Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
J Immunol ; 188(10): 4759-68, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22490865

RESUMO

Sphingosine analogues display immunosuppressive activities and thus have therapeutic potential in the treatment of autoimmune diseases. In this study, we investigated the effects of the sphingosine analogue AAL-R (FTY720 derivative) on dendritic cell (DC) response upon TLR stimulation. Unlike its known immunosuppressive activity, AAL-R increased TLR7-mediated DC responses by elevating the levels of MHC class I and costimulatory molecules and type I IFN expression and by enhancing the capacity of DCs to induce CD8(+) T cell proliferation. Importantly, the stimulatory activity of AAL-R was dependent on type I IFN signaling, as type I IFN receptor-deficient DCs failed to respond to AAL-R. Also, AAL-R activated p38 MAPK to increase type I IFN synthesis and TLR7-mediated DC maturation. These findings enhance our understanding of sphingosine regulation of the host immune system, in particular upon pathogenic infections.


Assuntos
Células Dendríticas/imunologia , Fatores Imunológicos/farmacologia , Interferon Tipo I/fisiologia , Glicoproteínas de Membrana/fisiologia , Propilenoglicóis/farmacologia , Transdução de Sinais/imunologia , Esfingosina/análogos & derivados , Receptor 7 Toll-Like/fisiologia , Regulação para Cima/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Cloridrato de Fingolimode , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Fosforilação/imunologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/farmacologia , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Proc Natl Acad Sci U S A ; 108(1): 331-6, 2011 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-21173229

RESUMO

Measles virus (MV), a member of the family Paramyxoviridae and an exclusively human pathogen, is among the most infectious viruses. A progressive fatal neurodegenerative complication, subacute sclerosing panencephalitis (SSPE), occurs during persistent MV infection of the CNS and is associated with biased hypermutations of the viral genome. The observed hypermutations of A-to-G are consistent with conversions catalyzed by the adenosine deaminase acting on RNA (ADAR1). To evaluate the role of ADAR1 in MV infection, we selectively disrupted expression of the IFN-inducible p150 ADAR1 isoform and found it caused embryonic lethality at embryo day (E) 11-E12. We therefore generated p150-deficient and WT mouse embryo fibroblast (MEF) cells stably expressing the MV receptor signaling lymphocyte activation molecule (SLAM or CD150). The p150(-/-) but not WT MEF cells displayed extensive syncytium formation and cytopathic effect (CPE) following infection with MV, consistent with an anti-MV role of the p150 isoform of ADAR1. MV titers were 3 to 4 log higher in p150(-/-) cells compared with WT cells at 21 h postinfection, and restoration of ADAR1 in p150(-/-) cells prevented MV cytopathology. In contrast to infection with MV, p150 disruption had no effect on vesicular stomatitis virus, reovirus, or lymphocytic choriomeningitis virus replication but protected against CPE resulting from infection with Newcastle disease virus, Sendai virus, canine distemper virus, and influenza A virus. Thus, ADAR1 is a restriction factor in the replication of paramyxoviruses and orthomyxoviruses.


Assuntos
Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Desenvolvimento Embrionário/genética , Mutação/genética , Vírus SSPE/genética , Panencefalite Esclerosante Subaguda/genética , Replicação Viral/genética , Animais , Antígenos CD/metabolismo , Linhagem Celular , Primers do DNA/genética , Imunofluorescência , Técnicas de Inativação de Genes , Proteínas de Fluorescência Verde , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas/genética , Proteínas de Ligação a RNA , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária
11.
J Exp Med ; 204(8): 1787-801, 2007 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-17664294

RESUMO

In conditions of T lymphopenia, interleukin (IL) 7 levels rise and, via T cell receptor for antigen-self-major histocompatibility complex (MHC) interaction, induce residual naive T cells to proliferate. This pattern of lymphopenia-induced "homeostatic" proliferation is typically quite slow and causes a gradual increase in total T cell numbers and differentiation into cells with features of memory cells. In contrast, we describe a novel form of homeostatic proliferation that occurs when naive T cells encounter raised levels of IL-2 and IL-15 in vivo. In this situation, CD8(+) T cells undergo massive expansion and rapid differentiation into effector cells, thus closely resembling the T cell response to foreign antigens. However, the responses induced by IL-2/IL-15 are not seen in MHC-deficient hosts, implying that the responses are driven by self-ligands. Hence, homeostatic proliferation of naive T cells can be either slow or fast, with the quality of the response to self being dictated by the particular cytokine (IL-7 vs. IL-2/IL-15) concerned. The relevance of the data to the gradual transition of naive T cells into memory-phenotype (MP) cells with age is discussed.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Interleucina-2/metabolismo , Animais , Membrana Celular/metabolismo , Proliferação de Células , Separação Celular , Memória Imunológica , Interleucina-15/metabolismo , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Subunidade beta de Receptor de Interleucina-2/biossíntese , Ligantes , Complexo Principal de Histocompatibilidade , Camundongos , Modelos Biológicos , Fenótipo
12.
Blood ; 118(1): 116-28, 2011 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-21540458

RESUMO

Immune responses lead to expression of immunoregulatory molecules on T cells, including natural killer (NK) receptors, such as CD94/NKG2A on CD8(+) T cells; these receptors restrain CD8(+) responses, thereby preventing T-cell exhaustion in chronic infections and limiting immunopathology. Here, we examined the requirements for inducing CD94/NKG2A on T cells responding to antigen. In vitro, moderate induction of CD94/NKG2A expression occurred after exposure of naive CD8(+) (but not CD4(+)) cells to CD3 ligation or specific peptide. Surprisingly, expression was inhibited by CD28/B7 costimulation. Such inhibition applied only to CD94/NKG2A and not other NK receptors (NKG2D) and was mediated by IL-2. Inhibition by IL-2 occurred via a NFAT cell-independent component of the calcineurin pathway, and CD94/NKG2A induction was markedly enhanced in the presence of calcineurin blockers, such as FK506 or using calcineurin-deficient T cells, both in vitro and in vivo. In addition to CD28-dependent inhibition by IL-2, CD94/NKG2A expression was impaired by several other cytokines (IL-4, IL-23, and transforming growth factor-ß) but enhanced by others (IL-6, IL-10, and IL-21). The complex interplay between these various stimuli may account for the variable expression of CD94/NKG2A during responses to different pathogens in vivo.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Calcineurina/metabolismo , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Subfamília D de Receptores Semelhantes a Lectina de Células NK/metabolismo , Animais , Antígenos CD28/imunologia , Antígenos CD28/metabolismo , Linfócitos T CD8-Positivos/imunologia , Calcineurina/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Expressão Gênica/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Interleucina-2/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Subfamília C de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília D de Receptores Semelhantes a Lectina de Células NK/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo
13.
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
14.
Proc Natl Acad Sci U S A ; 106(5): 1560-5, 2009 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-19164548

RESUMO

Pulmonary tissue damage resulting from influenza virus infection is caused by both the cytolytic activity of the virus and the host immune response. Immune-mediated injury results from T cell-mediated destruction of virus-infected cells and by release of cytokines and chemokines that attract polymorphonuclear leukocytes (PML) and macrophages to the infected site. The cytokines/chemokines potentiate dendritic cell (DC) activation and T cell expansion, which further enhances local damage. Here we report that immune modulation by local administration to the respiratory tract of sphingosine analog AAL-R significantly dampens the release of cytokines and chemokines while maintaining protective neutralizing antibody and cytotoxic T cell responses. As a result there was a marked reduction of infiltrating PML and macrophages into the lung and resultant pulmonary tissue injury. DC maturation was suppressed, which limited proliferation of specific antiviral T cells in the lung and draining lymph nodes. Further, AAL-R was effective in controlling CD8(+) T cell accumulation in the lungs even when given 4 days after initiation of influenza virus infection. These data indicate that sphingosine analogs display useful potential for controlling the immunopathology caused by influenza virus.


Assuntos
Citocinas/biossíntese , Influenza Humana/fisiopatologia , Esfingosina/farmacologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Células Apresentadoras de Antígenos/imunologia , Sequência de Bases , DNA Viral , Modelos Animais de Doenças , Humanos , Influenza Humana/imunologia , Influenza Humana/metabolismo , Pulmão/imunologia , Camundongos , Dados de Sequência Molecular , Testes de Neutralização , Linfócitos T Citotóxicos/imunologia
15.
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
16.
J Virol ; 84(16): 8124-31, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20519401

RESUMO

Sphingosine 1-phosphate (S1P)-metabolizing enzymes regulate the level of sphingolipids and have important biological functions. However, the effects of S1P-metabolizing enzymes on host defense against invading viruses remain unknown. In this study, we investigated the role of S1P-metabolizing enzymes in modulating cellular responses to influenza virus infection. Overexpression of S1P lyase (SPL), which induces the degradation of S1P, interfered with the amplification of infectious influenza virus. Accordingly, SPL-overexpressing cells were much more resistant than control cells to the cytopathic effects caused by influenza virus infection. SPL-mediated inhibition of virus-induced cell death was supported by impairment of the upregulation of the proapoptotic protein Bax, a critical factor for influenza virus cytopathogenicity. Importantly, influenza virus infection of SPL-overexpressing cells induced rapid activation of extracellular signal-regulated kinase (ERK) and STAT1 but not of p38 mitogen-activated protein kinase (MAPK), Akt, or c-Jun N-terminal kinase (JNK). Blockade of STAT1 expression or inhibition of Janus kinase (JAK) activity elevated the level of influenza virus replication in the cells, indicating that SPL protects cells from influenza virus via the activation of JAK/STAT signaling. In contrast to that of SPL, the overexpression of S1P-producing sphingosine kinase 1 heightened the cells' susceptibility to influenza virus infection, an effect that was reversed by the inhibition of its kinase activity, representing opposed enzymatic activity. These findings indicate that the modulation of S1P-metabolizing enzymes is crucial for controlling the host defense against infection with influenza virus. Thus, S1P-metabolizing enzymes are novel potential targets for the treatment of diseases caused by influenza virus infection.


Assuntos
Aldeído Liases/metabolismo , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N1/patogenicidade , Lisofosfolipídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Esfingosina/análogos & derivados , Linhagem Celular , Sobrevivência Celular , Efeito Citopatogênico Viral , Humanos , Transdução de Sinais , Esfingosina/metabolismo
17.
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
18.
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
19.
Adv Appl Microbiol ; 73: 83-101, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20800760

RESUMO

Type I interferon (IFN), as its name implies, 'interferes' with virus replication by activating numerous genes. Further, virus-induced type I IFN regulates the magnitude and functions of cells directing the host immune system. Importantly, recent exploration into how type I IFN operates following virus infection has advanced our understanding of its role with respect to modulation of host innate and adaptive immune responses. Such activities include the activation of antigen-presenting dendritic cells and the localization, expansion or differentiation of virus-specific T lymphocytes and antibody-producing B lymphocytes. However, type I IFN not only benefits the host but can also induce unnecessary or extremely pathogenic immune responses. This review focuses on such interactions and the manner in which type I IFN induces dynamic changes in the host immune network, particularly adaptive immune responses to viral invasion. Manipulating the type I IFN-mediated host immune response during virus infections could provide new immunotherapeutic interventions to remedy viral diseases and implement more effective and sustainable type I IFN therapy.


Assuntos
Sistema Imunitário/imunologia , Interferon gama/imunologia , Viroses/imunologia , Vírus/patogenicidade , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Humanos , Interferon gama/metabolismo , Camundongos , Linfócitos T/citologia , Linfócitos T/imunologia , Viroses/virologia , Vírus/imunologia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA