Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Biol Chem ; 294(37): 13755-13768, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31346033

RESUMEN

Protection of neuronal homeostasis is a major goal in the management of neurodegenerative diseases. Microtubule-associated Ser/Thr kinase 2 (MAST2) inhibits neurite outgrowth, and its inhibition therefore represents a potential therapeutic strategy. We previously reported that a viral protein (G-protein from rabies virus) capable of interfering with protein-protein interactions between the PDZ domain of MAST2 and the C-terminal moieties of its cellular partners counteracts MAST2-mediated suppression of neurite outgrowth. Here, we designed peptides derived from the native viral protein to increase the affinity of these peptides for the MAST2-PDZ domain. Our strategy involved modifying the length and flexibility of the noninteracting sequence linking the two subsites anchoring the peptide to the PDZ domain. Three peptides, Neurovita1 (NV1), NV2, and NV3, were selected, and we found that they all had increased affinities for the MAST2-PDZ domain, with Kd values decreasing from 1300 to 60 nm, while target selectivity was maintained. A parallel biological assay evaluating neurite extension and branching in cell cultures revealed that the NV peptides gradually improved neural activity, with the efficacies of these peptides for stimulating neurite outgrowth mirroring their affinities for MAST2-PDZ. We also show that NVs can be delivered into the cytoplasm of neurons as a gene or peptide. In summary, our findings indicate that virus-derived peptides targeted to MAST2-PDZ stimulate neurite outgrowth in several neuron types, opening up promising avenues for potentially using NVs in the management of neurodegenerative diseases.


Asunto(s)
Neuritas/metabolismo , Proyección Neuronal/efectos de los fármacos , Dominios PDZ/fisiología , Estimulantes del Sistema Nervioso Central/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Microtúbulos/metabolismo , Neuronas/metabolismo , Péptidos/metabolismo , Péptidos/farmacología , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Virus de la Rabia , Relación Estructura-Actividad , Proteínas Virales/metabolismo , Proteínas Virales/farmacología
2.
Vaccine ; 37(33): 4673-4680, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-29523449

RESUMEN

Rabies causes more than 60,000 human deaths annually in areas where the virus is endemic. Importantly, rabies is one of the few pathogens for which there is no treatment following the onset of clinical disease with the outcome of infection being death in almost 100% of cases. Whilst vaccination, and the combination of vaccine and rabies immunoglobulin treatment for post-exposure administration are available, no tools have been identified that can reduce or prevent rabies virus replication once clinical disease has initiated. The search for effective antiviral molecules to treat those that have already developed clinical disease associated with rabies virus infection is considered one of the most important goals in rabies research. The current study assesses a single chain antibody molecule (ScFv) based on a monoclonal antibody that potently neutralises rabies in vitro as a potential therapeutic candidate. The recombinant ScFv was generated in Nicotiana benthamiana by transient expression, and was chemically conjugated (ScFv/RVG) to a 29 amino acid peptide, specific for nicotinic acetylcholine receptor (nAchR) binding in the CNS. This conjugated molecule was able to bind nAchR in vitro and enter neuronal cells more efficiently than ScFv. The ability of the ScFv/RVG to neutralise virus in vivo was assessed using a staggered administration where the molecule was inoculated either four hours before, two days after or four days after infection. The ScFv/RVG conjugate was evaluated in direct comparison with HRIG and a potential antiviral molecule, Favipiravir (also known as T-705) to indicate whether there was greater bioavailability of the ScFv in the brains of treated mice. The study indicated that the approach taken with the ScFv/RVG conjugate may have utility in the design and implementation of novel tools targetting rabies virus infection in the brain.


Asunto(s)
Vacunas Antirrábicas/uso terapéutico , Virus de la Rabia/inmunología , Rabia/metabolismo , Anticuerpos de Cadena Única/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Barrera Hematoencefálica/metabolismo , Western Blotting , Línea Celular , Electroforesis en Gel de Poliacrilamida , Humanos , Ratones , Rabia/inmunología , Rabia/prevención & control , Vacunas Antirrábicas/inmunología , Virus de la Rabia/patogenicidad , Anticuerpos de Cadena Única/inmunología
3.
Plant Biotechnol J ; 15(10): 1331-1339, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28273388

RESUMEN

The biomedical applications of antibody engineering are developing rapidly and have been expanded to plant expression platforms. In this study, we have generated a novel antibody molecule in planta for targeted delivery across the blood-brain barrier (BBB). Rabies virus (RABV) is a neurotropic virus for which there is no effective treatment after entry into the central nervous system. This study investigated the use of a RABV glycoprotein peptide sequence to assist delivery of a rabies neutralizing single-chain antibody (ScFv) across an in cellulo model of human BBB. The 29 amino acid rabies virus peptide (RVG) recognizes the nicotinic acetylcholine receptor (nAchR) at neuromuscular junctions and the BBB. ScFv and ScFv-RVG fusion proteins were produced in Nicotiana benthamiana by transient expression. Both molecules were successfully expressed and purified, but the ScFv expression level was significantly higher than that of ScFv-RVG fusion. Both ScFv and ScFv-RVG fusion molecules had potent neutralization activity against RABVin cellulo. The ScFv-RVG fusion demonstrated increased binding to nAchR and entry into neuronal cells, compared to ScFv alone. Additionally, a human brain endothelial cell line BBB model was used to demonstrate that plant-produced ScFv-RVGP fusion could translocate across the cells. This study indicates that the plant-produced ScFv-RVGP fusion protein was able to cross the in celluloBBB and neutralize RABV.


Asunto(s)
Barrera Hematoencefálica , Glicoproteínas/inmunología , Fragmentos de Péptidos/inmunología , Planticuerpos/farmacología , Virus de la Rabia/inmunología , Proteínas Virales/inmunología , Anticuerpos Neutralizantes/biosíntesis , Línea Celular , Humanos , Planticuerpos/aislamiento & purificación , Planticuerpos/metabolismo , Plantas Modificadas Genéticamente , Receptores Nicotínicos/metabolismo , Proteínas Recombinantes de Fusión , Nicotiana
4.
J Biol Chem ; 291(32): 16699-708, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27246854

RESUMEN

The human protein tyrosine phosphatase non-receptor type 4 (PTPN4) prevents cell death induction in neuroblastoma and glioblastoma cell lines in a PDZ·PDZ binding motifs-dependent manner, but the cellular partners of PTPN4 involved in cell protection are unknown. Here, we described the mitogen-activated protein kinase p38γ as a cellular partner of PTPN4. The main contribution to the p38γ·PTPN4 complex formation is the tight interaction between the C terminus of p38γ and the PDZ domain of PTPN4. We solved the crystal structure of the PDZ domain of PTPN4 bound to the p38γ C terminus. We identified the molecular basis of recognition of the C-terminal sequence of p38γ that displays the highest affinity among all endogenous partners of PTPN4. We showed that the p38γ C terminus is also an efficient inducer of cell death after its intracellular delivery. In addition to recruiting the kinase, the binding of the C-terminal sequence of p38γ to PTPN4 abolishes the catalytic autoinhibition of PTPN4 and thus activates the phosphatase, which can efficiently dephosphorylate the activation loop of p38γ. We presume that the p38γ·PTPN4 interaction promotes cellular signaling, preventing cell death induction.


Asunto(s)
Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Complejos Multienzimáticos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 4/metabolismo , Transducción de Señal/fisiología , Muerte Celular , Línea Celular Tumoral , Humanos , Proteína Quinasa 12 Activada por Mitógenos/genética , Complejos Multienzimáticos/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 4/genética
5.
J Infect Dis ; 210(2): 214-23, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24493823

RESUMEN

Influenza A virus triggers a contagious respiratory disease that can cause considerable morbidity and mortality. Using an in vitro approach, we previously demonstrated that the pattern recognition receptor retinoic acid-inducible gene I (RIG-I) plays a key role in influenza A virus-mediated immune response. However, the importance of RIG-I signaling in vivo has not been thoroughly examined, because of the lack of an appropriate mouse models. To circumvent this issue, we generated a new transgenic mouse overexpressing LGP2 (hereafter, "LGP2 TG mice"), a major regulator of the RIG-I signaling pathway. The time course of several parameters was compared in infected wild-type and LGP2 TG mice. We found that LGP2 TG mice displayed significantly reduced inflammatory mediators and a lower leukocyte infiltration into the bronchoalveolar airspace. More importantly, LGP2 TG mice had a significant survival advantage. Hence, our in vivo study reveals that LGP2 is a major downregulator of the influenza A virus-triggered detrimental inflammatory response.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , ARN Helicasas/metabolismo , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Mediadores de Inflamación/análisis , Leucocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Transducción de Señal , Análisis de Supervivencia
6.
J Am Chem Soc ; 134(50): 20533-43, 2012 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-23171049

RESUMEN

PTEN phosphatase is a tumor suppressor controlling notably cell growth, proliferation and survival. The multisite phosphorylation of the PTEN C-terminal tail regulates PTEN activity and intracellular trafficking. The dynamical nature of such regulatory events represents a crucial dimension for timing cellular decisions. Here we show that NMR spectroscopy allows reporting on the order and kinetics of clustered multisite phosphorylation events. We first unambiguously identify in vitro seven bona fide sites modified by CK2 and GSK3ß kinases and two new sites on the PTEN C-terminal tail. Then, monitoring the formation of transient intermediate phosphorylated states, we determine the sequence of these reactions and calculate their apparent rate constants. Finally, we assess the dynamic formation of these phosphorylation events induced by endogenous kinases directly in extracts of human neuroblastoma cells. Taken together, our data indicate that two cascades of events controlled by CK2 and GSK3ß occur independently on two clusters of sites (S380-S385 and S361-S370) and that in each cluster the reactions follow an ordered model with a distributive kinetic mechanism. Besides emphasizing the ability of NMR to quantitatively and dynamically follow post-translational modifications, these results bring a temporal dimension on the establishment of PTEN phosphorylation cascades.


Asunto(s)
Fosfohidrolasa PTEN/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Resonancia Magnética Nuclear Biomolecular , Fosfohidrolasa PTEN/química , Fosforilación
7.
Sci Signal ; 5(237): ra58, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22894835

RESUMEN

PTEN (phosphatase and tensin homolog deleted on chromosome 10) and MAST2 (microtubule-associated serine and threonine kinase 2) interact with each other through the PDZ domain of MAST2 (MAST2-PDZ) and the carboxyl-terminal (C-terminal) PDZ domain-binding site (PDZ-BS) of PTEN. These two proteins function as negative regulators of cell survival pathways, and silencing of either one promotes neuronal survival. In human neuroblastoma cells infected with rabies virus (RABV), the C-terminal PDZ domain of the viral glycoprotein (G protein) can target MAST2-PDZ, and RABV infection triggers neuronal survival in a PDZ-BS-dependent fashion. These findings suggest that the PTEN-MAST2 complex inhibits neuronal survival and that viral G protein disrupts this complex through competition with PTEN for binding to MAST2-PDZ. We showed that the C-terminal sequences of PTEN and the viral G protein bound to MAST2-PDZ with similar affinities. Nuclear magnetic resonance structures of these complexes exhibited similar large interaction surfaces, providing a structural basis for their binding specificities. Additionally, the viral G protein promoted the nuclear exclusion of PTEN in infected neuroblastoma cells in a PDZ-BS-dependent manner without altering total PTEN abundance. These findings suggest that formation of the PTEN-MAST2 complex is specifically affected by the viral G protein and emphasize how disruption of a critical protein-protein interaction regulates intracellular PTEN trafficking. In turn, the data show how the viral protein might be used to decipher the underlying molecular mechanisms and to clarify how the subcellular localization of PTEN regulates neuronal survival.


Asunto(s)
Glicoproteínas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Moleculares , Neuronas/fisiología , Fosfohidrolasa PTEN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Virus de la Rabia/metabolismo , Proteínas Virales/metabolismo , Unión Competitiva , Western Blotting , Calorimetría , Línea Celular Tumoral , Supervivencia Celular/fisiología , Glicoproteínas/química , Humanos , Inmunohistoquímica , Marcaje Isotópico , Proteínas Asociadas a Microtúbulos/química , Neuronas/metabolismo , Resonancia Magnética Nuclear Biomolecular , Dominios PDZ/fisiología , Fosfohidrolasa PTEN/química , Proteínas Serina-Treonina Quinasas/química , Espectrometría de Fluorescencia , Proteínas Virales/química
8.
Structure ; 19(10): 1518-24, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22000519

RESUMEN

PTPN4, a human tyrosine phosphatase, protects cells against apoptosis. This protection could be abrogated by targeting the PDZ domain of this phosphatase with a peptide mimicking the C-terminal sequence of the G protein of an attenuated rabies virus strain. Here, we demonstrate that glioblastoma death is triggered upon intracellular delivery of peptides, either from viral origin or from known endogenous ligands of PTPN4-PDZ, such as the C terminus sequence of the glutamate receptor subunit GluN2A. The killing efficiency of peptides closely reflects their affinities for the PTPN4-PDZ. The crystal structures of two PTPN4-PDZ/peptide complexes allow us to pinpoint the main structural determinants of binding and to synthesize a peptide of high affinity for PTPN4-PDZ enhancing markedly its cell death capacity. These results allow us to propose a potential mechanism for the efficiency of peptides and provide a target and a robust framework for the design of new pro-death compounds.


Asunto(s)
Muerte Celular , Glioblastoma/patología , Dominios PDZ , Péptidos/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 4/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Citometría de Flujo , Glioblastoma/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Complejos Multiproteicos/metabolismo , Mutación Puntual , Unión Proteica , Estructura Secundaria de Proteína , Virus de la Rabia/química , Receptores de N-Metil-D-Aspartato/metabolismo , Alineación de Secuencia , Proteínas Virales/síntesis química , Proteínas Virales/farmacología
9.
J Neurovirol ; 17(4): 353-67, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21805057

RESUMEN

Rabies virus (RABV) is a neurotropic virus transmitted by the bite of an infected animal that triggers a fatal encephalomyelitis. During its migration in the nervous system (NS), RABV triggers an innate immune response, including a type I IFN response well known to limit viral infections. We showed that although the neuroinvasive RABV strain CVS-NIV dampens type I IFN signaling by inhibiting IRF3 phosphorylation and STAT2 translocation, an early and transient type I IFN response is still triggered in the infected neuronal cells and NS. This urged us to investigate the role of type I IFN on RABV infection. We showed that primary mouse neurons (DRGs) of type I IFN(α/ß) receptor deficient mice (IFNAR(-/-) mice) were more susceptible to RABV than DRGs of WT mice. In addition, exogenous type I IFN is partially efficient in preventing and slowing down infection in human neuroblastoma cells. Intra-muscular inoculation of type I IFNAR deficient mice [IFNAR(-/-) mice and NesCre ((+/-)) IFNAR ((flox/flox)) mice lacking IFNAR in neural cells of neuroectodermal origin only] with RABV reveals that the type I IFN response limits RABV dissemination in the inoculated muscle, slows down invasion of the spinal cord, and delays mortality. Thus, the type I IFN which is still produced in the NS during RABV infection is efficient enough to reduce neuroinvasiveness and pathogenicity and partially protect the host from fatal infection.


Asunto(s)
Interferón Tipo I , Neuronas/inmunología , Virus de la Rabia/fisiología , Rabia/inmunología , Receptor de Interferón alfa y beta/deficiencia , Transducción de Señal/inmunología , Médula Espinal/inmunología , Animales , Línea Celular Tumoral , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Inyecciones Intramusculares , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Interferón Tipo I/farmacología , Ratones , Ratones Noqueados , Neuroblastoma/inmunología , Neuroblastoma/patología , Neuroblastoma/virología , Neuronas/virología , Cultivo Primario de Células , Rabia/mortalidad , Rabia/patología , Rabia/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Médula Espinal/virología , Tasa de Supervivencia , Carga Viral/inmunología
10.
J Virol ; 85(13): 6657-68, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21525357

RESUMEN

The neurotropic rabies virus (RABV) has developed several evasive strategies, including immunoevasion, to successfully infect the nervous system (NS) and trigger a fatal encephalomyelitis. Here we show that expression of LGP2, a protein known as either a positive or negative regulator of the RIG-I-mediated innate immune response, is restricted in the NS. We used a new transgenic mouse model (LGP2 TG) overexpressing LGP2 to impair the innate immune response to RABV and thus revealed the role of the RIG-I-mediated innate immune response in RABV pathogenesis. After infection, LGP2 TG mice exhibited reduced expression of inflammatory/chemoattractive molecules, beta interferon (IFN-ß), and IFN-stimulated genes in their NS compared to wild-type (WT) mice, demonstrating the inhibitory function of LGP2 in the innate immune response to RABV. Surprisingly, LGP2 TG mice showed more viral clearance in the brain and lower morbidity than WT mice, indicating that the host innate immune response, paradoxically, favors RABV neuroinvasiveness and morbidity. LGP2 TG mice exhibited similar neutralizing antibodies and microglia activation to those of WT mice but showed a reduction of infiltrating CD4(+) T cells and less disappearance of infiltrating CD8(+) T cells. This occurred concomitantly with reduced neural expression of the IFN-inducible protein B7-H1, an immunoevasive protein involved in the elimination of infiltrated CD8(+) T cells. Our study shows that the host innate immune response favors the infiltration of T cells and, at the same time, promotes CD8(+) T cell elimination. Thus, to a certain extent, RABV exploits the innate immune response to develop its immunoevasive strategy.


Asunto(s)
Antígeno B7-1/metabolismo , Inmunidad Innata , Glicoproteínas de Membrana/metabolismo , Péptidos/metabolismo , ARN Helicasas/metabolismo , Virus de la Rabia/inmunología , Virus de la Rabia/patogenicidad , Rabia/inmunología , Animales , Antígeno B7-1/genética , Antígeno B7-H1 , Encéfalo/inmunología , Encéfalo/virología , Línea Celular , Línea Celular Tumoral , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células 3T3 NIH , Neuronas/inmunología , Neuronas/virología , Péptidos/genética , ARN Helicasas/genética , Rabia/virología , Linfocitos T/inmunología
11.
Sci Signal ; 3(105): ra5, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20086240

RESUMEN

The capacity of a rabies virus to promote neuronal survival (a signature of virulence) or death (a marker of attenuation) depends on the cellular partners recruited by the PDZ-binding site (PDZ-BS) of its envelope glycoprotein (G). Neuronal survival requires the selective association of the PDZ-BS of G with the PDZ domains of two closely related serine-threonine kinases, MAST1 and MAST2. Here, we found that a single amino acid change in the PDZ-BS triggered the apoptotic death of infected neurons and enabled G to interact with additional PDZ partners, in particular the tyrosine phosphatase PTPN4. Knockdown of PTPN4 abrogated virus-mediated apoptosis. Thus, we propose that attenuation of rabies virus requires expansion of the set of host PDZ proteins with which G interacts, which interferes with the finely tuned homeostasis required for survival of the infected neuron.


Asunto(s)
Virus de la Rabia/patogenicidad , Proteínas del Envoltorio Viral/fisiología , Sustitución de Aminoácidos , Animales , Apoptosis , Citoplasma , Ratones , Neuronas/virología , Dominios PDZ , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 4 , Rabia , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Virulencia
12.
J Immunol ; 180(11): 7506-15, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18490751

RESUMEN

Rabies virus is the etiological agent of an acute encephalitis, which in absence of post exposure treatment is fatal in almost all cases. Virus lethality rests on its ability to evade the immune response. In this study, we analyzed the role of the immuno-inhibitory molecule B7-H1 in this virus strategy. We showed that in the brain and spinal cord of mice, rabies virus infection resulted in significant up-regulation of B7-H1 expression, which is specifically expressed in infected neurons. Correlatively, clinical rabies in B7-H1(-/-) mice is markedly less severe than in wild-type mice. B7-H1(-/-) mice display resistance to rabies. Virus invasion is reduced and the level of migratory CD8 T cells increases into the nervous system, while CD4/CD8 ratio remains unchanged in the periphery. In vivo, neuronal B7-H1 expression is critically depending on TLR3 signaling and IFN-beta, because TLR3(-/-) mice--in which IFN-beta production is reduced--showed only a limited increase of B7-H1 transcripts after infection. These data provide evidence that neurons can express the B7-H1 molecule after viral stress or exposure to a particular cytokine environment. They show that the B7-H1/PD-1 pathway can be exploited locally and in an organ specific manner--here the nervous system--by a neurotropic virus to promote successful host invasion.


Asunto(s)
Antígenos CD/metabolismo , Antígeno B7-1/metabolismo , Encefalitis Viral/inmunología , Glicoproteínas de Membrana/metabolismo , Neuronas/metabolismo , Péptidos/metabolismo , Virus de la Rabia/inmunología , Rabia/inmunología , Animales , Antígenos CD/inmunología , Antígeno B7-1/inmunología , Antígeno B7-H1 , Encéfalo/inmunología , Encéfalo/metabolismo , Línea Celular Tumoral , Células Cultivadas , Encefalitis Viral/virología , Humanos , Interferón beta/inmunología , Interferón beta/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Mutantes , Neuronas/inmunología , Neuronas/virología , Péptidos/inmunología , Rabia/metabolismo , Rabia/virología , Médula Espinal/inmunología , Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Immunol ; 180(10): 6760-7, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18453596

RESUMEN

West Nile virus (WNV) is an emerging neurotropic flavivirus. We investigated the dynamics of immune cell recruitment in peripheral tissues and in the CNS during WNV encephalitis in an immunocompetent mouse model. In the periphery, immune cell expansion can successfully limit viremia and lymphoid tissue infection. However, viral clearance in the periphery is too late to prevent viral invasion of the CNS. In the CNS, innate immune cells, including microglia/macrophages, NK cells, and plasmacytoid dendritic cells, greatly expand as the virus invades the brain, whereas B and T cells are recruited after viral invasion, and fail to control the spread of the virus. Thus, the onset of WNV encephalitis was correlated both with CNS viral infection and with a large local increase of innate immune cells. Interestingly, we identify a new immune cell type: CD19(+)B220(-) BST-2(+), which we name G8-ICs. These cells appear during peripheral infection and enter the CNS. G8-ICs express high levels of MHC class II, stain for viral Ag, and are localized in the paracortical zone of lymph nodes, strongly suggesting they are previously unidentified APCs that appear in response to viral infection.


Asunto(s)
Antígenos CD19/metabolismo , Antígenos CD/metabolismo , Quimiotaxis de Leucocito/inmunología , Antígenos Comunes de Leucocito/metabolismo , Leucocitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Fiebre del Nilo Occidental/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Linfocitos B/inmunología , Linfocitos B/virología , Linaje de la Célula , Sistema Nervioso Central/citología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas Ligadas a GPI , Antígenos de Histocompatibilidad Clase II/metabolismo , Células Asesinas Naturales/inmunología , Leucocitos/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Macrófagos/inmunología , Ratones , Microglía/inmunología , Linfocitos T/inmunología , Linfocitos T/virología
14.
Hum Immunol ; 68(4): 294-302, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17400066

RESUMEN

Human Leukocyte Antigen (HLA)-G and E are nonclassical human MHC class I molecules. They may promote tolerance leading to virus and tumor immune escape. We recently described that the herpes simplex virus type 1 (HSV-1), a neurotropic virus inducing chronic infection and neuron latency, and rabies virus (RABV), a neuronotropic virus triggering acute neuron infection, up-regulate HLA-G expression in human neurons (NT2-N). Surface expression was only detected after RABV infection. We investigated here whether RABV and HSV-1 up-regulate HLA-E expression in human neuronal precursors (Ntera-2D/1). We found that RABV, not HSV-1, up-regulates HLA-E expression, nevertheless HLA-E could not be detected on the surface of RABV-infected Ntera-2D/1. Altogether these data suggest that HLA-G and not HLA-E could contribute to the immune escape of RABV. In contrast, there was no evidence that these molecules are used by latent HSV-1 infection. Thus, neurotropic viruses that escape the host immune response totally (RABV) or partially (HSV-1) regulate HLA-G expression on human neuronal cells differentially.


Asunto(s)
Antígenos HLA/genética , Herpesvirus Humano 1/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Neuronas/inmunología , Virus de la Rabia/inmunología , Línea Celular , Membrana Celular/inmunología , Antígenos HLA/biosíntesis , Antígenos HLA-G , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Neuronas/metabolismo , Neuronas/virología , Regulación hacia Arriba/inmunología , Antígenos HLA-E
15.
J Virol ; 79(24): 15226-37, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16306594

RESUMEN

HLA-G is a nonclassical human major histocompatibility complex class I molecule. It may promote tolerance, leading to acceptance of the semiallogeneic fetus and tumor immune escape. We show here that two viruses-herpes simplex virus type 1 (HSV-1), a neuronotropic virus inducing acute infection and neuron latency; and rabies virus (RABV), a neuronotropic virus triggering acute neuron infection-upregulate the neuronal expression of several HLA-G isoforms, including HLA-G1 and HLA-G5, the two main biologically active isoforms. RABV induces mostly HLA-G1, and HSV-1 induces mostly HLA-G3 and HLA-G5. HLA-G expression is upregulated in infected cells and neighboring uninfected cells. Soluble mediators, such as beta interferon (IFN-beta) and IFN-gamma, upregulate HLA-G expression in uninfected cells. The membrane-bound HLA-G1 isoform was detected on the surface of cultured RABV-infected neurons but not on the surface of HSV-1-infected cells. Thus, neuronotropic viruses that escape the host immune response totally (RABV) or partially (HSV-1) regulate HLA-G expression on human neuronal cells differentially. HLA-G may therefore be involved in the escape of certain viruses from the immune response in the nervous system.


Asunto(s)
Antígenos HLA/metabolismo , Herpesvirus Humano 1/fisiología , Antígenos de Histocompatibilidad Clase I/metabolismo , Sistema Nervioso/virología , Neuronas/metabolismo , Antígenos HLA/genética , Antígenos HLA-G , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Sistema Nervioso/patología , Neuronas/virología , ARN Mensajero/biosíntesis , Virus de la Rabia/fisiología , Transcripción Genética , Células Tumorales Cultivadas , Regulación hacia Arriba/inmunología
16.
J Virol ; 79(20): 12893-904, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16188991

RESUMEN

To study the capacity of human neurons to mount innate immunity responses to viral infections, we infected cells of a human postmitotic neuron-derivative cell line, NT2-N, with rabies virus (RABV) and herpes simplex type 1 (HSV-1). Changes in neuronal gene expression were analyzed by use of Affymetrix microarrays. Applying a twofold cutoff, RABV increased the transcription of 228 genes, and HSV-1 increased the transcription of 263 genes. The most striking difference between the two infections concerns genes involved in immunity. These genes represent 24% of the RABV-upregulated genes and only 4.9% of the HSV-1-upregulated genes. Following RABV infection, the most upregulated genes belong to the immunity cluster and included almost exclusively genes for beta interferon (IFN-beta) primary and secondary responses as well as genes for chemokines (CCL-5, CXCL-10) and inflammatory cytokines (interleukin 6 [IL-6], tumor necrosis factor alpha, interleukin 1 alpha). In contrast, HSV-1 infection did not increase IFN-beta gene transcripts and triggered the production of only IL-6 and interferon regulatory factor 1 mRNAs. The microarray results were confirmed by real-time PCR, immunocytochemistry, and enzyme-linked immunosorbent assay. Human neurons were found to express Toll-like receptor 3. They produced IFN-beta after treatment with poly(I:C) but not with lipopolysaccharide. Thus, human neurons can mount an innate immunity response to double-stranded RNA. These observations firmly establish that human neurons, in absence of glia, have the intrinsic machinery to sense virus infection.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 1/inmunología , Interferón beta/biosíntesis , Glicoproteínas de Membrana/metabolismo , Neuronas/inmunología , Neuronas/virología , Virus de la Rabia/inmunología , Rabia/virología , Receptores de Superficie Celular/metabolismo , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Herpes Simple/inmunología , Humanos , Inmunidad Innata , Inmunohistoquímica , Interferón beta/genética , Neuronas/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Rabia/inmunología , Receptor Toll-Like 3 , Receptores Toll-Like
17.
J Neurovirol ; 10(6): 372-82, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15765808

RESUMEN

Following its injection into the hindlimbs of mice, CVS, a highly pathogenic strain of rabies virus, invades the spinal cord and brain resulting in the death of the animal. In contrast, central nervous system (CNS) invasion by PV, a strain of attenuated pathogenicity, is restricted to the spinal cord and mice infected with this virus survive. Lymphocytes display transient migration into the infected CNS in fatal rabies and sustained migration in nonfatal rabies. The transient migration of T cells in fatal rabies is associated with an increase in T-cell apoptosis. We found that the early production of Fas ligand (FasL) mRNAs was up-regulated only in fatal rabies. FasL is produced by several neuronal cells and mainly in infected neurons. In mice lacking FasL (gld), infection with the neuroinvasive rabies virus strain was less severe, and the number of CD3 T cells undergoing apoptosis was smaller than that in normal mice. These data provide strong evidence that fatal rabies virus infection involves the early triggering of FasL production leading to the destruction of migratory T cells by the Fas/FasL apoptosis pathway. This mechanism could be in part responsible for the fact that T cells cannot control neuroinvasive rabies infection. Thus, rabies virus seems to use an immunosubversive strategy that takes advantage of the immune privilege status of the CNS.


Asunto(s)
Infecciones del Sistema Nervioso Central/inmunología , Sistema Nervioso Central/inmunología , Glicoproteínas de Membrana/metabolismo , Rabia/inmunología , Regulación hacia Arriba , Animales , Apoptosis , Complejo CD3/metabolismo , Movimiento Celular/inmunología , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Infecciones del Sistema Nervioso Central/patología , Infecciones del Sistema Nervioso Central/virología , Proteína Ligando Fas , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , ARN Mensajero/análisis , Virus de la Rabia/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Linfocitos T/virología , Receptor fas
18.
Biochimie ; 85(8): 777-88, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-14585545

RESUMEN

Rabies virus (RV) causes a non-lytic infection of neurons leading to a fatal myeloencephalitis in mammals including humans. By comparing the infection of the nervous system of mice by a highly pathogenic neuroinvasive strain of RV (CVS) and by a strain of attenuated pathogenicity (PV) with restricted brain invasion, we showed that RV neuroinvasiveness results of three factors: not only neurotropic RV avoids induced neuron cell death but also "protective" T cells that migrate into the infected nervous system are killed by apoptosis and finally inflammation of the infected nervous system is limited. Our data suggest that the preservation of the neuronal network, the limitation of the inflammation and the destruction of T cells that invade the CNS in response to the infection are crucial events for RV neuroinvasion and for transmission of RV to another animal.


Asunto(s)
Apoptosis/fisiología , Sistema Nervioso Central/virología , Virus de la Rabia/patogenicidad , Animales , Antígenos CD11/metabolismo , Complejo CD3/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Femenino , Humanos , Interferón gamma/genética , Interleucina-1/genética , Interleucina-6/genética , Células Jurkat , Ratones , Ratones Endogámicos BALB C , Neuroblastoma/patología , Neuroblastoma/virología , Neuronas/patología , Neuronas/virología , Reacción en Cadena de la Polimerasa/métodos , Rabia/patología , Rabia/virología , Médula Espinal/patología , Médula Espinal/virología , Linfocitos T/patología , Linfocitos T/virología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1 , Células Tumorales Cultivadas
19.
Virology ; 314(2): 549-61, 2003 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-14554083

RESUMEN

We report here that rabies virus strains, currently used to immunize wildlife against rabies, induce not only caspase-dependent apoptosis in the human lymphoblastoid Jurkat T cell line (Jurkat-vect), but also a caspase-independent pathway involving the apoptosis-inducing factor (AIF). In contrast, a strain of neurotropic RV that does not induce apoptosis did not activate caspases or induce AIF translocation. Bcl-2 overproduction in Jurkat T cells (Jurkat-Bcl-2) abolished both pathways. ERA infection and production were similar in Jurkat-vect and Jurkat-Bcl-2 cells, indicating Bcl-2 has no direct antiviral effects. Bcl-2 production is naturally upregulated by day 3 in ERA-infected Jurkat-vect cultures. The increase in Bcl-2 levels seems to be controlled by the virus infection itself and results in the establishment of long-term, persistently infected cultures that continue to produce virus. Thus, in infections with live RV vaccine strains, infected cells may be productive reservoirs of virus in the long term. This may account for the high efficacy of live rabies vaccines.


Asunto(s)
Apoptosis , Células Jurkat/virología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Vacunas Antirrábicas , Virus de la Rabia/fisiología , Factor Inductor de la Apoptosis , Caspasas/metabolismo , Línea Celular , Flavoproteínas/genética , Flavoproteínas/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Rabia/fisiopatología , Rabia/virología , Virus de la Rabia/patogenicidad , Vacunas Atenuadas
20.
J Virol ; 77(19): 10537-47, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12970438

RESUMEN

We showed that, unlike pathogenic rabies virus (RV) strain CVS, attenuated RV strain ERA triggers the caspase-dependent apoptosis of human cells. Furthermore, we observed that the induction of apoptosis is correlated with a particular virus antigen distribution: the overexpression of the viral G protein on the cell surface, with continuous localization on the cytoplasmic membrane, and large cytoplasmic inclusions of the N protein. To determine whether one of these two major RV proteins (G and N proteins) triggers apoptosis, we constructed transgenic Jurkat T-cell lines that drive tetracycline-inducible gene expression to produce the G and N proteins of ERA and CVS individually. The induction of ERA G protein (G-ERA) expression but not of ERA N protein expression resulted in apoptosis, and G-ERA was more efficient at triggering apoptosis than was CVS G protein. To test whether other viral proteins participated in the induction of apoptosis, human cells were infected with recombinant RV in which the G protein gene from the attenuated strain had been replaced by its virulent strain counterpart (CVS). Only RV containing the G protein from the nonpathogenic RV strain was able to trigger the apoptosis of human cells. Thus, the ability of RV strains to induce apoptosis is largely determined by the viral G protein.


Asunto(s)
Antígenos Virales , Apoptosis , Glicoproteínas/fisiología , Virus de la Rabia/patogenicidad , Proteínas del Envoltorio Viral/fisiología , Doxiciclina/farmacología , Glicoproteínas/genética , Humanos , Células Jurkat , Nucleocápside/fisiología , Proteínas de la Nucleocápside/fisiología , Virus de la Rabia/inmunología , Proteínas del Envoltorio Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA