Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38731896

RESUMO

Following infection, influenza viruses strive to establish a new host cellular environment optimized for efficient viral replication and propagation. Influenza viruses use or hijack numerous host factors and machinery not only to fulfill their own replication process but also to constantly evade the host's antiviral and immune response. For this purpose, influenza viruses appear to have formulated diverse strategies to manipulate the host proteins or signaling pathways. One of the most effective tactics is to specifically induce the degradation of the cellular proteins that are detrimental to the virus life cycle. Here, we summarize the cellular factors that are deemed to have been purposefully degraded by influenza virus infection. The focus is laid on the mechanisms for the protein ubiquitination and degradation in association with facilitated viral amplification. The fate of influenza viral infection of hosts is heavily reliant on the outcomes of the interplay between the virus and the host antiviral immunity. Understanding the processes of how influenza viruses instigate the protein destruction pathways could provide a foundation for the development of advanced therapeutics to target host proteins and conquer influenza.


Assuntos
Interações Hospedeiro-Patógeno , Orthomyxoviridae , Ubiquitinação , Replicação Viral , Humanos , Orthomyxoviridae/metabolismo , Orthomyxoviridae/fisiologia , Influenza Humana/metabolismo , Influenza Humana/virologia , Proteólise , Animais
2.
Viruses ; 15(9)2023 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-37766322

RESUMO

Following virus infections, type I interferons are synthesized to induce the expression of antiviral molecules and interfere with virus replication. The importance of early antiviral type I IFN response against virus invasion has been emphasized during COVID-19 as well as in studies on the microbiome. Further, type I IFNs can directly act on various immune cells to enhance protective host immune responses to viral infections. However, accumulating data indicate that IFN responses can be harmful to the host by instigating inflammatory responses or inducing T cell suppression during virus infections. Also, inhibition of lymphocyte and dendritic cell development can be caused by type I IFN, which is independent of the traditional signal transducer and activator of transcription 1 signaling. Additionally, IFNs were shown to impair airway epithelial cell proliferation, which may affect late-stage lung tissue recovery from the infection. As such, type I IFN-virus interaction research is diverse, including host antiviral innate immune mechanisms in cells, viral strategies of IFN evasion, protective immunity, excessive inflammation, immune suppression, and regulation of tissue repair. In this report, these IFN activities are summarized with an emphasis placed on the functions of type I IFNs recently observed during acute or chronic virus infections.


Assuntos
COVID-19 , Interferon Tipo I , Viroses , Humanos , Antivirais/uso terapêutico , Replicação Viral
3.
DNA Cell Biol ; 41(4): 331-335, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35325556

RESUMO

Sphingosine 1-phosphate lyase (SPL) is a critical component of sphingosine 1-phosphate (S1P) metabolism. SPL has been associated with several crucial cellular functions due to its role in S1P metabolism, but its role in viral infections is poorly understood. Studies show that SPL has an antiviral function against influenza A virus (IAV) by interacting with IKKɛ, promoting the type I interferon (IFN) innate immune response to IAV infection. However, a more recent study has revealed that IAV NS1 protein hampers this by triggering ubiquitination and subsequent degradation of SPL, which reduces the type I IFN innate immune response. In this study, we describe SPL, the type I IFN response, and known interactions between SPL and IAV.


Assuntos
Vírus da Influenza A , Influenza Humana , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Lisofosfolipídeos , Esfingosina/análogos & derivados
4.
Viruses ; 13(10)2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34696381

RESUMO

Ever since the immune regulatory strains of lymphocytic choriomeningitis virus (LCMV), such as Clone 13, were isolated, LCMV infection of mice has served as a valuable model for the mechanistic study of viral immune suppression and virus persistence. The exhaustion of virus-specific T cells was demonstrated during LCMV infection, and the underlying mechanisms have been extensively investigated using LCMV infection in mouse models. In particular, the mechanism for gradual CD8+ T cell exhaustion at molecular and transcriptional levels has been investigated. These studies revealed crucial roles for inhibitory receptors, surface markers, regulatory cytokines, and transcription factors, including PD-1, PSGL-1, CXCR5, and TOX in the regulation of T cells. However, the action mode for CD4+ T cell suppression is largely unknown. Recently, sphingosine kinase 2 was proven to specifically repress CD4+ T cell proliferation and lead to LCMV persistence. As CD4+ T cell regulation was also known to be important for viral persistence, research to uncover the mechanism for CD4+ T cell repression could help us better understand how viruses launch and prolong their persistence. This review summarizes discoveries derived from the study of LCMV in regard to the mechanisms for T cell suppression and approaches for the termination of viral persistence with special emphasis on CD8+ T cells.


Assuntos
Interações entre Hospedeiro e Microrganismos/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Infecção Persistente/imunologia , Linfócitos T/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/imunologia , Humanos , Ativação Linfocitária , Coriomeningite Linfocítica/virologia , Camundongos , Infecção Persistente/virologia , Linfócitos T/classificação
5.
Virology ; 558: 67-75, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33730651

RESUMO

The type I interferon (IFN)-mediated innate immune response is one of the central obstacles influenza A virus (IAV) must overcome in order to successfully replicate within the host. We have previously shown that sphingosine 1-phosphate (S1P) lyase (SPL) enhances IKKϵ-mediated type I IFN responses. Here, we demonstrate that the nonstructural protein 1 (NS1) of IAV counteracts the SPL-mediated antiviral response by inducing degradation of SPL. SPL was ubiquitinated and downregulated upon IAV infection or NS1 expression, whereas NS1-deficient IAV failed to elicit SPL ubiquitination or downregulation. Transiently overexpressed SPL increased phosphorylation of IKKϵ, resulting in enhanced expression of type I IFNs. However, this induction was markedly inhibited by IAV NS1. Collectively, this study reveals a novel strategy employed by IAV to subvert the type I IFN response, providing new insights into the interplay between IAV and host innate immunity.


Assuntos
Aldeído Liases/metabolismo , Interações entre Hospedeiro e Microrganismos/imunologia , Imunidade Inata , Vírus da Influenza A/imunologia , Proteólise , Proteínas não Estruturais Virais/imunologia , Células A549 , Aldeído Liases/genética , Regulação para Baixo , Células HEK293 , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Evasão da Resposta Imune , Influenza Humana , Fosforilação , Ubiquitinação
6.
J Gen Virol ; 102(3)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33416468

RESUMO

Chios mastic gum (CMG), a resin of the mastic tree (Pistacia lentiscus var. chia), has been used to treat multiple disorders caused by gastrointestinal malfunctions and bacterial infections for more than 2500 years. However, little is known about CMG's antiviral activity. CMG is known to influence multiple cellular processes such as cell proliferation, differentiation and apoptosis. As virus replication is largely dependent on the host cellular metabolism, it is conceivable that CMG regulates virus infectivity. Therefore, in this study, we evaluated CMG's potential as an antiviral drug to treat influenza A virus (IAV) infection. CMG treatment dramatically reduced the cytopathogenic effect and production of RNAs, proteins and infectious particles of IAV. Interestingly, CMG interfered with the early stage of the virus life cycle after viral attachment. Importantly, the administration of CMG greatly ameliorated morbidity and mortality in IAV-infected mice. The results suggest that CMG displays a potent anti-IAV activity by blocking the early stage of viral replication. Thus, mastic gum could be exploited as a novel therapeutic agent against IAV infection.


Assuntos
Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/patogenicidade , Resina Mástique/farmacologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Efeito Citopatogênico Viral/efeitos dos fármacos , Cães , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Células Madin Darby de Rim Canino , Resina Mástique/uso terapêutico , Infecções por Orthomyxoviridae/virologia , Virulência/efeitos dos fármacos , Ligação Viral , Replicação Viral/efeitos dos fármacos
7.
J Clin Invest ; 130(12): 6523-6538, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32897877

RESUMO

Chronic viral infections are often established by the exploitation of immune-regulatory mechanisms that result in nonfunctional T cell responses. Viruses that establish persistent infections remain a serious threat to human health. Sphingosine kinase 2 (SphK2) generates sphingosine 1-phosphate, which is a molecule known to regulate multiple cellular processes. However, little is known about SphK2's role during the host immune responses to viral infection. Here, we demonstrate that SphK2 functions during lymphocytic choriomeningitis virus Cl 13 (LCMV Cl 13) infection to limit T cell immune pathology, which subsequently aids in the establishment of virus-induced immunosuppression and the resultant viral persistence. The infection of Sphk2-deficient (Sphk2-/-) mice with LCMV Cl 13 led to the development of nephropathy and mortality via T cell-mediated immunopathology. Following LCMV infection, Sphk2-/- CD4+ T cells displayed increased activity and proliferation, and these cells promoted overactive LCMV Cl 13-specific CD8+ T cell responses. Notably, oral instillation of an SphK2-selective inhibitor promoted protective T cell responses and accelerated the termination of LCMV Cl 13 persistence in mice. Thus, SphK2 is indicated as an immunotherapeutic target for the control of persistent viral infections.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Nefropatias/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Fosfotransferases (Aceptor do Grupo Álcool)/imunologia , Animais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Nefropatias/genética , Nefropatias/patologia , Nefropatias/virologia , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/patologia , Camundongos , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/genética
8.
Viruses ; 12(6)2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32570695

RESUMO

Viruses have evolved to survive in hosts, presumably by devising meticulous strategies to elude or suppress host immunity [...].


Assuntos
Vírus da Diarreia Viral Bovina Tipo 1/imunologia , Herpesvirus Suídeo 1/imunologia , Evasão da Resposta Imune/imunologia , Vírus da Influenza A/imunologia , Vírus dos Macacos de Mason-Pfizer/imunologia , Linhagem Celular , Células HEK293 , Humanos
9.
J Virol ; 94(7)2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-31915279

RESUMO

Influenza A virus (IAV) utilizes multiple strategies to confront or evade host type I interferon (IFN)-mediated antiviral responses in order to enhance its own propagation within the host. One such strategy is to induce the degradation of type I IFN receptor 1 (IFNAR1) by utilizing viral hemagglutinin (HA). However, the molecular mechanism behind this process is poorly understood. Here, we report that a cellular protein, poly(ADP-ribose) polymerase 1 (PARP1), plays a critical role in mediating IAV HA-induced degradation of IFNAR1. We identified PARP1 as an interacting partner for IAV HA through mass spectrometry analysis. This interaction was confirmed by coimmunoprecipitation analyses. Furthermore, confocal fluorescence microscopy showed altered localization of endogenous PARP1 upon transient IAV HA expression or during IAV infection. Knockdown or inhibition of PARP1 rescued IFNAR1 levels upon IAV infection or HA expression, exemplifying the importance of PARP1 for IAV-induced reduction of IFNAR1. Notably, PARP1 was crucial for the robust replication of IAV, which was associated with regulation of the type I IFN receptor signaling pathway. These results indicate that PARP1 promotes IAV replication by controlling viral HA-induced degradation of host type I IFN receptor. Altogether, these findings provide novel insight into interactions between influenza virus and the host innate immune response and reveal a new function for PARP1 during influenza virus infection.IMPORTANCE Influenza A virus (IAV) infections cause seasonal and pandemic influenza outbreaks, which pose a devastating global health concern. Despite the availability of antivirals against influenza, new IAV strains continue to persist by overcoming the therapeutics. Therefore, much emphasis in the field is placed on identifying new therapeutic targets that can more effectively control influenza. IAV utilizes several tactics to evade host innate immunity, which include the evasion of antiviral type I interferon (IFN) responses. Degradation of type I IFN receptor (IFNAR) is one known method of subversion, but the molecular mechanism for IFNAR downregulation during IAV infection remains unclear. Here, we have found that a host protein, poly(ADP-ribose) polymerase 1 (PARP1), facilitates IFNAR degradation and accelerates IAV replication. The findings reveal a novel cellular target for the potential development of antivirals against influenza, as well as expand our base of knowledge regarding interactions between influenza and the host innate immunity.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Influenza Humana/virologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Células A549 , Animais , Antivirais/farmacologia , Chlorocebus aethiops , Cães , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Imunidade Inata , Influenza Humana/imunologia , Células Madin Darby de Rim Canino , Espectrometria de Massas , Microscopia Confocal , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Células Vero
10.
Viruses ; 11(12)2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31783527

RESUMO

The sphingosine 1-phosphate (S1P) metabolic pathway is a dynamic regulator of multiple cellular and disease processes. Identification of the immune regulatory role of the sphingosine analog FTY720 led to the development of the first oral therapy for the treatment of an autoimmune disease, multiple sclerosis. Furthermore, inhibitors of sphingosine kinase (SphK), which mediate S1P synthesis, are being evaluated as a therapeutic option for the treatment of cancer. In conjunction with these captivating discoveries, S1P and S1P-metabolizing enzymes have been revealed to display vital functions during virus infections. For example, S1P lyase, which is known for metabolizing S1P, inhibits influenza virus replication by promoting antiviral type I interferon innate immune responses. In addition, both isoforms of sphingosine kinase have been shown to regulate the replication or pathogenicity of many viruses. Pro- or antiviral activities of S1P-metabolizing enzymes appear to be dependent on diverse virus-host interactions and viral pathogenesis. This review places an emphasis on summarizing the functions of S1P-metabolizing enzymes during virus infections and discusses the opportunities for designing pioneering antiviral drugs by targeting these host enzymes.


Assuntos
Aldeído Liases/imunologia , Imunidade Inata , Lisofosfolipídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/imunologia , Esfingosina/análogos & derivados , Viroses/imunologia , Vírus/enzimologia , Aldeído Liases/antagonistas & inibidores , Antivirais/imunologia , Antivirais/uso terapêutico , Interações Hospedeiro-Patógeno , Humanos , Interferon Tipo I/uso terapêutico , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Esfingosina/metabolismo , Viroses/virologia , Replicação Viral , Vírus/genética , Vírus/imunologia , Vírus/patogenicidade
11.
Antiviral Res ; 158: 171-177, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30125617

RESUMO

Influenza continues to pose a threat to public health by causing illness and mortality in humans. Discovering host factors that regulate influenza virus propagation is vital for the development of novel drugs. We have previously reported that sphingosine kinase (SphK) 1 promotes influenza A virus (IAV) replication in vitro. Here we demonstrate that the other isoform of SphK, SphK2 promotes the replication of influenza A virus (IAV) in cultured cells, and temporary inhibition of SphK1 or SphK2 enhances the host defense against influenza in mice. IAV infection led to an increased expression and phosphorylation of SphK2 in host cells. Furthermore, pharmacologic inhibition or siRNA-based knockdown of SphK2 attenuated IAV replication in vitro. Notably, oral administration of an SphK2-specific inhibitor substantially improved the viability of mice following IAV infection. In addition, the local instillation of an SphK1-specific inhibitor or an inhibitor that globally blocks SphK1 and SphK2 provided protection to IAV-infected mice. Collectively, our results indicate that both SphK1 and SphK2 function as proviral factors during IAV infection in vivo. Therefore, SphK1 and SphK2 represent potential host targets for therapeutics against influenza.


Assuntos
Vírus da Influenza A/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Células A549 , Adamantano/análogos & derivados , Adamantano/farmacologia , Administração Oral , Amino Álcoois/farmacologia , Aminofenóis/farmacologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Vírus da Influenza A/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Isoformas de Proteínas , Piridinas/farmacologia , RNA Interferente Pequeno , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tiazóis/farmacologia , Replicação Viral
12.
Virology ; 522: 19-26, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30014854

RESUMO

Interferons (IFNs) exhibit forceful inhibitory activities against numerous viruses by inducing synthesis of anti-viral proteins or promoting immune cell functions, which help eradicate the vicious microbes. Consequently, the degree to which viruses evade or counterattack IFN responses influences viral pathogenicity. Viruses have developed many strategies to interfere with the synthesis of IFNs or IFN receptor signaling pathway. Furthermore, multiple viruses decrease levels of IFN receptors via diverse tactics, which include decreasing type I IFN receptor mRNA expression, blocking post-translational modification of the receptor, and degrading IFN receptors. Recently, influenza virus was found to induce CK1α-induced phosphorylation and subsequent degradation of the receptor for type I and II IFNs. In this review, viral mechanisms that remove IFN receptors are summarized with an emphasis on the mechanisms for virus-induced degradation of IFN receptors.


Assuntos
Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Receptores de Interferon/antagonistas & inibidores , Vírus/patogenicidade , Animais , Humanos
13.
J Clin Invest ; 128(6): 2459-2472, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29733298

RESUMO

Many Toll-like receptors (TLRs) signal through TNF receptor-associated factor 6 (TRAF6) to activate innate immune responses. Here, we show that somatic nuclear autoantigenic sperm protein (sNASP) binds to TRAF6 to prevent TRAF6 autoubiquitination in unstimulated macrophages. Following LPS stimulation, a complex consisting of sNASP, TRAF6, IRAK4, and casein kinase 2 (CK2) is formed. CK2 phosphorylates sNASP at serine 158, allowing sNASP to dissociate from TRAF6. Free TRAF6 is then autoubiquitinated, followed by activation of downstream signaling pathways. In sNasp S158A knockin (S158A-KI) mice, LPS-treated macrophages could not phosphorylate sNASP, which remained bound to TRAF6. S158A-KI mice were more susceptible to sepsis due to a marked reduction in IL-1ß, TNF-α, and IFN-γ production accompanied by an inability to clear bacteria and recruit leukocytes. Furthermore, phosphorylation-regulated release of sNASP from TRAF6 is observed following activation of TLR-1, -2, -4, -5, and -6. Thus, sNASP is a negative regulator of TLR signaling to modulate the innate immune response.


Assuntos
Imunidade Inata , Macrófagos/imunologia , Sepse/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Animais , Autoantígenos/genética , Autoantígenos/imunologia , Proteínas de Ciclo Celular , Citocinas/genética , Citocinas/imunologia , Células HEK293 , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/imunologia , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/patologia , Camundongos , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Células RAW 264.7 , Sepse/genética , Sepse/patologia , Transdução de Sinais/genética , Células THP-1 , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/imunologia , Receptores Toll-Like/genética
14.
J Virol ; 92(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29343571

RESUMO

Although influenza A virus (IAV) evades cellular defense systems to effectively propagate in the host, the viral immune-evasive mechanisms are incompletely understood. Our recent data showed that hemagglutinin (HA) of IAV induces degradation of type I IFN receptor 1 (IFNAR1). Here, we demonstrate that IAV HA induces degradation of type II IFN (IFN-γ) receptor 1 (IFNGR1), as well as IFNAR1, via casein kinase 1α (CK1α), resulting in the impairment of cellular responsiveness to both type I and II IFNs. IAV infection or transient HA expression induced degradation of both IFNGR1 and IFNAR1, whereas HA gene-deficient IAV failed to downregulate the receptors. IAV HA caused the phosphorylation and ubiquitination of IFNGR1, leading to the lysosome-dependent degradation of IFNGR1. Influenza viral HA strongly decreased cellular sensitivity to type II IFNs, as it suppressed the activation of STAT1 and the induction of IFN-γ-stimulated genes in response to exogenously supplied recombinant IFN-γ. Importantly, CK1α, but not p38 MAP kinase or protein kinase D2, was proven to be critical for HA-induced degradation of both IFNGR1 and IFNAR1. Pharmacologic inhibition of CK1α or small interfering RNA (siRNA)-based knockdown of CK1α repressed the degradation processes of both IFNGR1 and IFNAR1 triggered by IAV infection. Further, CK1α was shown to be pivotal for proficient replication of IAV. Collectively, the results suggest that IAV HA induces degradation of IFN receptors via CK1α, creating conditions favorable for viral propagation. Therefore, the study uncovers a new immune-evasive pathway of influenza virus.IMPORTANCE Influenza A virus (IAV) remains a grave threat to humans, causing seasonal and pandemic influenza. Upon infection, innate and adaptive immunity, such as the interferon (IFN) response, is induced to protect hosts against IAV infection. However, IAV seems to be equipped with tactics to evade the IFN-mediated antiviral responses, although the detailed mechanisms need to be elucidated. In the present study, we show that IAV HA induces the degradation of the type II IFN receptor IFNGR1 and thereby substantially attenuates cellular responses to IFN-γ. Of note, a cellular kinase, casein kinase 1α (CK1α), is crucial for IAV HA-induced degradation of both IFNGR1 and IFNAR1. Accordingly, CK1α is proven to positively regulate IAV propagation. Thus, this study unveils a novel strategy employed by IAV to evade IFN-mediated antiviral activities. These findings may provide new insights into the interplay between IAV and host immunity to impact influenza virus pathogenicity.


Assuntos
Caseína Quinase I/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Evasão da Resposta Imune , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/imunologia , Proteólise , Receptor de Interferon alfa e beta/imunologia , Receptores de Interferon/imunologia , Células A549 , Animais , Caseína Quinase I/genética , Chlorocebus aethiops , Cães , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Influenza Humana/genética , Influenza Humana/patologia , Células Madin Darby de Rim Canino , Proteína Quinase D2 , Proteínas Quinases/genética , Proteínas Quinases/imunologia , Receptor de Interferon alfa e beta/genética , Receptores de Interferon/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Células Vero , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Receptor de Interferon gama
15.
J Immunol ; 199(2): 677-687, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28600291

RESUMO

Sphingosine 1-phosphate (S1P) lyase (SPL) is an intracellular enzyme that mediates the irreversible degradation of the bioactive lipid S1P. We have previously reported that overexpressed SPL displays anti-influenza viral activity; however, the underlying mechanism is incompletely understood. In this study, we demonstrate that SPL functions as a positive regulator of IKKε to propel type I IFN-mediated innate immune responses against viral infection. Exogenous SPL expression inhibited influenza A virus replication, which correlated with an increase in type I IFN production and IFN-stimulated gene accumulation upon infection. In contrast, the lack of SPL expression led to an elevated cellular susceptibility to influenza A virus infection. In support of this, SPL-deficient cells were defective in mounting an effective IFN response when stimulated by influenza viral RNAs. SPL augmented the activation status of IKKε and enhanced the kinase-induced phosphorylation of IRF3 and the synthesis of type I IFNs. However, the S1P degradation-incompetent form of SPL also enhanced IFN responses, suggesting that SPL's pro-IFN function is independent of S1P. Biochemical analyses revealed that SPL, as well as the mutant form of SPL, interacts with IKKε. Importantly, when endogenous IKKε was downregulated using a small interfering RNA approach, SPL's anti-influenza viral activity was markedly suppressed. This indicates that IKKε is crucial for SPL-mediated inhibition of influenza virus replication. Thus, the results illustrate the functional significance of the SPL-IKKε-IFN axis during host innate immunity against viral infection.


Assuntos
Aldeído Liases/metabolismo , Quinase I-kappa B/metabolismo , Imunidade Inata , Vírus da Influenza A/imunologia , Interferon Tipo I/imunologia , Células A549 , Aldeído Liases/deficiência , Aldeído Liases/genética , Regulação para Baixo , Ativação Enzimática , Células HEK293 , Humanos , Quinase I-kappa B/genética , Vírus da Influenza A/fisiologia , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/biossíntese , Lisofosfolipídeos/metabolismo , Fosforilação , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Replicação Viral
16.
J Virol ; 90(5): 2403-17, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26676772

RESUMO

UNLABELLED: Influenza A virus (IAV) employs diverse strategies to circumvent type I interferon (IFN) responses, particularly by inhibiting the synthesis of type I IFNs. However, it is poorly understood if and how IAV regulates the type I IFN receptor (IFNAR)-mediated signaling mode. In this study, we demonstrate that IAV induces the degradation of IFNAR subunit 1 (IFNAR1) to attenuate the type I IFN-induced antiviral signaling pathway. Following infection, the level of IFNAR1 protein, but not mRNA, decreased. Indeed, IFNAR1 was phosphorylated and ubiquitinated by IAV infection, which resulted in IFNAR1 elimination. The transiently overexpressed IFNAR1 displayed antiviral activity by inhibiting virus replication. Importantly, the hemagglutinin (HA) protein of IAV was proved to trigger the ubiquitination of IFNAR1, diminishing the levels of IFNAR1. Further, influenza A viral HA1 subunit, but not HA2 subunit, downregulated IFNAR1. However, viral HA-mediated degradation of IFNAR1 was not caused by the endoplasmic reticulum (ER) stress response. IAV HA robustly reduced cellular sensitivity to type I IFNs, suppressing the activation of STAT1/STAT2 and induction of IFN-stimulated antiviral proteins. Taken together, our findings suggest that IAV HA causes IFNAR1 degradation, which in turn helps the virus escape the powerful innate immune system. Thus, the research elucidated an influenza viral mechanism for eluding the IFNAR signaling pathway, which could provide new insights into the interplay between influenza virus and host innate immunity. IMPORTANCE: Influenza A virus (IAV) infection causes significant morbidity and mortality worldwide and remains a major health concern. When triggered by influenza viral infection, host cells produce type I interferon (IFN) to block viral replication. Although IAV was shown to have diverse strategies to evade this powerful, IFN-mediated antiviral response, it is not well-defined if IAV manipulates the IFN receptor-mediated signaling pathway. Here, we uncovered that influenza viral hemagglutinin (HA) protein causes the degradation of type I IFN receptor subunit 1 (IFNAR1). HA promoted phosphorylation and polyubiquitination of IFNAR1, which facilitated the degradation of this receptor. The HA-mediated elimination of IFNAR1 notably decreased the cells' sensitivities to type I IFNs, as demonstrated by the diminished expression of IFN-induced antiviral genes. This discovery could help us understand how IAV regulates the host innate immune response to create an environment optimized for viral survival in host cells.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Vírus da Influenza A/imunologia , Vírus da Influenza A/fisiologia , Interferon Tipo I/metabolismo , Receptor de Interferon alfa e beta/antagonistas & inibidores , Animais , Linhagem Celular , Humanos , Fosforilação , Processamento de Proteína Pós-Traducional , Proteólise , Ubiquitinação
17.
Biochem Biophys Res Commun ; 467(4): 766-70, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26478433

RESUMO

Health policy precludes neonatal vaccination against influenza. Hence, morbidity and mortality are high under 6 months of age. Lactoferrin may activate diminished numbers of dysfunctional dendritic cells and reverse neonatal vaccine failures. Aluminum hydroxide/ALUM recruits neutrophils that secrete lactoferrin at deposition sites of antigen. We theorized lactoferrin + influenza antigen initiates an equivalent antibody response compared to ALUM. Three-day-old mice received subcutaneously 30 µg of H1N1 hemagglutinin + 200 µg of bovine lactoferrin versus hemagglutinin + ALUM. Controls received hemagglutinin, lactoferrin, or ALUM. After 21 days, sera measured anti-H1N1 (ELISA) and neutralizing antibody (plaque assays). ELISA detected equal antibody production with lactoferrin + hemagglutinin compared to hemagglutinin + ALUM; both sera also neutralized H1N1 virus at a 1:20 dilution (p < 0.01). Controls had no anti-H1N1 antibody. Neonates given lactoferrin had no anaphylaxis when challenged four weeks later. Lactoferrin is a safe and effective adjuvant for inducing antibody against influenza in neonates.


Assuntos
Adjuvantes Imunológicos/farmacologia , Lactoferrina/farmacologia , Infecções por Orthomyxoviridae/imunologia , Vacinação/métodos , Hidróxido de Alumínio/imunologia , Hidróxido de Alumínio/farmacologia , Animais , Animais Recém-Nascidos , Bovinos , Cães , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/farmacologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Lactoferrina/imunologia , Células Madin Darby de Rim Canino/virologia , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia
18.
J Immunol ; 194(9): 4339-49, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25810392

RESUMO

The ceramide family of lipids plays important roles in both cell structure and signaling in a diverse array of cell types, including immune cells. However, very little is known regarding how ceramide affects the activation of dendritic cells (DCs) in response to viral infection. In this study, we demonstrate that a synthetic ceramide analog (C8) stimulates DCs to increase the expansion of virus-specific T cells upon virus infection. Exogenously supplied C8 ceramide elevated the expression of DC maturation markers such as MHC class I and costimulatory molecules following infection with the clone 13 strain of lymphocytic choriomeningitis virus (LCMV) or influenza virus. Importantly, ceramide-conditioned, LCMV-infected DCs displayed an increased ability to promote expansion of virus-specific CD8(+) T cells when compared with virus-infected DCs. Furthermore, a locally instilled ceramide analog significantly increased virus-reactive T cell responses in vivo to both LCMV and influenza virus infections. Collectively, these findings provide new insights into ceramide-mediated regulation of DC responses against virus infection and help us establish a foundation for novel immune-stimulatory therapeutics.


Assuntos
Ceramidas/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Viroses/imunologia , Viroses/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Modelos Animais de Doenças , Fatores Imunológicos/farmacologia , Imunomodulação/efeitos dos fármacos , Ativação Linfocitária , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Subpopulações de Linfócitos T/efeitos dos fármacos
19.
Viral Immunol ; 27(2): 82-6, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24605791

RESUMO

Sphingosine analogs display diverse immunoregulatory activities with curative potential in autoimmune diseases and viral infections. Recently, the sphingosine analog AAL-R was shown to increase DC activation upon TLR7 stimulation. Here, we investigated the effect of AAL-R on activation of dendritic cells (DCs) infected by lymphocytic choriomeningitis virus (LCMV). Concomitant treatment of LCMV-infected DCs with AAL-R enhanced DC maturation and DC ability to stimulate and expand antiviral CD8(+) T cells. Importantly, AAL-R's stimulatory activity was abrogated in type I interferon (IFN) receptor-deficient DCs following LCMV infection. In support of this observation, AAL-R increased type I IFN production from DCs infected with LCMV. Taken together, the sphingosine analog could directly act on DCs to promote defensive host DC responses to the viral invasion via type I IFN signaling.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Fatores Imunológicos/imunologia , Esfingosina/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Fatores Imunológicos/metabolismo , Interferon Tipo I/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Esfingosina/análogos & derivados , Esfingosina/metabolismo
20.
Scientifica (Cairo) ; 2014: 793815, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24672735

RESUMO

Viruses attempt to create a distinctive cellular environment to favor viral replication and spread. Recent studies uncovered new functions of the sphingolipid signaling/metabolism during pathogenic virus infections. While sphingolipids such as sphingomyelin and ceramide were reported to influence the entry step of several viruses, sphingolipid-metabolizing enzymes could directly alter viral replication processes. Influenza virus was shown to increase the level of sphingosine kinase (SK) 1 to promote virus propagation. The mechanism involves regulation of intracellular signaling pathways, leading to the amplification of influenza viral RNA synthesis and nuclear export of viral ribonucleoprotein (RNP) complex. However, bovine viral diarrhea virus inhibits SK1 to enhance the efficacy of virus replication, demonstrating the presence of virus-specific strategies for modulation of the sphingolipid system. Therefore, investigating the sphingolipid metabolism and signaling in the context of virus replication could help us design innovative therapeutic approaches to improve human health.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA