Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Crit Care Med ; 50(6): e504-e515, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35067534

RESUMO

OBJECTIVES: Recent publications have shown that mitochondrial dynamics can govern the quality and quantity of extracellular mitochondria subsequently impacting immune phenotypes. This study aims to determine if pathologic mitochondrial fission mediated by Drp1/Fis1 interaction impacts extracellular mitochondrial content and macrophage function in sepsis-induced immunoparalysis. DESIGN: Laboratory investigation. SETTING: University laboratory. SUBJECTS: C57BL/6 and BALB/C mice. INTERVENTIONS: Using in vitro and murine models of endotoxin tolerance (ET), we evaluated changes in Drp1/Fis1-dependent pathologic fission and simultaneously measured the quantity and quality of extracellular mitochondria. Next, by priming mouse macrophages with isolated healthy mitochondria (MC) and damaged mitochondria, we determined if damaged extracellular mitochondria are capable of inducing tolerance to subsequent endotoxin challenge. Finally, we determined if inhibition of Drp1/Fis1-mediated pathologic fission abrogates release of damaged extracellular mitochondria and improves macrophage response to subsequent endotoxin challenge. MEASUREMENTS AND MAIN RESULTS: When compared with naïve macrophages (NMs), endotoxin-tolerant macrophages (ETM) demonstrated Drp1/Fis1-dependent mitochondrial dysfunction and higher levels of damaged extracellular mitochondria (Mitotracker-Green + events/50 µL: ETM = 2.42 × 106 ± 4,391 vs NM = 5.69 × 105 ± 2,478; p < 0.001). Exposure of NMs to damaged extracellular mitochondria (MH) induced cross-tolerance to subsequent endotoxin challenge, whereas MC had minimal effect (tumor necrosis factor [TNF]-α [pg/mL]: NM = 668 ± 3, NM + MH = 221 ± 15, and NM + Mc = 881 ± 15; p < 0.0001). Inhibiting Drp1/Fis1-dependent mitochondrial fission using heptapeptide (P110), a selective inhibitor of Drp1/Fis1 interaction, improved extracellular mitochondrial function (extracellular mitochondrial membrane potential, JC-1 [R/G] ETM = 7 ± 0.5 vs ETM + P110 = 19 ± 2.0; p < 0.001) and subsequently improved immune response in ETMs (TNF-α [pg/mL]; ETM = 149 ± 1 vs ETM + P110 = 1,150 ± 4; p < 0.0001). Similarly, P110-treated endotoxin tolerant mice had lower amounts of damaged extracellular mitochondria in plasma (represented by higher extracellular mitochondrial membrane potential, TMRM/MT-G: endotoxin tolerant [ET] = 0.04 ± 0.02 vs ET + P110 = 0.21 ± 0.02; p = 0.03) and improved immune response to subsequent endotoxin treatment as well as cecal ligation and puncture. CONCLUSIONS: Inhibition of Drp1/Fis1-dependent mitochondrial fragmentation improved macrophage function and immune response in both in vitro and in vivo models of ET. This benefit is mediated, at least in part, by decreasing the release of damaged extracellular mitochondria, which contributes to endotoxin cross-tolerance. Altogether, these data suggest that alterations in mitochondrial dynamics may play an important role in sepsis-induced immunoparalysis.


Assuntos
Dinaminas/metabolismo , Sepse , Animais , Dinaminas/genética , Dinaminas/farmacologia , Tolerância à Endotoxina , Endotoxinas , Humanos , Macrófagos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitocôndrias , Dinâmica Mitocondrial/fisiologia , Proteínas Mitocondriais , Sepse/patologia
2.
PLoS Pathog ; 16(8): e1008763, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32834002

RESUMO

The various sub-species of Salmonella enterica cause a range of disease in human hosts. The human-adapted Salmonella enterica serovar Typhi enters the gastrointestinal tract and invades systemic sites to cause enteric (typhoid) fever. In contrast, most non-typhoidal serovars of Salmonella are primarily restricted to gut tissues. Across Africa, invasive non-typhoidal Salmonella (iNTS) have emerged with an ability to spread beyond the gastrointestinal tract and cause systemic bloodstream infections with increased morbidity and mortality. To investigate this evolution in pathogenesis, we compared the genomes of African iNTS isolates with other Salmonella enterica serovar Typhimurium and identified several macA and macB gene variants unique to African iNTS. MacAB forms a tripartite efflux pump with TolC and is implicated in Salmonella pathogenesis. We show that macAB transcription is upregulated during macrophage infection and after antimicrobial peptide exposure, with macAB transcription being supported by the PhoP/Q two-component system. Constitutive expression of macAB improves survival of Salmonella in the presence of the antimicrobial peptide C18G. Furthermore, these macAB variants affect replication in macrophages and influence fitness during colonization of the murine gastrointestinal tract. Importantly, the infection outcome resulting from these macAB variants depends upon both the Salmonella Typhimurium genetic background and the host gene Nramp1, an important determinant of innate resistance to intracellular bacterial infection. The variations we have identified in the MacAB-TolC efflux pump in African iNTS may reflect evolution within human host populations that are compromised in their ability to clear intracellular Salmonella infections.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Proteínas de Bactérias/genética , Colite/patologia , Variação Genética , Macrófagos/imunologia , Salmonelose Animal/patologia , Salmonella typhimurium/imunologia , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Linhagem da Célula , Colite/induzido quimicamente , Colite/imunologia , Colite/microbiologia , Análise Mutacional de DNA , Modelos Animais de Doenças , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Salmonelose Animal/imunologia , Salmonelose Animal/microbiologia , Replicação Viral
3.
Cell Host Microbe ; 27(1): 54-67.e5, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31883922

RESUMO

Many intracellular bacteria can establish chronic infection and persist in tissues within granulomas composed of macrophages. Granuloma macrophages exhibit heterogeneous polarization states, or phenotypes, that may be functionally distinct. Here, we elucidate a host-pathogen interaction that controls granuloma macrophage polarization and long-term pathogen persistence during Salmonella Typhimurium (STm) infection. We show that STm persists within splenic granulomas that are densely populated by CD11b+CD11c+Ly6C+ macrophages. STm preferentially persists in granuloma macrophages reprogrammed to an M2 state, in part through the activity of the effector SteE, which contributes to the establishment of persistent infection. We demonstrate that tumor necrosis factor (TNF) signaling limits M2 granuloma macrophage polarization, thereby restricting STm persistence. TNF neutralization shifts granuloma macrophages toward an M2 state and increases bacterial persistence, and these effects are partially dependent on SteE activity. Thus, manipulating granuloma macrophage polarization represents a strategy for intracellular bacteria to overcome host restriction during persistent infection.


Assuntos
Granuloma/imunologia , Interações Hospedeiro-Patógeno/imunologia , Ativação de Macrófagos/imunologia , Infecções por Salmonella/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Granuloma/microbiologia , Humanos , Interleucina-4/metabolismo , Macrófagos/microbiologia , Camundongos , Salmonella typhimurium/imunologia , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Baço/citologia , Baço/microbiologia , Baço/patologia , Transativadores/metabolismo , Fatores de Virulência/metabolismo
4.
J Infect Dis ; 215(3): 440-451, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27932612

RESUMO

BACKGROUND: Legionella longbeachae (Llo) and Legionella pneumophila (Lpn) are the most common pneumonia-causing agents of the genus. Although both species can be lethal to humans and are highly prevalent, little is known about the molecular pathogenesis of Llo infections. In murine models of infection, Lpn infection is self-limited, whereas Llo infection is lethal. METHODS: We used mouse macrophages, human macrophages, human epithelial cells, and mouse infections in vivo to evaluate multiple parameters of the infection. RESULTS: We determined that the Llo Dot/Icm secretion system is critical for virulence. Different than Lpn, Llo disseminates and the animals develop a severe pulmonary failure, as demonstrated by lung mechanics and blood oxygenation assays. As compared to Lpn, Llo is immunologically silent and fails to trigger the production of cytokines in human pulmonary epithelial cells and in mouse and human macrophages. Infections in Tnfr1-/-, Ifng-/-, and Il12p40-/- mice supported the participation of cytokines for the resistance phenotype. CONCLUSIONS: Both Lpn and Llo require the Dot/Icm system for pathogenesis, but the infection outcome is strikingly different. Llo is immunologically silent, highly virulent, and lethal. The differences reported herein may reflect unappreciated clinical differences in patients infected with Lpn or Llo.


Assuntos
Legionella longbeachae/imunologia , Legionella longbeachae/patogenicidade , Legionelose/imunologia , Animais , Citocinas/metabolismo , Resistência à Doença/imunologia , Feminino , Humanos , Legionella pneumophila/imunologia , Legionelose/microbiologia , Legionelose/patologia , Legionelose/fisiopatologia , Leucócitos Mononucleares , Pulmão/fisiopatologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Especificidade da Espécie , Virulência
5.
Nat Commun ; 6: 10205, 2015 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-26687278

RESUMO

Coxiella burnetii is a highly infectious bacterium that promotes its own replication in macrophages by inhibiting several host cell responses. Here, we show that C. burnetii inhibits caspase-1 activation in primary mouse macrophages. By using co-infection experiments, we determine that the infection of macrophages with C. burnetii inhibits the caspase-11-mediated non-canonical activation of the NLRP3 inflammasome induced by subsequent infection with Escherichia coli or Legionella pneumophila. Genetic screening using flagellin mutants of L. pneumophila as a surrogate host, reveals a novel C. burnetii gene (IcaA) involved in the inhibition of caspase activation. Expression of IcaA in L. pneumophila inhibited the caspase-11 activation in macrophages. Moreover, icaA(-) mutants of C. burnetii failed to suppress the caspase-11-mediated inflammasome activation induced by L. pneumophila. Our data reveal IcaA as a novel C. burnetii effector protein that is secreted by the Dot/Icm type IV secretion system and interferes with the caspase-11-induced, non-canonical activation of the inflammasome.


Assuntos
Proteínas de Bactérias/imunologia , Coxiella burnetii/imunologia , Inflamassomos/imunologia , Febre Q/imunologia , Sistemas de Secreção Tipo IV/imunologia , Animais , Proteínas de Bactérias/genética , Caspases/genética , Caspases/imunologia , Caspases Iniciadoras , Coxiella burnetii/genética , Feminino , Regulação Bacteriana da Expressão Gênica , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Febre Q/genética , Febre Q/microbiologia , Sistemas de Secreção Tipo IV/genética
6.
Proc Natl Acad Sci U S A ; 110(35): E3321-30, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23942123

RESUMO

NAIP5/NLRC4 (neuronal apoptosis inhibitory protein 5/nucleotide oligomerization domain-like receptor family, caspase activation recruitment domain domain-containing 4) inflammasome activation by cytosolic flagellin results in caspase-1-mediated processing and secretion of IL-1ß/IL-18 and pyroptosis, an inflammatory cell death pathway. Here, we found that although NLRC4, ASC, and caspase-1 are required for IL-1ß secretion in response to cytosolic flagellin, cell death, nevertheless, occurs in the absence of these molecules. Cytosolic flagellin-induced inflammasome-independent cell death is accompanied by IL-1α secretion and is temporally correlated with the restriction of Salmonella Typhimurium infection. Despite displaying some apoptotic features, this peculiar form of cell death do not require caspase activation but is regulated by a lysosomal pathway, in which cathepsin B and cathepsin D play redundant roles. Moreover, cathepsin B contributes to NAIP5/NLRC4 inflammasome-induced pyroptosis and IL-1α and IL-1ß production in response to cytosolic flagellin. Together, our data describe a pathway induced by cytosolic flagellin that induces a peculiar form of cell death and regulates inflammasome-mediated effector mechanisms of macrophages.


Assuntos
Citosol/metabolismo , Flagelina/metabolismo , Inflamassomos/metabolismo , Lisossomos/metabolismo , Macrófagos/imunologia , Animais , Apoptose , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/patogenicidade , Receptor 5 Toll-Like/genética
7.
J Immunol ; 187(12): 6447-55, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22079982

RESUMO

Although NLRC4/IPAF activation by flagellin has been extensively investigated, the downstream signaling pathways and the mechanisms responsible for infection clearance remain unclear. In this study, we used mice deficient for the inflammasome components in addition to wild-type (WT) Legionella pneumophila or bacteria deficient for flagellin (flaA) or motility (fliI) to assess the pathways responsible for NLRC4-dependent growth restriction in vivo and ex vivo. By comparing infections with WT L. pneumophila, fliI, and flaA, we found that flagellin and motility are important for the colonization of the protozoan host Acanthamoeba castellanii. However, in macrophages and mammalian lungs, flagellin expression abrogated bacterial replication. The flagellin-mediated growth restriction was dependent on NLRC4, and although it was recently demonstrated that NLRC4 is able to recognize bacteria independent of flagellin, we found that the NLRC4-dependent restriction of L. pneumophila multiplication was fully dependent on flagellin. By examining infected caspase-1(-/-) mice and macrophages with flaA, fliI, and WT L. pneumophila, we could detect greater replication of flaA, which suggests that caspase-1 only partially accounted for flagellin-dependent growth restriction. Conversely, WT L. pneumophila multiplied better in macrophages and mice deficient for NLRC4 compared with that in macrophages and mice deficient for caspase-1, supporting the existence of a novel caspase-1-independent response downstream of NLRC4. This response operated early after macrophage infection and accounted for the restriction of bacterial replication within bacteria-containing vacuoles. Collectively, our data indicate that flagellin is required for NLRC4-dependent responses to L. pneumophila and that NLRC4 triggers caspase-1-dependent and -independent responses for bacterial growth restriction in macrophages and in vivo.


Assuntos
Acanthamoeba castellanii/microbiologia , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte/fisiologia , Flagelos/imunologia , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Acanthamoeba castellanii/enzimologia , Acanthamoeba castellanii/imunologia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Carga Bacteriana/imunologia , Proteínas de Bactérias/genética , Células da Medula Óssea/enzimologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/microbiologia , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/genética , Proteínas de Transporte/genética , Linhagem Celular , Feminino , Flagelos/enzimologia , Flagelos/genética , Flagelina/biossíntese , Flagelina/genética , Inflamassomos/deficiência , Inflamassomos/genética , Legionella pneumophila/genética , Locomoção/imunologia , Macrófagos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , ATPases Translocadoras de Prótons/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia
8.
J Biol Chem ; 285(42): 32087-95, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20702413

RESUMO

Innate immune recognition of flagellin is shared by transmembrane TLR5 and cytosolic Nlrc4 (NOD-like receptor family CARD (caspase activation recruitment domain) domain containing 4)/Naip5 (neuronal apoptosis inhibitory protein 5). TLR5 activates inflammatory genes through MYD88 pathway, whereas Nlrc4 and Naip5 assemble multiprotein complexes called inflammasomes, culminating in caspase-1 activation, IL-1ß/IL-18 secretion, and pyroptosis. Although both TLR5 and Naip5/Nlrc4 pathways cooperate to clear infections, little is known about the relative anti-pathogen effector mechanisms operating through each of them. Here we show that the cytosolic flagellin (FLA-BSDot) was able to activate iNOS, an enzyme previously associated with TLR5 pathway. Using Nlrc4- or Naip5-deficient macrophages, we found that both receptors are involved in iNOS activation by FLA-BSDot. Moreover, distinct from extracellular flagellin (FLA-BS), iNOS activation by intracellular flagellin is completely abrogated in the absence of caspase-1. Interestingly, IL-1ß and IL-18 do not seem to be important for FLA-BSDot-mediated iNOS production. Together, our data defined an additional anti-pathogen effector mechanism operated through Naip5 and Nlrc4 inflammasomes and illustrated a novel signaling transduction pathway that activates iNOS.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Inflamação/imunologia , Complexos Multiproteicos/imunologia , Proteína Inibidora de Apoptose Neuronal/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas de Ligação ao Cálcio/genética , Ativação Enzimática , Feminino , Flagelina/imunologia , Flagelina/farmacologia , Imunidade Inata/imunologia , Interleucina-18/imunologia , Interleucina-1beta/imunologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Proteína Inibidora de Apoptose Neuronal/genética , Transdução de Sinais/fisiologia , Receptor 5 Toll-Like/genética , Receptor 5 Toll-Like/metabolismo
9.
Infect Immun ; 77(4): 1700-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19204092

RESUMO

Paracoccidioidomycosis (PCM) is a systemic granulomatous disease caused by the dimorphic fungus Paracoccidioides brasiliensis. Anti-PCM vaccine formulations based on the secreted fungal cell wall protein (gp43) or the derived P10 sequence containing a CD4(+) T-cell-specific epitope have shown promising results. In the present study, we evaluated new anti-PCM vaccine formulations based on the intranasal administration of P. brasiliensis gp43 or the P10 peptide in combination with the Salmonella enterica FliC flagellin, an innate immunity agonist binding specifically to the Toll-like receptor 5, in a murine model. BALB/c mice immunized with gp43 developed high-specific-serum immunoglobulin G1 responses and enhanced interleukin-4 (IL-4) and IL-10 levels. On the other hand, mice immunized with recombinant purified flagellins genetically fused with P10 at the central hypervariable domain, either flanked or not by two lysine residues, or the synthetic P10 peptide admixed with purified FliC elicited a prevailing Th1-type immune response based on lung cell-secreted type 1 cytokines. Mice immunized with gp43 and FliC and intratracheally challenged with P. brasiliensis yeast cells had increased fungal proliferation and lung tissue damage. In contrast, mice immunized with the chimeric flagellins and particularly those immunized with P10 admixed with FliC reduced P. brasiliensis growth and lung damage. Altogether, these results indicate that S. enterica FliC flagellin modulates the immune response to P. brasiliensis P10 antigen and represents a promising alternative for the generation of anti-PCM vaccines.


Assuntos
Antígenos de Fungos , Flagelina , Proteínas Fúngicas , Vacinas Fúngicas , Glicoproteínas , Paracoccidioides/imunologia , Paracoccidioidomicose/prevenção & controle , Peptídeos , Animais , Anticorpos Antibacterianos/sangue , Especificidade de Anticorpos , Antígenos de Fungos/química , Antígenos de Fungos/genética , Antígenos de Fungos/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Flagelina/genética , Flagelina/imunologia , Flagelina/metabolismo , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/imunologia , Vacinas Fúngicas/administração & dosagem , Vacinas Fúngicas/genética , Vacinas Fúngicas/imunologia , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/imunologia , Humanos , Imunização , Pulmão/imunologia , Pulmão/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Paracoccidioides/patogenicidade , Paracoccidioidomicose/imunologia , Paracoccidioidomicose/microbiologia , Peptídeos/síntese química , Peptídeos/genética , Peptídeos/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Salmonella enterica/genética , Salmonella enterica/imunologia , Salmonella enterica/metabolismo
10.
Braz. j. microbiol ; 39(1): 44-49, Jan.-Mar. 2008. ilus, graf
Artigo em Inglês | LILACS, Sec. Est. Saúde SP | ID: lil-480672

RESUMO

Bacterial flagellins are important virulence-associated factors and strong inducers of inflammatory responses in mammalian hosts. Flagellins have also been investigated as potential vaccine adjuvants, either for induction of humoral or cellular immune responses, to different target antigens. In this study we investigated the adjuvant properties of three Salmonella enterica flagellins types (FliCd, FliCi and FljB) to an ovalbumin-derived CD8+ T cell-restricted epitope (OVA257264). Although mice immunized with the three tested flagellins elicited antigen-specific activated CD8+ T cells, only animals immunized with FliCi and FliCd flagellins admixed with ovalbumin mounted specific in vivo cytotoxic responses to peptide-pulsed target cells. The present results indicate that Salmonella flagellins are endowed with type-specific adjuvant effects toward murine CD8+ T cells, a feature that may impact their use as adjuvants for prophylatic or therapeutic vaccines.


As flagelinas bacterianas são importantes fatores associados à virulência e potentes indutores de resposta inflamatória em mamíferos. Estas moléculas são também investigadas como potencial adjuvante para uso em vacinas na indução de resposta imune humoral e celular para diferentes antígenos alvo. No presente estudo investigamos as propriedades adjuvantes de três tipos de flagelinas de Salmonella enterica (FliCd, FliCi e FljB) para um epítopo derivado da ovalbumina específico para células T CD8+. As três flagelinas testadas induziram respostas de células T CD8+ específicas em camundongos imunizados, porém, somente animais imunizados com as flagelinas FliCi e FliCd co-administradas com ovalbumina montaram resposta citotóxica específica in vivo para células-alvo pulsadas com peptídeo OVA. Os resultados apresentados indicam que flagelinas de Salmonella são dotadas de efeitos adjuvantes tipo-específico frente a células T CD8+ in vivo, uma característica que pode gerar impactos no uso dessas proteínas como adjuvantes em vacinas profiláticas ou terapêuticas.


Assuntos
Animais , Adjuvantes Imunológicos , Flagelina/análise , Flagelina/isolamento & purificação , Técnicas In Vitro , Linfócitos T , Salmonella enterica/isolamento & purificação , Vacinas/análise , Métodos , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA