Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nature ; 450(7168): 430-4, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17960134

RESUMEN

Engulfment and subsequent degradation of apoptotic cells is an essential step that occurs throughout life in all multicellular organisms. ELMO/Dock180/Rac proteins are a conserved signalling module for promoting the internalization of apoptotic cell corpses; ELMO and Dock180 function together as a guanine nucleotide exchange factor (GEF) for the small GTPase Rac, and thereby regulate the phagocyte actin cytoskeleton during engulfment. However, the receptor(s) upstream of the ELMO/Dock180/Rac module are still unknown. Here we identify brain-specific angiogenesis inhibitor 1 (BAI1) as a receptor upstream of ELMO and as a receptor that can bind phosphatidylserine on apoptotic cells. BAI1 is a seven-transmembrane protein belonging to the adhesion-type G-protein-coupled receptor family, with an extended extracellular region and no known ligands. We show that BAI1 functions as an engulfment receptor in both the recognition and subsequent internalization of apoptotic cells. Through multiple lines of investigation, we identify phosphatidylserine, a key 'eat-me' signal exposed on apoptotic cells, as a ligand for BAI1. The thrombospondin type 1 repeats within the extracellular region of BAI1 mediate direct binding to phosphatidylserine. As with intracellular signalling, BAI1 forms a trimeric complex with ELMO and Dock180, and functional studies suggest that BAI1 cooperates with ELMO/Dock180/Rac to promote maximal engulfment of apoptotic cells. Last, decreased BAI1 expression or interference with BAI1 function inhibits the engulfment of apoptotic targets ex vivo and in vivo. Thus, BAI1 is a phosphatidylserine recognition receptor that can directly recruit a Rac-GEF complex to mediate the uptake of apoptotic cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Angiogénicas/metabolismo , Apoptosis , Factores de Intercambio de Guanina Nucleótido/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rac/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Angiogénicas/genética , Animales , Línea Celular , Cricetinae , Cricetulus , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Ligandos , Ratones , Fagocitosis , Fosfatidilserinas/metabolismo , Unión Proteica , Timo/citología , Timo/metabolismo , Proteínas de Unión al GTP rac/genética
2.
Nat Struct Mol Biol ; 11(8): 756-62, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15247908

RESUMEN

The members of the Dock180 superfamily of proteins are novel guanine nucleotide exchange factors (GEF) for Rho family GTPases and are linked to multiple biological processes from worms to mammals. ELMO is a critical regulator of Dock180, and the Dock180-ELMO complex functions as a bipartite GEF for Rac. We identified a mechanism wherein the PH domain of ELMO, by binding the Dock180-Rac complex in trans, stabilizes Rac in the nucleotide-free transition state. Mutagenesis studies reveal that this ELMO PH domain-dependent regulation is essential for the Dock180-ELMO complex to function in phagocytosis and cell migration. Genetic rescue studies in Caenorhabditis elegans using ELMO and its homolog CED-12 support the above observations in vivo. These data reveal a new mode of action of PH domains and a novel, evolutionarily conserved mechanism by which a bipartite GEF can activate Rac.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/química , Proteínas de Unión al GTP rac/química , Animales , Proteínas Reguladoras de la Apoptosis , Células CHO , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Línea Celular , Movimiento Celular , Cricetinae , Proteínas del Citoesqueleto/metabolismo , Dimerización , Activación Enzimática , Glutatión Transferasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Immunoblotting , Microscopía Fluorescente , Mutagénesis , Mutación , Fagocitosis , Plásmidos/metabolismo , Pruebas de Precipitina , Estructura Terciaria de Proteína , Espectrometría de Fluorescencia , Factores de Tiempo , Transgenes
3.
Cancer Res ; 67(15): 7203-11, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671188

RESUMEN

A distinct feature of malignant gliomas is the intrinsic ability of single tumor cells to disperse throughout the brain, contributing to the failure of existing therapies to alter the progression and recurrence of these deadly brain tumors. Regrettably, the mechanisms underlying the inherent invasiveness of glioma cells are poorly understood. Here, we report for the first time that engulfment and cell motility 1 (ELMO1) and dedicator of cytokinesis 1 (Dock180), a bipartite Rac1 guanine nucleotide exchange factor (GEF), are evidently linked to the invasive phenotype of glioma cells. Immunohistochemical analysis of primary human glioma specimens showed high expression levels of ELMO1 and Dock180 in actively invading tumor cells in the invasive areas, but not in the central regions of these tumors. Elevated expression of ELMO1 and Dock180 was also found in various human glioma cell lines compared with normal human astrocytes. Inhibition of endogenous ELMO1 and Dock180 expression significantly impeded glioma cell invasion in vitro and in brain tissue slices with a concomitant reduction in Rac1 activation. Conversely, exogenous expression of ELMO1 and Dock180 in glioma cells with low level endogenous expression increased their migratory and invasive capacity in vitro and in brain tissue. These data suggest that the bipartite GEF, ELMO1 and Dock180, play an important role in promoting cancer cell invasion and could be potential therapeutic targets for the treatment of diffuse malignant gliomas.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Encefálicas/patología , Glioma/patología , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Neoplasias Encefálicas/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Glioma/metabolismo , Humanos , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Plásmidos/metabolismo , Transfección , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética
4.
Nat Commun ; 10(1): 4456, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575859

RESUMEN

Fertilization is essential for species survival. Although Izumo1 and Juno are critical for initial interaction between gametes, additional molecules necessary for sperm:egg fusion on both the sperm and the oocyte remain to be defined. Here, we show that phosphatidylserine (PtdSer) is exposed on the head region of viable and motile sperm, with PtdSer exposure progressively increasing during sperm transit through the epididymis. Functionally, masking phosphatidylserine on sperm via three different approaches inhibits fertilization. On the oocyte, phosphatidylserine recognition receptors BAI1, CD36, Tim-4, and Mer-TK contribute to fertilization. Further, oocytes lacking the cytoplasmic ELMO1, or functional disruption of RAC1 (both of which signal downstream of BAI1/BAI3), also affect sperm entry into oocytes. Intriguingly, mammalian sperm could fuse with skeletal myoblasts, requiring PtdSer on sperm and BAI1/3, ELMO2, RAC1 in myoblasts. Collectively, these data identify phosphatidylserine on viable sperm and PtdSer recognition receptors on oocytes as key players in sperm:egg fusion.


Asunto(s)
Oocitos/metabolismo , Fagocitos/metabolismo , Fosfatidilserinas/metabolismo , Interacciones Espermatozoide-Óvulo/fisiología , Espermatozoides/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Angiogénicas/metabolismo , Animales , Antígenos CD36/metabolismo , Proteínas del Citoesqueleto/metabolismo , Epidídimo , Femenino , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Mioblastos Esqueléticos , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Fosfatidilserinas/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Tirosina Quinasa c-Mer/metabolismo , Proteína de Unión al GTP rac1/metabolismo
5.
Curr Biol ; 14(24): 2208-16, 2004 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-15620647

RESUMEN

BACKGROUND: Phagocytosis of cells undergoing apoptosis is essential during development, cellular turnover, and wound healing. Failure to promptly clear apoptotic cells has been linked to autoimmune disorders. C. elegans CED-12 and mammalian ELMO are evolutionarily conserved scaffolding proteins that play a critical role in engulfment from worm to human. ELMO functions together with Dock180 (a guanine nucleotide exchange factor for Rac) to mediate Rac-dependent cytoskeletal reorganization during engulfment and cell migration. However, the components upstream of ELMO and Dock180 during engulfment remain elusive. RESULTS: Here, we define a conserved signaling module involving the small GTPase RhoG and its exchange factor TRIO, which functions upstream of ELMO/Dock180/Rac during engulfment. Complementary studies in C. elegans show that MIG-2 (which we identify as the homolog of mammalian RhoG) and UNC-73 (the TRIO homolog) also regulate corpse clearance in vivo, upstream of CED-12. At the molecular level, we identify a novel set of evolutionarily conserved Armadillo (ARM) repeats within CED-12/ELMO that mediate an interaction with activated MIG-2/RhoG; this, in turn, promotes Dock180-mediated Rac activation and cytoskeletal reorganization. CONCLUSIONS: The combination of in vitro and in vivo studies presented here identify two evolutionarily conserved players in engulfment, TRIO/UNC73 and RhoG/MIG-2, and the TRIO --> RhoG signaling module is linked by ELMO/CED-12 to Dock180-dependent Rac activation during engulfment. This work also identifies ARM repeats within CED-12/ELMO and their role in linking RhoG and Rac, two GTPases that function in tandem during engulfment.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Citoesqueleto/metabolismo , GTP Fosfohidrolasas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fagocitosis/fisiología , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rac/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis , Caenorhabditis elegans , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Humanos , Secuencias Repetitivas de Ácidos Nucleicos/genética , Proteínas de Unión al GTP rho
6.
Cell Rep ; 10(3): 307-316, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25600866

RESUMEN

Cancer develops after the acquisition of a collection of mutations that together create the cancer phenotype. How collections of mutations work together within a cell and whether there is selection for certain combinations of mutations are not well understood. We investigated this problem with a mathematical model of the Ras signaling network, including a computational random mutagenesis. Modeling and subsequent experiments revealed that mutations of the tumor suppressor gene NF1 can amplify the effects of other Ras pathway mutations, including weakly activating, noncanonical Ras mutants. Furthermore, analyzing recently available, large, cancer genomic data sets uncovered increased co-occurrence of NF1 mutations with mutations in other Ras network genes. Overall, these data suggest that combinations of Ras pathway mutations could serve the role of cancer "driver." More generally, this work suggests that mutations that result in network instability may promote cancer in a manner analogous to genomic instability.

7.
Nat Cell Biol ; 13(1): 79-86, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21170032

RESUMEN

Multicellular animals rapidly clear dying cells from their bodies. Many of the pathways that mediate this cell removal are conserved through evolution. Here, we identify srgp-1 as a negative regulator of cell clearance in both Caenorhabditis elegans and mammalian cells. Loss of srgp-1 function results in improved engulfment of apoptotic cells, whereas srgp-1 overexpression inhibits apoptotic cell corpse removal. We show that SRGP-1 functions in engulfing cells and functions as a GTPase activating protein (GAP) for CED-10 (Rac1). Interestingly, loss of srgp-1 function promotes not only the clearance of already dead cells, but also the removal of cells that have been brought to the verge of death through sublethal apoptotic, necrotic or cytotoxic insults. In contrast, impaired engulfment allows damaged cells to escape clearance, which results in increased long-term survival. We propose that C. elegans uses the engulfment machinery as part of a primitive, but evolutionarily conserved, survey mechanism that identifies and removes unfit cells within a tissue.


Asunto(s)
Apoptosis , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Línea Celular , Proteínas Activadoras de GTPasa/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Células 3T3 NIH , Fagocitosis , Unión Proteica , Interferencia de ARN , Homología de Secuencia de Aminoácido , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo
9.
J Biol Chem ; 281(9): 5928-37, 2006 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-16377631

RESUMEN

ERMs are closely related proteins involved in cell migration, cell adhesion, maintenance of cell shape, and formation of microvilli through their ability to cross-link the plasma membrane with the actin cytoskeleton. ELMO proteins are also known to regulate actin cytoskeleton reorganization through activation of the small GTPbinding protein Rac via the ELMO-Dock180 complex. Here we showed that ERM proteins associate directly with ELMO1 as purified recombinant proteins in vitro and at endogenous levels in intact cells. We mapped ERM binding on ELMO1 to the N-terminal 280 amino acids, which overlaps with the region required for binding to the GTPase RhoG, but is distinct from the C-terminal Dock180 binding region. Consistent with this, ELMO1 could simultaneously bind both radixin and Dock180, although radixin did not alter Rac activation via the Dock180-ELMO complex. Most interestingly, radixin binding did not affect ELMO binding to active RhoG and a trimeric complex of active RhoG-ELMO-radixin could be detected. Moreover, the three proteins colocalized at the plasma membrane. Finally, in contrast to most other ERM-binding proteins, ELMO1 binding occurred independently of the state of radixin C-terminal phosphorylation, suggesting an ELMO1 interaction with both the active and inactive forms of ERM proteins and implying a possible role of ELMO in localizing or retaining ERM proteins in certain cellular sites. Together these data suggest that ELMO1-mediated cytoskeletal changes may be coordinated with ERM protein crosslinking activity during dynamic cellular functions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Sitios de Unión , Línea Celular , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Complejos Multiproteicos , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/genética , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo
10.
J Cell Physiol ; 195(3): 435-45, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12704653

RESUMEN

Mesangial cells in diverse glomerular diseases become myofibroblast-like, characterized by activation of smooth muscle alpha-actin (alpha-SMA) expression. In cultured mesangial cells, serum-deprivation markedly increases alpha-SMA expression, cell size, and stress fiber formation. Since stress fibers are assembled from actin monomers, we investigated the hypothesis that alterations in stress fiber formation regulate alpha-SMA expression and hypertrophy. Human mesangial cells were treated with agents that disrupt or stabilize actin stress fibers. Depolymerization of actin stress fibers in serum-deprived cells with actin-depolymerizing agents, cytochalasin B (CytB) and latrunculin B (LatB), or with inhibitors of Rho-kinase, Y-27632 and HA-1077 decreased alpha-SMA mRNA as judged by Northern blot analysis. Western blot analysis showed that CytB also reduced alpha-SMA protein levels. In serum-fed cells, agents that stabilized actin stress fibers, jasplakinolide (Jas) and phalloidin, increased alpha-SMA mRNA and protein. Treatment of human or rat mesangial cells with CytB, LatB, or Y-27632 decreased alpha-SMA promoter activity. In contrast, Jas increased promoter activity 5.6-fold in rat mesangial cells. The presence of an RNA polymerase inhibitor blocked degradation of alpha-SMA mRNA in cells treated with CytB suggesting that destabilization of this message is dependent on a newly transcribed or rapidly degraded factor. Inhibition of actin polymerization by CytB, LatB, Y-27623, and HA-1077 inhibited incorporation of (3)[H]-leucine into newly synthesized protein. Additionally, CytB and LatB decreased cell volume as determined by flow cytometry. Collectively, these results indicate that the state of polymerization of the actin cytoskeleton regulates alpha-SMA expression, hypertrophy, and myofibroblast differentiation in mesangial cells.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Actinas/metabolismo , Depsipéptidos , Fibroblastos/citología , Fibroblastos/metabolismo , Mesangio Glomerular/citología , Citoesqueleto de Actina/efectos de los fármacos , Actinas/genética , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Citocalasina B/farmacología , Inhibidores Enzimáticos/farmacología , Fibroblastos/ultraestructura , Humanos , Péptidos y Proteínas de Señalización Intracelular , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Péptidos Cíclicos/farmacología , Faloidina/farmacología , Fenotipo , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Estabilidad del ARN , ARN Mensajero/metabolismo , Ratas , Tiazoles/farmacología , Tiazolidinas , Activación Transcripcional , Quinasas Asociadas a rho
11.
J Biol Chem ; 279(7): 6087-97, 2004 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-14638695

RESUMEN

Cell migration is essential throughout embryonic and adult life. In numerous cell systems, the small GTPase Rac is required for lamellipodia formation at the leading edge and movement ability. However, the molecular mechanisms leading to Rac activation during migration are still unclear. Recently, a mammalian superfamily of proteins related to the prototype member Dock180 has been identified with homologues in Drosophila and Caenorhabditis elegans. Here, we addressed the role of Dock180 and ELMO1 proteins, which function as a complex to mediate Rac activation, in mammalian cell migration. Using mutants of Dock180 and ELMO1 in a Transwell assay as well as transgenic rescue of a C. elegans mutant lacking CED-5 (Dock180 homologue), we identified specific regions of Dock180 and ELMO1 required for migration in vitro and in a whole animal model. In both systems, the Dock180.ELMO1 complex formation and the ability to activate Rac were required. We also found that ELMO1 regulated multiple Dock180 superfamily members to promote migration. Interestingly, deletion mutants of ELMO1 missing their first 531 or first 330 amino acids that can still bind and cooperate with Dock180 in Rac activation failed to promote migration, which correlated with the inability to localize to lamellipodia. This finding suggests that Rac activation by the ELMO.Dock180 complex at discrete intracellular locations mediated by the N-terminal 330 amino acids of ELMO1 rather than generalized Rac activation plays a role in cell migration.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Caenorhabditis elegans , Proteínas Portadoras/química , Adhesión Celular , Línea Celular , Movimiento Celular , Evolución Molecular , Genotipo , Glutatión Transferasa/metabolismo , Guanosina Trifosfato/química , Humanos , Immunoblotting , Microscopía Fluorescente , Mutación , Fagocitosis , Plásmidos/metabolismo , Pruebas de Precipitina , Estructura Terciaria de Proteína , Factores de Tiempo , Transfección , Proteínas de Unión al GTP rac/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA