Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Mol Cell ; 83(1): 105-120.e5, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36538933

RESUMO

The versatility of ubiquitination to control vast domains of eukaryotic biology is due, in part, to diversification through differently linked poly-ubiquitin chains. Deciphering signaling roles for some chain types, including those linked via K6, has been stymied by a lack of specificity among the implicated regulatory proteins. Forged through strong evolutionary pressures, pathogenic bacteria have evolved intricate mechanisms to regulate host ubiquitin during infection. Herein, we identify and characterize a deubiquitinase domain of the secreted effector LotA from Legionella pneumophila that specifically regulates K6-linked poly-ubiquitin. We demonstrate the utility of LotA for studying K6 poly-ubiquitin signals. We identify the structural basis of LotA activation and poly-ubiquitin specificity and describe an essential "adaptive" ubiquitin-binding domain. Without LotA activity during infection, the Legionella-containing vacuole becomes decorated with K6 poly-ubiquitin as well as the AAA ATPase VCP/p97/Cdc48. We propose that LotA's deubiquitinase activity guards Legionella-containing vacuole components from ubiquitin-dependent extraction.


Assuntos
Legionella pneumophila , Ubiquitina , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitinação , Poliubiquitina/genética , Poliubiquitina/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Enzimas Desubiquitinantes/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(23): e2122872119, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35653564

RESUMO

Adenosine diphosphate (ADP) ribosylation is a reversible posttranslational modification involved in the regulation of numerous cellular processes. Prototype ADP ribosyltransferases (ARTs) from many pathogenic bacteria are known to function as toxins, while other bacterial ARTs have just recently emerged. Recent studies have shown that bacteria also possess enzymes that function as poly-ADP ribose (ADPr) glycohydrolases (PARGs), which reverse poly-ADP ribosylation. However, how bacteria manipulate host target proteins by coordinated reactions of ARTs and ADPr hydrolases (ARHs) remains elusive. The intracellular bacterial pathogen Legionella pneumophila, the causative agent of Legionnaires' disease, transports a large array of effector proteins via the Dot/Icm type IV secretion system to host cells. The effector proteins, which mostly function as enzymes, modulate host cellular processes for the bacteria's benefit. In this study, we identified a pair of L. pneumophila effector proteins, Lpg0080 and Lpg0081, which function as an ART and an ARH, respectively. The two proteins were shown to coordinately modulate mitochondrial ADP/adenosine triphosphate (ATP) translocases (ANTs) by their enzymatic activities to conjugate ADPr to, and remove it from, a key arginine residue. The crystal structures of Lpg0081 and the Lpg0081:ADPr complex indicated that Lpg0081 is a macroD-type ARH with a noncanonical macrodomain, whose folding topology is strikingly distinct from that of the canonical macrodomain that is ubiquitously found in eukaryotic PARGs and ARHs. Our results illustrate that L. pneumophila has acquired an effector pair that coordinately manipulate mitochondrial activity via reversible chemical modification of ANTs.


Assuntos
Legionella pneumophila , Legionella , Difosfato de Adenosina , Trifosfato de Adenosina , Proteínas de Bactérias , Mitocôndrias/fisiologia , Translocases Mitocondriais de ADP e ATP
3.
Proc Natl Acad Sci U S A ; 119(48): e2206739119, 2022 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-36409909

RESUMO

The serious threats posed by drug-resistant bacterial infections and recent developments in synthetic biology have fueled a growing interest in genetically engineered phages with therapeutic potential. To date, many investigations on engineered phages have been limited to proof of concept or fundamental studies using phages with relatively small genomes or commercially available "phage display kits". Moreover, safeguards supporting efficient translation for practical use have not been implemented. Here, we developed a cell-free phage engineering and rebooting platform. We successfully assembled natural, designer, and chemically synthesized genomes and rebooted functional phages infecting gram-negative bacteria and acid-fast mycobacteria. Furthermore, we demonstrated the creation of biologically contained phages for the treatment of bacterial infections. These synthetic biocontained phages exhibited similar properties to those of a parent phage against lethal sepsis in vivo. This efficient, flexible, and rational approach will serve to accelerate phage biology studies and can be used for many practical applications, including phage therapy.


Assuntos
Infecções Bacterianas , Bacteriófagos , Terapia por Fagos , Humanos , Bacteriófagos/genética , Contenção de Riscos Biológicos , Biologia Sintética , Infecções Bacterianas/terapia
4.
J Cell Sci ; 135(6)2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35224642

RESUMO

The gram-negative bacterium, Legionella pneumophila is known to manipulate the host cellular functions. L. pneumophila secretes bacterial proteins called Legionella effectors into the host cytosol that are necessary for these manipulations. The Legionella effector Lpg1137 was identified as a serine protease responsible for the degradation of syntaxin 17 (Stx17). However, how Lpg1137 specifically recognizes and degrades Stx17 remained unknown. Given that Stx17 is localized in the ER, mitochondria-associated membrane (MAM), and mitochondria, Lpg1137 likely distributes to these compartments to recognize Stx17. Here, we show that the C-terminal region of Lpg1137 binds to phosphatidic acid (PA), a MAM and mitochondria-enriched phospholipid, and that this binding is required for the correct intracellular distribution of Lpg1137. Two basic residues in the C-terminal region of Lpg1137 are required for PA binding and their mutation causes mislocalization of Lpg1137. This mutant also fails to degrade Stx17 while retaining protease activity. Taken together, our data reveal that Lpg1137 utilizes PA for its distribution to the membranous compartments in which Stx17 is localized.


Assuntos
Legionella pneumophila , Legionella , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Legionella/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Ácidos Fosfatídicos/metabolismo , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo
5.
BMC Microbiol ; 24(1): 351, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39289639

RESUMO

BACKGROUND: Bacillus cereus is a Gram-positive, spore-forming bacterium that produces a spectrum of effectors integral to bacterial niche adaptation and the development of various infections. Among those is EsxA, whose secretion depends on the EssC component of the type VII secretion system (T7SS). EsxA's roles within the bacterial cell are poorly understood, although postulations indicate that it may be involved in sporulation. However, the T7SS repertoire in B. cereus has not been reported, and its functions are unestablished. METHODS: We used the type strain, B. cereus ATCC14579, to generate ΔessC mutant through homologous recombination using the homing endonuclease I-SceI mediated markerless gene replacement. Comparatively, we analyzed the culture supernatant of type strain and the ΔessC mutant through Liquid chromatography-tandem mass spectrometry (LC-MS/MS). We further generated T7SSb-specific gene mutations to explore the housekeeping roles of the T7SSb-dependent effectors. The sporulation process of B. cereus ATCC14579 and its mutants was observed microscopically through the classic Schaeffer-Fulton staining method. The spore viability of each strain in this study was established by enumerating the colony-forming units on LB agar. RESULTS: Through LC-MS/MS, we identified a pair of nearly identical (94%) effector proteins named EsxA belonging to the sagEsxA-like subfamily of the WXG100 protein superfamily in the culture supernatant of the wild type and none in the ΔessC mutant. Homology analysis of the T7SSb gene cluster among B. cereus strains revealed diversity from the 3' end of essC, encoding additional substrates. Deletions in esxA1 and esxA2 neither altered cellular morphology nor growth rate, but the ΔesxA1ΔesxA2 deletion resulted in significantly fewer viable spores and an overall slower sporulation process. Within 24 h culture, more than 80% of wild-type cells formed endospores compared to less than 5% in the ΔesxA1ΔesxA2 mutant. The maximum spore ratios for the wild type and ΔesxA1ΔesxA2 were 0.96 and 0.72, respectively. Altogether, these results indicated that EsxA1 and EsxA2 work cooperatively and are required for sporulation in B. cereus ATCC14567. CONCLUSION: B. cereus ATCC14579 possesses two nearly identical T7SSb-dependent effectors belonging to the sagEsxA-like proteins. Simultaneous deletion of genes encoding these effectors significantly delayed and reduced sporulation, a novel finding for EsxA.


Assuntos
Bacillus cereus , Proteínas de Bactérias , Esporos Bacterianos , Sistemas de Secreção Tipo VII , Bacillus cereus/genética , Bacillus cereus/metabolismo , Bacillus cereus/fisiologia , Bacillus cereus/crescimento & desenvolvimento , Esporos Bacterianos/genética , Esporos Bacterianos/crescimento & desenvolvimento , Esporos Bacterianos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Tipo VII/genética , Sistemas de Secreção Tipo VII/metabolismo , Espectrometria de Massas em Tandem , Mutação , Cromatografia Líquida
6.
Microbiol Immunol ; 66(2): 67-74, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34807482

RESUMO

The intracellular bacterial pathogen Legionella pneumophila utilizes the Dot/Icm type IV secretion system to translocate approximately 300 effector proteins to establish a replicative niche known as the Legionella-containing vacuole. The Dot/Icm system is classified as a type IVB secretion system, which is evolutionarily closely related to the I-type conjugation systems and is distinct from type IVA secretion systems, such as the Agrobacterium VirB/D4 system. Although both type IVA and IVB systems directly transport nucleic acids or proteins into the cytosol of recipient cells, the components and architecture of type IVB systems are much more complex than those of type IVA systems. Taking full advantage of rapidly developing cryo-electron microscopy techniques, the structural details of the transport apparatus and coupling complexes in the Dot/Icm system have been clarified in the past few years. In this review, we summarize recent progress in the structural studies of the L. pneumophila type IVB secretion system and the insights gained into the mechanisms of substrate recognition and transport.


Assuntos
Legionella pneumophila , Sistemas de Secreção Tipo IV , Proteínas de Bactérias , Microscopia Crioeletrônica , Vacúolos
7.
Curr Microbiol ; 78(4): 1267-1276, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33638001

RESUMO

The bacterium Staphylococcus aureus, which colonizes healthy human skin, may cause diseases, such as atopic dermatitis (AD). Treatment for such AD cases involves antibiotic use; however, alternate treatments are preferred owing to the development of antimicrobial resistance. This study aimed to characterize the novel bacteriophage SaGU1 as a potential agent for phage therapy to treat S. aureus infections. SaGU1 that infects S. aureus strains previously isolated from the skin of patients with AD was screened from sewage samples in Gifu, Japan. Its genome was sequenced and analyzed using bioinformatics tools, and the morphology, lytic activity, stability, and host range of the phage were determined. The SaGU1 genome was 140,909 bp with an average GC content of 30.2%. The viral chromosome contained 225 putative protein-coding genes and four tRNA genes, carrying neither toxic nor antibiotic resistance genes. Electron microscopy analysis revealed that SaGU1 belongs to the Myoviridae family. Stability tests showed that SaGU1 was heat-stable under physiological and acidic conditions. Host range testing revealed that SaGU1 can infect a broad range of S. aureus clinical isolates present on the skin of AD patients, whereas it did not kill strains of Staphylococcus epidermidis, which are symbiotic resident bacteria on human skin. Hence, our data suggest that SaGU1 is a potential candidate for developing a phage therapy to treat AD caused by pathogenic S. aureus.


Assuntos
Dermatite Atópica , Staphylococcus aureus , Genoma Viral , Humanos , Japão , Fagos de Staphylococcus/genética , Staphylococcus aureus/genética
8.
Cell Microbiol ; 20(7): e12840, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29543380

RESUMO

The intracellular bacterial pathogen, Legionella pneumophila, establishes the replicative niche as a result of the actions of a large array of effector proteins delivered via the Legionella Type 4 secretion system. Many effector proteins are expected to be involved in biogenesis and regulation of the Legionella-containing vacuole (LCV) that is highly decorated with ubiquitin. Here, we identified a Legionella deubiquitinase, designated LotA, by carrying out a genome analysis to find proteins resembling the eukaryotic ovarian tumour superfamily of cysteine proteases. LotA exhibits a dual ability to cleave ubiquitin chains that is dependent on 2 distinctive catalytic cysteine residues in the eukaryotic ovarian tumour domains. One cysteine dominantly contributes to the removal of ubiquitin from the LCVs by its polyubiquitin cleavage activity. The other specifically cleaves conjugated Lys6-linked ubiquitin. After delivered by the Type 4 secretion system, LotA localises on the LCVs via its PI(3)P-binding domain. The lipid-binding ability of LotA is crucial for ubiquitin removal from the vacuoles. We further analysed the functional interaction of the protein with the recently reported noncanonical ubiquitin ligases of L. pneumophila, revealing that the effector proteins are involved in coordinated regulation that contributes to bacterial growth in the host cells.


Assuntos
Enzimas Desubiquitinantes/metabolismo , Interações Hospedeiro-Patógeno , Legionella pneumophila/enzimologia , Legionella pneumophila/crescimento & desenvolvimento , Ubiquitina/metabolismo , Biologia Computacional , Mineração de Dados , Células HEK293 , Células HeLa , Humanos , Legionella pneumophila/genética , Metabolismo dos Lipídeos , Ligação Proteica , Vacúolos/metabolismo , Vacúolos/microbiologia
9.
PLoS Pathog ; 10(8): e1004321, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25144274

RESUMO

Etiological agents of acute, persistent, or relapsing clinical infections are often refractory to antibiotics due to multidrug resistance and/or antibiotic tolerance. Pseudomonas aeruginosa is an opportunistic Gram-negative bacterial pathogen that causes recalcitrant and severe acute chronic and persistent human infections. Here, we target the MvfR-regulated P. aeruginosa quorum sensing (QS) virulence pathway to isolate robust molecules that specifically inhibit infection without affecting bacterial growth or viability to mitigate selective resistance. Using a whole-cell high-throughput screen (HTS) and structure-activity relationship (SAR) analysis, we identify compounds that block the synthesis of both pro-persistence and pro-acute MvfR-dependent signaling molecules. These compounds, which share a benzamide-benzimidazole backbone and are unrelated to previous MvfR-regulon inhibitors, bind the global virulence QS transcriptional regulator, MvfR (PqsR); inhibit the MvfR regulon in multi-drug resistant isolates; are active against P. aeruginosa acute and persistent murine infections; and do not perturb bacterial growth. In addition, they are the first compounds identified to reduce the formation of antibiotic-tolerant persister cells. As such, these molecules provide for the development of next-generation clinical therapeutics to more effectively treat refractory and deleterious bacterial-human infections.


Assuntos
Antibacterianos/farmacologia , Descoberta de Drogas , Farmacorresistência Bacteriana/efeitos dos fármacos , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/efeitos dos fármacos , Percepção de Quorum/fisiologia , Animais , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Camundongos , Virulência/efeitos dos fármacos
10.
Elife ; 122024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38771316

RESUMO

Rab GTPases are representative targets of manipulation by intracellular bacterial pathogens for hijacking membrane trafficking. Legionella pneumophila recruits many Rab GTPases to its vacuole and exploits their activities. Here, we found that infection-associated regulation of Rab10 dynamics involves ubiquitin signaling cascades mediated by the SidE and SidC families of Legionella ubiquitin ligases. Phosphoribosyl-ubiquitination of Rab10 catalyzed by the SidE ligases is crucial for its recruitment to the bacterial vacuole. SdcB, the previously uncharacterized SidC-family effector, resides on the vacuole and contributes to retention of Rab10 at the late stages of infection. We further identified MavC as a negative regulator of SdcB. By the transglutaminase activity, MavC crosslinks ubiquitin to SdcB and suppresses its function, resulting in elimination of Rab10 from the vacuole. These results demonstrate that the orchestrated actions of many L. pneumophila effectors fine-tune the dynamics of Rab10 during infection.


Assuntos
Proteínas de Bactérias , Legionella pneumophila , Vacúolos , Proteínas rab de Ligação ao GTP , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab de Ligação ao GTP/genética , Legionella pneumophila/metabolismo , Legionella pneumophila/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Humanos , Vacúolos/metabolismo , Vacúolos/microbiologia , Interações Hospedeiro-Patógeno , Ubiquitinação , Animais , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética
11.
Nature ; 446(7136): 680-4, 2007 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-17377534

RESUMO

Semaphorins are axon guidance factors that assist growing axons in finding appropriate targets and forming synapses. Emerging evidence suggests that semaphorins are involved not only in embryonic development but also in immune responses. Semaphorin 7A (Sema7A; also known as CD108), which is a glycosylphosphatidylinositol-anchored semaphorin, promotes axon outgrowth through beta1-integrin receptors and contributes to the formation of the lateral olfactory tract. Although Sema7A has been shown to stimulate human monocytes, its function as a negative regulator of T-cell responses has also been reported. Thus, the precise function of Sema7A in the immune system remains unclear. Here we show that Sema7A, which is expressed on activated T cells, stimulates cytokine production in monocytes and macrophages through alpha1beta1 integrin (also known as very late antigen-1) as a component of the immunological synapse, and is critical for the effector phase of the inflammatory immune response. Sema7A-deficient (Sema7a-/-) mice are defective in cell-mediated immune responses such as contact hypersensitivity and experimental autoimmune encephalomyelitis. Although antigen-specific and cytokine-producing effector T cells can develop and migrate into antigen-challenged sites in Sema7a-/- mice, Sema7a-/- T cells fail to induce contact hypersensitivity even when directly injected into the antigen-challenged sites. Thus, the interaction between Sema7A and alpha1beta1 integrin is crucial at the site of inflammation. These findings not only identify a function of Sema7A as an effector molecule in T-cell-mediated inflammation, but also reveal a mechanism of integrin-mediated immune regulation.


Assuntos
Antígenos CD/metabolismo , Inflamação/imunologia , Integrina alfa1beta1/metabolismo , Semaforinas/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Antígenos CD/genética , Citocinas/metabolismo , Imunidade/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Monócitos/metabolismo , Semaforinas/deficiência , Semaforinas/genética , Transdução de Sinais
12.
Mol Microbiol ; 79(6): 1615-28, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21244531

RESUMO

Isoniazid (INH) is a key agent in the treatment of tuberculosis. In Mycobacterium tuberculosis, INH is converted to its active form by KatG, a catalase-peroxidase, and attacks InhA, which is essential for the synthesis of mycolic acids. We sequenced furA-katG and fabG1-inhA in 108 INH-resistant (INH(r) ) and 51 INH-susceptible (INH(s) ) isolates, and found three mutations in the furA-katG intergenic region (Int(g-7a) , Int(a-10c) and Int(g-12a) ) in four of 108 INH(r) isolates (4%), and the furA(c41t) mutation with an amino acid substitution in 18 INH(r) isolates (17%). These mutations were not found in any of 51 INH(s) isolates tested. We reconstructed these mutations in isogenic strains to determine whether they conferred INH resistance. We found that the Int(g-7a) , Int(a-10c) and Int(g-12a) single mutations in the furA-katG intergenic region decreased katG expression and conferred INH resistance. In contrast, the furA(c41t) mutation was not sufficient to confer INH resistance. These results suggested that downregulation of katG is a mechanism of INH resistance in M. tuberculosis and that mutations in the furA-katG intergenic region play a role in this resistance mechanism.


Assuntos
Antituberculosos/farmacologia , Proteínas de Bactérias/genética , Catalase/genética , Regulação para Baixo , Isoniazida/farmacologia , Mycobacterium tuberculosis/enzimologia , Proteínas de Bactérias/metabolismo , Catalase/metabolismo , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Farmacorresistência Bacteriana , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Mutação , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/genética , Tuberculose/microbiologia
13.
J Cell Biol ; 178(1): 167-78, 2007 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-17591922

RESUMO

Fibronectin (FN) is secreted as a disulfide-bonded FN dimer. Each subunit contains three types of repeating modules: FN-I, FN-II, and FN-III. The interactions of alpha5beta1 or alphav integrins with the RGD motif of FN-III repeat 10 (FN-III10) are considered an essential step in the assembly of FN fibrils. To test this hypothesis in vivo, we replaced the RGD motif with the inactive RGE in mice. FN-RGE homozygous embryos die at embryonic day 10 with shortened posterior trunk, absent tail bud-derived somites, and severe vascular defects resembling the phenotype of alpha5 integrin-deficient mice. Surprisingly, the absence of a functional RGD motif in FN did not compromise assembly of an FN matrix in mutant embryos or on mutant cells. Matrix assembly assays and solid-phase binding assays reveal that alphavbeta3 integrin assembles FN-RGE by binding an isoDGR motif in FN-I5, which is generated by the nonenzymatic rearrangement of asparagines (N) into an iso-aspartate (iso-D). Our findings demonstrate that FN contains a novel motif for integrin binding and fibril formation whose activity is controlled by amino acid modification.


Assuntos
Ácido Aspártico/metabolismo , Fibronectinas/química , Fibronectinas/metabolismo , Oligopeptídeos/química , Reticulina/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular Transformada , Dimerização , Dissulfetos/química , Embrião de Mamíferos , Matriz Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibronectinas/genética , Heterozigoto , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Camundongos , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Solubilidade
14.
Nat Commun ; 13(1): 5103, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-36042245

RESUMO

Intestinal barrier derangement allows intestinal bacteria and their products to translocate to the systemic circulation. Pseudomonas aeruginosa (PA) superimposed infection in critically ill patients increases gut permeability and leads to gut-driven sepsis. PA infections are challenging due to multi-drug resistance (MDR), biofilms, and/or antibiotic tolerance. Inhibition of the quorum-sensing transcriptional regulator MvfR(PqsR) is a desirable anti-PA anti-virulence strategy as MvfR controls multiple acute and chronic virulence functions. Here we show that MvfR promotes intestinal permeability and report potent anti-MvfR compounds, the N-Aryl Malonamides (NAMs), resulting from extensive structure-activity-relationship studies and thorough assessment of the inhibition of MvfR-controlled virulence functions. This class of anti-virulence non-native ligand-based agents has a half-maximal inhibitory concentration in the nanomolar range and strong target engagement. Using a NAM lead in monotherapy protects murine intestinal barrier function, abolishes MvfR-regulated small molecules, ameliorates bacterial dissemination, and lowers inflammatory cytokines. This study demonstrates the importance of MvfR in PA-driven intestinal permeability. It underscores the utility of anti-MvfR agents in maintaining gut mucosal integrity, which should be part of any successful strategy to prevent/treat PA infections and associated gut-derived sepsis in critical illness settings. NAMs provide for the development of crucial preventive/therapeutic monotherapy options against untreatable MDR PA infections.


Assuntos
Infecções por Pseudomonas , Sepse , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Proteínas de Bactérias/farmacologia , Biofilmes , Estado Terminal , Humanos , Camundongos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/fisiologia , Percepção de Quorum , Sepse/tratamento farmacológico , Virulência
15.
J Bacteriol ; 193(24): 7010, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22123763

RESUMO

We report the completely annotated genome sequence of Pseudomonas aeruginosa NCGM2.S1, a representative strain of a cluster endemic to Japan with a high level of resistance to carbapenem (MIC ≥ 128 µg/ml), amikacin (MIC ≥ 128 µg/ml), and fluoroquinolone (MIC ≥ 128 µg/ml).


Assuntos
Genoma Bacteriano , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética , Antibacterianos/farmacologia , Sequência de Bases , Farmacorresistência Bacteriana Múltipla , Japão/epidemiologia , Dados de Sequência Molecular , Infecções por Pseudomonas/epidemiologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/isolamento & purificação
16.
J Bacteriol ; 193(22): 6397, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22038962

RESUMO

We report the annotated genome sequence of multidrug-resistant Pseudomonas aeruginosa strain NCGM1179, which is highly resistant to carbapenems, aminoglycosides, and fluoroquinolones and is emerging at medical facilities in Japan.


Assuntos
Farmacorresistência Bacteriana Múltipla , Genoma Bacteriano , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética , Antibacterianos/farmacologia , Sequência de Bases , Humanos , Dados de Sequência Molecular , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/isolamento & purificação
17.
STAR Protoc ; 2(2): 100410, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33870219

RESUMO

The intracellular bacterial pathogen Legionella pneumophila exploits host cellular systems using approximately 300 effector proteins to establish a replicative niche known as the Legionella-containing vacuole (LCV). During infection, both host and bacterial proteins interactively function on the LCVs. Here, we describe a detailed step-by-step protocol to visualize proteins associated with LCVs in host cells. This protocol can aid in analyzing whether a protein of interest influences the subcellular localization of LCV-associated proteins during infection. For complete details on the use and execution of this protocol, please refer to Kitao et al. (2020).


Assuntos
Proteínas de Bactérias/análise , Técnicas Bacteriológicas/métodos , Imunofluorescência/métodos , Legionella pneumophila/química , Vacúolos , Proteínas de Bactérias/química , Técnicas de Cultura de Células , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Plasmídeos/genética , Plasmídeos/metabolismo , Transfecção , Vacúolos/química , Vacúolos/microbiologia
18.
J Antimicrob Chemother ; 65(7): 1382-6, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20478990

RESUMO

OBJECTIVES: To develop an easy-to-use method for the rapid detection of antibiotic-resistant bacteria. Here, a new immunochromatographic assay specific for aminoglycoside 6'-N-acetyltransferase AAC(6')-Iae was designed. AAC(6')-Iae is a significant marker molecule for multidrug-resistant (MDR) Pseudomonas aeruginosa isolates in Japan. METHODS: Monoclonal antibodies specific for AAC(6')-Iae were used to construct the assay. The assessment of the assay was performed using 116 P. aeruginosa clinical isolates obtained from hospitals in the Kanto area of Japan where little was known about AAC(6')-Iae producers. PCR analyses of the aac(6')-Iae and class 1 integron, antimicrobial susceptibility testing and PFGE analysis were performed to characterize positive strains. RESULTS: The detection limit of the assay was 1.0 x 10(5) cfu. Of 116 clinical isolates, 60 were positive for AAC(6')-Iae using the assay. The results of assessment with clinical isolates were fully consistent with those of aac(6')-Iae PCR analyses, showing no false positives or negatives. All positive strains detected by the assay showed MDR phenotypes that were resistant to several classes of antibiotic. PFGE analysis showed that 59 of 60 positive strains tightly clustered, and these included clonal expansions. CONCLUSIONS: The developed assay is an easy-to-use and reliable detection method for AAC(6')-Iae-producing MDR P. aeruginosa. This approach may be applicable for screening and investigation of antibiotic-resistant bacteria as an alternative to PCR analysis.


Assuntos
Acetiltransferases/análise , Técnicas Bacteriológicas/métodos , Cromatografia/métodos , Farmacorresistência Bacteriana Múltipla , Pseudomonas aeruginosa/enzimologia , Anticorpos Antibacterianos , Anticorpos Monoclonais , Proteínas de Bactérias/análise , Técnicas de Tipagem Bacteriana , Análise por Conglomerados , Infecção Hospitalar/microbiologia , Impressões Digitais de DNA , DNA Bacteriano/genética , Eletroforese em Gel de Campo Pulsado , Genótipo , Humanos , Imunoensaio/métodos , Integrons , Japão , Reação em Cadeia da Polimerase , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/isolamento & purificação , Sensibilidade e Especificidade
19.
Artigo em Inglês | MEDLINE | ID: mdl-32974222

RESUMO

The intracellular bacterial pathogen Legionella pneumophila employs bacteria-derived effector proteins in a variety of functions to exploit host cellular systems. The ubiquitination machinery constitutes a crucial eukaryotic system for the regulation of numerous cellular processes, and is a representative target for effector-mediated bacterial manipulation. L. pneumophila transports over 300 effector proteins into host cells through its Dot/Icm type IV secretion system. Among these, several effector proteins have been found to function as ubiquitin ligases, including unprecedented enzymes that catalyze ubiquitination through unconventional mechanisms. Recent studies have identified many L. pneumophila effector proteins that can interfere with ubiquitination. These effectors include proteins that are distantly related to the ovarian tumor protein superfamily described as deubiquitinases (DUBs), which regulate important signaling cascades in human cells. Intriguingly, L. pneumophila DUBs are not limited to enzymes that exhibit canonical DUB activity. Some L. pneumophila DUBs can catalyze the cleavage of the unconventional linkage between ubiquitin and substrates. Furthermore, novel mechanisms have been found that adversely affect the function of specific ubiquitin ligases; for instance, effector-mediated posttranslational modifications of ubiquitin ligases result in the inhibition of their activity. In the context of L. pneumophila infection, the existence of enzymes that reverse ubiquitination primarily relates to a fine tuning of biogenesis and remodeling of the Legionella-containing vacuole as a replicative niche. The complexity of the effector arrays reflects sophisticated strategies that bacteria have adopted to adapt their host environment and enable their survival in host cells. This review summarizes the current state of knowledge on the divergent mechanisms of the L. pneumophila effectors that can reverse ubiquitination, which is mediated by other effectors as well as the host ubiquitin machinery.


Assuntos
Legionella pneumophila , Legionella , Doença dos Legionários , Proteínas de Bactérias/metabolismo , Humanos , Legionella/metabolismo , Legionella pneumophila/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Ubiquitinação
20.
Cell Rep ; 32(10): 108107, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32905772

RESUMO

The intracellular bacterial pathogen Legionella pneumophila uses many effector proteins delivered by the bacterial type IV secretion system (T4SS) to hijack the early secretory pathway to establish its replicative niche, known as the Legionella-containing vacuole (LCV). On LCV biogenesis, the endoplasmic reticulum (ER) vesicular soluble N-ethylmaleimide-sensitive factor attachment protein receptors (v-SNARE) Sec22b is recruited to the bacterial phagosome and forms non-canonical pairings with target membrane SNAREs (t-SNAREs) from the plasma membrane. Here, we identify a Legionella deubiquitinase (DUB), LotB, that can modulate the early secretory pathway by interacting with coatomer protein complex I (COPI) vesicles when ectopically expressed. We show that Sec22b is ubiquitinated upon L. pneumophila infection in a T4SS-dependent manner and that, subsequently, LotB deconjugates K63-linked ubiquitins from Sec22b. The DUB activity of LotB stimulates dissociation of the t-SNARE syntaxin 3 (Stx3) from Sec22b, which resides on the LCV. Our study highlights a bacterial strategy manipulating the dynamics of infection-induced SNARE pairing using a bacterial DUB.


Assuntos
Enzimas Desubiquitinantes/metabolismo , Legionella pneumophila/patogenicidade , Proteínas de Bactérias/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa