Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(7): e1012379, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39037956

RESUMO

RNA helicases are involved in the innate immune response against pathogens, including bacteria and viruses; however, their mechanism in the human airway epithelial cells is still not fully understood. Here, we demonstrated that DEAH (Asp-Glu-Ala-His) box polypeptide 35 (DHX35), a member of the DExD/H (Asp-Glu-x-Asp/His)-box helicase family, boosts antiviral innate immunity in human airway epithelial cells. DHX35 knockdown attenuated the production of interferon-ß (IFN-ß), IL6, and CXCL10, whereas DHX35 overexpression increased their production. Upon stimulation, DHX35 was constitutively expressed, but it translocated from the nucleus into the cytosol, where it recognized cytosolic poly(I:C) and poly(dA:dT) via its HELICc domain. Mitochondrial antiviral signaling protein (MAVS) acted as an adaptor for DHX35 and interacted with the HELICc domain of DHX35 using amino acids 360-510. Interestingly, DHX35 interacted with retinoic acid-inducible gene 1 (RIG-I), enhanced the binding affinity of RIG-I with poly(I:C) and poly(dA:dT), and formed a signalsome with MAVS to activate interferon regulatory factor 3 (IRF3), NF-κB-p65, and MAPK signaling pathways. These results indicate that DHX35 not only acted as a cytosolic nucleic acid sensor but also synergized with RIG-I to enhance antiviral immunity in human airway epithelial cells. Our results demonstrate a novel molecular mechanism for DHX35 in RIG-I-mediated innate immunity and provide a novel candidate for drug and vaccine design to control viral infections in the human airway.


Assuntos
Proteína DEAD-box 58 , RNA Helicases DEAD-box , Imunidade Inata , Receptores Imunológicos , Humanos , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/imunologia , RNA Helicases DEAD-box/metabolismo , RNA Helicases DEAD-box/imunologia , Receptores Imunológicos/metabolismo , Poli I-C/imunologia , Poli I-C/farmacologia , RNA Helicases/metabolismo , RNA Helicases/imunologia , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Células HEK293
2.
Cell Rep ; 38(10): 110503, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35235832

RESUMO

Natural killer (NK) cells are innate immune cells that contribute to host defense against virus infections. NK cells respond to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro and are activated in patients with acute coronavirus disease 2019 (COVID-19). However, by which mechanisms NK cells detect SARS-CoV-2-infected cells remains largely unknown. Here, we show that the Non-structural protein 13 of SARS-CoV-2 encodes for a peptide that is presented by human leukocyte antigen E (HLA-E). In contrast with self-peptides, the viral peptide prevents binding of HLA-E to the inhibitory receptor NKG2A, thereby rendering target cells susceptible to NK cell attack. In line with these observations, NKG2A-expressing NK cells are particularly activated in patients with COVID-19 and proficiently limit SARS-CoV-2 replication in infected lung epithelial cells in vitro. Thus, these data suggest that a viral peptide presented by HLA-E abrogates inhibition of NKG2A+ NK cells, resulting in missing self-recognition.


Assuntos
COVID-19 , Antígenos de Histocompatibilidade Classe I , Células Matadoras Naturais , Metiltransferases , Subfamília C de Receptores Semelhantes a Lectina de Células NK , RNA Helicases , SARS-CoV-2 , Proteínas não Estruturais Virais , COVID-19/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Células Matadoras Naturais/imunologia , Metiltransferases/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Peptídeos/metabolismo , RNA Helicases/imunologia , Proteínas não Estruturais Virais/imunologia , Antígenos HLA-E
3.
Cell Rep ; 38(10): 110434, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35263596

RESUMO

Type I interferons (IFN-I) are essential to establish antiviral innate immunity. Unanchored (or free) polyubiquitin (poly-Ub) has been shown to regulate IFN-I responses. However, few unanchored poly-Ub interactors are known. To identify factors regulated by unanchored poly-Ub in a physiological setting, we developed an approach to isolate unanchored poly-Ub from lung tissue. We identified the RNA helicase DHX16 as a potential pattern recognition receptor (PRR). Silencing of DHX16 in cells and in vivo diminished IFN-I responses against influenza virus. These effects extended to members of other virus families, including Zika and SARS-CoV-2. DHX16-dependent IFN-I production requires RIG-I and unanchored K48-poly-Ub synthesized by the E3-Ub ligase TRIM6. DHX16 recognizes a signal in influenza RNA segments that undergo splicing and requires its RNA helicase motif for direct, high-affinity interactions with specific viral RNAs. Our study establishes DHX16 as a PRR that partners with RIG-I for optimal activation of antiviral immunity requiring unanchored poly-Ub.


Assuntos
Proteína DEAD-box 58 , Interferon Tipo I , RNA Helicases , RNA Viral , Receptores Imunológicos , Infecção por Zika virus , Zika virus , COVID-19 , Proteína DEAD-box 58/imunologia , Humanos , Imunidade Inata , Interferon Tipo I/imunologia , RNA Helicases/imunologia , Receptores Imunológicos/imunologia , SARS-CoV-2 , Proteínas com Motivo Tripartido , Zika virus/genética , Infecção por Zika virus/imunologia
4.
Signal Transduct Target Ther ; 7(1): 22, 2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35075101

RESUMO

As a highly pathogenic human coronavirus, SARS-CoV-2 has to counteract an intricate network of antiviral host responses to establish infection and spread. The nucleic acid-induced stress response is an essential component of antiviral defense and is closely related to antiviral innate immunity. However, whether SARS-CoV-2 regulates the stress response pathway to achieve immune evasion remains elusive. In this study, SARS-CoV-2 NSP5 and N protein were found to attenuate antiviral stress granule (avSG) formation. Moreover, NSP5 and N suppressed IFN expression induced by infection of Sendai virus or transfection of a synthetic mimic of dsRNA, poly (I:C), inhibiting TBK1 and IRF3 phosphorylation, and restraining the nuclear translocalization of IRF3. Furthermore, HEK293T cells with ectopic expression of NSP5 or N protein were less resistant to vesicular stomatitis virus infection. Mechanistically, NSP5 suppressed avSG formation and disrupted RIG-I-MAVS complex to attenuate the RIG-I-mediated antiviral immunity. In contrast to the multiple targets of NSP5, the N protein specifically targeted cofactors upstream of RIG-I. The N protein interacted with G3BP1 to prevent avSG formation and to keep the cofactors G3BP1 and PACT from activating RIG-I. Additionally, the N protein also affected the recognition of dsRNA by RIG-I. This study revealed the intimate correlation between SARS-CoV-2, the stress response, and innate antiviral immunity, shedding light on the pathogenic mechanism of COVID-19.


Assuntos
Proteases 3C de Coronavírus/genética , Proteínas do Nucleocapsídeo de Coronavírus/genética , Proteína DEAD-box 58/genética , DNA Helicases/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , RNA Helicases/genética , Proteínas com Motivo de Reconhecimento de RNA/genética , Proteínas de Ligação a RNA/genética , Receptores Imunológicos/genética , SARS-CoV-2/genética , Grânulos de Estresse/genética , Animais , Chlorocebus aethiops , Proteases 3C de Coronavírus/imunologia , Proteínas do Nucleocapsídeo de Coronavírus/imunologia , Proteína DEAD-box 58/imunologia , DNA Helicases/imunologia , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Evasão da Resposta Imune , Fosfoproteínas/genética , Fosfoproteínas/imunologia , Poli I-C/farmacologia , Proteínas de Ligação a Poli-ADP-Ribose/imunologia , Ligação Proteica , RNA Helicases/imunologia , Proteínas com Motivo de Reconhecimento de RNA/imunologia , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/imunologia , Proteínas de Ligação a RNA/imunologia , Receptores Imunológicos/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Vírus Sendai/genética , Vírus Sendai/imunologia , Transdução de Sinais , Grânulos de Estresse/efeitos dos fármacos , Grânulos de Estresse/imunologia , Grânulos de Estresse/virologia , Células Vero , Vesiculovirus/genética , Vesiculovirus/imunologia
5.
Proc Natl Acad Sci U S A ; 117(27): 15778-15788, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32571931

RESUMO

RIG-I, MDA5, and LGP2 comprise the RIG-I-like receptors (RLRs). RIG-I and MDA5 are essential pathogen recognition receptors sensing viral infections while LGP2 has been described as both RLR cofactor and negative regulator. After sensing and binding to viral RNA, including double-stranded RNA (dsRNA), RIG-I and MDA5 undergo cytosol-to-membrane relocalization to bind and signal through the MAVS adaptor protein on intracellular membranes, thus directing downstream activation of IRF3 and innate immunity. Here, we report examination of the dynamic subcellular localization of all three RLRs within the intracellular response to dsRNA and RNA virus infection. Observations from high resolution biochemical fractionation and electron microscopy, coupled with analysis of protein interactions and IRF3 activation, show that, in resting cells, microsome but not mitochondrial fractions harbor the central components to initiate innate immune signaling. LGP2 interacts with MAVS in microsomes, blocking the RIG-I/MAVS interaction. Remarkably, in response to dsRNA treatment or RNA virus infection, LGP2 is rapidly released from MAVS and redistributed to mitochondria, temporally correlating with IRF3 activation. We reveal that IRF3 activation does not take place on mitochondria but instead occurs at endoplasmic reticulum (ER)-derived membranes. Our observations suggest ER-derived membranes as key RLR signaling platforms controlled through inhibitory actions of LGP2 binding to MAVS wherein LGP2 translocation to mitochondria releases MAVS inhibition to facilitate RLR-mediated signaling of innate immunity.


Assuntos
Proteína DEAD-box 58/genética , Helicase IFIH1 Induzida por Interferon/genética , RNA Helicases/genética , Viroses/imunologia , Proteína DEAD-box 58/imunologia , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Fator Regulador 3 de Interferon/genética , Helicase IFIH1 Induzida por Interferon/imunologia , Mitocôndrias/genética , Mitocôndrias/imunologia , RNA Helicases/imunologia , RNA de Cadeia Dupla/genética , RNA Viral/genética , RNA Viral/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Viroses/genética , Viroses/virologia
6.
Immunology ; 160(1): 90-102, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32128816

RESUMO

Multifunctional interleukin 10 (IL10)+ Th1 cells have been implicated in favorable evolution of many infectious diseases, promoting an efficacious immune response while limiting immunopathology. Here, we investigated the presence of multifunctional CD4+ and CD8+ T-cells that expressed interferon gamma (IFNγ), IL10 and tumor necrosis factor (TNF), or its combinations during dengue infection. Peripheral blood mononuclear cells (PBMCs) from outpatients with dengue (mild dengue forms) and hospitalized patients (or patients with dengue with warning signs and severe dengue) were cultured in the presence of envelope (ENV) or NS3 peptide libraries of DENV during critical (hospitalization period) and convalescence phases. The production of IFNγ, IL10 and TNF by CD4+ and CD8+ T-cells was assessed by flow cytometry. Our data show that patients with mild dengue, when compared with patients with dengue with warning signs and severe dengue, presented higher frequencies of multifunctional T-cells like NS3-specific IFNγ/IL10-producing CD4+ T-cells in critical phase and NS3- and ENV-specific CD8+ T-cells producing IFNγ/IL10. In addition, NS3-specific CD8+ T-cells producing high levels of IFNγ/TNF and IFNγ/TNF/IL10 were also observed in the mild dengue group. We observed that multifunctional T-cells produced higher levels of cytokines as measured by intracellular content when compared with single producer T-cells. Importantly, multifunctional CD4+ and CD8+ T-cells producing IFNγ, TNF and IL10 simultaneously displayed positive correlation with platelet levels, suggesting a protective role of this population. The presence of IL10+ Th1 and IL10+ Tc1 multifunctional cells was associated with mild dengue presentation, suggesting that these cells play a role in clinical evolution of dengue infection.


Assuntos
Dengue/diagnóstico , Dengue/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Adolescente , Adulto , Idoso , Antígenos Virais/imunologia , Brasil , Estudos de Casos e Controles , Dengue/sangue , Vírus da Dengue/imunologia , Feminino , Voluntários Saudáveis , Humanos , Interferon gama/metabolismo , Interleucina-10/metabolismo , Masculino , Pessoa de Meia-Idade , Cultura Primária de Células , RNA Helicases/imunologia , Serina Endopeptidases/imunologia , Índice de Gravidade de Doença , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas não Estruturais Virais/imunologia , Adulto Jovem
7.
Infect Genet Evol ; 78: 104106, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31706079

RESUMO

Japanese encephalitis (JE) is a serious leading health complication emerging expansively that has severely affected the survival rate of human beings. This fatal disease is caused by JE Virus (JEV). The current study was carried out for designing a multi-epitope loaded peptide vaccine to prevent JEV. Based on reverse vaccinology and in silico approaches, octapeptide B-cell and hexapeptide T-cell epitopes belonging to five proteins, viz. E, prM, NS1, NS3 and NS5 of JEV were determined. Hydrophilicity, antigenicity, immunogenicity and aliphatic amino acids of the epitopes were estimated. Further, the epitopes were analyzed for different physicochemical parameters, e.g. total net charges, amino acid composition and Boman index. Out of all the epitopes, a total of four T-cell epitopes namely KRADSS, KRSRRS, SKRSRR and KECPDE and one B-cell epitope i.e. PKPCSKGD were found to have potential for raising immunity in human against the pathogen. Taking into account the outcome of this study, the pharmaceutical industries could initiate efforts to combine the identified epitopes together with adjuvant or carrier protein to develop a multi-epitope-loaded peptide vaccine against JEV. The peptide vaccine, being cost effective, could be administered as a prophylactic measure and in JEV infected individuals to combat the spread of this virus in human population. However, prior to administration into human beings, the vaccine must pass through several clinical trials.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/imunologia , Vacinas contra Encefalite Japonesa/imunologia , Aminoácidos/análise , Linfócitos B/imunologia , Epitopos/química , Epitopos/imunologia , Imunogenicidade da Vacina , Peptídeos/imunologia , RNA Helicases/imunologia , Serina Endopeptidases/imunologia , Proteínas não Estruturais Virais/imunologia
8.
Immun Inflamm Dis ; 7(4): 276-285, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31568656

RESUMO

INTRODUCTION: Although the role of dengue virus (DENV)-specific T cells in the pathogenesis of acute dengue infection is emerging, the functionality of virus-specific T cells associated with milder clinical disease has not been well studied. We sought to investigate how the functionality of DENV-NS3 and DENV-NS5 protein-specific T cells differ in patients with dengue fever (DF) and dengue hemorrhagic fever (DHF). METHODS: Using intracellular cytokine assays, we assessed the production of interferon γ (IFNγ), tumor necrosis factor-α (TNF-α), macrophage inflammatory protein-1ß (MIP-1ß), and CD107a expression in adult patients with acute DF (n = 21) and DHF (n = 22). RESULTS: Quadruple cytokine-producing, polyfunctional DENV-NS3- and DENV-NS5-specific T cells were more frequent in those with DF when compared to those with DHF. While DENV-NS3- and DENV-NS5-specific T cells in patients with DF expressed IFNγ > TNF-α > MIP-ß > CD107a, T cells of those with DHF predominantly expressed CD107a > MIP-1ß > IFNγ > TNF-α. Overall production of IFNγ or TNF-α by DENV-NS3- and DENV-NS5-specific T cells was significantly higher in patients with DF. The majority of NS3-specific T cells in patients with DF (78.6%) and DHF (68.9%) were single-cytokine producers; 76.6% of DENV-NS5-specific T cells in those with DF and 77.1% of those with DHF, produced only a single cytokine. However, no significant association was found with polyfunctional T-cell responses and the degree of viraemia. CONCLUSIONS: Our results suggest that the functional phenotype of DENV-specific T cells are likely to associate with clinical disease severity.


Assuntos
Citocinas/imunologia , Dengue/imunologia , Imunidade Celular , Linfócitos T/imunologia , Proteínas não Estruturais Virais/imunologia , Doença Aguda , Adulto , Dengue/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Helicases/imunologia , Serina Endopeptidases/imunologia , Índice de Gravidade de Doença , Linfócitos T/patologia
9.
mBio ; 10(3)2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31213553

RESUMO

The integrated stress response (ISR) is a cellular response system activated upon different types of stresses, including viral infection, to restore cellular homeostasis. However, many viruses manipulate this response for their own advantage. In this study, we investigated the association between murine norovirus (MNV) infection and the ISR and demonstrate that MNV regulates the ISR by activating and recruiting key ISR host factors. We observed that during MNV infection, there is a progressive increase in phosphorylated eukaryotic initiation factor 2α (p-eIF2α), resulting in the suppression of host translation, and yet MNV translation still progresses under these conditions. Interestingly, the shutoff of host translation also impacts the translation of key signaling cytokines such as beta interferon, interleukin-6, and tumor necrosis factor alpha. Our subsequent analyses revealed that the phosphorylation of eIF2α was mediated via protein kinase R (PKR), but further investigation revealed that PKR activation, phosphorylation of eIF2α, and translational arrest were uncoupled during infection. We further observed that stress granules (SGs) are not induced during MNV infection and that MNV can restrict SG nucleation and formation. We observed that MNV recruited the key SG nucleating protein G3BP1 to its replication sites and intriguingly the silencing of G3BP1 negatively impacts MNV replication. Thus, it appears that MNV utilizes G3BP1 to enhance replication but equally to prevent SG formation, suggesting an anti-MNV property of SGs. Overall, this study highlights MNV manipulation of SGs, PKR, and translational control to regulate cytokine translation and to promote viral replication.IMPORTANCE Viruses hijack host machinery and regulate cellular homeostasis to actively replicate their genome, propagate, and cause disease. In retaliation, cells possess various defense mechanisms to detect, destroy, and clear infecting viruses, as well as signal to neighboring cells to inform them of the imminent threat. In this study, we demonstrate that the murine norovirus (MNV) infection stalls host protein translation and the production of antiviral and proinflammatory cytokines. However, virus replication and protein translation still ensue. We show that MNV further prevents the formation of cytoplasmic RNA granules, called stress granules (SGs), by recruiting the key host protein G3BP1 to the MNV replication complex, a recruitment that is crucial to establishing and maintaining virus replication. Thus, MNV promotes immune evasion of the virus by altering protein translation. Together, this evasion strategy delays innate immune responses to MNV infection and accelerates disease onset.


Assuntos
Infecções por Caliciviridae/imunologia , Grânulos Citoplasmáticos/virologia , DNA Helicases/imunologia , Fator de Iniciação 2 em Eucariotos/imunologia , Evasão da Resposta Imune , Proteínas de Ligação a Poli-ADP-Ribose/imunologia , RNA Helicases/imunologia , Proteínas com Motivo de Reconhecimento de RNA/imunologia , eIF-2 Quinase/imunologia , Animais , Grânulos Citoplasmáticos/imunologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Camundongos , Fosforilação , Biossíntese de Proteínas , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
10.
Gene ; 695: 18-25, 2019 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-30738967

RESUMO

Dengue is a severe emerging arthropod borne viral disease occurring globally. Around two fifths of the world's population, or up to 3.9 billion people, are at a risk of dengue infection. Infection induces a life-long protective immunity to the homologous serotype but confers only partial and transient protection against subsequent infection caused by other serotypes. Thus, there is a need for a vaccine which is capable of providing a life- long protection against all the serotypes of dengue virus. In our study, comparative genomics of Dengue virus (DENV) was conducted to explore potential candidates for novel vaccine targets. From our analysis we successfully found 100% conserved epitopes in Envelope protein (RCPTQGE); NS3 (SAAQRRGR, PGTSGSPI); NS4A (QRTPQDNQL); NS4B (LQAKATREAQKRA) and NS5 proteins (QRGSGQV) in all DENV serotypes. Some serotype specific conserved motifs were also found in NS1, NS5, Capsid, PrM and Envelope proteins. Using comparative genomics and immunoinformatics approach, we could find conserved epitopes which can be explored as peptide vaccine candidates to combat dengue worldwide. Serotype specific epitopes can also be exploited for rapid diagnostics. All ten proteins are explored to find the conserved epitopes in DENV serotypes, thus making it the most extensively studied viral genome so far.


Assuntos
Vírus da Dengue/imunologia , Dengue/prevenção & controle , Epitopos/imunologia , Vacinas/imunologia , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/genética , Anticorpos Antivirais/imunologia , Dengue/imunologia , Vírus da Dengue/genética , Vírus da Dengue/patogenicidade , Epitopos/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , RNA Helicases/genética , RNA Helicases/imunologia , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Sorogrupo , Vacinas/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia
11.
Genome Res ; 28(8): 1136-1146, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29970450

RESUMO

Long interspersed nuclear element-1 (LINE-1 or L1) retrotransposons are normally suppressed in somatic tissues mainly due to DNA methylation and antiviral defense. However, the mechanism to suppress L1s may be disrupted in cancers, thus allowing L1s to act as insertional mutagens and cause genomic rearrangement and instability. Whereas the frequency of somatic L1 insertions varies greatly among individual tumors, much remains to be learned about underlying genetic, cellular, or environmental factors. Here, we report multiple correlates of L1 activity in stomach, colorectal, and esophageal tumors through an integrative analysis of cancer whole-genome and matched RNA-sequencing profiles. Clinical indicators of tumor progression, such as tumor grade and patient age, showed positive association. A potential L1 expression suppressor, TP53, was mutated in tumors with frequent L1 insertions. We characterized the effects of somatic L1 insertions on mRNA splicing and expression, and demonstrated an increased risk of gene disruption in retrotransposition-prone cancers. In particular, we found that a cancer-specific L1 insertion in an exon of MOV10, a key L1 suppressor, caused exon skipping and decreased expression of the affected allele due to nonsense-mediated decay in a tumor with a high L1 insertion load. Importantly, tumors with high immune activity, for example, those associated with Epstein-Barr virus infection or microsatellite instability, tended to carry a low number of L1 insertions in genomes with high expression levels of L1 suppressors such as APOBEC3s and SAMHD1 Our results indicate that cancer immunity may contribute to genome stability by suppressing L1 retrotransposition in gastrointestinal cancers.


Assuntos
Neoplasias Gastrointestinais/genética , Elementos Nucleotídeos Longos e Dispersos/genética , Retroelementos/genética , Proteína Supressora de Tumor p53/genética , Desaminase APOBEC-3G/genética , Linhagem Celular Tumoral , Metilação de DNA/genética , Neoplasias Gastrointestinais/imunologia , Neoplasias Gastrointestinais/patologia , Regulação Neoplásica da Expressão Gênica/imunologia , Instabilidade Genômica/genética , Instabilidade Genômica/imunologia , Humanos , Elementos Nucleotídeos Longos e Dispersos/imunologia , Mutagênese Insercional/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , RNA Helicases/genética , RNA Helicases/imunologia , Splicing de RNA/genética , Retroelementos/imunologia
12.
Cancer Res ; 78(15): 4292-4302, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29853604

RESUMO

Presence of cytotoxic CD8+ T cells (CTL) in tumor microenvironments (TME) is critical for the effectiveness of immune therapies and patients' outcome, whereas regulatory T(reg) cells promote cancer progression. Immune adjuvants, including double-stranded (ds)RNAs, which signal via Toll-like receptor-3 (TLR3) and helicase (RIG-I/MDA5) pathways, all induce intratumoral production of CTL-attractants, but also Treg attractants and suppressive factors, raising the question of whether induction of these opposing groups of immune mediators can be separated. Here, we use human tumor explant cultures and cell culture models to show that the (ds) RNA Sendai Virus (SeV), poly-I:C, and rintatolimod (poly-I:C12U) all activate the TLR3 pathway involving TRAF3 and IRF3, and induce IFNα, ISG-60, and CXCL10 to promote CTL chemotaxis to ex vivo-treated tumors. However, in contrast with SeV and poly I:C, rintatolimod did not activate the MAVS/helicase pathway, thus avoiding NFκB- and TNFα-dependent induction of COX2, COX2/PGE2-dependent induction of IDO, IL10, CCL22, and CXCL12, and eliminating Treg attraction. Induction of CTL-attractants by either poly I:C or rintatolimod was further enhanced by exogenous IFNα (enhancer of TLR3 expression), whereas COX2 inhibition enhanced the response to poly-I:C only. Our data identify the helicase/NFκB/TNFα/COX2 axis as the key suppressive pathway of dsRNA signaling in human TME and suggest that selective targeting of TLR3 or elimination of NFκB/TNFα/COX2-driven suppression may allow for selective enhancement of type-1 immunity.Significance: This study characterizes two different poly-I:C-induced signaling pathways in their induction of immunostimulatory and suppressive factors and suggests improved ways to reprogram the TME to enhance the antitumor efficacy of immunotherapies. Cancer Res; 78(15); 4292-302. ©2018 AACR.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Tolerância Imunológica/imunologia , Inflamação/imunologia , NF-kappa B/metabolismo , RNA Helicases/metabolismo , RNA de Cadeia Dupla/metabolismo , Microambiente Tumoral/imunologia , Adulto , Idoso , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Ciclo-Oxigenase 2/imunologia , Feminino , Humanos , Inflamação/metabolismo , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , NF-kappa B/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , RNA Helicases/imunologia , RNA de Cadeia Dupla/imunologia , Ratos , Transdução de Sinais/imunologia , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
13.
PLoS Pathog ; 14(6): e1007135, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29958302

RESUMO

The RNA helicase LGP2 (Laboratory of Genetics and Physiology 2) is a non-signaling member of the retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), whose pivotal role on innate immune responses against RNA viruses is being increasingly uncovered. LGP2 is known to work in synergy with melanoma differentiation-associated gene 5 (MDA5) to promote the antiviral response induced by picornavirus infection. Here, we describe the activity of the foot-and-mouth disease virus (FMDV) Leader protease (Lpro) targeting LGP2 for cleavage. When LGP2 and Lpro were co-expressed, cleavage products were observed in an Lpro dose-dependent manner while co-expression with a catalytically inactive Lpro mutant had no effect on LGP2 levels or pattern. We further show that Lpro localizes and immunoprecipitates with LGP2 in transfected cells supporting their interaction within the cytoplasm. Evidence of LGP2 proteolysis was also detected during FMDV infection. Moreover, the inhibitory effect of LGP2 overexpression on FMDV growth observed was reverted when Lpro was co-expressed, concomitant with lower levels of IFN-ß mRNA and antiviral activity in those cells. The Lpro target site in LGP2 was identified as an RGRAR sequence in a conserved helicase motif whose replacement to EGEAE abrogated LGP2 cleavage by Lpro. Taken together, these data suggest that LGP2 cleavage by the Leader protease of aphthoviruses may represent a novel antagonistic mechanism for immune evasion.


Assuntos
Endopeptidases/metabolismo , Vírus da Febre Aftosa/imunologia , Febre Aftosa/virologia , Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , RNA Helicases/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Endopeptidases/genética , Febre Aftosa/imunologia , Febre Aftosa/patologia , Vírus da Febre Aftosa/enzimologia , Células HEK293 , Humanos , RNA Helicases/genética , RNA Helicases/imunologia , Células Vero
14.
Arch Virol ; 163(6): 1407-1417, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29397456

RESUMO

Infection of chickens with virulent Newcastle disease virus (NDV) is associated with severe pathology and increased morbidity and mortality. The innate immune response contributes to the pathogenicity of NDV. As professional antigen-presenting cells, dendritic cells (DCs) play a unique role in innate immunity. However, the contribution of DCs to NDV infection has not been investigated in chickens. In this study, we selected two representative NDV strains, i.e., the velogenic NDV strain Chicken/Guangdong/GM/2014 (GM) and the lentogenic NDV strain La Sota, to investigate whether NDVs could infect LPS-activated chicken bone-derived marrow DCs (mature chicken BM-DCs). We compared the viral titres and innate immune responses in mature chicken BM-DCs following infection with those strains. Both NDV strains could infect mature chicken BM-DC, but the GM strain showed stronger replication capacity than the La Sota strain in mature chicken BM-DCs. Gene expression profiling showed that MDA5, LGP2, TLR3, TLR7, IFN-α, IFN-ß, IFN-γ, IL-1ß, IL-6, IL-18, IL-8, CCL5, IL-10, IL-12, MHC-I, and MHC-II levels were altered in mature DCs after infection with NDVs at all evaluated times postinfection. Notably, the GM strain triggered stronger innate immune responses than the La Sota strain in chicken BM-DCs. However, both strains were able to suppress the expression of some cytokines, such as IL-6 and IFN-α, in mature chicken DCs at 24 hpi. These data provide a foundation for further investigation of the role of chicken DCs in NDV infection.


Assuntos
Proteínas Aviárias/imunologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/patogenicidade , Doenças das Aves Domésticas/imunologia , Animais , Proteínas Aviárias/genética , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/virologia , Galinhas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/virologia , Perfilação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Interações Hospedeiro-Patógeno/genética , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/imunologia , Interferons/genética , Interferons/imunologia , Interleucinas/genética , Interleucinas/imunologia , Lipopolissacarídeos/farmacologia , Doença de Newcastle/genética , Doença de Newcastle/patologia , Doença de Newcastle/virologia , Vírus da Doença de Newcastle/imunologia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , RNA Helicases/genética , RNA Helicases/imunologia , Transdução de Sinais , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Virulência , Replicação Viral
15.
Biochem Biophys Res Commun ; 494(1-2): 227-233, 2017 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-29032202

RESUMO

Laboratory of genetics and physiology 2 (LGP2) and melanoma differentiation-associated gene 5 (MDA5) cooperatively detect viral RNA in the cytoplasm of Cardiovirus-infected cells and activate innate immune responses. Here, we evaluated whether the double-stranded RNA-binding protein PACT plays a role in this anti-viral response to further elucidate the mechanism. Immunoprecipitation experiments demonstrated that PACT interacts with LGP2 and that this interaction is enhanced by encephalomyocarditis virus (EMCV) infection. In vitro interaction analyses using purified recombinant proteins confirmed that the single-stranded Theiler's murine encephalitis virus genome enhanced the interaction between LGP2 and PACT. Small interfering RNA knockdown experiments further indicated that PACT is required for Cardiovirus-triggered interferon responses. To support this functional interaction with LGP2, overexpressed PACT was shown to enhance EMCV-triggered interferon promoter activity only when LGP2 and MDA5 were co-expressed but not when MDA5 is expressed alone. Together, our findings indicate a possible role of PACT in regulating the Cardiovirus-triggered immune responses mediated by MDA5 and LGP2, which opens the door to novel therapeutic strategies in interferon-related autoimmune diseases and cancer.


Assuntos
Infecções por Cardiovirus/imunologia , Vírus da Encefalomiocardite , Helicase IFIH1 Induzida por Interferon/imunologia , RNA Helicases/imunologia , Proteínas de Ligação a RNA/imunologia , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Linhagem Celular , Chlorocebus aethiops , RNA Helicases DEAD-box/antagonistas & inibidores , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/imunologia , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/imunologia , Técnicas de Silenciamento de Genes , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/genética , Helicase IFIH1 Induzida por Interferon/genética , Interferon beta/genética , Camundongos , Regiões Promotoras Genéticas , RNA Helicases/genética , RNA Interferente Pequeno/genética , RNA Viral/genética , RNA Viral/imunologia , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Ribonuclease III/antagonistas & inibidores , Ribonuclease III/genética , Ribonuclease III/imunologia , Células Vero
16.
Sci Rep ; 7(1): 6737, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28751780

RESUMO

Classical swine fever virus (CSFV) non-structural protein 3 (NS3) is a multifunctional non-structural protein that plays a major role in viral replication. However, how exactly NS3 exerts these functions remains unknown. Here, we identified tumour necrosis factor receptor-associated factor 6 (TRAF6) as a novel NS3-interacting protein via yeast two-hybrid analysis, co-immunoprecipitation, and glutathione S-transferase pull-down assays. Furthermore, we observed that TRAF6 overexpression significantly inhibited CSFV replication, and TRAF6 knockdown promoted CSFV replication in porcine alveolar macrophages. Additionally, TRAF6 was degraded during CSFV infection or NS3 expression exclusively, indicating that CSFV and TRAF6 were mutually antagonistic and that TRAF6 degradation might contribute to persistent CSFV replication. Moreover, nuclear factor-kappa B (NF-κB) activity and interferon (IFN)-ß and interleukin (IL)-6 expression were increased in TRAF6-overexpressing cells, whereas TRAF6-knockdown cells exhibited decreased NF-κB activity and IFN-ß and IL-6 levels. Notably, TRAF6 overexpression did not reduce CSFV replication following inhibition of NF-κB activation by p65 knockdown. Our findings revealed that TRAF6 inhibits CSFV replication via activation of NF-κB-signalling pathways along with increases in the expression of its targets IFN-ß and IL-6. This work addresses a novel aspect concerning the regulation of innate antiviral immune response during CSFV infection.


Assuntos
Vírus da Febre Suína Clássica/genética , Interações Hospedeiro-Patógeno/genética , Macrófagos Alveolares/virologia , Fator 6 Associado a Receptor de TNF/genética , Fator de Transcrição RelA/genética , Proteínas não Estruturais Virais/genética , Replicação Viral , Animais , Linhagem Celular , Vírus da Febre Suína Clássica/crescimento & desenvolvimento , Regulação da Expressão Gênica , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interferon beta/genética , Interferon beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Macrófagos Alveolares/imunologia , Ligação Proteica , Estabilidade Proteica , Proteólise , RNA Helicases/genética , RNA Helicases/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Transdução de Sinais , Suínos , Fator 6 Associado a Receptor de TNF/antagonistas & inibidores , Fator 6 Associado a Receptor de TNF/imunologia , Fator de Transcrição RelA/imunologia , Técnicas do Sistema de Duplo-Híbrido , Proteínas não Estruturais Virais/imunologia
17.
J Virol ; 90(24): 11259-11278, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27707928

RESUMO

Epidemiological studies suggest that India has the largest number of dengue virus infection cases worldwide. However, there is minimal information about the immunological responses in these patients. CD8 T cells are important in dengue, because they have been implicated in both protection and immunopathology. Here, we provide a detailed analysis of HLA-DR+ CD38+ and HLA-DR- CD38+ effector CD8 T cell subsets in dengue patients from India and Thailand. Both CD8 T cell subsets expanded and expressed markers indicative of antigen-driven proliferation, tissue homing, and cytotoxic effector functions, with the HLA-DR+ CD38+ subset being the most striking in these effector qualities. The breadth of the dengue-specific CD8 T cell response was diverse, with NS3-specific cells being the most dominant. Interestingly, only a small fraction of these activated effector CD8 T cells produced gamma interferon (IFN-γ) when stimulated with dengue virus peptide pools. Transcriptomics revealed downregulation of key molecules involved in T cell receptor (TCR) signaling. Consistent with this, the majority of these CD8 T cells remained IFN-γ unresponsive even after TCR-dependent polyclonal stimulation (anti-CD3 plus anti-CD28) but produced IFN-γ by TCR-independent polyclonal stimulation (phorbol 12-myristate 13-acetate [PMA] plus ionomycin). Thus, the vast majority of these proliferating, highly differentiated effector CD8 T cells probably acquire TCR refractoriness at the time the patient is experiencing febrile illness that leads to IFN-γ unresponsiveness. Our studies open novel avenues for understanding the mechanisms that fine-tune the balance between CD8 T cell-mediated protective versus pathological effects in dengue. IMPORTANCE: Dengue is becoming a global public health concern. Although CD8 T cells have been implicated both in protection and in the cytokine-mediated immunopathology of dengue, how the balance is maintained between these opposing functions remains unknown. We comprehensively characterized CD8 T cell subsets in dengue patients from India and Thailand and show that these cells expand massively and express phenotypes indicative of overwhelming antigenic stimulus and tissue homing/cytotoxic-effector functions but that a vast majority of them fail to produce IFN-γ in vitro Interestingly, the cells were fully capable of producing the cytokine when stimulated in a T cell receptor (TCR)-independent manner but failed to do so in TCR-dependent stimulation. These results, together with transcriptomics, revealed that the vast majority of these CD8 T cells from dengue patients become cytokine unresponsive due to TCR signaling insufficiencies. These observations open novel avenues for understanding the mechanisms that fine-tune the balance between CD8-mediated protective versus pathological effects.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica , Vírus da Dengue/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Transcriptoma/imunologia , ADP-Ribosil Ciclase 1/genética , ADP-Ribosil Ciclase 1/imunologia , Adolescente , Anticorpos/farmacologia , Antígenos CD28/antagonistas & inibidores , Antígenos CD28/genética , Antígenos CD28/imunologia , Complexo CD3/genética , Complexo CD3/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/virologia , Proliferação de Células/efeitos dos fármacos , Criança , Pré-Escolar , Vírus da Dengue/genética , Vírus da Dengue/crescimento & desenvolvimento , Vírus da Dengue/metabolismo , Feminino , Regulação da Expressão Gênica , Antígenos HLA-DR/genética , Antígenos HLA-DR/imunologia , Humanos , Imunidade Celular , Índia , Lactente , Interferon gama/genética , Interferon gama/imunologia , Ionomicina/farmacologia , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Cultura Primária de Células , RNA Helicases/genética , RNA Helicases/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Transdução de Sinais , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/virologia , Acetato de Tetradecanoilforbol/farmacologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia
18.
J Med Virol ; 88(8): 1448-52, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26792253

RESUMO

In Brazil, dengue is a public health problem with the occurrence of explosive epidemics. This study reports maternal and fetal deaths due to dengue and which tissues of placenta and umbilical cord were analyzed by molecular methods and immunohistochemistry. The dengue NS3 and NS1 detection revealed the viral presence in different cells from placenta and umbilical cord. In the latter, DENV-2 was detected at a viral titer of 1,02 × 10(4) amounts of viral RNA. It was shown that the DENV markers analyzed here may be an alternative approach for dengue fatal cases investigation, especially involving maternal and fetal death. J. Med. Virol. 88:1448-1452, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Vírus da Dengue , Dengue/virologia , Morte Fetal/etiologia , Morte Materna/etiologia , Placenta/virologia , Cordão Umbilical/virologia , Proteínas não Estruturais Virais/isolamento & purificação , Anticorpos Antivirais/imunologia , Antígenos Virais/genética , Brasil/epidemiologia , Dengue/epidemiologia , Vírus da Dengue/química , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Vírus da Dengue/isolamento & purificação , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Macrófagos/virologia , Placenta/citologia , Placenta/patologia , Gravidez , RNA Helicases/genética , RNA Helicases/imunologia , RNA Helicases/isolamento & purificação , RNA Viral/genética , RNA Viral/isolamento & purificação , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Serina Endopeptidases/isolamento & purificação , Testes Sorológicos , Cordão Umbilical/citologia , Cordão Umbilical/patologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Adulto Jovem
19.
J Biochem ; 159(3): 279-86, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26748340

RESUMO

Activation of antiviral innate immunity is triggered by cellular pattern recognition receptors. Retinoic acid inducible gene-I (RIG-I)-like receptors (RLRs) detect viral non-self RNA in cytoplasm of virus-infected cells and play a critical role in the clearance of the invaded viruses through production of antiviral cytokines. Among the three known RLRs, RIG-I and melanoma differentiation-associated gene 5 recognize distinct non-self signatures of viral RNA and activate antiviral signaling. Recent reports have clearly described the molecular machinery underlying the activation of RLRs and interactions with the downstream adaptor, mitochondrial antiviral signaling protein (MAVS). RLRs and MAVS are thought to form large multimeric filaments around cytoplasmic organelles depending on the presence of Lys63-linked ubiquitin chains. Furthermore, RLRs have been shown to localize to stress-induced ribonucleoprotein aggregate known as stress granules and utilize them as a platform for recognition/activation of signaling. In this review, we will focus on the current understanding of RLR-mediated signal activation and the interactions with stress-induced RNA granules.


Assuntos
Grânulos Citoplasmáticos/imunologia , Infecções por Vírus de DNA/imunologia , Imunidade Inata , Infecções por Vírus de RNA/imunologia , RNA Viral/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Grânulos Citoplasmáticos/virologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/imunologia , RNA Helicases DEAD-box/metabolismo , Humanos , Helicase IFIH1 Induzida por Interferon , Camundongos , Poliubiquitina/metabolismo , RNA Helicases/imunologia , RNA Helicases/metabolismo , Receptores Imunológicos , Receptores de Reconhecimento de Padrão/metabolismo , Ribonucleoproteínas/metabolismo , Transdução de Sinais , Estresse Fisiológico/imunologia
20.
Vaccine ; 33(32): 4004-12, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26079613

RESUMO

DNA vaccination is effective in inducing potent immunity in mice; however it appears to be less so in large animals. Increasing the dose of DNA plasmid to activate innate immunity has been shown to improve DNA vaccine adaptive immunity. Retinoic acid-inducible gene I (RIG-I) is a critical cytoplasmic double-stranded RNA pattern receptor required for innate immune activation in response to viral infection. RIG-I recognise viral RNA and trigger antiviral response, resulting in type I interferon (IFN) and inflammatory cytokine production. In an attempt to enhance the antibody response induced by BVDV DNA in cattle, we expressed BVDV truncated E2 (E2t) and NS3 codon optimised antigens from antibiotic free-plasmid vectors expressing a RIG-I agonist and designated either NTC E2t(co) and NTC NS3(co). To evaluate vaccine efficacy, groups of five BVDV-free calves were intramuscularly injected three times with NTC E2t(co) and NTC NS3(co) vaccine plasmids individually or in combination. Animals vaccinated with our (previously published) conventional DNA vaccines pSecTag/E2 and pTriExNS3 and plasmids expressing RIG-I agonist only presented both the positive and mock-vaccine groups. Our results showed that vaccines coexpressing E2t with a RIG-I agonist induced significantly higher E2 antigen specific antibody response (p<0.05). Additionally, E2t augmented the immune response to NS3 when the two vaccines were delivered in combination. Despite the lack of complete protection, on challenge day 4/5 calves vaccinated with NTC E2t(co) alone or NTC E2t(co) plus NTC NS3(co) had neutralising antibody titres exceeding 1/240 compared to 1/5 in the mock vaccine control group. Based on our results we conclude that co-expression of a RIG-I agonist with viral antigen could enhance DNA vaccine potency in cattle.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Doença das Mucosas por Vírus da Diarreia Viral Bovina/prevenção & controle , Vírus da Diarreia Viral Bovina/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Bovinos , Vírus da Diarreia Viral Bovina/genética , Modelos Animais de Doenças , Expressão Gênica , Vetores Genéticos , Injeções Intramusculares , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/imunologia , Plasmídeos , RNA Helicases/genética , RNA Helicases/imunologia , Resultado do Tratamento , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA