Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Cytokine Growth Factor Rev ; 60: 28-45, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33992887

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) elicits an interferon (IFN) deficiency state, which aggravates the type I interferon deficiency and slow IFN responses, which associate with e.g. aging and obesity. Additionally, SARS-CoV-2 may also elicit a cytokine storm, which accounts for disease progression and ultimately the urgent need of ventilator support. Based upon several reports, it has been argued that early treatment with IFN-alpha2 or IFN-beta, preferentially in the early disease stage, may prohibit disease progression. Similarly, preliminary studies have shown that JAK1/2 inhibitor treatment with ruxolitinib or baricitinib may decrease mortality by dampening the deadly cytokine storm, which - in addition to the virus itself - also contributes to multi-organ thrombosis and multi-organ failure. Herein, we describe the rationale for treatment with IFNs (alpha2 or beta) and ruxolitinib emphasizing the urgent need to explore these agents in the treatment of SARS-CoV-2 - both as monotherapies and in combination. In this context, we take advantage of several safety and efficacy studies in patients with the chronic myeloproliferative blood cancers (essential thrombocythemia, polycythemia vera and myelofibrosis) (MPNs), in whom IFN-alpha2 and ruxolitinib have been used successfully for the last 10 (ruxolitinib) to 30 years (IFN) as monotherapies and most recently in combination as well. In the context of these agents being highly immunomodulating (IFN boosting immune cells and JAK1/2 inhibitors being highly immunosuppressive and anti-inflammatory), we also discuss if statins and hydroxyurea, both agents possessing anti-inflammatory, antithrombotic and antiviral potentials, might be inexpensive agents to be repurposed in the treatment of SARS-CoV-2.


Assuntos
Tratamento Farmacológico da COVID-19 , Síndrome da Liberação de Citocina/virologia , Interferons/deficiência , Interferons/uso terapêutico , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , SARS-CoV-2/patogenicidade , Animais , COVID-19/imunologia , COVID-19/patologia , Ensaios Clínicos como Assunto , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/patologia , Humanos , SARS-CoV-2/imunologia
2.
Int J Mol Sci ; 22(5)2021 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-33806448

RESUMO

Lambda interferons mediate antiviral immunity by inducing interferon-stimulated genes (ISGs) in epithelial tissues. A common variant rs368234815TT/∆G creating functional gene from an IFNL4 pseudogene is associated with the expression of major ISGs in the liver but impaired clearance of hepatitis C. To explain this, we compared Halo-tagged and non-tagged IFNL3 and IFNL4 signaling in liver-derived cell lines. Transfection with non-tagged IFNL3, non-tagged IFNL4 and Halo-tagged IFNL4 led to a similar degree of JAK-STAT activation and ISG induction; however, the response to transfection with Halo-tagged IFNL3 was lower and delayed. Transfection with non-tagged IFNL3 or IFNL4 induced no transcriptome change in the cells lacking either IL10R2 or IFNLR1 receptor subunits. Cytosolic overexpression of signal peptide-lacking IFNL3 or IFNL4 in wild type cells did not interfere with JAK-STAT signaling triggered by interferons in the medium. Finally, expression profile changes induced by transfection with non-tagged IFNL3 and IFNL4 were highly similar. These data do not support the hypothesis about IFNL4-specific non-canonical signaling and point out that functional studies conducted with tagged interferons should be interpreted with caution.


Assuntos
Hepatócitos/imunologia , Hepatócitos/metabolismo , Interferons/genética , Interferons/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Linhagem Celular , Expressão Gênica , Técnicas de Inativação de Genes , Células Hep G2 , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Interferons/deficiência , Subunidade beta de Receptor de Interleucina-10/deficiência , Subunidade beta de Receptor de Interleucina-10/genética , Subunidade beta de Receptor de Interleucina-10/metabolismo , Interleucinas/deficiência , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Transfecção
3.
Viruses ; 12(12)2020 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-33322160

RESUMO

Innate immune interferons (IFNs), including type I and III IFNs, constitute critical antiviral mechanisms. Recent studies reveal that IFN dysregulation is key to determine COVID-19 pathogenesis. Effective IFN stimulation or prophylactic administration of IFNs at the early stage prior to severe COVID-19 may elicit an autonomous antiviral state, restrict the virus infection, and prevent COVID-19 progression. Inborn genetic flaws and autoreactive antibodies that block IFN response have been significantly associated with about 14% of patients with life-threatening COVID-19 pneumonia. In most severe COVID-19 patients without genetic errors in IFN-relevant gene loci, IFN dysregulation is progressively worsened and associated with the situation of pro-inflammation and immunopathy, which is prone to autoimmunity. In addition, the high correlation of severe COVID-19 with seniority, males, and individuals with pre-existing comorbidities will be plausibly explained by the coincidence of IFN aberrance in these situations. Collectively, current studies call for a better understanding of the IFN response regarding the spatiotemporal determination and subtype-specificity against SARS-CoV-2 infections, which are warranted to devise IFN-related prophylactics and therapies.


Assuntos
Antivirais/imunologia , COVID-19/imunologia , Interferons/imunologia , SARS-CoV-2/patogenicidade , Antivirais/uso terapêutico , COVID-19/patologia , Progressão da Doença , Humanos , Interferons/deficiência , Interferons/uso terapêutico , Cinética , Prognóstico , Transdução de Sinais , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Síndrome de Resposta Inflamatória Sistêmica/patologia , Tratamento Farmacológico da COVID-19
5.
Vet Parasitol ; 280: 109091, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32208305

RESUMO

Neospora caninum is considered one of the main causes of abortion in cattle but can also cause abortion in sheep. There is limited knowledge of the N. caninum population infecting sheep, and only one N. caninum isolate from a pregnant sheep from Japan has been reported. This study describes the in vitro isolation and genetic characterization of two new sheep isolates of N. caninum implicated in ovine reproductive failure. We used IFN-γ-knockout mice inoculated with PCR-positive brain homogenates from two clinically healthy but congenitally infected lambs at 4.5 months of age for parasite isolation. The lambs were born to dams from a sheep farm that had experienced pregnancy failure caused by N. caninum in successive generations. Tachyzoites were microscopically visualized in peritoneal flushes from all inoculated mice and were also observed in MARC-145 cell cultures within one week after inoculation with peritoneal flushes. Two N. caninum isolates, Nc-Spain11 and Nc-Spain12, were obtained from each lamb. The genotyping of the Nc-Spain11 and Nc-Spain12 isolates based on 9 microsatellite markers showed identical multilocus genotype (MLG). Comparison between a previous N. caninum genotype dataset including 80 MLGs from Argentinean, Spanish, Mexican, German and Scottish bovine isolates and the Japanese sheep isolate showed that the Nc-Spain11 and Nc-Spain12 MLG was unique and differed from the other MLGs. eBURST analyses showed that the Nc-Spain11 and Nc-Spain12 MLG was genetically clustered with other bovine MLGs and one ovine MLG, and the nearest genetic relationship was with an MLG from a bovine abortion collected in the same geographical area of Galicia.


Assuntos
Coccidiose/veterinária , Neospora/isolamento & purificação , Doenças dos Ovinos/parasitologia , Animais , Coccidiose/parasitologia , Feminino , Interferons/deficiência , Masculino , Camundongos , Camundongos Knockout/parasitologia , Neospora/genética , Análise de Sequência de DNA/veterinária , Ovinos , Espanha
7.
Rev Med Virol ; 28(6): e2008, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30209859

RESUMO

Increased sensitivity of cancer cells to viruses is a prerequisite for the success of oncolytic virotherapy. One of the major causes of such a phenotype is the disruption of innate antiviral defenses associated with dysfunction of type 1 interferons (IFNs) that permits unlimited replication of viruses in cancer cells. Defects in IFN pathways help cancer progression by providing additional advantages to tumor cells. However, while these defects promote the survival and accelerated proliferation of malignant cells, they facilitate viral replication and thus enhance the efficiency of viral oncolysis. This review describes a broad spectrum of defects in genes that participate in IFN induction and IFN response pathways. Expression levels and/or functional activities of these genes are frequently low or absent in cancer cells, making them sensitive to virus infection. Therefore, certain specific defects in IFN signaling cascades might serve as potential biomarkers to help in identifying individual cancer patients who are likely to benefit from oncolytic virotherapy.


Assuntos
Antineoplásicos/imunologia , Biomarcadores/análise , Interferons/deficiência , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Humanos
8.
J Virol ; 90(23): 10789-10799, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27681121

RESUMO

The interferon (IFN) response to viral pathogens is critical for host survival. In humans and mouse models, defects in IFN responses can result in lethal herpes simplex virus 1 (HSV-1) infections, usually from encephalitis. Although rare, HSV-1 can also cause fulminant hepatic failure, which is often fatal. Although herpes simplex encephalitis has been extensively studied, HSV-1 generalized infections and subsequent acute liver failure are less well understood. We previously demonstrated that IFN-αßγR-/- mice are exquisitely susceptible to liver infection following corneal infection with HSV-1. In this study, we used bone marrow chimeras of IFN-αßγR-/- (AG129) and wild-type (WT; 129SvEv) mice to probe the underlying IFN-dependent mechanisms that control HSV-1 pathogenesis. After infection, WT mice with either IFN-αßγR-/- or WT marrow exhibited comparable survival, while IFN-αßγR-/- mice with WT marrow had a significant survival advantage over their counterparts with IFN-αßγR-/- marrow. Furthermore, using bioluminescent imaging to maximize data acquisition, we showed that the transfer of IFN-competent hematopoietic cells controlled HSV-1 replication and damage in the livers of IFN-αßγR-/- mice. Consistent with this, the inability of IFN-αßγR-/- immune cells to control liver infection in IFN-αßγR-/- mice manifested as profoundly elevated aspartate transaminase (AST) and alanine transaminase (ALT) levels, indicative of severe liver damage. In contrast, IFN-αßγR-/- mice receiving WT marrow exhibited only modest elevations of AST and ALT levels. These studies indicate that IFN responsiveness of the immune system is a major determinant of viral tropism and damage during visceral HSV infections. IMPORTANCE: Herpes simplex virus 1 (HSV-1) infection is an incurable viral infection with the most significant morbidity and mortality occurring in neonates and patients with compromised immune systems. Severe pathologies from HSV include the blindness-inducing herpetic stromal keratitis, highly debilitating and lethal herpes simplex encephalitis, and generalized infections that can lead to herpes simplex virus-induced acute liver failure. While immune compromise is a known factor, the precise mechanisms that lead to generalized HSV infections are unknown. In this study, we used and developed a mouse model system in combination with real-time bioluminescence imaging to demonstrate the relative importance of the immune and nonimmune compartments for containing viral spread and promoting host survival after corneal infection. Our results shed light on the pathogenesis of HSV infections that lead to generalized infection and acute liver failure.


Assuntos
Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/patogenicidade , Interferons/metabolismo , Falência Hepática Aguda/imunologia , Animais , Modelos Animais de Doenças , Feminino , Herpes Simples/etiologia , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Humanos , Hospedeiro Imunocomprometido , Interferons/deficiência , Interferons/genética , Ceratite Herpética/etiologia , Ceratite Herpética/imunologia , Ceratite Herpética/virologia , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Falência Hepática Aguda/etiologia , Falência Hepática Aguda/virologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Quimera por Radiação/imunologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Replicação Viral/imunologia
9.
J Virol ; 90(19): 8592-604, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27440896

RESUMO

UNLABELLED: Hand, foot, and mouth disease (HFMD) has spread throughout the Asia-Pacific region, affecting millions of young children, who develop symptoms ranging from painful blisters around their mouths and hands to neurological complications. Many members of the genus Enterovirus (family Picornaviridae) cause HFMD. Enterovirus 71 (EV71) is one of the primary causative agents and has been linked to severe disease. Vaccine efficacy and pathogenesis studies for EV71 have been limited because there is a lack of suitable animal models. Previously, we generated a mouse-adapted EV71 (mEV71) capable of infecting 12-week-old interferon receptor-deficient AG129 mice and used the model to evaluate the efficacy of candidate HFMD vaccines. Here, we present data investigating the genetic correlates of EV71 adaptation and characterize the virus's tissue tropism in mice. Using reverse genetics, a VP1 mutation (K244E) was shown to be necessary for mEV71 virulence in adult mice. Another VP1 mutation (H37R) was required for mEV71 recovery on rhabdomyosarcoma (RD) cells. Viral loads determined by real-time reverse transcription (RT)-PCR confirmed the presence of mEV71 in the sera and multiple organs of mice. Histological analysis revealed signs of meningitis and encephalitis, characteristic of severe human disease. The further description of this model has provided insight into EV71 pathogenesis and demonstrates the importance of the VP1 region in facilitating mEV71 adaptation. IMPORTANCE: EV71 is a reemerging pathogen, and little is known about the genetic determinants involved in its pathogenesis. The absence of animal models has contributed to this lack of knowledge. The data presented here improve upon the existing animal models by characterizing a mouse-adapted strain of EV71. We determined that a VP1 mutation (K244E) was needed for EV71 virulence in adult AG129 mice. While this mutation was found previously for EV71 adaptation in 5-day-old BALB/c mice, neurotropic disease did not develop. Using interferon-deficient mice, we raised the age of susceptibility beyond 6 weeks and provided clear evidence that our model mimics severe human infections. The model can be exploited to identify determinants of EV71 virulence and to reveal molecular mechanisms that control the virus-host interaction, especially those associated with neurotropic disease. Furthermore, these data provide useful information regarding the importance of VP1, specifically position 244, in host adaptation and tissue dissemination.


Assuntos
Enterovirus Humano A/patogenicidade , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Proteínas Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Adulto , Estruturas Animais/virologia , Animais , Modelos Animais de Doenças , Encefalite Viral/patologia , Encefalite Viral/virologia , Enterovirus Humano A/genética , Humanos , Interferons/deficiência , Meningite Viral/patologia , Meningite Viral/virologia , Camundongos , Proteínas Mutantes/genética , Reação em Cadeia da Polimerase em Tempo Real , Genética Reversa , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Soro/virologia , Carga Viral , Proteínas Estruturais Virais/genética , Virulência , Fatores de Virulência/genética
10.
MAbs ; 7(2): 428-39, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25606664

RESUMO

Anifrolumab (anifrolumab) is an antagonist human monoclonal antibody that targets interferon α receptor 1 (IFNAR1). Anifrolumab has been developed to treat autoimmune diseases and is currently in clinical trials. To decipher the molecular basis of its mechanism of action, we engaged in multiple epitope mapping approaches to determine how it interacts with IFNAR1 and antagonizes the receptor. We identified the epitope of anifrolumab using enzymatic fragmentation, phage-peptide library panning and mutagenesis approaches. Our studies revealed that anifrolumab recognizes the SD3 subdomain of IFNAR1 with the critical residue R(279). Further, we solved the crystal structure of anifrolumab Fab to a resolution of 2.3 Å. Guided by our epitope mapping studies, we then used in silico protein docking of the anifrolumab Fab crystal structure to IFNAR1 and characterized the corresponding mode of binding. We find that anifrolumab sterically inhibits the binding of IFN ligands to IFNAR1, thus blocking the formation of the ternary IFN/IFNAR1/IFNAR2 signaling complex. This report provides the molecular basis for the mechanism of action of anifrolumab and may provide insights toward designing antibody therapies against IFNAR1.


Assuntos
Anticorpos Monoclonais/química , Mapeamento de Epitopos , Epitopos/química , Biblioteca de Peptídeos , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/química , Substituição de Aminoácidos , Animais , Anticorpos Monoclonais/uso terapêutico , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/metabolismo , Células CHO , Cricetinae , Cricetulus , Epitopos/genética , Interferons/antagonistas & inibidores , Interferons/química , Interferons/deficiência , Interferons/metabolismo , Masculino , Mutação de Sentido Incorreto , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo
11.
PLoS Negl Trop Dis ; 8(6): e2933, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24901990

RESUMO

The new world arenavirus Junín virus (JUNV) is the causative agent of Argentine hemorrhagic fever, a lethal human infectious disease. Adult laboratory mice are generally resistant to peripheral infection by JUNV. The mechanism underlying the mouse resistance to JUNV infection is largely unknown. We have reported that interferon receptor knockout mice succumb to JUNV infection, indicating the critical role of interferon in restricting JUNV infection in mice. Here we report that the pathogenic and vaccine strains of JUNV were highly sensitive to interferon in murine primary cells. Treatment with low concentrations of interferon abrogated viral NP protein expression in murine cells. The replication of both JUNVs was enhanced in IRF3/IRF7 deficient cells. In addition, the vaccine strain of JUNV displayed impaired growth in primary murine cells. Our data suggested a direct and potent role of host interferon response in restricting JUNV replication in mice. The defect in viral growth for vaccine JUNV might also partially explain its attenuation in mice.


Assuntos
Antivirais/farmacologia , Interferons/imunologia , Interferons/farmacologia , Vírus Junin/efeitos dos fármacos , Vírus Junin/imunologia , Animais , Células Cultivadas , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 7 de Interferon/deficiência , Interferons/deficiência , Vírus Junin/crescimento & desenvolvimento , Vírus Junin/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Virais/biossíntese , Replicação Viral/efeitos dos fármacos
12.
Vaccine ; 32(2): 258-64, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24252694

RESUMO

Japanese encephalitis virus (JEV) is the most common cause of viral encephalitis in Asia, and it is increasingly a global public health concern due to its recent geographic expansion. While commercial vaccines are available and used in some endemic countries, JEV continues to be a public health problem, with 50,000 cases reported annually. Research with virulent JEV in mouse models to develop new methods of prevention and treatment is restricted to BSL-3 containment facilities, confining these studies to investigators with access to these facilities. We have developed an adult small animal peripheral challenge model using interferon-deficient AG129 mice and the JEV live-attenuated vaccine SA14-14-2, thus requiring only BSL-2 containment. A low dose of virus (10PFU/0.1ml) induced 100% morbidity in infected mice. Increased body temperatures measured by implantable temperature transponders correlated with an increase in infectious virus and viral RNA in serum, spleen and brain as well as an increase in pro-inflammatory markers measured by a 58-biomarker multi-analyte profile (MAP) constructed during the course of infection. In the future, the MAP measurements can be used as a baseline for comparison in order to better assess the inhibition of disease progression by other prophylactic and therapeutic agents. The use of the AG129/JEV SA14-14-2 animal model makes vaccine and therapeutic studies feasible for laboratories with limited biocontainment facilities.


Assuntos
Modelos Animais de Doenças , Encefalite Japonesa/prevenção & controle , Vacinas contra Encefalite Japonesa/administração & dosagem , Animais , Vírus da Encefalite Japonesa (Espécie)/classificação , Interferons/deficiência , Camundongos , Vacinas Atenuadas/administração & dosagem , Carga Viral
13.
Georgian Med News ; (212): 24-32, 2012 Nov.
Artigo em Russo | MEDLINE | ID: mdl-23221135

RESUMO

Interferons deficiency has a negative influence on the development of infection and inflammation in general. The use in the complex of anti-inflammatory therapy of interferon inducers (Meglumine acridоnacetate, Tilorone), combining antiviral, immunomodulatory, interferon correction effects with etiopathogenic action leads to the correction of the interferon system defects and eliminate etiological infectious agents, that is confirmed by laboratory data and clinical efficacy.


Assuntos
Doenças dos Genitais Femininos/tratamento farmacológico , Doenças dos Genitais Femininos/imunologia , Indutores de Interferon/uso terapêutico , Interferons/deficiência , Adulto , Anti-Inflamatórios/uso terapêutico , Antivirais/uso terapêutico , Doença Crônica , Feminino , Doenças dos Genitais Femininos/microbiologia , Humanos , Meglumina/uso terapêutico , Pessoa de Meia-Idade , Doença Inflamatória Pélvica/tratamento farmacológico , Doença Inflamatória Pélvica/imunologia , Doença Inflamatória Pélvica/microbiologia , Tilorona/uso terapêutico , Cervicite Uterina/tratamento farmacológico , Cervicite Uterina/imunologia , Cervicite Uterina/microbiologia , Vaginite/tratamento farmacológico , Vaginite/imunologia , Vaginite/microbiologia , Vulvite/tratamento farmacológico , Vulvite/imunologia , Vulvite/microbiologia , Adulto Jovem
14.
Thorax ; 67(6): 517-25, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22213737

RESUMO

BACKGROUND: Rhinoviruses are important triggers of pulmonary exacerbations and possible contributors to long-term respiratory morbidity in cystic fibrosis (CF), but mechanisms leading to rhinovirus-induced CF exacerbations are poorly understood. It is hypothesised that there is a deficient innate immune response of the airway epithelium towards rhinovirus infection in CF. METHODS: Early innate immune responses towards rhinoviruses (RV-16, major-type and RV-1B, minor-type) in CF and non-CF bronchial epithelial cell lines and primary nasal and bronchial epithelial cells from patients with CF (n=13) and healthy controls (n=24) were studied. RESULTS: Rhinovirus RNA expression and virus release into supernatants was increased more than tenfold in CF cells compared with controls. CF cells expressed up to 1000 times less interferon (IFN) type I (ß) and type III (λ) mRNA and produced less than half of IFN-ß and IFN-λ protein compared with controls. In contrast, interleukin 8 production was not impaired, indicating a selective deficiency in the innate antiviral defence system. Deficient IFN production was paralleled by lower expression of IFN-stimulated genes including myxovirus resistance A, 2',5'-oligoadenylate synthetase, viperin and nitric oxide synthase 2. Addition of exogenous type I and III IFNs, particularly IFN-ß, restored antiviral pathways and virus control in CF cells, underscoring the crucial role of these molecules. CONCLUSIONS: This study describes a novel mechanism to explain the increased susceptibility of patients with CF to rhinovirus infections. A profound impairment of the antiviral early innate response in CF airway epithelial cells was identified, suggesting a potential use of IFNs in the treatment of rhinovirus-induced CF exacerbations.


Assuntos
Antivirais/farmacologia , Brônquios/imunologia , Fibrose Cística/imunologia , Interferon beta/imunologia , Interferons/deficiência , Interleucinas/imunologia , Infecções por Picornaviridae/imunologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , Rhinovirus/imunologia , Doença Aguda , Antivirais/imunologia , Brônquios/citologia , Brônquios/virologia , Células Cultivadas , Fibrose Cística/tratamento farmacológico , Fibrose Cística/virologia , Suscetibilidade a Doenças , Células Epiteliais/imunologia , Células Epiteliais/virologia , Humanos , Imunidade Inata/efeitos dos fármacos , Interferon Tipo I/imunologia , Interferon beta/deficiência , Interferon beta/farmacologia , Interferons/imunologia , Interleucinas/deficiência , Infecções por Picornaviridae/tratamento farmacológico , Infecções por Picornaviridae/metabolismo , Infecções por Picornaviridae/virologia , Podócitos/imunologia , Rhinovirus/efeitos dos fármacos , Resultado do Tratamento
15.
Vaccine ; 30(8): 1513-20, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22178727

RESUMO

Formulations of chimeric dengue vaccine (DENVax) viruses containing the pre-membrane (prM) and envelope (E) genes of serotypes 1-4 expressed in the context of the attenuated DENV-2 PDK-53 genome were tested for safety, immunogenicity and efficacy in interferon receptor knock-out mice (AG129). Monovalent formulations were safe and elicited robust neutralizing antibody responses to the homologous virus and only limited cross-reactivity to other serotypes. A single dose of monovalent DENVax-1, -2, or -3 vaccine provided eighty or greater percent protection against both wild-type (wt) DENV-1 (Mochizuki strain) and DENV-2 (New Guinea C strain) challenge viruses. A single dose of monovalent DENVax-4 also provided complete protection against wt DENV-1 challenge and significantly increased the survival times after challenge with wt DENV-2. In studies using tetravalent mixtures, DENVax ratios were identified that: (i) caused limited viremia, (ii) induced serotype-specific neutralizing antibodies to all four DENV serotypes with different hierarchies, and (iii) conferred full protection against clinical signs of disease following challenge with either wt DENV-1 or DENV-2 viruses. Overall, these data highlight the immunogenic profile of DENVax, a novel candidate tetravalent dengue vaccine and the advantage of sharing a common attenuated genomic backbone among the DENVax monovalent vaccines that confer protection against homologous or heterologous virus challenge.


Assuntos
Vacinas contra Dengue/efeitos adversos , Vacinas contra Dengue/imunologia , Interferons/deficiência , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Temperatura Corporal , Peso Corporal , Dengue/mortalidade , Dengue/patologia , Dengue/prevenção & controle , Vacinas contra Dengue/administração & dosagem , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Análise de Sobrevida , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia , Carga Viral , Viremia/prevenção & controle
16.
Curr Opin Virol ; 1(6): 487-96, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22347990

RESUMO

The three types of interferon (IFNs) are essential for immunity against at least some viruses in the mouse model of experimental infections, type I IFNs displaying the broadest and strongest anti-viral activity. Consistently, human genetic studies have shown that type II IFN is largely redundant for immunity against viruses in the course of natural infections. The precise contributions of human type I and III IFNs remain undefined. However, various inborn errors of anti-viral IFN immunity have been described, which can result in either broad or narrow immunological and viral phenotypes. The broad disorders impair the response to (STAT1, TYK2) or the production of at least type I and type III IFNs following multiple stimuli (NEMO), resulting in multiple viral infections at various sites, including herpes simplex encephalitis (HSE). The narrow disorders impair exclusively (TLR3) or mostly (UNC-93B, TRIF, TRAF3) the TLR3-dependent induction of type I and III IFNs, leading to HSE in apparently otherwise healthy individuals. These recent discoveries highlight the importance of human type I and III IFNs in protective immunity against viruses, including the TLR3-IFN pathway in protection against HSE.


Assuntos
Encefalite por Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Interferons/imunologia , Animais , Encefalite por Herpes Simples/virologia , Humanos , Interferons/deficiência , Interferons/genética
17.
Nature ; 468(7322): 452-6, 2010 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21085181

RESUMO

Cellular messenger RNA (mRNA) of higher eukaryotes and many viral RNAs are methylated at the N-7 and 2'-O positions of the 5' guanosine cap by specific nuclear and cytoplasmic methyltransferases (MTases), respectively. Whereas N-7 methylation is essential for RNA translation and stability, the function of 2'-O methylation has remained uncertain since its discovery 35 years ago. Here we show that a West Nile virus (WNV) mutant (E218A) that lacks 2'-O MTase activity was attenuated in wild-type primary cells and mice but was pathogenic in the absence of type I interferon (IFN) signalling. 2'-O methylation of viral RNA did not affect IFN induction in WNV-infected fibroblasts but instead modulated the antiviral effects of IFN-induced proteins with tetratricopeptide repeats (IFIT), which are interferon-stimulated genes (ISGs) implicated in regulation of protein translation. Poxvirus and coronavirus mutants that lacked 2'-O MTase activity similarly showed enhanced sensitivity to the antiviral actions of IFN and, specifically, IFIT proteins. Our results demonstrate that the 2'-O methylation of the 5' cap of viral RNA functions to subvert innate host antiviral responses through escape of IFIT-mediated suppression, and suggest an evolutionary explanation for 2'-O methylation of cellular mRNA: to distinguish self from non-self RNA. Differential methylation of cytoplasmic RNA probably serves as an example for pattern recognition and restriction of propagation of foreign viral RNA in host cells.


Assuntos
Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica/imunologia , Imunidade Inata/imunologia , Interferons/imunologia , Proteínas/metabolismo , Capuzes de RNA/metabolismo , RNA Viral/metabolismo , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Células Cultivadas , Coronavirus/enzimologia , Coronavirus/genética , Coronavirus/imunologia , Coronavirus/fisiologia , Fibroblastos , Regulação da Expressão Gênica/genética , Humanos , Imunidade Inata/genética , Interferons/deficiência , Interferons/genética , Metilação , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Modelos Imunológicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Poxviridae/enzimologia , Poxviridae/genética , Poxviridae/imunologia , Poxviridae/fisiologia , Biossíntese de Proteínas/imunologia , Proteínas/genética , Capuzes de RNA/genética , Capuzes de RNA/imunologia , RNA Viral/genética , RNA Viral/imunologia , Proteínas de Ligação a RNA , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Taxa de Sobrevida , Replicação Viral , Vírus do Nilo Ocidental/enzimologia , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia
18.
FEMS Immunol Med Microbiol ; 55(3): 422-31, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19243434

RESUMO

Streptococcus pyogenes is a major causative agent of tonsillitis or pharyngitis in children. Streptococcus pyogenes can persist in tonsils, and one-third of children treated with antibiotics continue to shed streptococci and have recurrent infections. Mouse nasal-associated lymphoid tissue (NALT) is functionally analogous to human oropharyngeal lymphoid tissues, and serves as a model for characterization of the mucosal innate immune response to S. pyogenes. Wild-type S. pyogenes induces transcription of both type I and interferon-gamma (IFN-gamma)-responsive genes, proinflammatory genes and acute-phase response proteins 24 h after intranasal infection. Invasion of NALT and the induction of the interferon response were not dependent on expression of antiphagocytic M protein. Intranasal infection induces a substantial influx of neutrophils into NALT at 24 h, which declines by 48 h after infection. Infection of IFN-gamma(-/-) [IFN-gamma knock-out mouse (GKO)] C57BL/6 mice with wild-type S. pyogenes resulted in local dissemination of bacteria to draining lymph nodes (LN), but did not lead to systemic infection by 48 h after infection. Infected GKO mice had an increased influx of neutrophils into NALT compared with immunocompetent mice. Thus, IFN-gamma-induced responses are required to prevent local dissemination of streptococci to the draining LN.


Assuntos
Interferons/biossíntese , Tecido Linfoide/imunologia , Tecido Linfoide/microbiologia , Streptococcus pyogenes/imunologia , Animais , Sobrevivência Celular , Contagem de Colônia Microbiana , Feminino , Perfilação da Expressão Gênica , Interferons/deficiência , Linfonodos/microbiologia , Tecido Linfoide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa Nasal/microbiologia , Neutrófilos/imunologia
19.
Virol J ; 5: 3, 2008 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-18190677

RESUMO

BACKGROUND: The competitiveness of two Tula hantavirus (TULV) isolates, TULV/Lodz and TULV/Moravia, was evaluated in interferon (IFN) -competent and IFN-deficient cells. The two isolates differ in the length of the open reading frame (ORF) encoding the nonstructural protein NSs, which has previously been shown to inhibit IFN response in infected cells. RESULTS: In IFN-deficient Vero E6 cells both TULV isolates survived equally well. In contrast, in IFN-competent MRC5 cells TULV/Lodz isolate, that possesses the NSs ORF for the full-length protein of 90 aa, survived for more consequent passages than TULV/Moravia isolate, which contains the ORF for truncated NSs protein (66-67 aa). CONCLUSION: Our data show that expression of a full-length NSs protein is beneficial for the virus survival and competitiveness in IFN-competent cells and not essential in IFN-deficient cells. These results suggest that the N-terminal aa residues are important for the full activity of the NSs protein.


Assuntos
Infecções por Hantavirus/virologia , Interferons/metabolismo , Orthohantavírus/química , Proteínas não Estruturais Virais/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Orthohantavírus/genética , Orthohantavírus/crescimento & desenvolvimento , Infecções por Hantavirus/imunologia , Humanos , Interferons/deficiência , Interferons/genética , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Alinhamento de Sequência , Inoculações Seriadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...