Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Cell Rep ; 38(8): 110414, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35196496

RESUMO

Inflammasome activation exacerbates infectious disease caused by pathogens such as Listeria monocytogenes, Staphylococcus aureus, and severe acute respiratory syndrome coronavirus 2. Although these pathogens activate host inflammasomes to regulate pathogen expansion, the mechanisms by which pathogen toxins contribute to inflammasome activation remain poorly understood. Here we show that activation of inflammasomes by Listeria infection is promoted by amino acid residue T223 of listeriolysin O (LLO) independently of its pore-forming activity. LLO T223 is critical for phosphorylation of the inflammasome adaptor ASC at amino acid residue Y144 through Lyn-Syk signaling, which is essential for ASC oligomerization. Notably, a Listeria mutant expressing LLO T223A is impaired in inducing ASC phosphorylation and inflammasome activation. Furthermore, the virulence of LLO T223A mutant is markedly attenuated in vivo due to impaired ability to activate the inflammasome. Our results reveal a function of a pathogen toxin that exacerbates infection by promoting phosphorylation of ASC.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Inflamassomos/metabolismo , Listeria monocytogenes/patogenicidade , Transdução de Sinais , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Proteínas Adaptadoras de Sinalização CARD/química , Proteínas Adaptadoras de Sinalização CARD/deficiência , Proteínas Adaptadoras de Sinalização CARD/genética , Edição de Genes , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Proteínas Hemolisinas/química , Proteínas Hemolisinas/genética , Interleucina-18/metabolismo , Listeria monocytogenes/metabolismo , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Sítio-Dirigida , Fosforilação , Quinase Syk/genética , Quinase Syk/metabolismo , Virulência , Quinases da Família src/genética , Quinases da Família src/metabolismo
2.
Mucosal Immunol ; 12(5): 1092-1103, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31278375

RESUMO

It is widely accepted that inflammasomes protect the host from microbial pathogens by inducing inflammatory responses through caspase-1 activation. Here, we show that the inflammasome components ASC and NLRP3 are required for resistance to pneumococcal pneumonia, whereas caspase-1 and caspase-11 are dispensable. In the lung of S. pneumoniae-infected mice, ASC and NLRP3, but not caspase-1/11, were required for optimal expression of several mucosal innate immune proteins. Among them, TFF2 and intelectin-1 appeared to be protective against pneumococcal pneumonia. During infection, ASC and NLRP3 maintained the expression of the transcription factor SPDEF, which can facilitate the expression of the mucosal defense factor genes. Moreover, activation of STAT6, a key regulator of Spdef expression, depended on ASC and NLRP3. Overexpression of these inflammasome proteins sustained STAT6 phosphorylation induced by type 2 cytokines. Collectively, this study suggests that ASC and NLRP3 promote airway mucosal innate immunity by an inflammasome-independent mechanism involving the STAT6-SPDEF pathway.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/genética , Homeostase , Imunidade Inata , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/metabolismo , Caspases Iniciadoras/metabolismo , Citocinas/metabolismo , Citometria de Fluxo , Genes Reporter , Imunidade nas Mucosas , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Infiltração de Neutrófilos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/microbiologia , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Streptococcus pneumoniae/imunologia
3.
FEMS Microbiol Lett ; 363(15)2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27377894

RESUMO

Invertebrate animal species that can withstand temperatures as high as 37°C, the human body temperature, are limited. In the present study, we utilized the two-spotted cricket, Gryllus bimaculatus, which lives in tropical and subtropical regions, as an animal model of human pathogenic bacterial infection. Injection of Pseudomonas aeruginosa or Staphylococcus aureus into the hemolymph killed crickets. Injected P. aeruginosa or S. aureus proliferated in the hemolymph until the cricket died. The ability of these pathogenic bacteria to kill the crickets was blocked by the administration of antibiotics. S. aureus gene-knockout mutants of virulence factors, including cvfA, agr and srtA, exhibited decreased killing ability compared with the parent strain. The dose at which 50% of crickets were killed by P. aeruginosa or S. aureus was not decreased at 37°C compared with that at 27°C. Injection of Listeria monocytogenes, which upregulates toxin expression at 37°C, killed crickets, and the dose at which 50% of crickets were killed was decreased at 37°C compared with that at 27°C. These findings suggest that the two-spotted cricket is a useful model animal for evaluating the virulence properties of various human pathogenic bacteria at variable temperature including 37°C.


Assuntos
Infecções Bacterianas/microbiologia , Gryllidae/microbiologia , Listeria monocytogenes/patogenicidade , Modelos Animais , Pseudomonas aeruginosa/patogenicidade , Staphylococcus aureus/patogenicidade , Aminoaciltransferases/deficiência , Aminoaciltransferases/genética , Animais , Antibacterianos/farmacologia , Infecções Bacterianas/tratamento farmacológico , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/deficiência , Cisteína Endopeptidases/genética , Técnicas de Inativação de Genes , Hemolinfa/microbiologia , Humanos , Infecções por Pseudomonas/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/genética , Temperatura , Transativadores/genética , Transativadores/metabolismo
4.
Front Microbiol ; 6: 1145, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26528279

RESUMO

Listeria monocytogenes has a well-characterized ability to cross the placental barrier, resulting in spontaneous abortion and fetal infections. However, the mechanisms resulting in infection-associated abortion are not fully understood. In this study, we demonstrate that the dephosphorylation of MAPK family proteins caused by L. monocytogenes infection of trophoblast giant (TG) cells, which are placental immune cells, contributes to infectious abortion. Dephosphorylation of c-Jun, p38, and ERK1/2 was observed in infected TG cells, causing the downregulation of cytoprotective heme oxygenase (HO)-1. Blocking the dephosphorylation of proteins, including MAPK family proteins, inhibited the decrease in HO-1 expression. Treatment with MAPK inhibitors inhibited bacterial internalization into TG cells. Moreover, Toll-like receptor 2 involved in the expression of MAPK family proteins. Infection with a listeriolysin O-deleted mutant impaired dephosphorylation of MAPK family proteins in TG cells and did not induce infectious abortion in a mouse model. These results suggest that inactivation of the MAPK pathway by L. monocytogenes induces TG cell death and causes infectious abortion.

5.
J Invest Dermatol ; 135(4): 1007-1015, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25493651

RESUMO

A large number of gamma delta T cells (γδ T cells) are located within epithelial tissues including the skin. In mice, epidermal and dermal γδ T cells consist of distinct subsets and have specific roles in cutaneous immune responses. A recent study demonstrated that γδ T cells and cutaneous dendritic cells migrate from the skin to the draining lymph nodes (LNs). However, it remains unclear whether they regulate the antigen-specific immune response within the LNs. Herein, we investigated their properties and role in the LNs using the Mycobacterium bovis bacille Calmette-Guérin (BCG) infection model. In vivo cell labeling analysis revealed that most of the migratory subset comprised dermal Vγ4(+) cells. This population transmigrated from the skin to the LNs in a Gi-coupled chemokine receptor-independent manner. By depleting Vγ4(+) cells, the intranodal expansion of CD8(+) T cell against BCG was significantly attenuated. In addition, in vitro analysis revealed that Vγ4(+) cells produced TNF-α and enhanced IL-12 production by dendritic cells. Taken together, these findings suggest that dermal Vγ4(+) cells are a unique subset that possesses a migratory potency to the skin-draining LNs and enhances the dendritic cell function therein.


Assuntos
Linfócitos T CD8-Positivos/citologia , Linfonodos/patologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Movimento Celular , Proliferação de Células , Regulação da Expressão Gênica , Interferon gama/metabolismo , Interleucina-12/metabolismo , Células de Langerhans/citologia , Células de Langerhans/metabolismo , Luz , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mycobacterium bovis/metabolismo , Pele/imunologia , Subpopulações de Linfócitos T/imunologia
6.
Eur J Immunol ; 44(12): 3696-707, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25251560

RESUMO

Listeria monocytogenes induces the formation of inflammasomes and subsequent caspase-1 activation, and the adaptor apoptosis-associated speck-like protein containing a CARD (ASC) is crucial for this response. However, the role of ASC in L. monocytogenes infection in vivo is unclear. In this study, we demonstrate that ASC has a detrimental effect on host defense against L. monocytogenes infection at a lethal dose (10(6) CFU), but not at a sublethal dose (10(3) CFU). During lethal L. monocytogenes infection, serum levels of IL-18 and IL-10 were markedly elevated in WT mice, but not in ASC KO mice. IL-18 KO mice were more resistant to lethal L. monocytogenes infection than WT mice and had lower levels of serum IL-10. Furthermore, blockade of IL-10 receptor resulted in a reduction in bacterial counts, suggesting that ASC and IL-18 might exacerbate L. monocytogenes infection through induction of IL-10. We noticed that maturation of IL-18 during lethal infection was partially independent of caspase-1, but was critically dependent on ASC. ASC was required for the elevation of serum neutrophil serine protease activity, which correlated with caspase-1-independent IL-18 maturation and IL-10 production. Collectively, these results suggest that ASC plays a detrimental role in lethal L. monocytogenes infection through IL-18 production in an inflammasome-dependent and -independent manner.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Inflamassomos/imunologia , Interleucina-10/imunologia , Interleucina-18/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Adaptadoras de Sinalização CARD , Inflamassomos/genética , Interleucina-10/genética , Interleucina-18/genética , Listeriose/genética , Listeriose/patologia , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/patologia , Receptores de Interleucina-10/genética , Receptores de Interleucina-10/imunologia , Serina Proteases/genética , Serina Proteases/imunologia
7.
Infect Immun ; 82(6): 2310-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24643540

RESUMO

Streptococcus pneumoniae, a Gram-positive bacterial pathogen, causes pneumonia, meningitis, and septicemia. Innate immune responses are critical for the control and pathology of pneumococcal infections. It has been demonstrated that S. pneumoniae induces the production of type I interferons (IFNs) by host cells and that type I IFNs regulate resistance and chemokine responses to S. pneumoniae infection in an autocrine/paracrine manner. In this study, we examined the effects of type I IFNs on macrophage proinflammatory cytokine production in response to S. pneumoniae. The production of interleukin-18 (IL-18), but not other cytokines tested, was significantly decreased by the absence or blockade of the IFN-α/ß receptor, suggesting that type I IFN signaling is necessary for IL-18 production. Type I IFN signaling was also required for S. pneumoniae-induced activation of caspase-1, a cysteine protease that plays a central role in maturation and secretion of IL-18. Earlier studies proposed that the AIM2 and NLRP3 inflammasomes mediate caspase-1 activation in response to S. pneumoniae. From our results, the AIM2 inflammasome rather than the NLRP3 inflammasome seemed to require type I IFN signaling for its optimal activation. Consistently, AIM2, but not NLRP3, was upregulated in S. pneumoniae-infected macrophages in a manner dependent on the IFN-α/ß receptor. Furthermore, type I IFN signaling was found to contribute to IL-18 production in pneumococcal pneumonia in vivo. Taken together, these results suggest that type I IFNs regulate S. pneumoniae-induced activation of the AIM2 inflammasome by upregulating AIM2 expression. This study revealed a novel role for type I IFNs in innate responses to S. pneumoniae.


Assuntos
Inflamassomos/fisiologia , Interferon Tipo I/fisiologia , Proteínas Nucleares/metabolismo , Infecções Pneumocócicas/metabolismo , Animais , Caspase 1/metabolismo , Citocinas/metabolismo , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Feminino , Imunidade Inata/fisiologia , Interleucina-18/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções Pneumocócicas/imunologia , Transdução de Sinais/fisiologia , Streptococcus pneumoniae
8.
Pathog Dis ; 70(1): 51-60, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23913588

RESUMO

Region of difference 1 (RD1) is a genomic locus in the Mycobacterium tuberculosis genome that has been shown to participate in the virulence of the bacterium, induction of cell death, and cytokine secretion in infected macrophages. In this study, we investigated the role of RD1 in interleukin-1α (IL-1α) secretion. M. tuberculosis H37Rv strain, but not a mutant strain deficient for RD1 (∆RD1), significantly induced IL-1α secretion from infected macrophages. Although IL-1α secretion was only observed in H37Rv-infected macrophages, there was no difference in the level of IL-1α transcription and pro-IL1α synthesis after infection with H37Rv and ∆RD1. Interestingly, ∆RD1 infection did not increase intracellular Ca(2+) levels, and Ca(2+) chelators markedly inhibited IL-1α secretion in response to H37Rv infection. Moreover, the inability of ∆RD1 to induce IL-1α secretion was restored by treatment with the calcium ionophore A23187. A significant increase in calpain activity was detected in macrophages infected with H37Rv, but not with ∆RD1, and calpain inhibitors abrogated IL-1α secretion. Taken together, these results suggest that in M. tuberculosis-infected macrophages, RD1 contributed to maturation and secretion of IL-1α by enhancing the influx of Ca(2+) followed by calpain activation.


Assuntos
Cálcio/metabolismo , Calpaína/metabolismo , Citoplasma/metabolismo , Interleucina-1alfa/metabolismo , Macrófagos/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Animais , Proteínas de Bactérias/genética , Feminino , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Transcrição Gênica/genética
9.
Nat Immunol ; 14(12): 1247-55, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24185614

RESUMO

The inflammasome adaptor ASC contributes to innate immunity through the activation of caspase-1. Here we found that signaling pathways dependent on the kinases Syk and Jnk were required for the activation of caspase-1 via the ASC-dependent inflammasomes NLRP3 and AIM2. Inhibition of Syk or Jnk abolished the formation of ASC specks without affecting the interaction of ASC with NLRP3. ASC was phosphorylated during inflammasome activation in a Syk- and Jnk-dependent manner, which suggested that Syk and Jnk are upstream of ASC phosphorylation. Moreover, phosphorylation of Tyr144 in mouse ASC was critical for speck formation and caspase-1 activation. Our results suggest that phosphorylation of ASC controls inflammasome activity through the formation of ASC specks.


Assuntos
Proteínas do Citoesqueleto/imunologia , Inflamassomos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas Quinases JNK Ativadas por Mitógeno/imunologia , Proteínas Tirosina Quinases/imunologia , Animais , Proteínas Reguladoras de Apoptose , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Caspase 1/imunologia , Caspase 1/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Células HEK293 , Humanos , Immunoblotting , Inflamassomos/genética , Inflamassomos/metabolismo , Interleucina-18/imunologia , Interleucina-18/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nigericina/farmacologia , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Fosforilação/imunologia , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Interferência de RNA , Quinase Syk , Tirosina/genética , Tirosina/imunologia , Tirosina/metabolismo
10.
Gene ; 530(1): 19-25, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23954874

RESUMO

Although mutations of autoimmune regulator (AIRE) gene are responsible for autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), presenting a wide spectrum of many characteristic and non-characteristic clinical features, some patients lack AIRE gene mutations. Therefore, something other than a mutation, such as dysregulation of AIRE gene, may be a causal factor for APECED or its related diseases. However, regulatory mechanisms for AIRE gene expression and/or translation have still remained elusive. We found that IL-2-stimulated CD4(+) T (IL-2T) cells showed a high expression of AIRE gene, but very low AIRE protein production, while Epstein-Barr virus-transformed B (EBV-B) cells express both AIRE gene and AIRE protein. By using microarray analysis, we could identify miR-220b as a possible regulatory mechanism for AIRE gene translation in IL-2T cells. Here we report that miR-220b significantly reduced the expression of AIRE protein in AIRE gene with 3'UTR region transfected 293T cells, whereas no alteration of AIRE protein production was observed in the open reading frame of AIRE gene alone transfected cells. In addition, anti-miR-220b reversed the inhibitory function of miR-220b for the expression of AIRE protein in AIRE gene with 3'UTR region transfected cells. Moreover, when AIRE gene transfected cells with mutated 3'UTR were transfected with miR-220b, no reduction of AIRE protein production was observed. Taken together, it was concluded that miR-220b inhibited the AIRE gene translation through the 3'UTR region of AIRE gene, indicating that miR-220b could serve as a regulator for human AIRE gene translation.


Assuntos
Interleucina-2/metabolismo , MicroRNAs/genética , Poliendocrinopatias Autoimunes/genética , Fatores de Transcrição/genética , Linfócitos T CD4-Positivos/metabolismo , Regulação da Expressão Gênica/genética , Herpesvirus Humano 4/genética , Humanos , Mutação , Poliendocrinopatias Autoimunes/imunologia , Poliendocrinopatias Autoimunes/virologia , Biossíntese de Proteínas , Proteína AIRE
11.
PLoS One ; 8(8): e73862, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991207

RESUMO

Although the autoimmune regulator (Aire) knockout (KO) mouse model has been reported to present various organ-specific autoimmune diseases depending on genetic background, autoimmune pancreatitis in mice of BALB/c background has not yet been reported. Here, we report that Aire KO mice with BALB/cAnN background showed significant lymphoid cell infiltration in the pancreas and stomach. To examine whether the phenotype in the pancreas and stomach is due to autoimmune reaction associated with autoantibody production, indirect immunofluorescence staining followed by Western blot analysis was performed. Consequently, the autoantibody against pancreas and stomach was detected in the sera of Aire KO mice, and the target antigen of the autoantibody was identified as protein disulfide isomerase-associated 2 (Pdia2), which was reported to be expressed preferentially in the pancreas and stomach. Thus, Aire KO mice of BALB/cAnN background can serve as a useful animal model for autoimmune gastro-pancreatitis with anti-Pdia2 autoantibody production.


Assuntos
Autoanticorpos/imunologia , Gastrite/imunologia , Pancreatite/imunologia , Poliendocrinopatias Autoimunes/imunologia , Isomerases de Dissulfetos de Proteínas/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Dados de Sequência Molecular , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
12.
Infect Genet Evol ; 16: 151-6, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23352891

RESUMO

Mycobacterium tuberculosis has a considerable degree of genetic variability resulting in different epidemiology and disease outcomes. We evaluated the pathogen-host cell interaction of two genetically closely-related multidrug-resistant M. tuberculosis strains of the Haarlem family, namely the strain M, responsible for an extensive multidrug-resistant tuberculosis outbreak, and its kin strain 410 which caused a single case in two decades. Intracellular growth and cytokine responses were evaluated in human monocyte-derived macrophages and dU937 macrophage-like cells. In monocyte-derived macrophages, strain M grew more slowly and induced lower levels of TNF-α and IL-10 than 410, contrasting with previous studies with other strains, where a direct correlation was observed between increased intracellular growth and epidemiological success. On the other hand, in dU937 cells, no difference in growth was observed between both strains, and strain M induced significantly higher TNF-α levels than strain 410. We found that both cell models differed critically in the expression of receptors for M. tuberculosis entry, which might explain the different infection outcomes. Our results in monocyte-derived macrophages suggest that strain M relies on a modest replication rate and cytokine induction, keeping a state of quiescence and remaining rather unnoticed by the host. Collectively, our results underscore the impact of M. tuberculosis intra-species variations on the outcome of host cell infection and show that results can differ depending on the in vitro infection model.


Assuntos
Macrófagos/microbiologia , Mycobacterium tuberculosis/genética , Tuberculose Resistente a Múltiplos Medicamentos/microbiologia , Análise de Variância , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Feminino , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Modelos Biológicos , Mycobacterium tuberculosis/patogenicidade
13.
J Immunol ; 189(11): 5113-7, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23100513

RESUMO

Although the NLRP3 inflammasome plays a pivotal role in host defense, its uncontrolled activation is associated with inflammatory disorders, suggesting that regulation of the inflammasome is important to prevent detrimental effects. Type I IFNs and long-term LPS stimulation were shown to negatively regulate NLRP3 activation. In this study, we found that endogenous NO is involved in the regulation of NLRP3 inflammasome activation by either IFN-ß pretreatment or long-term LPS stimulation. Furthermore, S-nitroso-N-acetylpenicillamine (SNAP), an NO donor, markedly inhibited NLRP3 inflammasome activation, whereas the AIM2 and NLRC4 inflammasomes were only partially inhibited by SNAP. An increase in mitochondrial reactive oxygen species induced by ATP was only modestly affected by SNAP treatment. Interestingly, S-nitrosylation of NLRP3 was detected in macrophages treated with SNAP, and this modification may account for the NO-mediated mechanism controlling inflammasome activation. Taken together, these results revealed a novel role for NO in regulating the NLRP3 inflammasome.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Inflamassomos/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Óxido Nítrico/metabolismo , S-Nitroso-N-Acetilpenicilamina/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Células Cultivadas , Proteínas de Ligação a DNA , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamassomos/imunologia , Interferon beta/imunologia , Interferon beta/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/imunologia , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Espécies Reativas de Oxigênio/metabolismo , S-Nitroso-N-Acetilpenicilamina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
14.
Blood ; 120(6): 1299-308, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22723549

RESUMO

Chronic infantile neurologic cutaneous and articular (CINCA) syndrome is an IL-1-driven autoinflammatory disorder caused mainly by NLRP3 mutations. The pathogenesis of CINCA syndrome patients who carry NLRP3 mutations as somatic mosaicism has not been precisely described because of the difficulty in separating individual cells based on the presence or absence of the mutation. Here we report the generation of NLRP3-mutant and nonmutant-induced pluripotent stem cell (iPSC) lines from 2 CINCA syndrome patients with somatic mosaicism, and describe their differentiation into macrophages (iPS-MPs). We found that mutant cells are predominantly responsible for the pathogenesis in these mosaic patients because only mutant iPS-MPs showed the disease relevant phenotype of abnormal IL-1ß secretion. We also confirmed that the existing anti-inflammatory compounds inhibited the abnormal IL-1ß secretion, indicating that mutant iPS-MPs are applicable for drug screening for CINCA syndrome and other NLRP3-related inflammatory conditions. Our results illustrate that patient-derived iPSCs are useful for dissecting somatic mosaicism and that NLRP3-mutant iPSCs can provide a valuable platform for drug discovery for multiple NLRP3-related disorders.


Assuntos
Síndromes Periódicas Associadas à Criopirina/patologia , Descoberta de Drogas/métodos , Células-Tronco Pluripotentes Induzidas/patologia , Modelos Teóricos , Mosaicismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Células Cultivadas , Síndromes Periódicas Associadas à Criopirina/tratamento farmacológico , Síndromes Periódicas Associadas à Criopirina/genética , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Lactente , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR
15.
Infect Immun ; 80(7): 2323-32, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22508860

RESUMO

Among a number of laboratory strains of Listeria monocytogenes used in experimental infection, strain LO28 is highly capable of inducing robust beta interferon (IFN-ß) production in infected macrophages. In this study, we investigated the molecular mechanism of the IFN-ß-inducing ability of LO28 by comparing it with that of strain EGD, a low-IFN-ß-inducing strain. It was found that LO28 secretes a large amount of IFN-ß-inducing factor, which turned out to be cyclic di-AMP. The secretion of cyclic di-AMP was dependent on MdrT, a multidrug resistance transporter, and LO28 exhibited a very high level of mdrT expression. The introduction of a null mutation into mdrT abolished the ability of LO28 to induce IFN-ß production. Examination of genes responsible for the regulation of mdrT expression revealed a spontaneous 188-bp deletion in tetR of LO28. By constructing recombinant strains of LO28 and EGD in which tetR from each strain was replaced, it was confirmed that the distinct ability of LO28 is attributable mostly to tetR mutation. We concluded that the strong IFN-ß-inducing ability of LO28 is due to a genetic defect in tetR resulting in the overexpression of mdrT and a concomitant increase in the secretion of cyclic di-AMP through MdrT.


Assuntos
Fosfatos de Dinucleosídeos/metabolismo , Interações Hospedeiro-Patógeno , Interferon beta/metabolismo , Listeria monocytogenes/patogenicidade , Macrófagos/imunologia , Macrófagos/microbiologia , Proteínas Repressoras/deficiência , Animais , DNA Bacteriano/química , DNA Bacteriano/genética , Feminino , Regulação Bacteriana da Expressão Gênica , Listeria monocytogenes/genética , Proteínas de Membrana Transportadoras/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas Repressoras/genética , Análise de Sequência de DNA , Deleção de Sequência
16.
Innate Immun ; 18(5): 764-73, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22419537

RESUMO

Modification of a lipid A moiety in Gram-negative bacterial LPS to a less acylated form is thought to facilitate bacterial evasion of host innate immunity, thereby enhancing pathogenicity. The contribution of less-acylated lipid A to interactions of whole bacterial cells with host cells (especially in humans) remains unclear. Mutant strains of Salmonella enterica serovar Typhimurium with fewer acylated groups were generated. The major lipid A form in wild-type (WT) and the mutant KCS237 strain is hexa-acylated; in mutant strains KCS311 and KCS324 it is penta-acylated; and in KCS369 it is tetra-acylated. WT and KCS237 formalin-killed and live bacteria, as well as their LPS, strongly stimulated production of pro-inflammatory cytokines in human U937 cells; this stimulation was suppressed by TLR4 suppressors. LPS of other mutants produced no agonistic activity, but strong antagonistic activity, while their formalin-killed and live bacteria preparations had weak agonistic and no antagonistic activity. Moreover, these less-acylated mutants had increased resistance to phagocytosis by U937 cells. Our results indicate that a decrease of one acyl group (from six to five) is enough to allow Salmonella to evade human innate immunity and that the antagonistic activity of less-acylated lipid A is not utilized for this evasion.


Assuntos
Lipídeo A/metabolismo , Macrófagos/imunologia , Salmonella typhi/imunologia , Receptor 4 Toll-Like/metabolismo , Febre Tifoide/imunologia , Acetilação , Anticorpos Bloqueadores/farmacologia , Citocinas/metabolismo , Humanos , Evasão da Resposta Imune , Imunidade Inata , Mediadores da Inflamação/metabolismo , Lipídeo A/genética , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Mutação/genética , Salmonella typhi/patogenicidade , Células U937
17.
Blood ; 119(2): 434-44, 2012 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-22123848

RESUMO

Adult T-cell leukemia (ATL) patients and human T-cell leukemia virus-1 (HTLV-1) infected individuals succumb to opportunistic infections. Cell mediated immunity is impaired, yet the mechanism of this impairment has remained elusive. The HTLV-1 basic leucine zipper factor (HBZ) gene is encoded in the minus strand of the viral DNA and is constitutively expressed in infected cells and ATL cells. To test the hypothesis that HBZ contributes to HTLV-1-associated immunodeficiency, we challenged transgenic mice that express the HBZ gene in CD4 T cells (HBZ-Tg mice) with herpes simplex virus type 2 or Listeria monocytogenes, and evaluated cellular immunity to these pathogens. HBZ-Tg mice were more vulnerable to both infections than non-Tg mice. The acquired immune response phase was specifically suppressed, indicating that cellular immunity was impaired in HBZ-Tg mice. In particular, production of IFN-γ by CD4 T cells was suppressed in HBZ-Tg mice. HBZ suppressed transcription from the IFN-γ gene promoter in a CD4 T cell-intrinsic manner by inhibiting nuclear factor of activated T cells and the activator protein 1 signaling pathway. This study shows that HBZ inhibits CD4 T-cell responses by directly interfering with the host cell-signaling pathway, resulting in impaired cell-mediated immunity in vivo.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Citocinas/metabolismo , Imunidade Celular/imunologia , Interferon gama/genética , Células Th1/imunologia , Proteínas Virais/fisiologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica , Herpes Simples/imunologia , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 2/patogenicidade , Humanos , Interferon gama/metabolismo , Listeria monocytogenes/patogenicidade , Listeriose/imunologia , Listeriose/metabolismo , Listeriose/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas , Proteínas dos Retroviridae , Células Th1/metabolismo , Fator de Transcrição AP-1/metabolismo
18.
J Immunol ; 187(9): 4890-9, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21957143

RESUMO

Streptococcus pneumoniae is a Gram-positive, extracellular bacterium that is responsible for significant mortality and morbidity worldwide. Pneumolysin (PLY), a cytolysin produced by all clinical isolates of the pneumococcus, is one of the most important virulence factors of this pathogen. We have previously reported that PLY is an essential factor for activation of caspase-1 and consequent secretion of IL-1ß and IL-18 in macrophages infected with S. pneumoniae. However, the host molecular factors involved in caspase-1 activation are still unclear. To further elucidate the mechanism of caspase-1 activation in macrophages infected with S. pneumoniae, we examined the involvement of inflammasomes in inducing this cellular response. Our study revealed that apoptosis-associated specklike protein containing a caspase recruitment domain (ASC), an adaptor protein for inflammasome receptors such as nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) and absent in melanoma 2 (AIM2), is essentially required for the induction of caspase-1 activation by S. pneumoniae. Caspase-1 activation was partially impaired in NLRP3(-/-) macrophages, whereas knockdown and knockout of AIM2 resulted in a clear decrease in caspase-1 activation in response to S. pneumoniae. These results suggest that ASC inflammasomes, including AIM2 and NLRP3, are critical for caspase-1 activation induced by S. pneumoniae. Furthermore, ASC(-/-) mice were more susceptible than wild-type mice to S. pneumoniae, with impaired secretion of IL-1ß and IL-18 into the bronchoalveolar lavage after intranasal infection, suggesting that ASC inflammasomes contribute to the protection of host from infection with PLY-producing S. pneumoniae.


Assuntos
Caspase 1/metabolismo , Proteínas do Citoesqueleto/fisiologia , Imunidade Inata , Inflamassomos/fisiologia , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/biossíntese , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/fisiologia , Caspase 1/deficiência , Caspase 1/genética , Linhagem Celular , Linhagem Celular Transformada , Células Cultivadas , Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA , Resistência à Doença/imunologia , Ativação Enzimática/imunologia , Feminino , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Nucleares/fisiologia , Infecções Pneumocócicas/enzimologia , Estreptolisinas/antagonistas & inibidores , Estreptolisinas/biossíntese
19.
J Med Microbiol ; 60(Pt 5): 582-591, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21233299

RESUMO

PPE37 is a member of the Mycobacterium tuberculosis proline-proline-glutamic acid (PPE) multigene family. Its expression is upregulated in bacteria that are phagocytosed by macrophages and is enhanced even more in bacteria isolated from the lungs of infected mice. This raises the possibility that PPE37 may play a role in the virulence of M. tuberculosis and led to this investigation of the function of PPE37. Recombinant bacterial strains, one expressing the M. tuberculosis PPE37 protein (Ms_ppe37) and another harbouring the vector alone (Ms_vec) were generated from the non-pathogenic Mycobacterium smegmatis. These bacterial strains were used to infect peritoneal exudate and bone marrow-derived macrophages. It was found that, despite the comparable intracellular survival between the two recombinant M. smegmatis strains, Ms_ppe37 induced a significantly lower level of tumour necrosis factor alpha and interleukin 6 in the infected macrophages compared with Ms_vec. Western blot analyses revealed that the activation levels of nuclear factor kappa B, mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase and MAPK/p38 were lower in macrophages infected with Ms_ppe37 than in macrophages infected with Ms_vec. These results suggest that PPE37 may have a potential role in interfering with the pro-inflammatory cytokine response of infected macrophages.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/imunologia , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Animais , Variação Antigênica , Sequência de Bases , Morte Celular , DNA Bacteriano/genética , Feminino , Genes Bacterianos , Vetores Genéticos , Interleucina-6/biossíntese , Interleucina-6/genética , Sistema de Sinalização das MAP Quinases , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Recombinação Genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Virulência/genética , Virulência/imunologia
20.
Int Immunol ; 22(12): 915-25, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21047981

RESUMO

A major concern still prevails as to the reason why various mycobacteria are able to persist within infected host in which protective immunity is generated. To address this question, we monitored the generation of protective T cells during infection with Mycobacterium bovis bacillus Calmette-Guérin (BCG). CD4(+) T cells obtained 3 weeks after infection conferred protection against Mycobacterium tuberculosis challenge and produced IFN-γ and tumor necrosis factor (TNF)-α upon antigen stimulation. However, these abilities were decreased after 6 weeks of infection even though BCG was not thoroughly eliminated from the host. We analyzed the expression of ligands for the CD28/CTLA-4 family receptors on antigen-presenting cells and found that the expression of PD-L1, a ligand for programmed cell death-1 (PD-1), was up-regulated later than 3 weeks of infection. We also found that bacterial numbers in the spleen of PD-1-deficient mice were significantly reduced compared with wild-type mice at 6 and 12 weeks after BCG infection. Furthermore, CD4(+) T cells of PD-1-deficient mice showed a higher ability to confer protection and produce IFN-γ and TNF-α even at 12 weeks after infection. These results indicate that the PD-1-PD-L1 pathway impairs T(h)1 immunity in the late stage of BCG infection, thereby facilitating the bacterial persistence in the host.


Assuntos
Antígenos de Diferenciação/metabolismo , Antígeno B7-1/metabolismo , Vacina BCG/imunologia , Interações Hospedeiro-Patógeno/imunologia , Glicoproteínas de Membrana/metabolismo , Mycobacterium bovis/imunologia , Peptídeos/metabolismo , Células Th1/imunologia , Tuberculose/imunologia , Animais , Antígenos de Diferenciação/genética , Antígeno B7-H1 , Interferon gama/biossíntese , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Morte Celular Programada 1 , Baço/microbiologia , Células Th1/metabolismo , Fatores de Tempo , Tuberculose/microbiologia , Fator de Necrose Tumoral alfa/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA