Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1173605, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37435074

RESUMO

Human metapneumovirus (HMPV) is a pneumovirus that may cause severe respiratory disease in humans. HMPV infection has been found to increase susceptibility to bacterial superinfections leading to increased morbidity and mortality. The molecular mechanisms underlying HMPV-mediated increase in bacterial susceptibility are poorly understood and largely understudied. Type I interferons (IFNs), while critical for antiviral defenses, may often have detrimental effects by skewing the host immune response and cytokine output of immune cells. It is currently unknown if HMPV skews the inflammatory response in human macrophages triggered by bacterial stimuli. Here we report that HMPV pre-infection impacts production of specific cytokines. HMPV strongly suppresses IL-1ß transcription in response to LPS or heat-killed Pseudomonas aeruginosa and Streptococcus pneumonia, while enhancing mRNA levels of IL-6, TNF-α and IFN-ß. We demonstrate that in human macrophages the HMPV-mediated suppression of IL-1ß transcription requires TANK-binding kinase 1 (TBK1) and signaling via the IFN-ß-IFNAR axis. Interestingly, our results show that HMPV pre-infection did not impair the LPS-stimulated activation of NF-κB and HIF-1α, transcription factors that stimulate IL-1ß mRNA synthesis in human cells. Furthermore, we determined that sequential HMPV-LPS treatment resulted in accumulation of the repressive epigenetic mark H3K27me3 at the IL1B promoter. Thus, for the first time we present data revealing the molecular mechanisms by which HMPV shapes the cytokine output of human macrophages exposed to bacterial pathogens/LPS, which appears to be dependent on epigenetic reprogramming at the IL1B promoter leading to reduced synthesis of IL-1ß. These results may improve current understanding of the role of type I IFNs in respiratory disease mediated not only by HMPV, but also by other respiratory viruses that are associated with superinfections.


Assuntos
Infecções Bacterianas , Interferon beta , Interleucina-1beta , Infecções por Paramyxoviridae , Superinfecção , Humanos , Citocinas , Metapneumovirus , Transcrição Gênica , Infecções Bacterianas/imunologia , Infecções por Paramyxoviridae/imunologia
2.
J Virol ; 95(3)2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33208442

RESUMO

Hepatitis C virus (HCV) infection triggers Golgi fragmentation through the Golgi-resident protein immunity-related GTPase M (IRGM). Here, we report the roles of NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) and ASC (apoptosis-associated speck-like protein containing a caspase activation and recruitment domain [CARD]), two inflammasome components, in the initial events leading to this fragmentation. We show that ASC resides at the Golgi with IRGM at homeostasis. Upon infection, ASC dissociates from both IRGM and the Golgi and associates with HCV-induced NLRP3. NLRP3 silencing inhibits Golgi fragmentation. ASC silencing disrupts the Golgi structure in both control and infected cells and reduces the localization of IRGM at the Golgi. IRGM depletion in the ASC-silenced cells cannot totally restore the Golgi structure. These data highlight a role for ASC, upstream of the formation of the inflammasome, in regulating IRGM through its control on the Golgi. A similar mechanism occurs in response to nigericin treatment, but not in cells infected with another member of the Flaviviridae family, Zika virus (ZIKV). We propose a model for a newly ascribed function of the inflammasome components in Golgi structural remodeling during certain stimuli.IMPORTANCE Numerous pathogens can affect cellular homeostasis and organelle dynamics. Hepatitis C virus (HCV) triggers Golgi fragmentation through the immunity-related GTPase M (IRGM), a resident Golgi protein, to enhance its lipid supply for replication. Here, we reveal the role of the inflammasome components NLRP3 and ASC in this process, thus uncovering a new interplay between effectors of inflammation and viral infection or stress. We show that the inflammasome component ASC resides at the Golgi under homeostasis and associates with IRGM. Upon HCV infection, ASC is recruited to NLRP3 and dissociates from IRGM, causing Golgi fragmentation. Our results uncover that aside from their known function in the inflammation response, these host defense regulators also ensure the maintenance of intact intracellular structure in homeostasis, while their activation relieves factors leading to Golgi remodeling.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Complexo de Golgi/fisiologia , Hepacivirus/isolamento & purificação , Hepatite C/virologia , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Apoptose , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas de Ligação ao GTP/genética , Complexo de Golgi/virologia , Hepatite C/metabolismo , Hepatite C/patologia , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética
3.
Mediators Inflamm ; 2019: 4964239, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31686982

RESUMO

Human metapneumovirus (HMPV) may cause severe respiratory disease. The early innate immune response to viruses like HMPV is characterized by induction of antiviral interferons (IFNs) and proinflammatory immune mediators that are essential in shaping adaptive immune responses. Although innate immune responses to HMPV have been comprehensively studied in mice and murine immune cells, there is less information on these responses in human cells, comparing different cell types infected with the same HMPV strain. The aim of this study was to characterize the HMPV-induced mRNA expression of critical innate immune mediators in human primary cells relevant for airway disease. In particular, we determined type I versus type III IFN expression in human epithelial cells and monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs). In epithelial cells, HMPV induced only low levels of IFN-ß mRNA, while a robust mRNA expression of IFN-λs was found in epithelial cells, MDMs, and MDDCs. In addition, we determined induction of the interferon regulatory factors (IRFs) IRF1, IRF3, and IRF7 and critical inflammatory cytokines (IL-6, IP-10, and IL-1ß). Interestingly, IRF1 mRNA was predominantly induced in MDMs and MDDCs. Overall, our results suggest that for HMPV infection of MDDCs, MDMs, NECs, and A549 cells (the cell types examined), cell type is a strong determinator of the ability of HMPV to induce different innate immune mediators. HMPV induces the transcription of IFN-ß and IRF1 to higher extents in MDMs and MDDCs than in A549s and NECs, whereas the induction of type III IFN-λ and IRF7 is considerable in MDMs, MDDCs, and A549 epithelial cells.


Assuntos
Imunidade Inata/fisiologia , Metapneumovirus/patogenicidade , Infecções por Paramyxoviridae/imunologia , Células A549 , Células Cultivadas , Quimiocina CXCL10/metabolismo , Imunofluorescência , Humanos , Imunidade Inata/genética , Interleucina-6/metabolismo , Macrófagos/metabolismo , Metapneumovirus/imunologia , Microscopia Confocal , Infecções por Paramyxoviridae/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais
4.
Viruses ; 11(10)2019 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-31635418

RESUMO

Viruses are the major causes of acute and chronic infectious diseases in the world. According to the World Health Organization, there is an urgent need for better control of viral diseases. Repurposing existing antiviral agents from one viral disease to another could play a pivotal role in this process. Here, we identified novel activities of obatoclax and emetine against herpes simplex virus type 2 (HSV-2), echovirus 1 (EV1), human metapneumovirus (HMPV) and Rift Valley fever virus (RVFV) in cell cultures. Moreover, we demonstrated novel activities of emetine against influenza A virus (FLUAV), niclosamide against HSV-2, brequinar against human immunodeficiency virus 1 (HIV-1), and homoharringtonine against EV1. Our findings may expand the spectrum of indications of these safe-in-man agents and reinforce the arsenal of available antiviral therapeutics pending the results of further in vitro and in vivo tests.


Assuntos
Antivirais/farmacologia , Vírus/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Compostos de Bifenilo/farmacologia , Técnicas de Cultura de Células , Chlorocebus aethiops , Cães , Emetina/farmacologia , Enterovirus Humano B/efeitos dos fármacos , HIV-1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Mepesuccinato de Omacetaxina/farmacologia , Humanos , Indóis , Células Madin Darby de Rim Canino , Niclosamida/farmacologia , Pirróis/farmacologia , Células Vero , Viroses/tratamento farmacológico , Vírus/classificação
5.
Front Immunol ; 10: 2186, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31636628

RESUMO

Viruses are one of the major causes of acute and chronic infectious diseases and thus a major contributor to the global burden of disease. Several studies have shown how viruses have evolved to hijack basic cellular pathways and evade innate immune response by modulating key host factors and signaling pathways. A collective view of these multiple studies could advance our understanding of virus-host interactions and provide new therapeutic perspectives for the treatment of viral diseases. Here, we performed an integrative meta-analysis to elucidate the 17 different host-virus interactomes. Network and bioinformatics analyses showed how viruses with small genomes efficiently achieve the maximal effect by targeting multifunctional and highly connected host proteins with a high occurrence of disordered regions. We also identified the core cellular process subnetworks that are targeted by all the viruses. Integration with functional RNA interference (RNAi) datasets showed that a large proportion of the targets are required for viral replication. Furthermore, we performed an interactome-informed drug re-purposing screen and identified novel activities for broad-spectrum antiviral agents against hepatitis C virus and human metapneumovirus. Altogether, these orthogonal datasets could serve as a platform for hypothesis generation and follow-up studies to broaden our understanding of the viral evasion landscape.


Assuntos
Interações entre Hospedeiro e Microrganismos , Mapas de Interação de Proteínas , Viroses/imunologia , Complexo I de Proteína do Envoltório/fisiologia , Biologia Computacional , Humanos , Evasão da Resposta Imune , Transdução de Sinais/fisiologia , Viroses/tratamento farmacológico , Replicação Viral
6.
Genome Announc ; 6(21)2018 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-29798923

RESUMO

Here, we report the complete genome sequences of human metapneumovirus (HMPV) prior to and after passaging in LLC-MK2 cells. Paired comparisons of the 13,335-nucleotide genomes revealed that the virus acquired the T10736C transition in its genome, which did not affect the amino acid sequences of HMPV proteins.

7.
Sci Rep ; 8(1): 883, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29343779

RESUMO

Thymic stromal lymphopoietin (TSLP) is associated with several allergic diseases including asthma. Two isoforms of TSLP exist in humans, a long form (lfTSLP) and a short form (sfTSLP), displaying distinct immunological functions. Recently, TSLP was found to be upregulated in human airway cells upon human metapneumovirus (hMPV) infection, yet it remains unclear if the two isoforms are regulated differently during hMPV infection. Importantly, the molecular mechanisms underlying hMPV-mediated TSLP induction remain undescribed. In this study, we characterized the expression and regulation of TSLP in hMPV-infected human airway cells. We demonstrated that hMPV strongly induced the expression of pro-inflammatory lfTSLP in human airway epithelial cells and lung fibroblasts. Further, knockdown of pattern recognition receptors retinoic acid-inducible gene I (RIG-I) or Toll-like receptor 3 (TLR3), as well as downstream signal transducers, abrogated hMPV-mediated lfTSLP induction. Importantly, silencing of TANK-binding kinase 1 (TBK1) also impaired hMPV-mediated lfTSLP induction, which could be attributed to compromised NF-κB activation. Overall, these results suggest that TBK1 may be instrumental for hMPV-mediated activation of NF-κB downstream RIG-I and TLR3, leading to a specific induction of lfTSLP in hMPV-infected human airway cells.


Assuntos
Citocinas/genética , Regulação da Expressão Gênica/genética , Inflamação/genética , Metapneumovirus/patogenicidade , Infecções por Paramyxoviridae/genética , Transdução de Sinais/genética , Células A549 , Asma/genética , Asma/virologia , Linhagem Celular Tumoral , Proteína DEAD-box 58/genética , Células Epiteliais/virologia , Humanos , Hipersensibilidade/genética , Hipersensibilidade/virologia , Inflamação/virologia , NF-kappa B/genética , Infecções por Paramyxoviridae/virologia , Proteínas Serina-Treonina Quinases/genética , Receptor 3 Toll-Like/genética , Linfopoietina do Estroma do Timo
8.
Proc Natl Acad Sci U S A ; 114(17): E3462-E3471, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28389568

RESUMO

Positive-stranded RNA viruses, such as hepatitis C virus (HCV), assemble their viral replication complexes by remodeling host intracellular membranes to a membranous web. The precise composition of these replication complexes and the detailed mechanisms by which they are formed are incompletely understood. Here we show that the human immunity-related GTPase M (IRGM), known to contribute to autophagy, plays a previously unrecognized role in this process. We show that IRGM is localized at the Golgi apparatus and regulates the fragmentation of Golgi membranes in response to HCV infection, leading to colocalization of Golgi vesicles with replicating HCV. Our results show that IRGM controls phosphorylation of GBF1, a guanine nucleotide exchange factor for Arf-GTPases, which normally operates in Golgi membrane dynamics and vesicle coating in resting cells. We also find that HCV triggers IRGM-mediated phosphorylation of the early autophagy initiator ULK1, thereby providing mechanistic insight into the role of IRGM in HCV-mediated autophagy. Collectively, our results identify IRGM as a key Golgi-situated regulator that links intracellular membrane remodeling by autophagy and Golgi fragmentation with viral replication.


Assuntos
Autofagia , Proteínas de Ligação ao GTP/metabolismo , Complexo de Golgi/metabolismo , Hepacivirus/fisiologia , Membranas Intracelulares/metabolismo , Replicação Viral/fisiologia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação ao GTP/genética , Complexo de Golgi/genética , Complexo de Golgi/virologia , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Membranas Intracelulares/virologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosforilação/genética
9.
PLoS One ; 11(5): e0155484, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27171557

RESUMO

Human metapneumovirus (hMPV) causes severe airway infection in children that may be caused by an unfavorable immune response. The nature of the innate immune response to hMPV in naturally occurring infections in children is largely undescribed, and it is unknown if inflammasome activation is implicated in disease pathogenesis. We examined nasopharynx aspirates and blood samples from hMPV-infected children without detectable co-infections. The expression of inflammatory and antiviral genes were measured in nasal airway secretions by relative mRNA quantification while blood plasma proteins were determined by a multiplex immunoassay. Several genes were significantly up-regulated at mRNA and protein level in the hMPV infected children. Most apparent was the expression of the chemokine IP-10, the pro-inflammatory cytokine IL-18 in addition to the interferon inducible gene ISG54. Interestingly, children experiencing more severe disease, as indicated by a severity index, had significantly more often up-regulation of the inflammasome-associated genes IL-1ß and NLRP3. Overall, our data point to cytokines, particularly inflammasome-associated, that might be important in hMPV mediated lung disease and the antiviral response in children with severe infection. Our study is the first to demonstrate that inflammasome components are associated with increased illness severity in hMPV-infected children.


Assuntos
Antivirais/uso terapêutico , Citocinas/genética , Metapneumovirus/patogenicidade , Infecções por Paramyxoviridae/genética , Infecções por Paramyxoviridae/virologia , Antivirais/farmacologia , Estudos de Casos e Controles , Criança , Pré-Escolar , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genótipo , Humanos , Lactente , Inflamassomos/genética , Inflamassomos/metabolismo , Interferons/genética , Interferons/metabolismo , Masculino , Metapneumovirus/efeitos dos fármacos , Nasofaringe/patologia , Nasofaringe/virologia , Infecções por Paramyxoviridae/sangue , Infecções por Paramyxoviridae/tratamento farmacológico
10.
Cell Signal ; 25(9): 1804-12, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23707530

RESUMO

Antiviral responses can be triggered by the cytoplasmic RNA helicase RIG-I that binds to viral RNA. RIG-I-mediated signaling stimulates the transcription factors IRF3 and NF-κB and their activation mechanisms have been intensively studied. Here we examined Sendai virus (SV)-mediated activation of the transcription factor CREB and the role of its feedback repressor ICER in production of endogenous antiviral proteins. We show that SV infection and the mitochondrial adapter protein MAVS promote CREB phosphorylation that is dependent upon p38 MAPK and MK2. ICER is induced by CREB and acts as a feedback repressor of CRE-dependent transcription. We found that SV infection stimulated induction of ICER mRNA and protein expression. Surprisingly, ectopic expression and siRNA-mediated knockdown of ICER revealed that ICER is a positive regulator of the production of antiviral IFN-ß and IP10 during SV infection. In contrast, ICER did not affect SV-elicited phosphorylation of IRF3, NF-κB or ATF2/c-Jun, transcription factors governing IFN-ß and IP10 synthesis. However, expression of ICER increased total IRF3 protein levels during SV infection. These results point to a novel role of ICER in antiviral immune signaling acting to increase levels of antiviral effectors.


Assuntos
Modulador de Elemento de Resposta do AMP Cíclico/imunologia , RNA Helicases DEAD-box/imunologia , Interações Hospedeiro-Patógeno , Interferon beta/imunologia , Infecções por Respirovirus/imunologia , Vírus Sendai/fisiologia , Fator 2 Ativador da Transcrição/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , AMP Cíclico/imunologia , Modulador de Elemento de Resposta do AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/imunologia , Proteína DEAD-box 58 , Células HEK293 , Humanos , Fator Regulador 3 de Interferon/imunologia , Interferon beta/genética , RNA Mensageiro/genética , Receptores Imunológicos , Infecções por Respirovirus/genética , Ativação Transcricional , eIF-2 Quinase/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
11.
J Lipids ; 2011: 532145, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21912747

RESUMO

Oxidized low-density lipoproteins (LDLs) play an important role during the development of atherosclerosis characterized by intimal inflammation and macrophage accumulation. A key component of LDL is lysophosphatidylcholine (lysoPC). LysoPC is a strong proinflammatory mediator, and its mechanism is uncertain, but it has been suggested to be mediated via the platelet activating factor (PAF) receptor. Here, we report that PAF triggers a pertussis toxin- (PTX-) sensitive intracellular signaling pathway leading to sequential activation of sPLA(2), PLD, cPLA(2), and AA release in human-derived monocytes. In contrast, lysoPC initiates two signaling pathways, one sequentially activating PLD and cPLA(2), and a second parallel PTX-sensitive pathway activating cPLA(2) with concomitant activation of sPLA(2), all leading to AA release. In conclusion, lysoPC and PAF stimulate AA release by divergent pathways suggesting involvement of independent receptors. Elucidation of monocyte lysoPC-specific signaling mechanisms will aid in the development of novel strategies for atherosclerosis prevention, diagnosis, and therapy.

12.
J Biol Chem ; 285(32): 24904-14, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20511230

RESUMO

The transcription factor interferon regulatory factor-3 (IRF3) regulates expression of type I interferon-beta and plays an important role in antiviral immunity. Despite the biological importance of IRF3, its in vivo phosphorylation pattern has not been reported. In this study, we have identified residues in IRF3 that are phosphorylated in vivo after infection with Sendai virus. We found that Sendai virus induced phosphorylation of the C-terminal residues Thr(390) and Ser(396), in addition to either Ser(385) or Ser(386). Moreover, Ser(173) and Ser(175) were constitutively phosphorylated. Ser(396) has previously been suggested to be the major target of the IRF3-activating kinase TBK1 (TANK-binding kinase-1), whereas Thr(390) has not previously been implicated in IRF3 regulation. Mutagenesis studies indicated that phosphorylation of Thr(390) promotes Ser(396) phosphorylation and binding to the coactivator cAMP-response element-binding protein. Taken together, our results show that IRF3 is subject to multiple interdependent phosphorylations, and we identify Thr(390) as a novel in vivo phosphorylation site that modulates the phosphorylation status of TBK1-targeted Ser(396).


Assuntos
Regulação Viral da Expressão Gênica , Fator Regulador 3 de Interferon/química , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Espectrometria de Massas/métodos , Dados de Sequência Molecular , Mutagênese , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Recombinantes/química , Vírus Sendai/metabolismo , Homologia de Sequência de Aminoácidos , Serina/química , Treonina/química
13.
J Biol Chem ; 285(22): 17011-9, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20351107

RESUMO

Interferon regulatory factors (IRFs) are crucial for transcription during innate immune responses. We have previously shown that the tyrosine kinase c-Src enhances IRF-3-dependent transcription in response to viral double-stranded RNA. In this study, we show that c-Src has distinct roles in Toll-like receptor (TLR)-mediated activation of IRF-5 and IRF-3. Surprisingly, c-Src inhibition markedly enhanced IRF-5 activation after treatment with unmethylated CpG, while suppressing IRF-3 activation. Also, CpG-elicited interleukin-6 mRNA production was increased, whereas IP10 mRNA synthesis was reduced in cells deficient in c-Src. Interestingly, c-Src regulated TLR-stimulated induction of activating transcription factor 3 (ATF3), a transcriptional repressor. Depletion of ATF3 by small interfering RNA markedly enhanced interleukin-6 production after CpG treatment, whereas IP10 production was reduced. These results demonstrate functional specificity for c-Src in TLR-stimulated responses and suggest that c-Src modulation and ATF3 activity may contribute to differential regulation of IRF-3- versus IRF-5-mediated gene expression.


Assuntos
Proteínas Tirosina Quinases/metabolismo , Receptores Toll-Like/metabolismo , Fator 3 Ativador da Transcrição/metabolismo , Animais , Proteína Tirosina Quinase CSK , Linhagem Celular , Ilhas de CpG , Humanos , Fator Regulador 3 de Interferon/metabolismo , Fatores Reguladores de Interferon/metabolismo , Interleucina-6/metabolismo , Camundongos , Modelos Biológicos , Interferência de RNA , Transdução de Sinais , Quinases da Família src
14.
J Biol Chem ; 284(28): 19122-31, 2009 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-19419966

RESUMO

Antiviral immune responses are initiated through Toll-like receptors (TLRs) and RIG-I (retinoic acid-inducible gene-I)-like RNA helicases that recognize nucleic acids from distinct viruses. In this study, we show that the tyrosine kinase c-Src participates in antiviral responses induced by the cytoplasmic RNA helicase RIG-I. Sendai virus (SV), which is recognized by RIG-I, induced c-Src phosphorylation. Functional impairment of c-Src through chemical inhibition or transient expression of a c-Src kinase-inactive mutant attenuated production of endogenous antiviral proteins after SV infection or after expression of RIG-I or its adapter protein MAVS. Importantly, SV-stimulated synthesis of antiviral proteins was significantly impaired in cells treated with c-Src small interfering RNA and in cells from c-Src-deficient mice. In addition, we found that c-Src interacted with components of the RIG-I pathway: RIG-I, MAVS, and TRAF3 (tumor necrosis factor receptor-associated factor-3). The interaction between c-Src and TRAF3 was found to occur within the RING domain of TRAF3. Taken together, our results suggest that c-Src enhances RIG-I-mediated signaling, acting at the level of TRAF3.


Assuntos
Antivirais/metabolismo , RNA Helicases DEAD-box/metabolismo , Regulação da Expressão Gênica , Fator 3 Associado a Receptor de TNF/metabolismo , Quinases da Família src/fisiologia , Animais , Proteína DEAD-box 58 , Genes Reporter , Humanos , Fator Regulador 3 de Interferon/metabolismo , Camundongos , Modelos Biológicos , RNA Interferente Pequeno/metabolismo , Receptores Imunológicos , Vírus Sendai/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo
15.
EMBO J ; 25(14): 3335-46, 2006 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-16858407

RESUMO

Double-stranded RNA (dsRNA) is produced during the replication cycle of most viruses and triggers antiviral immune responses through Toll-like receptor 3 (TLR3). However, the molecular mechanisms and subcellular compartments associated with dsRNA-TLR3-mediated signaling are largely unknown. Here we show that c-Src tyrosine kinase is activated by dsRNA in human monocyte-derived dendritic cells, and is recruited to TLR3 in a dsRNA-dependent manner. DsRNA-induced activation of interferon-regulatory factor 3 and signal transducer and activator of transcription 1 was abolished in Src kinase-deficient cells, and restored by adding back c-Src, suggesting a central role of c-Src in antiviral immunity. We also provide evidence that TLR3 is localized in the endoplasmic reticulum of unstimulated cells, moves to dsRNA-containing endosomes in response to dsRNA, and colocalizes with c-Src on endosomes containing dsRNA in the lumen. These results provide novel insight into the molecular mechanisms of TLR3-mediated signaling, which may contribute to the understanding of innate immune responses during viral infections.


Assuntos
Endossomos/enzimologia , Endossomos/virologia , Proteínas Tirosina Quinases/fisiologia , RNA de Cadeia Dupla/antagonistas & inibidores , Rhinovirus/fisiologia , Transdução de Sinais/fisiologia , Receptor 3 Toll-Like/fisiologia , Animais , Proteína Tirosina Quinase CSK , Células Cultivadas , Células Dendríticas , Endossomos/fisiologia , Ativação Enzimática/fisiologia , Células HeLa , Humanos , Fator Regulador 3 de Interferon/metabolismo , Camundongos , Camundongos Knockout , Proteínas Tirosina Quinases/metabolismo , RNA de Cadeia Dupla/fisiologia , Rhinovirus/genética , Fator de Transcrição STAT1/metabolismo , Receptor 3 Toll-Like/metabolismo , Quinases da Família src
16.
J Lipid Res ; 45(5): 831-8, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14754906

RESUMO

Oxidation and lipolytic remodeling of LDL are believed to stimulate LDL entrapment in the arterial wall, expanding the inflammatory response and promoting atherosclerosis. However, the cellular responses and molecular mechanisms underlying the atherogenic effects of lipolytically modified LDL are incompletely understood. Human THP-1 monocytes were prelabeled with [(3)H]arachidonic acid (AA) before incubation with LDL or LDL lipolytically modified by secretory PLA(2) (sPLA(2)) or bacterial sphingomyelinase (SMase). LDL elicited rapid and dose-dependent extracellular release of AA in monocytes. Interestingly, LDL modified by sPLA(2) or SMase displayed a marked increase in AA mobilization relative to native LDL, and this increase correlated with enhanced activity of cytosolic PLA(2) (cPLA(2)) assayed in vitro as well as increased monocyte tumor necrosis factor-alpha secretion. The AA liberation was attenuated by inhibitors toward cPLA(2) and sPLA(2), indicating that both PLA(2) enzymes participate in LDL-induced AA release. In conclusion, these results demonstrate that LDL lipolytically modified by sPLA(2) or SMase potentiates cellular AA release and cPLA(2) activation in human monocytes. From our results, we suggest novel atherogenic properties for LDL modified by sPLA(2) and SMase in AA release and signaling, which could contribute to the inflammatory gene expression observed in atherosclerosis.


Assuntos
Ácido Araquidônico/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Monócitos/efeitos dos fármacos , Fosfolipases A/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Técnicas de Cultura de Células , Humanos , Lipoproteínas LDL/química , Monócitos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
17.
FEBS Lett ; 555(2): 257-62, 2003 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-14644424

RESUMO

To determine if lysophosphatidylcholine (lysoPC) is able to induce proinflammatory changes in monocytes, its ability to stimulate arachidonic acid (AA) release, a product of phospholipase A2 (PLA(2)) activity, has been analyzed. LysoPC increased AA release in THP-1 and Mono Mac6 cells in a time- and concentration-dependent manner. The monocytes expressed both secretory and cytosolic PLA(2) enzymes and AA release was strongly reduced by cellular pretreatment with different PLA(2) inhibitors and by pertussis toxin, an inhibitor of G(i)-protein activation. This indicates that both cytosolic and secretory PLA(2) enzymes regulate specific lysoPC receptor-induced AA release, suggesting lysoPC participation in monocyte proinflammatory activation.


Assuntos
Ácido Araquidônico/metabolismo , Lisofosfatidilcolinas/farmacologia , Monócitos/metabolismo , Fosfolipases A/fisiologia , Ácido Araquidônico/química , Ácidos Araquidônicos/farmacologia , Radioisótopos de Carbono , Diferenciação Celular/fisiologia , Linhagem Celular , Citosol/enzimologia , Citosol/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Monócitos/efeitos dos fármacos , Ácido Oleico/química , Ácido Oleico/metabolismo , Organofosfonatos/farmacologia , Toxina Pertussis/farmacologia , Fosfolipases A/antagonistas & inibidores , Fosfolipases A/metabolismo , Fosfolipases A2 , Fatores de Tempo , Trítio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...