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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 125(Pt 18): 4241-52, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22641690

RESUMEN

In culture, cell confluence generates signals that commit actively growing keratinocytes to exit the cell cycle and differentiate to form a stratified epithelium. Using a comparative proteomic approach, we studied this 'confluence switch' and identified a new pathway triggered by cell confluence that regulates basement membrane (BM) protein composition by suppressing the uPA-uPAR-plasmin pathway. Indeed, confluence triggers adherens junction maturation and enhances TGF-ß and activin A activity, resulting in increased deposition of PAI-1 and perlecan in the BM. Extracellular matrix (ECM)-accumulated PAI-1 suppresses the uPA-uPAR-plasmin pathway and further enhances perlecan deposition by inhibiting its plasmin-dependent proteolysis. We show that perlecan deposition in the ECM strengthens cell adhesion, inhibits keratinocyte motility and promotes additional accumulation of PAI-1 in the ECM at confluence. In agreement, during wound-healing, perlecan concentrates at the wound-margin, where BM matures to stabilize keratinocyte adhesion. Our results demonstrate that confluence-dependent signaling orchestrates not only growth inhibition and differentiation, but also controls ECM proteolysis and BM formation. These data suggest that uncontrolled integration of confluence-dependent signaling, might favor skin disorders, including tumorigenesis, not only by promoting cell hyperproliferation, but also by altering protease activity and deposition of ECM components.


Asunto(s)
Matriz Extracelular/metabolismo , Fibrinolisina/metabolismo , Queratinocitos/metabolismo , Proteolisis , Transducción de Señal , Activinas/metabolismo , Uniones Adherentes/metabolismo , Animales , Membrana Basal/metabolismo , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Retroalimentación Fisiológica , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Queratinocitos/patología , Ratones , Plasminógeno/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Unión Proteica , Proteómica , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Cicatrización de Heridas
2.
Eur J Clin Microbiol Infect Dis ; 30(8): 965-72, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21311940

RESUMEN

Staphylococcus aureus, a major causative agent of human infection, produces a large array of virulence factors, including various toxins. Among them, the host RhoA GTPase ADP-ribosylating EDIN toxins are considered as potential virulence factors. Using the polymerase chain reaction (PCR) assay, we analyzed the virulence profile of 256 S. aureus isolates from various clinical sites of infections. We developed specific primers to detect the three isoforms of edin-encoding genes. We found a prevalence of 14% (36 bacteria) of edin-encoding genes among these clinical isolates. Strikingly, we found that 90% of all edin-bearing S. aureus isolates carried the type-C allele. Both the spa types and the profile of virulence factors of these edin-positive isolates are highly variable. Notably, we show for the first time that edin-C-positive isolates were more frequently recovered from deep-seated infections than other types of infections. Our present work, thus, strongly suggests that the presence of edin-C is a risk factor of S. aureus dissemination in tissues and, thus, represents a predictive marker for a pejorative evolution of staphylococcal infections.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Factores de Virulencia/biosíntesis , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Alelos , Proteínas Bacterianas/genética , Cartilla de ADN/genética , ADN Bacteriano/genética , Variación Genética , Genotipo , Humanos , Reacción en Cadena de la Polimerasa/métodos , Prevalencia , Staphylococcus aureus/genética , Staphylococcus aureus/aislamiento & purificación , Factores de Virulencia/genética
3.
J Cell Biol ; 145(2): 317-30, 1999 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-10209027

RESUMEN

Pollen tube cells elongate based on actin- dependent targeted secretion at the tip. Rho family small GTPases have been implicated in the regulation of related processes in animal and yeast cells. We have functionally characterized Rac type Rho family proteins that are expressed in growing pollen tubes. Expression of dominant negative Rac inhibited pollen tube elongation, whereas expression of constitutive active Rac induced depolarized growth. Pollen tube Rac was found to accumulate at the tip plasma membrane and to physically associate with a phosphatidylinositol monophosphate kinase (PtdIns P-K) activity. Phosphatidylinositol 4, 5-bisphosphate (PtdIns 4, 5-P2), the product of PtdIns P-Ks, showed a similar intracellular localization as Rac. Expression of the pleckstrin homology (PH)-domain of phospholipase C (PLC)-delta1, which binds specifically to PtdIns 4, 5-P2, inhibited pollen tube elongation. These results indicate that Rac and PtdIns 4, 5-P2 act in a common pathway to control polar pollen tube growth and provide direct evidence for a function of PtdIns 4, 5-P2 compartmentalization in the regulation of this process.


Asunto(s)
Arabidopsis/fisiología , Proteínas de Unión al GTP/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Polen/fisiología , Secuencia de Aminoácidos , Arabidopsis/ultraestructura , Células Cultivadas , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Glicosilación , Isoenzimas/metabolismo , Datos de Secuencia Molecular , Fosfolipasa C delta , Proteínas de Plantas/metabolismo , Estructuras de las Plantas , Plantas Tóxicas , Polen/ultraestructura , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Nicotiana , Transfección , Fosfolipasas de Tipo C/metabolismo , Proteínas de Unión al GTP rac
4.
Mol Biol Cell ; 8(12): 2437-47, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9398666

RESUMEN

Pretreatment of intact rabbit portal vein smooth muscle with the chimeric toxin DC3B (10(-6) M, 48 h; ; ) ADP-ribosylated endogenous RhoA, including cytosolic RhoA complexed with rhoGDI, and inhibited the tonic phase of phenylephrine-induced contraction and the Ca2+-sensitization of force by phenylephrine, endothelin and guanosine triphosphate (GTP)gammaS, but did not inhibit Ca2+-sensitization by phorbol dibutyrate. DC3B also inhibited GTPgammaS-induced translocation of cytosolic RhoA () to the membrane fraction. In DC3B-treated muscles the small fraction of membrane-associated RhoA could be immunoprecipitated, even after exposure to GTPgammaS, which prevents immunoprecipitation of non-ADP-ribosylated RhoA. Dissociation of cytosolic RhoA-rhoGDI complexes with SDS restored the immunoprecipitability and ADP ribosylatability of RhoA, indicating that both the ADP-ribosylation site (Asn 41) and RhoA insert loop (Wei et al., 1997) are masked by rhoGDI and that the long axes of the two proteins are in parallel in the heterodimer. We conclude that RhoA plays a significant role in G-protein-, but not protein kinase C-mediated, Ca2+ sensitization and that ADP ribosylation inhibits in vivo the Ca2+-sensitizing effect of RhoA by interfering with its binding to a membrane-associated effector.


Asunto(s)
ADP Ribosa Transferasas/farmacología , Adenosina Difosfato/metabolismo , Toxinas Botulínicas , Calcio/antagonistas & inhibidores , Toxina Diftérica/farmacología , Proteína de Unión al GTP rhoA/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Calcio/metabolismo , Calcio/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Endotelinas/antagonistas & inhibidores , Endotelinas/farmacología , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/antagonistas & inhibidores , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Técnicas In Vitro , Contracción Isométrica/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Fragmentos de Péptidos/farmacología , Fenilefrina/antagonistas & inhibidores , Fenilefrina/farmacología , Forbol 12,13-Dibutirato/farmacología , Vena Porta/efectos de los fármacos , Vena Porta/metabolismo , Vena Porta/fisiología , Pruebas de Precipitina , Unión Proteica/efectos de los fármacos , Conejos , Proteínas Recombinantes de Fusión/farmacología , Dodecil Sulfato de Sodio/farmacología , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico , Proteína de Unión al GTP rhoA/antagonistas & inhibidores
5.
Virologie (Montrouge) ; 11(4): 289-296, 2007 Aug 01.
Artículo en Francés | MEDLINE | ID: mdl-36131476

RESUMEN

The curvature of host cell membranes during the budding of HIV viruses occurs by an opposite topology, as compared to endocytosis and phagocytosis phenomena. This topology of vesicle formation is indeed observed during the formation of internal vesicles of late endosomal compartments called multivesicular bodies (MVB). Formation of these vesicles is controlled by ubiquitylation and the recruitment of the endosomal sorting complex required for transport (ESCRT). The sorting of HIV-1 structural protein Pr55gag into the viral bud requires the recruitment of the ESCRT complex, potentiated by Pr55gag ubiquitylation. Interestingly, in response to viral infection, type I interferon triggers the expression of the ubiquitin-like polypeptide ISG15 which then inhibits the recruitment of the ESCRT machinery by inhibiting Pr55gag ubiquitylation.

6.
Curr Top Microbiol Immunol ; 291: 177-90, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15981464

RESUMEN

The CNF1 toxin is produced by some uropathogenic (UPECs) andmeningitis-causing Escherichia coli strains. It belongs to a large family of bacterial virulence factors and toxins modifying cellular regulators of the actin cytoskeleton, namely the Rho GTPases. CNF1 autonomously enters the host cell cytosol, where it catalyzes the constitutive activation of Rho GTPases by deamidation. This activation is, however, attenuated because of activated Rho protein ubiquitin-mediated proteasomal degradation. Both Rho protein activation and deactivation confer phagocytic properties on epithelial and endothelial cells, as well as epithelial cell motility and cell-cell junction dynamics. Transcriptome analysis using DNA microarray revealed that endothelial cells respond to high doses of CNF1 by launching a genetic program of host alarm. This host cell reaction to CNF1 intoxication also indicates that degradation of activated Rho proteins by the proteasome may lead to a lowering of the threshold of the intoxicated cell inflammatory response. These results are consistent with growing evidence that Rho proteins control the cell inflammatory responses. It is tempting to assume that Rho deregulation may participate in various immunological disorders also involved in cancer.


Asunto(s)
Toxinas Bacterianas/metabolismo , Activadores de Enzimas/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Toxinas Bacterianas/toxicidad , Citotoxinas/toxicidad , Activación Enzimática , Proteínas de Escherichia coli/toxicidad , Humanos , Factores de Virulencia/toxicidad
7.
AIDS ; 12(8): 859-63, 1998 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-9631138

RESUMEN

OBJECTIVES: To study the antiviral efficacy of the recombinant immunotoxin DAB389CD4 against wild-type strains of HIV and to analyse its potential toxicity in non-infected peripheral blood mononuclear cells (PBMC). DESIGN AND METHODS: PBMC from HIV-seropositive patients were cultured in the presence of DAB389CD4. After 30 days in culture, viral load was assessed by quantification of RNA levels in supernatants and HIV-specific polymerase chain reaction (PCR) was performed for measuring proviral DNA as an indicator of remaining virus in cells. To study the toxicity of DAB389CD4, PBMC from healthy donors were isolated and cell viability and lymphocyte proliferation were assessed after immunotoxin treatment. RESULTS: DAB389CD4 presented a strong antiviral activity in five of the six primary isolates decreasing p24 production in cultures to undetectable levels and eliminating selectively HIV-infected cells as measured by HIV DNA-specific PCR. One viral isolate was resistant to DAB389CD4 treatment. The immunotoxin was active against both syncytial and non-syncytial HIV strains. DAB389CD4 was not toxic in non-infected PBMC as measured by different techniques: trypan blue exclusion, methyl thiazol tetrazolium oxidation, lymphocyte proliferation, and CD4 cell count. CONCLUSIONS: DAB389CD4 showed a strong antiviral and specific activity against primary HIV isolates by killing selectively HIV-infected cells without affecting non-infected cells. This antiviral effect produced the eradication of HIV in cultures and indicated the potential use of this drug as a new therapeutic tool in combination with antiretroviral drugs. This immunotoxin would be especially interesting in the context of the marginal populations of HIV-infected cells remaining after successful antiviral treatment.


Asunto(s)
Fármacos Anti-VIH/farmacología , Antígenos CD4/farmacología , VIH/fisiología , Inmunotoxinas/farmacología , Leucocitos Mononucleares/virología , Antígenos CD4/genética , Antígenos CD4/toxicidad , Supervivencia Celular , Células Cultivadas , Toxina Diftérica/genética , Relación Dosis-Respuesta a Droga , Proteína p24 del Núcleo del VIH/biosíntesis , Humanos , Inmunotoxinas/toxicidad , Fenotipo , Provirus , ARN Viral/análisis , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/toxicidad , Replicación Viral
8.
Eur Cytokine Netw ; 10(2): 227-36, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10400829

RESUMEN

The role of the polymorphonuclear leukocyte (PMN) cytoskeleton during the transmigration across colonic epithelial cells is not very well understood. In order to study the role of different components of the PMN cytoskeleton during transepithelial migration across a colonic epithelial cell monolayer (T84), PMN were preincubated with drugs affecting either the actin cytoskeleton (cytochalasin B, iota toxin of Clostridium perfringens, and phalloidin) or the microtubules (colchicine and taxol). The role of PMN myosin during transepithelial migration was investigated using the inhibitor 2,3-butanedione monoxime (BDM) and DC3B toxin. PMN intracellular Ca2+, during neutrophil adhesion and translocation across the epithelium, was assessed by the Ca2+ chelator 1, 2bis-(2-aminophenoxy)-ethane-N,N,N', N'-tetra-acetic acid tetrakis (acetoxymethyl) ester (BAPTA-AM). Transmigration of PMN was initiated by applying either interleukin-8 or formyl-met-leu-phe (fMLP). While colchicine and taxol preexposure did not influence PMN transepithelial migration, treatment with cytochalasin B, iota toxin, phalloidin, BDM, DC3B toxin and BAPTA-AM greatly diminished migration of PMN across T84 monolayers. Similarly, cell-cell contacts established between PMN and epithelial cells during the transmigration were diminished after treatment of PMN with iota toxin or cytochalasin B. These data show that the neutrophil actin cytokeleton and myosin, but not the microtubules, evoke a Ca2+ -dependent motility that facilitates migration across the colonic epithelial barrier.


Asunto(s)
ADP Ribosa Transferasas , Actinas/fisiología , Quimiotaxis de Leucocito/efectos de los fármacos , Interleucina-8/farmacología , Mucosa Intestinal/citología , Microtúbulos/fisiología , Miosinas/fisiología , Neutrófilos/fisiología , Toxinas Bacterianas/farmacología , Calcio/metabolismo , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Quelantes/farmacología , Colchicina/farmacología , Neoplasias del Colon/patología , Citocalasina B/farmacología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Diacetil/análogos & derivados , Diacetil/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Células Epiteliales/citología , Humanos , Microtúbulos/efectos de los fármacos , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/efectos de los fármacos , Neutrófilos/ultraestructura , Paclitaxel/farmacología , Faloidina/farmacología , Células Tumorales Cultivadas
9.
Clin Microbiol Infect ; 19(9): 875-80, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23176291

RESUMEN

Staphylococcus aureus is both a common colonizer of human skin and the most frequently isolated pathogen in diabetes foot infections (DFIs). The spread of DFI to soft tissue and bony structures is a major causal factor for lower-limb amputation. It is therefore of great importance to differentiate colonizing from infecting strains of S. aureus. Epidermal cell differentiation inhibitors known as EDIN and EDIN-like factors, a group of toxins targeting RhoA master regulator of the actin cytoskeleton, may confer virulence properties on S. aureus. In this study, for the first time, analysis of S. aureus strains, recovered in DFIs at an initial stage and during the follow-up, showed that 71.4% of edin-positive strains were associated with moderate-to-severe infections (grades 3 and 4 of the IDSA/IWGDF classification) compared with 28.6% of edin-positive strains associated with low-grade infections. Most of these strains were edin-B positive (86.7%) and belonged to CC25/28-MSSA (n = 10). One edin-B-positive ST152-MSSA strain was negative for the two highly prevalent predictive markers of infecting strains (lukDE and hlgv). Collectively, this points towards the edin-B encoding gene as a bonafide subsidiary predictive risk marker of DFI.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pie Diabético/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Factores de Virulencia/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Bacterianas/genética , Exotoxinas/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Staphylococcus aureus/clasificación , Staphylococcus aureus/aislamiento & purificación , Factores de Virulencia/genética , Proteína de Unión al GTP rhoA/metabolismo
10.
Clin Microbiol Infect ; 19(1): 85-90, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22268649

RESUMEN

Extraintestinal pathogenic Escherichia coli (ExPEC) strains, a major cause of bacteraemia, typically belong to phylogenetic group B2 and express diverse accessory traits that contribute to virulence in mouse infection models. However, their high genomic diversity obscures the relationship between virulence factors and severity of infection in patients. In this study, we analysed concomitantly the strain's expression of virulence in a mouse model, genomic determinants and the clinical severity of infection in 60 bacteraemic patients. We show that bacterial virulence based on an animal model study and virulence factor determination is not correlated with pejorative outcome of E. coli human blood infections.


Asunto(s)
Bacteriemia/microbiología , Infecciones por Escherichia coli/microbiología , Escherichia coli/patogenicidad , Análisis de Varianza , Animales , Distribución de Chi-Cuadrado , Modelos Animales de Enfermedad , Escherichia coli/genética , Femenino , Perfilación de la Expresión Génica , Genes Bacterianos , Humanos , Ratones , Modelos Estadísticos , Filogenia , Virulencia
12.
Nature ; 387(6634): 729-33, 1997 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-9192901

RESUMEN

Pathogenic Escherichia coli are responsible for a variety of diseases, including diarrhoea, haemolytic uraemic syndrome, kidney infection, septicaemia, pneumonia and meningitis. Toxins called cytotoxic necrotizing factors (CNFs) are among the virulence factors produced by uropathogenic (CNF1) or enteropathogenic (CNF2) E. coli strains that cause diseases in humans and animals, respectively. CNFs induce an increase in the content of actin stress fibres and focal contacts in cultured cells. Effects of CNFs on the actin cytoskeleton correlated with a decrease in the electrophoretic mobility of the GTP-binding protein Rho and indirect evidence indicates that CNF1 might constitutively activate Rho. Here we show that CNF1 catalyses the deamidation of a glutamine residue at position 63 of Rho, turning it into glutamic acid, which inhibits both intrinsic GTP hydrolysis and that stimulated by its GTPase-activating protein (GAP). Thus, this deamidation of glutamine 63 by CNF1 leads to the constitutive activation of Rho, and induces the reorganization of actin stress fibres. To our knowledge, CNF1 is the first example of a bacterial toxin acting by deamidation of a specific target protein.


Asunto(s)
Toxinas Bacterianas/farmacología , Citotoxinas/farmacología , Proteínas de Escherichia coli , Proteínas de Unión al GTP/metabolismo , Proteínas Activadoras de GTPasa , Glutamina/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Electroforesis en Gel de Poliacrilamida , Escherichia coli , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/química , Guanosina Trifosfato/metabolismo , Cinética , Datos de Secuencia Molecular , Células Vero , Proteínas de Unión al GTP rho , Proteína de Unión al GTP rhoA
13.
Mol Microbiol ; 24(5): 1061-70, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9220012

RESUMEN

Cytotoxic necrotizing factor type 1 (CNF1) induces, in epithelial cells, the development of stress fibres via the GTPase Rho pathway. We showed that CNF1 is able to modify Rho both in vitro and in vivo. Recombinant N-terminal 33kDa (CNF1Nter) and C-terminal 14.8-31.5 kDa (CNF1Cter) regions of the CNF1 protein allowed us to demonstrate that the N-terminal region contains the cell-binding domain of the toxin and that the C-terminal region is responsible for its catalytic activity. CNF1Nter lowered the activity of CNF1 when provided to cells before the toxin whereas CNF1Cter had no effect on CNF1 cell toxicity. CNF1Cter was sufficient to induce a typical CNF1 phenotype when microinjected into African green monkey kidney cells (Vero cells), and was able to modify Rho as previously reported for CNF1. The C-terminal domain lost its catalytic activity when deleted of various subdomains, suggesting a scattered distribution of catalytic-site amino acids. Elucidation of the CNF1 functional organization and analysis of amino acid homologies between CNFs (CNF1, CNF2), Pasteurella multocida toxin (PMT) and dermonecrotic toxin of Bordetella pertussis (DNT) allowed us to postulate that CNFs and DNT act on Rho via the same enzymatic activity located in their C-terminus, and that CNFs and PMT probably bind to analogous cell receptors.


Asunto(s)
Toxinas Bacterianas/metabolismo , Citotoxinas/metabolismo , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Secuencia de Aminoácidos , Animales , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Sitios de Unión , Catálisis , Chlorocebus aethiops , Citotoxinas/química , Citotoxinas/genética , Proteínas de Unión al GTP/metabolismo , Glutatión Transferasa/genética , Humanos , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/genética , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas , Células Vero , Proteínas de Unión al GTP rho
14.
Genes Dev ; 15(14): 1808-16, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11459830

RESUMEN

Plant water homeostasis is maintained by the phytohormone abscisic acid (ABA), which triggers stomatal pore closure in response to drought stress. We identified the Arabidopsis small guanosine triphosphatase (GTPase) protein AtRac1 as a central component in the ABA-mediated stomatal closure process. ABA treatment induced inactivation of AtRac GTPases and disruption of the guard cell actin cytoskeleton. In contrast, in the ABA-insensitive mutant abi1-1, which is impaired in stomatal closure, neither AtRac inactivation nor actin cytoskeleton disruption was observed on ABA treatment. These observations indicate that AtRac1 inactivation is a limiting step in the ABA-signaling cascade leading to stomatal closure. Consistent with these findings, expression of a dominant-positive mutant of AtRac1 blocked the ABA-mediated effects on actin cytoskeleton and stomatal closure in wild-type plants, whereas expression of a dominant-negative AtRac1 mutant recapitulated the ABA effects in the absence of the hormone. Moreover, the dominant-negative form of AtRac1 could also restore stomatal closure in abi1-1. These results define AtRac1 as a central element for plant adaptation to drought.


Asunto(s)
Ácido Abscísico/farmacología , Hojas de la Planta/fisiología , Proteínas de Plantas/antagonistas & inhibidores , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Secuencia de Aminoácidos , Arabidopsis , Biología Computacional , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Mutación , Hojas de la Planta/citología , Plantas Modificadas Genéticamente , Homología de Secuencia de Aminoácido
15.
Mol Microbiol ; 23(3): 445-57, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9044279

RESUMEN

After reaching early endosomes by receptor-mediated endocytosis, diphtheria toxin (DT) molecules have two possible fates. A large pool enters the degradative pathway whereas a few molecules become cytotoxic by translocating their catalytic fragment A (DTA) into the cytosol. Impairment of DT degradation by microtubule depolymerization does not block DT cytotoxicity. Therefore, DTA membrane translocation into the cytosol occurs from an endocytic compartment located upstream of late endosomes. Comparisons between early endosomes and endocytic carrier vesicles in a cell-free translocation assay have demonstrated that early endosomes are the earliest endocytic compartment from which DTA translocates. DTA translocation is ATP-dependent, requires early endosomal acidification, and is increased by the addition of cytosol. Cytosol-dependent DTA translocation is GTP gamma S-insensitive but is blocked by anti-beta COP antibodies.


Asunto(s)
Toxina Diftérica/fisiología , Endosomas/metabolismo , Fragmentos de Péptidos/fisiología , Animales , Proteínas Portadoras/metabolismo , Sistema Libre de Células , Invaginaciones Cubiertas de la Membrana Celular , Vesículas Cubiertas , Citosol/química , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Técnicas In Vitro , Lisosomas/metabolismo , Fusión de Membrana , Microscopía Confocal , Microscopía Fluorescente
16.
J Biol Chem ; 272(31): 19532-7, 1997 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9235957

RESUMEN

Cytotoxic necrotizing factor 1 (CNF1), a 110-kDa protein toxin from pathogenic Escherichia coli induces actin reorganization into stress fibers and retraction fibers in human epithelial cultured cells allowing them to spread. CNF1 is acting in the cytosol since microinjection of the toxin into HEp-2 cells mimics the effects of the externally applied CNF1. Incubation in vitro of CNF1 with recombinant small GTPases induces a modification of Rho (but not of Rac, Cdc42, Ras, or Rab6) as demonstrated by a discrete increase in the apparent molecular weight of the molecule. Preincubation of cells with CNF1 impairs the cytotoxic effects of Clostridium difficile toxin B, which inactivates Rho but not those of Clostridium sordellii LT toxin, which inhibits Ras and Rac. As shown for Rho-GTP, CNF1 activates, in a time- and dose-dependent manner, a cytoskeleton-associated phosphatidylinositol 4-phosphate 5-kinase. However, neither the phosphatidylinositol 4,5-bisphosphate (PIP2) nor the phosphatidylinositol 3,4-bisphosphate (PI 3,4-P2) or 3,4,5-trisphosphate (PIP3) cellular content were found increased in CNF1 treated HEp-2 cells. Cellular effects of CNF1 were not blocked by LY294002, a stable inhibitor of the phosphoinositide 3-kinase. Incubation of HEp-2 cells with CNF1 induces relocalization of myosin 2 in stress fibers but not in retraction fibers. Altogether, our data indicate that CNF1 is a toxin that selectively activates the Rho GTP-binding protein, thus inducing contractility and cell spreading.


Asunto(s)
Proteínas Bacterianas , Toxinas Bacterianas/farmacología , Citotoxinas/farmacología , Proteínas de Escherichia coli , Proteínas de Unión al GTP/efectos de los fármacos , Proteínas de la Membrana/efectos de los fármacos , Actinas/biosíntesis , Animales , Chlorocebus aethiops , Diacetil/análogos & derivados , Diacetil/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Miosinas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/efectos de los fármacos , Células Vero , Proteína de Unión al GTP rhoB
17.
Mol Cell ; 8(1): 115-27, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11511365

RESUMEN

Adhesion to fibronectin through the alpha5beta1 integrin enables endothelial cells to proliferate in response to growth factors, whereas adhesion to laminin through alpha2beta1 results in growth arrest under the same conditions. On laminin, endothelial cells fail to translate Cyclin D1 mRNA and activate CDK4 and CDK6. Activated Rac, but not MEK1, PI-3K, or Akt, rescues biosynthesis of cyclin D1 and progression through the G(1) phase. Conversely, dominant negative Rac prevents these events on fibronectin. Mitogens promote activation of Rac on fibronectin but not laminin. This process is mediated by SOS and PI-3K and requires coordinate upstream signals through Shc and FAK. These results indicate that Rac is a crucial mediator of the integrin-specific control of cell cycle in endothelial cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Quinasas CDC2-CDC28 , Endotelio Vascular/citología , Fase G1/fisiología , Integrinas/metabolismo , Proteínas Proto-Oncogénicas , Receptores de Fibronectina/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Northern Blotting , Caveolina 1 , Caveolinas/metabolismo , Adhesión Celular , Células Cultivadas , Medio de Cultivo Libre de Suero , Ciclina D1/genética , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Adhesiones Focales/metabolismo , Humanos , Immunoblotting , Insulina/farmacología , Integrinas/genética , Laminina/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Microscopía Fluorescente , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas/metabolismo , Receptores de Colágeno , Receptores de Fibronectina/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína SOS1/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
18.
Proc Natl Acad Sci U S A ; 92(23): 10629-33, 1995 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-7479854

RESUMEN

The rho family of GTP-binding proteins regulates actin filament organization. In unpolarized mammalian cells, rho proteins regulate the assembly of actin-containing stress fibers at the cell-matrix interface. Polarized epithelial cells, in contrast, are tall and cylindrical with well developed intercellular tight junctions that permit them to behave as biologic barriers. We report that rho regulates filamentous actin organization preferentially in the apical pole of polarized intestinal epithelial cells and, in so doing, influences the organization and permeability of the associated apical tight junctions. Thus, barrier function, which is an essential characteristic of columnar epithelia, is regulated by rho.


Asunto(s)
Actinas/fisiología , Toxinas Botulínicas , Polaridad Celular/fisiología , Proteínas de Unión al GTP/metabolismo , Intestinos/citología , Uniones Estrechas/fisiología , Proteínas de Unión al GTP rho , ADP Ribosa Transferasas/metabolismo , Actinas/ultraestructura , Western Blotting , Cadherinas/metabolismo , Adhesión Celular , Comunicación Celular , Células Cultivadas , Células Epiteliales , Técnica del Anticuerpo Fluorescente , Proteínas de Unión al GTP/genética , Humanos , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Pruebas de Precipitina , Proteínas Recombinantes/metabolismo , Transducción de Señal , Uniones Estrechas/ultraestructura , Proteína de la Zonula Occludens-1 , Proteína rhoC de Unión a GTP
19.
Cell Immunol ; 182(2): 105-12, 1997 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9514697

RESUMEN

Molecules of the tetraspan superfamily are engaged in multimolecular complexes containing other proteins such as beta 1 integrins and MHC antigens. Although their functions are not clear, they have been suggested to play a role in cell adhesion and migration, signal transduction, and costimulation. We have in this paper directly compared the functional properties of four tetraspans, CD9, CD53, CD81, and CD82. mAbs to any of these molecules were able to deliver a costimulatory signal for CD3-mediated activation of the T cell line Jurkat. CD82 mAbs were the most efficient in triggering this effect. Moreover, engagement of CD9, CD81, and CD82 induced the homotypic aggregation of the megakaryocytic cell line HEL, and inhibited the migration of this cell line. Similar results were obtained with the preB cell line NALM-6 using the CD9 and CD81 mAbs. The CD81 mAb 5A6 produced the strongest effects. Therefore, the tetraspans are recognized by mAbs which produce similar effects on the same cell lines. This is consistent with the tetraspans being included in large molecular complexes and possibly forming a tetraspan network (the tetraspan web). We also demonstrate that the tetraspans are likely to keep specific functional properties inside this network. Indeed, we have demonstrated that the human CD9 is able, like the monkey molecule, to upregulate the activity of the transmembrane precursor of heparin-binding EGF as a receptor for the diphtheria toxin when cotransfected in murine LM cells. Neither CD81, nor CD82 had such activity. By using chimeric CD9/CD81 molecules we demonstrate that this activity requires the second half of CD9, which contains the large extracellular loop, the fourth transmembrane region, and the last short cytoplasmic domain.


Asunto(s)
Antígenos CD/fisiología , Antígenos de Diferenciación de Linfocitos T/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Heparina/metabolismo , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana , Proteínas Proto-Oncogénicas , Animales , Anticuerpos Monoclonales , Antígenos CD/química , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/química , Antígenos de Diferenciación de Linfocitos T/genética , Secuencia de Bases , Adhesión Celular/inmunología , Línea Celular , Movimiento Celular/inmunología , Cartilla de ADN/genética , Toxina Diftérica/farmacología , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Péptidos y Proteínas de Señalización Intercelular , Interleucina-2/biosíntesis , Proteína Kangai-1 , Células L , Activación de Linfocitos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Ratones , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Tetraspanina 25 , Tetraspanina 28 , Tetraspanina 29 , Transfección , Regulación hacia Arriba
20.
J Biol Chem ; 271(17): 10217-24, 1996 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-8626586

RESUMEN

Lethal toxin (LT) from Clostridium sordellii is one of the high molecular mass clostridial cytotoxins. On cultured cells, it causes a rounding of cell bodies and a disruption of actin stress fibers. We demonstrate that LT is a glucosyltransferase that uses UDP-Glc as a cofactor to covalently modify 21-kDa proteins both in vitro and in vivo. LT glucosylates Ras, Rap, and Rac. In Ras, threonine at position 35 was identified as the target amino acid glucosylated by LT. Other related members of the Ras GTPase superfamily, including RhoA, Cdc42, and Rab6, were not modified by LT. Incubation of serum-starved Swiss 3T3 cells with LT prevents the epidermal growth factor-induced phosphorylation of mitogen-activated protein kinases ERK1 and ERK2, indicating that the toxin blocks Ras function in vivo. We also demonstrate that LT acts inside the cell and that the glucosylation reaction is required to observe its dramatic effect on cell morphology. LT is thus a powerful tool to inhibit Ras function in vivo.


Asunto(s)
Toxinas Bacterianas/metabolismo , Clostridium/patogenicidad , Proteínas de Unión al GTP/metabolismo , Glucosiltransferasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células 3T3 , Citoesqueleto de Actina/ultraestructura , Actinas/química , Secuencia de Aminoácidos , Animales , Toxinas Bacterianas/toxicidad , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Factor de Crecimiento Epidérmico/farmacología , GTP Fosfohidrolasas/metabolismo , Glucosa/metabolismo , Guanosina Trifosfato/metabolismo , Células HeLa/efectos de los fármacos , Células HeLa/ultraestructura , Humanos , Cinética , Ratones , Datos de Secuencia Molecular , Treonina/metabolismo , Uridina Difosfato Glucosa/metabolismo , Proteínas de Unión al GTP rac , Proteínas de Unión al GTP rap
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA