Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(36): e2308752120, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37639588

RESUMO

The causative agent of human Q fever, Coxiella burnetii, is highly adapted to infect alveolar macrophages by inhibiting a range of host responses to infection. Despite the clinical and biological importance of this pathogen, the challenges related to genetic manipulation of both C. burnetii and macrophages have limited our knowledge of the mechanisms by which C. burnetii subverts macrophages functions. Here, we used the related bacterium Legionella pneumophila to perform a comprehensive screen of C. burnetii effectors that interfere with innate immune responses and host death using the greater wax moth Galleria mellonella and mouse bone marrow-derived macrophages. We identified MceF (Mitochondrial Coxiella effector protein F), a C. burnetii effector protein that localizes to mitochondria and contributes to host cell survival. MceF was shown to enhance mitochondrial function, delay membrane damage, and decrease mitochondrial ROS production induced by rotenone. Mechanistically, MceF recruits the host antioxidant protein Glutathione Peroxidase 4 (GPX4) to the mitochondria. The protective functions of MceF were absent in primary macrophages lacking GPX4, while overexpression of MceF in human cells protected against oxidative stress-induced cell death. C. burnetii lacking MceF was replication competent in mammalian cells but induced higher mortality in G. mellonella, indicating that MceF modulates the host response to infection. This study reveals an important C. burnetii strategy to subvert macrophage cell death and host immunity and demonstrates that modulation of the host antioxidant system is a viable strategy to promote the success of intracellular bacteria.


Assuntos
Antioxidantes , Coxiella , Humanos , Animais , Camundongos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Estresse Oxidativo , Morte Celular , Mamíferos
2.
J Infect Dis ; 227(12): 1364-1375, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-36763010

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection triggers activation of the NLRP3 inflammasome, which promotes inflammation and aggravates severe COVID-19. Here, we report that SARS-CoV-2 induces upregulation and activation of human caspase-4/CASP4 (mouse caspase-11/CASP11), and this process contributes to NLRP3 activation. In vivo infections performed in transgenic hACE2 humanized mice, deficient or sufficient for Casp11, indicate that hACE2 Casp11-/- mice were protected from disease development, with the increased pulmonary parenchymal area, reduced clinical score of the disease, and reduced mortality. Assessing human samples from fatal cases of COVID-19, we found that CASP4 was expressed in patient lungs and correlated with the expression of inflammasome components and inflammatory mediators, including CASP1, IL1B, IL18, and IL6. Collectively, our data establish that CASP4/11 promotes NLRP3 activation and disease pathology, revealing a possible target for therapeutic interventions for COVID-19.


Assuntos
COVID-19 , Inflamassomos , Camundongos , Animais , Humanos , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Macrófagos/metabolismo , COVID-19/metabolismo , SARS-CoV-2/metabolismo , Camundongos Transgênicos
3.
PLoS Pathog ; 15(6): e1007886, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31251782

RESUMO

Inflammasomes are cytosolic multi-protein complexes that detect infection or cellular damage and activate the Caspase-1 (CASP1) protease. The NAIP5/NLRC4 inflammasome detects bacterial flagellin and is essential for resistance to the flagellated intracellular bacterium Legionella pneumophila. The effectors required downstream of NAIP5/NLRC4 to restrict bacterial replication remain unclear. Upon NAIP5/NLRC4 activation, CASP1 cleaves and activates the pore-forming protein Gasdermin-D (GSDMD) and the effector caspase-7 (CASP7). However, Casp1-/- (and Casp1/11-/-) mice are only partially susceptible to L. pneumophila and do not phenocopy Nlrc4-/-mice, because NAIP5/NLRC4 also activates CASP8 for restriction of L. pneumophila infection. Here we show that CASP8 promotes the activation of CASP7 and that Casp7/1/11-/- and Casp8/1/11-/- mice recapitulate the full susceptibility of Nlrc4-/- mice. Gsdmd-/- mice exhibit only mild susceptibility to L. pneumophila, but Gsdmd-/-Casp7-/- mice are as susceptible as the Nlrc4-/- mice. These results demonstrate that GSDMD and CASP7 are the key substrates downstream of NAIP5/NLRC4/CASP1/8 required for resistance to L. pneumophila.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Proteínas de Ligação ao Cálcio/imunologia , Caspase 1/imunologia , Caspase 7/imunologia , Caspase 8/imunologia , Inflamassomos/imunologia , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Proteína Inibidora de Apoptose Neuronal/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas de Ligação ao Cálcio/genética , Caspase 1/genética , Caspase 7/genética , Caspase 8/genética , Inflamassomos/genética , Peptídeos e Proteínas de Sinalização Intracelular , Doença dos Legionários/genética , Doença dos Legionários/patologia , Camundongos , Camundongos Knockout , Proteína Inibidora de Apoptose Neuronal/genética , Proteínas de Ligação a Fosfato
4.
Methods Mol Biol ; 1921: 305-319, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30694501

RESUMO

Legionella pneumophila is a gram-negative bacterium that infects many species of unicellular protozoa in freshwater environments. The human infection is accidental, and the bacteria may not have evolved strategies to bypass innate immune signaling in mammalian macrophages. Thus, L. pneumophila triggers many innate immune pathways including inflammasome activation. The inflammasomes are multimolecular platforms assembled in the host cell cytoplasm and lead to activation of inflammatory caspases. Inflammasome activation leads to secretion of inflammatory cytokines, such as IL-1ß and IL-18, and an inflammatory form of cell death called pyroptosis, which initiates with the induction of a pore in the macrophage membranes. In this chapter we provide detailed protocols to evaluate Legionella-induced inflammasome activation in macrophages, including real-time pore formation assay, western blotting to detect activation of inflammatory caspases (cleavage and pulldown), and the measurement of inflammatory cytokines.


Assuntos
Interações Hospedeiro-Patógeno , Inflamassomos/metabolismo , Legionella/fisiologia , Legionelose/metabolismo , Legionelose/microbiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Animais , Caspases/genética , Caspases/metabolismo , Citocinas/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Mediadores da Inflamação/metabolismo , Legionelose/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Knockout
5.
PLoS Pathog ; 13(8): e1006502, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28771586

RESUMO

Legionella pneumophila is a Gram-negative, flagellated bacterium that survives in phagocytes and causes Legionnaires' disease. Upon infection of mammalian macrophages, cytosolic flagellin triggers the activation of Naip/NLRC4 inflammasome, which culminates in pyroptosis and restriction of bacterial replication. Although NLRC4 and caspase-1 participate in the same inflammasome, Nlrc4-/- mice and their macrophages are more permissive to L. pneumophila replication compared with Casp1/11-/-. This feature supports the existence of a pathway that is NLRC4-dependent and caspase-1/11-independent. Here, we demonstrate that caspase-8 is recruited to the Naip5/NLRC4/ASC inflammasome in response to flagellin-positive bacteria. Accordingly, caspase-8 is activated in Casp1/11-/- macrophages in a process dependent on flagellin, Naip5, NLRC4 and ASC. Silencing caspase-8 in Casp1/11-/- cells culminated in macrophages that were as susceptible as Nlrc4-/- for the restriction of L. pneumophila replication. Accordingly, macrophages and mice deficient in Asc/Casp1/11-/- were more susceptible than Casp1/11-/- and as susceptible as Nlrc4-/- for the restriction of infection. Mechanistically, we found that caspase-8 activation triggers gasdermin-D-independent pore formation and cell death. Interestingly, caspase-8 is recruited to the Naip5/NLRC4/ASC inflammasome in wild-type macrophages, but it is only activated when caspase-1 or gasdermin-D is inhibited. Our data suggest that caspase-8 activation in the Naip5/NLRC4/ASC inflammasome enable induction of cell death when caspase-1 or gasdermin-D is suppressed.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Caspase 1/imunologia , Caspase 8/imunologia , Inflamassomos/imunologia , Doença dos Legionários/imunologia , Animais , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Ligação ao Cálcio , Caspase 1/metabolismo , Caspase 8/metabolismo , Modelos Animais de Doenças , Ativação Enzimática/imunologia , Ensaio de Imunoadsorção Enzimática , Técnicas de Silenciamento de Genes , Inflamassomos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Legionella pneumophila , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Inibidora de Apoptose Neuronal , Proteínas de Ligação a Fosfato , Reação em Cadeia da Polimerase em Tempo Real
6.
Cell Rep ; 20(4): 794-805, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28746866

RESUMO

Inflammasomes are multimeric protein complexes that initiate inflammatory cascades. Their activation is a hallmark of many infectious or inflammatory diseases. Their composition and activity are specified by proinflammatory stimuli. For example, the NLRP3 inflammasome is activated in response to cell damage and K+ efflux, whereas the AIM2 inflammasome is activated in response to cytosolic DNA. We used Legionella pneumophila, an intracellular bacterial pathogen that activates multiple inflammasomes, to elucidate the molecular mechanisms regulating inflammasome activation during infection. Upon infection, the AIM2 inflammasome engaged caspase-1 to induce pore formation in the cell membrane, which then caused K+-efflux-mediated activation of NLRP3. Thus, the AIM2 inflammasome amplifies signals of infection, triggering noncanonical activation of NLRP3. During infection, AIM2 and caspase-11 induced membrane damage, which was sufficient and essential for activating the NLRP3 inflammasome. Our data reveal that different inflammasomes regulate one another's activity to ensure an effective immune response to infection.


Assuntos
Caspase 1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Caspase 1/genética , Caspases/metabolismo , Caspases Iniciadoras , Proteínas de Ligação a DNA/genética , Feminino , Flagelina/genética , Flagelina/metabolismo , Inflamassomos/genética , Inflamassomos/imunologia , Legionella pneumophila/imunologia , Legionella pneumophila/patogenicidade , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Potássio/metabolismo
7.
J Leukoc Biol ; 101(4): 841-849, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27999148

RESUMO

Inflammasomes are multimeric protein complexes that assemble in the cytosol of many types of cells, including innate immune cells. The inflammasomes can be activated in response to infection or in response to stress signals that induce damage in the host cell membranes. These platforms trigger inflammatory processes, cell death, and the control of microbial replication. Many inflammasomes have been described so far, including NLRP3, NAIP/NLRC4, caspase-11, and AIM2. The ligand for NLRP3 is still unidentified, but the efflux of K+ is essential for NLRP3 activation. By contrast, inflammasomes, such as those composed of NAIP/NLRC4, caspase-11, and AIM2, can be activated by bacterial flagellin, LPS, and dsDNA. The knowledge of inflammasome biology has advanced tremendously in the last decade, fostered by the use of model organisms, such as Legionella pneumophila This bacterium evolved, infecting unicellular protozoa in freshwater environments, and the human infection is accidental. Thus, L. pneumophila did not evolve sophisticated mechanisms to inhibit mammalian innate immunity. For this reason, it has emerged as a very appropriate model of a pathogenic microbe for the investigation of inflammasome biology. In this review, we highlight the current information regarding the biology of inflammasomes and emphasize the advances achieved using L. pneumophila We also describe the inflammasomes activated in response to L. pneumophila infection and discuss the effector mechanisms that operate to clear the infection.


Assuntos
Inflamassomos/metabolismo , Legionella pneumophila/metabolismo , Animais , Caspases/metabolismo , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo
8.
J Infect Dis ; 211(2): 322-30, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25104770

RESUMO

Legionella pneumophila, the etiological agent of Legionnaires' disease, triggers activation of multiple innate immune pathways that lead to the restriction of bacterial replication in vivo. Despite the critical role for MyD88 in infection clearance, the receptors and mechanisms responsible for MyD88-mediated pulmonary bacterial clearance are still unclear. Here, we used flagellin mutants of L. pneumophila, which bypass the NAIP5/NLRC4-mediated restriction of bacterial replication, to assess the receptors involved in MyD88-mediated pulmonary bacterial clearance. By systematically comparing pulmonary clearance of L. pneumophila in C57BL/6 MyD88(-/-), TLR2(-/-), TLR3(-/-), TLR4(-/-), TLR9(-/-), IL-1R(-/-), and IL-18(-/-) mice, we found that, while the knockout of a single Toll-like receptor or interleukin 18 resulted only in minor impairment of bacterial clearance, deficiency in the interleukin 1 (IL-1) receptor led to a significant impairment. IL-1/MyD88-mediated pulmonary bacterial clearance occurs via processes involving the recruitment of neutrophils. Collectively, our data contribute to the understanding of the effector mechanisms involved in MyD88-mediated pulmonary bacterial clearance.


Assuntos
Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Pulmão/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Infiltração de Neutrófilos , Receptores de Interleucina-1/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA