Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
2.
Nat Immunol ; 21(8): 868-879, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32690950

RESUMO

STING is essential for control of infections and for tumor immunosurveillance, but it can also drive pathological inflammation. STING resides on the endoplasmic reticulum (ER) and traffics following stimulation to the ERGIC/Golgi, where signaling occurs. Although STING ER exit is the rate-limiting step in STING signaling, the mechanism that drives this process is not understood. Here we identify STEEP as a positive regulator of STING signaling. STEEP was associated with STING and promoted trafficking from the ER. This was mediated through stimulation of phosphatidylinositol-3-phosphate (PtdIns(3)P) production and ER membrane curvature formation, thus inducing COPII-mediated ER-to-Golgi trafficking of STING. Depletion of STEEP impaired STING-driven gene expression in response to virus infection in brain tissue and in cells from patients with STING-associated diseases. Interestingly, STING gain-of-function mutants from patients interacted strongly with STEEP, leading to increased ER PtdIns(3)P levels and membrane curvature. Thus, STEEP enables STING signaling by promoting ER exit.


Assuntos
Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais/fisiologia , Animais , Retículo Endoplasmático/imunologia , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Proteínas de Membrana/imunologia , Camundongos , Proteínas do Tecido Nervoso/imunologia , Proteínas Nucleares , Transporte Proteico/fisiologia
3.
EMBO J ; 37(8)2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29496741

RESUMO

Negative regulation of immune pathways is essential to achieve resolution of immune responses and to avoid excess inflammation. DNA stimulates type I IFN expression through the DNA sensor cGAS, the second messenger cGAMP, and the adaptor molecule STING Here, we report that STING degradation following activation of the pathway occurs through autophagy and is mediated by p62/SQSTM1, which is phosphorylated by TBK1 to direct ubiquitinated STING to autophagosomes. Degradation of STING was impaired in p62-deficient cells, which responded with elevated IFN production to foreign DNA and DNA pathogens. In the absence of p62, STING failed to traffic to autophagy-associated vesicles. Thus, DNA sensing induces the cGAS-STING pathway to activate TBK1, which phosphorylates IRF3 to induce IFN expression, but also phosphorylates p62 to stimulate STING degradation and attenuation of the response.


Assuntos
Nucleotidiltransferases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteína Sequestossoma-1/fisiologia , Animais , Autofagia , Linhagem Celular , DNA/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
4.
Fish Shellfish Immunol ; 87: 220-225, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30641186

RESUMO

MicroRNAs (miRNAs) are a type of small non-coding RNAs that participate in diverse cellular processes including microbial invasion and immune defense. In a previous study, we identified a large amount of Japanese flounder (Paralichthys olivaceus) miRNAs responsive to megalocytivirus infection. In the present study, we examined the function of one of these miRNAs, pol-miR-194a, in association with the infectivity of Edwardsiella tarda, an intracellular bacterial pathogen to many fish species including flounder. We found that pol-miR-194a was induced in expression to a significant extent in the spleen, liver, and gill of Japanese flounder infected by E. tarda. Transfection of flounder cells with pol-miR-194a mimic significantly enhanced the intracellular replication of E. tarda. pol-miR-194a was able to interact specifically with the 3'UTR of IRF7 in a negative manner, resulting in inhibition of IRF7 expression. Consistently, pol-miR-194a significantly blocked the promoter activity of type Ⅰ interferon. Taken together, these results indicate that pol-miR-194a plays an important role in the regulation of flounder immune response as well as microbial infection, and that pol-miR-194a probably serves as a target for E. tarda to manipulate and escape host immune defense.


Assuntos
Infecções por Enterobacteriaceae/imunologia , Doenças dos Peixes/imunologia , Linguados/imunologia , Interferon Tipo I/metabolismo , RNA Mensageiro/metabolismo , Animais , Edwardsiella tarda/fisiologia , Distribuição Aleatória
5.
BMC Genomics ; 15: 878, 2014 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-25297525

RESUMO

BACKGROUND: MicroRNAs (miRNAs) regulate gene expression by binding to mRNA transcripts in various biological processes. In mammals and birds, miRNAs are known to play vital parts in both host immune defense and viral infection. However, in lower vertebrates such as teleost, systematic investigations on host and viral miRNAs are lacking. RESULTS: In this study, we applied high-throughput sequencing technology to identify and analyze both host and viral miRNAs in Japanese flounder (Paralichthys olivaceus), an economically important teleost fish farmed widely in the world, infected with megalocytivirus at a timescale of 14 days divided into five different time points. The results showed that a total of 381 host miRNAs and 9 viral miRNAs were identified, the latter being all novel miRNAs that have no homologues in the currently available databases. Of the host miRNAs, 251 have been reported previously in flounder and other species, and 130 were discovered for the first time. The expression levels of 121 host miRNAs were significantly altered at 2 d to 14 d post-viral infection (pi), and these miRNAs were therefore classified as differentially expressed host miRNAs. The expression levels of all 9 viral miRNAs increased from 0 d pi to 10 d pi and then dropped from 10 d pi to 14 d pi. For the 121 differentially expressed host miRNAs and the 9 viral miRNAs, 243 and 48 putative target genes, respectively, were predicted in flounder. GO and KEGG enrichment analysis revealed that the putative target genes of both host and viral miRNAs were grouped mainly into the categories of immune response, signal transduction, and apoptotic process. CONCLUSIONS: The results of our study provide the first evidences that indicate existence in teleost fish (i) infection-responsive host and viral miRNAs that exhibit dynamic changes in expression profiles during the course of viral infection, and (ii) potential involvement of miRNAs in host-viral interaction.


Assuntos
Linguado/genética , Linguado/virologia , Interações Hospedeiro-Patógeno/genética , Iridoviridae/genética , Iridoviridae/fisiologia , MicroRNAs/genética , RNA Viral/genética , Animais , Infecções por Vírus de DNA/genética , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Análise de Sequência de RNA
6.
Fish Shellfish Immunol ; 39(2): 312-20, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24875010

RESUMO

IL-12p40, also called IL-12ß, is a subunit of the proinflammatory cytokines interleukin (IL)-12 and IL-23. In teleost, IL-12p40 homologues have been identified in several species, however, the biological function of fish IL-12p40 is essentially unknown. In this work, we reported the identification and analysis of an IL-12p40, OfIL-12p40, from rock bream (Oplegnathus fasciatus). OfIL-12p40 is composed of 361 amino acids and possesses a conserved IL-12p40 domain and a WSxWS signature motif characteristic of known IL-12p40. Constitutive expression of OfIL-12p40 occurred in multiple tissues and was highest in kidney. Experimental infection with bacterial pathogen upregulated the expression of OfIL-12p40 in kidney and spleen in a time-dependent manner. Purified recombinant OfIL-12p40 (rOfIL-12p40) stimulated the respiratory burst activity of peripheral blood leukocytes in a dose-dependent manner. rOfIL-12p40 also enhanced the resistance of rock bream against bacterial infection and upregulated the expression of innate immune genes in kidney. Taken together, these results indicate that OfIL-12p40 possesses cytokine-like property and plays a role in immune defense against bacterial infection.


Assuntos
Infecções Bacterianas/veterinária , Doenças dos Peixes/genética , Doenças dos Peixes/imunologia , Regulação da Expressão Gênica/imunologia , Subunidade p40 da Interleucina-12/genética , Perciformes , Proteínas Recombinantes/farmacologia , Motivos de Aminoácidos/genética , Análise de Variância , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Primers do DNA/genética , Subunidade p40 da Interleucina-12/imunologia , Rim/metabolismo , Plasmídeos/genética , Estrutura Terciária de Proteína , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Explosão Respiratória/fisiologia , Análise de Sequência de DNA , Baço/metabolismo
7.
Nat Commun ; 15(1): 2760, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38553448

RESUMO

The cGAS-STING pathway plays a crucial role in anti-tumoral responses by activating inflammation and reprogramming the tumour microenvironment. Upon activation, STING traffics from the endoplasmic reticulum (ER) to Golgi, allowing signalling complex assembly and induction of interferon and inflammatory cytokines. Here we report that cGAMP stimulation leads to a transient decline in ER cholesterol levels, mediated by Sterol O-Acyltransferase 1-dependent cholesterol esterification. This facilitates ER membrane curvature and STING trafficking to Golgi. Notably, we identify two cholesterol-binding motifs in STING and confirm their contribution to ER-retention of STING. Consequently, depletion of intracellular cholesterol levels enhances STING pathway activation upon cGAMP stimulation. In a preclinical tumour model, intratumorally administered cholesterol depletion therapy potentiated STING-dependent anti-tumoral responses, which, in combination with anti-PD-1 antibodies, promoted tumour remission. Collectively, we demonstrate that ER cholesterol sets a threshold for STING signalling through cholesterol-binding motifs in STING and we propose that this could be exploited for cancer immunotherapy.


Assuntos
Proteínas de Membrana , Neoplasias , Humanos , Proteínas de Membrana/metabolismo , Transdução de Sinais/fisiologia , Interferons/metabolismo , Nucleotidiltransferases/metabolismo , Neoplasias/terapia , Neoplasias/metabolismo , Retículo Endoplasmático/metabolismo , Microambiente Tumoral
8.
Commun Biol ; 7(1): 283, 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38454028

RESUMO

DNA is a danger signal sensed by cGAS to engage signaling through STING to activate innate immune functions. The best-studied downstream responses to STING activation include expression of type I interferon and inflammatory genes, but STING also activates other pathways, including apoptosis. Here, we report that STING-dependent induction of apoptosis in macrophages occurs through the intrinsic mitochondrial pathway and is mediated via IRF3 but acts independently of gene transcription. By intersecting four mass spectrometry datasets, we identify SAM68 as crucial for the induction of apoptosis downstream of STING activation. SAM68 is essential for the full activation of apoptosis. Still, it is not required for STING-mediated activation of IFN expression or activation of NF-κB. Mechanistic studies reveal that protein trafficking is required and involves SAM68 recruitment to STING upon activation, with the two proteins associating at the Golgi or a post-Golgi compartment. Collectively, our work identifies SAM68 as a STING-interacting protein enabling induction of apoptosis through this DNA-activated innate immune pathway.


Assuntos
Proteínas de Membrana , Transdução de Sinais , Proteínas de Membrana/metabolismo , Macrófagos/metabolismo , Proteínas de Ciclo Celular/metabolismo , DNA/metabolismo , Apoptose
9.
Dis Aquat Organ ; 104(3): 203-14, 2013 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-23759558

RESUMO

The family Iridoviridae consists of 5 genera of double-stranded DNA viruses, including the genus Megalocytivirus, which contains species that are important fish pathogens. In a previous study, we isolated the first rock bream iridovirus from China (RBIV-C1) and identified it as a member of the genus Megalocytivirus. In this report, we determined the complete genomic sequence of RBIV-C1 and examined its in vivo expression profiles. The genome of RBIV-C1 is 112333 bp in length, with a GC content of 55% and a coding density of 92%. RBIV-C1 contains 4584 simple sequence repeats, 89.8% of which are distributed among coding regions. A total of 119 potential open reading frames (ORFs) were identified in RBIV-C1, including the 26 core iridovirus genes; 41 ORFs encode proteins that are predicted to be associated with essential biological functions. RBIV-C1 exhibits the highest degree of sequence conservation and colinear arrangement of genes with orange-spotted grouper iridovirus (OSGIV) and rock bream iridovirus (RBIV). The pairwise nucleotide identities are 99.49% between RBIV-C1 and OSGIV and 98.69% between RBIV-C1 and RBIV. Compared to OSGIV, RBIV-C1 contains 11 insertions, 13 deletions, and 103 single nucleotide mutations. Whole-genome transcription analysis showed that following experimental infection of rock bream with RBIV-C1, all but 1 of the 119 ORFs were expressed at different time points and clustered into 3 hierarchical groups based on their expression patterns. These results provide new insights into the genetic nature and gene expression features of megalocytiviruses.


Assuntos
Regulação Viral da Expressão Gênica/fisiologia , Iridoviridae/genética , Animais , Genoma Viral , Mutação , Transcriptoma
10.
EBioMedicine ; 66: 103314, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33813142

RESUMO

BACKGROUND: Nucleic acids are potent stimulators of type I interferon (IFN-I) and antiviral defense, but may also promote pathological inflammation. A range of diseases are characterized by elevated IFN-I, including systemic lupus erythematosus (lupus). The DNA-activated cGAS-STING pathway is a major IFN-I-inducing pathway, and activation of signaling is dependent on trafficking of STING from the ER to the Golgi. METHODS: Here we used cell culture systems, a mouse lupus model, and material from lupus patients, to explore the mode of action of a STING antagonistic peptide, and its ability to modulate disease processes. FINDINGS: We report that the peptide ISD017 selectively inhibits all known down-stream activities of STING, including IFN-I, inflammatory cytokines, autophagy, and apoptosis. ISD017 blocks the essential trafficking of STING from the ER to Golgi through a mechanism dependent on the STING ER retention factor STIM1. Importantly, ISD017 blocks STING activity in vivo and ameliorates disease development in a mouse model for lupus. Finally, ISD017 treatment blocks pathological cytokine responses in cells from lupus patients with elevated IFN-I levels. INTERPRETATION: These data hold promise for beneficial use of STING-targeting therapy in lupus. FUNDING: The Novo Nordisk Foundation, The European Research Council, The Lundbeck Foundation, European Union under the Horizon 2020 Research, Deutsche Forschungsgemeinschaft, Chulalongkorn University.


Assuntos
Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Lúpus Eritematoso Sistêmico/etiologia , Lúpus Eritematoso Sistêmico/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Suscetibilidade a Doenças , Vesículas Extracelulares/metabolismo , Expressão Gênica , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Knockout , Transporte Proteico/efeitos dos fármacos
11.
Dev Comp Immunol ; 103: 103531, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31668931

RESUMO

MicroRNAs (miRNAs) are post-transcriptional regulators that play vital roles in diverse physiological processes including immunity. In this study, we investigated the regulatory mechanism and function of a novel Japanese flounder (Paralichthys olivaceus) miRNA, pol-miR-3p-2. pol-miR-3p-2 was responsive in expression to the infection of the bacterial pathogen Edwardsiella tarda. pol-miR-3p-2 negatively regulated the expression of p53 through interaction with the 3'UTR of p53. Overexpression of pol-miR-3p-2 promoted autophagy, resulting in augmented production of LC3-II, while knockdown of p53 increased the level of beclin, a key factor of autophagy. In vivo and in vitro studies showed that E. tarda infection induced autophagy in flounder, and pol-miR-3p-2 inhibited the infectivity of E. tarda. Together these results indicate that pol-miR-3p-2 regulates autophagy through the target gene p53, thus revealing a regulatory link between p53 and autophagy in teleost, and that pol-miR-3p-2 plays an important role in the immune defense against E. tarda.


Assuntos
Infecções por Enterobacteriaceae/veterinária , Doenças dos Peixes/imunologia , Proteínas de Peixes/imunologia , Linguado/imunologia , MicroRNAs/imunologia , Animais , Autofagia/fisiologia , Edwardsiella tarda , Infecções por Enterobacteriaceae/imunologia , Proteínas de Peixes/metabolismo , Linguado/genética , Linguado/metabolismo , Regulação da Expressão Gênica/imunologia , MicroRNAs/metabolismo , Proteína Supressora de Tumor p53/imunologia , Proteína Supressora de Tumor p53/metabolismo
12.
J Exp Med ; 217(7)2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32383759

RESUMO

Herpes simplex virus (HSV) is the main cause of viral encephalitis in the Western world, and the type I interferon (IFN) system is important for antiviral control in the brain. Here, we have compared Ifnb induction in mixed murine brain cell cultures by a panel of HSV1 mutants, each devoid of one mechanism to counteract the IFN-stimulating cGAS-STING pathway. We found that a mutant lacking the deubiquitinase (DUB) activity of the VP1-2 protein induced particularly strong expression of Ifnb and IFN-stimulated genes. HSV1 ΔDUB also induced elevated IFN expression in murine and human microglia and exhibited reduced viral replication in the brain. This was associated with increased ubiquitination of STING and elevated phosphorylation of STING, TBK1, and IRF3. VP1-2 associated directly with STING, leading to its deubiquitination. Recruitment of VP1-2 to STING was dependent on K150 of STING, which was ubiquitinated by TRIM32. Thus, the DUB activity of HSV1 VP1-2 is a major viral immune-evasion mechanism in the brain.


Assuntos
Encéfalo/virologia , Enzimas Desubiquitinantes/metabolismo , Herpesvirus Humano 1/metabolismo , Interferon Tipo I/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Virais/metabolismo , Animais , Encéfalo/patologia , Células Cultivadas , Citoplasma/metabolismo , DNA Viral/metabolismo , Células HEK293 , Humanos , Lisina/metabolismo , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Mutação/genética , Nucleotidiltransferases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Ubiquitina/metabolismo , Ubiquitinação , Replicação Viral/fisiologia
13.
J Interferon Cytokine Res ; 39(4): 191-204, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30855198

RESUMO

Incoming viruses challenge the cell with diverse foreign molecules, which need to be sensed quickly to initiate immune responses and to remove the viral components. In this study, we investigate the cellular requirements for sensing and degradation of incoming viral DNA and capsids during herpes simplex virus type 1 (HSV-1) infections. Using click chemistry labeling of the viral genome, we found that HSV-1 DNA was released from a subset of capsids into the cytosol early in infection. By next-generation sequencing of cyclic GMP-AMP (cGAMP) synthase (cGAS)-bound DNA from HSV-1-infected cells, we show that HSV-1 DNA was bound by the cytosolic DNA sensor cGAS. Activation of cGAS enzymatic activity by viral DNA did not require proteasomal activity, indicating that viral DNA release into the cytosol is not proteasome-dependent. However, induction of interferon (IFN)-ß expression was blocked by inhibition of the proteasome, suggesting a contribution of the proteasome to IFN-ß induction through the cGAS-stimulator of interferon genes pathway. Viral DNA was cleared from the cytosol within few hours, in a manner dependent on TREX1 and a cGAS-dependent process. Capsid material in the cytoplasm was also degraded rapidly. This was partially blocked by treatment with a proteasome inhibitor. This treatment led to accumulation of DNA-containing viral capsids near the nucleus and reduced nuclear entry of viral DNA. Thus, cells infected with HSV-1 use a panel of mechanisms to eliminate viral DNA and capsids. This represents a barrier for establishment of infection and potentially enables the host to gear the IFN-ß response to a level required for antiviral defense without causing immunopathology.


Assuntos
Capsídeo/imunologia , DNA Viral/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Animais , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Células Vero , Replicação Viral/genética , Replicação Viral/imunologia
14.
Nat Microbiol ; 4(4): 701-713, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30804548

RESUMO

The innate immune system is crucial for eventual control of infections, but may also contribute to pathology. Listeria monocytogenes is an intracellular Gram-positive bacteria and a major cause of food-borne disease. However, important knowledge on the interactions between L. monocytogenes and the immune system is still missing. Here, we report that Listeria DNA is sorted into extracellular vesicles (EVs) in infected cells and delivered to bystander cells to stimulate the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway. This was also observed during infections with Francisella tularensis and Legionella pneumophila. We identify the multivesicular body protein MVB12b as a target for TANK-binding kinase 1 phosphorylation, which is essential for the sorting of DNA into EVs and stimulation of bystander cells. EVs from Listeria-infected cells inhibited T-cell proliferation, and primed T cells for apoptosis. Collectively, we describe a pathway for EV-mediated delivery of foreign DNA to bystander cells, and suggest that intracellular bacteria exploit this pathway to impair antibacterial defence.


Assuntos
Vesículas Extracelulares/microbiologia , Listeria monocytogenes/fisiologia , Listeriose/metabolismo , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Vesículas Extracelulares/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Listeria monocytogenes/genética , Listeriose/genética , Listeriose/microbiologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Nucleotídeos Cíclicos , Nucleotidiltransferases/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas de Transporte Vesicular/genética
15.
Sci Rep ; 6: 28354, 2016 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-27311682

RESUMO

Megalocytivirus is a DNA virus that is highly infectious in a wide variety of marine and freshwater fish, including Japanese flounder (Paralichthys olivaceus), a flatfish that is farmed worldwide. However, the infection mechanism of megalocytivirus remains largely unknown. In this study, we investigated the function of a flounder microRNA, pol-miR-731, in virus-host interaction. We found that pol-miR-731 was induced in expression by megalocytivirus and promoted viral replication at the early infection stage. In vivo and in vitro studies revealed that pol-miR-731 (i) specifically suppresses the expression of interferon regulatory factor 7 (IRF7) and cellular tumor antigen p53 in a manner that depended on the integrity of the pol-miR-731 complementary sequences in the 3' untranslated regions of IRF7 and p53, (ii) disrupts megalocytivirus-induced Type I interferon response through IRF7, (iii) inhibits megalocytivirus-induced splenocyte apoptosis and cell cycle arrest through p53. Furthermore, overexpression of IRF7 and p53 abolished both the inhibitory effects of pol-miR-731 on these biological processes and its stimulatory effect on viral replication. These results disclosed a novel evasion mechanism of megalocytivirus mediated by a host miRNA. This study also provides the first evidence that a virus-induced host miRNA can facilitate viral infection by simultaneously suppressing several antiviral pathways.


Assuntos
Linguado/virologia , Fator Regulador 7 de Interferon/genética , Iridoviridae/fisiologia , MicroRNAs/genética , Proteína Supressora de Tumor p53/genética , Animais , Apoptose , Pontos de Checagem do Ciclo Celular , Proteínas de Peixes/genética , Linguado/genética , Linguado/imunologia , Interações Hospedeiro-Patógeno , Interferon Tipo I/genética , RNA Viral/genética , Replicação Viral
16.
Dev Comp Immunol ; 53(1): 96-104, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26162512

RESUMO

Prothymosin alpha (ProTα) is a small protein that in mammals is known to participate in diverse biological processes including immunomodulation. In teleost, the immunological function of ProTα is unknown. In the current study, we investigated the expression and function of the ProTα (named CsProTα) from the teleost fish tongue sole (Cynoglossus semilaevis). We found that CsProTα expression was abundant in immune relevant tissues and upregulated by megalocytivirus infection. Immunoblot detected secretion of CsProTα by peripheral blood leukocytes. Recombinant CsProTα (rCsProTα) as well as the C-terminal 11-residue (Ct11) were able to bind head kidney monocytes (HKM) and induce immune gene expression; however, the induction patterns caused by rCsProTα and Ct11 differed considerably. When introduced in vivo, rCsProTα and Ct11 significantly reduced megalocytivirus infection in fish tissues, whereas rCsProTα antibody significantly promoted viral replication. Blocking of Myd88 activity abolished the virus-inhibitory effect of rCsProTα but not Ct11. Taken together, these results demonstrate for the first time that both the intact protein and the C-terminal segment of a teleost ProTα can act like cytokines and induce antiviral immunity via, however, distinct signaling pathways that differ in the requirement of Myd88.


Assuntos
Doenças dos Peixes/imunologia , Linguados/imunologia , Iridoviridae/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Precursores de Proteínas/metabolismo , Timosina/análogos & derivados , Sequência de Aminoácidos , Animais , Sequência de Bases , Citocinas/imunologia , Doenças dos Peixes/virologia , Linguados/virologia , Dados de Sequência Molecular , Fator 88 de Diferenciação Mieloide/antagonistas & inibidores , Precursores de Proteínas/antagonistas & inibidores , Precursores de Proteínas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Alinhamento de Sequência , Análise de Sequência de DNA , Transdução de Sinais/imunologia , Timosina/antagonistas & inibidores , Timosina/genética , Timosina/metabolismo
17.
PLoS One ; 10(3): e0121282, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25815484

RESUMO

Megalocytivirus is a DNA virus with a broad host range among teleost fish. Although the complete genome sequences of a number of megalocytivirus isolates have been reported, the functions of most of the genes of this virus are unknown. In this study, we selected two megalocytivirus immunogens, P247 and P523, which were expressed during host infection and, when in the form of DNA vaccines (pCN247 and pCN523 respectively), elicited strong protectivity against lethal megalocytivirus challenge in a turbot (Scophthalmus maximus) model. Compared to control fish, fish vaccinated with pCN247 and pCN523 exhibited drastically reduced viral loads in tissues and high levels of survival rates. Immune response analysis showed that pCN247 and pCN523 (i) induced production of specific serum antibodies, (ii) caused generation of cytotoxic immune cells and specific memory immune cells that responded to secondary antigen stimulation, and (iii) upregulated the expression of genes involved in innate and adaptive immunity. To examine the potential role of P247 and P523 in viral infection, the expression of P247 and P523 was knocked down by siRNA. Subsequent in vivo infection study showed that P247 and P523 knockdown significantly impaired viral replication. Furthermore, whole-genome transcriptome analysis revealed that P247 and P523 knockdown altered the expression profiles of 26 and 41 viral genes, respectively, putatively participating in diverse aspects of viral infection. Taken together, these results indicate that P247 and P523 induce protective immunity in teleost and play fundamental roles essential to viral replication. These observations provide the first evidence that suggests a likely link between the protectivity of viral immunogens and their biological significance in viral replication.


Assuntos
Linguados/virologia , Interações Hospedeiro-Patógeno/genética , Iridoviridae/genética , Proteínas Virais/genética , Animais , Sequência de Bases , Doenças dos Peixes/genética , Doenças dos Peixes/imunologia , Linguados/genética , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade/genética , Iridoviridae/imunologia , Iridoviridae/patogenicidade , Proteínas Virais/imunologia , Viroses/genética , Viroses/imunologia , Replicação Viral/genética
18.
PLoS One ; 9(11): e112918, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25393122

RESUMO

Polyinosinic:polycytidylic acid (poly(I:C)) is a ligand of toll-like receptor (TLR) 3 that has been used as an immunostimulant in humans and mice against viral diseases based on its ability to enhance innate and adapt immunity. Antiviral effect of poly(I:C) has also been observed in teleost, however, the underling mechanism is not clear. In this study, we investigated the potential and signaling mechanism of poly(I:C) as an antiviral agent in a model of Japanese flounder (Paralichthys olivaceus) infected with megalocytivirus. We found that poly(I:C) exhibited strong antiviral activity and enhanced activation of head kidney macrophages and peripheral blood leukocytes. In vivo studies showed that (i) TLR3 as well as MDA5 knockdown reduced poly(I:C)-mediated immune response and antiviral activity to significant extents; (ii) when Myd88 was overexpressed in flounder, poly(I:C)-mediated antiviral activity was significantly decreased; (iii) when Myd88 was inactivated, the antiviral effect of poly(I:C) was significantly increased. Cellular study showed that (i) the NF-κB activity induced by poly(I:C) was upregulated in Myd88-overexpressing cells and unaffected in Myd88-inactivated cells; (ii) Myd88 overexpression inhibited and upregulated the expression of poly(I:C)-induced antiviral genes and inflammatory genes respectively; (iii) Myd88 inactivation enhanced the expression of the antiviral genes induced by poly(I:C). Taken together, these results indicate that poly(I:C) is an immunostimulant with antiviral potential, and that the immune response of poly(I:C) requires TLR3 and MDA5 and is negatively regulated by Myd88 in a manner not involving NK-κB. These results provide insights to the working mechanism of poly(I:C), TLR3, and Myd88 in fish.


Assuntos
RNA Helicases DEAD-box/imunologia , Proteínas de Peixes/imunologia , Linguado/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Poli I-C/farmacologia , Receptor 3 Toll-Like/imunologia , Animais , RNA Helicases DEAD-box/genética , Infecções por Vírus de DNA/genética , Infecções por Vírus de DNA/imunologia , Infecções por Vírus de DNA/veterinária , Doenças dos Peixes/genética , Doenças dos Peixes/imunologia , Proteínas de Peixes/genética , Linguado/genética , Iridoviridae/genética , Iridoviridae/imunologia , Camundongos , Fator 88 de Diferenciação Mieloide/genética , Receptor 3 Toll-Like/genética
19.
PLoS One ; 9(3): e91324, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24621602

RESUMO

Streptococcus iniae is a Gram-positive bacterium that is reckoned one of the most severe aquaculture pathogens. It has a broad host range among farmed marine and freshwater fish and can also cause zoonotic infection in humans. Here we report for the first time the complete genome sequence as well as the host factor-induced proteomic profile of a pathogenic S. iniae strain, SF1, a serotype I isolate from diseased fish. SF1 possesses a single chromosome of 2,149,844 base pairs, which contains 2,125 predicted protein coding sequences (CDS), 12 rRNA genes, and 45 tRNA genes. Among the protein-encoding CDS are genes involved in resource acquisition and utilization, signal sensing and transduction, carbohydrate metabolism, and defense against host immune response. Potential virulence genes include those encoding adhesins, autolysins, toxins, exoenzymes, and proteases. In addition, two putative prophages and a CRISPR-Cas system were found in the genome, the latter containing a CRISPR locus and four cas genes. Proteomic analysis detected 21 secreted proteins whose expressions were induced by host serum. Five of the serum-responsive proteins were subjected to immunoprotective analysis, which revealed that two of the proteins were highly protective against lethal S. iniae challenge when used as purified recombinant subunit vaccines. Taken together, these results provide an important molecular basis for future study of S. iniae in various aspects, in particular those related to pathogenesis and disease control.


Assuntos
Antígenos de Bactérias/imunologia , Genoma Bacteriano/genética , Proteômica , Análise de Sequência , Streptococcus/genética , Streptococcus/imunologia , Animais , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Sequência de Bases , Transporte Biológico/genética , Sistemas CRISPR-Cas , Espaço Extracelular/metabolismo , Linguados/imunologia , Linguados/microbiologia , Prófagos/fisiologia , Soro/imunologia , Streptococcus/metabolismo , Streptococcus/virologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
20.
Dev Comp Immunol ; 45(1): 35-42, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24525178

RESUMO

Viperin in mammals is known to be an antiviral protein that inhibits the replication of diverse DNA and RNA viruses. In teleost, viperin homologues have been identified in a large number of species and, in some cases, are stimulated in transcription by viruses. However, the biological significance of fish viperin protein in antiviral immunity has not been investigated. In this study, we identified a viperin homologue from rock bream (Oplegnathus fasciatus) (named OfVip) and examined its expression pattern, subcellular localization, and immune effect. We found that OfVip expression occurred in eight tissues, and experimental challenge of rock bream with the viral fish pathogen megalocytivirus upregulated OfVip expression in kidney, liver, and spleen. OfVip was localized in the endoplasmic reticulum under normal physiological conditions, and viral infection induced subcellular redistribution of OfVip. Transient transfection of cultured fish cells with an OfVip-expressing plasmid caused enhanced cellular resistance against megalocytivirus challenge. Consistently, in vivo study showed that rock bream overexpressing OfVip exhibited significantly reduced viral loads in tissues following experimental infection with megalocytivirus. Furthermore, OfVip upregulated the expression of a wide range of immune genes, including those that are known to participate in antiviral immunity. Taken together, these results indicate for the first time that a teleost viperin is a virus-responsive protein that is modulated in subcellular localization by viral infection, and that viperin regulates the immune reactions of the host fish in a manner that augments resistance against viral infection.


Assuntos
Infecções por Vírus de DNA/veterinária , Doenças dos Peixes/imunologia , Proteínas de Peixes/metabolismo , Imunidade Inata , Iridoviridae/imunologia , Perciformes/imunologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Sequência Conservada , Infecções por Vírus de DNA/imunologia , Resistência à Doença , Doenças dos Peixes/virologia , Proteínas de Peixes/genética , Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno , Dados de Sequência Molecular , Especificidade de Órgãos , Perciformes/metabolismo , Perciformes/virologia , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA