Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Infect Immun ; 92(6): e0013224, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38700334

RESUMO

Adherent and invasive Escherichia coli (AIEC) is a pathobiont that is involved in the onset and exacerbation of Crohn's disease. Although the inducible expression of virulence traits is a critical step for AIEC colonization in the host, the mechanism underlying AIEC colonization remains largely unclear. We here showed that the two-component signal transduction system CpxRA contributes to AIEC gut competitive colonization by activating type 1 fimbriae expression. CpxRA from AIEC strain LF82 functioned as a transcriptional regulator, as evidenced by our finding that an isogenic cpxRA mutant exhibits reduced expression of cpxP, a known regulon gene. Transcription levels of cpxP in LF82 increased in response to envelope stress, such as exposure to antimicrobials compromising the bacterial membrane, whereas the cpxRA mutant did not exhibit this response. Furthermore, we found that the cpxRA mutant exhibits less invasiveness into host cells than LF82, primarily due to reduced expression of the type 1 fimbriae. Finally, we found that the cpxRA mutant is impaired in gut competitive colonization in a mouse model. The colonization defects were reversed by the introduction of a plasmid encoding the cpxRA gene or expressing the type 1 fimbriae. Our findings indicate that modulating CpxRA activity could be a promising approach to regulating AIEC-involved Crohn's disease.


Assuntos
Aderência Bacteriana , Modelos Animais de Doenças , Células Epiteliais , Infecções por Escherichia coli , Escherichia coli , Fímbrias Bacterianas , Regulação Bacteriana da Expressão Gênica , Animais , Camundongos , Fímbrias Bacterianas/metabolismo , Fímbrias Bacterianas/genética , Escherichia coli/genética , Escherichia coli/patogenicidade , Células Epiteliais/microbiologia , Infecções por Escherichia coli/microbiologia , Aderência Bacteriana/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Virulência/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Intestinos/microbiologia , Feminino
2.
Microbiol Immunol ; 68(6): 206-211, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38644589

RESUMO

Colonization resistance, conferred by the host's microbiota through both direct and indirect protective actions, serves to protect the host from enteric infections. Here, we identified the specific members of the gut microbiota that impact gastrointestinal colonization by Citrobacter rodentium, a murine pathogen causing colonic crypt hyperplasia. The gut colonization levels of C. rodentium in C57BL/6 mice varied among breeding facilities, probably due to differences in microbiota composition. A comprehensive analysis of the microbiota revealed that specific members of the microbiota may influence gut colonization by C. rodentium, thus providing a potential link between the two.


Assuntos
Citrobacter rodentium , Infecções por Enterobacteriaceae , Microbioma Gastrointestinal , Trato Gastrointestinal , Camundongos Endogâmicos C57BL , Animais , Citrobacter rodentium/patogenicidade , Citrobacter rodentium/fisiologia , Infecções por Enterobacteriaceae/microbiologia , Camundongos , Trato Gastrointestinal/microbiologia , Colo/microbiologia , Colo/patologia , Fezes/microbiologia , RNA Ribossômico 16S/genética
3.
Infect Immun ; 90(3): e0066221, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-34978926

RESUMO

Adherent-invasive Escherichia coli (AIEC) is involved in onset and/or exacerbation of Crohn's disease (CD). AIEC adapts to the gut environment by altering gene expression programs, leading to successful gut-lumen colonization. However, the underlying mechanism of gut colonization is still far from clarified. Here, we show the role of UvrY, a response regulator of bacterial two-component signal transduction systems, in AIEC gut colonization. An AIEC mutant lacking the uvrY gene exhibited impairment of competitive colonization in the murine intestinal tract. UvrY contributes to functional expression of type 1 fimbriae by activating expression of small RNA CsrB, which confers adherence and invasion into epithelial cells on AIEC. In contrast, acetate suppresses the UvrY-dependent expression of type 1 fimbriae, resulting in less efficient cell invasion and attenuated gut colonization. Our findings might lead to therapeutic interventions for CD, in which inhibitions of UvrY activation and acetate supplementation reduce the colonization levels of AIEC by decreasing type 1 fimbria expression.


Assuntos
Doença de Crohn , Infecções por Escherichia coli , Acetatos/metabolismo , Animais , Aderência Bacteriana/genética , Doença de Crohn/microbiologia , Células Epiteliais/microbiologia , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/metabolismo , Mucosa Intestinal/metabolismo , Camundongos
4.
Infect Immun ; 90(7): e0018422, 2022 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-35652649

RESUMO

Long-chain-fatty-acid (LCFA) metabolism is a fundamental cellular process in bacteria that is involved in lipid homeostasis, energy production, and infection. However, the role of LCFA metabolism in Salmonella enterica serovar Typhimurium (S. Tm) gut infection remains unclear. Here, using a murine gastroenteritis infection model, we demonstrate involvement of LCFA metabolism in S. Tm gut colonization. The LCFA metabolism-associated transcriptional regulator FadR contributes to S. Tm gut colonization. fadR deletion alters the gene expression profile and leads to aberrant flagellar motility of S. Tm. Colonization defects in the fadR mutant are attributable to altered swimming behavior characterized by less frequently smooth swimming, resulting from reduced expression of the phase 2 flagellin FljB. Notably, changes in lipid LCFA composition by fadR deletion lead to reduced expression of fljB, which is restored by exogenous LCFA. Therefore, LCFA homeostasis may maintain proper flagellar motility by activating fljB expression, contributing to S. Tm gut colonization. Our findings improve the understanding of the effect of luminal LCFA on the virulence of enteric pathogens.


Assuntos
Flagelina , Salmonella typhimurium , Animais , Ácidos Graxos/metabolismo , Flagelina/metabolismo , Homeostase , Lipídeos , Camundongos , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo
5.
Infect Immun ; 88(6)2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32284369

RESUMO

Salmonella enterica serovar Typhimurium is an important foodborne pathogen that causes diarrhea. S. Typhimurium elicits inflammatory responses and colonizes the gut lumen by outcompeting the microbiota. Although evidence is accumulating with regard to the underlying mechanism, the infectious stage has not been adequately defined. Peptidoglycan amidases are widely distributed among bacteria and play a prominent role in peptidoglycan maintenance by hydrolyzing peptidoglycans. Amidase activation is required for the regulation of at least one of two cognate activators, NlpD or EnvC (also called YibP). Recent studies established that the peptidoglycan amidase AmiC-mediated cell division specifically confers a fitness advantage on S Typhimurium in the inflamed gut. However, it remains unknown which cognate activators are involved in the amidase activation and how the activators influence Salmonella sp. pathogenesis. Here, we characterize the role of two activators, NlpD and EnvC, in S Typhimurium cell division and gut infection. EnvC was found to contribute to cell division of S Typhimurium cells through the activation of AmiA and AmiC. The envC mutant exhibited impairments in gut infection, including a gut colonization defect and reduced ability to elicit inflammatory responses. Importantly, the colonization defect of the envC mutant was unrelated to the microbiota but was conferred by attenuated motility and chemotaxis of S Typhimurium cells, which were not observed in the amiA amiC mutant. Furthermore, the envC mutant was impaired in its induction of mucosal inflammation and sustained gut colonization. Collectively, our findings provide a novel insight into the peptidoglycan amidase/cognate activator circuits and their dependent pathogenesis.


Assuntos
Proteínas de Bactérias/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/fisiologia , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Ácido Desoxicólico/farmacologia , Escherichia coli/fisiologia , Lipoproteínas/genética , Lipoproteínas/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Testes de Sensibilidade Microbiana , Modelos Biológicos , Mutação , N-Acetil-Muramil-L-Alanina Amidase/genética , Salmonella typhimurium/efeitos dos fármacos
6.
Infect Immun ; 88(3)2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31818958

RESUMO

The twin-arginine translocation (Tat) system is involved in not only a wide array of cellular processes but also pathogenesis in many bacterial pathogens; thus, this system is expected to become a novel therapeutic target to treat infections. To the best of our knowledge, involvement of the Tat system has not been reported in the gut infection caused by Citrobacter rodentium Here, we studied the role of Tat in C. rodentium gut infection, which resembles human infection with enterohemorrhagic Escherichia coli (EHEC) and enteropathogenic E. coli (EPEC). A C. rodentium Tat loss-of-function mutant displayed prolonged gut colonization, which was explained by reduced inflammatory responses and, particularly, neutrophil infiltration. Further, the Tat mutant had colonization defects upon coinfection with the wild-type strain of C. rodentium The Tat mutant also became hypersensitive to bile acids, and an increase in fecal bile acids fostered C. rodentium clearance from the gut lumen. Finally, we show that the chain form of C. rodentium cells, induced by a Tat-dependent cell division defect, exhibits impaired resistance to bile acids. Our findings indicate that the Tat system is involved in gut colonization by C. rodentium, which is associated with neutrophil infiltration and resistance to bile acids. Interventions that target the Tat system, as well as luminal bile acids, might thus be promising therapeutic strategies to treat human EHEC and EPEC infections.


Assuntos
Citrobacter rodentium/patogenicidade , Infecções por Enterobacteriaceae/imunologia , Trato Gastrointestinal/microbiologia , Sistema de Translocação de Argininas Geminadas/fisiologia , Animais , Ácidos e Sais Biliares/metabolismo , Ácidos e Sais Biliares/farmacologia , Citrobacter rodentium/efeitos dos fármacos , Citrobacter rodentium/fisiologia , Infecções por Enterobacteriaceae/microbiologia , Trato Gastrointestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
7.
PLoS Pathog ; 14(10): e1007391, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30379938

RESUMO

Salmonella enterica serovar Typhimurium (S. Tm) is a cause of food poisoning accompanied with gut inflammation. Although mucosal inflammation is generally thought to be protective against bacterial infection, S. Tm exploits the inflammation to compete with commensal microbiota, thereby growing up to high densities in the gut lumen and colonizing the gut continuously at high levels. However, the molecular mechanisms underlying the beneficial effect of gut inflammation on S. Tm competitive growth are poorly understood. Notably, the twin-arginine translocation (Tat) system, which enables the transport of folded proteins outside bacterial cytoplasm, is well conserved among many bacterial pathogens, with Tat substrates including virulence factors and virulence-associated proteins. Here, we show that Tat and Tat-exported peptidoglycan amidase, AmiA- and AmiC-dependent cell division contributes to S. Tm competitive fitness advantage in the inflamed gut. S. Tm tatC or amiA amiC mutants feature a gut colonization defect, wherein they display a chain form of cells. The chains are attributable to a cell division defect of these mutants and occur in inflamed but not in normal gut. We demonstrate that attenuated resistance to bile acids confers the colonization defect on the S. Tm amiA amiC mutant. In particular, S. Tm cell chains are highly sensitive to bile acids as compared to single or paired cells. Furthermore, we show that growth media containing high concentrations of NaCl and sublethal concentrations of antimicrobial peptides induce the S. Tm amiA amiC mutant chain form, suggesting that gut luminal conditions such as high osmolarity and the presence of antimicrobial peptides impose AmiA- and AmiC-dependent cell division on S. Tm. Together, our data indicate that Tat and the Tat-exported amidases, AmiA and AmiC, are required for S. Tm luminal fitness in the inflamed gut, suggesting that these proteins might comprise effective targets for novel antibacterial agents against infectious diarrhea.


Assuntos
Amidoidrolases/metabolismo , Trato Gastrointestinal/microbiologia , Inflamação/microbiologia , Peptidoglicano/metabolismo , Salmonelose Animal/microbiologia , Salmonella typhimurium/fisiologia , Sistema de Translocação de Argininas Geminadas/metabolismo , Animais , Divisão Celular , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Salmonelose Animal/metabolismo , Salmonelose Animal/patologia
8.
Infect Immun ; 87(9)2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31235639

RESUMO

Salmonella enterica serovar Typhimurium (S. Typhimurium) induces inflammatory changes in the ceca of streptomycin-pretreated mice. In this mouse model of colitis, the type III secretion system 1 (T3SS-1) has been shown to induce rapid inflammatory change in the cecum at early points, 10 to 24 h after infection. Five proteins, SipA, SopA, SopB, SopD, and SopE2, have been identified as effectors involved in eliciting intestinal inflammation within this time range. In contrast, a T3SS-1-deficient strain was shown to exhibit inflammatory changes in the cecum at 72 to 120 h postinfection. However, the effectors eliciting T3SS-1-independent inflammation remain to be clarified. In this study, we focused on two T3SS-2 phenotypes, macrophage proliferation and cytotoxicity, to identify the T3SS-2 effectors involved in T3SS-1-independent inflammation. We identified a mutant strain that could not induce cytotoxicity in a macrophage-like cell line and that reduced intestinal inflammation in streptomycin-pretreated mice. We also identified five T3SS-2 effectors, SifA, SpvB, SseF, SseJ, and SteA, associated with T3SS-1-independent macrophage cytotoxicity. We then constructed a strain lacking T3SS-1 and all the five T3SS-2 effectors, termed T1S5. The S. Typhimurium T1S5 strain significantly reduced cytotoxicity in macrophages in the same manner as a mutant invA spiB strain (T1T2). Finally, the T1S5 strain elicited no inflammatory changes in the ceca of streptomycin-pretreated mice. We conclude that these five T3SS-2 effectors contribute to T3SS-1-independent inflammation.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/fisiologia , Colite/microbiologia , Salmonella enterica/patogenicidade , Estreptomicina/farmacologia , Sistemas de Secreção Tipo III/fisiologia , Animais , Ceco/patologia , Colite/patologia , Modelos Animais de Doenças , Macrófagos/patologia , Camundongos , Proteínas dos Microfilamentos/fisiologia , Salmonella enterica/metabolismo
9.
Infect Immun ; 86(7)2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29685984

RESUMO

Salmonella enterica, a common cause of diarrhea, has to colonize the gut lumen to elicit disease. In the gut, the pathogen encounters a vast array of environmental stresses that cause perturbations in the bacterial envelope. The CpxRA two-component system monitors envelope perturbations and responds by altering the bacterial gene expression profile. This allows Salmonella to survive under such harmful conditions. Therefore, CpxRA activation is likely to contribute to Salmonella gut infection. However, the role of the CpxRA-mediated envelope stress response in Salmonella-induced diarrhea is unclear. Here, we show that CpxRA is dispensable for the induction of colitis by S. enterica serovar Typhimurium, whereas it is required for gut colonization. We prove that CpxRA is expressed during gut infection and that the presence of antimicrobial peptides in growth media activates the expression of CpxRA-regulated genes. In addition, we demonstrate that a S Typhimurium strain lacking the cpxRA gene is able to cause colitis but is unable to continuously colonize the gut. Finally, we show that CpxRA-dependent gut colonization requires the host gut inflammatory response, while DegP, a CpxRA-regulated protease, is dispensable. Our findings reveal that the CpxRA-mediated envelope stress response plays a crucial role in Salmonella gut infection, suggesting that CpxRA might be a promising therapeutic target for infectious diarrhea.


Assuntos
Proteínas de Bactérias/fisiologia , Colite/etiologia , Trato Gastrointestinal/microbiologia , Proteínas Quinases/fisiologia , Salmonella typhimurium/fisiologia , Transdução de Sinais/fisiologia , Animais , Antibacterianos/farmacologia , Proteínas de Choque Térmico/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Periplásmicas/fisiologia , Serina Endopeptidases/fisiologia
10.
J Biol Chem ; 287(41): 34844-55, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-22896700

RESUMO

RegIIIß is a member of the C-type lectin family called RegIII. It is known to bind peptidoglycan, and its bactericidal activity shapes the interactions with commensal and pathogenic gut bacteria. However, little is known about its carbohydrate recognition specificity and the bactericidal mechanism, particularly against Gram-negative bacteria. Here, we show that RegIIIß can bind directly to LPS by recognizing the carbohydrate moiety of lipid A via a novel motif that is indispensable for its bactericidal activity. This bactericidal activity of RegIIIß could be inhibited by preincubation with LPS, lipid A, or gentiobiose. The latter is a disaccharide composed of two units of ß-(1→6)-linked d-glucose and resembles the carbohydrate moiety of lipid A. Therefore, this structural element may form a key target site recognized by RegIIIß. Using point-mutated RegIIIß proteins, we found that amino acid residues in two structural motifs termed "loop 1" and "loop 2," are important for peptidoglycan and lipid A binding (Arg-135, Asp-142) and for the bactericidal activity (Glu-134, Asn-136, Asp-142). Thus, the ERN motif and residue Asp-142 in the loop 2 are of critical importance for RegIIIß function. This provides novel insights into the carbohydrate recognition specificity of RegIIIß and explains its bactericidal activity against Gram-negative bacteria.


Assuntos
Antibacterianos/química , Bactérias Gram-Negativas/química , Lectinas Tipo C/química , Lipídeo A/química , Proteínas/química , Motivos de Aminoácidos , Animais , Antibacterianos/metabolismo , Dissacarídeos/química , Dissacarídeos/genética , Dissacarídeos/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Lipídeo A/metabolismo , Camundongos , Proteínas Associadas a Pancreatite , Mutação Puntual , Ligação Proteica , Proteínas/genética , Proteínas/metabolismo
11.
Mol Microbiol ; 80(5): 1186-203, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21435035

RESUMO

The type III secretion system (T3SS) encoded by Chromobacterium pathogenicity islands 1 and 1a (Cpi-1/-1a) is critical for Chromobacterium violaceum pathogenesis. T3SS-dependent virulence is commonly characterized by type III effector virulence function, but the full repertoire of the effector proteins of Cpi-1/-1a T3SS is unknown. In this study, we showed that expression of Cpi-1/-1a T3SS is controlled by the master regulator CilA. We used transcriptional profiling with DNA microarrays to define CilA regulon and identified genes encoding T3SS effectors whose translocation into host cells was dependent on Cpi-1/-1a T3SS. From these effectors, we found that CopE (CV0296) has similarities to a guanine nucleotide exchange factor (GEF) for Rho GTPases in its C-terminal portion. The N-terminal portions (1-81 amino acids) of CopE and a CivB as a putative chaperone were required for its translocation. CopE specifically activates Rac1 and Cdc42 followed by the induction of actin cytoskeletal rearrangement. Interestingly, C. violaceum invades human epithelial HeLa cells in a Cpi-1/-1a-encoded T3SS- and CopE-dependent manner. Finally, C. violaceum strains lacking copE and expressing a CopE-G168V deficient in GEF activity were attenuated for virulence in mice, suggesting that CopE contributes to the virulence of this pathogen.


Assuntos
Proteínas de Bactérias/metabolismo , Chromobacterium/metabolismo , Chromobacterium/patogenicidade , Células Epiteliais/microbiologia , Infecções por Bactérias Gram-Negativas/microbiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Motivos de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Linhagem Celular , Chromobacterium/química , Chromobacterium/genética , Feminino , Regulação Bacteriana da Expressão Gênica , Ilhas Genômicas , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Virulência
12.
Environ Microbiol Rep ; 14(4): 637-645, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35581157

RESUMO

Bifidobacterium bifidum possesses two extracellular sialidases (SiaBb1 and SiaBb2) that release free sialic acid from mucin sialoglycans, which can be utilized via cross-feeding by Bifidobacterium breve that, otherwise, is prevented from utilizing this nutrient source. Modification of sialic acids with O-acetyl esters is known to protect mucin glycans from degradation by bacterial sialidases. Compared to SiaBb2, SiaBb1 has an additional O-acetylesterase (Est) domain. We aimed to elucidate the role of the SiaBb1 Est domain from B. bifidum in sialic acid cross-feeding within Bifidobacterium. Pre-treatment of mucin secreted from bovine submaxillary glands (BSM) using His6 -tagged-Est and -SiaBb2 released a higher amount of sialic acid compared to the pre-treatment by His6 -SiaBb2. Growth of B. breve increased with an increase in nanE expression when supplemented with both His6 -Est- and His6 -SiaBb2-treated BSM. These results indicate that the esterase activity of the SiaBb1 Est domain enhances the efficiency of SiaBb2 to cleave sialic acid from mucin. This free sialic acid can be utilized by coexisting sialic acid scavenging B. breve via cross-feeding. Here, we provide the molecular mechanism underlying the unique sialoglycan degradation property of B. bifidum which is mediated by the complementary activities of SiaBb1 and SiaBb2 in the context of sialic acid cross-feeding.


Assuntos
Bifidobacterium bifidum , Bifidobacterium breve , Acetilesterase/genética , Acetilesterase/metabolismo , Animais , Bifidobacterium bifidum/metabolismo , Bifidobacterium breve/metabolismo , Bovinos , Proliferação de Células , Mucinas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/genética , Neuraminidase/metabolismo , Ácidos Siálicos/metabolismo
13.
Mol Microbiol ; 77(4): 855-72, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20545857

RESUMO

Chromobacterium violaceum is a Gram-negative bacterium that causes fatal septicaemia in humans and animals. C. violaceum ATCC 12472 possesses genes associated with two distinct type III secretion systems (T3SSs). One of these systems is encoded by Chromobacterium pathogenicity islands 1 and 1a (Cpi-1/-1a), another is encoded by Chromobacterium pathogenicity island 2 (Cpi-2). Here we show that C. violaceum causes fulminant hepatitis in a mouse infection model, and Cpi-1/-1a-encoded T3SS is required for its virulence. In addition, using C. violaceum strains with defined mutations in the genes that encode the Cpi-1/-1a or Cpi-2 locus in combination with cultured mammalian cell lines, we found that C. violaceum is able to induce cytotoxicity in a Cpi-1/-1a-dependent manner. Characterization of Chromobacterium-induced cytotoxicity revealed that cell lysis by C. violaceum infection involves the formation of pore structures on the host cell membrane, as demonstrated by protection by cytotoxicity in the presence of osmoprotectants. Finally, we demonstrated that CipB, a Cpi-1/-1a effector, is implicated in translocator-mediated pore formation and the ability of CipB to form a pore is essential for Chromobacterium-induced cytotoxicity. These results strongly suggest that Cpi-1/-1a-encoded T3SS is a virulence determinant that causes fatal infection by the induction of cell death in hepatocytes.


Assuntos
Sistemas de Secreção Bacterianos , Chromobacterium/crescimento & desenvolvimento , Chromobacterium/genética , Ilhas Genômicas , Fatores de Virulência/metabolismo , Animais , Secreções Corporais , Morte Celular , Chromobacterium/patogenicidade , Modelos Animais de Doenças , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Negativas/patologia , Hepatite Animal/microbiologia , Hepatite Animal/patologia , Hepatócitos/microbiologia , Hepatócitos/fisiologia , Camundongos , Virulência , Fatores de Virulência/genética
14.
PLoS One ; 16(3): e0248975, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33735297

RESUMO

Nuclear factor-kappa B (NF-κB) plays a critical role in the host defense against microbial pathogens. Many pathogens modulate NF-κB signaling to establish infection in their host. Salmonella enterica serovar Typhimurium (S. Typhimurium) possesses two type III secretion systems (T3SS-1 and T3SS-2) and directly injects many effector proteins into host cells. It has been reported that some effectors block NF-κB signaling, but the molecular mechanism of the inactivation of NF-κB signaling in S. Typhimurium is poorly understood. Here, we identified seven type III effectors-GogA, GtgA, PipA, SseK1, SseK2, SseK3, and SteE-that inhibited NF-κB activation in HeLa cells stimulated with TNF-α. We also determined that only GogA and GtgA are involved in regulation of the activation of NF-κB in HeLa cells infected with S. Typhimurium. GogA, GtgA, and PipA are highly homologous to one another and have the consensus zinc metalloprotease HEXXH motif. Our experiments demonstrated that GogA, GtgA, and PipA each directly cleaved NF-κB p65, whereas GogA and GtgA, but not PipA, inhibited the NF-κB activation in HeLa cells infected with S. Typhimurium. Further, expressions of the gogA or gtgA gene were induced under the SPI-1-and SPI-2-inducing conditions, but expression of the pipA gene was induced only under the SPI-2-inducing condition. We also showed that PipA was secreted into RAW264.7 cells through T3SS-2. Finally, we indicated that PipA elicits bacterial dissemination in the systemic stage of infection of S. Typhimurium via a T3SS-1-independent mechanism. Collectively, our results suggest that PipA, GogA and GtgA contribute to S. Typhimurium pathogenesis in different ways.


Assuntos
Proteínas de Bactérias/metabolismo , Salmonella typhimurium/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Animais , Células HeLa , Humanos , Metaloproteases/metabolismo , Camundongos , Camundongos Endogâmicos CBA , NF-kappa B/metabolismo , Células RAW 264.7 , Infecções por Salmonella/metabolismo , Zinco/metabolismo
15.
BMC Microbiol ; 10: 324, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21176126

RESUMO

BACKGROUND: The global ppGpp-mediated stringent response in pathogenic bacteria plays an important role in the pathogenesis of bacterial infections. In Salmonella enterica serovar Typhimurium (S. Typhimurium), several genes, including virulence genes, are regulated by ppGpp when bacteria are under the stringent response. To understand the control of virulence genes by ppGpp in S. Typhimurium, agarose 2-dimensional electrophoresis (2-DE) combined with mass spectrometry was used and a comprehensive 2-DE reference map of amino acid-starved S. Typhimurium strain SH100, a derivative of ATCC 14028, was established. RESULTS: Of the 366 examined spots, 269 proteins were successfully identified. The comparative analysis of the wild-type and ppGpp0 mutant strains revealed 55 proteins, the expression patterns of which were affected by ppGpp. Using a mouse infection model, we further identified a novel virulence-associated factor, STM3169, from the ppGpp-regulated and Salmonella-specific proteins. In addition, Salmonella strains carrying mutations in the gene encoding STM3169 showed growth defects and impaired growth within macrophage-like RAW264.7 cells. Furthermore, we found that expression of stm3169 was controlled by ppGpp and SsrB, a response regulator of the two-component system located on Salmonella pathogenicity island 2. CONCLUSIONS: A proteomic approach using a 2-DE reference map can prove a powerful tool for analyzing virulence factors and the regulatory network involved in Salmonella pathogenesis. Our results also provide evidence of a global response mediated by ppGpp in S. enterica.


Assuntos
Proteínas de Bactérias/metabolismo , Nucleotídeos de Guanina/deficiência , Macrófagos/microbiologia , Viabilidade Microbiana , Mutação , Proteômica , Infecções por Salmonella/microbiologia , Salmonella typhimurium/metabolismo , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Linhagem Celular , Eletroforese em Gel Bidimensional , Feminino , Regulação Bacteriana da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Salmonella typhimurium/química , Salmonella typhimurium/genética , Fatores de Virulência/química , Fatores de Virulência/genética
16.
J Crohns Colitis ; 14(10): 1462-1472, 2020 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-32227170

RESUMO

BACKGROUND AND AIMS: Regenerating islet-derived protein type 3 [Reg3] lectins are antimicrobial peptides at mucosal surfaces of the gut, whose expression is regulated by pathogenic gut microbes via interleukin-22- or Toll-like receptor signalling. In addition to antimicrobial effects, tissue protection is hypothesized, but has been poorly investigated in the gut. METHODS: We applied antibiotic-induced microbiota perturbations, gnotobiotic approaches and a dextran-sodium sulfate [DSS] colitis model to assess microbial Reg3 regulation in the intestines and its role in colitis. We also used an intestinal organoid model to investigate this axis in vitro. RESULTS: First, we studied whether gut commensals are involved in Reg3 expression in mice, and found that antibiotic-mediated reduction of Clostridia downregulated intestinal Reg3B. A loss in Clostridia was accompanied by a significant reduction of short-chain fatty acids [SCFAs], and knock-out [KO] mice for SCFA receptors GPR43 and GPR109 expressed less intestinal Reg3B/-G. Propionate was found to induce Reg3 in intestinal organoids and in gnotobiotic mice colonized with a defined, SCFA-producing microbiota. Investigating the role of Reg3B as a protective factor in colitis, we found that Reg3B-KO mice display increased inflammation and less crypt proliferation in the DSS colitis model. Propionate decreased colitis and increased proliferation. Treatment of organoids exposed to DSS with Reg3B or propionate reversed the chemical injury with a loss of expression of the stem-cell marker Lgr5 and Olfm4. CONCLUSIONS: Our results suggest that Clostridia can regulate Reg3-associated epithelial homeostasis through propionate signalling. We also provide evidence that the Reg3-propionate axis may be an important mediator of gut epithelial regeneration in colitis.


Assuntos
Colite , Microbioma Gastrointestinal/fisiologia , Interleucinas/metabolismo , Mucosa Intestinal , Proteínas Associadas a Pancreatite/imunologia , Propionatos , Receptores Toll-Like/metabolismo , Animais , Proliferação de Células , Colite/imunologia , Colite/microbiologia , Colite/terapia , Modelos Animais de Doenças , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Lectinas/imunologia , Camundongos , Propionatos/metabolismo , Propionatos/farmacologia , Fatores de Proteção , Transdução de Sinais/imunologia , Interleucina 22
17.
J Bacteriol ; 191(22): 6843-54, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19767440

RESUMO

The type III secretion system (T3SS) encoded by Salmonella pathogenicity island 2 (SPI-2) is involved in systemic infection and intracellular replication of Salmonella enterica serovar Typhimurium. In this study, we investigated the function of SsaE, a small cytoplasmic protein encoded within the SPI-2 locus, which shows structural similarity to the T3SS class V chaperones. An S. enterica serovar Typhimurium ssaE mutant failed to secrete SPI-2 translocator SseB and SPI-2-dependent effector PipB proteins. Coimmunoprecipitation and mass spectrometry analyses using an SsaE-FLAG fusion protein indicated that SsaE interacts with SseB and a putative T3SS-associated ATPase, SsaN. A series of deleted and point-mutated SsaE-FLAG fusion proteins revealed that the C-terminal coiled-coil domain of SsaE is critical for protein-protein interactions. Although SseA was reported to be a chaperone for SseB and to be required for its secretion and stability in the bacterial cytoplasm, an sseA deletion mutant was able to secrete the SseB in vitro when plasmid-derived SseB was overexpressed. In contrast, ssaE mutant strains could not transport SseB extracellularly under the same assay conditions. In addition, an ssaE(I55G) point-mutated strain that expresses the SsaE derivative lacking the ability to form a C-terminal coiled-coil structure showed attenuated virulence comparable to that of an SPI-2 T3SS null mutant, suggesting that the coiled-coil interaction of SsaE is absolutely essential for the functional SPI-2 T3SS and for Salmonella virulence. Based on these findings, we propose that SsaE recognizes translocator SseB and controls its secretion via SPI-2 type III secretion machinery.


Assuntos
Proteínas de Bactérias/fisiologia , Ilhas Genômicas/genética , Salmonella typhimurium/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Transporte Biológico/genética , Transporte Biológico/fisiologia , Imunoprecipitação , Espectrometria de Massas , Microscopia de Fluorescência , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Dados de Sequência Molecular , Ligação Proteica/genética , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , Salmonella typhimurium/genética , Homologia de Sequência de Aminoácidos
18.
Gut Microbes ; 9(2): 179-187, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-28985140

RESUMO

In the inflamed gut, the bactericidal lectin RegIIIß is massively produced by intestinal mucosa. RegIIIß binds peptidoglycan and lipid A respectively, and thus can kill certain Gram-positive and Gram-negative bacteria, including the gut commensal microbiota and enteropathogenic bacteria. Considering the expression pattern and bactericidal activity, RegIIIß is believed to be a host defense factor for protecting against the infection with enteropathogenic bacteria. However, it was poorly understood how RegIIIß recognizes the target bacteria and kill them, and how RegIIIß plays role(s) in infectious diarrhea. Therefore, our recent study has focused on RegIIIß-target recognition, killing of Gram-negative bacteria, and host protective functions of RegIIIß for infectious diarrhea inflicted by Salmonella Typhimurium. Here, we discuss novel insights into the protective role of RegIIIß in infectious diarrhea, and propose avenues towards novel therapeutic interventions for Salmonella diarrhea.


Assuntos
Antibacterianos/metabolismo , Diarreia/microbiologia , Mucosa Intestinal/microbiologia , Proteínas/metabolismo , Salmonelose Animal/microbiologia , Salmonella typhimurium/crescimento & desenvolvimento , Animais , Translocação Bacteriana , Bacteroides/crescimento & desenvolvimento , Bacteroides/metabolismo , Microbioma Gastrointestinal/fisiologia , Homeostase , Mucosa Intestinal/imunologia , Camundongos , Mutação , Proteínas/genética , Salmonella typhimurium/fisiologia , Vitamina B 6/biossíntese
19.
PLoS One ; 12(12): e0190095, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29267354

RESUMO

Mucosal barrier formed by cationic antimicrobial peptides (CAMPs) is believed to be crucial for host protection from pathogenic gut infection. However, some pathogens can develop resistance to the CAMPs to survive in hosts. Salmonella enterica is a common cause of acute diarrhea. During the course of this disease, the pathogen must continuously colonize the gut lumen, which contains CAMPs. However, it is incompletely understood whether the resistance of Salmonella strains to CAMPs contributes to the development of gut infections. PhoPQ two-component system-dependent lipid A modifications confer resistance to CAMPs in S. enterica serovar Typhimurium. Therefore, we introduced mutations into the PhoPQ-regulated genes in an S. Typhimurium strain, obtaining pagP ugtL and pmrA mutant strains. Each mutant strain demonstrated a distinct spectrum of the resistance to CAMPs. Using streptomycin mouse model for Salmonella diarrhea, we show that the pagP ugtL, but not pmrA, mutant strain had a gut colonization defect. Furthermore, the pagP ugtL, but not pmrA, mutant strain had decreased outer membrane integrity and susceptibility to magainin 2, an alpha-helical CAMP. Taken together, the PagP- and UgtL-dependent resistance to CAMPs was demonstrated to contribute to sustained colonization in the gut. This may be due to the robust outer membrane of S. Typhimurium, inducing the resistance to alpha-helical CAMPs such as α-defensins. Our findings indicate that the development of resistance to CAMPs is required for the S. Typhimurium gut infection.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Intestinos/microbiologia , Peptídeos/farmacologia , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/metabolismo
20.
Cell Host Microbe ; 21(2): 195-207, 2017 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-28111202

RESUMO

The bactericidal lectin RegIIIß is inducibly produced by intestinal epithelial cells as a defense against infection by enteropathogens. In the gut lumen, RegIIIß kills not only certain enteropathogens, but also some commensal bacteria; thus, RegIIIß is also thought to be an innate immune effector shaping microbiota composition and establishing intestinal homeostasis. Using the streptomycin mouse model for Salmonella colitis, we show that RegIIIß can promote sustained gut colonization of Salmonella Typhimurium and prolong enteropathy. RegIIIß expression was associated with suppression of Bacteroides spp. in the gut lumen, prolonged disease-associated alterations in colonic metabolism, and reduced luminal vitamin B6 levels. Supplementation with Bacteroides spp. or vitamin B6 accelerated pathogen clearance from the gut and remission of enteropathy. Our findings indicate that interventions at the level of RegIIIß and supplementation with Bacteroides spp. or vitamin B6 might open new avenues for therapeutic intervention in the context of Salmonella colitis.


Assuntos
Antibacterianos/farmacologia , Colite/tratamento farmacológico , Lectinas/farmacologia , Salmonelose Animal/tratamento farmacológico , Animais , Bacteroides/crescimento & desenvolvimento , Colite/microbiologia , Colo/microbiologia , Diarreia/tratamento farmacológico , Diarreia/microbiologia , Modelos Animais de Doenças , Microbioma Gastrointestinal , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Probióticos/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Salmonelose Animal/microbiologia , Salmonella enterica/efeitos dos fármacos , Salmonella enterica/crescimento & desenvolvimento , Estreptomicina , Vitamina B 6/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA