Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 35(10): e21917, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34533865

RESUMO

The resolution of inflammation facilitates proper wound healing and limits tissue repair short of exaggerated fibrotic scarring. The atypical chemokine receptor (ACKR)2/D6 scavenges inflammatory chemokines, while IFN-ß is a recently unveiled pro-resolving cytokine. Both effector molecules limit acute inflammatory episodes and promote their resolution in various organs. Here, we found fibrotic skin lesions from ACKR2-/- mice presented increased epidermal and dermal thickening, atrophy of the subcutaneous adipose tissue, augmented disorientation of collagen deposition, and enhanced deformation and loss of hair follicles compared to WT counterparts. In addition, affected skin sections from ACKR2-/- mice contained reduced levels of the pro-resolving mediators IFN-ß and IL-10, but increased levels of the pro-inflammatory chemokines CCL2 and 3, the pro-fibrotic cytokine TGF-ß, and the immune-stimulating cytokine IL-12. Notably, treatment with exogenous IFN-ß rescued, at least in part, all the pro-fibrotic outcomes and lesion size in ACKR2-/- mice and promoted expression of the pro-resolving enzyme 12/15-lipoxygenase (LO) in both ACKR2-/- and WT mice. Moreover, Ifnb-/- mice displayed enhanced pro-fibrotic indices upon exposure to bleomycin. These findings suggest ACKR2 is an important mediator in limiting inflammatory skin fibrosis and acts via IFN-ß production to promote the resolution of inflammation and minimize tissue scaring.


Assuntos
Alopecia/metabolismo , Fibrose/metabolismo , Interferon beta/metabolismo , Receptores de Quimiocinas/metabolismo , Pele/metabolismo , Alopecia/genética , Alopecia/patologia , Animais , Colágeno/metabolismo , Fibrose/genética , Fibrose/patologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Interferon beta/deficiência , Interferon beta/genética , Interleucina-10/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Receptores de Quimiocinas/deficiência , Receptores de Quimiocinas/genética , Pele/patologia
2.
Pathog Dis ; 79(1)2021 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-33289808

RESUMO

A vast proportion of coronavirus disease 2019 (COVID-19) individuals remain asymptomatic and can shed severe acute respiratory syndrome (SARS-CoV) type 2 virus to transmit the infection, which also explains the exponential increase in the number of COVID-19 cases globally. Furthermore, the rate of recovery from clinical COVID-19 in certain pockets of the globe is surprisingly high. Based on published reports and available literature, here, we speculated a few immunovirological mechanisms as to why a vast majority of individuals remain asymptomatic similar to exotic animal (bats and pangolins) reservoirs that remain refractile to disease development despite carrying a huge load of diverse insidious viral species, and whether such evolutionary advantage would unveil therapeutic strategies against COVID-19 infection in humans. Understanding the unique mechanisms that exotic animal species employ to achieve viral control, as well as inflammatory regulation, appears to hold key clues to the development of therapeutic versatility against COVID-19.


Assuntos
COVID-19/imunologia , Síndrome da Liberação de Citocina/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Receptores KIR/imunologia , Receptores Semelhantes a Lectina de Células NK/imunologia , Zoonoses/imunologia , Animais , Animais Exóticos/virologia , Doenças Assintomáticas , COVID-19/genética , COVID-19/transmissão , COVID-19/virologia , Quirópteros/virologia , Síndrome da Liberação de Citocina/genética , Síndrome da Liberação de Citocina/prevenção & controle , Síndrome da Liberação de Citocina/virologia , Reservatórios de Doenças , Eutérios/virologia , Expressão Gênica , Especificidade de Hospedeiro , Humanos , Tolerância Imunológica , Imunidade Inata , Interferon beta/deficiência , Interferon beta/genética , Interferon beta/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Monócitos/imunologia , Monócitos/virologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Receptores KIR/deficiência , Receptores KIR/genética , Receptores Semelhantes a Lectina de Células NK/deficiência , Receptores Semelhantes a Lectina de Células NK/genética , SARS-CoV-2/patogenicidade , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Zoonoses/genética , Zoonoses/transmissão , Zoonoses/virologia
3.
J Neurosci ; 40(11): 2357-2370, 2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32029532

RESUMO

DNA damage and type I interferons (IFNs) contribute to inflammatory responses after traumatic brain injury (TBI). TBI-induced activation of microglia and peripherally-derived inflammatory macrophages may lead to tissue damage and neurological deficits. Here, we investigated the role of IFN-ß in secondary injury after TBI using a controlled cortical impact model in adult male IFN-ß-deficient (IFN-ß-/-) mice and assessed post-traumatic neuroinflammatory responses, neuropathology, and long-term functional recovery. TBI increased expression of DNA sensors cyclic GMP-AMP synthase and stimulator of interferon genes in wild-type (WT) mice. IFN-ß and other IFN-related and neuroinflammatory genes were also upregulated early and persistently after TBI. TBI increased expression of proinflammatory mediators in the cortex and hippocampus of WT mice, whereas levels were mitigated in IFN-ß-/- mice. Moreover, long-term microglia activation, motor, and cognitive function impairments were decreased in IFN-ß-/- TBI mice compared with their injured WT counterparts; improved neurological recovery was associated with reduced lesion volume and hippocampal neurodegeneration in IFN-ß-/- mice. Continuous central administration of a neutralizing antibody to the IFN-α/ß receptor (IFNAR) for 3 d, beginning 30 min post-injury, reversed early cognitive impairments in TBI mice and led to transient improvements in motor function. However, anti-IFNAR treatment did not improve long-term functional recovery or decrease TBI neuropathology at 28 d post-injury. In summary, TBI induces a robust neuroinflammatory response that is associated with increased expression of IFN-ß and other IFN-related genes. Inhibition of IFN-ß reduces post-traumatic neuroinflammation and neurodegeneration, resulting in improved neurological recovery. Thus, IFN-ß may be a potential therapeutic target for TBI.SIGNIFICANCE STATEMENT TBI frequently causes long-term neurological and psychiatric changes in head injury patients. TBI-induced secondary injury processes including persistent neuroinflammation evolve over time and can contribute to chronic neurological impairments. The present study demonstrates that TBI is followed by robust activation of type I IFN pathways, which have been implicated in microglial-associated neuroinflammation and chronic neurodegeneration. We examined the effects of genetic or pharmacological inhibition of IFN-ß, a key component of type I IFN mechanisms to address its role in TBI pathophysiology. Inhibition of IFN-ß signaling resulted in reduced neuroinflammation, attenuated neurobehavioral deficits, and limited tissue loss long after TBI. These preclinical findings suggest that IFN-ß may be a potential therapeutic target for TBI.


Assuntos
Dano Encefálico Crônico/fisiopatologia , Lesões Encefálicas Traumáticas/fisiopatologia , Interferon beta/fisiologia , Degeneração Neural/etiologia , Animais , Dano Encefálico Crônico/etiologia , Lesões Encefálicas Traumáticas/complicações , Córtex Cerebral/metabolismo , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica , Hipocampo/metabolismo , Inflamação , Interferon beta/biossíntese , Interferon beta/deficiência , Interferon beta/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/etiologia , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/fisiologia , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/fisiopatologia , Distribuição Aleatória , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais , Regulação para Cima
4.
J Virol ; 94(1)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31619554

RESUMO

Type I interferons (IFNs) are key mediators of the innate immune response. Although members of this family of cytokines signal through a single shared receptor, biochemical and functional variation exists in response to different IFN subtypes. While previous work has demonstrated that type I IFNs are essential to control infection by chikungunya virus (CHIKV), a globally emerging alphavirus, the contributions of individual IFN subtypes remain undefined. To address this question, we evaluated CHIKV pathogenesis in mice lacking IFN-ß (IFN-ß knockout [IFN-ß-KO] mice or mice treated with an IFN-ß-blocking antibody) or IFN-α (IFN regulatory factor 7 knockout [IRF7-KO] mice or mice treated with a pan-IFN-α-blocking antibody). Mice lacking either IFN-α or IFN-ß developed severe clinical disease following infection with CHIKV, with a marked increase in foot swelling compared to wild-type mice. Virological analysis revealed that mice lacking IFN-α sustained elevated infection in the infected ankle and in distant tissues. In contrast, IFN-ß-KO mice displayed minimal differences in viral burdens within the ankle or at distal sites and instead had an altered cellular immune response. Mice lacking IFN-ß had increased neutrophil infiltration into musculoskeletal tissues, and depletion of neutrophils in IFN-ß-KO but not IRF7-KO mice mitigated musculoskeletal disease caused by CHIKV. Our findings suggest disparate roles for the IFN subtypes during CHIKV infection, with IFN-α limiting early viral replication and dissemination and IFN-ß modulating neutrophil-mediated inflammation.IMPORTANCE Type I interferons (IFNs) possess a range of biological activity and protect against a number of viruses, including alphaviruses. Despite signaling through a shared receptor, there are established biochemical and functional differences among the IFN subtypes. The significance of our research is in demonstrating that IFN-α and IFN-ß both have protective roles during acute chikungunya virus (CHIKV) infection but do so by distinct mechanisms. IFN-α limits CHIKV replication and dissemination, whereas IFN-ß protects from CHIKV pathogenesis by limiting inflammation mediated by neutrophils. Our findings support the premise that the IFN subtypes have distinct biological activities in the antiviral response.


Assuntos
Febre de Chikungunya/genética , Vírus Chikungunya/patogenicidade , Fator Regulador 7 de Interferon/genética , Interferon-alfa/genética , Interferon beta/genética , Neutrófilos/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Osso e Ossos/imunologia , Osso e Ossos/patologia , Osso e Ossos/virologia , Febre de Chikungunya/imunologia , Febre de Chikungunya/patologia , Febre de Chikungunya/virologia , Vírus Chikungunya/imunologia , Feminino , Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Inflamação , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/imunologia , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/deficiência , Interferon-alfa/imunologia , Interferon beta/antagonistas & inibidores , Interferon beta/deficiência , Interferon beta/imunologia , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Músculo Esquelético/virologia , Infiltração de Neutrófilos , Neutrófilos/patologia , Neutrófilos/virologia , Tarso Animal/imunologia , Tarso Animal/patologia , Tarso Animal/virologia , Replicação Viral
5.
Sci Rep ; 8(1): 4248, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29523863

RESUMO

Defective production of antiviral interferon (IFN)-ß is thought to contribute to rhinovirus-induced asthma exacerbations. These exacerbations are associated with elevated lung levels of lactate dehydrogenase (LDH), indicating occurrence of cell necrosis. We thus hypothesized that reduced lung IFN-ß could contribute to necrotic cell death in a model of asthma exacerbations. Wild-type and IFN-ß-/- mice were given saline or house dust mite (HDM) intranasally for 3 weeks to induce inflammation. Double-stranded RNA (dsRNA) was then given for additional 3 days to induce exacerbation. HDM induced an eosinophilic inflammation, which was not associated with increased expression of cleaved caspase-3, cleaved PARP or elevated bronchoalveolar lavage fluid (BALF) LDH levels in wild-type. However, exacerbation evoked by HDM + dsRNA challenges increased BALF levels of LDH, apoptotic markers and the necroptotic markers receptor-interacting protein (RIP)-3 and phosphorylation of mixed linage kinase domain-like protein (pMLKL), compared to HDM + saline. Absence of IFN-ß at exacerbation further increased BALF LDH and protein expression of pMLKL compared to wild-type. We demonstrate that cell death markers are increased at viral stimulus-induced exacerbation in mouse lungs, and that absence of IFN-ß augments markers of necroptotic cell death at exacerbation. Our data thus suggest a novel role of deficient IFN-ß production at viral-induced exacerbation.


Assuntos
Apoptose , Asma/metabolismo , Interferon beta/deficiência , Proteínas Quinases/metabolismo , Animais , Caspase 3/metabolismo , Feminino , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Necrose , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
6.
J Neurovirol ; 24(3): 273-290, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29476408

RESUMO

Zika virus (ZIKV) has received widespread attention because of its effect on the developing fetus. It is becoming apparent, however, that severe neurological sequelae, such as Guillian-Barrë syndrome (GBS), myelitis, encephalitis, and seizures can occur after infection of adults. This study demonstrates that a contemporary strain of ZIKV can widely infect astrocytes and neurons in the brain and spinal cord of adult, interferon α/ß receptor knockout mice (AG129 strain) and cause progressive hindlimb paralysis, as well as severe seizure-like activity during the acute phase of disease. The severity of hindlimb motor deficits correlated with increased numbers of ZIKV-infected lumbosacral spinal motor neurons and decreased numbers of spinal motor neurons. Electrophysiological compound muscle action potential (CMAP) amplitudes in response to stimulation of the lumbosacral spinal cord were reduced when obvious motor deficits were present. ZIKV immunoreactivity was high, intense, and obvious in tissue sections of the brain and spinal cord. Infection in the brain and spinal cord was also associated with astrogliosis as well as T cell and neutrophil infiltration. CMAP and histological analysis indicated that peripheral nerve and muscle functions were intact. Consequently, motor deficits in these circumstances appear to be primarily due to myelitis and possibly encephalitis as opposed to a peripheral neuropathy or a GBS-like syndrome. Thus, acute ZIKV infection of adult AG129 mice may be a useful model for ZIKV-induced myelitis, encephalitis, and seizure activity.


Assuntos
Encefalite/fisiopatologia , Transtornos Motores/fisiopatologia , Mielite/fisiopatologia , Convulsões/fisiopatologia , Infecção por Zika virus/fisiopatologia , Zika virus/patogenicidade , Potenciais de Ação/fisiologia , Animais , Astrócitos/imunologia , Astrócitos/patologia , Astrócitos/virologia , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/virologia , Modelos Animais de Doenças , Encefalite/imunologia , Encefalite/virologia , Feminino , Humanos , Interferon-alfa/deficiência , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/deficiência , Interferon beta/genética , Interferon beta/imunologia , Interferon gama/deficiência , Interferon gama/genética , Interferon gama/imunologia , Masculino , Camundongos , Camundongos Knockout , Transtornos Motores/imunologia , Transtornos Motores/virologia , Neurônios Motores/imunologia , Neurônios Motores/patologia , Neurônios Motores/virologia , Músculo Esquelético/fisiologia , Mielite/imunologia , Mielite/virologia , Neutrófilos/imunologia , Neutrófilos/patologia , Neutrófilos/virologia , Convulsões/imunologia , Convulsões/virologia , Medula Espinal/imunologia , Medula Espinal/patologia , Medula Espinal/virologia , Linfócitos T/imunologia , Linfócitos T/patologia , Linfócitos T/virologia , Zika virus/crescimento & desenvolvimento , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
7.
Immunology ; 152(3): 439-450, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28646573

RESUMO

CD11c+ dendritic cells (DCs) exert a critical role as antigen-presenting cells in regulating pathogenic T cells in multiple sclerosis (MS). To determine whether the therapeutic benefit of interferon-ß (IFN-ß) treatment for MS is in part influenced by IFN regulation of DC function, we examined the immunophenotype of DCs derived from IFN-ß+/+ and IFN-ß-/- mice using a myelin oligodendrocyte glycoprotein (MOG) peptide-induced mouse model of MS, experimental autoimmune encephalomyelitis (EAE). Our earlier work identified that IFN-ß-/- mice exhibit earlier onset and more rapid progression of neurological impairment compared with IFN-ß+/+ mice. In this study we show that lipopolysaccharide-/MOG peptide-stimulated IFN-ß-/- DCs secrete cytokines associated with pathological T helper type 17 rather than regulatory T-cell polarization and exhibit increased CD80 and MHCII expression when compared with stimulated IFN-ß+/+ DCs. IFN-ß-/- DCs from mice immunized to develop EAE induce greater proliferation of MOG-transgenic CD4+ T cells and promote interleukin-17 production by these T cells. Adoptive transfer of MOG peptide-primed IFN-ß-/- DCs into IFN-ß+/+ and IFN-ß-/- mice immunized to develop EAE resulted in their rapid migration into the central nervous system of recipient mice, before onset of disease, which we attribute to failed signal transducer and activator of transcription 1-mediated inhibition of CCR7. Taken together, our data support immunoregulatory roles for IFN-ß in the activation and migration of DCs during EAE.


Assuntos
Quimiotaxia , Células Dendríticas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Interferon beta/metabolismo , Transferência Adotiva , Animais , Antígeno CD11c/imunologia , Antígeno CD11c/metabolismo , Linhagem da Célula , Plasticidade Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/imunologia , Células Dendríticas/transplante , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Predisposição Genética para Doença , Interferon beta/deficiência , Interferon beta/genética , Interferon beta/imunologia , Lipopolissacarídeos/imunologia , Ativação Linfocitária , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/imunologia , Fragmentos de Peptídeos/imunologia , Fenótipo , Receptores CCR7/imunologia , Receptores CCR7/metabolismo , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
9.
J Virol ; 89(5): 2731-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25540366

RESUMO

UNLABELLED: Previously we found that following intranasal (i.n.) infection with neurotropic vesicular stomatitis virus (VSV) type I interferon receptor (IFNAR) triggering of neuroectodermal cells was critically required to constrain intracerebral virus spread. To address whether locally active IFN-ß was induced proximally, we studied spatiotemporal conditions of VSV-mediated IFN-ß induction. To this end, we performed infection studies with IFN-ß reporter mice. One day after intravenous (i.v.) VSV infection, luciferase induction was detected in lymph nodes. Upon i.n. infection, luciferase induction was discovered at similar sites with delayed kinetics, whereas on days 3 and 4 postinfection enhanced luciferase expression additionally was detected in the foreheads of reporter mice. A detailed analysis of cell type-specific IFN-ß reporter mice revealed that within the olfactory bulb IFN-ß was expressed by neuroectodermal cells, primarily by astrocytes and to a lesser extent by neurons. Importantly, locally induced type I IFN triggered distal parts of the brain as indicated by the analysis of ISRE-eGFP mice which after i.n. VSV infection showed enhanced green fluorescent protein (eGFP) expression throughout the brain. Compared to wild-type mice, IFN-ß(-/-) mice showed increased mortality to i.n. VSV infection, whereas upon i.v. infection no such differences were detected highlighting the biological significance of intracerebrally expressed IFN-ß. In conclusion, upon i.n. VSV instillation, IFN-ß responses mounted by astrocytes within the olfactory bulb critically contribute to the antiviral defense by stimulating distal IFN-ß-negative brain areas and thus arresting virus spread. IMPORTANCE: The central nervous system has long been considered an immune privileged site. More recently, it became evident that specialized immune mechanisms are active within the brain to control pathogens. Previously, we showed that virus, which entered the brain via the olfactory route, was arrested within the olfactory bulb by a type I IFN-dependent mechanism. Since peripheral type I IFN would not readily cross the blood-brain barrier and within the brain thus far no abundant type I IFN responses have been detected, here we addressed from where locally active IFN originated from. We found that upon intranasal VSV instillation, primarily astrocytes, and to a lesser extent neurons, were stimulated within the olfactory bulb to mount IFN-ß responses that also activated and protected distal brain areas. Our results are surprising because in other infection models astrocytes have not yet been identified as major type I IFN producers.


Assuntos
Astrócitos/imunologia , Encefalite Viral/imunologia , Interferon beta/metabolismo , Bulbo Olfatório/imunologia , Infecções por Rhabdoviridae/imunologia , Vesiculovirus/imunologia , Animais , Astrócitos/virologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Interferon beta/deficiência , Luciferases/análise , Luciferases/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/imunologia , Neurônios/virologia , Bulbo Olfatório/virologia , Análise de Sobrevida
10.
J Interferon Cytokine Res ; 34(5): 330-41, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24175628

RESUMO

To investigate the immunoregulatory effects of interferon (IFN)-ß on CD4+ T cells, we examined the response of CD4+ T cells from IFN-ß(+/+) and IFN-ß(-/-) mice to CD3/CD28 activation and to differentiation to Th17 lineage, analyzing the expression of signaling effectors, cell surface receptors, production of IL-17, and gene expression profiles. We provide evidence of increased phosphorylation of the membrane proximal kinase associated with TCR activation, ZAP-70, in IFN-ß(-/-) T cells compared with IFN-ß(+/+) T cells. Anti-CD3/anti-CD28 antibody stimulation of whole splenocytes or CD4+ T cells from IFN-ß(-/-) mice results in secretion of IL-17A, in contrast to identical stimulation of cells from IFN-ß(+/+) mice, which fails to increase IL-17A production. After CD3/CD28 activation, IFN-ß(-/-) CD4+ T cells express higher levels of IRF-4, required for Th17 differentiation, and increased expression of CCR6, IL-23R, IL-6R, and CXCR4, compared with activated IFN-ß(+/+) T cells. Notably, cell surface expression of IL-6R and IL-23R is significantly higher in the IFN-ß(-/-) CD4+ T cells, with an increased number of double-positive CCR6+IL-23R+ and IL-6R+IL-23R+ CD4+ T cells. On polarization to Th17 lineage, CD4+ T cells from IFN-ß(-/-) mice exhibit a more Th17-primed transcriptome compared with CD4+ T cells from IFN-ß(+/+) mice. Indeed, when CD4+ T cells from IFN-ß(+/+) mice are polarized to Th17 lineage in the presence of IFN-ß, many Th17-associated genes are down-regulated. Employing a MOG-peptide-induced experimental autoimmune encephalomyelitis model of multiple sclerosis, we identify a greater proportion of Th17 cells in the lymph nodes of IFN-ß(-/-) mice compared with IFN-ß(+/+) mice, and increased numbers of CD4+ T cells in the central nervous system of IFN-ß(-/-) mice, regardless of the stage of disease. Taken together, our data indicate an immunoregulatory role for IFN-ß in the suppression of Th17 cells.


Assuntos
Interferon beta/imunologia , Células Th17/imunologia , Animais , Interferon beta/deficiência , Camundongos , Fenótipo , Células Th17/citologia
11.
FEBS Lett ; 586(19): 3255-62, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22771905

RESUMO

IFN-ß is induced via a c-fos dependent mechanism that is present downstream of the receptor activator of NF-κB ligand (RANKL)-RANK signal transduction cascade during osteoclast differentiation. Increased production of IFN-ß in turn inhibits osteoclastogenesis. However, the mechanism by which IFN-ß exerts its suppressive function remains unclear. In the present study, we found that miR-155, an IFN-ß-induced miRNA, mediated the suppressive effect of IFN-ß on osteoclast differentiation by targeting SOCS1 and MITF, two essential regulators of osteoclastogenesis. These findings have not only demonstrated that miR-155 inhibits osteoclast differentiation, but also provided a new therapeutic target for treatment of osteoclast-mediated diseases.


Assuntos
Interferon beta/deficiência , Interferon beta/farmacologia , MicroRNAs/biossíntese , MicroRNAs/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Regiões 3' não Traduzidas , Animais , Sequência de Bases , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Regulação para Baixo , Células HEK293 , Humanos , Interferon beta/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição Associado à Microftalmia/genética , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Proteínas Recombinantes/farmacologia , Homologia de Sequência do Ácido Nucleico , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/genética
12.
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
13.
Gastroenterology ; 141(2): 674-85, 685.e1-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21683701

RESUMO

BACKGROUND & AIMS: The hepatitis C virus (HCV) serine protease NS3/4A can cleave mitochondria-associated antiviral signaling protein (MAVS) and block retinoic acid-inducible gene I-mediated interferon (IFN) responses. Although this mechanism is thought to have an important role in HCV-mediated innate immunosuppression, its significance in viral persistence is not clear. METHODS: We generated transgenic mice that express the HCV NS3/4A proteins specifically in the liver and challenged the animals with a recombinant vesicular stomatitis virus, a synthetic HCV genome, IFN alfa, or IFN beta. We evaluated the effects of HCV serine protease on the innate immune responses and their interactions. RESULTS: Expression of HCV NS3/4A resulted in cleavage of intrahepatic MAVS; challenge of transgenic mice with vesicular stomatitis virus or a synthetic HCV genome induced strong, type I IFN-mediated responses that were not significantly lower than those of control mice. Different challenge agents induced production of different ratios of IFN alfa and beta, resulting in different autophagic responses and vesicular trafficking patterns of endoplasmic reticulum- and mitochondria-associated viral proteins. IFN beta promoted degradation of the viral proteins by the autolysosome. Variant isoforms of MAVS were associated with distinct, type I IFN-mediated autophagic responses; these responses have a role in trafficking of viral components to endosomal compartments that contain Toll-like receptor-3. CONCLUSIONS: IFN beta mediates a distinct autophagic mechanism of antiviral host defense. MAVS has an important role in type I IFN-induced autophagic trafficking of viral proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Hepacivirus/imunologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Fígado/metabolismo , RNA Viral/imunologia , Vesiculovirus/imunologia , Proteínas não Estruturais Virais/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Autofagia/imunologia , Transporte Biológico , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Retículo Endoplasmático/imunologia , Expressão Gênica , Imunidade Inata , Fator Regulador 1 de Interferon/metabolismo , Interferon Tipo I/imunologia , Interferon-alfa/deficiência , Interferon-alfa/metabolismo , Interferon beta/deficiência , Interferon beta/metabolismo , Fígado/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Recombinantes , Transdução de Sinais
14.
J Immunol ; 186(1): 556-62, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21098223

RESUMO

Intratracheal administration of low molecular mass (LMM) hyaluronan (200 kDa) results in greater neutrophil infiltration in the lungs of TLR4(-/-) mice compared with that in wild-type mice. In general, enhanced neutrophil infiltration in tissue is due to cell influx; however, neutrophil apoptosis also plays an important role. We have assessed the effects of TLR4 in the regulation of neutrophil apoptosis in response to administration of LMM hyaluronan. We found that apoptosis of inflammatory neutrophils is impaired in TLR4(-/-) mice, an effect that depends upon the IFN-ß-mediated TRAIL/TRAILR system. IFN-ß levels were decreased in LMM hyaluronan-treated TLR4-deficient neutrophils. The treatment of inflammatory neutrophils with IFN-ß enhanced the levels of TRAIL and TRAILR 2. LMM hyaluronan-induced inflammatory neutrophil apoptosis was substantially prevented by anti-TRAIL neutralizing mAb. We conclude that decreased IFN-ß levels decrease the activity of the TRAIL/TRAILR system in TLR4-deficient neutrophils, leading to impaired apoptosis of neutrophils and resulting in abnormal accumulation of neutrophils in the lungs of LMM hyaluronan-treated mice. Thus, TLR4 plays a novel homeostatic role in noninfectious lung inflammation by accelerating the elimination of inflammatory neutrophils.


Assuntos
Apoptose/imunologia , Ácido Hialurônico/administração & dosagem , Ácido Hialurônico/fisiologia , Interferon beta/fisiologia , Neutrófilos/imunologia , Receptor 4 Toll-Like/fisiologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Homeostase/imunologia , Ácido Hialurônico/química , Mediadores da Inflamação/administração & dosagem , Mediadores da Inflamação/química , Mediadores da Inflamação/fisiologia , Interferon beta/deficiência , Interferon beta/genética , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peso Molecular , Ativação de Neutrófilo/efeitos dos fármacos , Ativação de Neutrófilo/imunologia , Infiltração de Neutrófilos/efeitos dos fármacos , Infiltração de Neutrófilos/imunologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética
15.
J Immunol ; 183(5): 3229-36, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19667093

RESUMO

Type I IFN is a major player in innate and adaptive immune responses. Besides, it is involved in organogenesis and tumor development. Generally, IFN responses are amplified by an autocrine loop with IFN-beta as the priming cytokine. However, due to the lack of sensitive detection systems, where and how type I IFN is produced in vivo is still poorly understood. In this study, we describe a luciferase reporter mouse, which allows tracking of IFN-beta gene induction in vivo. Using this reporter mouse, we reveal strong tissue-specific induction of IFN-beta following infection with influenza or La Crosse virus. Importantly, this reporter mouse also allowed us to visualize that IFN-beta is expressed constitutively in several tissues. As suggested before, low amounts of constitutively produced IFN might maintain immune cells in an activated state ready for a timely response to pathogens. Interestingly, thymic epithelial cells were the major source of IFN-beta under noninflammatory conditions. This relatively high constitutive expression was controlled by the NF Aire and might influence induction of tolerance or T cell development.


Assuntos
Genes Reporter , Mediadores da Inflamação/fisiologia , Interferon beta/biossíntese , Interferon beta/genética , Animais , Linhagem Celular Transformada , Células Cultivadas , Células Epiteliais/imunologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Feminino , Genes Reporter/imunologia , Mediadores da Inflamação/metabolismo , Vírus da Influenza A Subtipo H7N7/genética , Vírus da Influenza A Subtipo H7N7/imunologia , Interferon beta/deficiência , Vírus La Crosse/genética , Vírus La Crosse/imunologia , Luciferases de Vaga-Lume/genética , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Doença de Newcastle/genética , Doença de Newcastle/imunologia , Doença de Newcastle/patologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Timo/imunologia , Timo/patologia , Timo/virologia
16.
J Immunol ; 183(2): 1099-109, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19581626

RESUMO

Type I IFNs play a key role in linking the innate and adaptive arms of the immune system. Although produced rapidly in response to pathogens, IFNs are also produced at low levels in the absence of infection. In the present study, we demonstrate that constitutively produced IFNs are necessary in vivo to maintain dendritic cells in an "Ag presentation-competent" state. Conventional dendritic cells (cDCs) isolated from spleens of IFN-beta or IFNAR-deficient mice exhibit a highly impaired ability to present Ag and activate naive T cells. Microarray analysis of mRNA isolated from IFN-beta(-/-) and IFNAR(-/-) cDCs revealed diminished expression of two genes that encoded members of the heat shock protein 70 (Hsp70) family. Consistent with this observation, pharmacological inhibition of Hsp70 in cDCs from wild-type mice impaired their T cell stimulatory capacity. Similarly, the Ag presentation ability of splenic cDCs isolated from Hsp70.1/3(-/-) mice was also severely impaired in comparison to wild-type cDCs. Thus, constitutive IFN-beta expression regulates Hsp70 levels to help maintain dendritic cells in a competent state for efficient priming of effector T cells in vivo.


Assuntos
Apresentação de Antígeno , Células Dendríticas/imunologia , Regulação para Baixo , Interferon beta/fisiologia , Animais , Proteínas de Choque Térmico HSP70 , Fatores Imunológicos , Interferon beta/deficiência , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência , Baço/citologia , Linfócitos T/imunologia
17.
Virology ; 390(2): 338-47, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19539968

RESUMO

North American eastern equine encephalitis virus (NA-EEEV) strains cause high mortality in humans, whereas South American strains (SA-EEEV) are typically avirulent. To clarify mechanisms of SA-EEEV attenuation, we compared mouse-attenuated BeAr436087 SA-EEEV, considered an EEEV vaccine candidate, with mouse-virulent NA-EEEV strain, FL93-939. Although attenuated, BeAr436087 initially replicated more efficiently than FL93-939 in lymphoid and other tissues, inducing systemic IFN-alpha/beta release, whereas FL93-939 induced little. BeAr436087 was more virulent than FL93-939 in IFN-alpha/beta-deficient mice, confirming that type I IFN responses determined attenuation, but the viruses were similarly sensitive to IFN-alpha/beta priming in vitro. Infection with BeAr436087 protected against FL93-939 disease/death, even when given 8 h afterward, suggesting that the environment produced by BeAr436087 infection attenuated FL93-939. We conclude that avoidance of IFN-alpha/beta induction is a major virulence factor for FL93-939. Furthermore, BeAr436087 could be used for vaccination and therapeutic treatment in the event of exposure to NA-EEEV during a bioterrorism attack.


Assuntos
Vírus da Encefalite Equina do Leste/imunologia , Vírus da Encefalite Equina do Leste/patogenicidade , Interferon-alfa/imunologia , Interferon beta/imunologia , Animais , Humanos , Interferon-alfa/sangue , Interferon-alfa/deficiência , Interferon beta/sangue , Interferon beta/deficiência , Camundongos , Camundongos Knockout , Virulência
18.
J Virol ; 82(7): 3642-53, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18234805

RESUMO

The virion host shutoff (vhs) protein of herpes simplex virus (HSV) has endoribonuclease activity and rapidly reduces protein synthesis in infected cells through mRNA degradation. Herpes simplex virus 1 (HSV-1) and HSV-2 vhs mutants are highly attenuated in vivo, but replication and virulence are largely restored to HSV-2 vhs mutants in the absence of a type I interferon (IFN) response. The role of vhs in pathogenesis and the hindrance of the type I IFN response have classically been examined with viruses that completely lack vhs or express a truncated vhs protein. To determine whether RNase activity is the principal mechanism of vhs-mediated type I IFN resistance and virulence, we constructed a HSV-2 point mutant that synthesizes full-length vhs protein lacking RNase activity (RNase(-) virus). Wild-type and mutant HSV-2 vhs proteins coimmunoprecipitated with VP16 and VP22. vhs protein bearing the point mutation was packaged into the virion as efficiently as the wild-type vhs protein. Like a mutant encoding truncated vhs, the RNase(-) virus showed IFN-dependent replication that was restricted compared with that of the wild-type virus. The RNase(-) virus was highly attenuated in wild-type mice infected intravaginally, with reduced mucosal replication, disease severity, and spread to the nervous system comparable to those of the vhs truncation mutant. Surprisingly, in alpha/beta interferon (IFN-alpha/beta) receptor knockout mice, the vhs RNase mutant was more attenuated than the vhs truncation mutant in terms of disease severity and virus titer in vaginal swabs and central nervous system samples, suggesting that non-enzymatically active vhs protein interferes with efficient virus replication. Our results indicate that vhs enzymatic activity plays a complex role in vhs-mediated type I IFN resistance during HSV-2 infection.


Assuntos
Herpes Simples/virologia , Herpesvirus Humano 2/crescimento & desenvolvimento , Ribonucleases/genética , Ribonucleases/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Animais , Encéfalo/virologia , Feminino , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 2/imunologia , Imunoprecipitação , Interferon Tipo I/imunologia , Interferon-alfa/deficiência , Interferon beta/deficiência , Camundongos , Camundongos Knockout , Mutação Puntual , Ligação Proteica , Índice de Gravidade de Doença , Análise de Sobrevida , Vagina/virologia , Proteínas Estruturais Virais/metabolismo , Vírion/química , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Montagem de Vírus/fisiologia
19.
Eur J Immunol ; 38(2): 507-17, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18200500

RESUMO

Plasmacytoid dendritic cells (pDC) are specialized in massive production of type I interferons (IFN) upon viral infections. Activation of IFN regulatory factor (IRF)-7 is critically required for the synthesis of type I IFN in pDC. IRF-7 is highly expressed by resting pDC and translocates into the nucleus to initiate type I IFN transcription. In a previous work, we observed an impaired IFN-alpha production in enriched cord blood pDC following a TLR9 stimulation using CpG oligonucleotides. Herein, we show that highly purified pDC from cord blood exhibit a profound defect in their capacity to produce IFN-alpha/beta in response to TLR9 as well as to TLR7 ligation or human CMV or HSV-1 exposure. Microarray experiments indicate that expression of the majority of type I IFN subtypes induced by a TLR7 agonist is reduced in cord blood pDC. We next demonstrated a reduced nuclear translocation of IRF-7 in cord blood pDC following CpG and HSV stimulation as compared to adult pDC. We conclude that impaired IRF-7 translocation in cord blood pDC is associated with defective expression of type I IFN genes. Our data provide a molecular understanding for the decreased ability of cord blood pDC to produce type I IFN upon viral stimulation.


Assuntos
Células Dendríticas/imunologia , Sangue Fetal/citologia , Sangue Fetal/imunologia , Fator Regulador 7 de Interferon/deficiência , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Adulto , Células Cultivadas , Citomegalovirus/imunologia , Células Dendríticas/metabolismo , Sangue Fetal/virologia , Herpesvirus Humano 1/imunologia , Humanos , Recém-Nascido , Fator Regulador 7 de Interferon/agonistas , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/fisiologia , Interferon-alfa/deficiência , Interferon-alfa/metabolismo , Interferon beta/deficiência , Interferon beta/metabolismo , Ligantes
20.
J Gen Virol ; 88(Pt 10): 2627-2635, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17872512

RESUMO

In mammals the interferon (IFN) system is a central innate antiviral defence mechanism, while the involvement of RNA interference (RNAi) in antiviral response against RNA viruses is uncertain. Here, we tested whether RNAi is involved in the antiviral response in mammalian cells. To investigate the role of RNAi in influenza A virus-infected cells in the absence of IFN, we used Vero cells that lack IFN-alpha and IFN-beta genes. Our results demonstrate that knockdown of a key RNAi component, Dicer, led to a modest increase of virus production and accelerated apoptosis of influenza A virus-infected cells. These effects were much weaker in the presence of IFN. The results also show that in both Vero cells and the IFN-producing alveolar epithelial A549 cell line influenza A virus targets Dicer at mRNA and protein levels. Thus, RNAi is involved in antiviral response, and Dicer is important for protection against influenza A virus infection.


Assuntos
Vírus da Influenza A Subtipo H7N7/patogenicidade , Interferência de RNA/fisiologia , Ribonuclease III/metabolismo , Animais , Anticorpos Monoclonais , Linhagem Celular , Chlorocebus aethiops , Genes Reporter , Haplorrinos , Humanos , Interferon-alfa/deficiência , Interferon beta/deficiência , Luciferases/genética , Camundongos , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/isolamento & purificação , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonuclease III/imunologia , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA