Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
mSphere ; 7(6): e0048522, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36346241

RESUMO

Shigella flexneri uses a type 3 secretion system (T3SS) apparatus to inject virulence effector proteins into the host cell cytosol. Upon host cell contact, MxiE, an S. flexneri AraC-like transcriptional regulator, is required for the expression of a subset of T3SS effector genes encoded on the large virulence plasmid. Here, we defined the MxiE regulon using RNA-seq. We identified virulence plasmid- and chromosome-encoded genes that are activated in response to type 3 secretion in a MxiE-dependent manner. Bioinformatic analysis revealed that similar to previously known MxiE-dependent genes, chromosome-encoded genes yccE and yfdF contain a regulatory element known as the MxiE box, which is required for their MxiE-dependent expression. The significant AT enrichment of MxiE-dependent genes suggested the involvement of H-NS. Using a dominant negative H-NS system, we demonstrate that H-NS silences the expression of MxiE-dependent genes located on the virulence plasmid (ipaH7.8 and ospC1) and the chromosome (yccE and yfdF). Furthermore, we show that MxiE is no longer required for the expression of ipaH7.8, ospC1, yccE, and yfdF when H-NS silencing is relieved. Finally, we show that the H-NS anti-silencer VirB is not required for ipaH7.8 and yccE expression upon MxiE/IpgC overexpression. Based on these genetic studies, we propose a model of MxiE-dependent gene regulation in which MxiE counteracts H-NS-mediated silencing. IMPORTANCE The expression of horizontally acquired genes, including virulence genes, is subject to complex regulation involving xenogeneic silencing proteins, and counter-silencing mechanisms. The pathogenic properties of Shigella flexneri mainly rely on the acquisition of the type 3 secretion system (T3SS) and cognate effector proteins, whose expression is repressed by the xenogeneic silencing protein H-NS. Based on previous studies, releasing H-NS-mediated silencing mainly relies on two mechanisms involving (i) a temperature shift leading to the release of H-NS at the virF promoter, and (ii) the virulence factor VirB, which dislodges H-NS upon binding to specific motifs upstream of virulence genes, including those encoding the T3SS. In this study, we provide genetic evidence supporting the notion that, in addition to VirB, the AraC family member MxiE also contributes to releasing H-NS-mediated silencing in S. flexneri.


Assuntos
Proteínas de Bactérias , Proteínas de Ligação a DNA , Proteínas de Ligação a DNA/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Shigella flexneri/genética , Shigella flexneri/metabolismo , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
PLoS Pathog ; 18(2): e1010380, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35202448

RESUMO

S. flexneri is an important human pathogen that causes bacillary dysentery. During infection, S. flexneri invades colonic epithelial cells, hijacks the host cell cytoskeleton to move in the cytosol of infected cells, and spreads from cell to cell through formation of membrane protrusions that project into adjacent cells and resolve into double membrane vacuoles (DMVs). S. flexneri cell-to-cell spread requires the integrity of the bacterial type three secretion system (T3SS). However, the exact role of the T3SS effector proteins in the dissemination process remains poorly understood. Here, we investigated the role of the T3SS effector protein IpgB1 in S. flexneri dissemination. IpgB1 was previously characterized as a guanine nucleotide exchange factor (GEF) that contributes to invasion. In addition to the invasion defect, we showed that the ipgB1 mutant formed smaller infection foci in HT-29 cells. Complementation of this phenotype required the GEF activity of IpgB1. Using live confocal microscopy, we showed that the ipgB1 mutant is specifically impaired in DMV escape. Depletion of Rac1, the host cell target of IpgB1 during invasion, as well as pharmacological inhibition of Rac1 signaling, reduced cell-to-cell spread and DMV escape. In a targeted siRNA screen, we uncovered that RhoA depletion restored ipgB1 cell-to-cell spread and DMV escape, revealing a critical role for the IpgB1-Rac1 axis in antagonizing RhoA-mediated restriction of DMV escape. Using an infant rabbit model of shigellosis, we showed that the ipgB1 mutant formed fewer and smaller infection foci in the colon of infected animals, which correlated with attenuated symptoms of disease, including epithelial fenestration and bloody diarrhea. Our results demonstrate that, in addition to its role during invasion, IpgB1 modulates Rho family small GTPase signaling to promote cell-to-cell spread, DMV escape, and S. flexneri pathogenesis.


Assuntos
Disenteria Bacilar , Shigella flexneri , Proteínas rac1 de Ligação ao GTP , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Disenteria Bacilar/microbiologia , Células Epiteliais/metabolismo , Humanos , Coelhos , Shigella flexneri/genética , Shigella flexneri/metabolismo , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo , Vacúolos/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
3.
PLoS Pathog ; 18(2): e1010324, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35130324

RESUMO

The bacterial pathogen Shigella flexneri causes 270 million cases of bacillary dysentery worldwide every year, resulting in more than 200,000 deaths. S. flexneri pathogenic properties rely on its ability to invade epithelial cells and spread from cell to cell within the colonic epithelium. This dissemination process relies on actin-based motility in the cytosol of infected cells and formation of membrane protrusions that project into adjacent cells and resolve into double-membrane vacuoles (DMVs) from which the pathogen escapes, thereby achieving cell-to-cell spread. S. flexneri dissemination is facilitated by the type 3 secretion system (T3SS) through poorly understood mechanisms. Here, we show that the T3SS effector IpgD facilitates the resolution of membrane protrusions into DMVs during S. flexneri dissemination. The phosphatidylinositol 4-phosphatase activity of IpgD decreases PtdIns(4,5)P2 levels in membrane protrusions, thereby counteracting de novo cortical actin formation in protrusions, a process that restricts the resolution of protrusions into DMVs. Finally, using an infant rabbit model of shigellosis, we show that IpgD is required for efficient cell-to-cell spread in vivo and contributes to the severity of dysentery.


Assuntos
Proteínas de Bactérias/metabolismo , Extensões da Superfície Celular/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Shigella flexneri/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Actinas/metabolismo , Animais , Proteínas de Bactérias/genética , Extensões da Superfície Celular/microbiologia , Colo/microbiologia , Modelos Animais de Doenças , Disenteria Bacilar/microbiologia , Células HT29 , Interações Hospedeiro-Patógeno , Humanos , Monoéster Fosfórico Hidrolases/genética , Coelhos , Shigella flexneri/genética
4.
Curr Biol ; 31(11): 2469-2476.e5, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-33852873

RESUMO

Apoptotic cells are quickly and efficiently engulfed and removed via the process of efferocytosis by either professional phagocytes, such as macrophages, or non-professional phagocytes, including epithelial cells.1,2 In addition to debris removal, a key benefit of efferocytosis is that phagocytes engulfing apoptotic cells release anti-inflammatory mediators3,4 that help reduce local tissue inflammation;5 conversely, accumulation of uncleared apoptotic cells predisposes to a pro-inflammatory tissue milieu.6-8 Due to their high proliferative capacity, intestinal epithelial cells (iECs) are sensitive to inflammation, irradiation, and chemotherapy-induced DNA damage, leading to apoptosis. Mechanisms of iEC death in the context of irradiation has been studied,9,10 but phagocytosis of dying iECs is poorly understood. Here, we identify an unexpected efferocytic role for Paneth cells, which reside in intestinal crypts and are linked to innate immunity and maintenance of the stem cell niche in the crypt.11,12 Through a series of studies spanning in vitro efferocytosis, ex vivo intestinal organoids ("enteroids"), and in vivo Cre-mediated deletion of Paneth cells, we show that Paneth cells mediate apoptotic cell uptake of dying neighbors. The relevance of Paneth-cell-mediated efferocytosis was revealed ex vivo and in mice after low-dose cesium-137 (137Cs) irradiation, mimicking radiation therapies given to cancer patients often causing significant apoptosis of iECs. These data advance a new concept that Paneth cells can act as phagocytes and identify another way in which Paneth cells contribute to the overall health of the intestine. These observations also have implications for individuals undergoing chemotherapy or chronic inflammatory bowel disease.


Assuntos
Celulas de Paneth , Fagocitose , Animais , Apoptose , Humanos , Inflamação , Intestinos , Camundongos , Fagócitos
5.
Clin Infect Dis ; 72(11): e868-e871, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32940644

RESUMO

In a cohort of infants, we found that lack of the Lewis histo-blood group antigen was associated with increased susceptibility to shigellosis. Broadly inhibiting fucosylation in epithelial cells in vitro decreased invasion by Shigella flexneri. These results support a role for fucosylated glycans in susceptibility to shigellosis.


Assuntos
Disenteria Bacilar , Humanos , Lactente , Antígenos do Grupo Sanguíneo de Lewis
6.
Gut Microbes ; 11(3): 597-602, 2020 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-31570038

RESUMO

The bacterial pathogen Shigella flexneri causes more than 250 million cases of bacillary dysentery (blood in stool) every year across the world. This human-specific disease is characterized by profuse bloody diarrhea, dramatic ulceration of the colonic epithelium and immune cell infiltration of the colonic tissue. A major challenge in understanding the mechanisms supporting bacillary dysentery is the reliance on animal models that do not fully recapitulate the symptoms observed in humans, including bloody diarrhea. Here we outline advances provided by a recently developed infant rabbit model of bacillary dysentery. The infant rabbit model defines bacillary dysentery as a critical combination of massive vascular lesions and dramatic epithelial fenestration due to intracellular infection and cell-to-cell spread, respectively. The infant rabbit model provides an unprecedented framework for understanding how the cell biology of Shigella flexneri infection relates to pathogenesis.


Assuntos
Colo/microbiologia , Colo/patologia , Disenteria Bacilar/microbiologia , Disenteria Bacilar/patologia , Shigella flexneri/patogenicidade , Animais , Animais Recém-Nascidos/microbiologia , Colo/imunologia , Diarreia/imunologia , Diarreia/microbiologia , Modelos Animais de Doenças , Disenteria Bacilar/imunologia , Cobaias , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Coelhos
7.
mBio ; 10(4)2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31455648

RESUMO

Various bacterial pathogens display an intracellular lifestyle and spread from cell to cell through actin-based motility (ABM). ABM requires actin polymerization at the bacterial pole and is mediated by the expression of bacterial factors that hijack the host cell actin nucleation machinery or exhibit intrinsic actin nucleation properties. It is increasingly recognized that bacterial ABM factors, in addition to having a crucial task during the intracellular phase of infection, display "moonlighting" adhesin functions, such as bacterial aggregation, biofilm formation, and host cell adhesion/invasion. Here, we review our current knowledge of ABM factors and their additional functions, and we propose that intracellular ABM functions have evolved from ancestral, extracellular adhesin functions.


Assuntos
Actinas/metabolismo , Bactérias/patogenicidade , Fenômenos Fisiológicos Bacterianos , Biofilmes/crescimento & desenvolvimento , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Listeria/patogenicidade , Listeria/fisiologia , Shigella/patogenicidade , Shigella/fisiologia
8.
Cell Microbiol ; 21(12): e13098, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31414511

RESUMO

Shigella flexneri is an intracellular pathogen that disseminates in colonic epithelial cells through actin-based motility and formation of membrane protrusions at cell-cell contacts, that project into adjacent cells and resolve into vacuoles, from which the pathogen escapes, thereby achieving cell-to-cell spread. Actin nucleation at the bacterial pole relies on the recruitment of the nucleation-promoting factor N-WASP, which activates the actin nucleator ARP2/3. In cells, the vast majority of N-WASP exists as a complex with WIP. The involvement of WIP in N-WASP-dependent actin-based motility of various pathogens, including vaccinia virus and S. flexneri, has been highly controversial. Here, we show that WIPF2 was the only WIP family member expressed in the human colonic epithelial cell line HT-29, and its depletion impaired S. flexneri dissemination. WIPF2 depletion increased the number of cytosolic bacteria lacking actin tails (non-motile) and decreased the velocity of motile bacteria. This correlated with a decrease in the recruitment of N-WASP to the bacterial pole, and among N-WASP-positive bacteria, a decrease in actin tail-positive bacteria, suggesting that WIPF2 is required for N-WASP recruitment and activation at the bacterial pole. In addition, when motile bacteria formed protrusions, WIPF2 depletion decreased the number of membrane protrusions that successfully resolved into vacuoles.


Assuntos
Actinas/metabolismo , Movimento Celular/fisiologia , Disenteria Bacilar/metabolismo , Proteínas dos Microfilamentos/metabolismo , Shigella flexneri/metabolismo , Linhagem Celular Tumoral , Disenteria Bacilar/parasitologia , Células Epiteliais/metabolismo , Células Epiteliais/parasitologia , Células HT29 , Células HeLa , Humanos , Shigella flexneri/fisiologia , Vacúolos/metabolismo
9.
Infect Immun ; 87(7)2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30988059

RESUMO

Shigella flexneri is an intracellular bacterial pathogen that invades epithelial cells in the colonic mucosa, leading to bloody diarrhea. A previous study showed that S. flexneri forms biofilms in the presence of bile salts, through an unknown mechanism. Here, we investigated the potential role of adhesin-like autotransporter proteins in S. flexneri biofilm formation. BLAST search analysis revealed that the S. flexneri 2457T genome harbors 4 genes, S1242, S1289, S2406, and icsA, encoding adhesin-like autotransporter proteins. Deletion mutants of the S1242, S1289, S2406 and icsA genes were generated and tested for biofilm formation. Phenotypic analysis of the mutant strains revealed that disruption of icsA abolished bile salt-induced biofilm formation. IcsA is an outer membrane protein secreted at the bacterial pole that is required for S. flexneri actin-based motility during intracellular infection. In extracellular biofilms, IcsA was also secreted at the bacterial pole and mediated bacterial cell-cell contacts and aggregative growth in the presence of bile salts. Dissecting individual roles of bile salts showed that deoxycholate is a robust biofilm inducer compared to cholate. The release of the extracellular domain of IcsA through IcsP-mediated cleavage was greater in the presence of cholate, suggesting that the robustness of biofilm formation was inversely correlated with IcsA processing. Accordingly, deletion of icsP abrogated IcsA processing in biofilms and enhanced biofilm formation.


Assuntos
Proteínas de Bactérias/fisiologia , Ácidos e Sais Biliares/farmacologia , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Proteínas de Ligação a DNA/fisiologia , Shigella flexneri/fisiologia , Fatores de Transcrição/fisiologia , Sistemas de Secreção Tipo V/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/genética , Ácidos e Sais Biliares/metabolismo , Proteínas de Ligação a DNA/genética , Shigella flexneri/efeitos dos fármacos , Shigella flexneri/metabolismo , Fatores de Transcrição/genética
10.
Nat Commun ; 10(1): 1826, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31015451

RESUMO

The bacterial pathogen Shigella flexneri causes 270 million cases of bacillary dysentery (blood in stool) worldwide every year, resulting in more than 200,000 deaths. A major challenge in combating bacillary dysentery is the lack of a small-animal model that recapitulates the symptoms observed in infected individuals, including bloody diarrhea. Here, we show that similar to humans, infant rabbits infected with S. flexneri experience severe inflammation, massive ulceration of the colonic mucosa, and bloody diarrhea. T3SS-dependent invasion of epithelial cells is necessary and sufficient for mediating immune cell infiltration and vascular lesions. However, massive ulceration of the colonic mucosa, bloody diarrhea, and dramatic weight loss are strictly contingent on the ability of the bacteria to spread from cell to cell. The infant rabbit model features bacterial dissemination as a critical determinant of S. flexneri pathogenesis and provides a unique small-animal model for research and development of therapeutic interventions.


Assuntos
Diarreia/patologia , Disenteria Bacilar/patologia , Hemorragia Gastrointestinal/patologia , Shigella flexneri/patogenicidade , Sistemas de Secreção Tipo III/imunologia , Animais , Animais Recém-Nascidos/microbiologia , Colo/microbiologia , Colo/patologia , Diarreia/microbiologia , Modelos Animais de Doenças , Disenteria Bacilar/microbiologia , Células Epiteliais/microbiologia , Feminino , Hemorragia Gastrointestinal/microbiologia , Células HT29 , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Gravidez , Coelhos
11.
Gut Microbes ; 10(5): 615-630, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30712505

RESUMO

Shigella is one of the major enteric pathogens worldwide. We present a murine model of S. flexneri infection and investigate the role of zinc deficiency (ZD). C57BL/6 mice fed either standard chow (HC) or ZD diets were pretreated with an antibiotic cocktail and received S. flexneri strain 2457T orally. Antibiotic pre-treated ZD mice showed higher S. flexneri colonization than non-treated mice. ZD mice showed persistent colonization for at least 50 days post-infection (pi). S. flexneri-infected mice showed significant weight loss, diarrhea and increased levels of fecal MPO and LCN in both HC and ZD fed mice. S. flexneri preferentially colonized the colon, caused epithelial disruption and inflammatory cell infiltrate, and promoted cytokine production which correlated with weight loss and histopathological changes. Infection with S. flexneri ΔmxiG (critical for type 3 secretion system) did not cause weight loss or diarrhea, and had decreased stool shedding duration and tissue burden. Several biochemical changes related to energy, inflammation and gut-microbial metabolism were observed. Zinc supplementation increased weight gains and reduced intestinal inflammation and stool shedding in ZD infected mice. In conclusion, young antibiotic-treated mice provide a new model of oral S. flexneri infection, with ZD promoting prolonged infection outcomes.


Assuntos
Diarreia/patologia , Modelos Animais de Doenças , Disenteria Bacilar/patologia , Shigella flexneri/patogenicidade , Zinco/deficiência , Animais , Antibacterianos/administração & dosagem , Peso Corporal , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Diarreia/tratamento farmacológico , Diarreia/metabolismo , Diarreia/microbiologia , Disenteria Bacilar/tratamento farmacológico , Disenteria Bacilar/metabolismo , Disenteria Bacilar/microbiologia , Fezes/enzimologia , Fezes/microbiologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Metaboloma , Camundongos Endogâmicos C57BL , Mutação , Shigella flexneri/genética , Shigella flexneri/crescimento & desenvolvimento , Sistemas de Secreção Tipo III/genética
13.
Infect Immun ; 86(8)2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29844234

RESUMO

Shigella flexneri disseminates within the colonic mucosa by displaying actin-based motility in the cytosol of epithelial cells. Motile bacteria form membrane protrusions that project into adjacent cells and resolve into double-membrane vacuoles (DMVs) from which the bacteria escape, thereby achieving cell-to-cell spread. During dissemination, S. flexneri is targeted by LC3-dependent autophagy, a host cell defense mechanism against intracellular pathogens. The S. flexneri type III secretion system effector protein IcsB was initially proposed to counteract the recruitment of the LC3-dependent autophagy machinery to cytosolic bacteria. However, a recent study proposed that LC3 was recruited to bacteria in DMVs formed during cell-to-cell spread. To resolve the controversy and clarify the role of autophagy in S. flexneri infection, we tracked dissemination using live confocal microscopy and determined the spatial and temporal recruitment of LC3 to bacteria. This approach demonstrated that (i) LC3 was exclusively recruited to wild-type or icsB bacteria located in DMVs and (ii) the icsB mutant was defective in cell-to-cell spread due to failure to escape LC3-positive as well as LC3-negative DMVs. Failure of S. flexneri to escape DMVs correlated with late LC3 recruitment, suggesting that LC3 recruitment is the consequence and not the cause of DMV escape failure. Inhibition of autophagy had no positive impact on the spreading of wild-type or icsB mutant bacteria. Our results unambiguously demonstrate that IcsB is required for DMV escape during cell-to-cell spread, regardless of LC3 recruitment, and do not support the previously proposed notion that autophagy counters S. flexneri dissemination.


Assuntos
Autofagia , Proteínas de Bactérias/metabolismo , Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno , Proteínas Associadas aos Microtúbulos/metabolismo , Shigella flexneri/crescimento & desenvolvimento , Vacúolos/microbiologia , Proteínas de Bactérias/genética , Linhagem Celular , Disenteria Bacilar/fisiopatologia , Humanos , Microscopia Intravital , Microscopia Confocal , Mutação , Ligação Proteica , Análise Espaço-Temporal , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
14.
Infect Immun ; 86(5)2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29531136

RESUMO

Ethanolamine is a ubiquitous and essential molecule within a host. Significantly, bacterial pathogens exploit ethanolamine during infection to promote growth and regulate virulence. The ethanolamine permease EutH is dispensable for growth in vitro under standard conditions, whereas EutH is required for ethanolamine utilization at low pH. These findings suggested a model in which EutH facilitates diffusion of ethanolamine into the bacterial cell in acidic environments. To date, the ecological significance of this model has not been thoroughly investigated, and the importance of EutH to bacterial growth under physiologically relevant conditions is not known. During infection, immune cells internalize invading bacteria within an acidic, nutrient-depleted vacuole called the phagosome. Here, we investigated the hypothesis that EutH promotes bacterial survival following phagocytosis. Our findings indicate that EutH is important for survival and replication of the facultative intracellular pathogens Salmonella enterica serovar Typhimurium and Listeria monocytogenes during prolonged or transient exposure to the phagosome, respectively. Furthermore, in agreement with EutH being important in the acidic environment, neutralization of the vacuole abolished the requirement for EutH. Significantly, consistent with a role for EutH in promoting intramacrophage survival, EutH was not required during S Typhimurium local intestinal infection but specifically conferred an advantage upon dissemination to peripheral organs. These findings reveal a physiologically relevant and conserved role for EutH in spatiotemporal niche adaptation during infection.


Assuntos
Proteínas de Bactérias/fisiologia , Transporte Biológico/fisiologia , Etanolaminas/metabolismo , Listeria monocytogenes/patogenicidade , Macrófagos/patologia , Salmonella enterica/patogenicidade , Vacúolos/microbiologia
15.
mBio ; 8(4)2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28851846

RESUMO

Cardiolipin, an anionic phospholipid that resides at the poles of the inner and outer membranes, is synthesized primarily by the putative cardiolipin synthase ClsA in Shigella flexneri An S. flexneri clsA mutant had no cardiolipin detected within its membrane, grew normally in vitro, and invaded cultured epithelial cells, but it failed to form plaques in epithelial cell monolayers, indicating that cardiolipin is required for virulence. The clsA mutant was initially motile within the host cell cytoplasm but formed filaments and lost motility during replication and failed to spread efficiently to neighboring cells. Mutation of pbgA, which encodes the transporter for cardiolipin from the inner membrane to the outer membrane, also resulted in loss of plaque formation. The S. flexneri pbgA mutant had normal levels of cardiolipin in the inner membrane, but no cardiolipin was detected in the outer membrane. The pbgA mutant invaded and replicated normally within cultured epithelial cells but failed to localize the actin polymerization protein IcsA properly on the bacterial surface and was unable to spread to neighboring cells. The clsA mutant, but not the pbgA mutant, had increased phosphatidylglycerol in the outer membrane. This appeared to compensate partially for the loss of cardiolipin in the outer membrane, allowing some IcsA localization in the outer membrane of the clsA mutant. We propose a dual function for cardiolipin in S. flexneri pathogenesis. In the inner membrane, cardiolipin is essential for proper cell division during intracellular growth. In the outer membrane, cardiolipin facilitates proper presentation of IcsA on the bacterial surface.IMPORTANCE The human pathogen Shigella flexneri causes bacterial dysentery by invading colonic epithelial cells, rapidly multiplying within their cytoplasm, and then spreading intercellularly to neighboring cells. Worldwide, Shigella spp. infect hundreds of millions of people annually, with fatality rates up to 15%. Antibiotic treatment of Shigella infections is compromised by increasing antibiotic resistance, and there is no approved vaccine to prevent future infections. This has created a growing need to understand Shigella pathogenesis and identify new targets for antimicrobial therapeutics. Here we show a previously unknown role of phospholipids in S. flexneri pathogenesis. We demonstrate that cardiolipin is required in the outer membrane for proper surface localization of IcsA and in the inner membrane for cell division during growth in the host cell cytoplasm.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Cardiolipinas/biossíntese , Shigella flexneri/metabolismo , Shigella flexneri/patogenicidade , Actinas/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cardiolipinas/metabolismo , Citoplasma/metabolismo , Citoplasma/microbiologia , Proteínas de Ligação a DNA/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Mutação , Shigella flexneri/genética , Transferases (Outros Grupos de Fosfato Substituídos)/metabolismo , Virulência
16.
Infect Immun ; 84(6): 1806-1814, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27068088

RESUMO

Listeria monocytogenes is an intracellular pathogen that disseminates within the intestinal epithelium through acquisition of actin-based motility and formation of plasma membrane protrusions that project into adjacent cells. The resolution of membrane protrusions into vacuoles from which the pathogen escapes results in bacterial spread from cell to cell. This dissemination process relies on the mlp-actA-plcB operon, which encodes ActA, a bacterial nucleation-promoting factor that mediates actin-based motility, and PlcB, a phospholipase that mediates vacuole escape. Here we investigated the role of the metalloprotease Mpl in the dissemination process. In agreement with previous findings showing that Mpl is required for PlcB activation, infection of epithelial cells with the ΔplcB or Δmpl strains resulted in the formation of small infection foci. As expected, the ΔplcB strain displayed a strong defect in vacuole escape. However, the Δmpl strain showed an unexpected defect in the resolution of protrusions into vacuoles, in addition to the expected but mild defect in vacuole escape. The Δmpl strain displayed increased levels of ActA on the bacterial surface in protrusions. We mapped an Mpl-dependent processing site in ActA between amino acid residues 207 to 238. Similar to the Δmpl strain, the ΔactA207-238 strain displayed increased levels of ActA on the bacterial surface in protrusions. Although the ΔactA207-238 strain displayed wild-type actin-based motility, it formed small infection foci and failed to resolve protrusions into vacuoles. We propose that, in addition to its role in PlcB processing and vacuole escape, the metalloprotease Mpl is required for ActA processing and protrusion resolution.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Listeria monocytogenes/genética , Proteínas de Membrana/genética , Metaloendopeptidases/genética , Fosfolipases Tipo C/genética , Vacúolos/microbiologia , Sequência de Aminoácidos , Proteínas de Bactérias/imunologia , Sítios de Ligação , Membrana Celular/imunologia , Membrana Celular/microbiologia , Membrana Celular/ultraestrutura , Citoplasma/imunologia , Citoplasma/microbiologia , Citoplasma/ultraestrutura , Deleção de Genes , Células HeLa , Humanos , Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/imunologia , Proteínas de Membrana/imunologia , Metaloendopeptidases/imunologia , Óperon , Ligação Proteica , Fosfolipases Tipo C/imunologia , Vacúolos/imunologia , Vacúolos/ultraestrutura
17.
Artigo em Inglês | MEDLINE | ID: mdl-27014639

RESUMO

The intracellular pathogen Shigella flexneri is the causative agent of bacillary dysentery in humans. The disease is characterized by bacterial invasion of intestinal cells, dissemination within the colonic epithelium through direct spread from cell to cell, and massive inflammation of the intestinal mucosa. Here, we review the mechanisms supporting S. flexneri dissemination. The dissemination process primarily relies on actin assembly at the bacterial pole, which propels the pathogen throughout the cytosol of primary infected cells. Polar actin assembly is supported by polar expression of the bacterial autotransporter family member IcsA, which recruits the N-WASP/ARP2/3 actin assembly machinery. As motile bacteria encounter cell-cell contacts, they form plasma membrane protrusions that project into adjacent cells. In addition to the ARP2/3-dependent actin assembly machinery, protrusion formation relies on formins and myosins. The resolution of protrusions into vacuoles occurs through the collapse of the protrusion neck, leading to the formation of an intermediate membrane-bound compartment termed vacuole-like protrusions (VLPs). VLP formation requires tyrosine kinase and phosphoinositide signaling in protrusions, which relies on the integrity of the bacterial type 3 secretion system (T3SS). The T3SS is also required for escaping double membrane vacuoles through the activity of the T3SS translocases IpaB and IpaC, and the effector proteins VirA and IcsB. Numerous factors supporting envelope biogenesis contribute to IcsA exposure and maintenance at the bacterial pole, including LPS synthesis, membrane proteases, and periplasmic chaperones. Although less characterized, the assembly and function of the T3SS in the context of bacterial dissemination also relies on factors supporting envelope biogenesis. Finally, the dissemination process requires the adaptation of the pathogen to various cellular compartments through transcriptional and post-transcriptional mechanisms.


Assuntos
Actinas/metabolismo , Disenteria Bacilar/microbiologia , Mucosa Intestinal/patologia , Shigella flexneri/patogenicidade , Sistemas de Secreção Tipo III/metabolismo , Vacúolos/microbiologia , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/metabolismo , Transporte Biológico , Extensões da Superfície Celular , Proteínas de Ligação a DNA/biossíntese , Disenteria Bacilar/patologia , Células Epiteliais/microbiologia , Humanos , Mucosa Intestinal/microbiologia , Fatores de Transcrição/biossíntese , Fatores de Virulência/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo
18.
Mol Biol Cell ; 27(2): 247-59, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26609071

RESUMO

The Nck adaptor protein recruits cytosolic effectors such as N-WASP that induce localized actin polymerization. Experimental aggregation of Nck SH3 domains at the membrane induces actin comet tails--dynamic, elongated filamentous actin structures similar to those that drive the movement of microbial pathogens such as vaccinia virus. Here we show that experimental manipulation of the balance between unbranched/branched nucleation altered the morphology and dynamics of Nck-induced actin comets. Inhibition of linear, formin-based nucleation with the small-molecule inhibitor SMIFH2 or overexpression of the formin FH1 domain resulted in formation of predominantly circular-shaped actin structures with low mobility (actin blobs). These results indicate that formin-based linear actin polymerization is critical for the formation and maintenance of Nck-dependent actin comet tails. Consistent with this, aggregation of an exclusively branched nucleation-promoting factor (the VCA domain of N-WASP), with density and turnover similar to those of N-WASP in Nck comets, did not reconstitute dynamic, elongated actin comets. Furthermore, enhancement of branched Arp2/3-mediated nucleation by N-WASP overexpression caused loss of the typical actin comet tail shape induced by Nck aggregation. Thus the ratio of linear to dendritic nucleation activity may serve to distinguish the properties of actin structures induced by various viral and bacterial pathogens.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Oncogênicas/metabolismo , Animais , Dendritos/metabolismo , Proteínas Fetais/metabolismo , Forminas , Células HeLa , Humanos , Camundongos , Proteínas dos Microfilamentos/metabolismo , Células NIH 3T3 , Proteínas Nucleares/metabolismo , Relação Estrutura-Atividade , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Domínios de Homologia de src
19.
Small GTPases ; 6(2): 119-22, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26147090

RESUMO

Vaccinia virus dissemination relies on the recruitment of the nucleation promoting factor N-WASP underneath cell-associated extracellular virus (CEVs) and subsequent recruitment and activation of the ARP2/3 complex, a major actin nucleator of the host cell. We have recently discovered that, in addition to the N-WASP/ARP2/3 pathway, vaccinia actin-based motility also relies on the small GTPase Rac1 and its downstream effector the formin-type actin nucleator FHOD1. Here we discuss the potential signaling mechanisms supporting the integration of the N-WASP/ARP2/3 and Rac1/FHOD1 pathways. We suggest the existence of a receptor tyrosine kinase family member that would integrate the Src-dependent activation of the N-WASP/ARP2/3 pathway and the GTP exchange factor-dependent activation of the Rac1/FHOD1 pathway.


Assuntos
Vaccinia virus/fisiologia , Vacínia/metabolismo , Liberação de Vírus , Proteínas rac1 de Ligação ao GTP/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/metabolismo , Animais , Humanos , Vacínia/virologia , Vaccinia virus/metabolismo , Vaccinia virus/patogenicidade , Proteínas Virais/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...