metabolismo , Técnicas de Silenciamiento del Gen , Ratones , Transducción de Señal/genética , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA