Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Nat Immunol ; 14(3): 246-53, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23314004

RESUMO

Immune responses to vaccines require direct recognition of pathogen-associated molecular patterns (PAMPs) through pattern-recognition receptors (PRRs) on dendritic cells (DCs). Unlike vaccination, infection by a live pathogen often impairs DC function and inflicts additional damage on the host. Here we found that after infection with live influenza A virus, signaling through the interleukin 1 receptor (IL-1R) was required for productive priming of CD8(+) T cells, but signaling through the PRRs TLR7 and RIG-I was not. DCs activated by IL-1 in trans were both required and sufficient for the generation of virus-specific CD8(+) T cell immunity. Our data demonstrate a critical role for a bystander cytokine in the priming of CD8(+) T cells during infection with a live virus.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Vírus da Influenza A/imunologia , Receptores de Interleucina-1/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Animais , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular , Movimento Celular , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Interleucina-1/imunologia , Ativação Linfocitária , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Infecções por Orthomyxoviridae/imunologia , Receptores CCR7/biossíntese , Receptores de Superfície Celular , Receptores de Interleucina-1/genética , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais , Receptor 7 Toll-Like/metabolismo
2.
Nat Immunol ; 11(5): 404-10, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20383149

RESUMO

Influenza virus, a negative-stranded RNA virus that causes severe illness in humans and animals, stimulates the inflammasome through the Nod-like receptor NLRP3. However, the mechanism by which influenza virus activates the NLRP3 inflammasome is unknown. Here we show that the influenza virus M2 protein, a proton-selective ion channel important in viral pathogenesis, stimulates the NLRP3 inflammasome pathway. M2 channel activity was required for the activation of inflammasomes by influenza and was sufficient to activate inflammasomes in primed macrophages and dendritic cells. M2-induced activation of inflammasomes required its localization to the Golgi apparatus and was dependent on the pH gradient. Our results show a mechanism by which influenza virus infection activates inflammasomes and identify the sensing of disturbances in intracellular ionic concentrations as a previously unknown pathogen-recognition pathway.


Assuntos
Proteínas de Transporte/metabolismo , Células Dendríticas/metabolismo , Canais Iônicos/metabolismo , Macrófagos/metabolismo , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/fisiologia , Proteínas da Matriz Viral/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/patologia , Células Dendríticas/virologia , Engenharia Genética , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Complexo de Golgi/virologia , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Canais Iônicos/genética , Canais Iônicos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Macrófagos/virologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monensin/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Oncogênicas Virais/farmacologia , Orthomyxoviridae/patogenicidade , Infecções por Orthomyxoviridae/fisiopatologia , Cloreto de Potássio/farmacologia , Transporte Proteico/efeitos dos fármacos , Prótons , Deleção de Sequência/genética , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/imunologia , Receptor 7 Toll-Like/metabolismo , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Replicação Viral
3.
Int Immunol ; 33(9): 479-490, 2021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34161582

RESUMO

RNase T2, a ubiquitously expressed RNase, degrades RNAs in the endosomal compartments. RNA sensors, double-stranded RNA (dsRNA)-sensing Toll-like receptor 3 (TLR3) and single-stranded RNA (ssRNA)-sensing TLR7, are localized in the endosomal compartment in mouse macrophages. We here studied the role of RNase T2 in TLR3 and TLR7 responses in macrophages. Macrophages expressed RNase T2 and a member of the RNase A family RNase 4. RNase T2 was also expressed in plasmacytoid and conventional dendritic cells. Treatment with dsRNAs or type I interferon (IFN) up-regulated expression of RNase T2 but not RNase 4. RNase T2-deficiency in macrophages up-regulated TLR3 responses but impaired TLR7 responses. Mechanistically, RNase T2 degraded both dsRNAs and ssRNAs in vitro, and its mutants showed a positive correlation between RNA degradation and the rescue of altered TLR3 and TLR7 responses. H122A and C188R RNase T2 mutations, not H69A and E118V mutations, impaired both RNA degradation and the rescue of altered TLR3 and TLR7 responses. RNase T2 in bone marrow-derived macrophages was broadly distributed from early endosomes to lysosomes, and colocalized with the internalized TLR3 ligand poly(I:C). These results suggest that RNase T2-dependent RNA degradation in endosomes/lysosomes negatively and positively regulates TLR3 and TLR7 responses, respectively, in macrophages.


Assuntos
Endorribonucleases/metabolismo , Endossomos/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , RNA de Cadeia Dupla/metabolismo , Receptor 3 Toll-Like/metabolismo , Receptor 7 Toll-Like/metabolismo , Animais , Linhagem Celular , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células HEK293 , Humanos , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
4.
Proc Natl Acad Sci U S A ; 116(8): 3118-3125, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30718396

RESUMO

Although climate change may expand the geographical distribution of several vector-borne diseases, the effects of environmental temperature in host defense to viral infection in vivo are unknown. Here, we demonstrate that exposure of mice to the high ambient temperature of 36 °C impaired adaptive immune responses against infection with viral pathogens, influenza, Zika, and severe fever with thrombocytopenia syndrome phlebovirus. Following influenza virus infection, the high heat-exposed mice failed to stimulate inflammasome-dependent cytokine secretion and respiratory dendritic cell migration to lymph nodes. Although commensal microbiota composition remained intact, the high heat-exposed mice decreased their food intake and increased autophagy in lung tissue. Induction of autophagy in room temperature-exposed mice severely impaired virus-specific CD8 T cells and antibody responses following respiratory influenza virus infection. In addition, we found that administration of glucose or dietary short-chain fatty acids restored influenza virus-specific adaptive immune responses in high heat-exposed mice. These findings uncover an unexpected mechanism by which ambient temperature and nutritional status control virus-specific adaptive immune responses.


Assuntos
Imunidade Adaptativa/imunologia , Vírus da Influenza A/imunologia , Influenza Humana/imunologia , Phlebovirus/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Temperatura Alta , Humanos , Inflamassomos/imunologia , Vírus da Influenza A/patogenicidade , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Pulmão/imunologia , Pulmão/virologia , Camundongos , Phlebovirus/patogenicidade , Zika virus/imunologia , Zika virus/patogenicidade , Infecção por Zika virus
5.
Proc Natl Acad Sci U S A ; 116(47): 23653-23661, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31694883

RESUMO

The activation of innate immune receptors by pathogen-associated molecular patterns (PAMPs) is central to host defense against infections. On the other hand, these receptors are also activated by immunogenic damage-associated molecular patterns (DAMPs), typically released from dying cells, and the activation can evoke chronic inflammatory or autoimmune disorders. One of the best known receptors involved in the immune pathogenesis is Toll-like receptor 7 (TLR7), which recognizes RNA with single-stranded structure. However, the causative DAMP RNA(s) in the pathogenesis has yet to be identified. Here, we first developed a chemical compound, termed KN69, that suppresses autoimmunity in several established mouse models. A subsequent search for KN69-binding partners led to the identification of U11 small nuclear RNA (U11snRNA) as a candidate DAMP RNA involved in TLR7-induced autoimmunity. We then showed that U11snRNA robustly activated the TLR7 pathway in vitro and induced arthritis disease in vivo. We also found a correlation between high serum level of U11snRNA and autoimmune diseases in human subjects and established mouse models. Finally, by revealing the structural basis for U11snRNA's ability to activate TLR7, we developed more potent TLR7 agonists and TLR7 antagonists, which may offer new therapeutic approaches for autoimmunity or other immune-driven diseases. Thus, our study has revealed a hitherto unknown immune function of U11snRNA, providing insight into TLR7-mediated autoimmunity and its potential for further therapeutic applications.


Assuntos
Glicoproteínas de Membrana/agonistas , RNA Nuclear Pequeno/imunologia , Receptor 7 Toll-Like/agonistas , Adulto , Alarminas/química , Animais , Artrite Reumatoide/sangue , Artrite Reumatoide/imunologia , Doenças Autoimunes/sangue , Doenças Autoimunes/imunologia , Sequência de Bases , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Imunossupressores/síntese química , Imunossupressores/farmacologia , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Pessoa de Meia-Idade , RNA/imunologia , RNA/metabolismo , Ribonucleoproteínas Nucleares Pequenas/química , Ribonucleoproteínas Nucleares Pequenas/imunologia , Análise de Sequência de RNA , Receptor 7 Toll-Like/deficiência , Adulto Jovem
6.
Int Immunol ; 31(3): 167-173, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30535046

RESUMO

Toll-like receptor 8 (TLR8), a sensor for pathogen-derived single-stranded RNA (ssRNA), binds to uridine (Uri) and ssRNA to induce defense responses. We here show that cytidine (Cyd) with ssRNA also activated TLR8 in peripheral blood leukocytes (PBLs) and a myeloid cell line U937, but not in an embryonic kidney cell line 293T. Cyd deaminase (CDA), an enzyme highly expressed in leukocytes, deaminates Cyd to Uri. CDA expression enabled TLR8 response to Cyd and ssRNA in 293T cells. CDA deficiency and a CDA inhibitor both reduced TLR8 responses to Cyd and ssRNA in U937. The CDA inhibitor also reduced PBL response to Cyd and ssRNA. A Cyd analogue, azacytidine, is used for the therapy of myelodysplastic syndrome and acute myeloid leukemia. Azacytidine with ssRNA induced tumor necrosis factor-α expression in U937 and PBLs in a manner dependent on CDA and TLR8. These results suggest that CDA enables TLR8 activation by Cyd or its analogues with ssRNA through deaminating activity. Nucleoside metabolism might impact TLR8 responses in a variety of situations such as the treatment with nucleoside analogues.


Assuntos
Citidina Desaminase/metabolismo , Citidina/análogos & derivados , Citidina/metabolismo , Receptor 8 Toll-Like/metabolismo , Citidina/química , Humanos , Monócitos/metabolismo , Monócitos/patologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Células Tumorais Cultivadas , Células U937
7.
J Virol ; 92(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30021900

RESUMO

The nonstructural protein (NSs) of severe fever with thrombocytopenia syndrome phlebovirus (SFTSV) sequesters TANK-binding kinase 1 (TBK1) into NSs-induced cytoplasmic structures to inhibit the phosphorylation and nuclear translocation of interferon (IFN) regulatory factor 3 (IRF3) and subsequent interferon beta (IFN-ß) production. Although the C-terminal region of SFTSV NSs (NSs66-249) has been linked to the formation of NSs-induced cytoplasmic structures and inhibition of host IFN-ß responses, the role of the N-terminal region in antagonizing host antiviral responses remains to be defined. Here, we demonstrate that two conserved amino acids at positions 21 and 23 in the SFTSV and heartland virus (HRTV) NSs are essential for suppression of IRF3 phosphorylation and IFN-ß mRNA expression following infection with SFTSV or recombinant influenza virus lacking the NS1 gene. Surprisingly, formation of SFTSV/HRTV NSs-induced cytoplasmic structures is not essential for inhibition of host antiviral responses. Rather, an association between SFTSV/HRTV NSs and TBK1 is required for suppression of mitochondrial antiviral signaling protein (MAVS)-mediated activation of IFN-ß promoter activity. Although SFTSV NSs did not prevent the ubiquitination of TBK1, it associates with TBK1 through its N-terminal kinase domain (residues 1 to 307) to block the autophosphorylation of TBK1. Furthermore, we found that both wild-type NSs and the 21/23A mutant (NSs in which residues at positions 21 and 23 were replaced with alanine) of SFTSV suppressed NLRP3 inflammasome-dependent interleukin-1ß (IL-1ß) secretion, suggesting that the importance of these residues is restricted to TBK1-dependent IFN signaling. Together, our findings strongly implicate the two conserved amino acids at positions 21 and 23 of SFTSV/HRTV NSs in the inhibition of host interferon responses.IMPORTANCE Recognition of viruses by host innate immune systems plays a critical role not only in providing resistance to viral infection but also in the initiation of antigen-specific adaptive immune responses against viruses. Severe fever with thrombocytopenia syndrome (SFTS) is a newly emerging infectious disease caused by the SFTS phlebovirus (SFTSV), a highly pathogenic tick-borne phlebovirus. The 294-amino-acid nonstructural protein (NSs) of SFTSV associates with TANK-binding kinase 1 (TBK1), a key regulator of host innate antiviral immunity, to inhibit interferon beta (IFN-ß) production and enhance viral replication. Here, we demonstrate that two conserved amino acids at positions 21 and 23 in the NSs of SFTSV and heartland virus, another tick-borne phlebovirus, are essential for association with TBK1 and suppression of IFN-ß production. Our results provide important insight into the molecular mechanisms by which SFTSV NSs helps to counteract host antiviral strategies.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Fator Regulador 3 de Interferon/imunologia , Interferon beta/imunologia , Phlebovirus/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Proteínas não Estruturais Virais/imunologia , Sequência de Aminoácidos , Sequência Conservada , Regulação da Expressão Gênica , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Fator Regulador 3 de Interferon/genética , Interferon beta/antagonistas & inibidores , Interferon beta/genética , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Febre por Flebótomos/genética , Febre por Flebótomos/imunologia , Febre por Flebótomos/patologia , Febre por Flebótomos/virologia , Phlebovirus/patogenicidade , Fosforilação , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Alinhamento de Sequência , Índice de Gravidade de Doença , Transdução de Sinais , Ubiquitinação , Proteínas não Estruturais Virais/genética , Vírus não Classificados/imunologia , Vírus não Classificados/patogenicidade
8.
Biochem Biophys Res Commun ; 495(1): 353-359, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29108997

RESUMO

Fatty acid-binding protein 4 (FABP4), a cytosolic lipid chaperone predominantly expressed in adipocytes and macrophages, modulates lipid fluxes, trafficking, signaling, and metabolism. Recent studies have demonstrated that FABP4 regulates metabolic and inflammatory pathways, and in mouse models its inhibition can improve type 2 diabetes mellitus and atherosclerosis. However, the role of FABP4 in bacterial infection, metabolic crosstalk between host and pathogen, and bacterial pathogenesis have not been studied. As an obligate intracellular pathogen, Chlamydia pneumoniae needs to obtain nutrients such as ATP and lipids from host cells. Here, we show that C. pneumoniae successfully infects and proliferates in murine adipocytes by inducing hormone sensitive lipase (HSL)-mediated lipolysis. Chemical inhibition or genetic manipulation of HSL significantly abrogated the intracellular growth of C. pneumoniae in adipocytes. Liberated free fatty acids were utilized to generate ATP via ß-oxidation, which C. pneumoniae usurped for its replication. Strikingly, chemical inhibition or genetic silencing of FABP4 significantly abrogated C. pneumoniae infection-induced lipolysis and mobilization of liberated FFAs, resulting in reduced bacterial growth in adipocytes. Collectively, these results demonstrate that C. pneumoniae exploits host FABP4 to facilitate fat mobilization and intracellular replication in adipocytes. This work uncovers a novel strategy used by intracellular pathogens for acquiring energy via hijacking of the host lipid metabolism pathway.


Assuntos
Adipócitos/microbiologia , Adipócitos/fisiologia , Chlamydophila pneumoniae/fisiologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Mobilização Lipídica/fisiologia , Esterol Esterase/metabolismo , Células 3T3-L1 , Animais , Proliferação de Células/fisiologia , Chlamydophila pneumoniae/citologia , Camundongos
9.
J Virol ; 90(8): 4105-4114, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26865721

RESUMO

UNLABELLED: Inflammasomes are cytosolic multimolecular protein complexes that stimulate the activation of caspase-1 and the release of mature forms of interleukin-1ß (IL-1ß) and IL-18. We previously demonstrated that the influenza A virus M2 protein stimulates IL-1ß secretion following activation of the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. The nonstructural protein 1 (NS1) of influenza virus inhibits caspase-1 activation and IL-1ß secretion. However, the precise mechanism by which NS1 inhibits IL-1ß secretion remains unknown. Here, we showed that J774A.1 macrophages stably expressing the NS1 protein inhibited IL-1ß secretion after infection with recombinant influenza virus lacking the NS1 gene. Coimmunoprecipitation assay revealed that the NS1 protein interacts with NLRP3. Importantly, the NS1 protein inhibited the NLRP3/ASC-induced single-speck formation required for full activation of inflammasomes. The NS1 protein of other influenza virus strains, including a recent pandemic strain, also inhibited inflammasome-mediated IL-1ß secretion. The NS1 RNA-binding domain (basic residues 38 and 41) and TRIM25-binding domain (acidic residues 96 and 97) were required for suppression of NLRP3 inflammasome-mediated IL-1ß secretion. These results shed light on a mechanism by which the NS1 protein of influenza virus suppresses NLRP3 inflammasome-mediated IL-1ß secretion. IMPORTANCE: Innate immune sensing of influenza virus via pattern recognition receptors not only plays a key role in generating type I interferons but also triggers inflammatory responses. We previously demonstrated that the influenza A virus M2 protein activates the NLRP3 inflammasome, leading to the secretion of interleukin-1ß (IL-1ß) and IL-18 following the activation of caspase-1. Although the nonstructural protein 1 (NS1) of influenza virus inhibits IL-1ß secretion, the precise mechanism by which it achieves this remains to be defined. Here, we demonstrate that the NS1 protein interacts with NLRP3 to suppress NLRP3 inflammasome activation. J774A.1 macrophages stably expressing the NS1 protein suppressed NLRP3-mediated IL-1ß secretion. The NS1 RNA-binding domain (basic residues 38 and 41) and TRIM25-binding domain (acidic residues 96 and 97) are important for suppression of NLRP3 inflammasome-mediated IL-1ß secretion. These results will facilitate the development of new anti-inflammatory drugs.


Assuntos
Proteínas de Transporte/metabolismo , Inflamassomos/imunologia , Interleucina-1beta/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Sítios de Ligação , Proteínas de Transporte/antagonistas & inibidores , Células HEK293 , Células HeLa , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Membranas Mitocondriais/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , RNA/metabolismo , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/metabolismo
10.
Proc Natl Acad Sci U S A ; 110(44): 17963-8, 2013 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-24127597

RESUMO

Nod-like receptor family, pyrin domain-containing 3 (NLRP3), is involved in the early stages of the inflammatory response by sensing cellular damage or distress due to viral or bacterial infection. Activation of NLRP3 triggers its assembly into a multimolecular protein complex, termed "NLRP3 inflammasome." This event leads to the activation of the downstream molecule caspase-1 that cleaves the precursor forms of proinflammatory cytokines, such as interleukin 1 beta (IL-1ß) and IL-18, and initiates the immune response. Recent studies indicate that the reactive oxygen species produced by mitochondrial respiration is critical for the activation of the NLRP3 inflammasome by monosodium urate, alum, and ATP. However, the precise mechanism by which RNA viruses activate the NLRP3 inflammasome is not well understood. Here, we show that loss of mitochondrial membrane potential [ΔΨ(m)] dramatically reduced IL-1ß secretion after infection with influenza, measles, or encephalomyocarditis virus (EMCV). Reduced IL-1ß secretion was also observed following overexpression of the mitochondrial inner membrane protein, uncoupling protein-2, which induces mitochondrial proton leakage and dissipates ΔΨ(m). ΔΨ(m) was required for association between the NLRP3 and mitofusin 2, a mediator of mitochondrial fusion, after infection with influenza virus or EMCV. Importantly, the knockdown of mitofusin 2 significantly reduced the secretion of IL-1ß after infection with influenza virus or EMCV. Our results provide insight into the roles of mitochondria in NLRP3 inflammasome activation.


Assuntos
Proteínas de Transporte/imunologia , GTP Fosfo-Hidrolases/imunologia , Inflamassomos/imunologia , Infecções por Vírus de RNA/imunologia , Análise de Variância , Animais , Interleucina-1beta/imunologia , Canais Iônicos/metabolismo , Potencial da Membrana Mitocondrial/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Espécies Reativas de Oxigênio/metabolismo , Proteína Desacopladora 2
11.
Uirusu ; 65(1): 127-34, 2015.
Artigo em Japonês | MEDLINE | ID: mdl-26923967

RESUMO

Understanding the mechanisms by which influenza viruses are recognized by the innate immune system to elicit a protective adaptive immune response is essential for the development of effective vaccines. We have demonstrated that synthetic double-stranded RNA poly(I:C) is an effective adjuvant for intranasal influenza vaccine. Furthermore, we found that influenza virus activated the NLR family, pyrin domain-containing 3 (NLRP3) inflammasome via its M2 protein. Inflammasome activation in the lung coupled with priming signals from the commensal microbiota in the gut are essential for the generation of influenza virus-specific adaptive immune responses. These results provide a useful basis for developing effective vaccines against influenza viruses.


Assuntos
Imunidade Adaptativa/imunologia , Descoberta de Drogas , Imunidade Inata/imunologia , Vacinas contra Influenza , Orthomyxoviridae/imunologia , Adjuvantes Imunológicos , Administração Intranasal , Animais , Humanos , Inflamassomos , Vacinas contra Influenza/administração & dosagem , Macaca fascicularis , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Orthomyxoviridae/genética , Poli I-C , Polinucleotídeos , RNA de Cadeia Dupla , Proteínas da Matriz Viral
12.
PLoS Pathog ; 8(8): e1002857, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22916014

RESUMO

Nod-like receptors (NLRs) comprise a large family of intracellular pattern- recognition receptors. Members of the NLR family assemble into large multiprotein complexes, termed the inflammasomes. The NLR family, pyrin domain-containing 3 (NLRP3) is triggered by a diverse set of molecules and signals, and forms the NLRP3 inflammasome. Recent studies have indicated that both DNA and RNA viruses stimulate the NLRP3 inflammasome, leading to the secretion of interleukin 1 beta (IL-1ß) and IL-18 following the activation of caspase-1. We previously demonstrated that the proton-selective ion channel M2 protein of influenza virus activates the NLRP3 inflammasome. However, the precise mechanism by which NLRP3 recognizes viral infections remains to be defined. Here, we demonstrate that encephalomyocarditis virus (EMCV), a positive strand RNA virus of the family Picornaviridae, activates the NLRP3 inflammasome in mouse dendritic cells and macrophages. Although transfection with RNA from EMCV virions or EMCV-infected cells induced robust expression of type I interferons in macrophages, it failed to stimulate secretion of IL-1ß. Instead, the EMCV viroporin 2B was sufficient to cause inflammasome activation in lipopolysaccharide-primed macrophages. While cells untransfected or transfected with the gene encoding the EMCV non-structural protein 2A or 2C expressed NLRP3 uniformly throughout the cytoplasm, NLRP3 was redistributed to the perinuclear space in cells transfected with the gene encoding the EMCV 2B or influenza virus M2 protein. 2B proteins of other picornaviruses, poliovirus and enterovirus 71, also caused the NLRP3 redistribution. Elevation of the intracellular Ca(2+) level, but not mitochondrial reactive oxygen species and lysosomal cathepsin B, was important in EMCV-induced NLRP3 inflammasome activation. Chelation of extracellular Ca(2+) did not reduce virus-induced IL-1ß secretion. These results indicate that EMCV activates the NLRP3 inflammasome by stimulating Ca(2+) flux from intracellular storages to the cytosol, and highlight the importance of viroporins, transmembrane pore-forming viral proteins, in virus-induced NLRP3 inflammasome activation.


Assuntos
Cálcio/metabolismo , Infecções por Cardiovirus/metabolismo , Proteínas de Transporte/metabolismo , Vírus da Encefalomiocardite/metabolismo , Inflamassomos/metabolismo , Proteínas Virais/metabolismo , Animais , Infecções por Cardiovirus/genética , Proteínas de Transporte/genética , Vírus da Encefalomiocardite/genética , Células HEK293 , Células HeLa , Humanos , Inflamassomos/genética , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Virais/genética , Vírion/genética , Vírion/metabolismo
13.
Proc Natl Acad Sci U S A ; 108(13): 5354-9, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21402903

RESUMO

Although commensal bacteria are crucial in maintaining immune homeostasis of the intestine, the role of commensal bacteria in immune responses at other mucosal surfaces remains less clear. Here, we show that commensal microbiota composition critically regulates the generation of virus-specific CD4 and CD8 T cells and antibody responses following respiratory influenza virus infection. By using various antibiotic treatments, we found that neomycin-sensitive bacteria are associated with the induction of productive immune responses in the lung. Local or distal injection of Toll-like receptor (TLR) ligands could rescue the immune impairment in the antibiotic-treated mice. Intact microbiota provided signals leading to the expression of mRNA for pro-IL-1ß and pro-IL-18 at steady state. Following influenza virus infection, inflammasome activation led to migration of dendritic cells (DCs) from the lung to the draining lymph node and T-cell priming. Our results reveal the importance of commensal microbiota in regulating immunity in the respiratory mucosa through the proper activation of inflammasomes.


Assuntos
Imunidade Adaptativa/imunologia , Vírus da Influenza A/imunologia , Metagenoma , Infecções Respiratórias/imunologia , Infecções Respiratórias/microbiologia , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bactérias/efeitos dos fármacos , Bactérias/imunologia , Bactérias/patogenicidade , Células Dendríticas/imunologia , Homeostase , Interações Hospedeiro-Patógeno , Humanos , Inflamassomos , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/virologia , Receptores Toll-Like/imunologia
14.
NPJ Sci Food ; 8(1): 18, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38485724

RESUMO

Current treatment options for COVID-19 are limited, with many antivirals and immunomodulators restricted to the most severe cases and preventative care limited to vaccination. As the SARS-CoV-2 virus and its increasing variants threaten to become a permanent fixture of our lives, this new reality necessitates the development of cost-effective and accessible treatment options for COVID-19. Studies have shown that there are correlations between the gut microbiome and severity of COVID-19, especially with regards to production of physiologically beneficial short-chain fatty acids (SCFAs) by gut microbes. In this study, we used a Syrian hamster model to study how dietary consumption of the prebiotic inulin affected morbidity and mortality resulting from SARS-CoV-2 infection. After two weeks of observation, we discovered that inulin supplementation attenuated morbid weight loss and increased survival rate in hamster subjects. An analysis of microbiome community structure showed significant alterations in 15 genera. Notably, there were also small increases in fecal DCA and a significant increase in serum DCA, perhaps highlighting a role for this secondary bile acid in conferring protection against SARS-CoV-2. In light of these results, inulin and other prebiotics are promising targets for future investigation as preventative treatment options for COVID-19.

15.
Nat Med ; 12(4): 466-72, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16550188

RESUMO

Adult T-cell leukemia-lymphoma (ATLL) is a group of T-cell malignancies caused by infection with human T-lymphotropic virus type I (HTLV-I). Although the pathogenesis of ATLL remains incompletely understood, the viral regulatory protein Tax is centrally involved in cellular transformation. Here we describe the generation of HTLV-I Tax transgenic mice using the Lck proximal promoter to restrict transgene expression to developing thymocytes. After prolonged latency periods, transgenic mice developed diffuse large-cell lymphomas and leukemia with clinical, pathological and immunological features characteristic of acute ATLL. Transgenic mice were functionally immunocompromised and they developed opportunistic infections. Fulminant disease also developed rapidly in SCID mice after engraftment of lymphomatous cells from transgenic mice. Flow cytometry showed that the cells were CD4(-) and CD8(-), but CD44(+), CD25(+) and cytoplasmic CD3(+). This phenotype is indicative of a thymus-derived pre-T-cell phenotype, and disease development was associated with the constitutive activation of NF-kappaB. Our model accurately reproduces human disease and will provide a tool for analysis of the molecular events in transformation and for the development of new therapeutics.


Assuntos
Genes pX , Vírus Linfotrópico T Tipo 1 Humano/genética , Leucemia Linfoide/patologia , Neoplasias do Timo/patologia , Animais , Biomarcadores , Complexo CD3/imunologia , Complexo CD3/metabolismo , Mapeamento Cromossômico , Cromossomos , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Vírus Linfotrópico T Tipo 1 Humano/ultraestrutura , Humanos , Imuno-Histoquímica , Leucemia Linfoide/genética , Leucemia Linfoide/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Transplante de Neoplasias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias do Timo/imunologia , Transgenes , Transplante Homólogo
16.
Sci Rep ; 13(1): 4033, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36899059

RESUMO

In order to reduce infection risk of novel coronavirus (SARS-CoV-2), we developed nano-photocatalysts with nanoscale rutile TiO2 (4-8 nm) and CuxO (1-2 nm or less). Their extraordinarily small size leads to high dispersity and good optical transparency, besides large active surface area. Those photocatalysts can be applied to white and translucent latex paints. Although Cu2O clusters involved in the paint coating undergo gradual aerobic oxidation in the dark, the oxidized clusters are re-reduced under > 380 nm light. The paint coating inactivated the original and alpha variant of novel coronavirus under irradiation with fluorescent light for 3 h. The photocatalysts greatly suppressed binding ability of the receptor binding domain (RBD) of coronavirus (the original, alpha and delta variants) spike protein to the receptor of human cells. The coating also exhibited antivirus effects on influenza A virus, feline calicivirus, bacteriophage Qß and bacteriophage M13. The photocatalysts would be applied to practical coatings and lower the risk of coronavirus infection via solid surfaces.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Desnaturação Proteica , Glicoproteína da Espícula de Coronavírus/metabolismo
17.
Nat Commun ; 14(1): 3863, 2023 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-37391427

RESUMO

Fever is a common symptom of influenza and coronavirus disease 2019 (COVID-19), yet its physiological role in host resistance to viral infection remains less clear. Here, we demonstrate that exposure of mice to the high ambient temperature of 36 °C increases host resistance to viral pathogens including influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). High heat-exposed mice increase basal body temperature over 38 °C to enable more bile acids production in a gut microbiota-dependent manner. The gut microbiota-derived deoxycholic acid (DCA) and its plasma membrane-bound receptor Takeda G-protein-coupled receptor 5 (TGR5) signaling increase host resistance to influenza virus infection by suppressing virus replication and neutrophil-dependent tissue damage. Furthermore, the DCA and its nuclear farnesoid X receptor (FXR) agonist protect Syrian hamsters from lethal SARS-CoV-2 infection. Moreover, we demonstrate that certain bile acids are reduced in the plasma of COVID-19 patients who develop moderate I/II disease compared with the minor severity of illness group. These findings implicate a mechanism by which virus-induced high fever increases host resistance to influenza virus and SARS-CoV-2 in a gut microbiota-dependent manner.


Assuntos
COVID-19 , Microbioma Gastrointestinal , Vírus da Influenza A , Influenza Humana , Cricetinae , Animais , Camundongos , Humanos , SARS-CoV-2 , Temperatura Corporal , Febre , Ácidos e Sais Biliares , Mesocricetus
18.
J Exp Med ; 220(9)2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37462944

RESUMO

Loss-of-function mutations in the lysosomal nucleoside transporter SLC29A3 cause lysosomal nucleoside storage and histiocytosis: phagocyte accumulation in multiple organs. However, little is known about the mechanism by which lysosomal nucleoside storage drives histiocytosis. Herein, histiocytosis in Slc29a3-/- mice was shown to depend on Toll-like receptor 7 (TLR7), which senses a combination of nucleosides and oligoribonucleotides (ORNs). TLR7 increased phagocyte numbers by driving the proliferation of Ly6Chi immature monocytes and their maturation into Ly6Clow phagocytes in Slc29a3-/- mice. Downstream of TLR7, FcRγ and DAP10 were required for monocyte proliferation. Histiocytosis is accompanied by inflammation in SLC29A3 disorders. However, TLR7 in nucleoside-laden splenic monocytes failed to activate inflammatory responses. Enhanced production of proinflammatory cytokines was observed only after stimulation with ssRNAs, which would increase lysosomal ORNs. Patient-derived monocytes harboring the G208R SLC29A3 mutation showed enhanced survival and proliferation in a TLR8-antagonist-sensitive manner. These results demonstrated that TLR7/8 responses to lysosomal nucleoside stress drive SLC29A3 disorders.


Assuntos
Histiocitose , Receptor 7 Toll-Like , Animais , Camundongos , Citocinas/genética , Histiocitose/genética , Mutação/genética , Nucleosídeos , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética
19.
J Virol ; 85(24): 13019-26, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21994456

RESUMO

Inflammasomes are cytosolic protein complexes that stimulate the activation of caspase-1, which in turn induces the secretion of the inflammatory cytokines Interleukin-1ß (IL-1ß) and IL-18. Recent studies have indicated that the inflammasome known as the NOD-like-receptor-family, pyrin domain-containing 3 (NLRP3) inflammasome recognizes several RNA viruses, including the influenza and encephalomyocarditis viruses, whereas the retinoic acid-inducible gene I (RIG-I) inflammasome may detect vesicular stomatitis virus. We demonstrate that measles virus (MV) infection induces caspase-1-dependent IL-1ß secretion in the human macrophage-like cell line THP-1. Gene knockdown experiments indicated that IL-1ß secretion in MV-infected THP-1 cells was mediated by the NLRP3 inflammasome but not the RIG-I inflammasome. MV produces the nonstructural V protein, which has been shown to antagonize host innate immune responses. The recombinant MV lacking the V protein induced more IL-1ß than the parental virus. THP-1 cells stably expressing the V protein suppressed NLRP3 inflammasome-mediated IL-1ß secretion. Furthermore, coimmunoprecipitation assays revealed that the V protein interacts with NLRP3 through its carboxyl-terminal domain. NLRP3 was located in cytoplasmic granular structures in THP-1 cells stably expressing the V protein, but upon inflammasome activation, NLRP3 was redistributed to the perinuclear region, where it colocalized with the V protein. These results indicate that the V protein of MV suppresses NLRP3 inflammasome-mediated IL-1ß secretion by directly or indirectly interacting with NLRP3.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Evasão da Resposta Imune , Inflamassomos/imunologia , Interleucina-1beta/metabolismo , Vírus do Sarampo/imunologia , Vírus do Sarampo/patogenicidade , Fosfoproteínas/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ligação Proteica , Mapeamento de Interação de Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA