Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 6059, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36229487

RESUMEN

Extracellular matrix (ECM) elasticity is perceived by cells via focal adhesion structures, which transduce mechanical cues into chemical signalling to conform cell behavior. Although the contribution of ECM compliance to the control of cell migration or division is extensively studied, little is reported regarding infectious processes. We study this phenomenon with the extraintestinal Escherichia coli pathogen UTI89. We show that UTI89 takes advantage, via its CNF1 toxin, of integrin mechanoactivation to trigger its invasion into cells. We identify the HACE1 E3 ligase-interacting protein Optineurin (OPTN) as a protein regulated by ECM stiffness. Functional analysis establishes a role of OPTN in bacterial invasion and integrin mechanical coupling and for stimulation of HACE1 E3 ligase activity towards the Rac1 GTPase. Consistent with a role of OPTN in cell mechanics, OPTN knockdown cells display defective integrin-mediated traction force buildup, associated with limited cellular invasion by UTI89. Nevertheless, OPTN knockdown cells display strong mechanochemical adhesion signalling, enhanced Rac1 activation and increased cyclin D1 translation, together with enhanced cell proliferation independent of ECM stiffness. Together, our data ascribe a new function to OPTN in mechanobiology.


Asunto(s)
Ciclina D1 , Integrinas , División Celular , Ciclina D1/metabolismo , Integrinas/metabolismo , Mecanotransducción Celular/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteína de Unión al GTP rac1/metabolismo
3.
Blood Adv ; 5(5): 1523-1534, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33683342

RESUMEN

Dysregulated immune response is the key factor leading to unfavorable coronavirus disease 2019 (COVID-19) outcome. Depending on the pathogen-associated molecular pattern, the NLRP3 inflammasome can play a crucial role during innate immunity activation. To date, studies describing the NLRP3 response during severe acute respiratory syndrome coronavirus 2 infection in patients are lacking. We prospectively monitored caspase-1 activation levels in peripheral myeloid cells from healthy donors and patients with mild to critical COVID-19. The caspase-1 activation potential in response to NLRP3 inflammasome stimulation was opposed between nonclassical monocytes and CD66b+CD16dim granulocytes in severe and critical COVID-19 patients. Unexpectedly, the CD66b+CD16dim granulocytes had decreased nigericin-triggered caspase-1 activation potential associated with an increased percentage of NLRP3 inflammasome impaired immature neutrophils and a loss of eosinophils in the blood. In patients who recovered from COVID-19, nigericin-triggered caspase-1 activation potential in CD66b+CD16dim cells was restored and the proportion of immature neutrophils was similar to control. Here, we reveal that NLRP3 inflammasome activation potential differs among myeloid cells and could be used as a biomarker of a COVID-19 patient's evolution. This assay could be a useful tool to predict patient outcome. This trial was registered at www.clinicaltrials.gov as #NCT04385017.


Asunto(s)
COVID-19/sangre , Inflamasomas/metabolismo , Células Mieloides/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Biomarcadores/sangre , COVID-19/inmunología , Estudios de Casos y Controles , Humanos , Inflamasomas/sangre , Persona de Mediana Edad , Estudios Prospectivos , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación
4.
Nat Microbiol ; 6(3): 401-412, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33432150

RESUMEN

Inflammasomes are signalling platforms that are assembled in response to infection or sterile inflammation by cytosolic pattern recognition receptors. The consequent inflammasome-triggered caspase-1 activation is critical for the host defence against pathogens. During infection, NLRP3, which is a pattern recognition receptor that is also known as cryopyrin, triggers the assembly of the inflammasome-activating caspase-1 through the recruitment of ASC and Nek7. The activation of the NLRP3 inflammasome is tightly controlled both transcriptionally and post-translationally. Despite the importance of the NLRP3 inflammasome regulation in autoinflammatory and infectious diseases, little is known about the mechanism controlling the activation of NLRP3 and the upstream signalling that regulates the NLRP3 inflammasome assembly. We have previously shown that the Rho-GTPase-activating toxin from Escherichia coli cytotoxic necrotizing factor-1 (CNF1) activates caspase-1, but the upstream mechanism is unclear. Here, we provide evidence of the role of the NLRP3 inflammasome in sensing the activity of bacterial toxins and virulence factors that activate host Rho GTPases. We demonstrate that this activation relies on the monitoring of the toxin's activity on the Rho GTPase Rac2. We also show that the NLRP3 inflammasome is activated by a signalling cascade that involves the p21-activated kinases 1 and 2 (Pak1/2) and the Pak1-mediated phosphorylation of Thr 659 of NLRP3, which is necessary for the NLRP3-Nek7 interaction, inflammasome activation and IL-1ß cytokine maturation. Furthermore, inhibition of the Pak-NLRP3 axis decreases the bacterial clearance of CNF1-expressing UTI89 E. coli during bacteraemia in mice. Taken together, our results establish that Pak1 and Pak2 are critical regulators of the NLRP3 inflammasome and reveal the role of the Pak-NLRP3 signalling axis in vivo during bacteraemia in mice.


Asunto(s)
Bacteriemia/metabolismo , Toxinas Bacterianas/metabolismo , Infecciones por Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Bacteriemia/inmunología , Bacteriemia/microbiología , Carga Bacteriana , Toxinas Bacterianas/genética , Escherichia coli/genética , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Inmunidad Innata , Ratones , Fosforilación , Transducción de Señal , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/genética , Proteína RCA2 de Unión a GTP
5.
Cancer Cell ; 36(3): 268-287.e10, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31447347

RESUMEN

GAPDH is emerging as a key player in T cell development and function. To investigate the role of GAPDH in T cells, we generated a transgenic mouse model overexpressing GAPDH in the T cell lineage. Aged mice developed a peripheral Tfh-like lymphoma that recapitulated key molecular, pathological, and immunophenotypic features of human angioimmunoblastic T cell lymphoma (AITL). GAPDH induced non-canonical NF-κB pathway activation in mouse T cells, which was strongly activated in human AITL. We developed a NIK inhibitor to reveal that targeting the NF-κB pathway prolonged AITL-bearing mouse survival alone and in combination with anti-PD-1. These findings suggest the therapeutic potential of targeting NF-κB signaling in AITL and provide a model for future AITL therapeutic investigations.


Asunto(s)
Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Linfadenopatía Inmunoblástica/patología , Linfoma de Células T/patología , FN-kappa B/metabolismo , Linfocitos T/inmunología , Anciano , Animales , Línea Celular Tumoral , Linaje de la Célula/inmunología , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Células HEK293 , Humanos , Linfadenopatía Inmunoblástica/genética , Linfoma de Células T/tratamiento farmacológico , Linfoma de Células T/genética , Linfoma de Células T/inmunología , Masculino , Ratones Transgénicos , Persona de Mediana Edad , FN-kappa B/genética , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Quinasa de Factor Nuclear kappa B
6.
Sci Rep ; 8(1): 1410, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29362425

RESUMEN

The regulation of Rac1 by HACE1-mediated ubiquitination and proteasomal degradation is emerging as an essential element in the maintenance of cell homeostasis. However, how the E3 ubiquitin ligase activity of HACE1 is regulated remains undetermined. Using a proteomic approach, we identified serine 385 as a target of group-I PAK kinases downstream Rac1 activation by CNF1 toxin from pathogenic E. coli. Moreover, cell treatment with VEGF also promotes Ser-385 phosphorylation of HACE1. We have established in vitro that HACE1 is a direct target of PAK1 kinase activity. Mechanistically, we found that the phospho-mimetic mutant HACE1(S385E), as opposed to HACE1(S385A), displays a lower capacity to ubiquitinate Rac1 in cells. Concomitantly, phosphorylation of Ser-385 plays a pivotal role in controlling the oligomerization state of HACE1. Finally, Ser-385 phosphorylated form of HACE1 localizes in the cytosol away from its target Rac1. Together, our data point to a feedback inhibition of HACE1 ubiquitination activity on Rac1 by group-I PAK kinases.


Asunto(s)
Serina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Toxinas Bacterianas/farmacología , Línea Celular , Proteínas de Escherichia coli/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Fosforilación , Multimerización de Proteína , Proteómica , Ubiquitinación , Factor A de Crecimiento Endotelial Vascular/farmacología
7.
Nat Commun ; 8: 15839, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28643776

RESUMEN

Transendothelial cell macroaperture (TEM) tunnels control endothelium barrier function and are triggered by several toxins from pathogenic bacteria that provoke vascular leakage. Cellular dewetting theory predicted that a line tension of uncharacterized origin works at TEM boundaries to limit their widening. Here, by conducting high-resolution microscopy approaches we unveil the presence of an actomyosin cable encircling TEMs. We develop a theoretical cellular dewetting framework to interpret TEM physical parameters that are quantitatively determined by laser ablation experiments. This establishes the critical role of ezrin and non-muscle myosin II (NMII) in the progressive implementation of line tension. Mechanistically, fluorescence-recovery-after-photobleaching experiments point for the upstream role of ezrin in stabilizing actin filaments at the edges of TEMs, thereby favouring their crosslinking by NMIIa. Collectively, our findings ascribe to ezrin and NMIIa a critical function of enhancing line tension at the cell boundary surrounding the TEMs by promoting the formation of an actomyosin ring.


Asunto(s)
Actomiosina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actomiosina/química , Actomiosina/genética , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Células Endoteliales de la Vena Umbilical Humana/química , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Miosina Tipo IIA no Muscular/química , Miosina Tipo IIA no Muscular/genética , Tensión Superficial
8.
Sci Rep ; 7: 44779, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28317937

RESUMEN

The E3 ubiquitin ligase HACE1 is a potent tumor suppressor that controls cell proliferation and ubiquitylates the small GTPase Rac1 to target it to proteasomal degradation. Whether and how the activity of HACE1 is regulated by the N-terminal ankyrin (ANK) and the middle (MID) domains is ill defined. Here, we identified in the version 64 of the Catalogue of Somatic Mutations in Cancer (COSMIC) 13 missense mutations of hace1 located outside the HECT domain, and found that all lead to defective control of cell proliferation. In addition, several mutations located in the ankyrin domain displayed a dramatic reduction in Rac1 ubiquitylation associated with a decrease of colony formation in soft agar. 3D structure modelling of the 7 ankyrin-repeats coupled to functional analysis identified a surface epitope centered on one of the mutated residue, Gly-175, which is critical for controlling Rac1 binding and ubiquitylation. We also identified a role for the MID domain in conferring the specificity of association of HACE1 to the active form of Rac1. Our study of the functional interplay between HACE1 and Rac1 in cancer thus sheds a new light on the molecular mechanism of Rac1 ubiquitylation by HACE1 and the impact of its cancer-associated mutations in cell proliferation.


Asunto(s)
Mutación Missense/genética , Neoplasias/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteína de Unión al GTP rac1/metabolismo , Secuencia de Aminoácidos , Línea Celular , Proliferación Celular , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Unión Proteica , Dominios Proteicos , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo
9.
Mol Cell ; 62(1): 121-36, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26949039

RESUMEN

HECT-family E3 ligases ubiquitinate protein substrates to control virtually every eukaryotic process and are misregulated in numerous diseases. Nonetheless, understanding of HECT E3s is limited by a paucity of selective and potent modulators. To overcome this challenge, we systematically developed ubiquitin variants (UbVs) that inhibit or activate HECT E3s. Structural analysis of 6 HECT-UbV complexes revealed UbV inhibitors hijacking the E2-binding site and activators occupying a ubiquitin-binding exosite. Furthermore, UbVs unearthed distinct regulation mechanisms among NEDD4 subfamily HECTs and proved useful for modulating therapeutically relevant targets of HECT E3s in cells and intestinal organoids, and in a genetic screen that identified a role for NEDD4L in regulating cell migration. Our work demonstrates versatility of UbVs for modulating activity across an E3 family, defines mechanisms and provides a toolkit for probing functions of HECT E3s, and establishes a general strategy for systematic development of modulators targeting families of signaling proteins.


Asunto(s)
Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Animales , Dominio Catalítico , Línea Celular , Movimiento Celular , Perros , Células HCT116 , Humanos , Células de Riñón Canino Madin Darby , Modelos Moleculares , Organoides/citología , Organoides/metabolismo , Biblioteca de Péptidos , Ubiquitina/química , Ubiquitina/genética
10.
Toxins (Basel) ; 7(10): 4131-42, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26501320

RESUMEN

It is crucial to define risk factors that contribute to host invasion by Staphylococcus aureus. Here, we demonstrate that the chromosomally encoded EDIN-B isoform from S. aureus contributes to the onset of bacteremia during the course of pneumonia. Deletion of edinB in a European lineage community-acquired methicillin resistant S. aureus (CA-MRSA) strain (ST80-MRSA-IV) dramatically decreased the frequency and magnitude of bacteremia in mice suffering from pneumonia. This deletion had no effect on the bacterial burden in both blood circulation and lung tissues. Re-expression of wild-type EDIN-B, unlike the catalytically inactive mutant EDIN-R185E, restored the invasive characteristics of ST80-MRSA-IV.


Asunto(s)
Bacteriemia/microbiología , Proteínas Bacterianas/genética , Traslocación Bacteriana , Neumonía Bacteriana/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Animales , Traslocación Bacteriana/genética , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Staphylococcus aureus/aislamiento & purificación , Virulencia
11.
Cytoskeleton (Hoboken) ; 72(10): 542-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26403219

RESUMEN

It remains a challenge to decode the molecular basis of the long-term actin cytoskeleton rearrangements that are governed by the reprogramming of gene expression. Bacillus anthracis lethal toxin (LT) inhibits mitogen-activated protein kinase (MAPK) signaling, thereby modulating gene expression, with major consequences for actin cytoskeleton organization and the loss of endothelial barrier function. Using a laser ablation approach, we characterized the contractile and tensile mechanical properties of LT-induced stress fibers. These actin cables resist pulling forces that are transmitted at cell-matrix interfaces and at cell-cell discontinuous adherens junctions. We report that treating the cells with trichostatin A (TSA), a broad range inhibitor of histone deacetylases (HDACs), or with MS-275, which targets HDAC1, 2 and 3, induces stress fibers. LT decreased the cellular levels of HDAC1, 2 and 3 and reduced the global HDAC activity in the nucleus. Both the LT and TSA treatments induced Rnd3 expression, which is required for the LT-mediated induction of actin stress fibers. Furthermore, we reveal that treating the LT-intoxicated cells with garcinol, an inhibitor of histone acetyl-transferases (HATs), disrupts the stress fibers and limits the monolayer barrier dysfunctions. These data demonstrate the importance of modulating the flux of protein acetylation in order to control actin cytoskeleton organization and the endothelial cell monolayer barrier.


Asunto(s)
Actinas/química , Antígenos Bacterianos/química , Bacillus anthracis/química , Toxinas Bacterianas/química , Histonas/química , Fibras de Estrés/química , Acetilación , Uniones Adherentes , Comunicación Celular , Núcleo Celular/metabolismo , Células Endoteliales/citología , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ácidos Hidroxámicos/química , Luz , Microscopía Fluorescente , Resistencia a la Tracción
12.
Methods Mol Biol ; 827: 77-86, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22144268

RESUMEN

Rho GTPases including RhoA, Cdc42, and Rac1 are master regulators of cell cytoskeleton dynamic, thus controlling essential cellular processes notably cell polarity, migration and cytokinesis. These GTPases undergo a spatiotemporal regulation primarily controlled by cellular factors inducing both the exchange of GDP for GTP and the hydrolysis of GTP into GDP. Recent findings have unveiled another layer of complexity in the regulation of Rho proteins consisting in their ubiquitylation followed by their proteasomal degradation. Here, we describe how to assess the level of ubiquitylation of Rho proteins in cells, taking Rac1 as an example.


Asunto(s)
Ubiquitinación/fisiología , Proteínas de Unión al GTP rho/metabolismo , Animales , Western Blotting , Células CHO , Células Cultivadas , Cricetinae , Electroforesis en Gel de Poliacrilamida , Metales/metabolismo , Unión Proteica , Proteínas/aislamiento & purificación , Proteínas/metabolismo , Transfección , Proteínas de Unión al GTP rho/aislamiento & purificación
13.
Dev Cell ; 21(5): 959-65, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22036506

RESUMEN

Rac1 small GTPase controls essential aspects of cell biology and is a direct target of numerous bacterial virulence factors. The CNF1 toxin of pathogenic Escherichia coli addresses Rac1 to ubiquitin-proteasome system (UPS). We report the essential role of the tumor suppressor HACE1, a HECT-domain containing E3 ubiquitin-ligase, in the targeting of Rac1 to UPS. HACE1 binds preferentially GTP-bound Rac1 and catalyzes its polyubiquitylation. HACE1 expression increases the ubiquitylation of Rac1, when the GTPase is activated by point mutations or by the GEF-domain of Dbl. RNAi-mediated depletion of HACE1 blocks the ubiquitylation of active Rac1 and increases GTP-bound Rac1 cellular levels. HACE1 antagonizes cell isotropic spreading, a hallmark of Rac1 activation, and is required for endothelial cell monolayer invasion by bacteria. Together, these data establish the role of the HACE1 E3 ubiquitin-ligase in controlling Rac1 ubiquitylation and activity.


Asunto(s)
Biocatálisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteína de Unión al GTP rac1/metabolismo , Animales , Células CHO , Células Cultivadas , Cricetinae , Células HEK293 , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/biosíntesis
14.
Cell Host Microbe ; 10(5): 464-74, 2011 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-22100162

RESUMEN

RhoA-inhibitory bacterial toxins, such as Staphylococcus aureus EDIN toxin, induce large transendothelial cell macroaperture (TEM) tunnels that rupture the host endothelium barrier and promote bacterial dissemination. Host cells repair these tunnels by extending actin-rich membrane waves from the TEM edges. We reveal that cyclic-AMP signaling produced by Bacillus anthracis edema toxin (ET) also induces TEM formation, which correlates with increased vascular permeability. We show that ET-induced TEM formation resembles liquid dewetting, a physical process of nucleation and growth of holes within a thin liquid film. We also identify the cellular mechanisms of tunnel closure and reveal that the I-BAR domain protein Missing in Metastasis (MIM) senses de novo membrane curvature generated by the TEM, accumulates at the TEM edge, and triggers Arp2/3-dependent actin polymerization, which induces actin-rich membrane waves that close the TEM. Thus, the balance between ET-induced TEM formation and resealing likely determines the integrity of the host endothelium barrier.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/química , Carbunco/metabolismo , Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , AMP Cíclico/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Complejo 2-3 Proteico Relacionado con la Actina/genética , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Animales , Carbunco/microbiología , Bacillus anthracis/genética , Células Endoteliales de la Vena Umbilical Humana/microbiología , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/genética , Proteínas de Neoplasias/genética , Polimerizacion
15.
Traffic ; 12(5): 579-90, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21291504

RESUMEN

Rho GTPases, which are master regulators of both the actin cytoskeleton and membrane trafficking, are often hijacked by pathogens to enable their invasion of host cells. Here we report that the cytotoxic necrotizing factor-1 (CNF1) toxin of uropathogenic Escherichia coli (UPEC) promotes Rac1-dependent entry of bacteria into host cells. Our screen for proteins involved in Rac1-dependent UPEC entry identifies the Toll-interacting protein (Tollip) as a new interacting protein of Rac1 and its ubiquitinated forms. We show that knockdown of Tollip reduces CNF1-induced Rac1-dependent UPEC entry. Tollip depletion also reduces the Rac1-dependent entry of Listeria monocytogenes expressing InlB invasion protein. Moreover, knockdown of Tollip, Tom1 and clathrin, decreases CNF1 and Rac1-dependent internalization of UPEC. Finally, we show that Tollip, Tom1 and clathrin associate with Rac1 and localize at the site of bacterial entry. Collectively, these findings reveal a new link between Rac1 and Tollip, Tom1 and clathrin membrane trafficking components hijacked by pathogenic bacteria to allow their efficient invasion of host cells.


Asunto(s)
Infecciones Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Escherichia coli Uropatógena/metabolismo , Escherichia coli Uropatógena/patogenicidad , Proteína de Unión al GTP rac1/metabolismo , Animales , Línea Celular , Endocitosis/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Proteínas/metabolismo , Escherichia coli Uropatógena/citología , Proteína de Unión al GTP rac1/genética
16.
PLoS One ; 6(2): e14682, 2011 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-21339824

RESUMEN

Bacillus sphaericus strains that produce the binary toxin (Bin) are highly toxic to Culex and Anopheles mosquitoes, and have been used since the late 1980s as a biopesticide for the control of these vectors of infectious disease agents. The Bin toxin produced by these strains targets mosquito larval midgut epithelial cells where it binds to Cpm1 (Culex pipiens maltase 1) a digestive enzyme, and causes severe intracellular damage, including a dramatic cytoplasmic vacuolation. The intoxication of mammalian epithelial MDCK cells engineered to express Cpm1 mimics the cytopathologies observed in mosquito enterocytes following Bin ingestion: pore formation and vacuolation. In this study we demonstrate that Bin-induced vacuolisation is a transient phenomenon that affects autolysosomes. In addition, we show that this vacuolisation is associated with induction of autophagy in intoxicated cells. Furthermore, we report that after internalization, Bin reaches the recycling endosomes but is not localized either within the vacuolating autolysosomes or within any other degradative compartment. Our observations reveal that Bin elicits autophagy as the cell's response to intoxication while protecting itself from degradation through trafficking towards the recycling pathways.


Asunto(s)
Autofagia/efectos de los fármacos , Infecciones por Bacillaceae/patología , Toxinas Bacterianas/toxicidad , Animales , Anopheles/enzimología , Anopheles/genética , Infecciones por Bacillaceae/metabolismo , Bacillus/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacología , Células Cultivadas , Culex/enzimología , Culex/genética , Perros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/fisiología , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/patología , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo , Vacuolas/patología , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo
17.
J Cell Sci ; 123(Pt 14): 2491-501, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20592186

RESUMEN

Integrin receptors and their extracellular matrix ligands provide cues to cell proliferation, survival, differentiation and migration. Here, we show that alpha2beta1 integrin, when ligated to the basement membrane component laminin-1, triggers a proliferation arrest in primary endothelial cells. Indeed, in the presence of strong growth signals supplied by growth factors and fibronectin, alpha2beta1 engagement alters assembly of mature focal adhesions by alpha5beta1 and leads to impairment of downstream signaling and cell-cycle arrest in the G1 phase. Although the capacity of alpha5beta1 to signal for GTP loading of Rac is preserved, the joint engagement of alpha2beta1 interferes with membrane anchorage of Rac. Adapting the 'split-ubiquitin' sensor to screen for membrane-proximal alpha2 integrin partners, we identified the CD9 tetraspanin and further establish its requirement for destabilization of focal adhesions, control of Rac subcellular localization and growth arrest induced by alpha2beta1 integrin. Altogether, our data establish that alpha2beta1 integrin controls endothelial cell commitment towards quiescence by triggering a CD9-dependent dominant signaling.


Asunto(s)
Antígenos CD/metabolismo , Células Endoteliales/metabolismo , Integrina alfa2beta1/metabolismo , Laminina/farmacología , Glicoproteínas de Membrana/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Antígenos CD/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibronectinas/farmacología , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Humanos , Integrina alfa2beta1/agonistas , Integrina alfa5beta1/agonistas , Integrina alfa5beta1/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Tetraspanina 29
18.
PLoS One ; 5(1): e8925, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-20111723

RESUMEN

BACKGROUND: Salmonella pathogenesis engages host cells in two-way biochemical interactions: phagocytosis of bacteria by recruitment of cellular small GTP-binding proteins induced by the bacteria, and by triggering a pro-inflammatory response through activation of MAPKs and nuclear translocation of NF-kappaB. Worldwide interest in the use of functional foods containing probiotic bacteria for health promotion and disease prevention has increased significantly. Saccharomyces boulardii is a non-pathogenic yeast used as a probiotic in infectious diarrhea. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we reported that S. boulardii (Sb) protected mice from Salmonella enterica serovar Typhimurium (ST)-induced death and prevented bacterial translocation to the liver. At a molecular level, using T84 human colorectal cancer cells, we demonstrate that incubation with Sb before infection totally abolished Salmonella invasion. This correlates with a decrease of activation of Rac1. Sb preserved T84 barrier function and decreased ST-induced IL-8 synthesis. This anti-inflammatory effect was correlated with an inhibitory effect of Sb on ST-induced activation of the MAPKs ERK1/2, p38 and JNK as well as on activation of NF-kappaB. Electron and confocal microscopy experiments showed an adhesion of bacteria to yeast cells, which could represent one of the mechanisms by which Sb exerts its protective effects. CONCLUSIONS: Sb shows modulating effects on permeability, inflammation, and signal transduction pathway in T84 cells infected by ST and an in vivo protective effect against ST infection. The present results also demonstrate that Sb modifies invasive properties of Salmonella.


Asunto(s)
Saccharomyces/fisiología , Infecciones por Salmonella/prevención & control , Salmonella enterica/patogenicidad , Animales , Adhesión Bacteriana , Liofilización , Hígado/microbiología , Ratones , Microscopía Electrónica de Transmisión , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Probióticos , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/mortalidad , Transducción de Señal
19.
J Biol Chem ; 283(21): 14857-66, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18356164

RESUMEN

The proteins of the RAF family (A-RAF, B-RAF, and C-RAF) are serine/threonine kinases that play important roles in development, mature cell regulation, and cancer. Although it is widely held that their localization on membranes is an important aspect of their function, there are few data that address this aspect of their mode of action. Here, we report that each member of the RAF family exhibits a specific distribution at the level of cellular membranes and that C-RAF is the only isoform that directly targets mitochondria. We found that the RAF kinases exhibit intrinsic differences in terms of mitochondrial affinity and that C-RAF is the only isoform that binds this organelle efficiently. This affinity is conferred by the C-RAF amino-terminal domain and does not depend on the presence of RAS GTPases on the surface of mitochondria. Finally, we analyzed the consequences of C-RAF activation on mitochondria and observed that this event dramatically changes their morphology and their subcellular distribution. Our observations indicate that: (i) RAF kinases exhibit different localizations at the level of cellular membranes; (ii) C-RAF is the only isoform that directly binds mitochondria; and (iii) through its functional coupling with MEK, C-RAF regulates the shape and the cellular distribution of mitochondria.


Asunto(s)
Mitocondrias/enzimología , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Isoenzimas/metabolismo , Ratones , Ratones Noqueados , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-raf/deficiencia , Proteínas Proto-Oncogénicas c-raf/genética , Especificidad por Sustrato , Proteínas ras/metabolismo
20.
J Cell Biol ; 177(2): 343-54, 2007 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-17438076

RESUMEN

Glycosylphosphatidylinositol-anchored proteins (GPI-APs) are endocytosed by a clathrin- independent pathway into vesicles named GPI-AP-enriched early endosomal compartments (GEECs). We recently showed that the vacuolating toxin VacA secreted by Helicobacter pylori is endocytosed into the GEECs (Gauthier, N.C., P. Monzo, V. Kaddai, A. Doye, V. Ricci, and P. Boquet. 2005. Mol. Biol. Cell. 16:4852-4866). Unlike GPI-APs that are mostly recycled back to the plasma membrane, VacA reaches early endosomes (EEs) and then late endosomes (LEs), where vacuolation occurs. In this study, we used VacA to study the trafficking pathway between GEECs and LEs. We found that VacA routing from GEECs to LEs required polymerized actin. During this trafficking, VacA was transferred from GEECs to EEs associated with polymerized actin structures. The CD2-associated protein (CD2AP), a docking protein implicated in intracellular trafficking, bridged the filamentous actin (F-actin) structures with EEs containing VacA. CD2AP regulated those F-actin structures and was required to transfer VacA from GEECs to LEs. These results demonstrate that sorting from GEECs to LEs requires dynamic F-actin structures on EEs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Endosomas/metabolismo , Helicobacter pylori/química , Actinas/metabolismo , Citoesqueleto/metabolismo , Endocitosis , Glicosilfosfatidilinositoles/metabolismo , Células HeLa , Humanos , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...