Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Plant J ; 107(6): 1771-1787, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34250673

RESUMO

Upon immune activation, chloroplasts switch off photosynthesis, produce antimicrobial compounds and associate with the nucleus through tubular extensions called stromules. Although it is well established that chloroplasts alter their position in response to light, little is known about the dynamics of chloroplast movement in response to pathogen attack. Here, we report that during infection with the Irish potato famine pathogen Phytophthora infestans, chloroplasts accumulate at the pathogen interface, associating with the specialized membrane that engulfs the pathogen haustorium. The chemical inhibition of actin polymerization reduces the accumulation of chloroplasts at pathogen haustoria, suggesting that this process is partially dependent on the actin cytoskeleton. However, chloroplast accumulation at haustoria does not necessarily rely on movement of the nucleus to this interface and is not affected by light conditions. Stromules are typically induced during infection, embracing haustoria and facilitating chloroplast interactions, to form dynamic organelle clusters. We found that infection-triggered stromule formation relies on BRASSINOSTEROID INSENSITIVE 1-ASSOCIATED KINASE 1 (BAK1)-mediated surface immune signaling, whereas chloroplast repositioning towards haustoria does not. Consistent with the defense-related induction of stromules, effector-mediated suppression of BAK1-mediated immune signaling reduced stromule formation during infection. On the other hand, immune recognition of the same effector stimulated stromules, presumably via a different pathway. These findings implicate chloroplasts in a polarized response upon pathogen attack and point to more complex functions of these organelles in plant-pathogen interactions.


Assuntos
Cloroplastos/microbiologia , Interações Hospedeiro-Patógeno/fisiologia , Nicotiana/microbiologia , Phytophthora infestans/patogenicidade , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Cloroplastos/efeitos dos fármacos , Cloroplastos/imunologia , Dinitrobenzenos/farmacologia , Luz , Microscopia Confocal , Pinças Ópticas , Doenças das Plantas/microbiologia , Imunidade Vegetal , Folhas de Planta/efeitos dos fármacos , Folhas de Planta/microbiologia , Plantas Geneticamente Modificadas , Espécies Reativas de Oxigênio/metabolismo , Sulfanilamidas/farmacologia , Tiazolidinas/farmacologia , Nicotiana/efeitos dos fármacos , Nicotiana/genética , Nicotiana/imunologia
2.
Am J Physiol Gastrointest Liver Physiol ; 318(5): G870-G888, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32223302

RESUMO

Clostridioides difficile is an important nosocomial pathogen that produces toxins to cause life-threatening diarrhea and colitis. Toxins bind to epithelial receptors and promote the collapse of the actin cytoskeleton. C. difficile toxin activity is commonly studied in cancer-derived and immortalized cell lines. However, the biological relevance of these models is limited. Moreover, no model is available for examining C. difficile-induced enteritis, an understudied health problem. We hypothesized that human intestinal enteroids (HIEs) express toxin receptors and provide a new model to dissect C. difficile cytotoxicity in the small intestine. We generated biopsy-derived jejunal HIE and Vero cells, which stably express LifeAct-Ruby, a fluorescent label of F-actin, to monitor actin cytoskeleton rearrangement by live-cell microscopy. Imaging analysis revealed that toxins from pathogenic C. difficile strains elicited cell rounding in a strain-dependent manner, and HIEs were tenfold more sensitive to toxin A (TcdA) than toxin B (TcdB). By quantitative PCR, we paradoxically found that HIEs expressed greater quantities of toxin receptor mRNA and yet exhibited decreased sensitivity to toxins when compared with traditionally used cell lines. We reasoned that these differences may be explained by components, such as mucins, that are present in HIEs cultures, that are absent in immortalized cell lines. Addition of human-derived mucin 2 (MUC2) to Vero cells delayed cell rounding, indicating that mucus serves as a barrier to toxin-receptor binding. This work highlights that investigation of C. difficile infection in that HIEs can provide important insights into the intricate interactions between toxins and the human intestinal epithelium.NEW & NOTEWORTHY In this article, we developed a novel model of Clostridioides difficile-induced enteritis using jejunal-derived human intestinal enteroids (HIEs) transduced with fluorescently tagged F-actin. Using live-imaging, we identified that jejunal HIEs express high levels of TcdA and CDT receptors, are more sensitive to TcdA than TcdB, and secrete mucus, which delays toxin-epithelial interactions. This work also optimizes optically clear C. difficile-conditioned media suitable for live-cell imaging.


Assuntos
Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Enterite/microbiologia , Jejuno/microbiologia , ADP Ribose Transferases/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiologia , Citoesqueleto de Actina/ultraestrutura , Animais , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Forma Celular , Chlorocebus aethiops , Clostridioides difficile/metabolismo , Infecções por Clostridium/metabolismo , Infecções por Clostridium/patologia , Enterite/metabolismo , Enterite/patologia , Enterotoxinas/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Jejuno/metabolismo , Jejuno/ultraestrutura , Mucina-2/metabolismo , Organoides , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Fatores de Tempo , Células Vero , Virulência
3.
Cell Microbiol ; 21(6): e13014, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30702192

RESUMO

The major virulence determinant of Legionella pneumophila is the type IVB secretion system (T4BSS), which delivers approximately 330 effector proteins into the host cell to modulate various cellular processes. However, the functions of most effector proteins remain unclear. WipA, an effector, was the first phosphotyrosine phosphatase of Legionella with unknown function. In this study, we found that WipA induced relatively strong growth defects in yeast in a phosphatase activity-dependent manner. Phosphoproteomics data showed that WipA was likely involved into endocytosis, FcγR-mediated phagocytosis, tight junction, and regulation of actin cytoskeleton pathways. Western blotting further confirmed WipA dephosphorylates several proteins associated with actin polymerisation, such as p-N-WASP, p-ARP3, p-ACK1, and p-NCK1. Thus, we hypothesised that WipA targets N-WASP/ARP2/3 complex signalling pathway, leading to disturbance of actin polymerisation. Indeed, we demonstrated that WipA inhibits host F-actin polymerisation by reducing the G-actin to F-actin transition during L. penumophila infection. Furthermore, the intracellular proliferation of wipA/legK2 double mutant was significantly impaired at the late stage of infection, although the absence of WipA does not confer any further effect on actin polymerisation to the legK2 mutant. Collectively, this study provides unique insights into the WipA-mediated regulation of host actin polymerisation and assists us to elucidate the pathogenic mechanisms of L. pnuemophila infection.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Legionella pneumophila/enzimologia , Macrófagos/metabolismo , Fosfotirosina/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Fatores de Virulência/metabolismo , Citoesqueleto de Actina/microbiologia , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/química , Animais , Cromatografia Líquida , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidade , Doença dos Legionários/metabolismo , Macrófagos/microbiologia , Camundongos , Fagocitose/genética , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/toxicidade , Proteômica , Saccharomyces cerevisiae/crescimento & desenvolvimento , Saccharomyces cerevisiae/metabolismo , Transdução de Sinais/genética , Espectrometria de Massas em Tandem , Junções Íntimas/metabolismo
4.
Cell Microbiol ; 21(8): e13033, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31009148

RESUMO

Vibrio cholerae produced-Cholix toxin (Cholix) is a cytotoxin that ADP-ribosylates eukaryotic elongation factor 2, inhibiting protein synthesis, and inducing apoptosis. Here, we identified prohibitin (PHB) 1 and 2 as novel Cholix-interacting membrane proteins in immortalised human hepatocytes and HepG2 cells by Cholix immunoprecipitation assays. The expression level of PHB1 was decreased by Cholix after a 12hr incubation. Cholix-induced poly (ADP-ribose) polymerase (PARP) cleavage was significantly enhanced in PHB (PHB1 or PHB2) knockdown cells. In contrast, transiently overexpressed PHB in hepatocytes attenuated Cholix-induced Bax/Bak conformational changes and PARP cleavage. In addition, Cholix-induced reactive oxygen species production and accumulation of fragmented mitochondria were enhanced in PHB-knockdown cells. Furthermore, Cholix induced activation of Rho-associated coiled coil-containing protein kinase 1 (ROCK1), which was enhanced in PHB-knockdown cells, followed by actin filament depolymerisation and accumulation of tubulin in the blebbing cells. Inhibition of ROCK1 by siRNA or its inhibitor suppressed Cholix-induced PARP cleavage and reactive oxygen species generation. Our findings identify PHB as a new protein that interacts with Cholix and is involved in Cholix-induced mitochondrial dysfunction and cytoskeletal rearrangement by ROCK1 activation during apoptosis.


Assuntos
Fatores de Ribosilação do ADP/química , Toxinas Bacterianas/química , Fator de Iniciação 2 em Eucariotos/metabolismo , Interações Hospedeiro-Patógeno/genética , Espécies Reativas de Oxigênio/metabolismo , Proteínas Repressoras/genética , Vibrio cholerae/genética , ADP-Ribosilação , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiologia , Citoesqueleto de Actina/ultraestrutura , Sequência de Aminoácidos , Apoptose/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Linhagem Celular Transformada , Fator de Iniciação 2 em Eucariotos/genética , Regulação da Expressão Gênica , Células Hep G2 , Hepatócitos/metabolismo , Hepatócitos/microbiologia , Hepatócitos/patologia , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/microbiologia , Mitocôndrias/ultraestrutura , Proibitinas , Ligação Proteica , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Proteólise , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/deficiência , Transdução de Sinais , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Vibrio cholerae/metabolismo , Vibrio cholerae/patogenicidade , Virulência , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
5.
EMBO Rep ; 19(1): 29-42, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29141986

RESUMO

The interaction of Mycobacterium tuberculosis (Mtb) with pulmonary epithelial cells is critical for early stages of bacillus colonization and during the progression of tuberculosis. Entry of Mtb into epithelial cells has been shown to depend on F-actin polymerization, though the molecular mechanisms are still unclear. Here, we demonstrate that mycobacterial uptake into epithelial cells requires rearrangements of the actin cytoskeleton, which are regulated by ADP-ribosylation factor 1 (Arf1) and phospholipase D1 (PLD1), and is dependent on the M3 muscarinic receptor (M3R). We show that this pathway is controlled by Arf GTPase-activating protein 1 (ArfGAP1), as its silencing has an impact on actin cytoskeleton reorganization leading to uncontrolled uptake and replication of Mtb. Furthermore, we provide evidence that this pathway is critical for mycobacterial entry, while the cellular infection with other pathogens, such as Shigella flexneri and Yersinia pseudotuberculosis, is not affected. Altogether, these results reveal how cortical actin plays the role of a barrier to prevent mycobacterial entry into epithelial cells and indicate a novel role for ArfGAP1 as a restriction factor of host-pathogen interactions.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/genética , Proteínas Ativadoras de GTPase/genética , Interações Hospedeiro-Patógeno , Mycobacterium tuberculosis/patogenicidade , Alvéolos Pulmonares/metabolismo , Células A549 , Fator 1 de Ribosilação do ADP/genética , Fator 1 de Ribosilação do ADP/metabolismo , Citoesqueleto de Actina/microbiologia , Citoesqueleto de Actina/ultraestrutura , Actinas/metabolismo , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica , Humanos , Mycobacterium tuberculosis/fisiologia , Fosfolipase D/genética , Fosfolipase D/metabolismo , Polimerização , Alvéolos Pulmonares/microbiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Shigella flexneri/fisiologia , Transdução de Sinais , Especificidade da Espécie , Yersinia pseudotuberculosis/fisiologia
6.
Cell Microbiol ; 18(5): 705-19, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26537021

RESUMO

Salmonella are able to invade non-phagocytic cells such as intestinal epithelial cells by modulating the host actin cytoskeleton to produce membrane ruffles. Two type III effector proteins SopB and SopE play key roles to this modulation. SopE is a known guanine nucleotide exchange factor (GEF) capable of activating Rac1 and CDC42. SopB is a phosphatidylinositol 4-phosphatase and 5-phosphatase promoting membrane ruffles and invasion of Salmonella through undefined mechanisms. Previous studies have demonstrated that the 4-phosphatase activity of SopB is required for PtdIns-3-phosphate (PtdIns(3)P) accumulation and SopB-mediated invasion. We show here that both the 4-phosphatase as well as the 5-phosphatase activities of SopB are essential in ruffle formation and subsequent invasion. We found that the 5-phosphatase activity of SopB is likely responsible for generating PtdIns-3,4-bisphosphate (PtdIns(3,4)P(2)) and subsequent recruitment of sorting nexin 9 (SNX9), an actin modulating protein. Intriguingly, the 4-phosphatase activity is responsible for the dephosphorylation of PtdIns(3,4)P(2) into PtdIns(3)P. Alone, neither activity is sufficient for ruffling but when acting in conjunction with one another, the 4-phosphatase and 5-phosphatase activities led to SNX9-mediated ruffling and Salmonella invasion. This work reveals the unique ability of bacterial effector protein SopB to utilize both its 4- and 5-phosphatase activities to regulate phosphoinositide dynamics to promote bacterial entry.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas de Bactérias/metabolismo , Membrana Celular/metabolismo , Fosfatidilinositóis/metabolismo , Citoesqueleto de Actina/microbiologia , Actinas/metabolismo , Animais , Proteínas de Bactérias/genética , Células COS , Membrana Celular/microbiologia , Extensões da Superfície Celular , Chlorocebus aethiops , Células HeLa , Humanos , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade
7.
Infect Immun ; 84(6): 1826-1841, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27068087

RESUMO

Many bacterial pathogens subvert mammalian type IA phosphoinositide 3-kinase (PI3K) in order to induce their internalization into host cells. How PI3K promotes internalization is not well understood. Also unclear is whether type IA PI3K affects different pathogens through similar or distinct mechanisms. Here, we performed an RNA interference (RNAi)-based screen to identify components of the type IA PI3K pathway involved in invasin-mediated entry of Yersinia enterocolitica, an enteropathogen that causes enteritis and lymphadenitis. The 69 genes targeted encode known upstream regulators or downstream effectors of PI3K. A similar RNAi screen was previously performed with the food-borne bacterium Listeria monocytogenes The results of the screen with Y. enterocolitica indicate that at least nine members of the PI3K pathway are needed for invasin-mediated entry. Several of these proteins, including centaurin-α1, Dock180, focal adhesion kinase (FAK), Grp1, LL5α, LL5ß, and PLD2 (phospholipase D2), were recruited to sites of entry. In addition, centaurin-α1, FAK, PLD2, and mTOR were required for remodeling of the actin cytoskeleton during entry. Six of the human proteins affecting invasin-dependent internalization also promote InlB-mediated entry of L. monocytogenes Our results identify several host proteins that mediate invasin-induced effects on the actin cytoskeleton and indicate that a subset of PI3K pathway components promote internalization of both Y. enterocolitica and L. monocytogenes.


Assuntos
Citoesqueleto de Actina/metabolismo , Adesinas Bacterianas/genética , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Interações Hospedeiro-Patógeno , Listeria monocytogenes/genética , Yersinia enterocolitica/genética , Citoesqueleto de Actina/microbiologia , Citoesqueleto de Actina/ultraestrutura , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adesinas Bacterianas/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosfolipase D/genética , Fosfolipase D/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Yersinia enterocolitica/crescimento & desenvolvimento , Yersinia enterocolitica/metabolismo , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo
8.
J Cell Sci ; 127(Pt 1): 240-9, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24155331

RESUMO

Several bacterial pathogens hijack the actin assembly machinery and display intracellular motility in the cytosol of infected cells. At the cell cortex, intracellular motility leads to bacterial dissemination through formation of plasma membrane protrusions that resolve into vacuoles in adjacent cells. Here, we uncover a crucial role for actin network disassembly in dissemination of Listeria monocytogenes. We found that defects in the disassembly machinery decreased the rate of actin tail turnover but did not affect the velocity of the bacteria in the cytosol. By contrast, defects in the disassembly machinery had a dramatic impact on bacterial dissemination. Our results suggest a model of L. monocytogenes dissemination in which the disassembly machinery, through local recycling of the actin network in protrusions, fuels continuous actin assembly at the bacterial pole and concurrently exhausts cytoskeleton components from the network distal to the bacterium, which enables membrane apposition and resolution of protrusions into vacuoles.


Assuntos
Citoesqueleto de Actina/genética , Actinas/genética , Listeria monocytogenes/fisiologia , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiologia , Actinas/antagonistas & inibidores , Actinas/metabolismo , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Cofilina 1/genética , Cofilina 1/metabolismo , Citosol/metabolismo , Citosol/microbiologia , Regulação da Expressão Gênica , Fator de Maturação da Glia/genética , Fator de Maturação da Glia/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Microscopia Confocal , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Vacúolos/metabolismo , Vacúolos/microbiologia
9.
PLoS Pathog ; 9(4): e1003290, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593000

RESUMO

Plants are constantly exposed to a large and diverse array of microbes; however, most plants are immune to the majority of potential invaders and susceptible to only a small subset of pathogens. The cytoskeleton comprises a dynamic intracellular framework that responds rapidly to biotic stresses and supports numerous fundamental cellular processes including vesicle trafficking, endocytosis and the spatial distribution of organelles and protein complexes. For years, the actin cytoskeleton has been assumed to play a role in plant innate immunity against fungi and oomycetes, based largely on static images and pharmacological studies. To date, however, there is little evidence that the host-cell actin cytoskeleton participates in responses to phytopathogenic bacteria. Here, we quantified the spatiotemporal changes in host-cell cytoskeletal architecture during the immune response to pathogenic and non-pathogenic strains of Pseudomonas syringae pv. tomato DC3000. Two distinct changes to host cytoskeletal arrays were observed that correspond to distinct phases of plant-bacterial interactions i.e. the perception of microbe-associated molecular patterns (MAMPs) during pattern-triggered immunity (PTI) and perturbations by effector proteins during effector-triggered susceptibility (ETS). We demonstrate that an immediate increase in actin filament abundance is a conserved and novel component of PTI. Notably, treatment of leaves with a MAMP peptide mimic was sufficient to elicit a rapid change in actin organization in epidermal cells, and this actin response required the host-cell MAMP receptor kinase complex, including FLS2, BAK1 and BIK1. Finally, we found that actin polymerization is necessary for the increase in actin filament density and that blocking this increase with the actin-disrupting drug latrunculin B leads to enhanced susceptibility of host plants to pathogenic and non-pathogenic bacteria.


Assuntos
Citoesqueleto de Actina , Arabidopsis/imunologia , Arabidopsis/metabolismo , Pseudomonas syringae/imunologia , Receptores de Reconhecimento de Padrão , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiologia , Arabidopsis/microbiologia , Proteínas de Arabidopsis/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Imunidade Inata , Doenças das Plantas/imunologia , Doenças das Plantas/microbiologia , Folhas de Planta/imunologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Pseudomonas syringae/metabolismo , Pseudomonas syringae/patogenicidade , Transdução de Sinais , Tiazolidinas/farmacologia , Fatores de Virulência/metabolismo
10.
J Biol Chem ; 287(16): 13128-36, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22351770

RESUMO

Listeria monocytogenes is a bacterial pathogen that induces its own entry into a broad range of mammalian cells through interaction of the bacterial surface protein InlB with the cellular receptor Met, promoting an actin polymerization/depolymerization process that leads to pathogen engulfment. Phosphatidylinositol bisphosphate (PI[4,5]P(2)) and trisphosphate (PI[3,4,5]P(3)) are two major phosphoinositide species that function as molecular scaffolds, recruiting cellular effectors that regulate actin dynamics during L. monocytogenes infection. Because the phosphatidylinositol 5'-phosphatase OCRL dephosphorylates PI(4,5)P(2) and to a lesser extent PI(3,4,5)P(3), we investigated whether this phosphatase modulates cell invasion by L. monocytogenes. Inactivation of OCRL by small interfering RNA (siRNA) leads to an increase in the internalization levels of L. monocytogenes in HeLa cells. Interestingly, OCRL depletion does not increase but rather decreases the surface expression of the receptor Met, suggesting that OCRL controls bacterial internalization by modulating signaling cascades downstream of Met. Immuno-fluorescence microscopy reveals that endogenous and overexpressed OCRL are present at L. monocytogenes invasion foci; live-cell imaging additionally shows that actin depolymerization coincides with EGFP-OCRL-a accumulation around invading bacteria. Together, these observations suggest that OCRL promotes actin depolymerization during L. monocytogenes infection; in agreement with this hypothesis, OCRL depletion leads to an increase in actin, PI(4,5)P(2), and PI(3,4,5)P(3) levels at bacterial internalization foci. Furthermore, in cells knocked down for OCRL, transfection of enzymatically active EGFP-OCRL-a (but not of a phosphatase-dead enzyme) decreases the levels of intracellular L. monocytogenes and of actin associated with invading bacteria. These results demonstrate that through its phosphatase activity, OCRL restricts L. monocytogenes invasion by modulating actin dynamics at bacterial internalization sites.


Assuntos
Actinas/metabolismo , Listeria monocytogenes/metabolismo , Listeriose/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiologia , Proteínas de Bactérias/metabolismo , Células HeLa , Humanos , Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Proteínas de Membrana/metabolismo , Fagocitose/fisiologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolases/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , RNA Interferente Pequeno/genética , Virulência
11.
BMC Cell Biol ; 14: 2, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23294665

RESUMO

BACKGROUND: Disease manifestations of Staphylococcus aureus are connected to the fibronectin (Fn)-binding capacity of these Gram-positive pathogens. Fn deposition on the surface of S. aureus allows engagement of α5ß1 integrins and triggers uptake by host cells. For several integrin- and actin-associated cytoplasmic proteins, including FAK, Src, N-WASP, tensin and cortactin, a functional role during bacterial invasion has been demonstrated. As reorganization of the actin cytoskeleton is critical for bacterial entry, we investigated whether vinculin, an essential protein linking integrins with the actin cytoskeleton, may contribute to the integrin-mediated internalization of S. aureus. RESULTS: Complementation of vinculin in vinculin -/- cells, vinculin overexpression, as well as shRNA-mediated vinculin knock-down in different eukaryotic cell types demonstrate, that vinculin does not have a functional role during the integrin-mediated uptake of S. aureus. CONCLUSIONS: Our results suggest that vinculin is insignificant for the integrin-mediated uptake of S. aureus despite the critical role of vinculin as a linker between integrins and F-actin.


Assuntos
Receptores de Vitronectina/metabolismo , Staphylococcus aureus/fisiologia , Vinculina/metabolismo , Citoesqueleto de Actina/microbiologia , Animais , Aderência Bacteriana/fisiologia , Linhagem Celular , Fibronectinas/metabolismo , Células HEK293 , Humanos , Camundongos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Vitronectina/genética , Vinculina/antagonistas & inibidores , Vinculina/genética
12.
PLoS Pathog ; 7(9): e1002227, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21949650

RESUMO

The intestine is a common site for invasion by intracellular pathogens, but little is known about how pathogens restructure and exit intestinal cells in vivo. The natural microsporidian parasite N. parisii invades intestinal cells of the nematode C. elegans, progresses through its life cycle, and then exits cells in a transmissible spore form. Here we show that N. parisii causes rearrangements of host actin inside intestinal cells as part of a novel parasite exit strategy. First, we show that N. parisii infection causes ectopic localization of the normally apical-restricted actin to the basolateral side of intestinal cells, where it often forms network-like structures. Soon after this actin relocalization, we find that gaps appear in the terminal web, a conserved cytoskeletal structure that could present a barrier to exit. Reducing actin expression creates terminal web gaps in the absence of infection, suggesting that infection-induced actin relocalization triggers gap formation. We show that terminal web gaps form at a distinct stage of infection, precisely timed to precede spore exit, and that all contagious animals exhibit gaps. Interestingly, we find that while perturbations in actin can create these gaps, actin is not required for infection progression or spore formation, but actin is required for spore exit. Finally, we show that despite large numbers of spores exiting intestinal cells, this exit does not cause cell lysis. These results provide insight into parasite manipulation of the host cytoskeleton and non-lytic escape from intestinal cells in vivo.


Assuntos
Citoesqueleto de Actina/metabolismo , Caenorhabditis elegans/microbiologia , Microsporídios/patogenicidade , Esporos Fúngicos/metabolismo , Citoesqueleto de Actina/microbiologia , Animais , Caenorhabditis elegans/citologia , Interações Hospedeiro-Patógeno , Intestinos/citologia , Intestinos/parasitologia , Proteínas dos Microfilamentos/metabolismo , Microsporídios/metabolismo , Interferência de RNA , RNA Interferente Pequeno
13.
Subcell Biochem ; 59: 363-88, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22374097

RESUMO

Phosphoinositides are considered as highly dynamic players in the spatiotemporal organization of key signaling pathways, actin cytoskeleton rearrangements, establishment of cell polarity and intracellular vesicle trafficking. Their metabolism is accurately controlled and mutations in several phosphoinositide metabolizing enzymes take part in the development of human pathologies. Interestingly, evidence is accumulating that modulation of the phosphoinositide metabolism is critical for pathogenicity and virulence of many human pathogens. Given the importance of phosphoinositides, which link membrane and cytoskeleton dynamics to cell responses, it is not surprising that many invasive pathogens hijack their metabolism as part of their strategies to establish infection. In fact, according to their lifestyle, cellular pathogens use the phosphoinositide metabolism in order to trigger their uptake in nonphagocytic cells and/or modulate the maturation of the pathogen-containing vacuole to establish their replicative niche or escape in the cytosol and promote host cell survival. The last two decades have been marked by the discovery of different tactics used by cellular pathogens to modulate the phosphoinositide metabolism as part of their strategies to survive, proliferate and disseminate in a hostile environment.


Assuntos
Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Células Eucarióticas/microbiologia , Interações Hospedeiro-Patógeno , Fosfatidilinositóis/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiologia , Bactérias/crescimento & desenvolvimento , Bactérias/patogenicidade , Polaridade Celular , Células Eucarióticas/metabolismo , Células Eucarióticas/patologia , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Transporte Proteico , Proteínas Proto-Oncogênicas c-met/metabolismo , Vesículas Transportadoras/metabolismo
14.
Pathog Dis ; 79(4)2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33705517

RESUMO

Bacterial infection is a highly complex biological process involving a dynamic interaction between the invading microorganism and the host. Specifically, intracellular pathogens seize control over the host cellular processes including membrane dynamics, actin cytoskeleton, phosphoinositide metabolism, intracellular trafficking and immune defense mechanisms to promote their host colonization. To accomplish such challenging tasks, virulent bacteria deploy unique species-specific secreted effectors to evade and/or subvert cellular defense surveillance mechanisms to establish a replication niche. However, despite superficially similar infection strategies, diverse Rickettsia species utilize different effector repertoires to promote host colonization. This review will discuss our current understandings on how different Rickettsia species deploy their effector arsenal to manipulate host cellular processes to promote their intracytosolic life within the mammalian host.


Assuntos
Vetores Artrópodes/microbiologia , Interações Hospedeiro-Patógeno , Infecções por Rickettsia/microbiologia , Rickettsia/classificação , Rickettsia/patogenicidade , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiologia , Animais , Especificidade de Hospedeiro , Humanos , Redes e Vias Metabólicas , Ácaros/microbiologia , Fosfatidilinositóis/metabolismo , Ftirápteros/microbiologia , Filogenia , Rickettsia/crescimento & desenvolvimento , Rickettsia/metabolismo , Infecções por Rickettsia/genética , Infecções por Rickettsia/patologia , Sifonápteros/microbiologia , Especificidade da Espécie , Carrapatos/microbiologia
15.
J Cell Biol ; 220(9)2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34180943

RESUMO

Phagocytes engulf unwanted particles into phagosomes that then fuse with lysosomes to degrade the enclosed particles. Ultimately, phagosomes must be recycled to help recover membrane resources that were consumed during phagocytosis and phagosome maturation, a process referred to as "phagosome resolution." Little is known about phagosome resolution, which may proceed through exocytosis or membrane fission. Here, we show that bacteria-containing phagolysosomes in macrophages undergo fragmentation through vesicle budding, tubulation, and constriction. Phagosome fragmentation requires cargo degradation, the actin and microtubule cytoskeletons, and clathrin. We provide evidence that lysosome reformation occurs during phagosome resolution since the majority of phagosome-derived vesicles displayed lysosomal properties. Importantly, we show that clathrin-dependent phagosome resolution is important to maintain the degradative capacity of macrophages challenged with two waves of phagocytosis. Overall, our work suggests that phagosome resolution contributes to lysosome recovery and to maintaining the degradative power of macrophages to handle multiple waves of phagocytosis.


Assuntos
Citoesqueleto de Actina/metabolismo , Lisossomos/metabolismo , Microtúbulos/metabolismo , Fagocitose/fisiologia , Fagossomos/metabolismo , Citoesqueleto de Actina/microbiologia , Citoesqueleto de Actina/ultraestrutura , Actinas/genética , Actinas/metabolismo , Animais , Clatrina/genética , Clatrina/metabolismo , Escherichia coli/química , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Lisossomos/microbiologia , Lisossomos/ultraestrutura , Fusão de Membrana , Camundongos , Microtúbulos/microbiologia , Microtúbulos/ultraestrutura , Fagossomos/microbiologia , Fagossomos/ultraestrutura , Proteólise , Células RAW 264.7
16.
mBio ; 12(6): e0293921, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34781738

RESUMO

Direct cell-to-cell spreading of Listeria monocytogenes requires the bacteria to induce actin-based finger-like membrane protrusions in donor host cells that are endocytosed through caveolin-rich membrane invaginations by adjacent receiving cells. An actin shell surrounds these endocytic sites; however, its structure, composition, and functional significance remain elusive. Here, we show that the formin mDia1, but surprisingly not the Arp2/3 complex, is enriched at the membrane invaginations generated by L. monocytogenes during HeLa and Jeg-3 cell infections. Electron microscopy reveals a band of linear actin filaments that run along the longitudinal axis of the invagination membrane. Mechanistically, mDia1 expression is vital for the assembly of this F-actin shell. mDia1 is also required for the recruitment of Filamin A, a caveola-associated F-actin cross-linking protein, and caveolin-1 to the invaginations. Importantly, mixed-cell infection assays show that optimal caveolin-based L. monocytogenes cell-to-cell spreading correlates with the formation of the linear actin filament-containing shell by mDia1. IMPORTANCE Listeria monocytogenes spreads from one cell to another to colonize tissues. This cell-to-cell movement requires the propulsive force of an actin-rich comet tail behind the advancing bacterium, which ultimately distends the host plasma membrane into a slender bacterium-containing membrane protrusion. These membrane protrusions induce a corresponding invagination in the membrane of the adjacent host cell. The host cell that receives the protrusion utilizes caveolin-based endocytosis to internalize the structures, and filamentous actin lines these membrane invaginations. Here, we set out to determine the structure and function of this filamentous actin "shell." We demonstrate that the formin mDia1, but not the Arp2/3 complex, localizes to the invaginations. Morphologically, we show that this actin is organized into linear arrays and not branched dendritic networks. Mechanistically, we show that the actin shell is assembled by mDia1 and that mDia1 is required for efficient cell-to-cell transfer of L. monocytogenes.


Assuntos
Actinas/metabolismo , Membrana Celular/microbiologia , Forminas/metabolismo , Listeria monocytogenes/fisiologia , Listeriose/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiologia , Caveolina 1/genética , Caveolina 1/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Filaminas/genética , Filaminas/metabolismo , Forminas/genética , Células HeLa , Humanos , Listeria monocytogenes/genética , Listeriose/genética , Listeriose/microbiologia
17.
Microbiology (Reading) ; 156(Pt 2): 340-355, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19926655

RESUMO

Waddlia chondrophila is an obligate intracellular bacterium considered as a potential agent of abortion in both humans and bovines. This member of the order Chlamydiales multiplies rapidly within human macrophages and induces lysis of the infected cells. To understand how this Chlamydia-like micro-organism invades and proliferates within host cells, we investigated its trafficking within monocyte-derived human macrophages. Vacuoles containing W. chondrophila acquired the early endosomal marker EEA1 during the first 30 min following uptake. However, the live W. chondrophila-containing vacuoles never co-localized with late endosome and lysosome markers. Instead of interacting with the endosomal pathway, W. chondrophila immediately co-localized with mitochondria and, shortly after, with endoplasmic reticulum- (ER-) resident proteins such as calnexin and protein disulfide isomerase. The acquisition of mitochondria and ER markers corresponds to the beginning of bacterial replication. It is noteworthy that mitochondrion recruitment to W. chondrophila inclusions is prevented only by simultaneous treatment with the microtubule and actin cytoskeleton-disrupting agents nocodazole and cytochalasin D. In addition, brefeldin A inhibits the replication of W. chondrophila, supporting a role for COPI-dependent trafficking in the biogenesis of the bacterial replicating vacuole. W. chondrophila probably survives within human macrophages by evading the endocytic pathway and by associating with mitochondria and the ER. The intracellular trafficking of W. chondrophila in human macrophages represents a novel route that differs strongly from that used by other members of the order Chlamydiales.


Assuntos
Chlamydiales/fisiologia , Macrófagos/microbiologia , Citoesqueleto de Actina/microbiologia , Células Cultivadas , Chlamydiales/crescimento & desenvolvimento , Complexo I de Proteína do Envoltório/metabolismo , Citoplasma/microbiologia , Retículo Endoplasmático/microbiologia , Endossomos/microbiologia , Complexo de Golgi/metabolismo , Humanos , Macrófagos/metabolismo , Microtúbulos/microbiologia , Mitocôndrias/metabolismo
18.
FEBS Lett ; 592(22): 3658-3669, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29935019

RESUMO

The actin cytoskeleton and Rho GTPase signaling to actin assembly are prime targets of bacterial and viral pathogens, simply because actin is involved in all motile and membrane remodeling processes, such as phagocytosis, macropinocytosis, endocytosis, exocytosis, vesicular trafficking and membrane fusion events, motility, and last but not least, autophagy. This article aims at providing an overview of the most prominent pathogen-induced or -hijacked actin structures, and an outlook on how future research might uncover additional, equally sophisticated interactions.


Assuntos
Citoesqueleto de Actina/microbiologia , Citoesqueleto de Actina/virologia , Membrana Celular/microbiologia , Membrana Celular/virologia , Interações Hospedeiro-Patógeno , Citoesqueleto de Actina/metabolismo , Autofagia , Bactérias/patogenicidade , Membrana Celular/metabolismo , Humanos , Transdução de Sinais , Virulência , Vírus/patogenicidade
19.
J Vet Sci ; 19(2): 207-215, 2018 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-28693312

RESUMO

Pasteurella multocida serotype B:2 causes hemorrhagic septicemia in cattle and buffalo. The invasion mechanism of the bacterium when invading the bloodstream is unclear. This study aimed to characterize the effects of immunomodulatory molecules, namely dexamethasone and lipopolysaccharide, on the invasion efficiency of P. multocida serotype B:2 toward bovine aortic endothelial cells (BAECs) and the involvement of actin microfilaments in the invasion mechanism. The results imply that treatment of BAECs with lipopolysaccharide at 100 ng/mL for 24 h significantly increases the intracellular bacteria number per cell (p < 0.01) compared with those in untreated and dexamethasone-treated cells. The lipopolysaccharide-treated cells showed a significant decrease in F-actin expression and an increase in G-actin expression (p < 0.001), indicating actin depolymerization of BAECs. However, no significant differences were detected in the invasion efficiency and actin filament reorganization between the dexamethasone-treated and untreated cells. Transmission electron microscopy showed that P. multocida B:2 resided in a vacuolar compartment of dexamethasone-treated and untreated cells, whereas the bacteria resided in cellular membrane of lipopolysaccharide-treated cells. The results suggest that lipopolysaccharide destabilizes the actin filaments of BAECs, which could facilitate the invasion of P. multocida B:2 into BAECs.


Assuntos
Aorta/microbiologia , Endotélio Vascular/microbiologia , Lipopolissacarídeos/farmacologia , Pasteurella multocida/patogenicidade , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/microbiologia , Animais , Aorta/efeitos dos fármacos , Bovinos , Dexametasona/farmacologia , Endotélio Vascular/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Técnicas In Vitro , Microscopia Eletrônica de Transmissão , Pasteurella multocida/efeitos dos fármacos
20.
Sci Rep ; 7: 41252, 2017 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-28128281

RESUMO

The actin cytoskeleton is an attractive target for bacterial toxins. The ADP-ribosyltransferase TccC3 from the insect bacterial pathogen Photorhabdus luminescence modifies actin to force its aggregation. We intended to transport the catalytic part of this toxin preferentially into cancer cells using a toxin transporter (Protective antigen, PA) which was redirected to Epidermal Growth Factor Receptors (EGFR) or to human EGF receptors 2 (HER2), which are overexpressed in several cancer cells. Protective antigen of anthrax toxin forms a pore through which the two catalytic parts (lethal factor and edema factor) or other proteins can be transported into mammalian cells. Here, we used PA as a double mutant (N682A, D683A; mPA) which cannot bind to the two natural anthrax receptors. Each mutated monomer is fused either to EGF or to an affibody directed against the human EGF receptor 2 (HER2). We established a cellular model system composed of two cell lines representing HER2 overexpressing esophageal adenocarcinomas (EACs) and EGFR overexpressing esophageal squamous cell carcinomas (ESCCs). We studied the specificity and efficiency of the re-directed anthrax pore for transport of TccC3 toxin and established Photorhabdus luminescence TccC3 as a toxin suitable for the development of a targeted toxin selectively killing cancer cells.


Assuntos
ADP Ribose Transferases/química , ADP-Ribosilação/genética , Toxinas Bacterianas/química , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias Esofágicas/tratamento farmacológico , ADP Ribose Transferases/genética , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/microbiologia , Antígenos de Bactérias/química , Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Receptores ErbB/química , Receptores ErbB/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Photorhabdus/química , Receptor ErbB-2/química , Receptor ErbB-2/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa