Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Immunol ; 206(1): 154-163, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33219146

RESUMEN

Viral RNA in the cytoplasm of mammalian host cells is recognized by retinoic acid-inducible protein-I-like receptors (RLRs), which localize to cytoplasmic stress granules (SGs). Activated RLRs associate with the mitochondrial adaptor protein IPS-1, which activates antiviral host defense mechanisms, including type I IFN induction. It has remained unclear, however, how RLRs in SGs and IPS-1 in the mitochondrial outer membrane associate physically and engage in information transfer. In this study, we show that NUDT21, an RNA-binding protein that regulates alternative transcript polyadenylation, physically associates with IPS-1 and mediates its localization to SGs in response to transfection with polyinosinic-polycytidylic acid [poly(I:C)], a mimic of viral dsRNA. We found that despite its well-established function in the nucleus, a fraction of NUDT21 localizes to mitochondria in resting cells and becomes localized to SGs in response to poly(I:C) transfection. NUDT21 was also found to be required for efficient type I IFN induction in response to viral infection in both human HeLa cells and mouse macrophage cell line RAW264.7 cells. Our results together indicate that NUDT21 links RLRs in SGs to mitochondrial IPS-1 and thereby activates host defense responses to viral infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones por Cardiovirus/metabolismo , Factor de Especificidad de Desdoblamiento y Poliadenilación/metabolismo , Proteína 58 DEAD Box/metabolismo , Virus de la Encefalomiocarditis/fisiología , Mitocondrias/metabolismo , Enfermedad de Newcastle/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Receptores Inmunológicos/metabolismo , Vesículas Secretoras/metabolismo , Animales , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones , Poli I-C/inmunología , Transporte de Proteínas , Células RAW 264.7 , ARN Interferente Pequeño/genética , ARN Viral/inmunología , Estrés Fisiológico
2.
mBio ; 10(4)2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31409686

RESUMEN

Encephalomyocarditis virus (EMCV) is an animal pathogen and an important model organism, whose receptor requirements are poorly understood. Here, we employed a genome-wide haploid genetic screen to identify novel EMCV host factors. In addition to the previously described picornavirus receptors sialic acid and glycosaminoglycans, this screen unveiled important new host factors for EMCV. These factors include components of the fibroblast growth factor (FGF) signaling pathway, such as the potential receptors FGFR1 and ADAM9, a cell-surface metalloproteinase. By employing various knockout cells, we confirmed the importance of the identified host factors for EMCV infection. The largest reduction in infection efficiency was observed in cells lacking ADAM9. Pharmacological inhibition of the metalloproteinase activity of ADAM9 did not affect virus infection. Moreover, reconstitution of inactive ADAM9 in knockout cells restored susceptibility to EMCV, pointing to a proteinase-independent role of ADAM9 in mediating EMCV infection. Using neutralization assays with ADAM9-specific antiserum and soluble receptor proteins, we provided evidence for a role of ADAM9 in EMCV entry. Finally, binding assays showed that ADAM9 facilitates attachment of EMCV to the cell surface. Together, our findings reveal a role for ADAM9 as a novel receptor or cofactor for EMCV.IMPORTANCE EMCV is an animal pathogen that causes acute viral infections, usually myocarditis or encephalitis. It is thought to circulate mainly among rodents, from which it is occasionally transmitted to other animal species, including humans. EMCV causes fatal outbreaks of myocarditis and encephalitis in pig farms and zoos, making it an important veterinary pathogen. Although EMCV has been widely used as a model to study mechanisms of viral disease in mice, little is known about its entry mechanism. Here, we employ a haploid genetic screen for EMCV host factors and identify an essential role for ADAM9 in EMCV entry.


Asunto(s)
Proteínas ADAM/metabolismo , Infecciones por Cardiovirus/virología , Virus de la Encefalomiocarditis/fisiología , Proteínas de la Membrana/metabolismo , Internalización del Virus , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/genética , Animales , Infecciones por Cardiovirus/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Virus de la Encefalomiocarditis/metabolismo , Técnicas de Inactivación de Genes , Genoma Humano/genética , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Acoplamiento Viral , Replicación Viral
3.
Virology ; 516: 139-146, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29353210

RESUMEN

Cardioviruses cause diseases in many animals including, in rare cases, humans. Although they share common features with all picornaviruses, cardioviruses have unique properties that distinguish them from other family members, including enteroviruses. One feature shared by all picornaviruses is the covalent attachment of VPg to the 5' end of genomic RNA via a phosphotyrosyl linkage. For enteroviruses, this linkage is cleaved by a host cell protein, TDP2. Since TDP2 is divergently required during enterovirus infections, we determined if TDP2 is necessary during infection by the prototype cardiovirus, EMCV. We found that EMCV yields are reduced in the absence of TDP2. We observed a decrease in viral protein accumulation and viral RNA replication in the absence of TDP2. In contrast to enterovirus infections, we found that TDP2 is modified at peak times of EMCV infection. This finding suggests a unique mechanism for cardioviruses to regulate TDP2 activity during infection.


Asunto(s)
Infecciones por Cardiovirus/metabolismo , Cardiovirus/metabolismo , Proteínas Nucleares/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Factores de Transcripción/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Proteínas Virales/metabolismo , Animales , Cardiovirus/genética , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/virología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Proteínas de Unión al ADN , Fibroblastos/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Hidrolasas Diéster Fosfóricas/genética , Transporte de Proteínas , Proteolisis , ARN Viral/genética , ARN Viral/metabolismo , Factores de Transcripción/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Proteínas Virales/genética , Replicación Viral
4.
J Neurovirol ; 23(5): 696-703, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28741149

RESUMEN

Seizure disorders are often associated with infectious etiologies. Infection, via the intracerebral (i.c.) route, of C57BL/6J mice with the Daniels (DA) strain of Theiler's murine encephalomyelitis virus (TMEV) results in approximately 50% of the mice developing acute behavioral seizures. TMEV-DA is the wild-type strain of the virus that replicates within the parenchyma of the brain. A variant of TMEV-DA, TMEV-H101, does not replicate within the parenchyma of the brain. However, infection with TMEV-H101 via the i.c. route still results in approximately 40% of the mice developing acute behavioral seizures. Infiltrating macrophages producing interleukin-6 (IL-6) have been implicated in the induction of acute seizures following TMEV-DA infection. We examined macrophage infiltration and microglial activation within the brain and cytokine levels in the periphery in mice infected with TMEV-DA or TMEV-H101 and assessed the effects of the addition of recombinant IL-6 to the periphery in wild-type and IL-6 knockout mice infected with TMEV-DA. We found that pathologic levels of IL-6 in the periphery may play a role in the development of seizures when viral replication within the brain is limited. Examination of the role played by the peripheral immune system in the development of seizures/epilepsy in the TMEV-induced seizure model, the first viral infection driven model for epilepsy, could lead to the elucidation of novel therapeutics.


Asunto(s)
Infecciones por Cardiovirus/complicaciones , Infecciones por Cardiovirus/inmunología , Interleucina-6/inmunología , Convulsiones/metabolismo , Convulsiones/virología , Animales , Infecciones por Cardiovirus/metabolismo , Interleucina-6/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Microglía/metabolismo , Theilovirus/inmunología
5.
Immunobiology ; 222(2): 350-357, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27665995

RESUMEN

Viral infections can give rise to secondary bacterial infections. In the present study, we examined the role of invariant natural killer T (iNKT) cells in lipopolysaccharide (LPS)-induced lethal shock during encephalomyocarditis virus (EMCV) infection. Wild-type (WT) mice and Jα18 gene knockout (Jα18 KO) mice were inoculated with EMCV, 5days prior to challenging with LPS. The survival rate of Jα18 KO mice subjected to EMCV and LPS was significantly higher than that of WT mice. TNF-α and nitric oxide (NO) production were increased in WT mice, than that in Jα18 KO mice, after the administration of EMCV and LPS. EMCV infection increased the number of iNKT cells and IFN-γ production by iNKT cells in WT mice. Moreover, EMCV infection enhanced the expression of Toll-like receptor 4 (TLR4) in the lung and spleen. IFN-γ also increased the expression of TLR4 in splenocytes. These findings indicated that EMCV infection activated iNKT cells, and IFN-γ secreted from the iNKT cells up-regulated the expression of TLR4 in various tissues. As a result, EMCV-infected mice were susceptible to LPS and easily developed the lethal shock. In conclusion, iNKT cells were involved in the development of LPS-induced lethal shock during EMCV infection.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/metabolismo , Virus de la Encefalomiocarditis/inmunología , Lipopolisacáridos/efectos adversos , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Choque Séptico/etiología , Choque Séptico/metabolismo , Animales , Biomarcadores , Infecciones por Cardiovirus/mortalidad , Infecciones por Cardiovirus/virología , Coinfección , Citocinas/metabolismo , Modelos Animales de Enfermedad , Lipopolisacáridos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Óxido Nítrico/metabolismo , Choque Séptico/mortalidad , Factor de Necrosis Tumoral alfa/metabolismo
6.
Biochem Biophys Res Commun ; 480(2): 187-193, 2016 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-27743889

RESUMEN

LGP2 and MDA5 cooperate to detect viral RNA in the cytoplasm of Picornavirus-infected cells and activate innate immune responses. To further define regulatory components of RNA recognition by LGP2/MDA5, a yeast two-hybrid screen was used to identify LGP2-interacting proteins. The screening has identified the TAR-RNA binding protein (TRBP), which is known to be an essential factor for RNA interference (RNAi). Immuno-precipitation experiments demonstrated that TRBP interacted specifically with LGP2 but not with related RIG-I-like receptors, RIG-I or MDA5. siRNA knockdown experiments indicate that TRBP is important for Cardiovirus-triggered interferon responses, but TRBP is not involved in Sendai virus-triggered interferon response that is mediated mainly by RIG-I. To support functional interaction with LGP2, overexpressed TRBP increased Cardiovirus-triggered interferon promoter activity only when LGP2 and MDA5 are co-expressed but not MDA5 alone. Together, our findings illustrate a possible connection between an RNAi-regulatory factor and antiviral RNA recognition that is specifically required for a branch of the virus induced innate immune response.


Asunto(s)
Infecciones por Cardiovirus/metabolismo , Interacciones Huésped-Patógeno , Proteínas de Unión al ARN/metabolismo , Animales , Cardiovirus/patogenicidad , Infecciones por Cardiovirus/inmunología , Chlorocebus aethiops , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Células HEK293 , Humanos , Helicasa Inducida por Interferón IFIH1/genética , Interferón beta/genética , Ratones , Regiones Promotoras Genéticas , ARN Helicasas/genética , ARN Helicasas/metabolismo , ARN Interferente Pequeño , Proteínas de Unión al ARN/genética , Receptores Inmunológicos , Virus Sendai/patogenicidad , Células Vero
7.
Sci Rep ; 6: 28699, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-27345730

RESUMEN

Neurologic complications associated with viral encephalitis, including seizures and cognitive impairment, are a global health issue, especially in children. We previously showed that hippocampal injury during acute picornavirus infection in mice is associated with calpain activation and is the result of neuronal death triggered by brain-infiltrating inflammatory monocytes. We therefore hypothesized that treatment with a calpain inhibitor would protect neurons from immune-mediated bystander injury. C57BL/6J mice infected with the Daniel's strain of Theiler's murine encephalomyelitis virus were treated with the FDA-approved drug ritonavir using a dosing regimen that resulted in plasma concentrations within the therapeutic range for calpain inhibition. Ritonavir treatment significantly reduced calpain activity in the hippocampus, protected hippocampal neurons from death, preserved cognitive performance, and suppressed seizure escalation, even when therapy was initiated 36 hours after disease onset. Calpain inhibition by ritonavir may be a powerful tool for preserving neurons and cognitive function and preventing neural circuit dysregulation in humans with neuroinflammatory disorders.


Asunto(s)
Calpaína/antagonistas & inhibidores , Infecciones por Cardiovirus/tratamiento farmacológico , Inhibidores de Cisteína Proteinasa/farmacología , Fármacos Neuroprotectores/farmacología , Ritonavir/farmacología , Theilovirus/metabolismo , Enfermedad Aguda , Animales , Calpaína/metabolismo , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/patología , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/virología , Ratones
8.
Gene ; 587(1): 33-41, 2016 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-27085482

RESUMEN

4.5SH and 4.5SI RNA are two abundant small non-coding RNAs specific for several related rodent families including Muridae. These RNAs have a number of common characteristics such as the short length (about 100nt), transcription by RNA polymerase III, and origin from Short Interspersed Elements (SINEs). However, their stabilities in cells substantially differ: the half-life of 4.5SH RNA is about 20min, while that of 4.5SI RNA is 22h. Here we studied the influence of cell stress such as heat shock or viral infection on these two RNAs. We found that the level of 4.5SI RNA did not change in stressed cells; whereas heat shock increased the abundance of 4.5SH RNA 3.2-10.5 times in different cell lines; and viral infection, 5 times. Due to the significant difference in the turnover rates of these two RNAs, a similar activation of their transcription by heat shock increases the level of the short-lived 4.5SH RNA and has minor effect on the level of the long-lived 4.5SI RNA. In addition, the accumulation of 4.5SH RNA results not only from the induction of its transcription but also from a substantial retardation of its decay. To our knowledge, it is the first example of a short-lived non-coding RNA whose elongated lifetime contributes significantly to its accumulation in stressed cells.


Asunto(s)
Estabilidad del ARN , ARN Interferente Pequeño/metabolismo , Estrés Fisiológico , Animales , Infecciones por Cardiovirus/metabolismo , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Virus de la Encefalomiocarditis/fisiología , Ratones , ARN Interferente Pequeño/genética , Ratas , Transcripción Genética
9.
PLoS Pathog ; 11(9): e1005185, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26406250

RESUMEN

Cardioviruses, including encephalomyocarditis virus (EMCV) and the human Saffold virus, are small non-enveloped viruses belonging to the Picornaviridae, a large family of positive-sense RNA [(+)RNA] viruses. All (+)RNA viruses remodel intracellular membranes into unique structures for viral genome replication. Accumulating evidence suggests that picornaviruses from different genera use different strategies to generate viral replication organelles (ROs). For instance, enteroviruses (e.g. poliovirus, coxsackievirus, rhinovirus) rely on the Golgi-localized phosphatidylinositol 4-kinase III beta (PI4KB), while cardioviruses replicate independently of the kinase. By which mechanisms cardioviruses develop their ROs is currently unknown. Here we show that cardioviruses manipulate another PI4K, namely the ER-localized phosphatidylinositol 4-kinase III alpha (PI4KA), to generate PI4P-enriched ROs. By siRNA-mediated knockdown and pharmacological inhibition, we demonstrate that PI4KA is an essential host factor for EMCV genome replication. We reveal that the EMCV nonstructural protein 3A interacts with and is responsible for PI4KA recruitment to viral ROs. The ensuing phosphatidylinositol 4-phosphate (PI4P) proved important for the recruitment of oxysterol-binding protein (OSBP), which delivers cholesterol to EMCV ROs in a PI4P-dependent manner. PI4P lipids and cholesterol are shown to be required for the global organization of the ROs and for viral genome replication. Consistently, inhibition of OSBP expression or function efficiently blocked EMCV RNA replication. In conclusion, we describe for the first time a cellular pathway involved in the biogenesis of cardiovirus ROs. Remarkably, the same pathway was reported to promote formation of the replication sites of hepatitis C virus, a member of the Flaviviridae family, but not other picornaviruses or flaviviruses. Thus, our results highlight the convergent recruitment by distantly related (+)RNA viruses of a host lipid-modifying pathway underlying formation of viral replication sites.


Asunto(s)
Infecciones por Cardiovirus/metabolismo , Virus de la Encefalomiocarditis/fisiología , Interacciones Huésped-Parásitos/fisiología , Metabolismo de los Lípidos/fisiología , Replicación Viral/fisiología , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Animales , Western Blotting , Hepacivirus/fisiología , Humanos , Inmunoprecipitación , Microscopía Fluorescente , Fosfatos de Fosfatidilinositol/metabolismo , Picornaviridae , Virus ARN , ARN Interferente Pequeño , Receptores de Esteroides/metabolismo , Transfección
10.
Neurosci Lett ; 593: 124-8, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25796181

RESUMEN

Changes in the expression of xCT, the specific subunit of system xc(-) or the cystine/glutamate antiporter, have been associated with several neurological disorders and system xc(-) was recently proposed as a potential target for the development of new treatment strategies for multiple sclerosis (MS). In this study we used Theiler's murine encephalomyelitis virus (TMEV) infection, both in vitro and in vivo, as a model to further evaluate the involvement of system xc(-) in MS. Protein levels of xCT, as well as activity of system xc(-) were unaffected in RAW264.7 macrophages after infection with the demyelinating DA strain of TMEV. Also, protein expression of xCT remained stable in spinal cord and brain of FVB mice 1-2 and 6 weeks after intracranial injection of the DA strain of TMEV. These results demonstrate that TMEV infection of macrophages or FVB mice has no effect on system xc(-) and as such cannot be used as a model to study the involvement of system xc(-) in MS.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Infecciones por Cardiovirus/metabolismo , Macrófagos/metabolismo , Theilovirus/fisiología , Animales , Encéfalo/metabolismo , Infecciones por Cardiovirus/virología , Cisteína/metabolismo , Femenino , Macrófagos/virología , Ratones , Médula Espinal/metabolismo
11.
Brain Pathol ; 25(6): 712-23, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25495532

RESUMEN

Microglia and macrophages play a central role for demyelination in Theiler's murine encephalomyelitis (TME) virus infection, a commonly used infectious model for chronic-progressive multiple sclerosis. In order to determine the dynamic changes of microglia/macrophage polarization in TME, the spinal cord of Swiss Jim Lambert (SJL) mice was investigated by gene expression profiling and immunofluorescence. Virus persistence and demyelinating leukomyelitis were confirmed by immunohistochemistry and histology. Electron microscopy revealed continuous myelin loss together with abortive myelin repair during the late chronic infection phase indicative of incomplete remyelination. A total of 59 genes out of 151 M1- and M2-related genes were differentially expressed in TME virus-infected mice over the study period. The onset of virus-induced demyelination was associated with a dominating M1 polarization, while mounting M2 polarization of macrophages/microglia together with sustained prominent M1-related gene expression was present during the chronic-progressive phase. Molecular results were confirmed by immunofluorescence, showing an increased spinal cord accumulation of CD16/32(+) M1-, arginase-1(+) M2- and Ym1(+) M2-type cells associated with progressive demyelination. The present study provides a comprehensive database of M1-/M2-related gene expression involved in the initiation and progression of demyelination supporting the hypothesis that perpetuating interaction between virus and macrophages/microglia induces a vicious circle with persistent inflammation and impaired myelin repair in TME.


Asunto(s)
Infecciones por Cardiovirus/metabolismo , Encefalomielitis/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Theilovirus , Animales , Infecciones por Cardiovirus/patología , Encefalomielitis/patología , Femenino , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Macrófagos/ultraestructura , Macrófagos/virología , Ratones , Análisis por Micromatrices , Microglía/ultraestructura , Microglía/virología , Microscopía Electrónica , Vaina de Mielina/patología , Vaina de Mielina/ultraestructura , Neuroinmunomodulación/fisiología , Médula Espinal/metabolismo , Médula Espinal/ultraestructura , Médula Espinal/virología
12.
J Neurovirol ; 20(5): 485-95, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25052192

RESUMEN

Theiler's murine encephalomyelitis virus (TMEV) induces demyelination in susceptible strains of mice (SJL/J) through an immunopathological process that is mediated by CD4(+) Th1 T cell. These T cells are chemoattracted to the central nervous system by chemokines. Hence, in this study, we focused on the production of the chemokine "interferon-gamma-inducible protein 10 kDa," or IP-10/CXCL10, by cultured SJL/J mouse astrocytes infected with the BeAn strain of TMEV and its capacity to attract activated T cells. The analysis of the whole murine genome by DNA hybridization with cRNAs from mock- and TMEV-infected cultures revealed the upregulation of six sequences that potentially encode for CXCL10. This increased CXCL10 expression was validated by PCR and qPCR. The presence of this chemokine was further demonstrated by enzyme-linked immunoassay (ELISA). Significantly, astrocytes from BALB/c mice, a strain resistant to demyelination, did not produce CXCL10. The secreted CXCL10 was biologically active, inducing chemoattraction of activated lymphocytes. The inflammatory cytokines, IL-1α, IFN-γ, and TNF-α, were strong inducers of CXCL10 in astrocytes. Serum from TMEV-infected SJL/J but not BALB/c mice contains CXCL10, the levels of which peak at the onset of the clinical disease. Finally, this in vitro inflammation model was fully inhibited by 17ß-estradiol and four selective estrogen receptor modulators, as demonstrated by ELISA and qPCR.


Asunto(s)
Astrocitos/virología , Infecciones por Cardiovirus/inmunología , Quimiocina CXCL10/biosíntesis , Linfocitos T/inmunología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Western Blotting , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/metabolismo , Células Cultivadas , Quimiocina CXCL10/genética , Ensayo de Inmunoadsorción Enzimática , Hibridación in Situ , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Theilovirus/inmunología , Regulación hacia Arriba
13.
J Virol ; 88(18): 10748-57, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25008915

RESUMEN

UNLABELLED: Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are essential intracellular detectors of viral RNA. They contribute to the type I interferon (IFN) response that is crucial for host defense against viral infections. Given the potent antiviral and proinflammatory activities elicited by the type I IFNs, induction of the type I IFN response is tightly regulated. Members of the tripartite motif (TRIM) family of proteins have recently emerged as key regulators of antiviral immunity. We show that TRIM13, an E3 ubiquitin ligase, is expressed in immune cells and is upregulated in bone marrow-derived macrophages upon stimulation with inducers of type I IFN. TRIM13 interacts with MDA5 and negatively regulates MDA5-mediated type I IFN production in vitro, acting upstream of IFN regulatory factor 3. We generated Trim13(-/-) mice and show that upon lethal challenge with encephalomyocarditis virus (EMCV), which is sensed by MDA5, Trim13(-/-) mice produce increased amounts of type I IFNs and survive longer than wild-type mice. Trim13(-/-) murine embryonic fibroblasts (MEFs) challenged with EMCV or poly(I · C) also show a significant increase in beta IFN (IFN-ß) levels, but, in contrast, IFN-ß responses to the RIG-I-detected Sendai virus were diminished, suggesting that TRIM13 may play a role in positively regulating RIG-I function. Together, these results demonstrate that TRIM13 regulates the type I IFN response through inhibition of MDA5 activity and that it functions nonredundantly to modulate MDA5 during EMCV infection. IMPORTANCE: The type I interferon (IFN) response is crucial for host defense against viral infections, and proper regulation of this pathway contributes to maintaining immune homeostasis. Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are intracellular detectors of viral RNA that induce the type I IFN response. In this study, we show that expression of the gene tripartite motif 13 (Trim13) is upregulated in response to inducers of type I IFN and that TRIM13 interacts with both MDA5 and RIG-I in vitro. Through the use of multiple in vitro and in vivo model systems, we show that TRIM13 is a negative regulator of MDA5-mediated type I IFN production and may also impact RIG-I-mediated type I IFN production by enhancing RIG-I activity. This places TRIM13 at a key junction within the viral response pathway and identifies it as one of the few known modulators of MDA5 activity.


Asunto(s)
Infecciones por Cardiovirus/enzimología , ARN Helicasas DEAD-box/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Virus de la Encefalomiocarditis/fisiología , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/virología , ARN Helicasas DEAD-box/genética , Proteínas de Unión al ADN/genética , Femenino , Fibroblastos/enzimología , Fibroblastos/metabolismo , Fibroblastos/virología , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Interferón-alfa/genética , Interferón beta/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/genética
14.
Microbiol Immunol ; 58(7): 361-74, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24866463

RESUMEN

Deletion of cellular isoform of prion protein (PrP(C)) increases neuronal predisposition to damage by modulating apoptosis and the negative consequences of oxidative stress. In vivo studies have demonstrated that PrP(C)-deficient mice are more prone to seizure, depression, and induction of epilepsy and experience extensive cerebral damage following ischemic challenge or viral infection. In addition, adenovirus-mediated overexpression of PrP(C) reduces brain damage in rat models of cerebral ischemia. In experimental autoimmune encephalomyelitis, PrP(C)-deficient mice reportedly have a more aggressive disease onset and less clinical improvement during the chronic phase than wild-type mice mice. In mice given oral dextran sulfate, PrP(C) has a potential protective role against inflammatory bowel disease. PrP(C)-deficient mice demonstrate significantly greater increases in blood glucose concentrations after intraperitoneal injection of glucose than wild-type mice. Further in vivo challenges to PrP gene-deficient models and conditional knockout models with siRNA and in vivo administration of PrP-ligating agents may assist in refining knowledge of the lymphoid function of PrP(C) and predicting the effects of anti-PrP treatment on the immune system. Together, these findings indicate that PrP(C) may have multiple neuroprotective and anti-inflammatory roles, which explains why this protein is so widely expressed.


Asunto(s)
Susceptibilidad a Enfermedades , Priones/genética , Estrés Fisiológico , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Isquemia Encefálica/metabolismo , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/metabolismo , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Depresión/genética , Depresión/inmunología , Depresión/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Epilepsia/genética , Epilepsia/inmunología , Epilepsia/metabolismo , Técnicas de Inactivación de Genes , Humanos , Inmunidad/genética , Ratones , Ratones Noqueados , Priones/metabolismo , Interferencia de ARN
15.
Virus Res ; 177(2): 222-5, 2013 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-24036175

RESUMEN

The Theiler's murine encephalomyelitis virus (TMEV) leader (L) protein zinc-finger domain was mutated to study its role in cell death in infection of the murine macrophage cell line M1-D, revealing that an intact zinc-finger domain is required for full apoptotic activity. A functional L zinc-finger domain was also required for activation of p38 MAPK that results in phosphorylation and activation of p53, and in turn, alteration of the conformation of the anti-apoptotic proteins Puma and Mcl-1, leading to the release of pro-apoptotic Bax and apoptosis through the intrinsic pathway. TMEV infection also inhibits host protein synthesis, a stress shown by others to induce apoptosis. Since inhibition of host protein synthesis follows rather than precedes activation of MKK3/6 and p38, it seems less likely that it triggers apoptosis in infected cells. Finally, we showed that the levels of reactive oxygen species following infection were consistent with apoptotic rather than necrotic cell death. Thus, these experiments support an important role for the TMEV L protein zinc-finger domain in apoptosis in an infected murine macrophage line.


Asunto(s)
Apoptosis , Infecciones por Cardiovirus/veterinaria , Macrófagos/citología , Enfermedades de los Roedores/fisiopatología , Enfermedades de los Roedores/virología , Theilovirus/genética , Proteínas Virales/química , Proteínas Virales/genética , Animales , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/virología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Mutación , Estructura Terciaria de Proteína , Enfermedades de los Roedores/genética , Enfermedades de los Roedores/metabolismo , Theilovirus/química , Theilovirus/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Virales/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
17.
Circ Heart Fail ; 6(2): 326-34, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23271791

RESUMEN

BACKGROUND: Viral myocarditis is among the most common causes of heart failure in children and young adults. The RNA helicase melanoma differentiation-associated gene-5 (MDA5) is critical for host antiviral responses against members of the Picornaviridae family, such as encephalomyocarditis virus (EMCV). MDA5-knockout mice are highly susceptible to EMCV infection and develop significant myocardial injury and left ventricular dysfunction. However, the mechanisms by which MDA5 signaling within cardiac myocytes contributes to the host response against viral infection have not been defined. METHODS AND RESULTS: We generated cardiac-specific MDA5 transgenic (alpha-myosin heavy chain [αMHC]-MDA5) mice. These mice showed increased baseline cardiac expression of antiviral cytokines and increased cellular infiltration but no alterations in cardiac function and structure. αMHC-MDA5 mice were less susceptible to EMCV infection and had a significantly lower cardiac viral load compared with littermate control mice. The severity of myocarditis, prevalence of cardiac myocyte apoptosis, and cleavage of caspase 3 after EMCV infection were attenuated in αMHC-MDA5 mice. Furthermore, αMHC-MDA5 mice were protected against EMCV-induced myocardial dysfunction. CONCLUSIONS: Our data suggest that myocardial MDA5 may be a key molecule in protecting the heart from direct viral injury and myocardial dysfunction.


Asunto(s)
Infecciones por Cardiovirus/metabolismo , ARN Helicasas DEAD-box/metabolismo , Virus Maus Elberfeld/patogenicidad , Miocarditis/prevención & control , Miocardio/metabolismo , Animales , Apoptosis , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/virología , Caspasa 3/metabolismo , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Modelos Animales de Enfermedad , Femenino , Genotipo , Helicasa Inducida por Interferón IFIH1 , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocarditis/genética , Miocarditis/metabolismo , Miocarditis/patología , Miocarditis/fisiopatología , Miocarditis/virología , Miocardio/patología , Fenotipo , Volumen Sistólico , Factores de Tiempo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Izquierda/prevención & control , Disfunción Ventricular Izquierda/virología , Función Ventricular Izquierda , Replicación Viral
18.
J Biol Chem ; 287(52): 43651-64, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23109342

RESUMEN

Autophagy is a tightly regulated mechanism that mediates sequestration, degradation, and recycling of cellular proteins, organelles, and pathogens. Several proteins associated with autophagy regulate host responses to viral infections. Ribonuclease L (RNase L) is activated during viral infections and cleaves cellular and viral single-stranded RNAs, including rRNAs in ribosomes. Here we demonstrate that direct activation of RNase L coordinates the activation of c-Jun N-terminal kinase (JNK) and double-stranded RNA-dependent protein kinase (PKR) to induce autophagy with hallmarks as accumulation of autophagic vacuoles, p62(SQSTM1) degradation and conversion of Microtubule-associated Protein Light Chain 3-I (LC3-I) to LC3-II. Accordingly, treatment of cells with pharmacological inhibitors of JNK or PKR and mouse embryonic fibroblasts (MEFs) lacking JNK1/2 or PKR showed reduced autophagy levels. Furthermore, RNase L-induced JNK activity promoted Bcl-2 phosphorylation, disrupted the Beclin1-Bcl-2 complex and stimulated autophagy. Viral infection with Encephalomyocarditis virus (EMCV) or Sendai virus led to higher levels of autophagy in wild-type (WT) MEFs compared with RNase L knock out (KO) MEFs. Inhibition of RNase L-induced autophagy using Bafilomycin A1 or 3-methyladenine suppressed viral growth in initial stages; in later stages autophagy promoted viral replication dampening the antiviral effect. Induction of autophagy by activated RNase L is independent of the paracrine effects of interferon (IFN). Our findings suggest a novel role of RNase L in inducing autophagy affecting the outcomes of viral pathogenesis.


Asunto(s)
Autofagia , Endorribonucleasas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , eIF-2 Quinasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/patología , Línea Celular Tumoral , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Virus de la Encefalomiocarditis/fisiología , Endorribonucleasas/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/metabolismo , Infecciones por Respirovirus/patología , Virus Sendai/fisiología , Proteína Sequestosoma-1 , Replicación Viral/fisiología , eIF-2 Quinasa/genética
19.
PLoS Pathog ; 8(8): e1002857, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22916014

RESUMEN

Nod-like receptors (NLRs) comprise a large family of intracellular pattern- recognition receptors. Members of the NLR family assemble into large multiprotein complexes, termed the inflammasomes. The NLR family, pyrin domain-containing 3 (NLRP3) is triggered by a diverse set of molecules and signals, and forms the NLRP3 inflammasome. Recent studies have indicated that both DNA and RNA viruses stimulate the NLRP3 inflammasome, leading to the secretion of interleukin 1 beta (IL-1ß) and IL-18 following the activation of caspase-1. We previously demonstrated that the proton-selective ion channel M2 protein of influenza virus activates the NLRP3 inflammasome. However, the precise mechanism by which NLRP3 recognizes viral infections remains to be defined. Here, we demonstrate that encephalomyocarditis virus (EMCV), a positive strand RNA virus of the family Picornaviridae, activates the NLRP3 inflammasome in mouse dendritic cells and macrophages. Although transfection with RNA from EMCV virions or EMCV-infected cells induced robust expression of type I interferons in macrophages, it failed to stimulate secretion of IL-1ß. Instead, the EMCV viroporin 2B was sufficient to cause inflammasome activation in lipopolysaccharide-primed macrophages. While cells untransfected or transfected with the gene encoding the EMCV non-structural protein 2A or 2C expressed NLRP3 uniformly throughout the cytoplasm, NLRP3 was redistributed to the perinuclear space in cells transfected with the gene encoding the EMCV 2B or influenza virus M2 protein. 2B proteins of other picornaviruses, poliovirus and enterovirus 71, also caused the NLRP3 redistribution. Elevation of the intracellular Ca(2+) level, but not mitochondrial reactive oxygen species and lysosomal cathepsin B, was important in EMCV-induced NLRP3 inflammasome activation. Chelation of extracellular Ca(2+) did not reduce virus-induced IL-1ß secretion. These results indicate that EMCV activates the NLRP3 inflammasome by stimulating Ca(2+) flux from intracellular storages to the cytosol, and highlight the importance of viroporins, transmembrane pore-forming viral proteins, in virus-induced NLRP3 inflammasome activation.


Asunto(s)
Calcio/metabolismo , Infecciones por Cardiovirus/metabolismo , Proteínas Portadoras/metabolismo , Virus de la Encefalomiocarditis/metabolismo , Inflamasomas/metabolismo , Proteínas Virales/metabolismo , Animales , Infecciones por Cardiovirus/genética , Proteínas Portadoras/genética , Virus de la Encefalomiocarditis/genética , Células HEK293 , Células HeLa , Humanos , Inflamasomas/genética , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Virales/genética , Virión/genética , Virión/metabolismo
20.
Int J Mol Sci ; 13(6): 7365-7374, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22837699

RESUMEN

The single-stranded RNA encephalomyocarditis virus (EMCV) can replicate in the central nervous system (CNS) and lead to prominent brain lesions in the stratum pyramidale hippocampus and the stratum granulosum cerebelli. Activated microglia cells infected by EMCV produce a massive burst of reactive oxygen species (ROS) via NADPH oxidase 2 (NOX2) activation, leading to neuronal death. Balancing this effect is mechanisms by which ROS are eliminated from the CNS. Cellular prion protein (PrP(C)) plays an important antioxidant role and contributes to cellular defense against EMCV infection. This review introduces recent knowledge on brain injury induced by EMCV infection via ROS generation as well as the involvement of various mediators and regulators in the pathogenesis.


Asunto(s)
Infecciones por Cardiovirus/metabolismo , Virus de la Encefalomiocarditis/metabolismo , Hipocampo/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Infecciones por Cardiovirus/patología , Hipocampo/patología , Hipocampo/virología , Humanos , Microglía/patología , Microglía/virología , NADPH Oxidasa 2 , Proteínas PrPC/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA