Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Microbes Infect ; 26(4): 105321, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38461968

RESUMO

Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year globally. RABV infection is characterized by the suppression of the interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) that initiates IFN signaling currently remain elusive. Here, we showed that RABV RNAs are primarily recognized by the RIG-I RLR, resulting in an IFN response in the infected cells, but this response varied according to the type of RABV used. Pathogenic RABV strain RNAs, Tha, were poorly detected in the cytosol by RIG-I and therefore caused a weak antiviral response. However, we revealed a strong IFN activity triggered by the attenuated RABV vaccine strain RNAs, SAD, mediated by RIG-I. We characterized two major 5' copy-back defective interfering (5'cb DI) genomes generated during SAD replication. Furthermore, we identified an interaction between 5'cb DI genomes, and RIG-I correlated with a high stimulation of the type I IFN signaling. This study indicates that wild-type RABV RNAs poorly activate the RIG-I pathway, while the presence of 5'cb DIs in the live-attenuated vaccine strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection thus strongly stimulates the IFN response.


Assuntos
Proteína DEAD-box 58 , Vírus da Raiva , Humanos , Linhagem Celular , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Receptores Imunológicos/metabolismo , RNA Viral/genética , Transdução de Sinais , Replicação Viral
2.
J Immunol ; 210(9): 1247-1256, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36939421

RESUMO

Retinoic acid-inducible gene I (RIG-I) is essential for activating host cell innate immunity to regulate the immune response against many RNA viruses. We previously identified that a small molecule compound, KIN1148, led to the activation of IFN regulatory factor 3 (IRF3) and served to enhance protection against influenza A virus (IAV) A/California/04/2009 infection. We have now determined direct binding of KIN1148 to RIG-I to drive expression of IFN regulatory factor 3 and NF-κB target genes, including specific immunomodulatory cytokines and chemokines. Intriguingly, KIN1148 does not lead to ATPase activity or compete with ATP for binding but activates RIG-I to induce antiviral gene expression programs distinct from type I IFN treatment. When administered in combination with a vaccine against IAV, KIN1148 induces both neutralizing Ab and IAV-specific T cell responses compared with vaccination alone, which induces comparatively poor responses. This robust KIN1148-adjuvanted immune response protects mice from lethal A/California/04/2009 and H5N1 IAV challenge. Importantly, KIN1148 also augments human CD8+ T cell activation. Thus, we have identified a small molecule RIG-I agonist that serves as an effective adjuvant in inducing noncanonical RIG-I activation for induction of innate immune programs that enhance adaptive immune protection of antiviral vaccination.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , Humanos , Animais , Camundongos , Proteína DEAD-box 58/metabolismo , Virus da Influenza A Subtipo H5N1/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Adjuvantes Imunológicos , Antivirais/farmacologia , Imunidade Inata
3.
Viruses ; 14(7)2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35891387

RESUMO

Pathogen-associated molecular patterns, including cytoplasmic DNA and double-strand (ds)RNA trigger the induction of interferon (IFN) and antiviral states protecting cells and organisms from pathogens. Here we discovered that the transfection of human airway cell lines or non-transformed fibroblasts with 24mer dsRNA mimicking the cellular micro-RNA (miR)29b-1* gives strong anti-viral effects against human adenovirus type 5 (AdV-C5), influenza A virus X31 (H3N2), and SARS-CoV-2. These anti-viral effects required blunt-end complementary RNA strands and were not elicited by corresponding single-strand RNAs. dsRNA miR-29b-1* but not randomized miR-29b-1* mimics induced IFN-stimulated gene expression, and downregulated cell adhesion and cell cycle genes, as indicated by transcriptomics and IFN-I responsive Mx1-promoter activity assays. The inhibition of AdV-C5 infection with miR-29b-1* mimic depended on the IFN-alpha receptor 2 (IFNAR2) and the RNA-helicase retinoic acid-inducible gene I (RIG-I) but not cytoplasmic RNA sensors MDA5 and ZNFX1 or MyD88/TRIF adaptors. The antiviral effects of miR29b-1* were independent of a central AUAU-motif inducing dsRNA bending, as mimics with disrupted AUAU-motif were anti-viral in normal but not RIG-I knock-out (KO) or IFNAR2-KO cells. The screening of a library of scrambled short dsRNA sequences identified also anti-viral mimics functioning independently of RIG-I and IFNAR2, thus exemplifying the diverse anti-viral mechanisms of short blunt-end dsRNAs.


Assuntos
COVID-19 , Interferon Tipo I , MicroRNAs , Antivirais/farmacologia , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/metabolismo , RNA Helicases DEAD-box/genética , Humanos , Vírus da Influenza A Subtipo H3N2/genética , Interferon Tipo I/genética , RNA de Cadeia Dupla , SARS-CoV-2
4.
J Control Release ; 347: 476-488, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35577151

RESUMO

Despite success in vaccinating populations against SARS-CoV-2, concerns about immunity duration, continued efficacy against emerging variants, protection from infection and transmission, and worldwide vaccine availability remain. Molecular adjuvants targeting pattern recognition receptors (PRRs) on antigen-presenting cells (APCs) could improve and broaden the efficacy and durability of vaccine responses. Native SARS-CoV-2 infection stimulates various PRRs, including toll-like receptors (TLRs) and retinoic acid-inducible gene I (RIG-I)-like receptors. We hypothesized that targeting PRRs using molecular adjuvants on nanoparticles (NPs) along with a stabilized spike protein antigen could stimulate broad and efficient immune responses. Adjuvants targeting TLR4 (MPLA), TLR7/8 (R848), TLR9 (CpG), and RIG-I (PUUC) delivered on degradable polymer NPs were combined with the S1 subunit of spike protein and assessed in vitro with isogeneic mixed lymphocyte reactions (isoMLRs). For in vivo studies, the adjuvant-NPs were combined with stabilized spike protein or spike-conjugated NPs and assessed using a two-dose intranasal or intramuscular vaccination model in mice. Combination adjuvant-NPs simultaneously targeting TLR and RIG-I receptors (MPLA+PUUC, CpG+PUUC, and R848+PUUC) differentially induced T cell proliferation and increased proinflammatory cytokine secretion by APCs in vitro. When delivered intranasally, MPLA+PUUC NPs enhanced CD4+CD44+ activated memory T cell responses against spike protein in the lungs while MPLA NPs increased anti-spike IgA in the bronchoalveolar (BAL) fluid and IgG in the blood. Following intramuscular delivery, PUUC NPs induced strong humoral immune responses, characterized by increases in anti-spike IgG in the blood and germinal center B cell populations (GL7+ and BCL6+ B cells) in the draining lymph nodes (dLNs). MPLA+PUUC NPs further boosted spike protein-neutralizing antibody titers and T follicular helper cell populations in the dLNs. These results suggest that protein subunit vaccines with particle-delivered molecular adjuvants targeting TLR4 and RIG-I could lead to robust and unique route-specific adaptive immune responses against SARS-CoV-2.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Proteína DEAD-box 58 , Nanopartículas , Receptores Imunológicos , Receptor 4 Toll-Like , Adjuvantes Imunológicos , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19/imunologia , Sistemas de Liberação de Medicamentos , Imunidade Humoral , Imunoglobulina G , Camundongos , Nanopartículas/química , Receptores Imunológicos/agonistas , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Receptor 4 Toll-Like/agonistas
5.
Biochem Biophys Res Commun ; 588: 90-96, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34953211

RESUMO

Here we investigated the gender difference in murine cholangitis resembling human primary biliary cholangitis (PBC) caused by synthetic double-stranded RNA, and underlying hepatic innate immune responses. Female C57Bl/6 mice given repeated injections of polyinosinic-polycytidylic acid (poly I:C) for 24 weeks developed overt cholangitis with positive serum anti-mitochondria-M2 antibody, whereas male mice showed minimal pathological changes without induction in autoantibody. Poly I:C induced hepatic inflammatory cytokines and type-I interferons predominantly in females. Hepatic expression levels of toll-like receptor (TLR) 3 and melanoma differentiation-associated protein (MDA) 5 were equivalent in both genders; however, both mRNA and protein levels of retinoic acid-inducible gene (RIG)-I were nearly doubled in female livers. Following 4-week injections of poly I:C, not only hepatic RIG-I, but also TLR3 and MDA5 showed female-predominance. Moreover, hepatic RIG-I levels were 25% lower in ovariectomized mice, whereas supplementation of 17 ß-estradiol enhanced hepatic RIG-I expression, as well as cytokine induction. These results clearly indicate that hepatic RIG-I expression is potentiated by estrogen, and triggers gender-dependent hepatic innate immune response against double-stranded RNA, which most likely play a pivotal role in the pathogenesis of autoimmune cholangiopathies including PBC.


Assuntos
Colangite/patologia , RNA de Cadeia Dupla/efeitos adversos , Caracteres Sexuais , Animais , Autoanticorpos/sangue , Colangite/sangue , Colangite/imunologia , Citocinas/metabolismo , Proteína DEAD-box 58/metabolismo , Estrogênios/farmacologia , Feminino , Helicase IFIH1 Induzida por Interferon/metabolismo , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Poli I-C/efeitos adversos , Receptores de Reconhecimento de Padrão/metabolismo , Receptor 3 Toll-Like/metabolismo
6.
Mol Pharm ; 18(2): 679-698, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32491861

RESUMO

Current influenza virus vaccines are focused on humoral immunity and are limited by the short duration of protection, narrow cross-strain efficacy, and suboptimal immunogenicity. Here, we combined two chemically and biologically distinct adjuvants, an oil-in-water nanoemulsion (NE) and RNA-based agonists of RIG-I, to determine whether the diverse mechanisms of these adjuvants could lead to improved immunogenicity and breadth of protection against the influenza virus. NE activates TLRs, stimulates immunogenic apoptosis, and enhances cellular antigen uptake, leading to a balanced TH1/TH2/TH17 response when administered intranasally. RIG-I agonists included RNAs derived from Sendai and influenza viral defective interfering RNAs (IVT DI, 3php, respectively) and RIG-I/TLR3 agonist, poly(I:C) (pIC), which induce IFN-Is and TH1-polarized responses. NE/RNA combined adjuvants potentially allow for costimulation of multiple innate immune receptor pathways, more closely mimicking patterns of activation occurring during natural viral infection. Mice intranasally immunized with inactivated A/Puerto Rico/8/1934 (H1N1) (PR/8) adjuvanted with NE/IVT DI or NE/3php (but not NE/pIC) showed synergistic enhancement of systemic PR/8-specific IgG with significantly greater avidity and virus neutralization activity than the individual adjuvants. Notably, NE/IVT DI induced protective neutralizing titers after a single immunization. Hemagglutinin stem-specific antibodies were also improved, allowing recognition of heterologous and heterosubtypic hemagglutinins. All NE/RNAs elicited substantial PR/8-specific sIgA. Finally, a unique cellular response with enhanced TH1/TH17 immunity was induced with the NE/RNAs. These results demonstrate that the enhanced immunogenicity of the adjuvant combinations was synergistic and not simply additive, highlighting the potential value of a combined adjuvant approach for improving the efficacy of vaccination against the influenza virus.


Assuntos
Proteína DEAD-box 58/metabolismo , Portadores de Fármacos/química , Vacinas contra Influenza/administração & dosagem , Influenza Humana/prevenção & controle , RNA Interferente Pequeno/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Cães , Emulsões , Feminino , Humanos , Imunidade Celular , Imunidade Humoral , Imunidade nas Mucosas , Imunogenicidade da Vacina , Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Camundongos , Nanopartículas/química , Poli I-C/administração & dosagem , Cultura Primária de Células , RNA Interferente Pequeno/imunologia , Vacinação/métodos
7.
J Ethnopharmacol ; 268: 113555, 2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33152425

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Carvacrol, a monoterpene phenol from Mosla chinensis Maxim, which is a commonly Chinese herbal medicine. The most important pharmacology of it is dispelling exogenous evils by increasing perspiration. And it is the gentleman medicine in the Chinese herbal compound prescription of Xin-Jia-Xiang-Ru-Yin, mainly for the treatment of summer colds with dampness including influenza virus A infection. AIM OF THE STUDY: Our preliminary study verified that the Xin-Jia-Xiang-Ru-Yin could inhibit acute lung injury of mice with influenza virus A infection. And there have been some reports implicating the high antimicrobial activity of carvacrol for a wide range of product preservation, but little research including the effects of it on viral infection. The aim of this study was to reveal the antiviral effects of carvacrol, the main constituent in Mosla chinensis Maxim. MATERIALS AND METHODS: Initially, C57BL/6 mice were grouped and intranasally administered FM1 virus to construct viral infection models. After treatment with ribavirin and carvacrol for 5 days, all mice were euthanized, and specimens were immediately obtained. Histology, flow cytometry and Meso Scale Discovery (MSD) analysis were used to analyze pathological changes in lung tissue, the expression levels of cytokines and the differentiation and proportion of CD4+ T cells subsets, while Western blot and qRT-PCR were used to detect the expression of related proteins and mRNA. RESULTS: Carvacrol attenuated lung tissue damage, the proportions of Th1, Th2, Th17 and Treg in CD4+ T cells and the relative proportions of Th1/Th2 and Th17/Treg cells. Carvacrol inhibited the expression of inflammation-associated cytokines including IFN-γ, IL-2, IL-4, IL-5, IL-12 and TNF-ɑ, IL-1, IL-10, IL-6. Decreased levels of TLR7, MyD88, IRAK4, TRAK6, NF-κB, RIG-I, IPS-I and IRF mRNA in carvacrol-treated mice were observed comparing to the mice in VC group. Further, the total expression of RIG-I, MyD88 and NF-κB proteins had increased significantly in the VC group but reduced obviously in the group treated with ribavirin or carvacrol. CONCLUSIONS: These results indicate that carvacrol is a potential alternative treatment for the excessive immune response induced by influenza virus A infection, the cold-fighting effect of Mosla chinensis Maxim may depend on the anti-virus of carvacrol.


Assuntos
Alphainfluenzavirus/efeitos dos fármacos , Cimenos/farmacologia , Proteína DEAD-box 58/antagonistas & inibidores , Imunidade Inata/efeitos dos fármacos , Glicoproteínas de Membrana/antagonistas & inibidores , Receptor 7 Toll-Like/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/metabolismo , Animais , Cimenos/uso terapêutico , Proteína DEAD-box 58/imunologia , Proteína DEAD-box 58/metabolismo , Feminino , Imunidade Inata/imunologia , Alphainfluenzavirus/imunologia , Alphainfluenzavirus/metabolismo , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Receptor 7 Toll-Like/imunologia , Receptor 7 Toll-Like/metabolismo , Replicação Viral/imunologia
8.
Leukemia ; 34(4): 1017-1026, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31740809

RESUMO

Retinoic acid-inducible gene-I (RIG-I) is a cytoplasmic immune receptor sensing viral RNA. It triggers the release of type I interferons (IFN) and proinflammatory cytokines inducing an adaptive cellular immune response. We investigated the therapeutic potential of systemic RIG-I activation by short 5'-triphosphate-modified RNA (ppp-RNA) for the treatment of acute myeloid leukemia (AML) in the syngeneic murine C1498 AML tumor model. ppp-RNA treatment significantly reduced tumor burden, delayed disease onset and led to complete remission including immunological memory formation in a substantial proportion of animals. Therapy-induced tumor rejection was dependent on CD4+ and CD8+ T cells, but not on NK or B cells, and relied on intact IFN and mitochondrial antiviral signaling protein (MAVS) signaling in the host. Interestingly, ppp-RNA treatment induced programmed death ligand 1 (PD-L1) expression on AML cells and established therapeutic sensitivity to anti-PD-1 checkpoint blockade in vivo. In immune-reconstituted humanized mice, ppp-RNA treatment reduced the number of patient-derived xenografted (PDX) AML cells in blood and bone marrow while concomitantly enhancing CD3+ T cell counts in the respective tissues. Due to its ability to establish a state of full remission and immunological memory, our findings show that ppp-RNA treatment is a promising strategy for the immunotherapy of AML.


Assuntos
Anticorpos Neutralizantes/farmacologia , Proteína DEAD-box 58/imunologia , Imunoterapia/métodos , Leucemia Mieloide Aguda/terapia , RNA de Cadeia Dupla/farmacologia , Receptores Virais/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Proteína DEAD-box 58/genética , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica , Xenoenxertos , Humanos , Memória Imunológica/efeitos dos fármacos , Interferons/genética , Interferons/imunologia , Isoenxertos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/mortalidade , Camundongos , Receptores Virais/agonistas , Receptores Virais/genética , Indução de Remissão , Transdução de Sinais , Análise de Sobrevida , Resultado do Tratamento
9.
Int J Cancer ; 146(7): 2007-2018, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31800094

RESUMO

Ovarian cancer (OC) is the most lethal gynecological malignancy, with platinum-based chemotherapy remaining the mainstay for adjuvant treatment after surgery. The lack of indication for immunotherapy may at least in part result from the lack of suitable biomarkers allowing stratification of potentially responding patients. In this monocentric study of 141 cases with OC, we used real-time quantitative PCR to assess the expression of retinoic acid-inducible gene-I (RIG-I) in primary tumor and healthy ovarian control tissues. RIG-I expression was correlated to various clinicopathological characteristics as well as to a set of molecular and immunological markers. The prognostic significance of RIG-I expression was queried in univariate and multivariate analyses and validated in an independent cohort. RIG-I was overexpressed in the cancerous ovary and correlated with a higher tumor grade. The more aggressive Type-II cancers and cancers with inactivating p53 mutations exhibited higher RIG-I expression. RIG-I levels were also elevated in cancers that recurred after remission or were platinum-refractory. Survival analyses disclosed RIG-I as an independent marker of poor outcome in OC. Continuative analyses revealed the molecular and immunological correlates of RIG-I expression in the tumor microenvironment, including interferon production and a distinct immune-regulatory signature involving checkpoint molecules (PD-L1/PD-1), the RNA-editing enzyme ADAR1 and the regulatory T cell-specific transcription factor FoxP3. We conclude that high RIG-I expression associates with poor outcome in OC, which is explainable by local immunosuppression in the tumor bed. RIG-I expression may inform checkpoint blockade and/or RIG-I agonistic targeting in a subset of high-risk OC patients.


Assuntos
Biomarcadores Tumorais , Proteína DEAD-box 58/genética , Neoplasias Ovarianas/etiologia , Neoplasias Ovarianas/mortalidade , Evasão Tumoral/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Neoplasias Ovarianas/diagnóstico , Prognóstico , Modelos de Riscos Proporcionais , Receptores Imunológicos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Adulto Jovem
10.
Cancer Immunol Immunother ; 68(9): 1479-1492, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31463653

RESUMO

RIG-I is a cytosolic RNA sensor that recognizes short 5' triphosphate RNA, commonly generated during virus infection. Upon activation, RIG-I initiates antiviral immunity, and in some circumstances, induces cell death. Because of this dual capacity, RIG-I has emerged as a promising target for cancer immunotherapy. Previously, a sequence-optimized RIG-I agonist (termed M8) was generated and shown to stimulate a robust immune response capable of blocking viral infection and to function as an adjuvant in vaccination strategies. Here, we investigated the potential of M8 as an anti-cancer agent by analyzing its ability to induce cell death and activate the immune response. In multiple cancer cell lines, M8 treatment strongly activated caspase 3-dependent apoptosis, that relied on an intrinsic NOXA and PUMA-driven pathway that was dependent on IFN-I signaling. Additionally, cell death induced by M8 was characterized by the expression of markers of immunogenic cell death-related damage-associated molecular patterns (ICD-DAMP)-calreticulin, HMGB1 and ATP-and high levels of ICD-related cytokines CXCL10, IFNß, CCL2 and CXCL1. Moreover, M8 increased the levels of HLA-ABC expression on the tumor cell surface, as well as up-regulation of genes involved in antigen processing and presentation. M8 induction of the RIG-I pathway in cancer cells favored dendritic cell phagocytosis and induction of co-stimulatory molecules CD80 and CD86, together with increased expression of IL12 and CXCL10. Altogether, these results highlight the potential of M8 in cancer immunotherapy, with the capacity to induce ICD-DAMP on tumor cells and activate immunostimulatory signals that synergize with current therapies.


Assuntos
Antineoplásicos/uso terapêutico , Células Dendríticas/imunologia , Melanoma/tratamento farmacológico , Nelfinavir/análogos & derivados , Alarminas/imunologia , Apresentação de Antígeno/efeitos dos fármacos , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Calreticulina/metabolismo , Caspase 3/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Proteína DEAD-box 58/antagonistas & inibidores , Proteína HMGB1/metabolismo , Humanos , Imunização , Interferons/metabolismo , Terapia de Alvo Molecular , Nelfinavir/farmacologia , Nelfinavir/uso terapêutico , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores Imunológicos , Transdução de Sinais
11.
Eur J Pharmacol ; 860: 172543, 2019 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-31323223

RESUMO

Ergosterol peroxide has been shown to exhibit anti-tumor, antioxidant and anti-bacterial properties. However, the effects of ergosterol peroxide isolated from the herbal Baphicacanthus cusia root on influenza virus infection remain poorly understood. In the present study, ergosterol peroxide (compound 22) was obtained from the B. cusia root and subjected to investigation regarding its immunoregulatory effect on influenza A virus (IAV)-induced inflammation in A549 human alveolar epithelial cells. The structure of compound 22 isolated from B. cusia root. was elucidated by NMR analyses. Structure determination showed that the chemical structure of compound 22 closely resembles that of ergosterol peroxide. We observed that ergosterol peroxide treatment significantly suppressed IAV-induced upregulation of RIG-I expression. Additionally, ergosterol peroxide inhibited the activation of RIG-I downstream signaling pathways, including p38 MAP kinase and NF-κB, which ultimately resulted in the reduced production of an array of pro-inflammatory mediators and interferons (IFN-ß and IFN-λ1). Interestingly, inhibitory effects of ergosterol peroxide on the expression of IFNs did not affect the expression of antiviral effectors or enhance viral replication. On the other hand, ergosterol peroxide effectively abolished the amplified production of pro-inflammatory mediators in cells pretreated with IFN-ß (500 ng/ml) prior to IAV infection. Moreover, Annexin V and Hoechst 33258 staining revealed that increased apoptosis of IAV-infected cells was reversed by the presence of ergosterol peroxide. Our findings suggest that ergosterol peroxide from the B. cusia root suppressed IAV-associated inflammation and apoptosis via blocking RIG-I signaling, which may serve as a supplementary approach to the treatment of influenza.


Assuntos
Apoptose/efeitos dos fármacos , Proteína DEAD-box 58/metabolismo , Ergosterol/análogos & derivados , Vírus da Influenza A Subtipo H1N1/fisiologia , Transdução de Sinais/efeitos dos fármacos , Células A549 , Animais , Cães , Ergosterol/química , Ergosterol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/patologia , Inflamação/virologia , Mediadores da Inflamação/metabolismo , Interferons/biossíntese , Células Madin Darby de Rim Canino , Receptores Imunológicos
12.
Inflammopharmacology ; 27(6): 1255-1263, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30783895

RESUMO

Influenza viruses can bring about acute respiratory diseases and are a potential hazard to human health. Antiviral drugs are the main ways to control the influenza virus infection except the vaccine. In this study, the immune regulation activity of pterodontic acid isolated from Laggera pterodonta induced by influenza A virus in vitro was evaluated. In studies on anti-influenza activity, our results showed that it maybe target the influenza protein of polymerase basic 1 (PB1), polymerase basic 2 (PB2), polymerase acid (PA), nuclear protein (NP), non-structural protein (NS), and matrix protein (M) but not hemagglutinin (HA) and neuraminidase (NA). In studies on immune regulation, our results demonstrated that pterodontic acid can inhibit the Retinoic acid inducible gene-I (RIG-I) expression in mRNA and protein level at 100 µg/ml, then further to clarify its action on the signalling pathway, The results indicated that pterodontic acid can inhibit the Tumor Necrosis Factor-related Apoptosis-inducing Ligand/Fas Ligand (TRAIL/Fasl) expression in mRNA level at 100 µg/ml; the cleaved caspase 3/7, p-NF-KB, and p-ERK were all suppressed in protein level by pterodontic acid at 100 µg/ml. This confirmed its mechanism that restrained the nuclear export of viral RNPs. The interferon system was also affected, the STAT1, IFN-α, IFN-ß expression were also inhibited by pterodontic acid at 25-100 µg/ml and also, the important programmed death-ligand of PD-L1 and PD-L2 was inhibited at 50-100 µg/ml. The mechanisms of pterodontic acid against influenza virus infection may be a cascade inhibition and it has the anti-inflammatory activity, which has no side effect, and can be as a supplement drug in clinical influenza virus infection.


Assuntos
Antivirais/farmacologia , Asteraceae/química , Antígeno B7-H1/fisiologia , Proteína DEAD-box 58/antagonistas & inibidores , Vírus da Influenza A/efeitos dos fármacos , Interferon Tipo I/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Proteína 2 Ligante de Morte Celular Programada 1/antagonistas & inibidores , Sesquiterpenos/farmacologia , Células A549 , Antígeno B7-H1/antagonistas & inibidores , Humanos , Vírus da Influenza A/fisiologia , Proteína 2 Ligante de Morte Celular Programada 1/fisiologia , Receptores Imunológicos , Ribonucleoproteínas/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores
13.
Mol Ther ; 27(3): 507-517, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30545600

RESUMO

Local immunomodulation can be a promising strategy to augment the efficacy and decrease off-target toxicities associated with cancer treatment. Pancreatic cancer is resistant to immunotherapies due to the immunosuppressive tumor microenvironment. Herein, we investigated a therapeutic approach involving delivery of a short interfering double-stranded RNA (dsRNA), specific to Bcl2, with 5' triphosphate ends, by lipid calcium phosphate nanoparticles, in an orthotopic allograft KPC model of pancreatic cancer. Retinoic acid-inducible gene I (RIG-I)-like receptors can bind to 5' triphosphate dsRNA (ppp dsRNA), a pathogen-associated molecular pattern, producing type I interferon, while Bcl2 silencing can drive apoptosis of cancer cells. Our approach demonstrated a robust enrichment of tumor tissue with therapeutic nanoparticles and enabled a significant tumor growth inhibition, prolonging median overall survival. Nanoparticles encapsulating dual-therapeutic ppp dsRNA allowed strong induction in levels of pro-inflammatory Th1 cytokines, further increasing proportions of CD8+ T cells over regulatory T cells, M1 over M2 macrophages, and decreased levels of immunosuppressive B regulatory and plasma cells in the tumor microenvironment. Thus, these results provide a new immunotherapy approach for pancreatic cancer.


Assuntos
Nanopartículas/química , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Apoptose/fisiologia , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Fosfatos de Cálcio/química , Proteína DEAD-box 58/metabolismo , Feminino , Imunidade Inata/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo
14.
Front Immunol ; 9: 711, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29686682

RESUMO

The innate immune system provides the first line of defense against pathogen infection though also influences pathways involved in cancer immunosurveillance. The innate immune system relies on a limited set of germ line-encoded sensors termed pattern recognition receptors (PRRs), signaling proteins and immune response factors. Cytosolic receptors mediate recognition of danger damage-associated molecular patterns (DAMPs) signals. Once activated, these sensors trigger multiple signaling cascades, converging on the production of type I interferons and proinflammatory cytokines. Recent studies revealed that PRRs respond to nucleic acids (NA) released by dying, damaged, cancer cells, as danger DAMPs signals, and presence of signaling proteins across cancer types suggests that these signaling mechanisms may be involved in cancer biology. DAMPs play important roles in shaping adaptive immune responses through the activation of innate immune cells and immunological response to danger DAMPs signals is crucial for the host response to cancer and tumor rejection. Furthermore, PRRs mediate the response to NA in several vaccination strategies, including DNA immunization. As route of double-strand DNA intracellular entry, DNA immunization leads to expression of key components of cytosolic NA-sensing pathways. The involvement of NA-sensing mechanisms in the antitumor response makes these pathways attractive drug targets. Natural and synthetic agonists of NA-sensing pathways can trigger cell death in malignant cells, recruit immune cells, such as DCs, CD8+ T cells, and NK cells, into the tumor microenvironment and are being explored as promising adjuvants in cancer immunotherapies. In this minireview, we discuss how cGAS-STING and RIG-I-MAVS pathways have been targeted for cancer treatment in preclinical translational researches. In addition, we present a targeted selection of recent clinical trials employing agonists of cytosolic NA-sensing pathways showing how these pathways are currently being targeted for clinical application in oncology.


Assuntos
Imunoterapia , Neoplasias/imunologia , Neoplasias/metabolismo , Ácidos Nucleicos/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Citosol/imunologia , Proteína DEAD-box 58/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , Humanos , Imunidade Inata , Helicase IFIH1 Induzida por Interferon/antagonistas & inibidores , Proteínas de Membrana/agonistas , Proteínas de Membrana/metabolismo , Neoplasias/terapia , Ácidos Nucleicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Vacinas de DNA/genética , Vacinas de DNA/imunologia
15.
Nucleic Acids Res ; 46(10): 5239-5249, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29534222

RESUMO

The emergence of RNA-based therapeutics demands robust and economical methods to produce RNA with few byproducts from aberrant activity. While in vitro transcription using the bacteriophage T7 RNA polymerase is one such popular method, its transcripts are known to display an immune-stimulatory activity that is often undesirable and uncontrollable. We here showed that the immune-stimulatory activity of T7 transcript is contributed by its aberrant activity to initiate transcription from a promoter-less DNA end. This activity results in the production of an antisense RNA that is fully complementary to the intended sense RNA product, and consequently a long double-stranded RNA (dsRNA) that can robustly stimulate a cytosolic pattern recognition receptor, MDA5. This promoter-independent transcriptional activity of the T7 RNA polymerase was observed for a wide range of DNA sequences and lengths, but can be suppressed by altering the transcription reaction with modified nucleotides or by reducing the Mg2+ concentration. The current work thus not only offers a previously unappreciated mechanism by which T7 transcripts stimulate the innate immune system, but also shows that the immune-stimulatory activity can be readily regulated.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Helicase IFIH1 Induzida por Interferon/imunologia , RNA de Cadeia Dupla/metabolismo , Proteínas Virais/metabolismo , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , RNA Polimerases Dirigidas por DNA/genética , Células HEK293 , Humanos , Imunidade Inata/fisiologia , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/metabolismo , Interferon beta/genética , Magnésio/farmacologia , Nucleotídeos/genética , Nucleotídeos/metabolismo , Regiões Promotoras Genéticas , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/imunologia , Receptores Imunológicos , Transcrição Gênica/efeitos dos fármacos , Proteínas Virais/genética
16.
Bioconjug Chem ; 29(3): 742-747, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29350913

RESUMO

Retinoic acid-inducible gene I (RIG-I) is a cytosolic pattern recognition receptor (PRR) that potently activates antiviral innate immunity upon recognition of 5' triphosphorylated double-stranded RNA (pppRNA). Accordingly, RNA ligands of the RIG-I pathway have recently emerged as promising antiviral agents, vaccine adjuvants, and cancer immunotherapeutics. However, RIG-I is expressed constitutively in virtually all cell types, and therefore administration of RIG-I agonists causes risk for systemic inflammation and possible dose-limiting toxicities. Here, we establish proof-of-concept and initial design criteria for pppRNA prodrugs capable of activating the RIG-I pathway in response to specific environmental stimuli. We show that covalent conjugation of poly(ethylene glycol) (PEG) to the 3' end of the complementary strand, i.e., on the same side but opposite strand as the 5' triphosphate group, can generate a synthetic overhang that prevents RIG-I activation. Additionally, conjugation of PEG through a cleavable linker-here, a reducible disulfide bond-allows for removal of the synthetic overhang and restoration of immunostimulatory activity. Furthermore, we demonstrate that blockade of RIG-I activation via synthetic overhangs is dependent on PEG molecular weight, with a critical molecular weight between 550 and 1000 Da required to inhibit activity. Additionally, we demonstrate that blockade of RIG-I activity is conjugation site-dependent, as ligation of PEG to the opposite end of the RNA did not influence ligand activity. Collectively, this work demonstrates that conjugation of synthetic polymer overhangs to pppRNA through cleavable linkers is a viable strategy for the development of environmentally triggerable RIG-I-targeting prodrugs.


Assuntos
Proteína DEAD-box 58/imunologia , Polietilenoglicóis/química , Pró-Fármacos/química , RNA/química , Células A549 , Descoberta de Drogas , Humanos , Interferon beta/imunologia , Fosforilação , Polietilenoglicóis/síntese química , Polietilenoglicóis/farmacologia , Pró-Fármacos/síntese química , Pró-Fármacos/farmacologia , RNA/síntese química , RNA/farmacologia , Receptores Imunológicos , Transdução de Sinais/efeitos dos fármacos
17.
World J Gastroenterol ; 23(32): 5895-5903, 2017 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-28932081

RESUMO

AIM: To investigate the effect of (-)-epigallocatechin-3-gallate (EGCG) on polyinosinic-polycytidylic acid (poly I:C)-triggered intracellular innate immunity against hepatitis C virus (HCV) in hepatocytes. METHODS: A cell culture model of HCV infection was generated by infecting a hepatoma cell line, Huh7, with HCV JFH-1 strain (JFH-1-Huh7). Poly I:C with a high molecular weight and EGCG were used to stimulate the JFH-1-Huh7 cells. Real-time reverse transcription-polymerase chain reaction was used to detect the expression levels of intracellular mRNAs and of intracellular and extracellular HCV RNA. Enzyme-linked immunosorbent assay was used to evaluate the interferon (IFN)-λ1 protein level in the cell culture supernatant. Immunostaining was used to examine HCV core protein expression in Huh7 cells. RESULTS: Our recent study showed that HCV replication could impair poly I:C-triggered intracellular innate immune responses in hepatocytes. In the current study, we showed that EGCG treatment significantly increased the poly I:C-induced expression of Toll-like receptor 3 (TLR3), retinoic acid-inducible gene I, and IFN-λ1 in JFH-1-Huh7 cells. In addition, supplementation with EGCG increased the poly I:C-mediated antiviral activity in JFH-1-Huh7 cells at the intracellular and extracellular HCV RNA and protein levels. Further investigation of the mechanisms showed that EGCG treatment significantly enhanced the poly I:C-induced expression of IFN-regulatory factor 9 and several antiviral IFN-stimulated genes, including ISG15, ISG56, myxovirus resistance A, and 2'-5'-oligoadenylate synthetase 1, which encode the key antiviral elements in the IFN signaling pathway. CONCLUSION: Our observations provide experimental evidence that EGCG has the ability to enhance poly I:C-induced intracellular antiviral innate immunity against HCV replication in hepatocytes.


Assuntos
Antivirais/farmacologia , Catequina/análogos & derivados , Hepacivirus/fisiologia , Hepatite C/imunologia , Imunidade Inata/efeitos dos fármacos , Interferon gama/imunologia , Poli I-C/imunologia , Antivirais/uso terapêutico , Catequina/farmacologia , Catequina/uso terapêutico , Linhagem Celular Tumoral , Proteína DEAD-box 58/imunologia , Proteína DEAD-box 58/metabolismo , Ensaio de Imunoadsorção Enzimática , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Hepatócitos , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Interferon gama/metabolismo , RNA Viral/isolamento & purificação , Receptores Imunológicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Receptor 3 Toll-Like/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologia
18.
J Gen Virol ; 98(6): 1282-1293, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28631605

RESUMO

Retinoic acid inducible gene (RIG-I)-mediated innate immunity plays a pivotal role in defence against virus infections. Previously we have shown that Sendai virus (SeV) defective interfering (DI) RNA functions as an exclusive and potent RIG-I ligand in DI-RNA-rich SeV-Cantell infected cells. To further understand how RIG-I is activated during SeV infection, we used a different interferon (IFN)-inducing SeV strain, recombinant SeVΔC, which, in contrast to SeV-Cantell is believed to stimulate IFN production due to the lack of the SeV IFN antagonist protein C. Surprisingly, we found that in SevΔC-infected cells, DI RNAs also functioned as an exclusive RIG-I ligand. Infections with wild-type SeV failed to generate any RIG-I-associated immunostimulatory RNA and this correlated with the lack of DI genomes in infected cells, as well as with the absence of cellular innate immune responses. Supplementation of the C protein in the context of SeVΔC infection led to a reduction in the number of DI RNAs, further supporting the potential role of the C protein as a negative regulator of DI generation and/or accumulation. Our findings indicate that limiting DI genome production is an important function of viral IFN antagonist proteins.


Assuntos
Proteína DEAD-box 58/metabolismo , Deleção de Genes , Regulação Viral da Expressão Gênica , RNA Interferente Pequeno/metabolismo , Vírus Sendai/imunologia , Proteínas Virais/genética , Adjuvantes Imunológicos/metabolismo , Teste de Complementação Genética , Células HeLa , Humanos , RNA Viral/metabolismo , Receptores Imunológicos , Vírus Sendai/genética
19.
Vaccine ; 35(15): 1964-1971, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28279563

RESUMO

Vaccine adjuvants are essential to drive a protective immune response in cases where vaccine antigens are weakly immunogenic, where vaccine antigen is limited, or where an increase in potency is needed for a specific population, such as the elderly. To discover novel vaccine adjuvants, we used a high-throughput screen (HTS) designed to identify small-molecule agonists of the RIG-I-like receptor (RLR) pathway leading to interferon regulatory factor 3 (IRF3) activation. RLRs are a group of cytosolic pattern-recognition receptors that are essential for the recognition of viral nucleic acids during infection. Upon binding of viral nucleic acid ligands, the RLRs become activated and signal to transcription factors, including IRF3, to initiate an innate immune transcriptional program to control virus infection. Among our HTS hits were a series of benzothiazole compounds from which we designed the lead analog, KIN1148. KIN1148 induced dose-dependent IRF3 nuclear translocation and specific activation of IRF3-responsive promoters. Prime-boost immunization of mice with a suboptimal dose of a monovalent pandemic influenza split virus H1N1 A/California/07/2009 vaccine plus KIN1148 protected against a lethal challenge with mouse-adapted influenza virus (A/California/04/2009) and induced an influenza virus-specific IL-10 and Th2 response by T cells derived from lung and lung-draining lymph nodes. Prime-boost immunization with vaccine plus KIN1148, but not prime immunization alone, induced antibodies capable of inhibiting influenza virus hemagglutinin and neutralizing viral infectivity. Nevertheless, a single immunization with vaccine plus KIN1148 provided increased protection over vaccine alone and reduced viral load in the lungs after challenge. These findings suggest that protection was at least partially mediated by a cellular immune component and that the induction of Th2 and immunoregulatory cytokines by a KIN1148-adjuvanted vaccine may be particularly beneficial for ameliorating the immunopathogenesis that is associated with influenza viruses.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Benzotiazóis/administração & dosagem , Proteína DEAD-box 58/metabolismo , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Fator Regulador 3 de Interferon/metabolismo , Adjuvantes Imunológicos/isolamento & purificação , Animais , Benzotiazóis/isolamento & purificação , Linhagem Celular , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Ensaios de Triagem em Larga Escala , Humanos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/prevenção & controle , Receptores Imunológicos , Análise de Sobrevida
20.
Cell Signal ; 28(1): 25-33, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26518362

RESUMO

Photochemotherapy using 8-methoxypsoralen in combination with UVA radiation (PUVA) is an effective treatment for various skin dermatosis including psoriasis however its molecular mechanism is not clear. Previously we demonstrated that PUVA differentially regulates miRNA expression profile with a significant up-regulation of hsa-miR-4516. To study in detail the molecular mechanism of PUVA in keratinocytes, we investigated the genome wide transcriptomic changes using Illumina whole genome gene expression beadchip. Microarray analysis revealed 1932 differentially expressed gene and their Insilico analysis revealed Retinoic Acid Inducible Gene-I (RIG-1) signaling, apoptosis and p53 pathway to be associated with PUVA induced effects. We demonstrate that miR-4516 mediated down-regulation of UBE2N promotes p53 nuclear translocation and pro-apoptotic activity of PUVA is independent of IRF3 but is mediated by the RIG-I in a p53 and NFκB dependent manner. Additionally, PUVA inactivated the AKT/mTOR pathway in concert with inhibition of autophagy and suppressed cell migration. Taken together this study broadens our understanding about the mechanism of action of PUVA providing possible new strategy targeting proapoptotic function of RIG-1, a regulator of innate immune response or p53 for psoriasis therapy.


Assuntos
Apoptose , RNA Helicases DEAD-box/genética , Perfilação da Expressão Gênica , Queratinócitos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose/efeitos dos fármacos , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Humanos , Fator Regulador 3 de Interferon/metabolismo , Metoxaleno/farmacologia , MicroRNAs/genética , Terapia PUVA/métodos , Receptores Imunológicos , Pele/efeitos dos fármacos , Neoplasias Cutâneas/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA