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











Base de dados
Intervalo de ano de publicação
1.
Nat Microbiol ; 9(3): 727-736, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38374245

RESUMO

In sub-Saharan Africa, multidrug-resistant non-typhoidal Salmonella serovars are a common cause of fatal bloodstream infection. Malnutrition is a predisposing factor, but the underlying mechanisms are unknown. Here we show that vitamin A deficiency, one of the most prevalent micronutrient deficits afflicting African children, increases susceptibility to disseminated non-typhoidal Salmonella disease in mice and impairs terminal neutrophil maturation. Immature neutrophils had reduced expression of Slc11a1, a gene that encodes a metal ion transporter generally thought to restrict pathogen growth in macrophages. Adoptive transfer of SLC11A1-proficient neutrophils, but not SLC11A1-deficient neutrophils, reduced systemic Salmonella burden in Slc11a1-/- mice or mice with vitamin A deficiency. Loss of terminal granulopoiesis regulator CCAAT/enhancer-binding protein ϵ (C/EBPϵ) also decreased neutrophil-mediated control of Salmonella, but not that mediated by peritoneal macrophages. Susceptibility to infection increased in Cebpe-/- Slc11a1+/+ mice compared with wild-type controls, in an Slc11a1-expression-dependent manner. These data suggest that SLC11A1 deficiency impairs Salmonella control in part by blunting neutrophil-mediated defence.


Assuntos
Salmonelose Animal , Deficiência de Vitamina A , Criança , Camundongos , Humanos , Animais , Neutrófilos , Salmonella , Macrófagos
2.
Infect Immun ; 90(3): e0001322, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35100011

RESUMO

Research on Brucella pathogenesis has focused primarily on its ability to cause persistent intracellular infection of the mononuclear phagocyte system. At these sites, Brucella abortus evades innate immunity, which results in low-level inflammation and chronic infection of phagocytes. In contrast, the host response in the placenta during infection is characterized by severe inflammation and extensive extracellular replication of B. abortus. Despite the importance of reproductive disease caused by Brucella infection, our knowledge of the mechanisms involved in placental inflammation and abortion is limited. To understand the immune responses specifically driving placental pathology, we modeled placental B. abortus infection in pregnant mice. B. abortus infection caused an increase in the production of tumor necrosis factor alpha (TNF-α), specifically in the placenta. We found that placental expression levels of Tnfa and circulating TNF-α were dependent on the induction of endoplasmic reticulum stress and the B. abortus type IV secretion system (T4SS) effector protein VceC. Blockade of TNF-α reduced placental inflammation and improved fetal viability in mice. This work sheds light on a tissue-specific response of the placenta to B. abortus infection that may be important for bacterial transmission via abortion in the natural host species.


Assuntos
Brucelose Bovina , Brucelose , Animais , Brucella abortus/fisiologia , Brucelose/microbiologia , Bovinos , Feminino , Inflamação , Camundongos , Placenta , Gravidez , Fator de Necrose Tumoral alfa/metabolismo
3.
Infect Immun ; 86(9)2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29986892

RESUMO

Disseminated infections with nontyphoidal Salmonella (NTS) are a significant cause of child mortality in sub-Saharan Africa. NTS infection in children is clinically associated with malaria, suggesting that malaria compromises the control of disseminated NTS infection. To study the mechanistic basis for increased NTS susceptibility, we utilized a model of concurrent infection with Salmonella enterica serotype Typhimurium and Plasmodium yoelii nigeriensis (P. yoelii). Underlying malaria blunted monocyte expression of Ly6C, a marker for inflammatory activation, and impaired recruitment of inflammatory cells to the liver. Hepatic mononuclear phagocytes expressed lower levels of inducible nitric oxide synthase, tumor necrosis factor alpha, and granulocyte-macrophage colony-stimulating factor and showed increased levels of production of interleukin-10 and heme oxygenase-1, indicating that the underlying malaria modifies the activation state and inflammatory response of mononuclear phagocytes to NTS. P. yoelii infection also increased intracellular iron levels in liver mononuclear cells, as evidenced by elevated levels of ferritin and by the rescue of an S Typhimurium tonB feoB mutant defective for iron uptake. In addition, concurrent P. yoelii infection partially rescued the systemic colonization defect of an S Typhimurium spiB mutant defective for type III secretion system 2 (T3SS-2), indicating that the ability of phagocytic cells to limit the spread of S Typhimurium is impaired during concurrent P. yoelii infection. These results show that concurrent malaria increases susceptibility to disseminated NTS infection by blunting macrophage bactericidal mechanisms and providing an essential nutrient that enhances bacterial growth.


Assuntos
Ferro/metabolismo , Macrófagos/fisiologia , Malária/complicações , Plasmodium yoelii/imunologia , Infecções por Salmonella/imunologia , África Subsaariana , Animais , Antígenos Ly/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Macrófagos/imunologia , Malária/imunologia , Camundongos , Camundongos Endogâmicos CBA , Monócitos/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/imunologia
4.
Infect Immun ; 86(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29203548

RESUMO

Treatment of intracellular bacterial pathogens with antibiotic therapy often requires a long course of multiple drugs. A barrier to developing strategies that enhance antibiotic efficacy against these pathogens is our poor understanding of the intracellular nutritional environment that maintains bacterial persistence. The intracellular pathogen Brucella abortus survives and replicates preferentially in alternatively activated macrophages (AAMs); however, knowledge of the metabolic adaptations promoting exploitation of this niche is limited. Here we show that one mechanism promoting enhanced survival in AAMs is a shift in macrophage arginine utilization from production of nitric oxide (NO) to biosynthesis of polyamines, induced by interleukin 4 (IL-4)/IL-13 treatment. Production of polyamines by infected AAMs promoted both intracellular survival of B. abortus and chronic infection in mice, as inhibition of macrophage polyamine synthesis or inactivation of the putative putrescine transporter encoded by potIHGF reduced both intracellular survival in AAMs and persistence in mice. These results demonstrate that increased intracellular availability of polyamines induced by arginase-1 expression in IL-4/IL-13-induced AAMs promotes chronic persistence of B. abortus within this niche and suggest that targeting of this pathway may aid in eradicating chronic infection.


Assuntos
Brucella abortus/fisiologia , Brucelose/microbiologia , Macrófagos/fisiologia , Poliaminas/metabolismo , Animais , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia
5.
Trends Immunol ; 38(10): 758-767, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28823510

RESUMO

NOD1 and NOD2 are pattern recognition receptors of the innate immune system with well-established roles in sensing fragments of bacterial peptidoglycan. In addition to their role as microbial sensors, recent evidence indicates that nucleotide-binding oligomerization domains (NODs) can also recognize a broader array of danger signals. Indeed, recent work has expanded the roles of NOD1 and NOD2 to encompass not only sensing of infections with viruses and parasites but also perceiving perturbations of cellular processes such as regulation of the actin cytoskeleton and maintenance of endoplasmic reticulum homeostasis. This review will comment on recent progress and point out emerging questions in these areas.


Assuntos
Doença de Crohn/imunologia , Diabetes Mellitus Tipo 2/imunologia , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Obesidade/imunologia , Parasitos/imunologia , Vírus/imunologia , Animais , Estresse do Retículo Endoplasmático , Humanos , Peptidoglicano , Transdução de Sinais
6.
Science ; 357(6351): 570-575, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28798125

RESUMO

Perturbation of the gut-associated microbial community may underlie many human illnesses, but the mechanisms that maintain homeostasis are poorly understood. We found that the depletion of butyrate-producing microbes by antibiotic treatment reduced epithelial signaling through the intracellular butyrate sensor peroxisome proliferator-activated receptor γ (PPAR-γ). Nitrate levels increased in the colonic lumen because epithelial expression of Nos2, the gene encoding inducible nitric oxide synthase, was elevated in the absence of PPAR-γ signaling. Microbiota-induced PPAR-γ signaling also limits the luminal bioavailability of oxygen by driving the energy metabolism of colonic epithelial cells (colonocytes) toward ß-oxidation. Therefore, microbiota-activated PPAR-γ signaling is a homeostatic pathway that prevents a dysbiotic expansion of potentially pathogenic Escherichia and Salmonella by reducing the bioavailability of respiratory electron acceptors to Enterobacteriaceae in the lumen of the colon.


Assuntos
Disbiose/metabolismo , Disbiose/microbiologia , Enterobacteriaceae/patogenicidade , Microbioma Gastrointestinal , Óxido Nítrico Sintase Tipo II/metabolismo , PPAR gama/metabolismo , Proteína 4 Semelhante a Angiopoietina/genética , Anilidas/farmacologia , Animais , Antibacterianos/farmacologia , Butiratos/metabolismo , Células CACO-2 , Clostridium/efeitos dos fármacos , Clostridium/metabolismo , Colite/metabolismo , Colite/microbiologia , Colo/metabolismo , Colo/microbiologia , Disbiose/induzido quimicamente , Disbiose/genética , Enterobacteriaceae/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Expressão Gênica , Homeostase , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nitratos/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/genética , Oxirredução , PPAR gama/antagonistas & inibidores , PPAR gama/genética , Transdução de Sinais , Estreptomicina/farmacologia
7.
PLoS Pathog ; 13(8): e1006566, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28817719

RESUMO

Th1 cells can be activated by TCR-independent stimuli, but the importance of this pathway in vivo and the precise mechanisms involved require further investigation. Here, we used a simple model of non-cognate Th1 cell stimulation in Salmonella-infected mice to examine these issues. CD4 Th1 cell expression of both IL-18R and DR3 was required for optimal IFN-γ induction in response to non-cognate stimulation, while IL-15R expression was dispensable. Interestingly, effector Th1 cells generated by immunization rather than live infection had lower non-cognate activity despite comparable IL-18R and DR3 expression. Mice lacking T cell intrinsic expression of MyD88, an important adapter molecule in non-cognate T cell stimulation, exhibited higher bacterial burdens upon infection with Salmonella, Chlamydia or Brucella, suggesting that non-cognate Th1 stimulation is a critical element of efficient bacterial clearance. Thus, IL-18R and DR3 are critical players in non-cognate stimulation of Th1 cells and this response plays an important role in protection against intracellular bacteria.


Assuntos
Infecções Bacterianas/imunologia , Ativação Linfocitária/imunologia , Receptores de Interleucina-18/biossíntese , Membro 25 de Receptores de Fatores de Necrose Tumoral/biossíntese , Células Th1/imunologia , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Interleucina-18/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Receptores de Interleucina-18/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Células Th1/metabolismo
8.
Int J Med Microbiol ; 306(8): 604-610, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27760693

RESUMO

Salmonella enterica serotype Typhimurium is able to expand in the lumen of the inflamed intestine through mechanisms that have not been fully resolved. Here we utilized streptomycin-pretreated mice and dextran sodium sulfate (DSS)-treated mice to investigate how pathways for S. Typhimurium iron acquisition contribute to pathogen expansion in the inflamed intestine. Competitive infection with an iron uptake-proficient S. Typhimurium strain and mutant strains lacking tonB feoB, feoB, tonB or iroN in streptomycin pretreated mice demonstrated that ferric iron uptake requiring IroN and TonB conferred a fitness advantage during growth in the inflamed intestine. However, the fitness advantage conferred by ferrous iron uptake mechanisms was independent of inflammation and was only apparent in models where the normal microbiota composition had been disrupted by antibiotic treatment.


Assuntos
Gastroenterite/microbiologia , Intestinos/microbiologia , Ferro/metabolismo , Redes e Vias Metabólicas/genética , Infecções por Salmonella/microbiologia , Salmonella typhimurium/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bovinos , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
9.
mBio ; 7(4)2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27435462

RESUMO

UNLABELLED: Salmonella enterica serovar Typhimurium can cross the epithelial barrier using either the invasion-associated type III secretion system (T3SS-1) or a T3SS-1-independent mechanism that remains poorly characterized. Here we show that flagellum-mediated motility supported a T3SS-1-independent pathway for entering ileal Peyer's patches in the mouse model. Flagellum-dependent invasion of Peyer's patches required energy taxis toward nitrate, which was mediated by the methyl-accepting chemotaxis protein (MCP) Tsr. Generation of nitrate in the intestinal lumen required inducible nitric oxide synthase (iNOS), which was synthesized constitutively in the mucosa of the terminal ileum but not in the jejunum, duodenum, or cecum. Tsr-mediated invasion of ileal Peyer's patches was abrogated in mice deficient for Nos2, the gene encoding iNOS. We conclude that Tsr-mediated energy taxis enables S Typhimurium to migrate toward the intestinal epithelium by sensing host-derived nitrate, thereby contributing to invasion of Peyer's patches. IMPORTANCE: Nontyphoidal Salmonella serovars, such as S. enterica serovar Typhimurium, are a common cause of gastroenteritis in immunocompetent individuals but can also cause bacteremia in immunocompromised individuals. While the invasion-associated type III secretion system (T3SS-1) is important for entry, S Typhimurium strains lacking a functional T3SS-1 can still cross the intestinal epithelium and cause a disseminated lethal infection in mice. Here we observed that flagellum-mediated motility and chemotaxis contributed to a T3SS-1-independent pathway for invasion and systemic dissemination to the spleen. This pathway required the methyl-accepting chemotaxis protein (MCP) Tsr and energy taxis toward host-derived nitrate, which we found to be generated by inducible nitric oxide synthase (iNOS) in the ileal mucosa prior to infection. Collectively, our data suggest that S Typhimurium enhances invasion by actively migrating toward the intestinal epithelium along a gradient of host-derived nitrate emanating from the mucosal surface of the ileum.


Assuntos
Proteínas de Bactérias/metabolismo , Quimiotaxia , Endocitose , Células Epiteliais/microbiologia , Proteínas de Membrana/metabolismo , Nitratos/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/patogenicidade , Animais , Ceco/enzimologia , Modelos Animais de Doenças , Metabolismo Energético , Flagelos/fisiologia , Ilhas Genômicas , Intestino Delgado/enzimologia , Locomoção , Camundongos , Óxido Nítrico Sintase Tipo II/análise , Salmonella typhimurium/metabolismo , Salmonella typhimurium/fisiologia
10.
Microbiol Spectr ; 4(2)2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-27227304

RESUMO

Many bacterial pathogens can cause acute infections that are cleared with the onset of adaptive immunity, but a subset of these pathogens can establish persistent, and sometimes lifelong, infections. While bacteria that cause chronic infections are phylogenetically diverse, they share common features in their interactions with the host that enable a protracted period of colonization. This article will compare the persistence strategies of two chronic pathogens from the Proteobacteria, Brucella abortus and Salmonella enterica serovar Typhi, to consider how these two pathogens, which are very different at the genomic level, can utilize common strategies to evade immune clearance to cause chronic intracellular infections of the mononuclear phagocyte system.


Assuntos
Bactérias/patogenicidade , Infecções Bacterianas/microbiologia , Animais , Anticorpos Antibacterianos/imunologia , Bactérias/imunologia , Infecções Bacterianas/imunologia , Doença Crônica , Citocinas/imunologia , Interações Hospedeiro-Parasita , Humanos , Evasão da Resposta Imune , Sistema Imunitário , Imunidade Inata , Macrófagos/imunologia
11.
Nature ; 532(7599): 394-7, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-27007849

RESUMO

Endoplasmic reticulum (ER) stress is a major contributor to inflammatory diseases, such as Crohn disease and type 2 diabetes. ER stress induces the unfolded protein response, which involves activation of three transmembrane receptors, ATF6, PERK and IRE1α. Once activated, IRE1α recruits TRAF2 to the ER membrane to initiate inflammatory responses via the NF-κB pathway. Inflammation is commonly triggered when pattern recognition receptors (PRRs), such as Toll-like receptors or nucleotide-binding oligomerization domain (NOD)-like receptors, detect tissue damage or microbial infection. However, it is not clear which PRRs have a major role in inducing inflammation during ER stress. Here we show that NOD1 and NOD2, two members of the NOD-like receptor family of PRRs, are important mediators of ER-stress-induced inflammation in mouse and human cells. The ER stress inducers thapsigargin and dithiothreitol trigger production of the pro-inflammatory cytokine IL-6 in a NOD1/2-dependent fashion. Inflammation and IL-6 production triggered by infection with Brucella abortus, which induces ER stress by injecting the type IV secretion system effector protein VceC into host cells, is TRAF2, NOD1/2 and RIP2-dependent and can be reduced by treatment with the ER stress inhibitor tauroursodeoxycholate or an IRE1α kinase inhibitor. The association of NOD1 and NOD2 with pro-inflammatory responses induced by the IRE1α/TRAF2 signalling pathway provides a novel link between innate immunity and ER-stress-induced inflammation.


Assuntos
Estresse do Retículo Endoplasmático , Inflamação/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Transdução de Sinais , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella abortus/imunologia , Brucella abortus/patogenicidade , Linhagem Celular , Ditiotreitol/farmacologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/patologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Endorribonucleases/antagonistas & inibidores , Feminino , Humanos , Imunidade Inata , Inflamação/induzido quimicamente , Interleucina-6/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1/imunologia , Proteína Adaptadora de Sinalização NOD2/imunologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator 2 Associado a Receptor de TNF/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , Tapsigargina/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos
12.
Immunobiology ; 221(3): 468-74, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26626201

RESUMO

Co-infections with malaria and non-typhoidal Salmonella serotypes (NTS) can present as life-threatening bacteremia, in contrast to self-resolving NTS diarrhea in healthy individuals. In previous work with our mouse model of malaria/NTS co-infection, we showed increased gut mastocytosis and increased ileal and plasma histamine levels that were temporally associated with increased gut permeability and bacterial translocation. Here, we report that gut mastocytosis and elevated plasma histamine are also associated with malaria in an animal model of falciparum malaria, suggesting a broader host distribution of this biology. In support of mast cell function in this phenotype, malaria/NTS co-infection in mast cell-deficient mice was associated with a reduction in gut permeability and bacteremia. Further, antihistamine treatment reduced bacterial translocation and gut permeability in mice with malaria, suggesting a contribution of mast cell-derived histamine to GI pathology and enhanced risk of bacteremia during malaria/NTS co-infection.


Assuntos
Histamina/metabolismo , Malária/metabolismo , Malária/parasitologia , Mastócitos/metabolismo , Mucosa/metabolismo , Mucosa/parasitologia , Animais , Coinfecção , Modelos Animais de Doenças , Feminino , Histamina/sangue , Antagonistas dos Receptores Histamínicos/farmacologia , Macaca mulatta , Malária/tratamento farmacológico , Malária/imunologia , Malária Falciparum/imunologia , Malária Falciparum/metabolismo , Mastócitos/imunologia , Mastócitos/patologia , Mastocitose/imunologia , Mastocitose/metabolismo , Camundongos , Camundongos Knockout , Mucosa/efeitos dos fármacos , Mucosa/microbiologia , Permeabilidade , Infecções por Salmonella/imunologia , Infecções por Salmonella/metabolismo
13.
Infect Immun ; 83(4): 1546-55, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25644011

RESUMO

To discern virulent from innocuous microbes, the innate immune system senses events associated with bacterial access to immunoprivileged sites such as the host cell cytosol. One such pathway is triggered by the cytosolic delivery of flagellin, the major subunit of the flagellum, by bacterial secretion systems. This leads to inflammasome activation and subsequent proinflammatory cell death (pyroptosis) of the infected phagocyte. In this study, we demonstrate that the causative agent of typhoid fever, Salmonella enterica serovar Typhi, can partially subvert this critical innate immune recognition event. The transcriptional regulator TviA, which is absent from Salmonella serovars associated with human gastroenteritis, repressed the expression of flagellin during infection of human macrophage-like (THP-1) cells. This mechanism allowed S. Typhi to dampen inflammasome activation, leading to reduced interleukin-1ß (IL-1ß) secretion and diminished cell death. Likewise, the introduction of the tviA gene in nontyphoidal Salmonella enterica serovar Typhimurium reduced flagellin-induced pyroptosis. These data suggest that gene regulation of virulence factors enables S. Typhi to evade innate immune recognition by concealing a pathogen-induced process from being sensed by the inflammasome.


Assuntos
Apoptose/genética , Proteínas de Bactérias/imunologia , Flagelina/biossíntese , Macrófagos/imunologia , Salmonella typhi/patogenicidade , Fatores de Transcrição/imunologia , Animais , Proteínas Reguladoras de Apoptose/imunologia , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos , Células da Medula Óssea/imunologia , Células da Medula Óssea/microbiologia , Proteínas de Ligação ao Cálcio/imunologia , Linhagem Celular , Regulação Bacteriana da Expressão Gênica , Humanos , Inflamassomos/imunologia , Interleucina-1beta/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Salmonella typhi/genética , Salmonella typhi/imunologia , Fatores de Transcrição/genética , Fatores de Virulência/genética
15.
Cell Host Microbe ; 14(2): 159-70, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23954155

RESUMO

Eradication of persistent intracellular bacterial pathogens with antibiotic therapy is often slow or incomplete. However, strategies to augment antibiotics are hampered by our poor understanding of the nutritional environment that sustains chronic infection. Here we show that the intracellular pathogen Brucella abortus survives and replicates preferentially in alternatively activated macrophages (AAMs), which are more abundant during chronic infection. A metabolic shift induced by peroxisome proliferator-activated receptor γ (PPARγ), which increases intracellular glucose availability, is identified as a causal mechanism promoting enhanced bacterial survival in AAMs. Glucose uptake was crucial for increased replication of B. abortus in AAMs, and for chronic infection, as inactivation of the bacterial glucose transporter gluP reduced both intracellular survival in AAMs and persistence in mice. Thus, a shift in intracellular nutrient availability induced by PPARγ promotes chronic persistence of B. abortus within AAMs, and targeting this pathway may aid in eradicating chronic infection.


Assuntos
Brucella abortus/fisiologia , Glucose/metabolismo , Ativação de Macrófagos , Macrófagos/microbiologia , Viabilidade Microbiana , PPAR gama/metabolismo , Animais , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/imunologia , Brucella abortus/metabolismo , Macrófagos/imunologia , Camundongos
16.
PLoS Pathog ; 9(6): e1003454, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23818855

RESUMO

Evasion of host immune responses is a prerequisite for chronic bacterial diseases; however, the underlying mechanisms are not fully understood. Here, we show that the persistent intracellular pathogen Brucella abortus prevents immune activation of macrophages by inducing CD4(+)CD25(+) T cells to produce the anti-inflammatory cytokine interleukin-10 (IL-10) early during infection. IL-10 receptor (IL-10R) blockage in macrophages resulted in significantly higher NF-kB activation as well as decreased bacterial intracellular survival associated with an inability of B. abortus to escape the late endosome compartment in vitro. Moreover, either a lack of IL-10 production by T cells or a lack of macrophage responsiveness to this cytokine resulted in an increased ability of mice to control B. abortus infection, while inducing elevated production of pro-inflammatory cytokines, which led to severe pathology in liver and spleen of infected mice. Collectively, our results suggest that early IL-10 production by CD25(+)CD4(+) T cells modulates macrophage function and contributes to an initial balance between pro-inflammatory and anti-inflammatory cytokines that is beneficial to the pathogen, thereby promoting enhanced bacterial survival and persistent infection.


Assuntos
Brucella abortus/imunologia , Brucelose/imunologia , Linfócitos T CD4-Positivos/imunologia , Interleucina-10/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Viabilidade Microbiana/imunologia , Viabilidade Microbiana/efeitos da radiação , Animais , Brucelose/genética , Brucelose/patologia , Linfócitos T CD4-Positivos/patologia , Linhagem Celular , Interleucina-10/genética , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/imunologia
17.
Infect Immun ; 81(10): 3515-26, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23690397

RESUMO

Coinfection with malaria and nontyphoidal Salmonella serotypes (NTS) can cause life-threatening bacteremia in humans. Coinfection with malaria is a recognized risk factor for invasive NTS, suggesting that malaria impairs intestinal barrier function. Here, we investigated mechanisms and strategies for prevention of coinfection pathology in a mouse model. Our findings reveal that malarial-parasite-infected mice, like humans, develop L-arginine deficiency, which is associated with intestinal mastocytosis, elevated levels of histamine, and enhanced intestinal permeability. Prevention or reversal of L-arginine deficiency blunts mastocytosis in ileal villi as well as bacterial translocation, measured as numbers of mesenteric lymph node CFU of noninvasive Escherichia coli Nissle and Salmonella enterica serotype Typhimurium, the latter of which is naturally invasive in mice. Dietary supplementation of malarial-parasite-infected mice with L-arginine or L-citrulline reduced levels of ileal transcripts encoding interleukin-4 (IL-4), a key mediator of intestinal mastocytosis and macromolecular permeability. Supplementation with L-citrulline also enhanced epithelial adherens and tight junctions in the ilea of coinfected mice. These data suggest that increasing L-arginine bioavailability via oral supplementation can ameliorate malaria-induced intestinal pathology, providing a basis for testing nutritional interventions to reduce malaria-associated mortality in humans.


Assuntos
Arginina/deficiência , Bacteriemia/imunologia , Intestinos/citologia , Malária/complicações , Mastócitos/fisiologia , Salmonelose Animal/microbiologia , Animais , Bacteriemia/microbiologia , Citrulina , Feminino , Intestinos/imunologia , Intestinos/patologia , Camundongos , Permeabilidade , Plasmodium yoelii , Salmonelose Animal/patologia
18.
mBio ; 4(1): e00418-12, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23422410

RESUMO

Host cytokine responses to Brucella abortus infection are elicited predominantly by the deployment of a type IV secretion system (T4SS). However, the mechanism by which the T4SS elicits inflammation remains unknown. Here we show that translocation of the T4SS substrate VceC into host cells induces proinflammatory responses. Ectopically expressed VceC interacted with the endoplasmic reticulum (ER) chaperone BiP/Grp78 and localized to the ER of HeLa cells. ER localization of VceC required a transmembrane domain in its N terminus. Notably, the expression of VceC resulted in reorganization of ER structures. In macrophages, VceC was required for B. abortus-induced inflammation by induction of the unfolded protein response by a process requiring inositol-requiring transmembrane kinase/endonuclease 1. Altogether, these findings suggest that translocation of the T4SS effector VceC induces ER stress, which results in the induction of proinflammatory host cell responses during B. abortus infection. IMPORTANCE Brucella species are pathogens that require a type IV secretion system (T4SS) to survive in host cells and to maintain chronic infection. By as-yet-unknown pathways, the T4SS also elicits inflammatory responses in infected cells. Here we show that inflammation caused by the T4SS results in part from the sensing of a T4SS substrate, VceC, that localizes to the endoplasmic reticulum (ER), an intracellular site of Brucella replication. Possibly via binding of the ER chaperone BiP, VceC causes ER stress with concomitant expression of proinflammatory cytokines. Thus, induction of the unfolded protein response may represent a novel pathway by which host cells can detect pathogens deploying a T4SS.


Assuntos
Sistemas de Secreção Bacterianos , Brucella abortus/metabolismo , Brucella abortus/patogenicidade , Resposta a Proteínas não Dobradas , Fatores de Virulência/metabolismo , Animais , Citocinas/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/ultraestrutura , Chaperona BiP do Retículo Endoplasmático , Feminino , Células HeLa , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Virulência/toxicidade
19.
Cell Microbiol ; 15(6): 942-960, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23227931

RESUMO

Brucella are facultative intracellular bacteria that cause chronic infections by limiting innate immune recognition. It is currently unknown whether Brucella FliC flagellin, the monomeric subunit of flagellar filament, is sensed by the host during infection. Here, we used two mutants of Brucella melitensis, either lacking or overexpressing flagellin, to show that FliC hinders bacterial replication in vivo. The use of cells and mice genetically deficient for different components of inflammasomes suggested that FliC was a target of the cytosolic innate immune receptor NLRC4 in vivo but not in macrophages in vitro where the response to FliC was nevertheless dependent on the cytosolic adaptor ASC, therefore suggesting a new pathway of cytosolic flagellin sensing. However, our work also suggested that the lack of TLR5 activity of Brucella flagellin and the regulation of its synthesis and/or delivery into host cells are both part of the stealthy strategy of Brucella towards the innate immune system. Nevertheless, as a flagellin-deficient mutant of B. melitensis wasfound to cause histologically demonstrable injuries in the spleen of infected mice, we suggested that recognition of FliC plays a role in the immunological stand-off between Brucella and its host, which is characterized by a persistent infection with limited inflammatory pathology.


Assuntos
Brucella melitensis/patogenicidade , Brucelose/fisiopatologia , Flagelina/imunologia , Flagelina/metabolismo , Imunidade Inata/fisiologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Brucella melitensis/imunologia , Brucella melitensis/metabolismo , Brucelose/metabolismo , Brucelose/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular , Colo/microbiologia , Colo/patologia , Modelos Animais de Doenças , Feminino , Flagelina/genética , Humanos , Técnicas In Vitro , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação/genética , Baço/microbiologia , Baço/patologia , Receptor 5 Toll-Like/metabolismo
20.
Science ; 337(6093): 477-81, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22722251

RESUMO

Defensins are antimicrobial peptides that contribute broadly to innate immunity, including protection of mucosal tissues. Human α-defensin (HD) 6 is highly expressed by secretory Paneth cells of the small intestine. However, in contrast to the other defensins, it lacks appreciable bactericidal activity. Nevertheless, we report here that HD6 affords protection against invasion by enteric bacterial pathogens in vitro and in vivo. After stochastic binding to bacterial surface proteins, HD6 undergoes ordered self-assembly to form fibrils and nanonets that surround and entangle bacteria. This self-assembly mechanism occurs in vivo, requires histidine-27, and is consistent with x-ray crystallography data. These findings support a key role for HD6 in protecting the small intestine against invasion by diverse enteric pathogens and may explain the conservation of HD6 throughout Hominidae evolution.


Assuntos
Imunidade Inata , Imunidade nas Mucosas , Intestino Delgado/imunologia , alfa-Defensinas/química , alfa-Defensinas/metabolismo , Adesinas Bacterianas/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Linhagem Celular , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/ultraestrutura , Intestino Delgado/microbiologia , Intestino Delgado/ultraestrutura , Substâncias Macromoleculares/química , Substâncias Macromoleculares/imunologia , Substâncias Macromoleculares/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Modelos Moleculares , Nanoestruturas , Celulas de Paneth/imunologia , Celulas de Paneth/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Salmonelose Animal/imunologia , Salmonelose Animal/microbiologia , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Salmonella typhimurium/ultraestrutura , Yersinia enterocolitica/imunologia , Yersinia enterocolitica/patogenicidade , alfa-Defensinas/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA