Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Appl Environ Microbiol ; 87(16): e0081221, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34106011

RESUMO

Increasing problems with antibiotic resistance have directed interest toward phage therapy in the aquaculture industry. However, phage resistance evolving in target bacteria is considered a challenge. To investigate how phage resistance influences the fish pathogen Flavobacterium columnare, two wild-type bacterial isolates, FCO-F2 and FCO-F9, were exposed to phages (FCO-F2 to FCOV-F2, FCOV-F5, and FCOV-F25, and FCO-F9 to FCL-2, FCOV-F13, and FCOV-F45), and resulting phenotypic and genetic changes in bacteria were analyzed. Bacterial viability first decreased in the exposure cultures but started to increase after 1 to 2 days, along with a change in colony morphology from original rhizoid to rough, leading to 98% prevalence of the rough morphotype. Twenty-four isolates (including four isolates from no-phage treatments) were further characterized for phage resistance, antibiotic susceptibility, motility, adhesion, and biofilm formation, protease activity, whole-genome sequencing, and virulence in rainbow trout fry. The rough isolates arising in phage exposure were phage resistant with low virulence, whereas rhizoid isolates maintained phage susceptibility and high virulence. Gliding motility and protease activity were also related to the phage susceptibility. Observed mutations in phage-resistant isolates were mostly located in genes encoding the type IX secretion system, a component of the Bacteroidetes gliding motility machinery. However, not all phage-resistant isolates had mutations, indicating that phage resistance in F. columnare is a multifactorial process, including both genetic mutations and changes in gene expression. Phage resistance may not, however, be a challenge for development of phage therapy against F. columnare infections since phage resistance is associated with decreases in bacterial virulence. IMPORTANCE Phage resistance of infectious bacteria is a common phenomenon posing challenges for the development of phage therapy. Along with a growing world population and the need for increased food production, constantly intensifying animal farming has to face increasing problems of infectious diseases. Columnaris disease, caused by Flavobacterium columnare, is a worldwide threat for salmonid fry and juvenile farming. Without antibiotic treatments, infections can lead to 100% mortality in a fish stock. Phage therapy of columnaris disease would reduce the development of antibiotic-resistant bacteria and antibiotic loads by the aquaculture industry, but phage-resistant bacterial isolates may become a risk. However, phenotypic and genetic characterization of phage-resistant F. columnare isolates in this study revealed that they are less virulent than phage-susceptible isolates and thus not a challenge for phage therapy against columnaris disease. This is valuable information for the fish farming industry globally when considering phage-based prevention and curing methods for F. columnare infections.


Assuntos
Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos/genética , Bacteriófagos/fisiologia , Doenças dos Peixes/microbiologia , Infecções por Flavobacteriaceae/veterinária , Flavobacterium/citologia , Flavobacterium/patogenicidade , Flavobacterium/virologia , Animais , Proteínas de Bactérias/imunologia , Sistemas de Secreção Bacterianos/imunologia , Bacteriófagos/genética , Peixes , Infecções por Flavobacteriaceae/microbiologia , Flavobacterium/imunologia , Mutação , Virulência
2.
Acta Crystallogr F Struct Biol Commun ; 77(Pt 6): 171-176, 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-34100775

RESUMO

GldL is an inner-membrane protein that is essential for the function of the type IX secretion system (T9SS) in Flavobacterium johnsoniae. The complex that it forms with GldM is supposed to act as a new rotary motor involved in the gliding motility of the bacterium. In the context of structural studies of GldL to gain information on the assembly and function of the T9SS, two camelid nanobodies were selected, produced and purified. Their interaction with the cytoplasmic domain of GldL was characterized and their crystal structures were solved. These nanobodies will be used as crystallization chaperones to help in the crystallization of the cytoplasmic domain of GldL and could also help to solve the structure of the complex using molecular replacement.


Assuntos
Proteínas de Bactérias/imunologia , Sistemas de Secreção Bacterianos/imunologia , Flavobacterium/química , Anticorpos de Domínio Único/química , Animais , Camelus , Cristalografia por Raios X , Cinética , Modelos Moleculares , Domínios Proteicos , Multimerização Proteica , Espalhamento de Radiação , Termodinâmica
3.
Trends Microbiol ; 27(2): 176-187, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30442534

RESUMO

The excretion of cytoplasmic and signal-peptide-less proteins (ECP) by microorganisms and eukaryotes remains a fascinating topic. In principle, it appears to be a waste of energy. However, it turns out that - extracellularly - some cytoplasmic proteins (CPs) exert a completely different function such as contributing to pathogenicity or evasion of the immune system. Such CPs have been referred to as 'moonlighting' proteins. ECP is boosted by many endogenous or external factors that impair the membrane or cell wall structure. There are also differences regarding their mode of release. In some microorganisms they appear to be released directly, while in others they are embedded in membrane vesicles, or bound to the cell envelope. Some CPs might be promising candidates for vaccine developments against major bacterial pathogens.


Assuntos
Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/metabolismo , Citoplasma/metabolismo , Proteínas de Bactérias/imunologia , Sistemas de Secreção Bacterianos/imunologia , Vacinas Bacterianas , Biofilmes , Membrana Celular/imunologia , Membrana Celular/metabolismo , Parede Celular/imunologia , Parede Celular/metabolismo , Proteínas da Matriz Extracelular , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/fisiologia , Evasão da Resposta Imune , Ligação Proteica
4.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 12): 810-816, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30511676

RESUMO

The type VI secretion system (T6SS) comprises needle-shaped multisubunit complexes that play a role in the microbial defense systems of Gram-negative bacteria. Some Gram-negative bacteria harboring a T6SS deliver toxic effector proteins into the cytoplasm or periplasm of competing bacteria in order to lyse and kill them. To avoid self-cell disruption, these bacteria have cognate immunity proteins that inhibit their toxic effector proteins. T6SS amidase effector protein 4 (Tae4) and T6SS amidase immunity protein 4 (Tai4) are a representative of the toxic effector-immunity pairs of the T6SS. Here, the three-dimensional structures of Tai4 and the Tae4-Tai4 complex from Agrobacterium tumefaciens are reported at 1.55 and 1.9 Šresolution, respectively. A structural comparison with other Tae4-Tai4 homologs revealed similarities and differences in the catalytic and inhibitory mechanisms among the Tae4 and Tai4 family proteins.


Assuntos
Agrobacterium tumefaciens/química , Agrobacterium tumefaciens/imunologia , Proteínas de Bactérias/química , Proteínas de Bactérias/imunologia , Sistemas de Secreção Bacterianos/química , Sistemas de Secreção Bacterianos/imunologia , Agrobacterium tumefaciens/genética , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos/genética , Cristalização , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
5.
PLoS Pathog ; 14(6): e1007139, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29912964

RESUMO

Tuberculosis is the deadliest infectious disease worldwide. Although the BCG vaccine is widely used, it does not efficiently protect against pulmonary tuberculosis and an improved tuberculosis vaccine is therefore urgently needed. Mycobacterium tuberculosis uses different ESX/Type VII secretion (T7S) systems to transport proteins important for virulence and host immune responses. We recently reported that secretion of T7S substrates belonging to the mycobacteria-specific Pro-Glu (PE) and Pro-Pro-Glu (PPE) proteins of the PGRS (polymorphic GC-rich sequences) and MPTR (major polymorphic tandem repeat) subfamilies required both a functional ESX-5 system and a functional PPE38/71 protein for secretion. Inactivation of ppe38/71 and the resulting loss of PE_PGRS/PPE-MPTR secretion were linked to increased virulence of M. tuberculosis strains. Here, we show that a predicted total of 89 PE_PGRS/PPE-MPTR surface proteins are not exported by certain animal-adapted strains of the M. tuberculosis complex including M. bovis. This Δppe38/71-associated secretion defect therefore also occurs in the M. bovis-derived tuberculosis vaccine BCG and could be partially restored by introduction of the M. tuberculosis ppe38-locus. Epitope mapping of the PPE-MPTR protein PPE10, further allowed us to monitor T-cell responses in splenocytes from BCG/M. tuberculosis immunized mice, confirming the dependence of PPE10-specific immune-induction on ESX-5/PPE38-mediated secretion. Restoration of PE_PGRS/PPE-MPTR secretion in recombinant BCG neither altered global antigenic presentation or activation of innate immune cells, nor protective efficacy in two different mouse vaccination-infection models. This unexpected finding stimulates a reassessment of the immunomodulatory properties of PE_PGRS/PPE-MPTR proteins, some of which are contained in vaccine formulations currently in clinical evaluation.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/imunologia , Proteínas de Membrana/imunologia , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose/imunologia , Animais , Proteínas de Bactérias/genética , Feminino , Genoma Bacteriano , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Família Multigênica , Tuberculose/prevenção & controle , Virulência
6.
Cell Rep ; 23(4): 1072-1084, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29694886

RESUMO

The pathogenic potential of Mycobacterium tuberculosis largely depends on ESX secretion systems exporting members of the multigenic Esx, Esp, and PE/PPE protein families. To study the secretion and regulation patterns of these proteins while circumventing immune cross-reactions due to their extensive sequence homologies, we developed an approach that relies on the recognition of their MHC class II epitopes by highly discriminative T cell receptors (TCRs) of a panel of T cell hybridomas. The latter were engineered so that each expresses a unique fluorescent reporter linked to specific antigen recognition. The resulting polychromatic and multiplexed imaging assay enabled us to measure the secretion of mycobacterial effectors inside infected host cells. We applied this novel technology to a large panel of mutants, clinical isolates, and host-cell types to explore the host-mycobacteria interplay and its impact on the intracellular bacterial secretome, which also revealed the unexpected capacity of phagocytes from lung granuloma to present mycobacterial antigens via MHC class II.


Assuntos
Sistemas de Secreção Bacterianos/imunologia , Epitopos de Linfócito T/imunologia , Granuloma do Sistema Respiratório , Mycobacterium tuberculosis/imunologia , Fagócitos , Tuberculose Pulmonar , Animais , Linhagem Celular Tumoral , Granuloma do Sistema Respiratório/imunologia , Granuloma do Sistema Respiratório/microbiologia , Granuloma do Sistema Respiratório/patologia , Antígenos de Histocompatibilidade Classe II/imunologia , Camundongos , Fagócitos/imunologia , Fagócitos/microbiologia , Fagócitos/patologia , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/patologia
7.
Curr Issues Mol Biol ; 25: 61-80, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28875940

RESUMO

Macrophages represent one of the first lines of host immune defenses against the invasion of pathogenic bacteria. Many receptors, immune signaling pathways and cellular processes in macrophages, including Toll-like receptors, Nod-like receptors, phagocytosis, autophagy and programmed cell death, are involved in combating the infection of bacterial pathogens. For efficient colonization in the host, bacterial pathogens have evolved diverse mechanisms to interfere with macrophage functions to evade host defenses. The major weapons utilized by bacterial pathogens are protein toxins and effectors secreted via specific bacterial secretion systems, including type I-VII secretion apparatuses. In recent years, great advances have been achieved in understanding how bacterial toxins and effectors subvert immune signaling and cellular processes of macrophages. In this review, we focus on the toxins and effectors that modulate the phagocytosis, intracellular immune signaling pathways, autophagy and programmed cell death processes of macrophages from the bacterium Legionella pneumophila, Shigella flexneri, Listeria monocytogenes, Salmonella spp., Yersinia spp., enteropathogenic E. coli and Mycobacterium tuberculosis.


Assuntos
Sistemas de Secreção Bacterianos/imunologia , Toxinas Bacterianas/biossíntese , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Positivas/imunologia , Evasão da Resposta Imune , Macrófagos/imunologia , Animais , Apoptose , Autofagia , Sistemas de Secreção Bacterianos/genética , Toxinas Bacterianas/genética , Regulação da Expressão Gênica , Bactérias Gram-Negativas/crescimento & desenvolvimento , Bactérias Gram-Positivas/crescimento & desenvolvimento , Humanos , Imunidade Inata , Macrófagos/microbiologia , Macrófagos/patologia , Proteínas NLR/genética , Proteínas NLR/imunologia , Fagocitose , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
8.
J Biol Chem ; 292(40): 16677-16687, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28842484

RESUMO

Numerous Gram-negative pathogens infect eukaryotes and use the type III secretion system (T3SS) to deliver effector proteins into host cells. One important T3SS feature is an extracellular needle with an associated tip complex responsible for assembly of a pore-forming translocon in the host cell membrane. Shigella spp. cause shigellosis, also called bacillary dysentery, and invade colonic epithelial cells via the T3SS. The tip complex of Shigella flexneri contains invasion plasmid antigen D (IpaD), which initially regulates secretion and provides a physical platform for the translocon pore. The tip complex represents a promising therapeutic target for many important T3SS-containing pathogens. Here, in an effort to further elucidate its function, we created a panel of single-VH domain antibodies (VHHs) that recognize distinct epitopes within IpaD. These VHHs recognized the in situ tip complex and modulated the infectious properties of Shigella Moreover, structural elucidation of several IpaD-VHH complexes provided critical insights into tip complex formation and function. Of note, one VHH heterodimer could reduce Shigella hemolytic activity by >80%. Our observations along with previous findings support the hypothesis that the hydrophobic translocator (IpaB in Shigella) likely binds to a region within the tip protein that is structurally conserved across all T3SS-possessing pathogens, suggesting potential therapeutic avenues for managing infections by these pathogens.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Sistemas de Secreção Bacterianos/imunologia , Epitopos/imunologia , Shigella flexneri/imunologia , Anticorpos de Cadeia Única/imunologia , Animais , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos/genética , Camelídeos Americanos , Evolução Molecular Direcionada , Epitopos/genética , Shigella flexneri/genética
9.
Infect Immun ; 84(1): 230-40, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26502912

RESUMO

The oral Gram-negative anaerobic bacterium Porphyromonas gingivalis is an important pathogen involved in chronic periodontitis. Among its virulence factors, the major extracellular proteinases, Arg-gingipain and Lys-gingipain, are of interest given their abilities to degrade host proteins and process other virulence factors. Gingipains possess C-terminal domains (CTDs) and are translocated to the cell surface or into the extracellular milieu by the type IX secretion system (T9SS). Gingipains contribute to the colonial pigmentation of the bacterium on blood agar. In this study, Omp17, the PGN_0300 gene product, was found in the outer membrane fraction. A mutant lacking Omp17 did not show pigmentation on blood agar and showed reduced proteolytic activity of the gingipains. CTD-containing proteins were released from bacterial cells without cleavage of the CTDs in the omp17 mutant. Although synthesis of the anionic polysaccharide (A-LPS) was not affected in the omp17 mutant, the processing of and A-LPS modification of CTD-containing proteins was defective. PorU, a C-terminal signal peptidase that cleaves the CTDs of other CTD-containing proteins, was not detected in any membrane fraction of the omp17 mutant, suggesting that the defective maturation of CTD-containing proteins by impairment of Omp17 is partly due to loss of function of PorU. In the mouse subcutaneous infection experiment, the omp17 mutant was less virulent than the wild type. These results suggested that Omp17 is involved in P. gingivalis virulence.


Assuntos
Adesinas Bacterianas/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Sistemas de Secreção Bacterianos/imunologia , Cisteína Endopeptidases/metabolismo , Porphyromonas gingivalis/patogenicidade , Fatores de Virulência/genética , Adesinas Bacterianas/imunologia , Animais , Cisteína Endopeptidases/imunologia , Cisteína Endopeptidases Gingipaínas , Camundongos , Camundongos Endogâmicos BALB C , Periodontite/microbiologia , Transporte Proteico
10.
PLoS One ; 10(10): e0139803, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26451946

RESUMO

Shiga toxin-producing Escherichia coli (STEC) serotype O103 is a zoonotic pathogen that is capable of causing hemorrhagic colitis and hemolytic uremic syndrome (HUS) in humans. The main animal reservoir for STEC is ruminants and hence reducing the levels of this pathogen in cattle could ultimately lower the risk of STEC infection in humans. During the process of infection, STECO103 uses a Type III Secretion System (T3SS) to secrete effector proteins (T3SPs) that result in the formation of attaching and effacing (A/E) lesions. Vaccination of cattle with STEC serotype O157 T3SPs has previously been shown to be effective in reducing shedding of STECO157 in a serotype-specific manner. In this study, we tested the ability of rabbit polyclonal sera against individual STECO103 T3SPs to block adherence of the organism to HEp-2 cells. Our results demonstrate that pooled sera against EspA, EspB, EspF, NleA and Tir significantly lowered the adherence of STECO103 relative to pre-immune sera. Likewise, pooled anti-STECO103 sera were also able to block adherence by STECO157. Vaccination of mice with STECO103 recombinant proteins induced strong IgG antibody responses against EspA, EspB, NleA and Tir but not against EspF. However, the vaccine did not affect fecal shedding of STECO103 compared to the PBS vaccinated group over the duration of the experiment. Cross reactivity studies using sera against STECO103 recombinant proteins revealed a high degree of cross reactivity with STECO26 and STECO111 proteins implying that sera against STECO103 proteins could potentially provide neutralization of attachment to epithelial cells by heterologous STEC serotypes.


Assuntos
Anticorpos Antibacterianos/farmacologia , Sistemas de Secreção Bacterianos/imunologia , Escherichia coli Êntero-Hemorrágica/imunologia , Proteínas de Escherichia coli/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Aderência Bacteriana/imunologia , Bovinos , Linhagem Celular , Reações Cruzadas , Escherichia coli Êntero-Hemorrágica/patogenicidade , Vacinas contra Escherichia coli/imunologia , Vacinas contra Escherichia coli/farmacologia , Humanos , Imunoglobulina G/imunologia , Camundongos , Coelhos
11.
J Immunol ; 195(3): 815-9, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26109648

RESUMO

Murine NLR family, apoptosis inhibitory protein (Naip)1, Naip2, and Naip5/6 are host sensors that detect the cytosolic presence of needle and rod proteins from bacterial type III secretion systems and flagellin, respectively. Previous studies using human-derived macrophage-like cell lines indicate that human macrophages sense the cytosolic needle protein, but not bacterial flagellin. In this study, we show that primary human macrophages readily sense cytosolic flagellin. Infection of primary human macrophages with Salmonella elicits robust cell death and IL-1ß secretion that is dependent on flagellin. We show that flagellin detection requires a full-length isoform of human Naip. This full-length Naip isoform is robustly expressed in primary macrophages from healthy human donors, but it is drastically reduced in monocytic tumor cells, THP-1, and U937, rendering them insensitive to cytosolic flagellin. However, ectopic expression of full-length Naip rescues the ability of U937 cells to sense flagellin. In conclusion, human Naip functions to activate the inflammasome in response to flagellin, similar to murine Naip5/6.


Assuntos
Sistemas de Secreção Bacterianos/imunologia , Flagelina/imunologia , Inflamassomos/imunologia , Macrófagos/imunologia , Proteína Inibidora de Apoptose Neuronal/imunologia , Células Cultivadas , Humanos , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Proteína Inibidora de Apoptose Neuronal/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Salmonella/imunologia , Infecções por Salmonella/imunologia , Células U937
12.
Infect Immun ; 83(4): 1507-22, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25644012

RESUMO

The type III secretion system is employed by many pathogens, including the genera Yersinia, Shigella, Pseudomonas, and Salmonella, to deliver effector proteins into eukaryotic cells. The injectisome needle is formed by the polymerization of a single protein, e.g., YscF (Yersinia pestis), PscF (Pseudomonas aeruginosa), PrgI (Salmonella enterica SPI-1), SsaG (Salmonella enterica SPI-2), or MxiH (Shigella flexneri). In this study, we demonstrated that the N termini of some needle proteins, particularly the N terminus of YscF from Yersinia pestis, influences host immune responses. The N termini of several needle proteins were truncated and tested for the ability to induce inflammatory responses in a human monocytic cell line (THP-1 cells). Truncated needle proteins induced proinflammatory cytokines to different magnitudes than the corresponding wild-type proteins, except SsaG. Notably, N-terminally truncated YscF induced significantly higher activation of NF-κB and/or AP-1 and higher induction of proinflammatory cytokines, suggesting that a function of the N terminus of YscF is interference with host sensing of YscF, consistent with Y. pestis pathogenesis. To directly test the ability of the N terminus of YscF to suppress cytokine induction, a YscF-SsaG chimera with 15 N-terminal amino acids from YscF added to SsaG was constructed. The chimeric YscF-SsaG induced lower levels of cytokines than wild-type SsaG. However, the addition of 15 random amino acids to SsaG had no effect on NF-κB/AP-1 activation. These results suggest that the N terminus of YscF can function to decrease cytokine induction, perhaps contributing to a favorable immune environment leading to survival of Y. pestis within the eukaryotic host.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Citocinas/metabolismo , Proteínas de Membrana/imunologia , Proteínas Recombinantes de Fusão/imunologia , Sequência de Aminoácidos , Proteínas de Bactérias/farmacologia , Sistemas de Secreção Bacterianos/imunologia , Linhagem Celular , Citocinas/biossíntese , Células HeLa , Humanos , Evasão da Resposta Imune/genética , Inflamação/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Monócitos/imunologia , NF-kappa B/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Salmonella enterica/genética , Salmonella enterica/imunologia , Salmonella enterica/patogenicidade , Alinhamento de Sequência , Deleção de Sequência/genética , Transdução de Sinais/imunologia , Fator de Transcrição AP-1/metabolismo , Yersinia pestis/genética , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade
13.
PLoS Pathog ; 11(2): e1004650, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25658322

RESUMO

Mycobacterium tuberculosis (Mtb) uses efficient strategies to evade the eradication by professional phagocytes, involving--as recently confirmed--escape from phagosomal confinement. While Mtb determinants, such as the ESX-1 type VII secretion system, that contribute to this phenomenon are known, the host cell factors governing this important biological process are yet unexplored. Using a newly developed flow-cytometric approach for Mtb, we show that macrophages expressing the phagosomal bivalent cation transporter Nramp-1, are much less susceptible to phagosomal rupture. Together with results from the use of the phagosome acidification inhibitor bafilomycin, we demonstrate that restriction of phagosomal acidification is a prerequisite for mycobacterial phagosomal rupture and cytosolic contact. Using different in vivo approaches including an enrichment and screen for tracking rare infected phagocytes carrying the CD45.1 hematopoietic allelic marker, we here provide first and unique evidence of M. tuberculosis-mediated phagosomal rupture in mouse spleen and lungs and in numerous phagocyte types. Our results, linking the ability of restriction of phagosome acidification to cytosolic access, provide an important conceptual advance for our knowledge on host processes targeted by Mtb evasion strategies.


Assuntos
Sistemas de Secreção Bacterianos/imunologia , Proteínas de Transporte de Cátions/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Fagossomos/imunologia , Tuberculose/imunologia , Animais , Proteínas de Transporte de Cátions/genética , Linhagem Celular Tumoral , Humanos , Concentração de Íons de Hidrogênio , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Mutantes , Fagossomos/genética , Tuberculose/genética , Tuberculose/patologia
14.
Infect Immun ; 83(4): 1276-85, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25605762

RESUMO

Melioidosis is an infectious disease of high mortality for humans and other animal species; it is prevalent in tropical regions worldwide. The pathogenesis of melioidosis depends on the ability of its causative agent, the Gram-negative bacterium Burkholderia pseudomallei, to enter and survive in host cells. B. pseudomallei can escape from the phagosome into the cytosol of phagocytic cells where it replicates and acquires actin-mediated motility, avoiding killing by the autophagy-dependent process, LC3 (microtubule-associated protein light chain 3)-associated phagocytosis (LAP). The type III secretion system cluster 3 (TTSS3) facilitates bacterial escape from phagosomes, although the mechanism has not been fully elucidated. Given the recent identification of small-molecule inhibitors of the TTSS ATPase, we sought to determine the potential of the predicted TTSS3 ATPase, encoded by bsaS, as a target for chemotherapeutic treatment of infection. A B. pseudomallei bsaS deletion mutant was generated and used as a control against which to assess the effect of inhibitor treatment. Infection of RAW 264.7 cells with wild-type bacteria and subsequent treatment with the ATPase inhibitor compound 939 resulted in reduced intracellular bacterial survival, reduced escape from phagosomes, and increased colocalization with both LC3 and the lysosomal marker LAMP1 (lysosome-associated membrane protein 1). These changes were similar to those observed for infection of RAW 264.7 cells with the bsaS deletion mutant. We propose that treatment with the ATPase inhibitor compound 939 decreased intracellular bacterial survival through a reduced ability of bacteria to escape from phagosomes and increased killing via LAP. Therefore, small-molecule inhibitors of the TTSS3 ATPase have potential as therapeutic treatments against melioidosis.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Sistemas de Secreção Bacterianos/imunologia , Burkholderia pseudomallei/efeitos dos fármacos , Burkholderia pseudomallei/enzimologia , Melioidose/tratamento farmacológico , Animais , Proteínas de Bactérias/genética , Burkholderia pseudomallei/genética , Linhagem Celular , Feminino , Evasão da Resposta Imune , Estimativa de Kaplan-Meier , Melioidose/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Associadas aos Microtúbulos/imunologia , Fagocitose/imunologia , Fatores de Virulência/genética
16.
PLoS Pathog ; 10(11): e1004479, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25375398

RESUMO

Clinical infections by Pseudomonas aeruginosa, a deadly Gram-negative, opportunistic pathogen of immunocompromised hosts, often involve the formation of antibiotic-resistant biofilms. Although biofilm formation has been extensively studied in vitro on glass or plastic surfaces, much less is known about biofilm formation at the epithelial barrier. We have previously shown that when added to the apical surface of polarized epithelial cells, P. aeruginosa rapidly forms cell-associated aggregates within 60 minutes of infection. By confocal microscopy we now show that cell-associated aggregates exhibit key characteristics of biofilms, including the presence of extracellular matrix and increased resistance to antibiotics compared to planktonic bacteria. Using isogenic mutants in the type III secretion system, we found that the translocon, but not the effectors themselves, were required for cell-associated aggregation on the surface of polarized epithelial cells and at early time points in a murine model of acute pneumonia. In contrast, the translocon was not required for aggregation on abiotic surfaces, suggesting a novel function for the type III secretion system during cell-associated aggregation. Supernatants from epithelial cells infected with wild-type bacteria or from cells treated with the pore-forming toxin streptolysin O could rescue aggregate formation in a type III secretion mutant, indicating that cell-associated aggregation requires one or more host cell factors. Our results suggest a previously unappreciated function for the type III translocon in the formation of P. aeruginosa biofilms at the epithelial barrier and demonstrate that biofilms may form at early time points of infection.


Assuntos
Sistemas de Secreção Bacterianos/imunologia , Biofilmes , Células Epiteliais/imunologia , Pneumonia Bacteriana/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/fisiologia , Animais , Sistemas de Secreção Bacterianos/genética , Modelos Animais de Doenças , Cães , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Células Madin Darby de Rim Canino , Camundongos , Mutação , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/patologia , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/patologia
17.
Nihon Saikingaku Zasshi ; 69(3): 503-11, 2014.
Artigo em Japonês | MEDLINE | ID: mdl-25186641

RESUMO

Legionella are gram-negative bacteria ubiquitously found in freshwater and soil environments. Once inhaled by humans, Legionella infection could result in a severe form of pneumonia known as Legionellosis. Legionella translocate ~300 effector proteins into host cells via the Dot/Icm type IV secretion system, which is central to Legionella pathogenesis. Here I describe a brief review on recent advances in research on the molecular basis of Legionella-eukaryotic-cell interaction.


Assuntos
Proteínas de Bactérias/imunologia , Proteínas de Bactérias/fisiologia , Sistemas de Secreção Bacterianos/imunologia , Sistemas de Secreção Bacterianos/fisiologia , Células Eucarióticas/imunologia , Células Eucarióticas/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Legionella pneumophila/genética , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Doença dos Legionários/microbiologia , Animais , Água Doce/microbiologia , Humanos , Legionella pneumophila/patogenicidade , Macrófagos Alveolares/microbiologia , Filogenia , Microbiologia do Solo
18.
PLoS Pathog ; 10(8): e1004346, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25165815

RESUMO

The mammalian immune system has the ability to discriminate between pathogens and innocuous microbes by detecting conserved molecular patterns. In addition to conserved microbial patterns, the mammalian immune system may recognize distinct pathogen-induced processes through a mechanism which is poorly understood. Previous studies have shown that a type III secretion system (T3SS) in Yersinia pseudotuberculosis leads to decreased survival of this bacterium in primary murine macrophages by unknown mechanisms. Here, we use colony forming unit assays and fluorescence microscopy to investigate how the T3SS triggers killing of Yersinia in macrophages. We present evidence that Yersinia outer protein E (YopE) delivered by the T3SS triggers intracellular killing response against Yersinia. YopE mimics eukaryotic GTPase activating proteins (GAPs) and inactivates Rho GTPases in host cells. Unlike wild-type YopE, catalytically dead YopER144A is impaired in restricting Yersinia intracellular survival, highlighting that the GAP activity of YopE is detected as a danger signal. Additionally, a second translocated effector, YopT, counteracts the YopE triggered killing effect by decreasing the translocation level of YopE and possibly by competing for the same pool of Rho GTPase targets. Moreover, inactivation of Rho GTPases by Clostridium difficile Toxin B mimics the effect of YopE and promotes increased killing of Yersinia in macrophages. Using a Rac inhibitor NSC23766 and a Rho inhibitor TAT-C3, we show that macrophages restrict Yersinia intracellular survival in response to Rac1 inhibition, but not Rho inhibition. In summary, our findings reveal that primary macrophages sense manipulation of Rho GTPases by Yersinia YopE and actively counteract pathogenic infection by restricting intracellular bacterial survival. Our results uncover a new mode of innate immune recognition in response to pathogenic infection.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Sistemas de Secreção Bacterianos/imunologia , Proteínas Ativadoras de GTPase/imunologia , Imunidade Inata/imunologia , Macrófagos/imunologia , Animais , Immunoblotting , Macrófagos/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Yersinia pseudotuberculosis/imunologia
19.
PLoS Pathog ; 10(7): e1004207, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24992093

RESUMO

Delivery of microbial products into the mammalian cell cytosol by bacterial secretion systems is a strong stimulus for triggering pro-inflammatory host responses. Here we show that Salmonella enterica serovar Typhi (S. Typhi), the causative agent of typhoid fever, tightly regulates expression of the invasion-associated type III secretion system (T3SS-1) and thus fails to activate these innate immune signaling pathways. The S. Typhi regulatory protein TviA rapidly repressed T3SS-1 expression, thereby preventing RAC1-dependent, RIP2-dependent activation of NF-κB in epithelial cells. Heterologous expression of TviA in S. enterica serovar Typhimurium (S. Typhimurium) suppressed T3SS-1-dependent inflammatory responses generated early after infection in animal models of gastroenteritis. These results suggest that S. Typhi reduces intestinal inflammation by limiting the induction of pathogen-induced processes through regulation of virulence gene expression.


Assuntos
Sistemas de Secreção Bacterianos/imunologia , Gastroenterite/imunologia , Imunidade Inata , Salmonella typhi/imunologia , Febre Tifoide/imunologia , Fatores de Virulência/imunologia , Animais , Sistemas de Secreção Bacterianos/genética , Bovinos , Modelos Animais de Doenças , Gastroenterite/genética , Gastroenterite/patologia , Regulação Bacteriana da Expressão Gênica/genética , Regulação Bacteriana da Expressão Gênica/imunologia , Células HeLa , Humanos , Camundongos , Salmonella typhi/genética , Salmonella typhi/patogenicidade , Febre Tifoide/genética , Febre Tifoide/patologia , Fatores de Virulência/genética
20.
Infect Immun ; 82(9): 3927-38, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25001604

RESUMO

Brucella spp. are facultative intracellular Gram-negative coccobacilli responsible for brucellosis, a worldwide zoonosis. We observed that Brucella melitensis is able to persist for several weeks in the blood of intraperitoneally infected mice and that transferred blood at any time point tested is able to induce infection in naive recipient mice. Bacterial persistence in the blood is dramatically impaired by specific antibodies induced following Brucella vaccination. In contrast to Bartonella, the type IV secretion system and flagellar expression are not critically required for the persistence of Brucella in blood. ImageStream analysis of blood cells showed that following a brief extracellular phase, Brucella is associated mainly with the erythrocytes. Examination by confocal microscopy and transmission electron microscopy formally demonstrated that B. melitensis is able to invade erythrocytes in vivo. The bacteria do not seem to multiply in erythrocytes and are found free in the cytoplasm. Our results open up new areas for investigation and should serve in the development of novel strategies for the treatment or prophylaxis of brucellosis. Invasion of erythrocytes could potentially protect the bacterial cells from the host's immune response and hamper antibiotic treatment and suggests possible Brucella transmission by bloodsucking insects in nature.


Assuntos
Brucella melitensis/imunologia , Eritrócitos/imunologia , Animais , Sistemas de Secreção Bacterianos/imunologia , Vacina contra Brucelose/imunologia , Brucelose/imunologia , Brucelose/microbiologia , Eritrócitos/microbiologia , Flagelos/imunologia , Flagelos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...