Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34353909

RESUMO

Perturbation of the endoplasmic reticulum (ER), a central organelle of the cell, can have critical consequences for cellular homeostasis. An elaborate surveillance system known as ER quality control ensures that cells can respond and adapt to stress via the unfolded protein response (UPR) and that only correctly assembled proteins reach their destination. Interestingly, several bacterial pathogens hijack the ER to establish an infection. However, it remains poorly understood how bacterial pathogens exploit ER quality-control functions to complete their intracellular cycle. Brucella spp. replicate extensively within an ER-derived niche, which evolves into specialized vacuoles suited for exit from infected cells. Here we present Brucella-secreted protein L (BspL), a Brucella abortus effector that interacts with Herp, a central component of the ER-associated degradation (ERAD) machinery. We found that BspL enhances ERAD at the late stages of the infection. BspL targeting of Herp and ERAD allows tight control of the kinetics of autophagic Brucella-containing vacuole formation, delaying the last step of its intracellular cycle and cell-to-cell spread. This study highlights a mechanism by which a bacterial pathogen hijacks ERAD components for fine regulation of its intracellular trafficking.


Assuntos
Proteínas de Bactérias/metabolismo , Brucella abortus/patogenicidade , Brucelose/metabolismo , Animais , Proteínas de Bactérias/genética , Brucella abortus/metabolismo , Brucelose/microbiologia , Retículo Endoplasmático/metabolismo , Degradação Associada com o Retículo Endoplasmático , Células HeLa , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Fator de Transcrição CHOP/genética , Sistemas de Secreção Tipo IV/metabolismo , Proteína 1 de Ligação a X-Box/genética
2.
PLoS Pathog ; 16(4): e1007979, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32298382

RESUMO

Brucella species are facultative intracellular Gram-negative bacteria relevant to animal and human health. Their ability to establish an intracellular niche and subvert host cell pathways to their advantage depends on the delivery of bacterial effector proteins through a type IV secretion system. Brucella Toll/Interleukin-1 Receptor (TIR)-domain-containing proteins BtpA (also known as TcpB) and BtpB are among such effectors. Although divergent in primary sequence, they interfere with Toll-like receptor (TLR) signaling to inhibit the innate immune responses. However, the molecular mechanisms implicated still remain unclear. To gain insight into the functions of BtpA and BtpB, we expressed them in the budding yeast Saccharomyces cerevisiae as a eukaryotic cell model. We found that both effectors were cytotoxic and that their respective TIR domains were necessary and sufficient for yeast growth inhibition. Growth arrest was concomitant with actin depolymerization, endocytic block and a general decrease in kinase activity in the cell, suggesting a failure in energetic metabolism. Indeed, levels of ATP and NAD+ were low in yeast cells expressing BtpA and BtpB TIR domains, consistent with the recently described enzymatic activity of some TIR domains as NAD+ hydrolases. In human epithelial cells, both BtpA and BtpB expression reduced intracellular total NAD levels. In infected cells, both BtpA and BtpB contributed to reduction of total NAD, indicating that their NAD+ hydrolase functions are active intracellularly during infection. Overall, combining the yeast model together with mammalian cells and infection studies our results show that BtpA and BtpB modulate energy metabolism in host cells through NAD+ hydrolysis, assigning a novel role for these TIR domain-containing effectors in Brucella pathogenesis.


Assuntos
Proteínas de Bactérias/metabolismo , Brucella abortus/crescimento & desenvolvimento , Brucelose/metabolismo , Hidrolases/metabolismo , NAD/metabolismo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Fatores de Virulência/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Brucella abortus/metabolismo , Brucelose/microbiologia , Células HeLa , Humanos , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Fatores de Virulência/genética
3.
EMBO J ; 36(13): 1869-1887, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28483816

RESUMO

Bacterial pathogens often subvert the innate immune system to establish a successful infection. The direct inhibition of downstream components of innate immune pathways is particularly well documented but how bacteria interfere with receptor proximal events is far less well understood. Here, we describe a Toll/interleukin 1 receptor (TIR) domain-containing protein (PumA) of the multi-drug resistant Pseudomonas aeruginosa PA7 strain. We found that PumA is essential for virulence and inhibits NF-κB, a property transferable to non-PumA strain PA14, suggesting no additional factors are needed for PumA function. The TIR domain is able to interact with the Toll-like receptor (TLR) adaptors TIRAP and MyD88, as well as the ubiquitin-associated protein 1 (UBAP1), a component of the endosomal-sorting complex required for transport I (ESCRT-I). These interactions are not spatially exclusive as we show UBAP1 can associate with MyD88, enhancing its plasma membrane localization. Combined targeting of UBAP1 and TLR adaptors by PumA impedes both cytokine and TLR receptor signalling, highlighting a novel strategy for innate immune evasion.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Evasão da Resposta Imune , Glicoproteínas de Membrana/antagonistas & inibidores , Fator 88 de Diferenciação Mieloide/antagonistas & inibidores , Pseudomonas aeruginosa/patogenicidade , Receptores de Interleucina-1/antagonistas & inibidores , Receptores Toll-Like/antagonistas & inibidores , Fatores de Virulência/metabolismo , Proteínas de Bactérias/metabolismo , Linhagem Celular , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Humanos , Pseudomonas aeruginosa/imunologia
4.
PLoS Pathog ; 13(1): e1006092, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28060920

RESUMO

Toll/interleukin-1 receptor (TIR) domains in Toll-like receptors are essential for initiating and propagating the eukaryotic innate immune signaling cascade. Here, we investigate TirS, a Staphylococcus aureus TIR mimic that is part of a novel bacterial invasion mechanism. Its ectopic expression in eukaryotic cells inhibited TLR signaling, downregulating the NF-kB pathway through inhibition of TLR2, TLR4, TLR5, and TLR9. Skin lesions induced by the S. aureus knockout tirS mutant increased in a mouse model compared with wild-type and restored strains even though the tirS-mutant and wild-type strains did not differ in bacterial load. TirS also was associated with lower neutrophil and macrophage activity, confirming a central role in virulence attenuation through local inflammatory responses. TirS invariably localizes within the staphylococcal chromosomal cassettes (SCC) containing the fusC gene for fusidic acid resistance but not always carrying the mecA gene. Of note, sub-inhibitory concentration of fusidic acid increased tirS expression. Epidemiological studies identified no link between this effector and clinical presentation but showed a selective advantage with a SCCmec element with SCC fusC/tirS. Thus, two key traits determining the success and spread of bacterial infections are linked.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Glicoproteínas de Membrana/genética , Proteínas de Ligação às Penicilinas/genética , Receptores de Interleucina-1/genética , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Ácido Fusídico/farmacologia , Células HEK293 , Humanos , Macrófagos/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Neutrófilos/imunologia , Receptores de Interleucina-1/imunologia , Transdução de Sinais/imunologia , Infecções Cutâneas Estafilocócicas/tratamento farmacológico , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Receptores Toll-Like/genética
6.
Int J Mol Sci ; 19(1)2018 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-29320462

RESUMO

The increasing threat of Acinetobacter baumannii as a nosocomial pathogen is mainly due to the occurrence of multidrug-resistant strains that are associated with the real problem of its eradication from hospital wards. The particular ability of this pathogen to form biofilms contributes to its persistence, increases antibiotic resistance, and promotes persistent/device-related infections. We previously demonstrated that virstatin, which is a small organic compound known to decrease virulence of Vibrio cholera via an inhibition of T4-pili expression, displayed very promising activity to prevent A. baumannii biofilm development. Here, we examined the antibiofilm activity of mono-unsaturated chain fatty acids, palmitoleic (PoA), and myristoleic (MoA) acids, presenting similar action on V. cholerae virulence. We demonstrated that PoA and MoA (at 0.02 mg/mL) were able to decrease A. baumannii ATCC 17978 biofilm formation up to 38% and 24%, respectively, presented a biofilm dispersing effect and drastically reduced motility. We highlighted that these fatty acids decreased the expression of the regulator abaR from the LuxIR-type quorum sensing (QS) communication system AbaIR and consequently reduced the N-acyl-homoserine lactone production (AHL). This effect can be countered by addition of exogenous AHLs. Besides, fatty acids may have additional non-targeted effects, independent from QS. Atomic force microscopy experiments probed indeed that PoA and MoA could also act on the initial adhesion process in modifying the material interface properties. Evaluation of fatty acids effect on 22 clinical isolates showed a strain-dependent antibiofilm activity, which was not correlated to hydrophobicity or pellicle formation ability of the tested strains, and suggested a real diversity in cell-to-cell communication systems involved in A. baumannii biofilm formation.


Assuntos
Acinetobacter baumannii/fisiologia , Biofilmes/efeitos dos fármacos , Ácidos Graxos Insaturados/farmacologia , Percepção de Quorum/efeitos dos fármacos , Acil-Butirolactonas/metabolismo , Ácidos Graxos Monoinsaturados/farmacologia , Microscopia de Força Atômica
7.
Environ Microbiol ; 19(10): 4349-4364, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28925528

RESUMO

The natural habitats and potential reservoirs of the nosocomial pathogen Acinetobacter baumannii are poorly defined. Here, we put forth and tested the hypothesis of avian reservoirs of A. baumannii. We screened tracheal and rectal swab samples from livestock (chicken, geese) and wild birds (white stork nestlings) and isolated A. baumannii from 3% of sampled chicken (n = 220), 8% of geese (n = 40) and 25% of white stork nestlings (n = 661). Virulence of selected avian A. baumannii isolates was comparable to that of clinical isolates in the Galleria mellonella infection model. Whole genome sequencing revealed the close relationship of an antibiotic-susceptible chicken isolate from Germany with a multidrug-resistant human clinical isolate from China and additional linkages between livestock isolates and human clinical isolates related to international clonal lineages. Moreover, we identified stork isolates related to human clinical isolates from the United States. Multilocus sequence typing disclosed further kinship between avian and human isolates. Avian isolates do not form a distinct clade within the phylogeny of A. baumannii, instead they diverge into different lineages. Further, we provide evidence that A. baumannii is constantly present in the habitats occupied by storks. Collectively, our study suggests A. baumannii could be a zoonotic organism that may disseminate into livestock.


Assuntos
Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/classificação , Acinetobacter baumannii/genética , Galinhas/microbiologia , Reservatórios de Doenças/microbiologia , Gansos/microbiologia , Células A549 , Acinetobacter baumannii/isolamento & purificação , Animais , Antibacterianos , Sequência de Bases , Linhagem Celular , China , Infecção Hospitalar/microbiologia , Farmacorresistência Bacteriana Múltipla/genética , Genoma Bacteriano/genética , Alemanha , Hospitais , Humanos , Tipagem de Sequências Multilocus , Filogenia , Polônia , Análise de Sequência de DNA , Estados Unidos , Sequenciamento Completo do Genoma
8.
Nat Rev Immunol ; 6(2): 136-47, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16491138

RESUMO

A key determinant for the survival of intracellular pathogens is their ability to subvert the cellular processes of the host to establish a compartment that allows replication. Although most microorganisms internalized by host cells are efficiently cleared following fusion with lysosomes, many pathogens have evolved mechanisms to escape this degradation. In this Review, we provide insight into the molecular processes that are targeted by pathogens that interact with the endoplasmic reticulum and thereby subvert the immune response, ensure their survival intracellularly and cause disease. We also discuss how the endoplasmic reticulum 'strikes back' and controls microbial growth.


Assuntos
Bactérias/patogenicidade , Retículo Endoplasmático/imunologia , Imunidade Celular , Fagócitos/imunologia , Toxoplasma/patogenicidade , Vírus/patogenicidade , Animais , Bactérias/imunologia , Retículo Endoplasmático/microbiologia , Retículo Endoplasmático/parasitologia , Retículo Endoplasmático/virologia , Humanos , Toxoplasma/imunologia , Vírus/imunologia
9.
PLoS Pathog ; 8(11): e1002983, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23166489

RESUMO

Bacterial cyclic glucans are glucose polymers that concentrate within the periplasm of alpha-proteobacteria. These molecules are necessary to maintain the homeostasis of the cell envelope by contributing to the osmolarity of Gram negative bacteria. Here, we demonstrate that Brucella ß 1,2 cyclic glucans are potent activators of human and mouse dendritic cells. Dendritic cells activation by Brucella ß 1,2 cyclic glucans requires TLR4, MyD88 and TRIF, but not CD14. The Brucella cyclic glucans showed neither toxicity nor immunogenicity compared to LPS and triggered antigen-specific CD8(+) T cell responses in vivo. These cyclic glucans also enhanced antigen-specific CD4(+) and CD8(+) T cell responses including cross-presentation by different human DC subsets. Brucella ß 1,2 cyclic glucans increased the memory CD4(+) T cell responses of blood mononuclear cells exposed to recombinant fusion proteins composed of anti-CD40 antibody and antigens from both hepatitis C virus and Mycobacterium tuberculosis. Thus cyclic glucans represent a new class of adjuvants, which might contribute to the development of effective antimicrobial therapies.


Assuntos
Adjuvantes Imunológicos , Brucella/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Glucanos/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Animais , Brucella/química , Células Cultivadas , Glucanos/química , Glucanos/farmacologia , Humanos , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
10.
J Infect Dis ; 207(7): 1075-83, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23303808

RESUMO

Brucellae replicate in a vacuole derived from the endoplasmic reticulum (ER) in epithelial cells, macrophages, and dendritic cells. In animals, trophoblasts are also key cellular targets where brucellae efficiently replicate in association with the ER. Therefore, we investigated the ability of Brucella spp. to infect human trophoblasts using both immortalized and primary trophoblasts. Brucella extensively proliferated within different subpopulations of trophoblasts, suggesting that they constitute an important niche in cases where the fetal-maternal barrier is breached. In extravillous trophoblasts (EVTs), B. abortus and B. suis replicated within single-membrane acidic lysosomal membrane-associated protein 1-positive inclusions, whereas B. melitensis replicated in the ER-derived compartment. Furthermore, B. melitensis but not B. abortus nor B. suis interfered with the invasive capacity of EVT-like cells in vitro. Because EVTs are essential for implantation during early stages of pregnancy, the nature of the replication niche may have a central role during Brucella-associated abortion in infected women.


Assuntos
Brucella abortus/crescimento & desenvolvimento , Brucella suis/crescimento & desenvolvimento , Trofoblastos/microbiologia , Autofagia , Carga Bacteriana , Brucella abortus/metabolismo , Brucella abortus/patogenicidade , Brucella melitensis/crescimento & desenvolvimento , Brucella melitensis/metabolismo , Brucella melitensis/patogenicidade , Brucella suis/metabolismo , Brucella suis/patogenicidade , Brucelose/microbiologia , Brucelose/patologia , Calnexina/metabolismo , Células Cultivadas , Feminino , Humanos , Proteínas de Membrana Lisossomal/metabolismo , Viabilidade Microbiana , Microscopia de Fluorescência , Placenta/metabolismo , Placenta/microbiologia , Placenta/patologia , Gravidez , Tetraspanina 30/metabolismo , Trofoblastos/metabolismo , Trofoblastos/patologia
11.
Microbiol Spectr ; 11(3): e0517422, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37052493

RESUMO

Acinetobacter baumannii is an antibiotic-resistant, Gram-negative pathogen that causes a multitude of nosocomial infections. However, pathogenicity mechanisms and the host cell response during infection remain unclear. In this study, we determined virulence traits of A. baumannii clinical isolates belonging to the most widely disseminated international clonal lineage, international cluster 2 (IC2), in vitro and in vivo. Complexome profiling of primary human endothelial cells with A. baumannii revealed that mitochondria, and in particular complexes of the electron transport chain, are important host cell targets. Infection with highly virulent A. baumannii remodelled assembly of mitochondrial protein complexes and led to metabolic adaptation. These were characterized by reduced mitochondrial respiration and glycolysis in contrast to those observed in infection with low-pathogenicity A. baumannii. Perturbation of oxidative phosphorylation, destabilization of mitochondrial ribosomes, and interference with mitochondrial metabolic pathways were identified as important pathogenicity mechanisms. Understanding the interaction of human host cells with the current global A. baumannii clone is the basis to identify novel therapeutic targets. IMPORTANCE Virulence traits of Acinetobacter baumannii isolates of the worldwide most prevalent international clonal lineage, IC2, remain largely unknown. In our study, multidrug-resistant IC2 clinical isolates differed substantially in their virulence potential despite their close genetic relatedness. Our data suggest that, at least for some isolates, mitochondria are important target organelles during infection of primary human endothelial cells. Complexes of the respiratory chain were extensively remodelled after infection with a highly virulent A. baumannii strain, leading to metabolic adaptation characterized by severely reduced respiration and glycolysis. Perturbations of both mitochondrial morphology and mitoribosomes were identified as important pathogenicity mechanisms. Our data might help to further decipher the molecular mechanisms of A. baumannii and host mitochondrial interaction during infection.


Assuntos
Infecções por Acinetobacter , Acinetobacter baumannii , Humanos , Acinetobacter baumannii/genética , Células Endoteliais , Infecções por Acinetobacter/tratamento farmacológico , Farmacorresistência Bacteriana Múltipla/genética , Antibacterianos/farmacologia , Proteínas Mitocondriais/uso terapêutico
12.
Nat Commun ; 14(1): 102, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36609656

RESUMO

The cell nucleus is a primary target for intracellular bacterial pathogens to counteract immune responses and hijack host signalling pathways to cause disease. Here we identify two Brucella abortus effectors, NyxA and NyxB, that interfere with host protease SENP3, and this facilitates intracellular replication of the pathogen. The translocated Nyx effectors directly interact with SENP3 via a defined acidic patch (identified from the crystal structure of NyxB), preventing nucleolar localisation of SENP3 at late stages of infection. By sequestering SENP3, the effectors promote cytoplasmic accumulation of nucleolar AAA-ATPase NVL and ribosomal protein L5 (RPL5) in effector-enriched structures in the vicinity of replicating bacteria. The shuttling of ribosomal biogenesis-associated nucleolar proteins is inhibited by SENP3 and requires the autophagy-initiation protein Beclin1 and the SUMO-E3 ligase PIAS3. Our results highlight a nucleomodulatory function of two Brucella effectors and reveal that SENP3 is a crucial regulator of the subcellular localisation of nucleolar proteins during Brucella infection, promoting intracellular replication of the pathogen.


Assuntos
Brucelose , Proteínas Nucleares , Humanos , Proteínas Nucleares/metabolismo , Núcleo Celular/metabolismo , Brucella abortus/metabolismo , Nucléolo Celular/metabolismo , Brucelose/microbiologia , Chaperonas Moleculares/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo
13.
Cell Microbiol ; 13(8): 1261-74, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21707904

RESUMO

Type IV secretion systems (T4SS) are specialized protein complexes used by many bacterial pathogens for the delivery of effector molecules that subvert varied host cellular processes. Brucella spp. are facultative intracellular pathogens capable of survival and replication inside mammalian cells. Brucella T4SS (VirB) is essential to subvert lysosome fusion and to create an organelle permissive for replication. One possible role for VirB is to translocate effector proteins that modulate host cellular functions for the biogenesis of the replicative organelle. We hypothesized that proteins with eukaryotic domains or protein-protein interaction domains, among others, would be good candidates for modulation of host cell functions. To identify these candidates, we performed an in silico screen looking for proteins with distinctive features. Translocation of 84 potential substrates was assayed using adenylate cyclase reporter. By this approach, we identified six proteins that are delivered to the eukaryotic cytoplasm upon infection of macrophage-like cells and we could determine that four of them, encoded by genes BAB1_1043, BAB1_2005, BAB1_1275 and BAB2_0123, require a functional T4SS for their delivery. We confirmed VirB-mediated translocation of one of the substrates by immunofluorescence confocal microscopy, and we found that the N-terminal 25 amino acids are required for its delivery into cells.


Assuntos
Proteínas de Bactérias/metabolismo , Brucella abortus/metabolismo , Brucella abortus/patogenicidade , Macrófagos/microbiologia , Fatores de Virulência/metabolismo , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Fusão Gênica Artificial , Proteínas de Bactérias/genética , Brucella abortus/genética , Brucelose/microbiologia , Linhagem Celular , Biologia Computacional/métodos , Modelos Animais de Doenças , Genes Reporter , Genômica/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Confocal , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Virulência/genética
14.
Cell Microbiol ; 13(7): 1044-58, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21501366

RESUMO

Bacteria of the Brucella genus are facultative intracellular class III pathogens. These bacteria are able to control the intracellular trafficking of their vacuole, presumably by the use of yet unknown translocated effectors. To identify such effectors, we used a high-throughput yeast two-hybrid screen to identify interactions between putative human phagosomal proteins and predicted Brucella spp. proteins. We identified a specific interaction between the human small GTPase Rab2 and a Brucella spp. protein named RicA. This interaction was confirmed by GST-pull-down with the GDP-bound form of Rab2. A TEM-ß-lactamase-RicA fusion was translocated from Brucella abortus to RAW264.7 macrophages during infection. This translocation was not detectable in a strain deleted for the virB operon, coding for the type IV secretion system. However, RicA secretion in a bacteriological culture was still observed in a ΔvirB mutant. In HeLa cells, a ΔricA mutant recruits less GTP-locked myc-Rab2 on its Brucella-containing vacuoles, compared with the wild-type strain. We observed altered kinetics of intracellular trafficking and faster proliferation of the B. abortusΔricA mutant in HeLa cells, compared with the wild-type control. Altogether, the data reported here suggest RicA as the first reported effector with a proposed function for B. abortus.


Assuntos
Proteínas de Bactérias/metabolismo , Brucella abortus/patogenicidade , Interações Hospedeiro-Patógeno , Mapeamento de Interação de Proteínas , Fatores de Virulência/metabolismo , Proteína rab2 de Ligação ao GTP/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Células Epiteliais/microbiologia , Deleção de Genes , Humanos , Macrófagos/microbiologia , Camundongos , Fagossomos/metabolismo , Fagossomos/microbiologia , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido , Virulência , Fatores de Virulência/genética
15.
mSystems ; 7(1): e0048821, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35103489

RESUMO

The spread of antibiotic-resistant Acinetobacter baumannii poses a significant threat to public health worldwide. This nosocomial bacterial pathogen can be associated with life-threatening infections, particularly in intensive care units. A. baumannii is mainly described as an extracellular pathogen with restricted survival within cells. This study shows that a subset of A. baumannii clinical isolates extensively multiply within nonphagocytic immortalized and primary cells without the induction of apoptosis and with bacterial clusters visible up to 48 h after infection. This phenotype was observed for the A. baumannii C4 strain associated with high mortality in a hospital outbreak and the A. baumannii ABC141 strain, which was isolated from the skin but was found to be hyperinvasive. Intracellular multiplication of these A. baumannii strains occurred within spacious single membrane-bound vacuoles, labeled with the lysosomal associate membrane protein (LAMP1). However, these compartments excluded lysotracker, an indicator of acidic pH, suggesting that A. baumannii can divert its trafficking away from the lysosomal degradative pathway. These compartments were also devoid of autophagy features. A high-content microscopy screen of 43 additional A. baumannii clinical isolates highlighted various phenotypes, and (i) the majority of isolates remained extracellular, (ii) a significant proportion was capable of invasion and limited persistence, and (iii) three more isolates efficiently multiplied within LAMP1-positive vacuoles, one of which was also hyperinvasive. These data identify an intracellular niche for specific A. baumannii clinical isolates that enables extensive multiplication in an environment protected from host immune responses and out of reach of many antibiotics. IMPORTANCE Multidrug-resistant Acinetobacter baumannii isolates are associated with significant morbidity and mortality in hospitals worldwide. Understanding their pathogenicity is critical for improving therapeutic management. Although A. baumannii can steadily adhere to surfaces and host cells, most bacteria remain extracellular. Recent studies have shown that a small proportion of bacteria can invade cells but present limited survival. We have found that some A. baumannii clinical isolates can establish a specialized intracellular niche that sustains extensive intracellular multiplication for a prolonged time without induction of cell death. We propose that this intracellular compartment allows A. baumannii to escape the cell's normal degradative pathway, protecting bacteria from host immune responses and potentially hindering antibiotic accessibility. This may contribute to A. baumannii persistence, relapsing infections, and enhanced mortality in susceptible patients. A high-content microscopy-based screen confirmed that this pathogenicity trait is present in other clinical A. baumannii isolates. There is an urgent need for new antibiotics or alternative antimicrobial approaches, particularly to combat carbapenem-resistant A. baumannii. The discovery of an intracellular niche for this pathogen, as well as hyperinvasive isolates, may help guide the development of antimicrobial therapies and diagnostics in the future.


Assuntos
Infecções por Acinetobacter , Acinetobacter baumannii , Anti-Infecciosos , Humanos , Acinetobacter baumannii/genética , Incidência , beta-Lactamases/genética , Farmacorresistência Bacteriana Múltipla , Testes de Sensibilidade Microbiana , Infecções por Acinetobacter/tratamento farmacológico , Antibacterianos/farmacologia , Anti-Infecciosos/farmacologia
16.
Eur J Immunol ; 40(12): 3458-71, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21108467

RESUMO

Control of pulmonary pathogens constitutes a challenging task as successful immune responses need to be mounted without damaging the lung parenchyma. Using immunofluorescence microscopy and flow cytometry, we analyzed in the mouse the initial innate immune response that follows intranasal inoculation of Brucella abortus. Bacteria were absent from parenchymal dendritic cells (DC) but present in alveolar macrophages in which they replicated. When the number of alveolar macrophages was reduced prior to Brucella infection, small numbers of pulmonary DC were infected and a massive recruitment of TNF-α- and iNOS-producing DC ensued. Coincidentally, Brucella disseminated to the lung-draining mediastinal lymph nodes (LN) where they replicated in both migratory DC and migratory alveolar macrophages. Together, these results demonstrate that alveolar macrophages are critical regulators of the initial innate immune response against Brucella within the lungs and show that pulmonary DC and alveolar macrophages play rather distinct roles in the control of microbial burden.


Assuntos
Brucella abortus/imunologia , Brucelose/imunologia , Células Dendríticas/metabolismo , Linfonodos/imunologia , Macrófagos Alveolares/metabolismo , Animais , Carga Bacteriana , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/patogenicidade , Brucelose/microbiologia , Brucelose/transmissão , Contagem de Células , Movimento Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Imunidade Inata , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Linfonodos/microbiologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
17.
PLoS Pathog ; 5(6): e1000487, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19557163

RESUMO

The intracellular pathogen Brucella abortus survives and replicates inside host cells within an endoplasmic reticulum (ER)-derived replicative organelle named the "Brucella-containing vacuole" (BCV). Here, we developed a subcellular fractionation method to isolate BCVs and characterize for the first time the protein composition of its replicative niche. After identification of BCV membrane proteins by 2 dimensional (2D) gel electrophoresis and mass spectrometry, we focused on two eukaryotic proteins: the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and the small GTPase Rab 2 recruited to the vacuolar membrane of Brucella. These proteins were previously described to localize on vesicular and tubular clusters (VTC) and to regulate the VTC membrane traffic between the endoplasmic reticulum (ER) and the Golgi. Inhibition of either GAPDH or Rab 2 expression by small interfering RNA strongly inhibited B. abortus replication. Consistent with this result, inhibition of other partners of GAPDH and Rab 2, such as COPI and PKC iota, reduced B. abortus replication. Furthermore, blockage of Rab 2 GTPase in a GDP-locked form also inhibited B. abortus replication. Bacteria did not fuse with the ER and instead remained in lysosomal-associated membrane vacuoles. These results reveal an essential role for GAPDH and the small GTPase Rab 2 in B. abortus virulence within host cells.


Assuntos
Brucella abortus/citologia , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Proteína rab2 de Ligação ao GTP/metabolismo , Animais , Brucella abortus/crescimento & desenvolvimento , Linhagem Celular , Membrana Celular/química , Membrana Celular/microbiologia , Sobrevivência Celular , Retículo Endoplasmático/microbiologia , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/química , Interações Hospedeiro-Patógeno/fisiologia , Imuno-Histoquímica , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Via Secretória/fisiologia , Vacúolos/química , Vacúolos/enzimologia , Vacúolos/microbiologia , Proteína rab2 de Ligação ao GTP/química
18.
FEMS Microbiol Rev ; 45(6)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34223888

RESUMO

Nicotinamide adenine dinucleotide (NAD+) is a major cofactor in redox reactions in all life-forms. A stable level of NAD+ is vital to ensure cellular homeostasis. Some pathogens can modulate NAD+ metabolism to their advantage and even utilize or cleave NAD+ from the host using specialized effectors known as ADP-ribosyltransferase toxins and NADases, leading to energy store depletion, immune evasion or even cell death. This review explores recent advances in the field of bacterial NAD+-targeting toxins, highlighting the relevance of NAD+ modulation as an emerging pathogenesis strategy. In addition, we discuss the role of specific NAD+-targeting toxins in niche colonization and bacterial lifestyle as components of toxin/antitoxin systems and key players in interbacterial competition. Understanding the mechanisms of toxicity, regulation and secretion of these toxins will provide interesting leads in the search for new antimicrobial treatments in the fight against infectious diseases.


Assuntos
Toxinas Bacterianas , NAD , ADP Ribose Transferases , Bactérias , NAD+ Nucleosidase
19.
Infect Immun ; 78(5): 2283-91, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20194591

RESUMO

Brucella abortus is a facultative intracellular bacterial pathogen that causes abortion in domestic animals and undulant fever in humans. The mechanism of virulence of Brucella spp. is not yet fully understood. Therefore, it is crucial to identify new molecules that can function as virulence factors to better understand the host-pathogen interplay. Herein, we identified the gene encoding the phosphoglycerate kinase (PGK) of B. abortus strain 2308. To test the role of PGK in Brucella pathogenesis, a pgk deletion mutant was constructed. Replacement of the wild-type pgk by recombination was demonstrated by Southern and Western blot analyses. The B. abortus Delta pgk mutant strain exhibited extreme attenuation in bone marrow-derived macrophages and in vivo in BALB/c, C57BL/6, 129/Sv, and interferon regulatory factor-1 knockout (IRF-1 KO) mice. Additionally, at 24 h postinfection the Delta pgk mutant was not found within the same endoplasmic reticulum-derived compartment as the wild-type bacteria, but, instead, over 60% of Brucella-containing vacuoles (BCVs) retained the late endosomal/lysosomal marker LAMP1. Furthermore, the B. abortus Delta pgk deletion mutant was used as a live vaccine. Challenge experiments revealed that the Delta pgk mutant strain induced protective immunity in 129/Sv or IRF-1 KO mice that was superior to the protection conferred by commercial strain 19 or RB51. Finally, the results shown here demonstrated that Brucella PGK is critical for full bacterial virulence and that a Delta pgk mutant may serve as a potential vaccine candidate in future studies.


Assuntos
Proteínas de Bactérias/genética , Vacina contra Brucelose/imunologia , Brucella abortus/enzimologia , Brucella abortus/patogenicidade , Brucelose/prevenção & controle , Fosfoglicerato Quinase/deficiência , Animais , Vacina contra Brucelose/genética , Brucella abortus/imunologia , Brucelose/imunologia , Células Cultivadas , Contagem de Colônia Microbiana , DNA Bacteriano/química , DNA Bacteriano/genética , Feminino , Deleção de Genes , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Análise de Sequência de DNA , Baço/microbiologia , Análise de Sobrevida , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
20.
PLoS Pathog ; 4(2): e21, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18266466

RESUMO

Brucella is an intracellular pathogen able to persist for long periods of time within the host and establish a chronic disease. We show that soon after Brucella inoculation in intestinal loops, dendritic cells from ileal Peyer's patches become infected and constitute a cell target for this pathogen. In vitro, we found that Brucella replicates within dendritic cells and hinders their functional activation. In addition, we identified a new Brucella protein Btp1, which down-modulates maturation of infected dendritic cells by interfering with the TLR2 signaling pathway. These results show that intracellular Brucella is able to control dendritic cell function, which may have important consequences in the development of chronic brucellosis.


Assuntos
Brucella abortus/patogenicidade , Brucelose/microbiologia , Células Dendríticas/microbiologia , Interações Hospedeiro-Patógeno , Receptor 2 Toll-Like/metabolismo , Animais , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/imunologia , Brucelose/imunologia , Brucelose/patologia , Sobrevivência Celular , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Íleo/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nódulos Linfáticos Agregados/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA