Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Cell ; 173(4): 817-819, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29727669

RESUMO

IRF8 is a master transcription factor for immune cell development. In this issue, Karki et al. reveal that IRF8 governs the constitutive expression of genes encoding for NAIP proteins that are critical for the innate immune sensing of bacteria.


Assuntos
Inflamassomos , Proteína Inibidora de Apoptose Neuronal/genética , Diferenciação Celular , Regulação da Expressão Gênica , Fatores Reguladores de Interferon/genética
2.
Nat Immunol ; 20(5): 527-533, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30962589

RESUMO

Monitoring of the cytosolic compartment by the innate immune system for pathogen-encoded products or pathogen activities often enables the activation of a subset of caspases. In most cases, the cytosolic surveillance pathways are coupled to activation of caspase-1 via canonical inflammasome complexes. A related set of caspases, caspase-11 in rodents and caspase-4 and caspase-5 in humans, monitors the cytosol for bacterial lipopolysaccharide (LPS). Direct activation of caspase-11, caspase-4 and caspase-5 by intracellular LPS elicits the lytic cell death called 'pyroptosis', which occurs in multiple cell types. The pyroptosis is executed by the pore-forming protein GSDMD, which is activated by cleavage mediated by caspase-11, caspase-4 or caspase-5. In monocytes, formation of GSDMD pores can induce activation of the NLRP3 inflammasome for maturation of the cytokines IL-1ß and IL-18. Caspase-11-mediated pyroptosis in response to cytosolic LPS is critical for antibacterial defense and septic shock. Here we review the emerging literature on the sensing of cytosolic LPS and its regulation and pathophysiological functions.


Assuntos
Caspases/imunologia , Citosol/imunologia , Imunidade Inata/imunologia , Lipopolissacarídeos/imunologia , Animais , Caspases/metabolismo , Citosol/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipopolissacarídeos/metabolismo , Modelos Imunológicos , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/metabolismo , Proteínas de Ligação a Fosfato , Piroptose/imunologia
3.
Cell ; 165(4): 792-800, 2016 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-27153493

RESUMO

Canonical activation of the inflammasome is critical to promote caspase-1-dependent maturation of the proinflammatory cytokines IL-1ß and IL-18, as well as to induce pyroptotic cell death in response to pathogens and endogenous danger signals. Recent discoveries, however, are beginning to unveil new components of the inflammasome machinery as well as the full spectrum of inflammasome functions, extending their influence beyond canonical functions to regulation of eicosanoid storm, autophagy, and metabolism. In addition, the receptor components of the inflammasome can also regulate diverse biological processes, such as cellular proliferation, gene transcription, and tumorigenesis, all of which are independent of their inflammasome complex-forming capabilities. Here, we review these recent advances that are shaping our understanding of the complex biology of the inflammasome and its constituents.


Assuntos
Inflamassomos/fisiologia , Transdução de Sinais , Animais , Morte Celular , Humanos , Inflamassomos/imunologia , Inflamação/imunologia , Inflamação/metabolismo
4.
Cell ; 165(5): 1106-1119, 2016 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-27156449

RESUMO

Sensing of lipopolysaccharide (LPS) in the cytosol triggers caspase-11 activation and is central to host defense against Gram-negative bacterial infections and to the pathogenesis of sepsis. Most Gram-negative bacteria that activate caspase-11, however, are not cytosolic, and the mechanism by which LPS from these bacteria gains access to caspase-11 in the cytosol remains elusive. Here, we identify outer membrane vesicles (OMVs) produced by Gram-negative bacteria as a vehicle that delivers LPS into the cytosol triggering caspase-11-dependent effector responses in vitro and in vivo. OMVs are internalized via endocytosis, and LPS is released into the cytosol from early endosomes. The use of hypovesiculating bacterial mutants, compromised in their ability to generate OMVs, reveals the importance of OMVs in mediating the cytosolic localization of LPS. Collectively, these findings demonstrate a critical role for OMVs in enabling the cytosolic entry of LPS and, consequently, caspase-11 activation during Gram-negative bacterial infections.


Assuntos
Bactérias Gram-Negativas/citologia , Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/microbiologia , Lipopolissacarídeos/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Citosol/metabolismo , Ativação Enzimática , Bactérias Gram-Negativas/química , Imunidade Inata , Inflamação/imunologia , Inflamação/microbiologia , Interleucina-1/imunologia , Camundongos
5.
Immunity ; 50(1): 51-63.e5, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30635239

RESUMO

Interferon-inducible human oligoadenylate synthetase-like (OASL) and its mouse ortholog, Oasl2, enhance RNA-sensor RIG-I-mediated type I interferon (IFN) induction and inhibit RNA virus replication. Here, we show that OASL and Oasl2 have the opposite effect in the context of DNA virus infection. In Oasl2-/- mice and OASL-deficient human cells, DNA viruses such as vaccinia, herpes simplex, and adenovirus induced increased IFN production, which resulted in reduced virus replication and pathology. Correspondingly, ectopic expression of OASL in human cells inhibited IFN induction through the cGAS-STING DNA-sensing pathway. cGAS was necessary for the reduced DNA virus replication observed in OASL-deficient cells. OASL directly and specifically bound to cGAS independently of double-stranded DNA, resulting in a non-competitive inhibition of the second messenger cyclic GMP-AMP production. Our findings define distinct mechanisms by which OASL differentially regulates host IFN responses during RNA and DNA virus infection and identify OASL as a negative-feedback regulator of cGAS.


Assuntos
2',5'-Oligoadenilato Sintetase/metabolismo , Infecções por Vírus de DNA/imunologia , Vírus de DNA/fisiologia , Infecções por Vírus de RNA/imunologia , Vírus de RNA/imunologia , 2',5'-Oligoadenilato Sintetase/genética , Animais , AMP Cíclico/metabolismo , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nucleotidiltransferases/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais , Células THP-1 , Replicação Viral
6.
Immunity ; 49(3): 413-426.e5, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30170814

RESUMO

Inflammasome-activated caspase-1 cleaves gasdermin D to unmask its pore-forming activity, the predominant consequence of which is pyroptosis. Here, we report an additional biological role for gasdermin D in limiting cytosolic DNA surveillance. Cytosolic DNA is sensed by Aim2 and cyclic GMP-AMP synthase (cGAS) leading to inflammasome and type I interferon responses, respectively. We found that gasdermin D activated by the Aim2 inflammasome suppressed cGAS-driven type I interferon response to cytosolic DNA and Francisella novicida in macrophages. Similarly, interferon-ß (IFN-ß) response to F. novicida infection was elevated in gasdermin D-deficient mice. Gasdermin D-mediated negative regulation of IFN-ß occurred in a pyroptosis-, interleukin-1 (IL-1)-, and IL-18-independent manner. Mechanistically, gasdermin D depleted intracellular potassium (K+) via membrane pores, and this K+ efflux was necessary and sufficient to inhibit cGAS-dependent IFN-ß response. Thus, our findings have uncovered an additional interferon regulatory module involving gasdermin D and K+ efflux.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Francisella/fisiologia , Infecções por Bactérias Gram-Negativas/imunologia , Inflamassomos/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose/genética , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Humanos , Interferon Tipo I/metabolismo , Interleucina-1/metabolismo , Interleucina-18/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Proteínas de Ligação a Fosfato , Potássio/metabolismo , RNA Interferente Pequeno/genética
7.
Cell ; 150(3): 606-19, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22819539

RESUMO

Systemic infections with Gram-negative bacteria are characterized by high mortality rates due to the "sepsis syndrome," a widespread and uncontrolled inflammatory response. Though it is well recognized that the immune response during Gram-negative bacterial infection is initiated after the recognition of endotoxin by Toll-like receptor 4, the molecular mechanisms underlying the detrimental inflammatory response during Gram-negative bacteremia remain poorly defined. Here, we identify a TRIF pathway that licenses NLRP3 inflammasome activation by all Gram-negative bacteria. By engaging TRIF, Gram-negative bacteria activate caspase-11. TRIF activates caspase-11 via type I IFN signaling, an event that is both necessary and sufficient for caspase-11 induction and autoactivation. Caspase-11 subsequently synergizes with the assembled NLRP3 inflammasome to regulate caspase-1 activation and leads to caspase-1-independent cell death. These events occur specifically during infection with Gram-negative, but not Gram-positive, bacteria. The identification of TRIF as a regulator of caspase-11 underscores the importance of TLRs as master regulators of inflammasomes during Gram-negative bacterial infection.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Caspases/metabolismo , Citrobacter rodentium/metabolismo , Escherichia coli Êntero-Hemorrágica/metabolismo , Inflamassomos/metabolismo , Interferons/metabolismo , Animais , Proteínas de Transporte/metabolismo , Caspases Iniciadoras , Citrobacter rodentium/imunologia , Escherichia coli Êntero-Hemorrágica/imunologia , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/imunologia , Bactérias Gram-Positivas/metabolismo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Transdução de Sinais
8.
Nat Immunol ; 14(1): 52-60, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23160153

RESUMO

Interleukin 1 (IL-1) is an important mediator of innate immunity but can also promote inflammatory tissue damage. During chronic infections such as tuberculosis, the beneficial antimicrobial role of IL-1 must be balanced with the need to prevent immunopathology. By exogenously controlling the replication of Mycobacterium tuberculosis in vivo, we obviated the requirement for antimicrobial immunity and discovered that both IL-1 production and infection-induced immunopathology were suppressed by lymphocyte-derived interferon-γ (IFN-γ). This effect was mediated by nitric oxide (NO), which we found specifically inhibited assembly of the NLRP3 inflammasome via thiol nitrosylation. Our data indicate that the NO produced as a result of adaptive immunity is indispensable in modulating the destructive innate inflammatory responses elicited during persistent infections.


Assuntos
Proteínas de Transporte/metabolismo , Interleucina-1beta/metabolismo , Mycobacterium tuberculosis/imunologia , Óxido Nítrico/metabolismo , Tuberculose/imunologia , Animais , Proteínas de Transporte/genética , Células Cultivadas , Humanos , Imunidade Inata , Inflamassomos/metabolismo , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Óxido Nítrico/imunologia , Modificação Traducional de Proteínas/genética , Modificação Traducional de Proteínas/imunologia , Multimerização Proteica/genética , Multimerização Proteica/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
9.
Nat Immunol ; 14(6): 543-53, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23644505

RESUMO

Phagocytosis is a fundamental cellular process that is pivotal for immunity as it coordinates microbial killing, innate immune activation and antigen presentation. An essential step in this process is phagosome acidification, which regulates many functions of these organelles that allow phagosomes to participate in processes that are essential to both innate and adaptive immunity. Here we report that acidification of phagosomes containing Gram-positive bacteria is regulated by the NLRP3 inflammasome and caspase-1. Active caspase-1 accumulates on phagosomes and acts locally to control the pH by modulating buffering by the NADPH oxidase NOX2. These data provide insight into a mechanism by which innate immune signals can modify cellular defenses and establish a new function for the NLRP3 inflammasome and caspase-1 in host defense.


Assuntos
Proteínas de Transporte/imunologia , Caspase 1/imunologia , Inflamassomos/imunologia , Glicoproteínas de Membrana/imunologia , NADPH Oxidases/imunologia , Fagossomos/imunologia , Animais , Proteínas de Transporte/metabolismo , Caspase 1/metabolismo , Células Cultivadas , Ativação Enzimática/imunologia , Citometria de Fluxo , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Concentração de Íons de Hidrogênio , Immunoblotting , Inflamassomos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fagocitose/imunologia , Fagossomos/metabolismo , Fagossomos/microbiologia , Fagossomos/ultraestrutura , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Staphylococcus aureus/imunologia , Staphylococcus aureus/fisiologia
10.
Nat Immunol ; 13(4): 333-42, 2012 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-22430786

RESUMO

Innate immune responses have the ability to both combat infectious microbes and drive pathological inflammation. Inflammasome complexes are a central component of these processes through their regulation of interleukin 1ß (IL-1ß), IL-18 and pyroptosis. Inflammasomes recognize microbial products or endogenous molecules released from damaged or dying cells both through direct binding of ligands and indirect mechanisms. The potential of the IL-1 family of cytokines to cause tissue damage and chronic inflammation emphasizes the importance of regulating inflammasomes. Many regulatory mechanisms have been identified that act as checkpoints for attenuating inflammasome signaling at multiple steps. Here we discuss the various regulatory mechanisms that have evolved to keep inflammasome signaling in check to maintain immunological balance.


Assuntos
Imunidade Inata/imunologia , Inflamassomos/imunologia , Transdução de Sinais/imunologia , Animais , Humanos , Inflamassomos/metabolismo
11.
Nat Immunol ; 12(3): 222-30, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21151103

RESUMO

Autophagy, a cellular process for organelle and protein turnover, regulates innate immune responses. Here we demonstrate that depletion of the autophagic proteins LC3B and beclin 1 enhanced the activation of caspase-1 and secretion of interleukin 1ß (IL-1ß) and IL-18. Depletion of autophagic proteins promoted the accumulation of dysfunctional mitochondria and cytosolic translocation of mitochondrial DNA (mtDNA) in response to lipopolysaccharide (LPS) and ATP in macrophages. Release of mtDNA into the cytosol depended on the NALP3 inflammasome and mitochondrial reactive oxygen species (ROS). Cytosolic mtDNA contributed to the secretion of IL-1ß and IL-18 in response to LPS and ATP. LC3B-deficient mice produced more caspase-1-dependent cytokines in two sepsis models and were susceptible to LPS-induced mortality. Our study suggests that autophagic proteins regulate NALP3-dependent inflammation by preserving mitochondrial integrity.


Assuntos
Autofagia , Proteínas de Transporte/imunologia , DNA Mitocondrial , Imunidade Inata , Inflamassomos/imunologia , Animais , Caspase 1/imunologia , Citometria de Fluxo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR
13.
Nat Immunol ; 11(5): 395-402, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20351692

RESUMO

Inflammasomes regulate the activity of caspase-1 and the maturation of interleukin 1beta (IL-1beta) and IL-18. AIM2 has been shown to bind DNA and engage the caspase-1-activating adaptor protein ASC to form a caspase-1-activating inflammasome. Using Aim2-deficient mice, we identify a central role for AIM2 in regulating caspase-1-dependent maturation of IL-1beta and IL-18, as well as pyroptosis, in response to synthetic double-stranded DNA. AIM2 was essential for inflammasome activation in response to Francisella tularensis, vaccinia virus and mouse cytomegalovirus and had a partial role in the sensing of Listeria monocytogenes. Moreover, production of IL-18 and natural killer cell-dependent production of interferon-gamma, events critical in the early control of virus replication, were dependent on AIM2 during mouse cytomegalovirus infection in vivo. Collectively, our observations demonstrate the importance of AIM2 in the sensing of both bacterial and viral pathogens and in triggering innate immunity.


Assuntos
Infecções por Vírus de DNA/imunologia , Vírus de DNA/imunologia , Francisella tularensis/imunologia , Células Matadoras Naturais/metabolismo , Listeriose/imunologia , Macrófagos/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Tularemia/imunologia , Animais , Proteínas Reguladoras de Apoptose , Proteínas Adaptadoras de Sinalização CARD , Caspase 1/genética , Caspase 1/imunologia , Caspase 1/metabolismo , Linhagem Celular , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Proteínas do Citoesqueleto/genética , DNA/imunologia , Infecções por Vírus de DNA/genética , Infecções por Vírus de DNA/metabolismo , Vírus de DNA/crescimento & desenvolvimento , Vírus de DNA/patogenicidade , Proteínas de Ligação a DNA , Francisella tularensis/patogenicidade , Humanos , Imunidade Inata , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/microbiologia , Células Matadoras Naturais/patologia , Células Matadoras Naturais/virologia , Listeriose/genética , Listeriose/metabolismo , Ativação Linfocitária/genética , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo , Tularemia/genética , Tularemia/metabolismo , Carga Viral/genética , Carga Viral/imunologia
14.
Immunity ; 37(1): 96-107, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22840842

RESUMO

Yersinia pestis, the causative agent of plague, is able to suppress production of inflammatory cytokines IL-18 and IL-1ß, which are generated through caspase-1-activating nucleotide-binding domain and leucine-rich repeat (NLR)-containing inflammasomes. Here, we sought to elucidate the role of NLRs and IL-18 during plague. Lack of IL-18 signaling led to increased susceptibility to Y. pestis, producing tetra-acylated lipid A, and an attenuated strain producing a Y. pseudotuberculosis-like hexa-acylated lipid A. We found that the NLRP12 inflammasome was an important regulator controlling IL-18 and IL-1ß production after Y. pestis infection, and NLRP12-deficient mice were more susceptible to bacterial challenge. NLRP12 also directed interferon-γ production via induction of IL-18, but had minimal effect on signaling to the transcription factor NF-κB. These studies reveal a role for NLRP12 in host resistance against pathogens. Minimizing NLRP12 inflammasome activation may have been a central factor in evolution of the high virulence of Y. pestis.


Assuntos
Inflamassomos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peste/imunologia , Peste/metabolismo , Yersinia pestis/imunologia , Animais , Inflamassomos/imunologia , Interferon gama/biossíntese , Interleucina-18/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peste/mortalidade , Transdução de Sinais
16.
Proc Natl Acad Sci U S A ; 111(21): 7765-70, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24828532

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) is an extracellular pathogen that causes hemorrhagic colitis and hemolytic uremic syndrome. The proinflammatory cytokine, interleukin-1ß, has been linked to hemolytic uremic syndrome. Here we identify the nucleotide-binding domain and leucine rich repeat containing family, pyrin domain containing 3 (NLRP3) inflammasome as an essential mediator of EHEC-induced IL-1ß. Whereas EHEC-specific virulence factors were dispensable for NLRP3 activation, bacterial nucleic acids such as RNA:DNA hybrids and RNA gained cytosolic access and mediated inflammasome-dependent responses. Consistent with a direct role for RNA:DNA hybrids in inflammasome activation, delivery of synthetic EHEC RNA:DNA hybrids into the cytosol triggered NLRP3-dependent responses, and introduction of RNase H, which degrades such hybrids, into infected cells specifically inhibited inflammasome activation. Notably, an E. coli rnhA mutant, which is incapable of producing RNase H and thus harbors increased levels of RNA:DNA hybrid, induced elevated levels of NLRP3-dependent caspase-1 activation and IL-1ß maturation. Collectively, these findings identify RNA:DNA hybrids of bacterial origin as a unique microbial trigger of the NLRP3 inflammasome.


Assuntos
Proteínas de Transporte/metabolismo , DNA de Cadeia Simples/metabolismo , Escherichia coli Êntero-Hemorrágica/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Inflamassomos/imunologia , Interleucina-1beta/imunologia , RNA/metabolismo , Animais , Sequência de Bases , Proteínas de Transporte/imunologia , Caspase 1/imunologia , DNA de Cadeia Simples/genética , Ensaio de Imunoadsorção Enzimática , Proteínas de Escherichia coli/genética , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Dados de Sequência Molecular , Proteína 3 que Contém Domínio de Pirina da Família NLR , RNA/genética , Proteínas Ribossômicas/genética
17.
J Immunol ; 193(5): 2519-2530, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25063877

RESUMO

Inflammasomes are central mediators of host defense to a wide range of microbial pathogens. The nucleotide-binding domain and leucine-rich repeat containing family (NLR), pyrin domain-containing 3 (NLRP3) inflammasome plays a key role in triggering caspase-1-dependent IL-1ß maturation and resistance to fungal dissemination in Candida albicans infection. ß-Glucans are major components of fungal cell walls that trigger IL-1ß secretion in both murine and human immune cells. In this study, we sought to determine the contribution of ß-glucans to C. albicans-induced inflammasome responses in mouse dendritic cells. We show that the NLRP3-apoptosis-associated speck-like protein containing caspase recruitment domain protein-caspase-1 inflammasome is absolutely critical for IL-1ß production in response to ß-glucans. Interestingly, we also found that both complement receptor 3 (CR3) and dectin-1 play a crucial role in coordinating ß-glucan-induced IL-1ß processing as well as a cell death response. In addition to the essential role of caspase-1, we identify an important role for the proapoptotic protease caspase-8 in promoting ß-glucan-induced cell death and NLRP3 inflammasome-dependent IL-1ß maturation. A strong requirement for CR3 and caspase-8 also was found for NLRP3-dependent IL-1ß production in response to heat-killed C. albicans. Taken together, these results define the importance of dectin-1, CR3, and caspase-8, in addition to the canonical NLRP3 inflammasome, in mediating ß-glucan- and C. albicans-induced innate responses in dendritic cells. Collectively, these findings establish a novel link between ß-glucan recognition receptors and the inflammatory proteases caspase-8 and caspase-1 in coordinating cytokine secretion and cell death in response to immunostimulatory fungal components.


Assuntos
Candida albicans/imunologia , Candidíase/imunologia , Caspase 8/imunologia , Polissacarídeos Fúngicos/imunologia , Interleucina-1beta/imunologia , Lectinas Tipo C/imunologia , Antígeno de Macrófago 1/imunologia , beta-Glucanas/imunologia , Animais , Candida albicans/genética , Candidíase/genética , Candidíase/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Caspase 8/genética , Morte Celular/genética , Morte Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Polissacarídeos Fúngicos/genética , Humanos , Interleucina-1beta/genética , Lectinas Tipo C/genética , Antígeno de Macrófago 1/genética , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR
18.
J Immunol ; 193(4): 1911-9, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25015823

RESUMO

TLR4 interactor with leucine-rich repeats (TRIL) is a brain-enriched accessory protein that is important in TLR3 and TLR4 signaling. In this study, we generated Tril(-/-) mice and examined TLR responses in vitro and in vivo. We found a role for TRIL in both TLR4 and TLR3 signaling in mixed glial cells, consistent with the high level of expression of TRIL in these cells. We also found that TRIL is a modulator of the innate immune response to LPS challenge and Escherichia coli infection in vivo. Tril(-/-) mice produce lower levels of multiple proinflammatory cytokines and chemokines specifically within the brain after E. coli and LPS challenge. Collectively, these data uncover TRIL as a mediator of innate immune responses within the brain, where it enhances neuronal cytokine responses to infection.


Assuntos
Encéfalo/imunologia , Proteínas de Transporte/imunologia , Imunidade Inata/imunologia , Proteínas de Membrana/imunologia , Receptor 3 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Células Cultivadas , Quimiocina CCL5/biossíntese , Escherichia coli/imunologia , Infecções por Escherichia coli/imunologia , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-6/biossíntese , Lipopolissacarídeos , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroglia/imunologia , Poli I-C/farmacologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/biossíntese
19.
J Immunol ; 191(7): 3514-8, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23997220

RESUMO

Mycobacterium tuberculosis extracellular DNA gains access to the host cell cytosol via the ESX-1 secretion system. It is puzzling that this extracellular DNA of M. tuberculosis does not induce activation of the AIM2 inflammasome because AIM2 recognizes cytosolic DNA. In this study, we show that nonvirulent mycobacteria such as Mycobacterium smegmatis induce AIM2 inflammasome activation, which is dependent on their strong induction of IFN-ß production. In contrast, M. tuberculosis, but not an ESX-1-deficient mutant, inhibits the AIM2 inflammasome activation induced by either M. smegmatis or transfected dsDNA. The inhibition does not involve changes in host cell AIM2 mRNA or protein levels but led to decreased activation of caspase-1. We furthermore demonstrate that M. tuberculosis inhibits IFN-ß production and signaling, which was partially responsible for the inhibition of AIM2 activation. In conclusion, we report a novel immune evasion mechanism of M. tuberculosis that involves the ESX-1-dependent, direct or indirect, suppression of the host cell AIM2 inflammasome activation during infection.


Assuntos
Sistemas de Secreção Bacterianos/fisiologia , Inflamassomos/metabolismo , Interferon beta/metabolismo , Interleucina-1beta/metabolismo , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/metabolismo , Proteínas Nucleares/metabolismo , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Proteínas de Ligação a DNA , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética
20.
J Immunol ; 190(10): 5216-25, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23585680

RESUMO

Vascular disrupting agents such as 5,6-dimethylxanthenone-4-acetic acid (DMXAA) represent a novel approach for cancer treatment. DMXAA has potent antitumor activity in mice and, despite significant preclinical promise, failed human clinical trials. The antitumor activity of DMXAA has been linked to its ability to induce type I IFNs in macrophages, although the molecular mechanisms involved are poorly understood. In this study, we identify stimulator of IFN gene (STING) as a direct receptor for DMXAA leading to TANK-binding kinase 1 and IFN regulatory factor 3 signaling. Remarkably, the ability to sense DMXAA was restricted to murine STING. Human STING failed to bind to or signal in response to DMXAA. Human STING also failed to signal in response to cyclic dinucleotides, conserved bacterial second messengers known to bind and activate murine STING signaling. Collectively, these findings detail an unexpected species-specific role for STING as a receptor for an anticancer drug and uncover important insights that may explain the failure of DMXAA in clinical trials for human cancer.


Assuntos
Leucócitos Mononucleares/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Xantonas/metabolismo , Xantonas/farmacologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Células HEK293 , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/efeitos dos fármacos , Interferon beta/metabolismo , Leucócitos Mononucleares/imunologia , Macrófagos/imunologia , Camundongos , NF-kappa B/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA