Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.048
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 17(12): e1009586, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34941903

RESUMO

The cell envelope is essential for viability in all domains of life. It retains enzymes and substrates within a confined space while providing a protective barrier to the external environment. Destabilising the envelope of bacterial pathogens is a common strategy employed by antimicrobial treatment. However, even in one of the best studied organisms, Escherichia coli, there remain gaps in our understanding of how the synthesis of the successive layers of the cell envelope are coordinated during growth and cell division. Here, we used a whole-genome phenotypic screen to identify mutants with a defective cell envelope. We report that loss of yhcB, a conserved gene of unknown function, results in loss of envelope stability, increased cell permeability and dysregulated control of cell size. Using whole genome transposon mutagenesis strategies, we report the comprehensive genetic interaction network of yhcB, revealing all genes with a synthetic negative and a synthetic positive relationship. These genes include those previously reported to have a role in cell envelope biogenesis. Surprisingly, we identified genes previously annotated as essential that became non-essential in a ΔyhcB background. Subsequent analyses suggest that YhcB functions at the junction of several envelope biosynthetic pathways coordinating the spatiotemporal growth of the cell, highlighting YhcB as an as yet unexplored antimicrobial target.


Assuntos
Parede Celular/genética , Proteínas de Escherichia coli/genética , Lipopolissacarídeos/genética , Oxirredutases/genética , Peptidoglicano/genética , Divisão Celular/genética , Membrana Celular/genética , Membrana Celular/microbiologia , Parede Celular/microbiologia , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/genética , Lipopolissacarídeos/biossíntese , Mutagênese , Fosfolipídeos/biossíntese , Fosfolipídeos/genética
2.
Proc Natl Acad Sci U S A ; 117(5): 2634-2644, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31964834

RESUMO

During invasion of host cells, Chlamydia pneumoniae secretes the effector protein CPn0678, which facilitates internalization of the pathogen by remodeling the target cell's plasma membrane and recruiting sorting nexin 9 (SNX9), a central multifunctional endocytic scaffold protein. We show here that the strongly amphipathic N-terminal helix of CPn0678 mediates binding to phospholipids in both the plasma membrane and synthetic membranes, and is sufficient to induce extensive membrane tubulations. CPn0678 interacts via its conserved C-terminal polyproline sequence with the Src homology 3 domain of SNX9. Thus, SNX9 is found at bacterial entry sites, where C. pneumoniae is internalized via EGFR-mediated endocytosis. Moreover, depletion of human SNX9 significantly reduces internalization, whereas ectopic overexpression of CPn0678-GFP results in a dominant-negative effect on endocytotic processes in general, leading to the uptake of fewer chlamydial elementary bodies and diminished turnover of EGFR. Thus, CPn0678 is an early effector involved in regulating the endocytosis of C. pneumoniae in an EGFR- and SNX9-dependent manner.


Assuntos
Membrana Celular/química , Infecções por Chlamydia/microbiologia , Chlamydia/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Infecções por Chlamydia/genética , Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/fisiopatologia , Endocitose , Interações Hospedeiro-Patógeno , Humanos , Nexinas de Classificação/genética , Nexinas de Classificação/metabolismo
3.
PLoS Pathog ; 16(11): e1009016, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33216805

RESUMO

The opportunistic pathogen Streptococcus pneumoniae has dual lifestyles: one of an asymptomatic colonizer in the human nasopharynx and the other of a deadly pathogen invading sterile host compartments. The latter triggers an overwhelming inflammatory response, partly driven via pore forming activity of the cholesterol dependent cytolysin (CDC), pneumolysin. Although pneumolysin-induced inflammation drives person-to-person transmission from nasopharynx, the primary reservoir for pneumococcus, it also contributes to high mortality rates, creating a bottleneck that hampers widespread bacterial dissemination, thus acting as a double-edged sword. Serotype 1 ST306, a widespread pneumococcal clone, harbours a non-hemolytic variant of pneumolysin (Ply-NH). Performing crystal structure analysis of Ply-NH, we identified Y150H and T172I as key substitutions responsible for loss of its pore forming activity. We uncovered a novel inter-molecular cation-π interaction, governing formation of the transmembrane ß-hairpins (TMH) in the pore state of Ply, which can be extended to other CDCs. H150 in Ply-NH disrupts this interaction, while I172 provides structural rigidity to domain-3, through hydrophobic interactions, inhibiting TMH formation. Loss of pore forming activity enabled improved cellular invasion and autophagy evasion, promoting an atypical intracellular lifestyle for pneumococcus, a finding that was corroborated in in vivo infection models. Attenuation of inflammatory responses and tissue damage promoted tolerance of Ply-NH-expressing pneumococcus in the lower respiratory tract. Adoption of this altered lifestyle may be necessary for ST306 due to its limited nasopharyngeal carriage, with Ply-NH, aided partly by loss of its pore forming ability, facilitating a benign association of SPN in an alternative, intracellular host niche.


Assuntos
Adaptação Fisiológica , Inflamação/microbiologia , Mutação com Perda de Função , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/fisiologia , Estreptolisinas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Membrana Celular/microbiologia , Colesterol/metabolismo , Citoplasma/microbiologia , Feminino , Humanos , Camundongos , Modelos Estruturais , Perforina/genética , Perforina/metabolismo , Alinhamento de Sequência , Streptococcus pneumoniae/genética , Estreptolisinas/genética
4.
PLoS Pathog ; 16(8): e1008734, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32853279

RESUMO

AMPylation, the post-translational modification with adenosine monophosphate (AMP), is catalyzed by effector proteins from a variety of pathogens. Legionella pneumophila is thus far the only known pathogen that, in addition to encoding an AMPylase (SidM/DrrA), also encodes a deAMPylase, called SidD, that reverses SidM-mediated AMPylation of the vesicle transport GTPase Rab1. DeAMPylation is catalyzed by the N-terminal phosphatase-like domain of SidD. Here, we determined the crystal structure of full length SidD including the uncharacterized C-terminal domain (CTD). A flexible loop rich in aromatic residues within the CTD was required to target SidD to model membranes in vitro and to the Golgi apparatus within mammalian cells. Deletion of the loop (Δloop) or substitution of its aromatic phenylalanine residues rendered SidD cytosolic, showing that the hydrophobic loop is the primary membrane-targeting determinant of SidD. Notably, deletion of the two terminal alpha helices resulted in a CTD variant incapable of discriminating between membranes of different composition. Moreover, a L. pneumophila strain producing SidDΔloop phenocopied a L. pneumophila ΔsidD strain during growth in mouse macrophages and displayed prolonged co-localization of AMPylated Rab1 with LCVs, thus revealing that membrane targeting of SidD via its CTD is a critical prerequisite for its ability to catalyze Rab1 deAMPylation during L. pneumophila infection.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Membrana Celular/microbiologia , Legionella pneumophila/enzimologia , Doença dos Legionários/microbiologia , Monofosfato de Adenosina/metabolismo , Animais , Proteínas de Bactérias/genética , Feminino , Complexo de Golgi/metabolismo , Humanos , Legionella pneumophila/química , Legionella pneumophila/genética , Camundongos , Domínios Proteicos
5.
Nat Immunol ; 11(1): 55-62, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19898471

RESUMO

Autophagy is emerging as a crucial defense mechanism against bacteria, but the host intracellular sensors responsible for inducing autophagy in response to bacterial infection remain unknown. Here we demonstrated that the intracellular sensors Nod1 and Nod2 are critical for the autophagic response to invasive bacteria. By a mechanism independent of the adaptor RIP2 and transcription factor NF-kappaB, Nod1 and Nod2 recruited the autophagy protein ATG16L1 to the plasma membrane at the bacterial entry site. In cells homozygous for the Crohn's disease-associated NOD2 frameshift mutation, mutant Nod2 failed to recruit ATG16L1 to the plasma membrane and wrapping of invading bacteria by autophagosomes was impaired. Our results link bacterial sensing by Nod proteins to the induction of autophagy and provide a functional link between Nod2 and ATG16L1, which are encoded by two of the most important genes associated with Crohn's disease.


Assuntos
Autofagia , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Animais , Proteínas Relacionadas à Autofagia , Bactérias/metabolismo , Proteínas de Transporte/genética , Linhagem Celular , Membrana Celular/microbiologia , Membrana Celular/ultraestrutura , Células Cultivadas , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica , Microscopia de Fluorescência , Mutação , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD2/genética , Transfecção
6.
Proc Natl Acad Sci U S A ; 116(51): 25649-25658, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31757855

RESUMO

Phthiocerol dimycocerosate (DIM) is a major virulence factor of the pathogen Mycobacterium tuberculosis (Mtb). While this lipid promotes the entry of Mtb into macrophages, which occurs via phagocytosis, its molecular mechanism of action is unknown. Here, we combined biophysical, cell biology, and modeling approaches to reveal the molecular mechanism of DIM action on macrophage membranes leading to the first step of Mtb infection. Matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry showed that DIM molecules are transferred from the Mtb envelope to macrophage membranes during infection. Multiscale molecular modeling and 31P-NMR experiments revealed that DIM adopts a conical shape in membranes and aggregates in the stalks formed between 2 opposing lipid bilayers. Infection of macrophages pretreated with lipids of various shapes uncovered a general role for conical lipids in promoting phagocytosis. Taken together, these results reveal how the molecular shape of a mycobacterial lipid can modulate the biological response of macrophages.


Assuntos
Lipídeos/química , Macrófagos/microbiologia , Mycobacterium tuberculosis , Tuberculose/microbiologia , Linhagem Celular , Membrana Celular/química , Membrana Celular/microbiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Macrófagos/química , Simulação de Dinâmica Molecular , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/patogenicidade , Mycobacterium tuberculosis/fisiologia , Ressonância Magnética Nuclear Biomolecular
7.
J Cell Sci ; 132(9)2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-31040222

RESUMO

Septins are widely recognized as a component of the cytoskeleton that is essential for cell division, and new work has shown that septins can recognise cell shape by assembling into filaments on membrane regions that display micrometer-scale curvature (e.g. at the cytokinetic furrow). Moreover, infection biology studies have illuminated important roles for septins in mediating the outcome of host-microbe interactions. In this Review, we discuss a selection of mechanistic insights recently gained from studying three infection paradigms: the rice blast fungus Magnaporthe oryzae, the poxvirus family member vaccinia virus and the Gram-negative bacterium Shigella flexneri These studies have respectively discovered that higher-order septin assemblies enable fungal invasion into plant cells, entrap viral particles at the plasma membrane and recognize dividing bacterial cells for delivery to lysosomes. Collectively, these insights illustrate how studying septin biology during microbial infection can provide fundamental advances in both cell and infection biology, and suggest new concepts underlying infection control.


Assuntos
Interações entre Hospedeiro e Microrganismos/fisiologia , Oryza/microbiologia , Oryza/virologia , Doenças das Plantas , Septinas , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Citoesqueleto/metabolismo , Citoesqueleto/microbiologia , Magnaporthe/patogenicidade , Doenças das Plantas/microbiologia , Doenças das Plantas/virologia , Septinas/biossíntese , Septinas/química , Septinas/genética , Septinas/metabolismo , Shigella flexneri/patogenicidade , Vaccinia virus/patogenicidade
8.
PLoS Pathog ; 15(6): e1007851, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31242273

RESUMO

Enteropathogenic E. coli (EPEC) is an extracellular diarrheagenic human pathogen which infects the apical plasma membrane of the small intestinal enterocytes. EPEC utilizes a type III secretion system to translocate bacterial effector proteins into its epithelial hosts. This activity, which subverts numerous signaling and membrane trafficking pathways in the infected cells, is thought to contribute to pathogen virulence. The molecular and cellular mechanisms underlying these events are not well understood. We investigated the mode by which EPEC effectors hijack endosomes to modulate endocytosis, recycling and transcytosis in epithelial host cells. To this end, we developed a flow cytometry-based assay and imaging techniques to track endosomal dynamics and membrane cargo trafficking in the infected cells. We show that type-III secreted components prompt the recruitment of clathrin (clathrin and AP2), early (Rab5a and EEA1) and recycling (Rab4a, Rab11a, Rab11b, FIP2, Myo5b) endocytic machineries to peripheral plasma membrane infection sites. Protein cargoes, e.g. transferrin receptors, ß1 integrins and aquaporins, which exploit the endocytic pathways mediated by these machineries, were also found to be recruited to these sites. Moreover, the endosomes and cargo recruitment to infection sites correlated with an increase in cargo endocytic turnover (i.e. endocytosis and recycling) and transcytosis to the infected plasma membrane. The hijacking of endosomes and associated endocytic activities depended on the translocated EspF and Map effectors in non-polarized epithelial cells, and mostly on EspF in polarized epithelial cells. These data suggest a model whereby EPEC effectors hijack endosomal recycling mechanisms to mislocalize and concentrate host plasma membrane proteins in endosomes and in the apically infected plasma membrane. We hypothesize that these activities contribute to bacterial colonization and virulence.


Assuntos
Membrana Celular/metabolismo , Endocitose , Endossomos/metabolismo , Escherichia coli Enteropatogênica/metabolismo , Infecções por Escherichia coli/metabolismo , Proteínas de Membrana/metabolismo , Membrana Celular/microbiologia , Membrana Celular/patologia , Endossomos/microbiologia , Endossomos/patologia , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/patologia , Células HeLa , Humanos
9.
PLoS Pathog ; 15(7): e1007959, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31339948

RESUMO

The enteric bacterial pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium), utilizes two type III secretion systems (T3SSs) to invade host cells, survive and replicate intracellularly. T3SS1 and its dedicated effector proteins are required for bacterial entry into non-phagocytic cells and establishment and trafficking of the nascent Salmonella-containing vacuole (SCV). Here we identify the first T3SS1 effector required to maintain the integrity of the nascent SCV as SopF. SopF associates with host cell membranes, either when translocated by bacteria or ectopically expressed. Recombinant SopF binds to multiple phosphoinositides in protein-lipid overlays, suggesting that it targets eukaryotic cell membranes via phospholipid interactions. In yeast, the subcellular localization of SopF is dependent on the activity of Mss4, a phosphatidylinositol 4-phosphate 5-kinase that generates PI(4,5)P2 from PI(4)P, indicating that membrane recruitment of SopF requires specific phospholipids. Ectopically expressed SopF partially colocalizes with specific phosphoinositide pools present on the plasma membrane in mammalian cells and with cytoskeletal-associated markers at the leading edge of cells. Translocated SopF concentrates on plasma membrane ruffles and around intracellular bacteria, presumably on the SCV. SopF is not required for bacterial invasion of non-phagocytic cells but is required for maintenance of the internalization vacuole membrane as infection with a S. Typhimurium ΔsopF mutant led to increased lysis of the SCV compared to wild type bacteria. Our structure-function analysis shows that the carboxy-terminal seven amino acids of SopF are essential for its membrane association in host cells and to promote SCV membrane stability. We also describe that SopF and another T3SS1 effector, SopB, act antagonistically to modulate nascent SCV membrane dynamics. In summary, our study highlights that a delicate balance of type III effector activities regulates the stability of the Salmonella internalization vacuole.


Assuntos
Salmonella typhimurium/fisiologia , Sistemas de Secreção Tipo III/fisiologia , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Células HeLa , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Camundongos , Fosfatidilinositóis/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidade , Sistemas de Secreção Tipo III/química , Sistemas de Secreção Tipo III/genética , Vacúolos/metabolismo , Vacúolos/microbiologia
10.
Infect Immun ; 88(2)2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31740527

RESUMO

The mechanisms by which interferon gamma (IFN-γ) controls the replication of cytosolic pathogens independent of responses, such as the generation of reactive oxygen species/reactive nitrogen species (ROS/RNS), have not been fully elucidated. In the current study, we developed a model using Francisella tularensis, the causative agent of tularemia, in which pathways triggered by IFN-γ commonly associated with bacterial control were not required. Using this model, we demonstrated that IFN-γ-mediated production of itaconate and its ability to impair host mitochondrial function, independent of activity on the pathogen, were central for the restriction of bacterial replication in vitro and in vivo We then demonstrate that IFN-γ-driven itaconate production was dispensable, as directly targeting complex II using cell membrane-permeable metabolites also controlled infection. Together, these findings show that while reprogramming of mitochondrial metabolism is a key factor in IFN-γ control of intracellular bacteria, the development of antimicrobial strategies based on targeting host mitochondrial metabolism independent of this cytokine may be an effective therapeutic approach.


Assuntos
Francisella tularensis/efeitos dos fármacos , Interferon gama/farmacologia , Mitocôndrias/efeitos dos fármacos , Animais , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Citosol/metabolismo , Citosol/microbiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Mitocôndrias/microbiologia , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Succinatos/farmacologia , Tularemia/tratamento farmacológico , Tularemia/metabolismo , Tularemia/microbiologia
11.
Microbiology (Reading) ; 166(10): 947-965, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32886602

RESUMO

Bacterial flagella have many established roles beyond swimming motility. Despite clear evidence of flagella-dependent adherence, the specificity of the ligands and mechanisms of binding are still debated. In this study, the molecular basis of Escherichia coli O157:H7 and Salmonella enterica serovar Typhimurium flagella binding to epithelial cell cultures was investigated. Flagella interactions with host cell surfaces were intimate and crossed cellular boundaries as demarcated by actin and membrane labelling. Scanning electron microscopy revealed flagella disappearing into cellular surfaces and transmission electron microscopy of S. Typhiumurium indicated host membrane deformation and disruption in proximity to flagella. Motor mutants of E. coli O157:H7 and S. Typhimurium caused reduced haemolysis compared to wild-type, indicating that membrane disruption was in part due to flagella rotation. Flagella from E. coli O157 (H7), EPEC O127 (H6) and S. Typhimurium (P1 and P2 flagella) were shown to bind to purified intracellular components of the actin cytoskeleton and directly increase in vitro actin polymerization rates. We propose that flagella interactions with host cell membranes and cytoskeletal components may help prime intimate attachment and invasion for E. coli O157:H7 and S. Typhimurium, respectively.


Assuntos
Membrana Celular/microbiologia , Citoesqueleto/metabolismo , Escherichia coli O157/fisiologia , Flagelos/metabolismo , Salmonella typhimurium/fisiologia , Actinas/química , Actinas/metabolismo , Actinas/ultraestrutura , Animais , Aderência Bacteriana , Membrana Celular/metabolismo , Membrana Celular/patologia , Membrana Celular/ultraestrutura , Células Cultivadas , Citoesqueleto/ultraestrutura , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Flagelos/genética , Flagelos/ultraestrutura , Interações Hospedeiro-Patógeno , Humanos , Microscopia Eletrônica , Mutação , Polimerização , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo
12.
Cell Microbiol ; 21(4): e13016, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30740852

RESUMO

Membrane bilayers of eukaryotic cells are an amalgam of lipids and proteins that distinguish organelles and compartmentalise cellular functions. The mammalian cell has evolved mechanisms to sense membrane tension or damage and respond as needed. In the case of the plasma membrane and phagosomal membrane, these bilayers act as a barrier to microorganisms and are a conduit by which the host interacts with pathogens, including fungi such as Candida, Cryptococcus, Aspergillus, or Histoplasma species. Due to their size, morphological flexibility, ability to produce long filaments, secrete pathogenicity factors, and their potential to replicate within the phagosome, fungi can assault host membranes in a variety of physical and biochemical ways. In addition, the recent discovery of a fungal pore-forming peptide toxin further highlights the importance of membrane biology in the outcomes between host and fungal cells. In this review, we discuss the apparent "stretching" of membranes as a sophisticated biological response and the role of vesicular transport in combating membrane stress and damage. We also review the known pathogenicity factors and physical properties of fungal pathogens in the context of host membranes and discuss how this may contribute to pathogenic interactions between fungal and host cells.


Assuntos
Membrana Celular/metabolismo , Membrana Celular/microbiologia , Fungos/patogenicidade , Candida/patogenicidade , Cryptococcus/patogenicidade , Fagossomos/metabolismo
13.
Nature ; 509(7499): 230-4, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24739967

RESUMO

Efferocytosis, the process by which dying or dead cells are removed by phagocytosis, has an important role in development, tissue homeostasis and innate immunity. Efferocytosis is mediated, in part, by receptors that bind to exofacial phosphatidylserine (PS) on cells or cellular debris after loss of plasma membrane asymmetry. Here we show that a bacterial pathogen, Listeria monocytogenes, can exploit efferocytosis to promote cell-to-cell spread during infection. These bacteria can escape the phagosome in host cells by using the pore-forming toxin listeriolysin O (LLO) and two phospholipase C enzymes. Expression of the cell surface protein ActA allows L. monocytogenes to activate host actin regulatory factors and undergo actin-based motility in the cytosol, eventually leading to formation of actin-rich protrusions at the cell surface. Here we show that protrusion formation is associated with plasma membrane damage due to LLO's pore-forming activity. LLO also promotes the release of bacteria-containing protrusions from the host cell, generating membrane-derived vesicles with exofacial PS. The PS-binding receptor TIM-4 (encoded by the Timd4 gene) contributes to efficient cell-to-cell spread by L. monocytogenes in macrophages in vitro and growth of these bacteria is impaired in Timd4(-/-) mice. Thus, L. monocytogenes promotes its dissemination in a host by exploiting efferocytosis. Our results indicate that PS-targeted therapeutics may be useful in the fight against infections by L. monocytogenes and other bacteria that use similar strategies of cell-to-cell spread during infection.


Assuntos
Extensões da Superfície Celular/microbiologia , Listeria monocytogenes/fisiologia , Fagocitose , Actinas/metabolismo , Animais , Toxinas Bacterianas/metabolismo , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Membrana Celular/patologia , Extensões da Superfície Celular/metabolismo , Citoplasma/metabolismo , Citoplasma/microbiologia , Feminino , Células HeLa , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Listeria monocytogenes/patogenicidade , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas de Membrana/metabolismo , Camundongos , Fagossomos/metabolismo , Fagossomos/microbiologia , Fosfatidilserinas/metabolismo , Fosfolipases Tipo C/metabolismo , Vacúolos/metabolismo , Vacúolos/microbiologia
14.
Cell Mol Life Sci ; 76(20): 4165-4178, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31076805

RESUMO

Efficient cell-to-cell transfer of Listeria monocytogenes (L. monocytogenes) requires the proper formation of actin-rich membrane protrusions. To date, only the host proteins ezrin, the binding partner of ezrin, CD44, as well as cyclophilin A (CypA) have been identified as crucial components for L. monocytogenes membrane protrusion stabilization and, thus, efficient cell-to-cell movement of the microbes. Here, we examine the classical binding partner of CypA, CD147, and find that this membrane protein is also hijacked by the bacteria for their cellular dissemination. CD147 is enriched at the plasma membrane surrounding the membrane protrusions as well as the resulting invaginations generated in neighboring cells. In cells depleted of CD147, these actin-rich structures appear similar to those generated in CypA depleted cells as they are significantly shorter and more contorted as compared to their straighter counterparts formed in wild-type control cells. The presence of malformed membrane protrusions hampers the ability of L. monocytogenes to efficiently disseminate from CD147-depleted cells. Our findings uncover another important host protein needed for L. monocytogenes membrane protrusion formation and efficient microbial dissemination.


Assuntos
Basigina/genética , Membrana Celular/microbiologia , Interações Hospedeiro-Patógeno/genética , Listeria monocytogenes/fisiologia , Shigella flexneri/fisiologia , Células A549 , Actinas/genética , Actinas/metabolismo , Animais , Basigina/antagonistas & inibidores , Basigina/metabolismo , Células CACO-2 , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Ciclofilina A/deficiência , Ciclofilina A/genética , Endocitose , Fibroblastos/microbiologia , Fibroblastos/ultraestrutura , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Listeria monocytogenes/patogenicidade , Listeria monocytogenes/ultraestrutura , Camundongos , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Shigella flexneri/patogenicidade , Shigella flexneri/ultraestrutura , Transdução de Sinais
15.
Adv Exp Med Biol ; 1267: 101-115, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32894479

RESUMO

Pathogenic bacteria colonize or disseminate into cells and tissues by inducing large-scale remodeling of host membranes. The physical phenomena underpinning these massive membrane extension and deformation are poorly understood. Invasive strategies of pathogens have been recently enriched by the description of a spectacular mode of opening of large transendothelial cell macroaperture (TEM) tunnels correlated to the dissemination of EDIN-producing strains of Staphylococcus aureus via a hematogenous route or to the induction of gelatinous edema triggered by the edema toxin from Bacillus anthracis. Remarkably, these highly dynamic tunnels close rapidly after they reach a maximal size. Opening and closure of TEMs in cells lasts for hours without inducing endothelial cell death. Multidisciplinary studies have started to provide a broader perspective of both the molecular determinants controlling cytoskeleton organization at newly curved membranes generated by the opening of TEMs and the physical processes controlling the dynamics of these tunnels. Here we discuss the analogy between the opening of TEM tunnels and the physical principles of dewetting, stemming from a parallel between membrane tension and surface tension. This analogy provides a broad framework to investigate biophysical constraints in cell membrane dynamics and their diversion by certain invasive microbial agents.


Assuntos
Bactérias/patogenicidade , Membrana Celular/microbiologia , Membrana Celular/patologia , Células Endoteliais/microbiologia , Células Endoteliais/patologia , Molhabilidade , Membrana Celular/metabolismo , Edema/metabolismo , Edema/microbiologia , Edema/patologia , Células Endoteliais/metabolismo , Humanos , Tensão Superficial
16.
Molecules ; 25(23)2020 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-33291521

RESUMO

In our previous study, temporin-GHaR (GHaR) showed potent antimicrobial activity with strong hemolytic toxicity. To overcome its weakness, we designed GHaR6R, GHaR7R, GHaR8R, GHaR9R, and GHaR9W by changing the number of positive charges and the hydrophobic surface of GHaR. With the exception of GHaR7R, the hemolytic toxicity of the derived peptides had been reduced, and the antimicrobial activities remained close to the parent peptide (except for GHaR9R). GHaR6R, GHaR7R, GHaR8R, and GHaR9W exhibited a great bactericidal effect on Streptococcus mutans (S. mutans), which is one of the main pathogens causing dental caries. According to the membrane permeation and scanning electron microscope (SEM) analysis, these derived peptides targeted to the cell membranes of planktonic bacteria, contributing to the disruption of the membrane integrity and leakage of the intracellular contents. Moreover, they inhibited the formation of biofilms and eradicated the mature biofilms of S. mutans. Compared with GHaR7R, the derived peptides showed less cytotoxicity to human oral epithelial cells (HOECs). The derived peptides are expected to be the molecular templates for designing antibacterial agents to prevent dental caries.


Assuntos
Anti-Infecciosos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Biofilmes/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Peptídeos/farmacologia , Streptococcus mutans/efeitos dos fármacos , Membrana Celular/microbiologia , Cárie Dentária/microbiologia , Células Epiteliais/microbiologia , Eritrócitos/microbiologia , Humanos , Testes de Sensibilidade Microbiana/métodos
17.
J Infect Dis ; 219(1): 145-153, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29733369

RESUMO

Background: Listeria generate actin-rich tubular protrusions at the plasma membrane that propel the bacteria into neighboring cells. The precise molecular mechanisms governing the formation of these protrusions remain poorly defined. Methods: In this study, we demonstrate that the prolyl cis-trans isomerase (PPIase) cyclophilin A (CypA) is hijacked by Listeria at membrane protrusions used for cell-to-cell spreading. Results: Cyclophilin A localizes within the F-actin of these structures and is crucial for their proper formation, as cells depleted of CypA have extended actin-rich structures that are misshaped and are collapsed due to changes within the F-actin network. The lack of structural integrity within the Listeria membrane protrusions hampers the microbes from spreading from CypA null cells. Conclusions: Our results demonstrate a crucial role for CypA during Listeria infections.


Assuntos
Extensões da Superfície Celular/metabolismo , Extensões da Superfície Celular/microbiologia , Ciclofilina A/metabolismo , Listeria/metabolismo , Listeriose/metabolismo , Células A549 , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actinas/ultraestrutura , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Extensões da Superfície Celular/ultraestrutura , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células HeLa , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Listeria/patogenicidade , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidade , Peptidilprolil Isomerase/metabolismo
18.
J Infect Dis ; 220(3): 494-504, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-30938438

RESUMO

Daptomycin resistance in enterococci is often mediated by the LiaFSR system, which orchestrates the cell membrane stress response. Activation of LiaFSR through the response regulator LiaR generates major changes in cell membrane function and architecture (membrane adaptive response), permitting the organism to survive the antibiotic attack. Here, using a laboratory strain of Enterococcus faecalis, we developed a novel Caenorhabditis elegans model of daptomycin therapy and showed that disrupting LiaR-mediated cell membrane adaptation restores the in vivo activity of daptomycin. The LiaR effect was also seen in a clinical strain of daptomycin-resistant Enterococcus faecium, using a murine model of peritonitis. Furthermore, alteration of the cell membrane response increased the ability of human polymorphonuclear neutrophils to readily clear both E. faecalis and multidrug-resistant E. faecium. Our results provide proof of concept that targeting the cell membrane adaptive response restores the in vivo activity of antibiotics, prevents resistance, and enhances the ability of the innate immune system to kill infecting bacteria.


Assuntos
Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecium/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Neutrófilos/efeitos dos fármacos , Animais , Proteínas de Bactérias , Membrana Celular/microbiologia , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Camundongos , Testes de Sensibilidade Microbiana/métodos , Neutrófilos/microbiologia
19.
Semin Cell Dev Biol ; 72: 124-132, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28647534

RESUMO

The membrane attack complex (MAC) is the pore-forming toxin of the complement system, a relatively early evolutionary acquisition that confers upon complement the capacity to directly kill pathogens. The MAC is more than just a bactericidal missile, having the capacity when formed on self-cells to initiate a host of cell activation events that can have profound consequences for tissue homeostasis in the face of infection or injury. Although the capacity of complement to directly kill pathogens has been recognised for over a century, and the pore-forming killing mechanism for at least 50 years, there remains considerable uncertainty regarding precisely how MAC mediates its killing and cell activation activities. A recent burst of new information on MAC structure provides context and opportunity to re-assess the ways in which MAC kills bacteria and modulates cell functions. In this brief review we will describe key aspects of MAC evolution, function and structure and seek to use the new structural information to better explain how the MAC works.


Assuntos
Bactérias/imunologia , Infecções Bacterianas/imunologia , Membrana Celular/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Animais , Bactérias/classificação , Infecções Bacterianas/microbiologia , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/ultraestrutura , Humanos , Microscopia Eletrônica , Modelos Moleculares , Conformação Proteica
20.
J Biol Chem ; 293(23): 8982-8993, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29685888

RESUMO

Many Gram-negative bacterial pathogens use a type III secretion system to infect eukaryotic cells. The injection of bacterial toxins or protein effectors via this system is accomplished through a plasma membrane channel formed by two bacterial proteins, termed translocators, whose assembly and membrane-insertion mechanisms are currently unclear. Here, using purified proteins we demonstrate that the translocators PopB and PopD in Pseudomonas aeruginosa assemble heterodimers in membranes, leading to stably inserted hetero-complexes. Using site-directed fluorescence labeling with an environment-sensitive probe, we found that hydrophobic segments in PopD anchor the translocator to the membrane, but without adopting a typical transmembrane orientation. A fluorescence dual-quenching assay revealed that the presence of PopB changes the conformation adopted by PopD segments in membranes. Furthermore, analysis of PopD's interaction with human cell membranes revealed that PopD adopts a distinctive conformation when PopB is present. An N-terminal region of PopD is only exposed to the host cytosol when PopB is present. We conclude that PopB assists with the proper insertion of PopD in cell membranes, required for the formation of a functional translocon and host infection.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiologia , Sistemas de Secreção Tipo III/metabolismo , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Células HeLa , Humanos , Interações Hidrofóbicas e Hidrofílicas , Multimerização Proteica , Infecções por Pseudomonas/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA