Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 47(1): 12-14, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723545

RESUMEN

Sickness in mammals can lead to cognition deficits, although the underlying mechanisms remain elusive. In a recent Nature Medicine article, Garré et al. (2017) report that sickness-induced cortical dendritic spine loss and impaired memory formation is mediated by CX3CR1+ monocyte-derived TNF-α.


Asunto(s)
Espinas Dendríticas/fisiología , Trastornos Mentales/inmunología , Monocitos/fisiología , Neuronas Motoras/fisiología , Red Nerviosa , Plasticidad Neuronal , Virosis/inmunología , Animales , Receptor 1 de Quimiocinas CX3C , Humanos , Memoria , Trastornos Mentales/etiología , Trastornos Mentales/psicología , Ratones , Monocitos/virología , Neuronas Motoras/virología , Poli I-C/inmunología , Receptores de Quimiocina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Virosis/complicaciones , Virosis/psicología
2.
J Virol ; 98(7): e0039724, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38869283

RESUMEN

Enterovirus D68 (EV-D68) is an emerging pathogen that can cause severe respiratory and neurologic disease [acute flaccid myelitis (AFM)]. Intramuscular (IM) injection of neonatal Swiss Webster (SW) mice with US/IL/14-18952 (IL52), a clinical isolate from the 2014 EV-D68 epidemic, results in many of the pathogenic features of human AFM, including viral infection of the spinal cord, death of motor neurons, and resultant progressive paralysis. In distinction, CA/14-4231 (CA4231), another clinical isolate from the 2014 EV-D68 outbreak, does not cause paralysis in mice, does not grow in the spinal cord, and does not cause motor neuron loss following IM injection. A panel of chimeric viruses containing sequences from IL52 and CA4231 was used to demonstrate that VP1 is the main determinant of EV-D68 neurovirulence following IM injection of neonatal SW mice. VP1 contains four amino acid differences between IL52 and CA4231. Mutations resulting in substituting these four amino acids (CA4231 residues into the IL52 polyprotein) completely abolished neurovirulence. Conversely, mutations resulting in substituting VP1 IL52 amino acid residues into the CA4231 polyprotein created a virus that induced paralysis to the same degree as IL52. Neurovirulence following infection of neonatal SW mice with parental and chimeric viruses was associated with viral growth in the spinal cord. IMPORTANCE: Emerging viruses allow us to investigate mutations leading to increased disease severity. Enterovirus D68 (EV-D68), once the cause of rare cases of respiratory illness, recently acquired the ability to cause severe respiratory and neurologic disease. Chimeric viruses were used to demonstrate that viral structural protein VP1 determines growth in the spinal cord, motor neuron loss, and paralysis following intramuscular (IM) injection of neonatal Swiss Webster (SW) mice with EV-D68. These results have relevance for predicting the clinical outcome of future EV-D68 epidemics as well as targeting retrograde transport as a potential strategy for treating virus-induced neurologic disease.


Asunto(s)
Proteínas de la Cápside , Enfermedades Virales del Sistema Nervioso Central , Modelos Animales de Enfermedad , Enterovirus Humano D , Infecciones por Enterovirus , Mielitis , Enfermedades Neuromusculares , Animales , Enterovirus Humano D/patogenicidad , Enterovirus Humano D/genética , Enterovirus Humano D/fisiología , Mielitis/virología , Ratones , Infecciones por Enterovirus/virología , Infecciones por Enterovirus/patología , Enfermedades Neuromusculares/virología , Enfermedades Neuromusculares/patología , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Enfermedades Virales del Sistema Nervioso Central/virología , Enfermedades Virales del Sistema Nervioso Central/patología , Humanos , Médula Espinal/virología , Médula Espinal/patología , Neuronas Motoras/virología , Neuronas Motoras/patología , Animales Recién Nacidos , Virulencia , Parálisis/virología
3.
J Neuroinflammation ; 21(1): 236, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39334427

RESUMEN

West Nile virus (WNV) and Usutu virus (USUV) are closely related flaviviruses with differing capacities to cause neurological disease in humans. WNV is thought to use a transneural route of neuroinvasion along motor neurons and causes severe motor deficits. The potential for use of transneural routes of neuroinvasion by USUV has not been investigated experimentally, and evidence from the few clinical case reports of USUV-associated neuroinvasive disease is lacking. We hypothesised that, compared with WNV, USUV is less able to infect motor neurons, and therefore determined the susceptibility of human induced pluripotent stem cell (iPSC)-derived spinal cord motor neurons to infection. Both viruses could grow to high titres in iPSC-derived neural cultures. However, USUV could not productively infect motor neurons due to restriction by the antiviral response, which was not induced upon WNV infection. Inhibition of the antiviral response allowed for widespread infection and transportation of USUV along motor neurons within a compartmented culture system. These results show a stark difference in the ability of these two viruses to evade initiation of intrinsic antiviral immunity. Our data suggests that USUV cannot infect motor neurons in healthy individuals but in case of immunodeficiency may pose a risk for motor-related neurological disease and transneural invasion.


West Nile virus, but not Usutu virus, can productively infect human motor neurons as a possible route of neuroinvasion.


Asunto(s)
Flavivirus , Células Madre Pluripotentes Inducidas , Neuronas Motoras , Virus del Nilo Occidental , Humanos , Virus del Nilo Occidental/fisiología , Virus del Nilo Occidental/patogenicidad , Neuronas Motoras/virología , Células Cultivadas , Flavivirus/fisiología , Células Madre Pluripotentes Inducidas/virología , Infecciones por Flavivirus/virología , Fiebre del Nilo Occidental/virología , Animales , Médula Espinal/virología
4.
J Virol ; 97(10): e0095923, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37772825

RESUMEN

IMPORTANCE: Viral encephalomyelitis outcome is dependent on host responses to neuronal infection. Interferon (IFN) is an important component of the innate response, and IFN regulatory factor (IRF) 7 is an inducible transcription factor for the synthesis of IFN-α. IRF7-deficient mice develop fatal paralysis after CNS infection with Sindbis virus, while wild-type mice recover. Irf7 -/- mice produce low levels of IFN-α but high levels of IFN-ß with induction of IFN-stimulated genes, so the reason for this difference is not understood. The current study shows that Irf7 -/- mice developed inflammation earlier but failed to clear virus from motor neuron-rich regions of the brainstem and spinal cord. Levels of IFN-γ and virus-specific antibody were comparable, indicating that IRF7 deficiency does not impair expression of these known viral clearance factors. Therefore, IRF7 is either necessary for the neuronal response to currently identified mediators of clearance or enables the production of additional antiviral factor(s) needed for clearance.


Asunto(s)
Infecciones por Alphavirus , Encefalomielitis , Factor 7 Regulador del Interferón , Virus Sindbis , Animales , Ratones , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Tronco Encefálico/virología , Encefalomielitis/inmunología , Encefalomielitis/virología , Inflamación/virología , Factor 7 Regulador del Interferón/deficiencia , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferón beta/inmunología , Interferón beta/metabolismo , Neuronas Motoras/virología , Virus Sindbis/inmunología , Médula Espinal/virología
5.
J Virol ; 95(12)2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33827951

RESUMEN

During viral infection, the dynamic virus-host relationship is constantly in play. Many cellular proteins, such as RNA-binding proteins (RBPs), have been shown to mediate antiviral responses during viral infection. Here, we report that the RBP FUS/TLS (fused in sarcoma/translocated in liposarcoma) acts as a host-restricting factor against infection with coxsackievirus B3 (CVB3). Mechanistically, we found that deletion of FUS leads to increased viral RNA transcription and enhanced internal ribosome entry site (IRES)-driven translation, with no apparent impact on viral RNA stability. We further demonstrated that FUS physically interacts with the viral genome, which may contribute to direct inhibition of viral RNA transcription/translation. Moreover, we identified a novel function for FUS in regulating host innate immune response. We show that in the absence of FUS, gene expression of type I interferons and proinflammatory cytokines elicited by viral or bacterial infection is significantly impaired. Emerging evidence suggests a role for stress granules (SGs) in antiviral innate immunity. We further reveal that knockout of FUS abolishes the ability to form SGs upon CVB3 infection or poly(I·C) treatment. Finally, we show that, to avoid FUS-mediated antiviral response and innate immunity, CVB3 infection results in cytoplasmic mislocalization and cleavage of FUS through the enzymatic activity of viral proteases. Together, our findings in this study identify FUS as a novel host antiviral factor which restricts CVB3 replication through direct inhibition of viral RNA transcription and protein translation and through regulation of host antiviral innate immunity.IMPORTANCE Enteroviruses are common human pathogens, including those that cause myocarditis (coxsackievirus B3 [CVB3]), poliomyelitis (poliovirus), and hand, foot, and mouth disease (enterovirus 71). Understanding the virus-host interaction is crucial for developing means of treating and preventing diseases caused by these pathogens. In this study, we explored the interplay between the host RNA-binding protein FUS/TLS and CVB3 and found that FUS/TLS restricts CVB3 replication through direct inhibition of viral RNA transcription/translation and through regulation of cellular antiviral innate immunity. To impede the antiviral role of FUS, CVB3 targets FUS for mislocalization and cleavage. Findings from this study provide novel insights into interactions between CVB3 and FUS, which may lead to novel therapeutic interventions against enterovirus-induced diseases.


Asunto(s)
Enterovirus Humano B/inmunología , Enterovirus Humano B/fisiología , Inmunidad Innata , Proteína FUS de Unión a ARN/metabolismo , Proteasas Virales 3C/metabolismo , Animales , Antivirales/farmacología , Autofagia , Línea Celular , Cisteína Endopeptidasas/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Citoplasma/metabolismo , Gránulos Citoplasmáticos/metabolismo , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Genoma Viral , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Sitios Internos de Entrada al Ribosoma , Ratones , Neuronas Motoras/virología , Poli I-C/farmacología , Biosíntesis de Proteínas , ARN Viral/genética , ARN Viral/metabolismo , Proteína FUS de Unión a ARN/genética , Estrés Fisiológico , Transcripción Genética , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
6.
Mol Ther ; 28(8): 1887-1901, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32470325

RESUMEN

Spinal muscular atrophy (SMA) is a neuromuscular disease mainly caused by mutations or deletions in the survival of motor neuron 1 (SMN1) gene and characterized by the degeneration of motor neurons and progressive muscle weakness. A viable therapeutic approach for SMA patients is a gene replacement strategy that restores functional SMN expression using adeno-associated virus serotype 9 (AAV9) vectors. Currently, systemic or intra-cerebrospinal fluid (CSF) delivery of AAV9-SMN is being explored in clinical trials. In this study, we show that the postnatal delivery of an AAV9 that expresses SMN under the control of the neuron-specific promoter synapsin selectively targets neurons without inducing re-expression in the peripheral organs of SMA mice. However, this approach is less efficient in restoring the survival and neuromuscular functions of SMA mice than the systemic or intra-CSF delivery of an AAV9 in which SMN is placed under the control of a ubiquitous promoter. This study suggests that further efforts are needed to understand the extent to which SMN is required in neurons and peripheral organs for a successful therapeutic effect.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/virología , Atrofia Muscular Espinal/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Animales , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Locomoción , Ratones , Atrofia Muscular Espinal/tratamiento farmacológico , Fenotipo , Pronóstico , Regiones Promotoras Genéticas , Médula Espinal/metabolismo , Médula Espinal/patología , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Transducción Genética , Resultado del Tratamiento
7.
J Virol ; 93(16)2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31167912

RESUMEN

Enterovirus D68 (EV-D68) is an emerging virus that has been identified as a cause of recent outbreaks of acute flaccid myelitis (AFM), a poliomyelitis-like spinal cord syndrome that can result in permanent paralysis and disability. In experimental mouse models, EV-D68 spreads to, infects, and kills spinal motor neurons following infection by various routes of inoculation. The topography of virus-induced motor neuron loss correlates with the pattern of paralysis. The mechanism(s) by which EV-D68 spreads to target motor neurons remains unclear. We sought to determine the capacity of EV-D68 to spread by the neuronal route and to determine the role of known EV-D68 receptors, sialic acid and intracellular adhesion molecule 5 (ICAM-5), in neuronal infection. To do this, we utilized a microfluidic chamber culture system in which human induced pluripotent stem cell (iPSC) motor neuron cell bodies and axons can be compartmentalized for independent experimental manipulation. We found that EV-D68 can infect motor neurons via their distal axons and spread by retrograde axonal transport to the neuronal cell bodies. Virus was not released from the axons via anterograde axonal transport after infection of the cell bodies. Prototypic strains of EV-D68 depended on sialic acid for axonal infection and transport, while contemporary circulating strains isolated during the 2014 EV-D68 outbreak did not. The pattern of infection did not correspond with the ICAM-5 distribution and expression in either human tissue, the mouse model, or the iPSC motor neurons.IMPORTANCE Enterovirus D68 (EV-D68) infections are on the rise worldwide. Since 2014, the United States has experienced biennial spikes in EV-D68-associated acute flaccid myelitis (AFM) that have left hundreds of children paralyzed. Much remains to be learned about the pathogenesis of EV-D68 in the central nervous system (CNS). Herein we investigated the mechanisms of EV-D68 CNS invasion through neuronal pathways. A better understanding of EV-D68 infection in experimental models may allow for better prevention and treatment strategies of EV-D68 CNS disease.


Asunto(s)
Transporte Axonal , Enterovirus Humano D/fisiología , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/virología , Interacciones Huésped-Patógeno , Neuronas Motoras/metabolismo , Neuronas Motoras/virología , Ácido N-Acetilneuramínico/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Enfermedades Virales del Sistema Nervioso Central/metabolismo , Enfermedades Virales del Sistema Nervioso Central/virología , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Neuronas Motoras/citología , Mielitis/metabolismo , Mielitis/virología , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neuromusculares/metabolismo , Enfermedades Neuromusculares/virología , Parálisis/etiología
8.
J Neurovirol ; 24(3): 273-290, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29476408

RESUMEN

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.


Asunto(s)
Encefalitis/fisiopatología , Trastornos Motores/fisiopatología , Mielitis/fisiopatología , Convulsiones/fisiopatología , Infección por el Virus Zika/fisiopatología , Virus Zika/patogenicidad , Potenciales de Acción/fisiología , Animales , Astrocitos/inmunología , Astrocitos/patología , Astrocitos/virología , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Modelos Animales de Enfermedad , Encefalitis/inmunología , Encefalitis/virología , Femenino , Humanos , Interferón-alfa/deficiencia , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferón beta/deficiencia , Interferón beta/genética , Interferón beta/inmunología , Interferón gamma/deficiencia , Interferón gamma/genética , Interferón gamma/inmunología , Masculino , Ratones , Ratones Noqueados , Trastornos Motores/inmunología , Trastornos Motores/virología , Neuronas Motoras/inmunología , Neuronas Motoras/patología , Neuronas Motoras/virología , Músculo Esquelético/fisiología , Mielitis/inmunología , Mielitis/virología , Neutrófilos/inmunología , Neutrófilos/patología , Neutrófilos/virología , Convulsiones/inmunología , Convulsiones/virología , Médula Espinal/inmunología , Médula Espinal/patología , Médula Espinal/virología , Linfocitos T/inmunología , Linfocitos T/patología , Linfocitos T/virología , Virus Zika/crecimiento & desarrollo , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
9.
J Neurovirol ; 23(2): 186-204, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27761801

RESUMEN

Neurological respiratory deficits are serious outcomes of West Nile virus (WNV) disease. WNV patients requiring intubation have a poor prognosis. We previously reported that WNV-infected rodents also appear to have respiratory deficits when assessed by whole-body plethysmography and diaphragmatic electromyography. The purpose of this study was to determine if the nature of the respiratory deficits in WNV-infected rodents is neurological and if deficits are due to a disorder of brainstem respiratory centers, cervical spinal cord (CSC) phrenic motor neuron (PMN) circuitry, or both. We recorded phrenic nerve (PN) activity and found that in WNV-infected mice, PN amplitude is reduced, corroborating a neurological basis for respiratory deficits. These results were associated with a reduction in CSC motor neuron number. We found no dramatic deficits, however, in brainstem-mediated breathing rhythm generation or responses to hypercapnia. PN frequency and pattern parameters were normal, and all PN parameters changed appropriately upon a CO2 challenge. Histological analysis revealed generalized microglia activation, astrocyte reactivity, T cell and neutrophil infiltration, and mild histopathologic lesions in both the brainstem and CSC, but none of these were tightly correlated with PN function. Similar results in PN activity, brainstem function, motor neuron number, and histopathology were seen in WNV-infected hamsters, except that histopathologic lesions were more severe. Taken together, the results suggest that respiratory deficits in acute WNV infection are primarily due to a lower motor neuron disorder affecting PMNs and the PN rather than a brainstem disorder. Future efforts should focus on markers of neuronal dysfunction, axonal degeneration, and myelination.


Asunto(s)
Tronco Encefálico/inmunología , Neuronas Motoras/inmunología , Nervio Frénico/inmunología , Médula Espinal/inmunología , Fiebre del Nilo Occidental/inmunología , Animales , Astrocitos/inmunología , Astrocitos/patología , Astrocitos/virología , Tronco Encefálico/patología , Tronco Encefálico/virología , Recuento de Células , Cricetulus , Electromiografía/métodos , Femenino , Humanos , Masculino , Ratones , Microglía/inmunología , Microglía/patología , Microglía/virología , Neuronas Motoras/patología , Neuronas Motoras/virología , Conducción Nerviosa , Infiltración Neutrófila , Nervio Frénico/patología , Nervio Frénico/virología , Médula Espinal/patología , Médula Espinal/virología , Linfocitos T/inmunología , Linfocitos T/patología , Linfocitos T/virología , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/patogenicidad , Virus del Nilo Occidental/fisiología
10.
Neurobiol Dis ; 94: 226-36, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27370226

RESUMEN

The concomitant expression of neuronal TAR DNA binding protein 43 (TDP-43) and human endogenous retrovirus-K (ERVK) is a hallmark of ALS. Since the involvement of TDP-43 in retrovirus replication remains controversial, we sought to evaluate whether TDP-43 exerts an effect on ERVK expression. In this study, TDP-43 bound the ERVK promoter in the context of inflammation or proteasome inhibition, with no effect on ERVK transcription. However, over-expression of ALS-associated aggregating forms of TDP-43, but not wild-type TDP-43, significantly enhanced ERVK viral protein accumulation. Human astrocytes and neurons further demonstrated cell-type specific differences in their ability to express and clear ERVK proteins during inflammation and proteasome inhibition. Astrocytes, but not neurons, were able to clear excess ERVK proteins through stress granule formation and autophagy. In vitro findings were validated in autopsy motor cortex tissue from patients with ALS and neuro-normal controls. We further confirmed marked enhancement of ERVK in cortical neurons of patients with ALS. Despite evidence of enhanced stress granule and autophagic response in ALS cortical neurons, these cells failed to clear excess ERVK protein accumulation. This highlights how multiple cellular pathways, in conjunction with disease-associated mutations, can converge to modulate the expression and clearance of viral gene products from genomic elements such as ERVK. In ALS, ERVK protein aggregation is a novel aspect of TDP-43 misregulation contributing towards the pathology of this neurodegenerative disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/metabolismo , Retrovirus Endógenos/metabolismo , Neuronas Motoras/virología , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/virología , Astrocitos/metabolismo , Astrocitos/virología , Autofagia/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas Motoras/patología , Mutación/genética , Proteínas Virales/metabolismo
11.
J Biol Chem ; 289(23): 16148-63, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24753246

RESUMEN

Rabies pseudotyped lentiviral vectors have great potential in gene therapy, not least because of their ability to transduce neurons following their distal axonal application. However, very little is known about the molecular processes that underlie their retrograde transport and cell transduction. Using multiple labeling techniques and confocal microscopy, we demonstrated that pseudotyping with rabies virus envelope glycoprotein (RV-G) enabled the axonal retrograde transport of two distinct subtypes of lentiviral vector in motor neuron cultures. Analysis of this process revealed that these vectors trafficked through Rab5-positive endosomes and accumulated within a non-acidic Rab7 compartment. RV-G pseudotyped vectors were co-transported with both the tetanus neurotoxin-binding fragment and the membrane proteins thought to mediate rabies virus endocytosis (neural cell adhesion molecule, nicotinic acetylcholine receptor, and p75 neurotrophin receptor), thus demonstrating that pseudotyping with RV-G targets lentiviral vectors for transport along the same pathway exploited by several toxins and viruses. Using motor neurons cultured in compartmentalized chambers, we demonstrated that axonal retrograde transport of these vectors was rapid and efficient; however, it was not able to transduce the targeted neurons efficiently, suggesting that impairment in processes occurring after arrival of the viral vector in the soma is responsible for the low transduction efficiency seen in vivo, which suggests a novel area for improvement of gene therapy vectors.


Asunto(s)
Transporte Axonal , Vectores Genéticos , Lentivirus/genética , Neuronas Motoras/metabolismo , Virus de la Rabia/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Endocitosis , Células HEK293 , Humanos , Neuronas Motoras/virología , Ratas , Proteínas del Envoltorio Viral/genética
12.
Gene Ther ; 21(5): 522-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24572783

RESUMEN

Systemic and intracerebrospinal fluid delivery of adeno-associated virus serotype 9 (AAV9) has been shown to achieve widespread gene delivery to the central nervous system (CNS). However, after systemic injection, the neurotropism of the vector has been reported to vary according to age at injection, with greater neuronal transduction in newborns and preferential glial cell tropism in adults. This difference has not yet been reported after cerebrospinal fluid (CSF) delivery. The present study analyzed both neuronal and glial cell transduction in the CNS of cats according to age of AAV9 CSF injection. In both newborns and young cats, administration of AAV9-GFP in the cisterna magna resulted in high levels of motor neurons (MNs) transduction from the cervical (84±5%) to the lumbar (99±1%) spinal cord, demonstrating that the remarkable tropism of AAV9 for MNs is not affected by age at CSF delivery. Surprisingly, numerous oligodendrocytes were also transduced in the brain and in the spinal cord white matter of young cats, but not of neonates, indicating that (i) age of CSF delivery influences the tropism of AAV9 for glial cells and (ii) AAV9 intracisternal delivery could be relevant for both the treatment of MN and demyelinating disorders.


Asunto(s)
Encéfalo/virología , Dependovirus/genética , Vectores Genéticos/genética , Neuronas Motoras/virología , Oligodendroglía/virología , Factores de Edad , Animales , Encéfalo/citología , Gatos , Líquido Cefalorraquídeo/virología , Terapia Genética , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/genética , Neuronas Motoras/citología , Neuroglía/citología , Neuronas/citología , Oligodendroglía/citología , Médula Espinal/citología , Médula Espinal/virología , Transducción Genética
13.
Emerg Microbes Infect ; 13(1): 2382235, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39017655

RESUMEN

Enterovirus A71 (EV-A71) causes Hand, Foot, and Mouth Disease and has been clinically associated with neurological complications. However, there is a lack of relevant models to elucidate the neuropathology of EV-A71 and its mechanism, as the current models mainly utilize animal models or immortalized cell lines. In this study, we established a human motor neuron model for EV-A71 infection. Single cell transcriptomics of a mixed neuronal population reveal higher viral RNA load in motor neurons, suggesting higher infectivity and replication of EV-A71 in motor neurons. The elevated RNA load in motor neurons correlates with the downregulation of ferritin-encoding genes. Subsequent analysis confirms that neurons infected with EV-A71 undergo ferroptosis, as evidenced by increased levels of labile Fe2+ and peroxidated lipids. Notably, the Fe2+ chelator Deferoxamine improves mitochondrial function and promotes survival of motor neurons by 40% after EV-A71 infection. These findings deepen understanding of the molecular pathogenesis of EV-A71 infection, providing insights which suggest that improving mitochondrial respiration and inhibition of ferroptosis can mitigate the impact of EV-A71 infection in the central nervous system.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Ferroptosis , Neuronas Motoras , Ferroptosis/efectos de los fármacos , Humanos , Enterovirus Humano A/fisiología , Enterovirus Humano A/genética , Enterovirus Humano A/efectos de los fármacos , Neuronas Motoras/virología , Neuronas Motoras/metabolismo , Infecciones por Enterovirus/virología , Infecciones por Enterovirus/metabolismo , Replicación Viral , Mitocondrias/metabolismo , Deferoxamina/farmacología , Carga Viral , Hierro/metabolismo , Ferritinas/metabolismo , Ferritinas/genética
14.
Neurobiol Dis ; 47(2): 174-83, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22521461

RESUMEN

Corticospinal motor neurons (CSMN) are the cortical component of motor neuron circuitry, which controls voluntary movement and degenerates in diseases such as amyotrophic lateral sclerosis, primary lateral sclerosis and hereditary spastic paraplegia. By using dual labeling combined with molecular marker analysis, we identified AAV2-2 mediated retrograde transduction as an effective approach to selectively target CSMN without affecting other neuron populations both in wild-type and hSOD1(G93A) transgenic ALS mice. This approach reveals very precise details of cytoarchitectural defects within vulnerable neurons in vivo. We report that CSMN vulnerability is marked by selective degeneration of apical dendrites especially in layer II/III of the hSOD1(G93A) mouse motor cortex, where cortical input to CSMN function is vastly modulated. While our findings confirm the presence of astrogliosis and microglia activation, they do not lend support to their direct role for the initiation of CSMN vulnerability. This study enables development of targeted gene replacement strategies to CSMN in the cerebral cortex, and reveals CSMN cortical modulation defects as a potential cause of neuronal vulnerability in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/virología , Dendritas/patología , Dependovirus/fisiología , Neuronas Motoras/patología , Tractos Piramidales/patología , Transducción de Señal/fisiología , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Dendritas/química , Dendritas/virología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/metabolismo , Neuronas Motoras/virología , Tractos Piramidales/metabolismo , Tractos Piramidales/virología
15.
Ann Neurol ; 69(1): 141-51, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21280084

RESUMEN

OBJECTIVE: Amyotrophic lateral sclerosis (ALS) is characterized by the progressive loss of motor neurons, of unknown etiology. Previous studies showed reverse transcriptase in serum of ALS patients at levels comparable to human immunodeficiency virus-infected patients; however, the source and significance of the retroviral elements is uncertain. METHODS: Expression of a human endogenous retrovirus (HERV-K) was determined in autopsy brain tissue of patients with ALS and compared to control populations by real-time polymerase chain reaction followed by sequencing of the amplified genes and confirmed by immunostaining. RESULTS: HERV-K pol transcripts were increased in patients with ALS compared to those with chronic systemic illness, but could not be detected in Parkinson disease or in the accidental death controls. Sequencing revealed several actively transcribed loci in the HML-2 and 3 subfamilies of HERV-K, with a specific pattern of expression including intact open reading frames and the transcription of a unique locus in ALS. The frequency of intact pol transcripts was highest in the motor cortex, and the reverse transcriptase protein was localized to cortical neurons of ALS patients. HERV-K expression strongly correlated with TDP-43, a multifunctional protein known to be dysregulated in ALS. INTERPRETATION: We have identified a specific pattern of HERV-K expression in ALS, which may potentially define the pathophysiology of ALS. Targeting of activated genome-encoded retroviral elements may open new prospects for the treatment of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/virología , Encéfalo/fisiopatología , Encéfalo/virología , Retrovirus Endógenos/aislamiento & purificación , Neuronas Motoras/virología , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/fisiopatología , Corteza Cerebral/fisiopatología , Corteza Cerebral/virología , Citogenética , Retrovirus Endógenos/genética , Femenino , Expresión Génica , Frecuencia de los Genes/genética , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/estadística & datos numéricos , Activación Transcripcional/fisiología , Proteínas Virales/genética
16.
PLoS Pathog ; 5(5): e1000442, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19461877

RESUMEN

Axonal transport is responsible for the movement of signals and cargo between nerve termini and cell bodies. Pathogens also exploit this pathway to enter and exit the central nervous system. In this study, we characterised the binding, endocytosis and axonal transport of an adenovirus (CAV-2) that preferentially infects neurons. Using biochemical, cell biology, genetic, ultrastructural and live-cell imaging approaches, we show that interaction with the neuronal membrane correlates with coxsackievirus and adenovirus receptor (CAR) surface expression, followed by endocytosis involving clathrin. In axons, long-range CAV-2 motility was bidirectional with a bias for retrograde transport in nonacidic Rab7-positive organelles. Unexpectedly, we found that CAR was associated with CAV-2 vesicles that also transported cargo as functionally distinct as tetanus toxin, neurotrophins, and their receptors. These results suggest that a single axonal transport carrier is capable of transporting functionally distinct cargoes that target different membrane compartments in the soma. We propose that CAV-2 transport is dictated by an innate trafficking of CAR, suggesting an unsuspected function for this adhesion protein during neuronal homeostasis.


Asunto(s)
Adenoviridae/metabolismo , Transporte Axonal , Axones/virología , Neuronas Motoras/virología , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Carbocianinas/metabolismo , Células Cultivadas , Vesículas Cubiertas por Clatrina/ultraestructura , Vesículas Cubiertas por Clatrina/virología , Invaginaciones Cubiertas de la Membrana Celular/ultraestructura , Invaginaciones Cubiertas de la Membrana Celular/virología , Receptor de Androstano Constitutivo , Endocitosis , Endosomas/metabolismo , Endosomas/virología , Colorantes Fluorescentes/metabolismo , Ganglios Espinales/metabolismo , Ganglios Espinales/ultraestructura , Ganglios Espinales/virología , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Neuronas Motoras/metabolismo , Neuronas Motoras/ultraestructura , Proteínas del Tejido Nervioso/metabolismo , Ratas , Nervio Ciático/metabolismo , Proteínas de Transporte Vesicular/metabolismo
17.
mBio ; 12(6): e0271221, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34781742

RESUMEN

Poliomyelitis-like illness is a common clinical manifestation of neurotropic viral infections. Functional loss and death of motor neurons often lead to reduced muscle tone and paralysis, causing persistent motor sequelae among disease survivors. Despite several reports demonstrating the molecular basis of encephalopathy, the pathogenesis behind virus-induced flaccid paralysis remained largely unknown. The present study for the first time aims to elucidate the mechanism responsible for limb paralysis by studying clinical isolates of Japanese encephalitis virus (JEV) and Chandipura virus (CHPV) responsible for causing acute flaccid paralysis (AFP) in vast regions of Southeast Asia and the Indian subcontinent. An experimental model for studying virus-induced AFP was generated by intraperitoneal injection of 10-day-old BALB/c mice. Progressive decline in motor performance of infected animals was observed, with paralysis being correlated with death of motor neurons (MNs). Furthermore, we demonstrated that upon infection, MNs undergo an extrinsic apoptotic pathway in a RIG-I-dependent fashion via transcription factors pIRF-3 and pIRF-7. Both gene-silencing experiments using specific RIG-I-short interfering RNA and in vivo morpholino abrogated cellular apoptosis, validating the important role of pattern recognition receptor (PRR) RIG-I in MN death. Hence, from our experimental observations, we hypothesize that host innate response plays a significant role in deterioration of motor functioning upon neurotropic virus infections. IMPORTANCE Neurotropic viral infections are an increasingly common cause of immediate or delayed neuropsychiatric sequelae, cognitive impairment, and movement disorders or, in severe cases, death. Given the highest reported disability-adjusted life years and mortality rate worldwide, a better understanding of molecular mechanisms for underlying clinical manifestations like AFP will help in development of more effective tools for therapeutic solutions.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central/metabolismo , Enfermedades Virales del Sistema Nervioso Central/fisiopatología , Proteína 58 DEAD Box/metabolismo , Virus de la Encefalitis Japonesa (Especie)/fisiología , Neuronas Motoras/citología , Mielitis/metabolismo , Mielitis/fisiopatología , Enfermedades Neuromusculares/metabolismo , Enfermedades Neuromusculares/fisiopatología , Vesiculovirus/fisiología , Animales , Muerte Celular , Enfermedades Virales del Sistema Nervioso Central/genética , Enfermedades Virales del Sistema Nervioso Central/virología , Proteína 58 DEAD Box/genética , Virus de la Encefalitis Japonesa (Especie)/genética , Femenino , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Masculino , Ratones , Actividad Motora , Neuronas Motoras/metabolismo , Neuronas Motoras/virología , Mielitis/genética , Mielitis/virología , Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/virología , Vesiculovirus/genética
18.
J Virol ; 83(10): 4995-5004, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19244317

RESUMEN

Poliovirus (PV), when injected intramuscularly into the calf, is incorporated into the sciatic nerve and causes an initial paralysis of the inoculated limb in transgenic (Tg) mice carrying the human PV receptor (hPVR/CD155) gene. We have previously demonstrated that a fast retrograde axonal transport process is required for PV dissemination through the sciatic nerves of hPVR-Tg mice and that intramuscularly inoculated PV causes paralytic disease in an hPVR-dependent manner. Here we showed that hPVR-independent axonal transport of PV was observed in hPVR-Tg and non-Tg mice, indicating that several different pathways for PV axonal transport exist in these mice. Using primary motor neurons (MNs) isolated from these mice or rats, we demonstrated that the axonal transport of PV requires several kinetically different motor machineries and that fast transport relies on a system involving cytoplasmic dynein. Unexpectedly, the hPVR-independent axonal transport of PV was not observed in cultured MNs. Thus, PV transport machineries in cultured MNs and in vivo differ in their hPVR requirements. These results suggest that the axonal trafficking of PV is carried out by several distinct pathways and that MNs in culture and in the sciatic nerve in situ are intrinsically different in the uptake and axonal transport of PV.


Asunto(s)
Transporte Axonal , Dineínas/metabolismo , Neuronas Motoras/virología , Poliovirus/fisiología , Receptores Virales/metabolismo , Animales , Células Cultivadas , Chlorocebus aethiops , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Poliomielitis/metabolismo , Poliomielitis/virología , Poliovirus/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Ciático/metabolismo , Nervio Ciático/virología , Células Vero
19.
J Neurovirol ; 16(1): 93-100, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20166837

RESUMEN

Human neurologic illness following infection with West Nile virus (WNV) may include meningitis, encephalitis, and acute flaccid paralysis (AFP). Most WNV-associated AFP is due to involvement of the spinal motor neurons producing an anterior (polio)myelitis. WNV poliomyelitis is typically characterized by acute and rapidly progressing limb weakness occurring early in the course of illness, which is followed by death or clinical plateauing with subsequent improvement to varying degrees. We describe four cases of WNV poliomyelitis in which the limb weakness was characterized by an atypical temporal pattern, including one case with onset several weeks after illness onset, and three cases developing relapsing or recurrent limb weakness following a period of clinical plateauing or improvement. Delayed onset or recurrent features may be due to persistence of viral infection or delayed neuroinvasion with delayed injury by excitotoxic or other mechanisms, by immune-mediated mechanisms, or a combination thereof. Further clinical and pathogenesis studies are needed to better understand the mechanisms for these phenomena. Clinicians should be aware of these clinical patterns in patients with WNV poliomyelitis.


Asunto(s)
Extremidades/fisiopatología , Debilidad Muscular/fisiopatología , Fiebre del Nilo Occidental/complicaciones , Virus del Nilo Occidental , Adulto , Anciano , Extremidades/patología , Extremidades/virología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas Motoras/patología , Neuronas Motoras/virología , Debilidad Muscular/etiología , Debilidad Muscular/patología , Conducción Nerviosa , Poliomielitis/etiología , Poliomielitis/fisiopatología , Recurrencia , Factores de Tiempo , Fiebre del Nilo Occidental/virología
20.
Reproduction ; 139(3): 645-53, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19965985

RESUMEN

In contrast to the uterus, the cervix is well innervated during pregnancy and the density of nerve fibers increases before birth. To assess neural connections between the cervix and the spinal cord, the cervix of pregnant mice was injected with the trans-synaptic retrograde neural tract tracer pseudorabies virus (PRV). After 5 days, the virus was present in nerve cells and fibers in specific areas of the sensory, autonomic, and motor subdivisions of the thoracolumbar spinal cord. In nonpregnant controls, the virus was predominantly distributed in laminae I-III in the dorsal gray sensory areas with the heaviest label in the substantia gelatinosa compared with the autonomic or motor areas. Labeled cells and processes were sparse in other regions, except for a prominent cluster in the intermediolateral column (lamina VII). Photomicrographs of spinal cord sections were digitized, and the total area with the virus was estimated. Compared with nonpregnant controls, the area with PRV was significantly decreased in all the spinal cord subdivisions in pregnant mice except in the intermediolateral column. However, areas with the virus were equivalent in mice injected with PRV at 4 days or 1 day before birth. These findings suggest that the predominant innervation of the murine cervix is from the sensory regions of the thoracolumbar spinal cord, and that these connections diminish with pregnancy. The results raise the possibility that the remaining connections from sensory and autonomic subdivisions, particularly the intermediolateral column, of the thoracolumbar spinal cord may be important for increased density of nerve fibers in the cervix as pregnancy nears term.


Asunto(s)
Cuello del Útero/inervación , Vías Nerviosas/fisiología , Técnicas de Trazados de Vías Neuroanatómicas , Preñez , Médula Espinal/fisiología , Animales , Recuento de Células , Cuello del Útero/citología , Cuello del Útero/virología , Femenino , Herpesvirus Suido 1/fisiología , Ratones , Ratones Endogámicos C3H , Neuronas Motoras/citología , Neuronas Motoras/fisiología , Neuronas Motoras/virología , Fibras Nerviosas/fisiología , Fibras Nerviosas/virología , Técnicas de Trazados de Vías Neuroanatómicas/métodos , Trazadores del Tracto Neuronal/farmacología , Neuronas Aferentes/citología , Neuronas Aferentes/fisiología , Neuronas Aferentes/virología , Embarazo , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA