Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
PLoS Pathog ; 16(12): e1009152, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33370401

RESUMO

Streptococcus pneumoniae or pneumococcus (PN) is a major causative agent of bacterial meningitis with high mortality in young infants and elderly people worldwide. The mechanism underlying PN crossing of the blood brain barrier (BBB) and specifically, the role of non-endothelial cells of the neurovascular unit that control the BBB function, remains poorly understood. Here, we show that the astroglial connexin 43 (aCx43), a major gap junctional component expressed in astrocytes, plays a predominant role during PN meningitis. Following intravenous PN challenge, mice deficient for aCx43 developed milder symptoms and showed severely reduced bacterial counts in the brain. Immunofluorescence analysis of brain slices indicated that PN induces the aCx43-dependent destruction of the network of glial fibrillary acid protein (GFAP), an intermediate filament protein specifically expressed in astrocytes and up-regulated in response to brain injury. PN also induced nuclear shrinkage in astrocytes associated with the loss of BBB integrity, bacterial translocation across endothelial vessels and replication in the brain cortex. We found that aCx4-dependent astrocyte damages could be recapitulated using in vitro cultured cells upon challenge with wild-type PN but not with a ply mutant deficient for the pore-forming toxin pneumolysin (Ply). Consistently, we showed that purified Ply requires Cx43 to promote host cell plasma membrane permeabilization in a process involving the Cx43-dependent release of extracellular ATP and prolonged increase of cytosolic Ca2+ in host cells. These results point to a critical role for astrocytes during PN meningitis and suggest that the cytolytic activity of the major virulence factor Ply at concentrations relevant to bacterial infection requires co-opting of connexin plasma membrane channels.


Assuntos
Astrócitos/metabolismo , Conexina 43/metabolismo , Meningite Pneumocócica , Estreptolisinas/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Streptococcus pneumoniae/metabolismo , Streptococcus pneumoniae/patogenicidade , Virulência/fisiologia , Fatores de Virulência/metabolismo
2.
PLoS Comput Biol ; 17(1): e1008169, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33411763

RESUMO

Streptococcus pyogenes (Group A streptococcus; GAS) is an important human pathogen responsible for mild to severe, life-threatening infections. GAS expresses a wide range of virulence factors, including the M family proteins. The M proteins allow the bacteria to evade parts of the human immune defenses by triggering the formation of a dense coat of plasma proteins surrounding the bacteria, including IgGs. However, the molecular level details of the M1-IgG interaction have remained unclear. Here, we characterized the structure and dynamics of this interaction interface in human plasma on the surface of live bacteria using integrative structural biology, combining cross-linking mass spectrometry and molecular dynamics (MD) simulations. We show that the primary interaction is formed between the S-domain of M1 and the conserved IgG Fc-domain. In addition, we show evidence for a so far uncharacterized interaction between the A-domain and the IgG Fc-domain. Both these interactions mimic the protein G-IgG interface of group C and G streptococcus. These findings underline a conserved scavenging mechanism used by GAS surface proteins that block the IgG-receptor (FcγR) to inhibit phagocytic killing. We additionally show that we can capture Fab-bound IgGs in a complex background and identify XLs between the constant region of the Fab-domain and certain regions of the M1 protein engaged in the Fab-mediated binding. Our results elucidate the M1-IgG interaction network involved in inhibition of phagocytosis and reveal important M1 peptides that can be further investigated as future vaccine targets.


Assuntos
Antígenos de Bactérias , Proteínas da Membrana Bacteriana Externa , Proteínas de Transporte , Imunoglobulina G , Streptococcus pyogenes , Antígenos de Bactérias/química , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Espectrometria de Massas , Simulação de Dinâmica Molecular , Fagocitose , Ligação Proteica , Streptococcus pyogenes/química , Streptococcus pyogenes/metabolismo , Fatores de Virulência/química , Fatores de Virulência/metabolismo
3.
J Infect Dis ; 223(12): 2174-2185, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33151309

RESUMO

Citrobacter freundii is a significant cause of human infections, responsible for food poisoning, diarrhea, and urinary tract infections. We previously identified a highly cytotoxic and adhesive C. freundii strain CF74 expressing a type VI secretion system (T6SS). In this study, we showed that in mice-derived macrophages, C. freundii CF74 activated the Nucleotide Oligomerization Domain -Like Receptor Family, Pyrin Domain Containing 3(NLRP3) inflammasomes in a T6SS-dependent manner. The C. freundii T6SS activated the inflammasomes mainly through caspase 1 and mediated pyroptosis of macrophages by releasing the cleaved gasdermin-N domain. The CF74 T6SS was required for flagellin-induced interleukin 1ß release by macrophages. We further show that the T6SS tail component and effector, hemolysin co-regulation protein-2 (Hcp-2), was necessary and sufficient to trigger NLRP3 inflammasome activation. In vivo, the T6SS played a key role in mediating interleukin 1ß secretion and the survival of mice during C. freundii infection in mice. These findings provide novel insights into the role of T6SS in the pathogenesis of C. freundii.


Assuntos
Citrobacter freundii , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Sistemas de Secreção Tipo VI , Animais , Caspase 1 , Citrobacter freundii/patogenicidade , Inflamassomos/imunologia , Interleucina-1beta , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Piroptose
4.
EMBO J ; 36(17): 2567-2580, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28701483

RESUMO

The role of second messengers in the diversion of cellular processes by pathogens remains poorly studied despite their importance. Among these, Ca2+ virtually regulates all known cell processes, including cytoskeletal reorganization, inflammation, or cell death pathways. Under physiological conditions, cytosolic Ca2+ increases are transient and oscillatory, defining the so-called Ca2+ code that links cell responses to specific Ca2+ oscillatory patterns. During cell invasion, Shigella induces atypical local and global Ca2+ signals. Here, we show that by hydrolyzing phosphatidylinositol-(4,5)bisphosphate, the Shigella type III effector IpgD dampens inositol-(1,4,5)trisphosphate (InsP3) levels. By modifying InsP3 dynamics and diffusion, IpgD favors the elicitation of long-lasting local Ca2+ signals at Shigella invasion sites and converts Shigella-induced global oscillatory responses into erratic responses with atypical dynamics and amplitude. Furthermore, IpgD eventually inhibits InsP3-dependent responses during prolonged infection kinetics. IpgD thus acts as a pathogen regulator of the Ca2+ code implicated in a versatility of cell functions. Consistent with this function, IpgD prevents the Ca2+-dependent activation of calpain, thereby preserving the integrity of cell adhesion structures during the early stages of infection.


Assuntos
Proteínas de Bactérias/metabolismo , Cálcio/metabolismo , Disenteria Bacilar/metabolismo , Interações Hospedeiro-Patógeno , Monoéster Fosfórico Hidrolases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Shigella flexneri/fisiologia , Calpaína/metabolismo , Adesão Celular , Células HeLa , Humanos , Transdução de Sinais
5.
PLoS Pathog ; 15(11): e1008123, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31725806

RESUMO

Adherent Invasive Escherichia coli (AIEC) strains recovered from Crohn's disease lesions survive and multiply within macrophages. A reference strain for this pathovar, AIEC LF82, forms microcolonies within phagolysosomes, an environment that prevents commensal E. coli multiplication. Little is known about the LF82 intracellular growth status, and signals leading to macrophage intra-vacuolar multiplication. We used single-cell analysis, genetic dissection and mathematical models to monitor the growth status and cell cycle regulation of intracellular LF82. We found that within macrophages, bacteria may replicate or undergo non-growing phenotypic switches. This switch results from stringent response firing immediately after uptake by macrophages or at later stages, following genotoxic damage and SOS induction during intracellular replication. Importantly, non-growers resist treatment with various antibiotics. Thus, intracellular challenges induce AIEC LF82 phenotypic heterogeneity and non-growing bacteria that could provide a reservoir for antibiotic-tolerant bacteria responsible for relapsing infections.


Assuntos
Antibacterianos/farmacologia , Doença de Crohn/microbiologia , Farmacorresistência Bacteriana , Infecções por Escherichia coli/microbiologia , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/patogenicidade , Macrófagos/microbiologia , Aderência Bacteriana , Comunicação Celular , Células Cultivadas , Escherichia coli/efeitos dos fármacos , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/genética , Humanos , Macrófagos/efeitos dos fármacos , Resposta SOS em Genética/efeitos dos fármacos
6.
Mol Microbiol ; 105(2): 211-226, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28486768

RESUMO

Nanomachines belonging to the type IV filament (Tff) superfamily serve a variety of cellular functions in prokaryotes, including motility, adhesion, electrical conductance, competence and secretion. The type 2 secretion system (T2SS) Tff member assembles a short filament called pseudopilus that promotes the secretion of folded proteins from the periplasm across the outer membrane of Gram-negative bacteria. A combination of structural, biochemical, imaging, computational and in vivo approaches had led to a working model for the assembled nanomachine. High-resolution cryo-electron microscopy and tomography provided the first view of several homologous Tff nanomachines in the cell envelope and revealed the structure of the outer membrane secretin channel, challenging current models of the overall stoichiometry of the T2SS. In addition, recent insights into exoprotein substrate features and interactions with the T2SS have led to new questions about the dynamics of the system and the role of the plasma membrane in substrate presentation. This micro-review will highlight recent advances in the field of type 2 secretion and discuss approaches that can be used to reach a mechanistic understanding of exoprotein recognition, integration into the machine and secretion.


Assuntos
Sistemas de Secreção Tipo II/metabolismo , Sistemas de Secreção Tipo II/ultraestrutura , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/metabolismo , Sequência de Bases , Microscopia Crioeletrônica/métodos , Bactérias Gram-Negativas/metabolismo , Proteínas de Membrana/metabolismo , Modelos Moleculares , Periplasma/metabolismo , Ligação Proteica , Dobramento de Proteína , Secretina/química , Relação Estrutura-Atividade
7.
Mol Microbiol ; 101(6): 924-41, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27260845

RESUMO

Type II secretion systems (T2SSs) promote secretion of folded proteins playing important roles in nutrient acquisition, adaptation and virulence of Gram-negative bacteria. Protein secretion is associated with the assembly of type 4 pilus (T4P)-like fibres called pseudopili. Initially membrane embedded, pseudopilin and T4 pilin subunits share conserved transmembrane segments containing an invariant Glu residue at the fifth position, E5. Mutations of E5 in major T4 pilins and in PulG, the major pseudopilin of the Klebsiella T2SS abolish fibre assembly and function. Among the four minor pseudopilins, only PulH required E5 for secretion of pullulanase, the substrate of the Pul T2SS. Mass-spectrometry analysis of pili resulting from the co-assembly of PulG(E5A) variant and PulG(WT) ruled out an E5 role in pilin processing and N-methylation. A bacterial two-hybrid analysis revealed interactions of the full-length pseudopilins PulG and PulH with the PulJ-PulI-PulK priming complex and with the assembly factors PulM and PulF. Remarkably, PulG(E5A) and PulH(E5A) variants were defective in interaction with PulM but not with PulF, and co-purification experiments confirmed the E5-dependent interaction between native PulM and PulG. These results reveal the role of E5 in a recruitment step critical for assembly of the functional T2SS, likely relevant to T4P assembly systems.


Assuntos
Proteínas de Fímbrias/metabolismo , Klebsiella/metabolismo , Sistemas de Secreção Tipo II/metabolismo , Sequência de Aminoácidos , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/metabolismo , Glutamina/metabolismo , Glicosídeo Hidrolases/metabolismo , Klebsiella/genética , Klebsiella oxytoca/genética , Klebsiella oxytoca/metabolismo , Dobramento de Proteína
8.
PLoS Pathog ; 11(7): e1005013, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26132339

RESUMO

Type III secretion systems (T3SSs) are specialized macromolecular machines critical for bacterial virulence, and allowing the injection of bacterial effectors into host cells. The T3SS-dependent injection process requires the prior insertion of a protein complex, the translocon, into host cell membranes consisting of two-T3SS hydrophobic proteins, associated with pore-forming activity. In all described T3SS to date, a hydrophilic protein connects one hydrophobic component to the T3SS needle, presumably insuring the continuum between the hollow needle and the translocon. In the case of Enteropathogenic Escherichia coli (EPEC), the hydrophilic component EspA polymerizes into a filament connecting the T3SS needle to the translocon composed of the EspB and EspD hydrophobic proteins. Here, we identify EspA and EspD as targets of EspC, a serine protease autotransporter of Enterobacteriaceae (SPATE). We found that in vitro, EspC preferentially targets EspA associated with EspD, but was less efficient at proteolyzing EspA alone. Consistently, we found that EspC did not regulate EspA filaments at the surface of primed bacteria that was devoid of EspD, but controlled the levels of EspD and EspA secreted in vitro or upon cell contact. While still proficient for T3SS-mediated injection of bacterial effectors and cytoskeletal reorganization, an espC mutant showed increased levels of cell-associated EspA and EspD, as well as increased pore formation activity associated with cytotoxicity. EspP from enterohaemorrhagic E. coli (EHEC) also targeted translocator components and its activity was interchangeable with that of EspC, suggesting a common and important function of these SPATEs. These findings reveal a novel regulatory mechanism of T3SS-mediated pore formation and cytotoxicity control during EPEC/EHEC infection.


Assuntos
Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Proteínas de Bactérias/metabolismo , Eletroforese em Gel de Poliacrilamida , Escherichia coli Enteropatogênica/metabolismo , Células HeLa , Humanos , Imuno-Histoquímica , Microscopia Confocal , Mutagênese Sítio-Dirigida
9.
Cell Microbiol ; 17(2): 174-82, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25469430

RESUMO

The actin cytoskeleton is key to the barrier function of epithelial cells, by permitting the establishment and maintenance of cell-cell junctions and cell adhesion to the basal matrix. Actin exists under monomeric and polymerized filamentous form and its polymerization following activation of nucleation promoting factors generates pushing forces, required to propel intracellular microorganisms in the host cell cytosol or for the formation of cell extensions that engulf bacteria. Actin filaments can associate with adhesion receptors at the plasma membrane via cytoskeletal linkers. Membrane anchored to actin filaments are then subjected to the retrograde flow that may pull membrane-bound bacteria inside the cell. To induce its internalization by normally non-phagocytic cells, bacteria need to establish adhesive contacts and trick the cell into apply pulling forces, and/or to generate protrusive forces that deform the membrane surrounding its contact site. In this review, we will focus on recent findings on actin cytoskeleton reorganization within epithelial cells during invasion and cell-to-cell spreading by the enteroinvasive pathogen Shigella, the causative agent of bacillary dysentery.


Assuntos
Citoesqueleto de Actina/metabolismo , Endocitose , Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno , Shigella/crescimento & desenvolvimento
11.
J Cell Sci ; 125(Pt 21): 4999-5004, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22899718

RESUMO

Filopodia are thin cell extensions sensing the environment. They play an essential role during cell migration, cell-cell or cell-matrix adhesion, by initiating contacts and conveying signals to the cell cortex. Pathogenic microorganisms can hijack filopodia to invade cells by inducing their retraction towards the cell body. Because their dynamics depend on a discrete number of actin filaments, filopodia provide a model of choice to study elementary events linked to adhesion and downstream signalling. However, the determinants controlling filopodial sensing are not well characterized. In this study, we used beads functionalized with different ligands that triggered filopodial retraction when in contact with filopodia of epithelial cells. With optical tweezers, we were able to measure forces stalling the retraction of a single filopodium. We found that the filopodial stall force depends on the coating of the bead. Stall forces reached 8 pN for beads coated with the ß1 integrin ligand Yersinia Invasin, whereas retraction was stopped with a higher force of 15 pN when beads were functionalized with carboxyl groups. In all cases, stall forces increased in relation to the density of ligands contacting filopodial tips and were independent of the optical trap stiffness. Unexpectedly, a discrete and small number of Shigella type three secretion systems induced stall forces of 10 pN. These results suggest that the number of receptor-ligand interactions at the filopodial tip determines the maximal retraction force exerted by filopodia but a discrete number of clustered receptors is sufficient to induce high retraction stall forces.


Assuntos
Células Epiteliais/ultraestrutura , Pseudópodes/ultraestrutura , Shigella/fisiologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Sistemas de Secreção Bacterianos , Fenômenos Biomecânicos , Adesão Celular , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Integrina beta1/metabolismo , Ligantes , Microscopia Confocal , Microesferas , Pinças Ópticas , Ligação Proteica , Pseudópodes/microbiologia , Pseudópodes/fisiologia , Análise de Célula Única , Imagem com Lapso de Tempo
12.
Life Sci Alliance ; 7(8)2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38834194

RESUMO

Vinculin is a cytoskeletal linker strengthening cell adhesion. The Shigella IpaA invasion effector binds to vinculin to promote vinculin supra-activation associated with head-domain-mediated oligomerization. Our study investigates the impact of mutations of vinculin D1D2 subdomains' residues predicted to interact with IpaA VBS3. These mutations affected the rate of D1D2 trimer formation with distinct effects on monomer disappearance, consistent with structural modeling of a closed and open D1D2 conformer induced by IpaA. Notably, mutations targeting the closed D1D2 conformer significantly reduced Shigella invasion of host cells as opposed to mutations targeting the open D1D2 conformer and later stages of vinculin head-domain oligomerization. In contrast, all mutations affected the formation of focal adhesions (FAs), supporting the involvement of vinculin supra-activation in this process. Our findings suggest that IpaA-induced vinculin supra-activation primarily reinforces matrix adhesion in infected cells, rather than promoting bacterial invasion. Consistently, shear stress studies pointed to a key role for IpaA-induced vinculin supra-activation in accelerating and strengthening cell-matrix adhesion.


Assuntos
Adesão Celular , Adesões Focais , Vinculina , Vinculina/metabolismo , Vinculina/genética , Humanos , Adesões Focais/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Mutação , Interações Hospedeiro-Patógeno , Células HeLa , Ligação Proteica , Shigella/metabolismo , Shigella/genética , Antígenos de Bactérias/metabolismo , Antígenos de Bactérias/genética , Disenteria Bacilar/microbiologia , Disenteria Bacilar/metabolismo
13.
Cell Rep ; 42(4): 112405, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37071535

RESUMO

Upon activation, vinculin reinforces cytoskeletal anchorage during cell adhesion. Activating ligands classically disrupt intramolecular interactions between the vinculin head and tail domains that bind to actin filaments. Here, we show that Shigella IpaA triggers major allosteric changes in the head domain, leading to vinculin homo-oligomerization. Through the cooperative binding of its three vinculin-binding sites (VBSs), IpaA induces a striking reorientation of the D1 and D2 head subdomains associated with vinculin oligomerization. IpaA thus acts as a catalyst producing vinculin clusters that bundle actin at a distance from the activation site and trigger the formation of highly stable adhesions resisting the action of actin relaxing drugs. Unlike canonical activation, vinculin homo-oligomers induced by IpaA appear to keep a persistent imprint of the activated state in addition to their bundling activity, accounting for stable cell adhesion independent of force transduction and relevant to bacterial invasion.


Assuntos
Proteínas de Bactérias , Shigella , Proteínas de Bactérias/metabolismo , Antígenos de Bactérias/metabolismo , Actinas/metabolismo , Vinculina/metabolismo , Shigella/metabolismo , Ligação Proteica
14.
J Biol Chem ; 286(26): 23214-21, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21525010

RESUMO

Internalization of Shigella into host epithelial cells, where the bacteria replicates and spreads to neighboring cells, requires a type 3 secretion system (T3SS) effector coined IpaA. IpaA binds directly to and activates the cytoskeletal protein vinculin after injection in the host cell cytosol, and this was previously thought to be directed by two amphipathic α-helical vinculin-binding sites (VBS) found in the C-terminal tail domain of IpaA. Here, we report a third VBS, IpaA-VBS3, that is located N-terminal to the other two VBSs of IpaA and show that one IpaA molecule can bind up to three vinculin molecules. Biochemical in vitro Shigella invasion assays and the 1.6 Å crystal structure of the vinculin·IpaA-VBS3 complex showed that IpaA-VBS3 is functionally redundant with the other two IpaA-VBSs in cell invasion and in activating the latent F-actin binding functions of vinculin. Multiple VBSs in IpaA are reminiscent of talin, which harbors 11 VBSs. However, most of the talin VBSs have low affinity and are buried in helix bundles, whereas all three of the VBSs of IpaA are high affinity, readily available, and in close proximity to each other in the IpaA structure. Although deletion of IpaA-VBS3 has no detectable effects on Shigella invasion of epithelial cells, deletion of all three VBSs impaired bacterial invasion to levels found in an ipaA null mutant strain. Thus, IpaA-directed mimicry of talin in activating vinculin occurs through three high affinity VBSs that are essential for Shigella pathogenesis.


Assuntos
Shigella flexneri/química , Shigella flexneri/metabolismo , Vinculina/química , Vinculina/metabolismo , Antígenos de Bactérias , Proteínas de Bactérias , Sítios de Ligação , Cristalografia por Raios X , Disenteria Bacilar/genética , Disenteria Bacilar/metabolismo , Células HeLa , Humanos , Mimetismo Molecular/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Shigella flexneri/genética , Shigella flexneri/patogenicidade , Talina/química , Talina/genética , Talina/metabolismo , Vinculina/genética
15.
Infect Immun ; 80(7): 2548-57, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22526677

RESUMO

Shigella flexneri, the causative agent of bacillary dysentery, induces massive cytoskeletal rearrangement, resulting in its entry into nonphagocytic epithelial cells. The bacterium-engulfing membrane ruffles are formed by polymerizing actin, a process activated through injected bacterial effectors that target host small GTPases and tyrosine kinases. Once inside the host cell, S. flexneri escapes from the endocytic vacuole within minutes to move intra- and intercellularly. We quantified the fluorescence signals from fluorescently tagged host factors that are recruited to the site of pathogen entry and vacuolar escape. Quantitative time lapse fluorescence imaging revealed simultaneous recruitment of polymerizing actin, small GTPases of the Rho family, and tyrosine kinases. In contrast, we found that actin surrounding the vacuole containing bacteria dispersed first from the disassembling membranes, whereas other host factors remained colocalized with the membrane remnants. Furthermore, we found that the disassembly of the membrane remnants took place rapidly, within minutes after bacterial release into the cytoplasm. Superresolution visualization of galectin 3 through photoactivated localization microscopy characterized these remnants as small, specular, patchy structures between 30 and 300 nm in diameter. Using our experimental setup to track the time course of infection, we identified the S. flexneri effector IpgB1 as an accelerator of the infection pace, specifically targeting the entry step, but not vacuolar progression or escape. Together, our studies show that bacterial entry into host cells follows precise kinetics and that this time course can be targeted by the pathogen.


Assuntos
Citoesqueleto/metabolismo , Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno , Shigella flexneri/patogenicidade , Citoplasma/microbiologia , Células HeLa , Humanos , Microscopia de Fluorescência , Fatores de Tempo , Imagem com Lapso de Tempo , Vacúolos/microbiologia
16.
EMBO J ; 27(2): 447-57, 2008 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18188151

RESUMO

Type III secretion (T3S) systems are largely used by pathogenic gram-negative bacteria to inject multiple effectors into eukaryotic cells. Upon cell contact, these bacterial microinjection devices insert two T3S substrates into host cell membranes, forming a so-called 'translocon' that is required for targeting of type III effectors in the cell cytosol. Here, we show that secretion of the translocon component IpaC of invasive Shigella occurs at the level of one bacterial pole during cell invasion. Using IpaC fusions with green fluorescent protein variants (IpaCi), we show that the IpaC cytoplasmic pool localizes at an old or new bacterial pole, where secretion occurs upon T3S activation. Deletions in ipaC identified domains implicated in polar localization. Only polar IpaCi derivatives inhibited T3S, while IpaCi fusions with diffuse cytoplasmic localization had no detectable effect on T3S. Moreover, the deletions that abolished polar localization led to secretion defects when introduced in ipaC. These results indicate that cytoplasmic polar localization directs secretion of IpaC at the pole of Shigella, and may represent a mandatory step for T3S.


Assuntos
Antígenos de Bactérias/metabolismo , Citoplasma/metabolismo , Shigella/metabolismo , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Citoplasma/microbiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Imunoprecipitação , Cinética , Microscopia Confocal , Microscopia de Fluorescência , Mutação , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Shigella/genética , Shigella/crescimento & desenvolvimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
Front Cell Infect Microbiol ; 12: 1012533, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36389142

RESUMO

Shigella, the causative agent of bacillary dysentery, subvert cytoskeletal and trafficking processes to invade and replicate in epithelial cells using an arsenal of bacterial effectors translocated through a type III secretion system. Here, we review the various roles of the type III effector IpgD, initially characterized as phosphatidylinositol 4,5 bisphosphate (PI4,5P2) 4-phosphatase. By decreasing PI4,5P2 levels, IpgD triggers the disassembly of cortical actin filaments required for bacterial invasion and cell migration. PI5P produced by IpgD further stimulates signaling pathways regulating cell survival, macropinosome formation, endosomal trafficking and dampening of immune responses. Recently, IpgD was also found to exhibit phosphotransferase activity leading to PI3,4P2 synthesis adding a new flavor to this multipotent bacterial enzyme. The substrate of IpgD, PI4,5P2 is also the main substrate hydrolyzed by endogenous phospholipases C to produce inositoltriphosphate (InsP3), a major Ca2+ second messenger. Hence, beyond the repertoire of effects associated with the direct diversion of phoshoinositides, IpgD indirectly down-regulates InsP3-mediated Ca2+ release by limiting InsP3 production. Furthermore, IpgD controls the intracellular lifestyle of Shigella promoting Rab8/11 -dependent recruitment of the exocyst at macropinosomes to remove damaged vacuolar membrane remnants and promote bacterial cytosolic escape. IpgD thus emerges as a key bacterial effector for the remodeling of host cell membranes.


Assuntos
Disenteria Bacilar , Shigella , Humanos , Fosfatidilinositóis/metabolismo , Shigella flexneri/metabolismo , Disenteria Bacilar/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo
18.
Sci Adv ; 8(45): eabo1461, 2022 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-36351022

RESUMO

Mechanosensing is an integral part of many physiological processes including stem cell differentiation, fibrosis, and cancer progression. Two major mechanosensing systems-focal adhesions and mechanosensitive ion channels-can convert mechanical features of the microenvironment into biochemical signals. We report here unexpectedly that the mechanosensitive calcium-permeable channel Piezo1, previously perceived to be diffusive on plasma membranes, binds to matrix adhesions in a force-dependent manner, promoting cell spreading, adhesion dynamics, and calcium entry in normal but not in most cancer cells tested except some glioblastoma lines. A linker domain in Piezo1 is needed for binding to adhesions, and overexpression of the domain blocks Piezo1 binding to adhesions, decreasing adhesion size and cell spread area. Thus, we suggest that Piezo1 is a previously unidentified component of focal adhesions in nontransformed cells that catalyzes adhesion maturation and growth through force-dependent calcium signaling, but this function is absent in most cancer cells.

19.
Nat Cell Biol ; 5(8): 720-6, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12844145

RESUMO

Shigella flexneri, the causative agent of bacillar dystentery, invades the colonic mucosa where it elicits an intense inflammatory reaction responsible for destruction of the epithelium. During cell invasion, contact with host cells activates the type-III secretion of the Shigella IpaB and IpaC proteins. IpaB and IpaC are inserted into host cell plasma membranes and trigger initial signals that result in actin polymerization, while allowing cytosolic access of other bacterial effectors that further reorganize the cytoskeleton. After internalization, Shigella moves intracellularly and forms protrusions that infect neighbouring cells, promoting bacterial dissemination across the epithelium. Here, we show that during cell invasion, Shigella induces transient peaks in intracellular calcium concentration that are dependent on a functional type-III secretory apparatus. In addition, Shigella invasion induces the opening of Connexin 26 (Cx26) hemichannels in an actin- and phospholipase-C-dependent manner, allowing release of ATP into the medium. The released ATP, in turn, increases bacterial invasion and spreading, as well as calcium signalling induced by Shigella. These results provide evidence that pathogen-induced opening of connexin channels promotes signalling events that favour bacterial invasion and dissemination.


Assuntos
Comunicação Celular/fisiologia , Conexinas/metabolismo , Células Epiteliais/microbiologia , Mucosa Intestinal/metabolismo , Shigella flexneri/fisiologia , Actinas/metabolismo , Trifosfato de Adenosina/metabolismo , Sinalização do Cálcio/fisiologia , Conexina 26 , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células HeLa , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia
20.
J Cell Biol ; 175(3): 465-75, 2006 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-17088427

RESUMO

Shigella flexneri, the causative agent of bacillary dysentery, injects invasin proteins through a type III secretion apparatus upon contacting the host cell, which triggers pathogen internalization. The invasin IpaA is essential for S. flexneri pathogenesis and binds to the cytoskeletal protein vinculin to facilitate host cell entry. We report that IpaA harbors two vinculin-binding sites (VBSs) within its C-terminal domain that bind to and activate vinculin in a mutually exclusive fashion. Only the highest affinity C-terminal IpaA VBS is necessary for efficient entry and cell-cell spread of S. flexneri, whereas the lower affinity VBS appears to contribute to vinculin recruitment at entry foci of the pathogen. Finally, the crystal structures of vinculin in complex with the VBSs of IpaA reveal the mechanism by which IpaA subverts vinculin's functions, where S. flexneri utilizes a remarkable level of molecular mimicry of the talin-vinculin interaction to activate vinculin. Mimicry of vinculin's interactions may therefore be a general mechanism applied by pathogens to infect the host cell.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Disenteria Bacilar/metabolismo , Mimetismo Molecular , Shigella flexneri/patogenicidade , Vinculina/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Células CACO-2 , Cristalização , Citoesqueleto/metabolismo , Citoesqueleto/microbiologia , Células HeLa , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Conformação Proteica , Transporte Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Shigella flexneri/metabolismo , Talina/metabolismo , Vinculina/química , Vinculina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA