Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Immunity ; 44(4): 889-900, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27084119

RESUMO

Metagenomic studies show that diverse resident viruses inhabit the healthy gut; however, little is known about the role of these viruses in the maintenance of gut homeostasis. We found that mice treated with antiviral cocktail displayed more severe dextran sulfate sodium (DSS)-induced colitis compared with untreated mice. DSS-induced colitis was associated with altered enteric viral abundance and composition. When wild-type mice were reconstituted with Toll-like receptor 3 (TLR3) or TLR7 agonists or inactivated rotavirus, colitis symptoms were significantly ameliorated. Mice deficient in both TLR3 and TLR7 were more susceptible to DSS-induced experimental colitis. In humans, combined TLR3 and TLR7 genetic variations significantly influenced the severity of ulcerative colitis. Plasmacytoid dendritic cells isolated from inflamed mouse colon produced interferon-ß in a TLR3 and TLR7-dependent manner. These results imply that recognition of resident viruses by TLR3 and TLR7 is required for protective immunity during gut inflammation.


Assuntos
Colite/imunologia , Trato Gastrointestinal/virologia , Interferon beta/imunologia , Glicoproteínas de Membrana/imunologia , Rotavirus/imunologia , Receptor 3 Toll-Like/imunologia , Receptor 7 Toll-Like/imunologia , Animais , Antivirais/farmacologia , Colite/induzido quimicamente , Células Dendríticas/imunologia , Sulfato de Dextrana , Microbioma Gastrointestinal , Trato Gastrointestinal/imunologia , Humanos , Inflamação/imunologia , Interferon beta/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , RNA Ribossômico 16S/genética , Receptor 3 Toll-Like/genética , Receptor 7 Toll-Like/genética
2.
Immunity ; 42(4): 744-55, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25862092

RESUMO

The microbiota stimulates inflammation, but the signaling pathways and the members of the microbiota involved remain poorly understood. We found that the microbiota induces interleukin-1ß (IL-1ß) release upon intestinal injury and that this is mediated via the NLRP3 inflammasome. Enterobacteriaceae and in particular the pathobiont Proteus mirabilis, induced robust IL-1ß release that was comparable to that induced by the pathogen Salmonella. Upon epithelial injury, production of IL-1ß in the intestine was largely mediated by intestinal Ly6C(high) monocytes, required chemokine receptor CCR2 and was abolished by deletion of IL-1ß in CCR2(+) blood monocytes. Furthermore, colonization with P. mirabilis promoted intestinal inflammation upon intestinal injury via the production of hemolysin, which required NLRP3 and IL-1 receptor signaling in vivo. Thus, upon intestinal injury, selective members of the microbiota stimulate newly recruited monocytes to induce NLRP3-dependent IL-1ß release, which promotes inflammation in the intestine.


Assuntos
Proteínas de Transporte/imunologia , Inflamassomos/imunologia , Interleucina-1beta/imunologia , Microbiota/imunologia , Monócitos/imunologia , Simbiose/imunologia , Animais , Antígenos Ly/genética , Antígenos Ly/imunologia , Proteínas de Transporte/genética , Regulação da Expressão Gênica , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/imunologia , Inflamassomos/genética , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Interleucina-1beta/genética , Intestinos/imunologia , Intestinos/lesões , Intestinos/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/microbiologia , Monócitos/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Infecções por Proteus/genética , Infecções por Proteus/imunologia , Infecções por Proteus/microbiologia , Infecções por Proteus/patologia , Proteus mirabilis/imunologia , Receptores CCR2/genética , Receptores CCR2/imunologia , Salmonella/imunologia , Infecções por Salmonella/genética , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Infecções por Salmonella/patologia , Transdução de Sinais
3.
Eur J Immunol ; 51(6): 1461-1472, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33548071

RESUMO

Blocking the mevalonate pathway for cholesterol reduction by using statin may have adverse effects including statin-induced colitis. Moreover, one of the predisposing factors for colitis is an imbalanced CD4+ T cell, which can be observed on the complete deletion of HMG-CoA reductase (HMGCR), a target of statins. In this study, we inquired geranylgeranyl pyrophosphate (GGPP) is responsible for maintaining the T-cell homeostasis. Following dextran sulfate sodium (DSS)-induced colitis, simvastatin increased the severity of disease, while cotreatment with GGPP, but not with cholesterol, reversed the disease magnitude. GGPP ameliorated DSS-induced colitis by increasing Treg cells. GGPP amplified Treg differentiation through increased IL-2/STAT 5 signaling. GGPP prenylated Ras protein, a prerequisite for extracellular signal-regulated kinase (ERK) pathway activation, leading to increased IL-2 production. Higher simvastatin dose increased the severity of colitis. GGPP ameliorated simvastatin-increased colitis by increasing Treg cells. Treg cells, which have the capacity to suppress inflammatory T cells and were generated through IL-2/STAT5 signaling, increased IL-2 production through prenylation and activation of the Ras/ERK pathway.


Assuntos
Anticolesterolemiantes/administração & dosagem , Linfócitos T CD4-Positivos/imunologia , Colite/imunologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/imunologia , Doenças Inflamatórias Intestinais/imunologia , Interleucina-2/metabolismo , Fosfatos de Poli-Isoprenil/uso terapêutico , Sinvastatina/administração & dosagem , Linfócitos T Reguladores/imunologia , Animais , Anticolesterolemiantes/efeitos adversos , Diferenciação Celular , Células Cultivadas , Colite/etiologia , Sulfato de Dextrana , Modelos Animais de Doenças , Homeostase , Humanos , Hidroximetilglutaril-CoA Redutases/genética , Ativação Linfocitária , Ácido Mevalônico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Sinvastatina/efeitos adversos
4.
Nanomedicine ; 37: 102438, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34256061

RESUMO

Highly pathogenic avian influenza viruses (HPAIVs) pose a significant threat to human health, with high mortality rates, and require effective vaccines. We showed that, harnessed with novel RNA-mediated chaperone function, hemagglutinin (HA) of H5N1 HPAIV could be displayed as an immunologically relevant conformation on self-assembled chimeric nanoparticles (cNP). A tri-partite monomeric antigen was designed including: i) an RNA-interaction domain (RID) as a docking tag for RNA to enable chaperna function (chaperna: chaperone + RNA), ii) globular head domain (gd) of HA as a target antigen, and iii) ferritin as a scaffold for 24 mer-assembly. The immunization of mice with the nanoparticles (~46 nm) induced a 25-30 fold higher neutralizing capacity of the antibody and provided cross-protection from homologous and heterologous lethal challenges. This study suggests that cNP assembly is conducive to eliciting antibodies against the conserved region in HA, providing potent and broad protective efficacy.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Vacinas contra Influenza/imunologia , Influenza Aviária/imunologia , RNA/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/uso terapêutico , Aves/virologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/uso terapêutico , Humanos , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/patogenicidade , Vacinas contra Influenza/química , Vacinas contra Influenza/uso terapêutico , Influenza Aviária/prevenção & controle , Influenza Aviária/virologia , Camundongos , Nanopartículas/química , Nanopartículas/uso terapêutico , Pandemias , RNA/genética , RNA/uso terapêutico
5.
Immunology ; 158(3): 219-229, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31478196

RESUMO

The role of symbiotic bacteria in the development of antigen-specific immunity remains poorly understood. Previous studies showed that sensing of symbiotic bacteria by nucleotide-binding oligomerization domain-containing protein 2 (Nod2) regulates antibody responses in response to nasal immunization with antigen and cholera toxin (CT). In this study, we examined the role of the microbiota in the adjuvant activity of CT induced after oral immunization with antigen. Germ-free (GF) mice showed impaired production of antibody responses and T-cell-specific cytokines after oral immunization when compared with that observed in conventionally raised mice. Similar to GF mice, Nod2-deficient mice showed reduced humoral responses upon oral immunization with antigen and CT. Treatment with CT enhanced the production of interleukin-1ß (IL-1ß), but not tumor necrosis factor-α or IL-12p40, induced by stimulation of dendritic cells with muramyl dipeptide, the Nod2 ligand. Mechanistically, the enhanced production of IL-1ß induced by muramyl dipeptide and CT stimulation required Nod2 and was mediated by both increased synthesis of pro-IL-1ß and caspase-1 activation. Furthermore, antigen-specific antibody and cytokine responses induced by CT were impaired in orally immunized IL-1ß-deficient mice. Collectively, our results indicate that Nod2 stimulation by symbiotic bacteria contributes to optimal CT-mediated antigen-specific oral vaccination through the induction of IL-1ß production.


Assuntos
Adjuvantes Imunológicos/farmacologia , Toxina da Cólera/farmacologia , Células Dendríticas/imunologia , Interleucina-1beta/imunologia , Microbiota/imunologia , Proteína Adaptadora de Sinalização NOD2/imunologia , Administração Oral , Animais , Subunidade p40 da Interleucina-12/imunologia , Camundongos , Camundongos Knockout , Proteína Adaptadora de Sinalização NOD2/genética , Fator de Necrose Tumoral alfa/imunologia
6.
J Immunol ; 199(4): 1362-1371, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28674182

RESUMO

Enteric pathogens including Salmonella enteric serovar Typhimurium can breach the epithelial barrier of the host and spread to systemic tissues. In response to infection, the host activates innate immune receptors via the signaling molecule MyD88, which induces protective inflammatory and antimicrobial responses. Most of these innate immune responses have been studied in hematopoietic cells, but the role of MyD88 signaling in other cell types remains poorly understood. Surprisingly, we found that Dermo1-Cre;Myd88fl/fl mice with mesenchymal cell-specific deficiency of MyD88 were less susceptible to orogastric and i.p. STyphimurium infection than their Myd88fl/fl littermates. The reduced susceptibility of Dermo1-Cre;Myd88fl/fl mice to infection was associated with lower loads of S. Typhimurium in the liver and spleen. Mutant analyses revealed that S. Typhimurium employs its virulence type III secretion system 2 to promote its growth through MyD88 signaling pathways in mesenchymal cells. Inflammatory monocytes function as a major cell population for systemic dissemination of S. Typhimurium Mechanistically, mesenchymal cell-specific MyD88 signaling promoted CCL2 production in the liver and spleen and recruitment of inflammatory monocytes to systemic organs in response to STyphimurium infection. Consistently, MyD88 signaling in mesenchymal cells enhanced the number of phagocytes including Ly6ChiLy6G- inflammatory monocytes harboring STyphimurium in the liver. These results suggest that S. Typhimurium promotes its systemic growth and dissemination through MyD88 signaling pathways in mesenchymal cells.


Assuntos
Monócitos/imunologia , Monócitos/microbiologia , Fator 88 de Diferenciação Mieloide/metabolismo , Salmonelose Animal/imunologia , Salmonelose Animal/microbiologia , Salmonella typhimurium/patogenicidade , Animais , Antígenos Ly/análise , Carga Bacteriana , Quimiocina CCL2/biossíntese , Imunidade Inata , Fígado/imunologia , Fígado/microbiologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/metabolismo , Transdução de Sinais , Baço/imunologia , Baço/microbiologia , Sistemas de Secreção Tipo III/metabolismo
8.
J Infect Dis ; 207(1): 50-60, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23100560

RESUMO

Interleukin (IL)-1 is a well-known cytokine for the initiation of innate immunity in bacterial infection. However, the underlying mechanism of IL-1 on the respiratory infection is not fully elucidated. We studied how IL-1 contributes to the host defense against Streptococcus pneumoniae. IL-1R(-/-) mice showed high mortality, local cytokine storm, and substantial infiltrates in the lower respiratory tract after intratracheal challenge with S. pneumoniae. The IL-1-deficient condition did not suppress the propagation of bacteria in the lung, although the recruitment and the bacteria-killing ability of neutrophils (CD11b(+)Ly6C(+)Ly6G(+)) were not defective compared with wild-type mice. Unexpectedly, we found that the transcription of fibrinogen alpha and gamma genes were highly activated in the lungs of wild-type mice after the infection, whereas no significant changes were found in IL-1R(-/-) mice. Of note, synthesis of fibrinogen was dependent on the IL-1-IL-6-Stat3 cascade. Treatment with recombinant fibrinogen improved survival and bacterial propagation in the IL-1R(-/-) mice and blockade of the coagulation increased the susceptibility of wild-type mice to pneumococcal pneumonia. Our findings suggest that IL-1 signaling leads to the synthesis of fibrinogen in the lung after pneumococcus infection and is followed by coagulation, which contributes to the control of bacterial infection in the pulmonary tract.


Assuntos
Coagulação Sanguínea/imunologia , Interleucina-1/imunologia , Pulmão/imunologia , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae/imunologia , Animais , Suscetibilidade a Doenças , Regulação para Baixo , Fibrinogênio/metabolismo , Imunidade Inata , Pulmão/metabolismo , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Neutrófilos/imunologia , Pneumonia Pneumocócica/tratamento farmacológico , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/mortalidade , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Proteínas Recombinantes , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Streptococcus pneumoniae/efeitos dos fármacos , Análise de Sobrevida , Trombina/metabolismo
9.
Semin Immunopathol ; 46(1-2): 2, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38990345

RESUMO

The gut microbiota, housing trillions of microorganisms within the gastrointestinal tract, has emerged as a critical regulator of host health and homeostasis. Through complex metabolic interactions, these microorganisms produce a diverse range of metabolites that substantially impact various physiological processes within the host. This review aims to delve into the intricate relationships of gut microbiota-derived metabolites and their influence on the host homeostasis. We will explore how these metabolites affect crucial aspects of host physiology, including metabolism, mucosal integrity, and communication among gut tissues. Moreover, we will spotlight the potential therapeutic applications of targeting these metabolites to restore and sustain host equilibrium. Understanding the intricate interplay between gut microbiota and their metabolites is crucial for developing innovative strategies to promote wellbeing and improve outcomes of chronic diseases.


Assuntos
Microbioma Gastrointestinal , Homeostase , Humanos , Microbioma Gastrointestinal/imunologia , Animais , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/imunologia , Metaboloma , Interações entre Hospedeiro e Microrganismos/imunologia
10.
Exp Mol Med ; 56(1): 192-209, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38200153

RESUMO

Tumor necrosis factor superfamily (TNFSF) resistance contributes to the development and progression of tumors and resistance to various cancer therapies. Tumor-intrinsic alterations involved in the adaptation to the TNFSF response remain largely unknown. Here, we demonstrate that protein kinase C substrate 80K-H (PRKCSH) abundance in lung cancers boosts oncogenic IGF1R activation, leading to TNFSF resistance. PRKCSH abundance is correlated with IGF1R upregulation in lung cancer tissues. Specifically, PRKCSH interacts with IGF1R and extends its half-life. The PRKCSH-IGF1R axis in tumor cells impairs caspase-8 activation, increases Mcl-1 expression, and inhibits caspase-9, leading to an imbalance between cell death and survival. PRKCSH deficiency augmented the antitumor effects of natural killer (NK) cells, representative TNFSF effector cells, in a tumor xenograft IL-2Rg-deficient NOD/SCID (NIG) mouse model. Our data suggest that PRKCSH plays a critical role in TNFSF resistance and may be a potential target to improve the efficacy of NK cell-based cancer therapy.


Assuntos
Neoplasias Pulmonares , Animais , Camundongos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Meia-Vida , Linhagem Celular Tumoral , Camundongos Endogâmicos NOD , Camundongos SCID , Fatores de Necrose Tumoral/metabolismo , Proteínas de Ligação ao Cálcio , Glucosidases/metabolismo , Receptor IGF Tipo 1/metabolismo
11.
Emerg Microbes Infect ; 13(1): 2362392, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38808613

RESUMO

Japanese encephalitis (JE), caused by the Japanese encephalitis virus (JEV) infection, continues to pose significant public health challenges worldwide despite efficient vaccines. The virus is classified into five genotypes, among which genotype V (GV) was not detected for a long period after its initial isolation in 1952, until reports emerged from China and the Republic of Korea (ROK) since 2009. The characteristics of the virus are crucial in estimating its potential epidemiological impact. However, characterization of GV JEVs has so far been limited to two strains: Muar, the original isolate, and XZ0934, isolated in China. Two additional ROK GV JEV isolates, NCCP 43279 and NCCP 43413, are currently available, but their characteristics have not been explored. Our phylogenetic analysis revealed that GV virus sequences from the ROK segregate into two clades. NCCP 43279 and NCCP 43413 belong to different clades and exhibit distinct in vitro phenotypes. NCCP 43279 forms larger plaques but demonstrates inefficient propagation in cell culture compared to NCCP 43413. In vivo, NCCP 43279 induces higher morbidity and mortality in mice than NCCP 43413. Notably, NCCP 43279 shows more severe blood-brain barrier damage, suggesting superior brain invasion capabilities. Consistent with its higher virulence, NCCP 43279 displays more pronounced histopathological and immunopathological outcomes. In conclusion, our study confirms that the two ROK isolates are not only classified into different clades but also exhibit distinct in vitro and in vivo characteristics.


Assuntos
Vírus da Encefalite Japonesa (Espécie) , Encefalite Japonesa , Genótipo , Filogenia , Vírus da Encefalite Japonesa (Espécie)/genética , Vírus da Encefalite Japonesa (Espécie)/isolamento & purificação , Vírus da Encefalite Japonesa (Espécie)/classificação , Animais , República da Coreia/epidemiologia , Encefalite Japonesa/virologia , Encefalite Japonesa/veterinária , Encefalite Japonesa/epidemiologia , Camundongos , Humanos , Virulência , Linhagem Celular , Feminino
12.
Emerg Microbes Infect ; 13(1): 2343910, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38618740

RESUMO

Japanese encephalitis (JE), caused by the Japanese encephalitis virus (JEV), is a highly threatening disease with no specific treatment. Fortunately, the development of vaccines has enabled effective defense against JE. However, re-emerging genotype V (GV) JEV poses a challenge as current vaccines are genotype III (GIII)-based and provide suboptimal protection. Given the isolation of GV JEVs from Malaysia, China, and the Republic of Korea, there is a concern about the potential for a broader outbreak. Under the hypothesis that a GV-based vaccine is necessary for effective defense against GV JEV, we developed a pentameric recombinant antigen using cholera toxin B as a scaffold and mucosal adjuvant, which was conjugated with the E protein domain III of GV by genetic fusion. This GV-based vaccine antigen induced a more effective immune response in mice against GV JEV isolates compared to GIII-based antigen and efficiently protected animals from lethal challenges. Furthermore, a bivalent vaccine approach, inoculating simultaneously with GIII- and GV-based antigens, showed protective efficacy against both GIII and GV JEVs. This strategy presents a promising avenue for comprehensive protection in regions facing the threat of diverse JEV genotypes, including both prevalent GIII and GI as well as emerging GV strains.


Assuntos
Vírus da Encefalite Japonesa (Espécie) , Encefalite Japonesa , Genótipo , Vacinas contra Encefalite Japonesa , Vírus da Encefalite Japonesa (Espécie)/genética , Vírus da Encefalite Japonesa (Espécie)/imunologia , Vírus da Encefalite Japonesa (Espécie)/classificação , Animais , Encefalite Japonesa/prevenção & controle , Encefalite Japonesa/imunologia , Encefalite Japonesa/virologia , Vacinas contra Encefalite Japonesa/imunologia , Vacinas contra Encefalite Japonesa/administração & dosagem , Vacinas contra Encefalite Japonesa/genética , Camundongos , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Humanos , Camundongos Endogâmicos BALB C , Feminino , Antígenos Virais/imunologia , Antígenos Virais/genética , Eficácia de Vacinas , Toxina da Cólera/genética , Toxina da Cólera/imunologia
13.
Br J Pharmacol ; 2024 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-38852992

RESUMO

BACKGROUND AND PURPOSE: Only limited therapeutic agents have been developed for non-alcoholic steatohepatitis (NASH). Glabridin, a promising anti-obesity candidate, has only limited druggability due to its low in vivo chemical stability and bioavailability. Therefore, we developed vutiglabridin (VUTI), which is based on a glabridin backbone, and investigated its mechanism of action in treating NASH in animal models. EXPERIMENTAL APPROACH: Anti-NASH effects of VUTI were determined in in vitro fatty liver models, spheroids of primary human hepatocytes and L02 normal liver cell lines. To identify VUTI possible cellular target/s, biotin-labelled VUTI was synthesized and underwent chemical proteomic analysis. Further, the evaluation of VUTI therapeutic efficacy was carried out using an amylin-NASH and high-fat (HF) diet-induced obese (DIO) mouse models. This was carried out using transcriptomic, lipidomic and proteomic analyses of the livers from the amylin-NASH mouse model. KEY RESULTS: VUTI treatment markedly reduces hepatic steatosis, fibrosis and inflammation by promoting lipid catabolism, activating autophagy and improving mitochondrial dysfunction, all of which are hallmarks of effective NASH treatment. The cellular target of VUTI was identified as paraoxonase 2 (PON2), a newly proposed protein target for the treatment of NASH, VUTI enhanced PON2 activity. The results using PON2 knockdown cells demonstrated that PON2 is important for VUTI- activation of autophagy, promoting mitochondrial function, decreasing oxidative stress and alleviating lipid accumulation under lipotoxic condition. CONCLUSION AND IMPLICATIONS: Our data demonstrated that VUTI is a promising therapeutic for NASH. Targeting PON2 may be important for improving liver function in various immune-metabolic diseases including NASH.

14.
Eur J Immunol ; 42(3): 618-28, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22105301

RESUMO

The role of TLR signaling in linking the innate and adaptive immune systems has been a controversial issue that remains to be solved. Here, we determined whether MyD88-dependent TLR signals are required for the generation of B-cell responses during chronic Salmonella infection. Oral administration of recombinant attenuated Salmonella enterica serovar Typhimurium vaccine (RASV) strain in MyD88(-/-) mice resulted in chronic infection. Infection was accompanied by enlarged germinal centers and hypergammaglobulinemia with anti-double-stranded DNA (dsDNA)-specific Ab in sera, and the deposition of immune complexes in the kidneys, suggesting onset of autoimmunity. CD4(+) T cells expressing PD-1, CXCR5, ICOS, and IL-21 were dramatically increased in chronically infected mice, indicating the expansion of follicular helper T (Tfh)-like cells. Of note, the depletion of CD4(+) T cells completely blocked the generation of polyclonal IgG Ab in sera after oral RASV challenge. Inflammatory myeloid cells expressing CD11b and Gr-1 accumulated in high numbers in the spleen of MyD88(-/-) mice. Interestingly, the blockade of PD-1 or ICOS significantly reduced the hypergammaglobulinemia and dsDNA-specific autoantibody production. Overall, these results suggest that Tfh-like cells in chronic bacterial infection trigger autoimmune hypergammaglobulinemia in a PD-1- and ICOS-dependent manner.


Assuntos
Autoimunidade/imunologia , Linfócitos B/imunologia , Hipergamaglobulinemia/microbiologia , Fator 88 de Diferenciação Mieloide/deficiência , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Hipergamaglobulinemia/imunologia , Imunoglobulina G/sangue , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Interleucinas/economia , Interleucinas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/imunologia , Receptor de Morte Celular Programada 1 , RNA/química , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores CXCR5/genética , Receptores CXCR5/imunologia , Infecções por Salmonella/microbiologia , Organismos Livres de Patógenos Específicos
15.
PLoS Pathog ; 7(2): e1001304, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21383977

RESUMO

Type I interferon (IFN-I) plays a critical role in the homeostasis of hematopoietic stem cells and influences neutrophil influx to the site of inflammation. IFN-I receptor knockout (Ifnar1⁻/⁻) mice develop significant defects in the infiltration of Ly6C(hi) monocytes in the lung after influenza infection (A/PR/8/34, H1N1). Ly6C(hi) monocytes of wild-type (WT) mice are the main producers of MCP-1 while the alternatively generated Ly6C(int) monocytes of Ifnar1⁻/⁻ mice mainly produce KC for neutrophil influx. As a consequence, Ifnar1⁻/⁻ mice recruit more neutrophils after influenza infection than do WT mice. Treatment of IFNAR1 blocking antibody on the WT bone marrow (BM) cells in vitro failed to differentiate into Ly6C(hi) monocytes. By using BM chimeric mice (WT BM into Ifnar1⁻/⁻ and vice versa), we confirmed that IFN-I signaling in hematopoietic cells is required for the generation of Ly6C(hi) monocytes. Of note, WT BM reconstituted Ifnar1⁻/⁻ chimeric mice with increased numbers of Ly6C(hi) monocytes survived longer than influenza-infected Ifnar1⁻/⁻ mice. In contrast, WT mice that received Ifnar1⁻/⁻ BM cells with alternative Ly6C(int) monocytes and increased numbers of neutrophils exhibited higher mortality rates than WT mice given WT BM cells. Collectively, these data suggest that IFN-I contributes to resistance of influenza infection by control of monocytes and neutrophils in the lung.


Assuntos
Monócitos/metabolismo , Monócitos/patologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Pneumonia Viral/patologia , Receptor de Interferon alfa e beta/fisiologia , Animais , Biomarcadores/metabolismo , Western Blotting , Células da Medula Óssea/metabolismo , Diferenciação Celular , Feminino , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Pneumonia/etiologia , Pneumonia/patologia , Pneumonia Viral/imunologia , Pneumonia Viral/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
16.
J Immunol ; 187(6): 3044-52, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21832166

RESUMO

Previous studies demonstrated cross talk between mucosal and reproductive organs during secretory IgA Ab induction. In this study, we aimed to clarify the underlying mechanisms of this cross talk. We found significantly higher titers of Ag-specific secretory IgA Ab in the vaginal wash after mucosal vaccination by both the intranasal (i.n.) and the intravaginal routes but not by the s.c. route. Interestingly, Ag-specific IgA Ab-secreting cells (ASCs) were found mainly in the uterus but not in the cervix and vaginal canal after i.n. vaccination. The fact that most Ag-specific IgA ASCs isolated from the uteri of vaccinated mice migrated toward mucosa-associated epithelial chemokine (MEC)/CCL28 suggests dominant expression of CCR10 on the IgA ASCs. Further, IgA ASCs in the uteri of vaccinated mice were reduced drastically in mice treated with neutralizing anti-MEC/CCL28 Ab. Most intriguingly, the female sex hormone estrogen directly regulated MEC/CCL28 expression and was augmented by i.n. vaccination with cholera toxin or stimulators for innate immunity. Further, blockage of estrogen function in the uterus by oral administration of the estrogen antagonist raloxifene significantly inhibited migration of Ag-specific IgA ASCs after i.n. vaccination with OVA plus cholera toxin. Taken together, these data strongly suggest that CCR10(+) IgA ASCs induced by mucosal vaccination via the i.n. route migrate into the uterus in a MEC/CCL28-dependent manner and that estrogen might have a crucial role in the protection against genital infection by regulating MEC/CCL28 expression in the uterus.


Assuntos
Quimiocinas CC/biossíntese , Estrogênios/imunologia , Imunidade nas Mucosas/imunologia , Plasmócitos/imunologia , Receptores CCR10/metabolismo , Útero/imunologia , Administração Intranasal , Animais , Células 3T3 BALB , Western Blotting , Quimiocinas CC/imunologia , Quimiotaxia de Leucócito/imunologia , Toxina da Cólera/administração & dosagem , Toxina da Cólera/imunologia , Ensaio de Imunoadsorção Enzimática , Estrogênios/metabolismo , Feminino , Imunoglobulina A Secretora/imunologia , Imunoglobulina A Secretora/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Plasmócitos/metabolismo , Receptores CCR10/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Útero/metabolismo , Vacinação
17.
J Immunol ; 186(12): 6999-7005, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21572021

RESUMO

Although the induction mechanism of secretory IgA has been well studied, that of IgG in the mucosal compartments is not well understood. In this study, vitamin A deficiency was convincingly shown to be associated with increased IgG in serum and intestinal fluid. We found increased numbers of IgG-secreting B cells in the lamina propria of the small intestine and mesenteric lymph node (MLN) of vitamin A-deficient (VAD) mice. Of note, IFN-γ secreted by MLN dendritic cells (DCs) was significantly augmented in VAD mice, unlike control mice, and CD103(+) DCs were the main subsets to secrete IFN-γ. The aberrant increase of IgG in VAD mice can be ascribable to IFN-γ, because IFN-γ(-/-) VAD mice have normal IgG levels and the addition of rIFN-γ increased IgG production by B cells cocultured with MLN DCs from IFN-γ(-/-) VAD mice. Oral feeding of antibiotics resulted in significant reduction of IgG in VAD mice, indicating a critical role for altered commensal bacteria for IgG class-switching recombination in the absence of vitamin A. Collectively, vitamin A deficiency provokes the generation of IFN-γ-secreting CD103(+) DCs, which may be a critical regulator for IgG generation in the MLN.


Assuntos
Células Dendríticas/imunologia , Imunoglobulina G/biossíntese , Interferon gama/imunologia , Linfonodos/imunologia , Deficiência de Vitamina A/imunologia , Animais , Antígenos CD/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Cadeias alfa de Integrinas/imunologia , Interferon gama/metabolismo , Mucosa Intestinal/imunologia , Mesentério/citologia , Mesentério/imunologia , Camundongos
18.
Int Immunopharmacol ; 122: 110628, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37454634

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) is typically managed using medications such as 5-aminosalicylic acid (5-ASA), glucocorticoids, anti-TNFα Ab, or anti-IL-12/23 Ab. However, some patients do not respond well to these treatments or frequently experience relapses. Therefore, alternative therapeutic options are needed. Since the activation of the inflammasome is crucial to the pathogenesis of IBD, inhibiting the inflammasome may be beneficial for patients. MATERIALS AND METHODS: We tested the efficacy of taurodeoxycholate (TDCA), which is a known G-protein coupled receptor 19 (GPCR19) agonist, in a mouse colitis model induced by dextran sodium sulfate (DSS). RESULTS: In the mouse colitis model, TDCA prevented loss of body weight, shortening of the colon, production of pro-inflammatory cytokines, infiltration of pro-inflammatory cells, and mucosal ulceration in the colon. In vitro, TDCA inhibited the activation of NF-κB in bone marrow-derived macrophages (BMDMs) by activating the cAMP-PKA axis. TDCA downregulated the expression of purinergic receptor P2X7 (P2X7R) and enhanced the colocalization of P2X7R with GPCR19, and inhibited the Ca2+ mobilization of BMDMs when stimulated with ATP or BzATP, which plays a pivotal role in activating the NLRP3 inflammasome (N3I) via P2X7R. TDCA inhibited the oligomerization of NLRP3-ASC and downregulated the expression of NLRP3 and ASC, as well as suppressed the maturation of pro-caspase-1 and pro-IL-1ß. TDCA also increased the percentage of M2 macrophages while decreasing the number of M1 macrophages, Th1, Th2, and Th17 cells in the colon. CONCLUSION: TDCA ameliorated DSS-induced colitis in mice, possibly by inhibiting both the priming phase (via the GPCR19-cAMP-PKA-NF-κB axis) and the activation phase (via the GPCR19-P2X7R-NLRP3-Caspase 1-IL-1ß axis) of N3I signaling.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Camundongos , Animais , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , NF-kappa B/metabolismo , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Sulfato de Dextrana , Camundongos Endogâmicos C57BL
19.
Commun Biol ; 6(1): 272, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36922564

RESUMO

Myeloid-derived suppressor cells (MDSCs) consist of monocytic (M-) MDSCs and polymorphonuclear (PMN-) MDSCs that contribute to an immunosuppressive environment in tumor-bearing hosts. However, research on the phenotypic and functional heterogeneity of MDSCs in tumor-bearing hosts and across different disease stage is limited. Here we subdivide M-MDSCs based on CD115 expression and report that CD115- M-MDSCs are functionally distinct from CD115+ M-MDSCs. CD115- M-MDSCs increased in bone marrow and blood as tumors progressed. Transcriptome analysis revealed that CD115- M-MDSCs expressed higher levels of neutrophil-related genes. Moreover, isolated CD115- M-MDSCs had higher potential to be differentiated into PMN-MDSCs compared with CD115+ M-MDSCs. Of note, CD115- M-MDSCs were able to differentiate into both olfactomedin 4 (OLFM4)hi and OLFM4lo PMN-MDSCs, whereas CD115+ M-MDSCs differentiated into a smaller proportion of OLFM4lo PMN-MDSCs. In vivo, M-MDSC to PMN-MDSC differentiation occurred most frequently in bone marrow while M-MDSCs preferentially differentiated into tumor-associated macrophages in the tumor mass. Our study reveals the presence of previously unrecognized subtypes of CD115- M-MDSCs in tumor-bearing hosts and demonstrates their cellular plasticity during tumorigenesis.


Assuntos
Células Supressoras Mieloides , Neoplasias , Humanos , Células Supressoras Mieloides/metabolismo , Neoplasias/patologia , Monócitos , Neutrófilos , Carcinogênese/metabolismo , Fator Estimulador de Colônias de Granulócitos
20.
Microbiome ; 11(1): 260, 2023 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-37996951

RESUMO

BACKGROUND: The modulation of immune responses by probiotics is crucial for local and systemic immunity. Recent studies have suggested a correlation between gut microbiota and lung immunity, known as the gut-lung axis. However, the evidence and mechanisms underlying this axis remain elusive. RESULTS: In this study, we screened various Lactobacillus (L.) strains for their ability to augment type I interferon (IFN-I) signaling using an IFN-α/ß reporter cell line. We identified L. paracasei (MI29) from the feces of healthy volunteers, which showed enhanced IFN-I signaling in vitro. Oral administration of the MI29 strain to wild-type B6 mice for 2 weeks resulted in increased expression of IFN-stimulated genes and pro-inflammatory cytokines in the lungs. We found that MI29-treated mice had significantly increased numbers of CD11c+PDCA-1+ plasmacytoid dendritic cells and Ly6Chi monocytes in the lungs compared with control groups. Pre-treatment with MI29 for 2 weeks resulted in less weight loss and lower viral loads in the lung after a sub-lethal dose of influenza virus infection. Interestingly, IFNAR1-/- mice did not show enhanced viral resistance in response to oral MI29 administration. Furthermore, metabolic profiles of MI29-treated mice revealed changes in fatty acid metabolism, with MI29-derived fatty acids contributing to host defense in a Gpr40/120-dependent manner. CONCLUSIONS: These findings suggest that the newly isolated MI29 strain can activate host defense immunity and prevent infections caused by the influenza virus through the gut-lung axis. Video Abstract.


Assuntos
Doenças Transmissíveis , Influenza Humana , Lacticaseibacillus paracasei , Infecções por Orthomyxoviridae , Orthomyxoviridae , Humanos , Camundongos , Animais , Pulmão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA