Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Mol Cell ; 62(1): 121-36, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26949039

RESUMO

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.


Assuntos
Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Domínio Catalítico , Linhagem Celular , Movimento Celular , Cães , Células HCT116 , Humanos , Células Madin Darby de Rim Canino , Modelos Moleculares , Organoides/citologia , Organoides/metabolismo , Biblioteca de Peptídeos , Ubiquitina/química , Ubiquitina/genética
2.
Traffic ; 12(5): 579-90, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21291504

RESUMO

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.


Assuntos
Infecções Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Escherichia coli Uropatogênica/metabolismo , Escherichia coli Uropatogênica/patogenicidade , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Endocitose/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Proteínas/metabolismo , Escherichia coli Uropatogênica/citologia , Proteínas rac1 de Ligação ao GTP/genética
3.
J Cell Sci ; 123(Pt 14): 2491-501, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20592186

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Células Endoteliais/metabolismo , Integrina alfa2beta1/metabolismo , Laminina/farmacologia , Glicoproteínas de Membrana/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Antígenos CD/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibronectinas/farmacologia , Adesões Focais/genética , Adesões Focais/metabolismo , Humanos , Integrina alfa2beta1/agonistas , Integrina alfa5beta1/agonistas , Integrina alfa5beta1/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Tetraspanina 29
4.
J Cell Biol ; 177(2): 343-54, 2007 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-17438076

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas do Citoesqueleto/metabolismo , Endossomos/metabolismo , Helicobacter pylori/química , Actinas/metabolismo , Citoesqueleto/metabolismo , Endocitose , Glicosilfosfatidilinositóis/metabolismo , Células HeLa , Humanos , Transporte Proteico
6.
Nat Commun ; 13(1): 6059, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36229487

RESUMO

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.


Assuntos
Ciclina D1 , Integrinas , Divisão Celular , Ciclina D1/metabolismo , Integrinas/metabolismo , Mecanotransdução Celular/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Proteínas rac1 de Ligação ao GTP/metabolismo
7.
J Cell Biol ; 173(5): 809-19, 2006 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-16754962

RESUMO

The GTPase RhoA is a major regulator of the assembly of actin stress fibers and the contractility of the actomyosin cytoskeleton. The epidermal cell differentiation inhibitor (EDIN) and EDIN-like ADP-ribosyltransferases of Staphylococcus aureus catalyze the inactivation of RhoA, producing actin cable disruption. We report that purified recombinant EDIN and EDIN-producing S. aureus provoke large transcellular tunnels in endothelial cells that we have named macroapertures (MAs). These structures open transiently, followed by the appearance of actin-containing membrane waves extending over the aperture. Disruption of actin cables, either directly or indirectly, through rhoA RNAi knockdown also triggers the formation of MAs. Intoxication of endothelial monolayers by EDIN produces a loss of barrier function and provides direct access of the endothelium basement membrane to S. aureus.


Assuntos
ADP Ribose Transferases/farmacologia , Proteínas de Bactérias/farmacologia , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , ADP Ribose Transferases/isolamento & purificação , Proteínas de Bactérias/isolamento & purificação , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Interferência de RNA/fisiologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Staphylococcus aureus/química , Staphylococcus aureus/enzimologia , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
8.
Nat Microbiol ; 6(3): 401-412, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33432150

RESUMO

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.


Assuntos
Bacteriemia/metabolismo , Toxinas Bacterianas/metabolismo , Infecções por Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Bacteriemia/imunologia , Bacteriemia/microbiologia , Carga Bacteriana , Toxinas Bacterianas/genética , Escherichia coli/genética , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Imunidade Inata , Camundongos , Fosforilação , Transdução de Sinais , Quinases Ativadas por p21/metabolismo , Proteínas rac de Ligação ao GTP/genética , Proteína RAC2 de Ligação ao GTP
9.
Blood Adv ; 5(5): 1523-1534, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33683342

RESUMO

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.


Assuntos
COVID-19/sangue , Inflamassomos/metabolismo , Células Mieloides/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Biomarcadores/sangue , COVID-19/imunologia , Estudos de Casos e Controles , Humanos , Inflamassomos/sangue , Pessoa de Meia-Idade , Estudos Prospectivos , SARS-CoV-2/imunologia , SARS-CoV-2/isolamento & purificação
10.
Mol Biol Cell ; 17(6): 2489-97, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16540523

RESUMO

Ubiquitylation of RhoA has emerged as an important aspect of both the virulence of Escherichia coli producing cytotoxic necrotizing factor (CNF) 1 toxin and the establishment of the polarity of eukaryotic cells. Owing to the molecular activity of CNF1, we have investigated the relationship between permanent activation of RhoA catalyzed by CNF1 and subsequent ubiquitylation of RhoA by Smurf1. Using Smurf1-deficient cells and by RNA interference (RNAi)-mediated Smurf1 knockdown, we demonstrate that Smurf1 is a rate-limiting and specific factor of the ubiquitin-mediated proteasomal degradation of activated RhoA. We further show that the cancer cell lines HEp-2, human embryonic kidney 293 and Vero are specifically deficient in ubiquitylation of either activated Rac, Cdc42, or Rho, respectively. In contrast, CNF1 produced the cellular depletion of all three isoforms of Rho proteins in the primary human cell types we have tested. We demonstrate that ectopic expression of Smurf1 in Vero cells, deficient for RhoA ubiquitylation, restores ubiquitylation of the activated forms of RhoA. We conclude here that Smurf1 ubiquitylates activated RhoA and that, in contrast to human primary cell types, some cancer cell lines have a lower ubiquitylation capacity of specific Rho proteins. Thus, both CNF1 and transforming growth factor-beta trigger activated RhoA ubiquitylation through Smurf1 ubiquitin-ligase.


Assuntos
Toxinas Bacterianas/farmacologia , Proteínas de Escherichia coli/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Linhagem Celular , Humanos , Rim , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitina/efeitos dos fármacos , Ubiquitina-Proteína Ligases/genética , Proteína rhoA de Ligação ao GTP/efeitos dos fármacos
11.
Cancer Cell ; 36(3): 268-287.e10, 2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31447347

RESUMO

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.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Linfadenopatia Imunoblástica/patologia , Linfoma de Células T/patologia , NF-kappa B/metabolismo , Linfócitos T/imunologia , Idoso , Animais , Linhagem Celular Tumoral , Linhagem da Célula/imunologia , Conjuntos de Dados como Assunto , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/genética , Células HEK293 , Humanos , Linfadenopatia Imunoblástica/genética , Linfoma de Células T/tratamento farmacológico , Linfoma de Células T/genética , Linfoma de Células T/imunologia , Masculino , Camundongos Transgênicos , Pessoa de Meia-Idade , NF-kappa B/genética , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Quinase Induzida por NF-kappaB
12.
Mol Biol Cell ; 16(10): 4852-66, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16055501

RESUMO

The vacuolating cytotoxin VacA is a major virulence factor of Helicobacter pylori, a bacterium responsible for gastroduodenal ulcers and cancer. VacA associates with lipid rafts, is endocytosed, and reaches the late endocytic compartment where it induces vacuolation. We have investigated the endocytic and intracellular trafficking pathways used by VacA, in HeLa and gastric AGS cells. We report here that VacA was first bound to plasma-membrane domains localized above F-actin structures that were controlled by the Rac1 GTPase. VacA was subsequently pinocytosed by a clathrin-independent mechanism into cell peripheral early endocytic compartments lacking caveolin 1, the Rab5 effector early endosomes antigen-1 (EEA1) and transferrin. These compartments took up fluid-phase (as evidenced by the accumulation of fluorescent dextran) and glycosylphosphatidylinositol-anchored proteins (GPI-APs). VacA pinocytosis was controlled by Cdc42 and did not require cellular tyrosine kinases, dynamin 2, ADP-ribosylating factor 6, or RhoA GTPase activities. VacA was subsequently routed to EEA1-sorting endosomes and then sorted to late endosomes. During all these different endocytic steps, VacA was continuously associated with detergent resistant membrane domains. From these results we propose that VacA might be a valuable probe to study raft-associated molecules, pinocytosed by a clathrin-independent mechanism, and routed to the degradative compartment.


Assuntos
Proteínas de Bactérias/fisiologia , Toxinas Bacterianas/metabolismo , Clatrina/fisiologia , Endossomos/fisiologia , Helicobacter pylori/fisiologia , Pinocitose/fisiologia , Fatores de Virulência/fisiologia , Proteína cdc42 de Ligação ao GTP/fisiologia , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/metabolismo , Actinas/metabolismo , Caveolina 1/metabolismo , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Dinamina II/metabolismo , Humanos , Microdomínios da Membrana/fisiologia , Proteínas de Membrana/metabolismo , Transferrina/metabolismo , Proteínas de Transporte Vesicular , Proteínas rab5 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
13.
Sci Rep ; 8(1): 1410, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29362425

RESUMO

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.


Assuntos
Serina/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Quinases Ativadas por p21/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Toxinas Bacterianas/farmacologia , Linhagem Celular , Proteínas de Escherichia coli/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Fosforilação , Multimerização Proteica , Proteômica , Ubiquitinação , Fator A de Crescimento do Endotélio Vascular/farmacologia
14.
Sci Rep ; 7: 44779, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28317937

RESUMO

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.


Assuntos
Mutação de Sentido Incorreto/genética , Neoplasias/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Proteínas rac1 de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Proliferação de Células , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Ligação Proteica , Domínios Proteicos , Relação Estrutura-Atividade , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
15.
Nat Commun ; 8: 15839, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28643776

RESUMO

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.


Assuntos
Actomiosina/metabolismo , Proteínas do Citoesqueleto/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actomiosina/química , Actomiosina/genética , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Miosina não Muscular Tipo IIA/química , Miosina não Muscular Tipo IIA/genética , Tensão Superficial
16.
Methods Enzymol ; 406: 447-56, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16472677

RESUMO

The CNF1 toxin is produced by uropathogenic and meningitis-causing Escherichia coli. CNF1 catalyzes the constitutive activation of Rho proteins by deamidation. The threshold of activation of Rho proteins by CNF1 is, however, attenuated because of a concomitant decrease of their cellular levels. Depletion of activated-Rac1 is catalyzed by ubiquitin-mediated proteasomal degradation. Consequently, we show by effector-binding pull-down that co-treatment of intoxicated cells with the MG132 proteasome-inhibitor results in a higher level of activation of Rac, as well as RhoA and Cdc42. We show that CNF1 induces the transient recruitment of Rho proteins to cellular membranes. Interestingly, at the difference of Rac and Cdc42, the inhibition of the proteasome during CNF1 treatment does not result in a significant accumulation of RhoA to cellular membranes. Using an in vivo ubiquitylation assay, we evidence that mutation of the geranylgeranyl acceptor cysteine of Rac1 (Rac1C189G) abolished the sensitivity of permanently activated-Rac1 to ubiquitylation, whereas Rac1C189G remained able to bind to the effector-binding domain of p21-PAK. Collectively, these results indicate that association with the cellular membranes is a necessary step for activated-Rac1 ubiquitylation.


Assuntos
Toxinas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Toxinas Bacterianas/isolamento & purificação , Membrana Celular/metabolismo , Células Cultivadas , Endotélio Vascular , Escherichia coli/metabolismo , Proteínas de Escherichia coli/isolamento & purificação , Humanos , Complexos Ubiquitina-Proteína Ligase/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo
17.
Toxins (Basel) ; 7(10): 4131-42, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26501320

RESUMO

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.


Assuntos
Bacteriemia/microbiologia , Proteínas de Bactérias/genética , Translocação Bacteriana , Pneumonia Bacteriana/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Animais , Translocação Bacteriana/genética , Modelos Animais de Doenças , Feminino , Deleção de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Staphylococcus aureus/isolamento & purificação , Virulência
18.
Cytoskeleton (Hoboken) ; 72(10): 542-56, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26403219

RESUMO

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.


Assuntos
Actinas/química , Antígenos de Bactérias/química , Bacillus anthracis/química , Toxinas Bacterianas/química , Histonas/química , Fibras de Estresse/química , Acetilação , Junções Aderentes , Comunicação Celular , Núcleo Celular/metabolismo , Células Endoteliais/citologia , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Ácidos Hidroxâmicos/química , Luz , Microscopia de Fluorescência , Resistência à Tração
19.
Methods Mol Biol ; 827: 77-86, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22144268

RESUMO

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.


Assuntos
Ubiquitinação/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Western Blotting , Células CHO , Células Cultivadas , Cricetinae , Eletroforese em Gel de Poliacrilamida , Metais/metabolismo , Ligação Proteica , Proteínas/isolamento & purificação , Proteínas/metabolismo , Transfecção , Proteínas rho de Ligação ao GTP/isolamento & purificação
20.
PLoS One ; 6(2): e14682, 2011 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-21339824

RESUMO

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.


Assuntos
Autofagia/efeitos dos fármacos , Infecções por Bacillaceae/patologia , Toxinas Bacterianas/toxicidade , Animais , Anopheles/enzimologia , Anopheles/genética , Infecções por Bacillaceae/metabolismo , Bacillus/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Células Cultivadas , Culex/enzimologia , Culex/genética , Cães , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/fisiologia , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/patologia , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Vacúolos/efeitos dos fármacos , Vacúolos/metabolismo , Vacúolos/patologia , alfa-Glucosidases/genética , alfa-Glucosidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa