Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
J Virol ; 97(3): e0000323, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36877049

RESUMEN

Pseudorabies virus (PRV) infection activates inflammatory responses to release robust proinflammatory cytokines, which are critical for controlling viral infection and clearance of PRV. However, the innate sensors and inflammasomes involved in the production and secretion of proinflammatory cytokines during PRV infection remain poorly studied. In this study, we report that the transcription and expression levels of some proinflammatory cytokines, including interleukin 1ß (IL-1ß), IL-6, and tumor necrosis factor alpha (TNF-α), are upregulated in primary peritoneal macrophages and in mice during PRV infection. Mechanistically, Toll-like receptor 2 (TLR2), TLR3, TLR4, and TLR5 were induced by the PRV infection to enhance the transcription levels of pro-IL-1ß, pro-IL-18, and gasdermin D (GSDMD). Additionally, we found that PRV infection and transfection of its genomic DNA triggered AIM2 inflammasome activation, apoptosis-related speckle-like protein (ASC) oligomerization, and caspase-1 activation to enhance the secretion of IL-1ß and IL-18, which was mainly dependent on GSDMD, but not GSDME, in vitro and in vivo. Taken together, our findings reveal that the activation of the TLR2-TLR3-TRL4-TLR5-NF-κB axis and AIM2 inflammasome, as well as GSDMD, is required for proinflammatory cytokine release, which resists the PRV replication and plays a critical role in host defense against PRV infection. Our findings provide novel clues to prevent and control PRV infection. IMPORTANCE PRV can infect several mammals, including pigs, other livestock, rodents, and wild animals, causing huge economic losses. As an emerging and reemerging infectious disease, the emergence of PRV virulent isolates and increasing human PRV infection cases indicate that PRV is still a high risk to public health. It has been reported that PRV infection leads to robust release of proinflammatory cytokines through activating inflammatory responses. However, the innate sensor that activates IL-1ß expression and the inflammasome involved in the maturation and secretion of proinflammatory cytokines during PRV infection remain poorly studied. In this study, our findings reveal that, in mice, activation of the TLR2-TLR3-TRL4-TLR5-NF-κB axis and AIM2 inflammasome, as well as GSDMD, is required for proinflammatory cytokine release during PRV infection, and it resists PRV replication and plays a critical role in host defense against PRV infection. Our findings provide novel clues to prevent and control PRV infection.


Asunto(s)
Herpesvirus Suido 1 , Inflamasomas , FN-kappa B , Animales , Humanos , Ratones , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Herpesvirus Suido 1/metabolismo , Inflamasomas/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Mamíferos , FN-kappa B/metabolismo , Porcinos , Receptor Toll-Like 2/genética , Receptor Toll-Like 3 , Receptor Toll-Like 5 , Transducción de Señal , Encefalitis Viral/metabolismo
2.
Vet Res ; 54(1): 25, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36918933

RESUMEN

Pseudorabies virus (PRV) causes viral encephalitis, a devastating disease with high mortality worldwide. Curcumin (CUR) can reduce inflammatory damage by altering the phenotype of microglia; however, whether and how these changes mediate resistance to PRV-induced encephalitis is still unclear. In this study, BV2 cells were infected with/without PRV for 24 h and further treated with/without CUR for 24 h. The results indicated that CUR promoted the polarization of PRV-infected BV2 cells from the M1 phenotype to the M2 phenotype and reversed PRV-induced mitochondrial dysfunction. Furthermore, M1 BV2 cell secretions induced signalling pathways leading to apoptosis in PC-12 neuronal cells, and this effect was abrogated by the secretions of M2 BV2 cells. RNA sequencing and bioinformatics analysis predicted that this phenotypic shift may be due to changes in energy metabolism. Furthermore, Western blot analysis showed that CUR inhibited the increase in AMP-activated protein kinase (AMPK) phosphorylation, glycolysis, and triacylglycerol synthesis and the reduction in oxidative phosphorylation induced by PRV infection. Moreover, the ATP levels in M2 BV2 cells were higher than those in M1 cells. Furthermore, CUR prevented the increase in mortality, elevated body temperature, slowed growth, nervous system excitation, brain tissue congestion, vascular cuffing, and other symptoms of PRV-induced encephalitis in vivo. Thus, this study demonstrated that CUR protected against PRV-induced viral encephalitis by switching the phenotype of BV2 cells, thereby protecting neurons from inflammatory injury, and this effect was mediated by improving mitochondrial function and the AMPK/NF-κB p65-energy metabolism-related pathway.


Asunto(s)
Curcumina , Encefalitis Viral , Encefalitis , Herpesvirus Suido 1 , Seudorrabia , Animales , Curcumina/efectos adversos , Curcumina/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/farmacología , Microglía/metabolismo , Encefalitis/inducido químicamente , Encefalitis/metabolismo , Encefalitis/veterinaria , Fenotipo , Encefalitis Viral/metabolismo , Encefalitis Viral/veterinaria
3.
J Neurovirol ; 28(1): 151-157, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35212942

RESUMEN

Primary human herpesvirus 6 (HHV-6) infection is sometimes accompanied by acute encephalopathy with reduced subcortical diffusion (AED) in immunocompetent children. We investigated exosomal microRNA (miRNA) expression profiles in cerebrospinal fluid (CSF) and sera of patients with HHV-6-associated AED (n = 5) and febrile seizure (FS) (n = 5) using high-throughput sequencing. A total of 176 and 663 miRNAs were identified in CSF and serum exosomes, respectively. Comparative analysis determined that some miRNAs (miR-381-3p, miR-155) were exclusively expressed in the CSF exosomes of AED but not of FS patients, suggesting their potential application as novel diagnostic biomarkers for AED.


Asunto(s)
Encefalitis Viral , Exosomas , Herpesvirus Humano 6 , MicroARNs , Infecciones por Roseolovirus , Niño , Encefalitis Viral/genética , Encefalitis Viral/metabolismo , Exosomas/genética , Exosomas/metabolismo , Herpesvirus Humano 6/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , MicroARNs/genética , Infecciones por Roseolovirus/genética
4.
Trends Neurosci ; 45(2): 158-170, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34906391

RESUMEN

Microglia are resident immune cells of the central nervous system (CNS) with multiple functions in health and disease. Their response during encephalitis depends on whether inflammation is triggered in a sterile or infectious manner, and in the latter case on the type of the infecting pathogen. Even though recent technological innovations advanced the understanding of the broad spectrum of microglia responses during viral encephalitis (VE), it is not entirely clear which microglia gene expression profiles are associated with antiviral and detrimental activities. Here, we review novel approaches to study microglia and the latest concepts of their function in VE. Improved understanding of microglial functions will be essential for the development of new therapeutic interventions for VE.


Asunto(s)
Encefalitis Viral , Microglía , Sistema Nervioso Central , Encefalitis Viral/metabolismo , Humanos , Inflamación/metabolismo , Microglía/metabolismo
5.
Front Immunol ; 12: 753683, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899705

RESUMEN

Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.


Asunto(s)
Efecto Citopatogénico Viral , Modelos Animales de Enfermedad , Encefalitis Viral/metabolismo , Parechovirus/patogenicidad , Infecciones por Picornaviridae/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Autofagia , Línea Celular Tumoral , Corteza Cerebral/virología , Chlorocebus aethiops , Citocinas/biosíntesis , Citocinas/genética , Encefalitis Viral/patología , Encefalitis Viral/virología , Glioblastoma/patología , Hipocampo/virología , Humanos , Inflamación , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Interferón Tipo I/farmacología , Interferones/biosíntesis , Interferones/genética , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Neuroblastoma/patología , Parechovirus/efectos de los fármacos , Parechovirus/fisiología , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Piroptosis , Células Vero , Replicación Viral/efectos de los fármacos , Interferón lambda
6.
Exp Neurol ; 346: 113845, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34454938

RESUMEN

Viral infection of the central nervous system (CNS) can cause lasting neurological decline in surviving patients and can present with symptoms resembling Parkinson's disease (PD). The mechanisms underlying postencephalitic parkinsonism remain unclear but are thought to involve increased innate inflammatory signaling in glial cells, resulting in persistent neuroinflammation. We therefore studied the role of glial cells in regulating neuropathology in postencephalitic parkinsonism by studying the involvement of astrocytes in loss of dopaminergic neurons and aggregation of α-synuclein protein following infection with western equine encephalitis virus (WEEV). Infections were conducted in both wildtype mice and in transgenic mice lacking NFκB inflammatory signaling in astrocytes. For 2 months following WEEV infection, we analyzed glial activation, neuronal loss and protein aggregation across multiple brain regions, including the substantia nigra pars compacta (SNpc). These data revealed that WEEV induces loss of SNpc dopaminergic neurons, persistent activation of microglia and astrocytes that precipitates widespread aggregation of α-synuclein in the brain of C57BL/6 mice. Microgliosis and macrophage infiltration occurred prior to activation of astrocytes and was followed by opsonization of ⍺-synuclein protein aggregates in the cortex, hippocampus and midbrain by the complement protein, C3. Astrocyte-specific NFκB knockout mice had reduced gliosis, α-synuclein aggregate formation and neuronal loss. These data suggest that astrocytes play a critical role in initiating PD-like pathology following encephalitic infection with WEEV through innate immune inflammatory pathways that damage dopaminergic neurons, possibly by hindering clearance of ⍺-synuclein aggregates. Inhibiting glial inflammatory responses could therefore represent a potential therapy strategy for viral parkinsonism.


Asunto(s)
Astrocitos/metabolismo , Neuronas Dopaminérgicas/metabolismo , Encefalitis Viral/metabolismo , Mediadores de Inflamación/metabolismo , Agregado de Proteínas/fisiología , alfa-Sinucleína/metabolismo , Animales , Astrocitos/inmunología , Neuronas Dopaminérgicas/inmunología , Virus de la Encefalitis Equina del Oeste/inmunología , Virus de la Encefalitis Equina del Oeste/metabolismo , Encefalitis Viral/inmunología , Femenino , Humanos , Mediadores de Inflamación/inmunología , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/fisiología
7.
Methods Mol Biol ; 2311: 109-130, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34033080

RESUMEN

The complexity of the central nervous system (CNS) is not recapitulated in cell culture models. Thin slicing and subsequent culture of CNS tissue has become a valued means to study neuronal and glial biology within the context of the physiologically relevant tissue milieu. Modern membrane-interface slice culturing methodology allows for straightforward access to both CNS tissue and feeding medium, enabling experimental manipulations and analyses that would otherwise be impossible in vivo. CNS slices can be successfully maintained in culture for up to several weeks for investigation of evolving pathology and long-term intervention in models of chronic neurologic disease.Herein, membrane-interface slice culture models for studying viral encephalitis and myelitis are detailed, with emphasis on the use of these models for investigation of pathogenesis and evaluation of novel treatment strategies. We describe techniques to (1) generate brain and spinal cord slices from rodent donors, (2) virally infect slices, (3) monitor viral replication, (4) assess virally induced injury/apoptosis, (5) characterize "CNS-specific" cytokine production, and, (6) treat slices with cytokines/pharmaceuticals. Although our focus is on CNS viral infection, we anticipate that the described methods can be adapted to address a wide range of investigations within the fields of neuropathology, neuroimmunology, and neuropharmacology.


Asunto(s)
Encéfalo/virología , Encefalitis Viral/virología , Mielitis/virología , Médula Espinal/virología , Animales , Animales Recién Nacidos , Antivirales/farmacología , Apoptosis , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Citocinas/metabolismo , Encefalitis Viral/tratamiento farmacológico , Encefalitis Viral/metabolismo , Encefalitis Viral/patología , Interacciones Huésped-Patógeno , Mediadores de Inflamación/metabolismo , Ratones , Mielitis/tratamiento farmacológico , Mielitis/metabolismo , Mielitis/patología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/patología , Técnicas de Cultivo de Tejidos , Replicación Viral
8.
Front Immunol ; 12: 667478, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025669

RESUMEN

Viral encephalitis is the most common cause of encephalitis. It is responsible for high morbidity rates, permanent neurological sequelae, and even high mortality rates. The host immune response plays a critical role in preventing or clearing invading pathogens, especially when effective antiviral treatment is lacking. However, due to blockade of the blood-brain barrier, it remains unclear how peripheral immune cells contribute to the fight against intracerebral viruses. Here, we report that peripheral injection of an antibody against human Tim-3, an immune checkpoint inhibitor widely expressed on immune cells, markedly attenuated vesicular stomatitis virus (VSV) encephalitis, marked by decreased mortality and improved neuroethology in mice. Peripheral injection of Tim-3 antibody enhanced the recruitment of immune cells to the brain, increased the expression of major histocompatibility complex-I (MHC-I) on macrophages, and as a result, promoted the activation of VSV-specific CD8+ T cells. Depletion of macrophages abolished the peripheral injection-mediated protection against VSV encephalitis. Notably, for the first time, we found a novel post-translational modification of MHC-I by Tim-3, wherein, by enhancing the expression of MARCH9, Tim-3 promoted the proteasome-dependent degradation of MHC-I via K48-linked ubiquitination in macrophages. These results provide insights into the immune response against intracranial infections; thus, manipulating the peripheral immune cells with Tim-3 antibody to fight viruses in the brain may have potential applications for combating viral encephalitis.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Células Presentadoras de Antígenos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encefalitis Viral/prevención & control , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Infecciones por Rhabdoviridae/prevención & control , Vesiculovirus/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/virología , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/virología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Encefalitis Viral/inmunología , Encefalitis Viral/metabolismo , Encefalitis Viral/virología , Células HEK293 , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inyecciones Intraperitoneales , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Células RAW 264.7 , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/metabolismo , Infecciones por Rhabdoviridae/virología , Ubiquitinación , Células Vero , Vesiculovirus/patogenicidad , Carga Viral
9.
Neuromolecular Med ; 23(1): 25-46, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33547562

RESUMEN

Neuro-inflammation accompanies numerous neurological disorders and conditions where it can be associated with a progressive neurodegenerative pathology. In a similar manner, alterations in sphingolipid metabolism often accompany or are causative features in degenerative neurological conditions. These include dementias, motor disorders, autoimmune conditions, inherited metabolic disorders, viral infection, traumatic brain and spinal cord injury, psychiatric conditions, and more. Sphingolipids are major regulators of cellular fate and function in addition to being important structural components of membranes. Their metabolism and signaling pathways can also be regulated by inflammatory mediators. Therefore, as certain sphingolipids exert distinct and opposing cellular roles, alterations in their metabolism can have major consequences. Recently, regulation of bioactive sphingolipids by neuro-inflammatory mediators has been shown to activate a neuronal NADPH oxidase 2 (NOX2) that can provoke damaging oxidation. Therefore, the sphingolipid-regulated neuronal NOX2 serves as a mechanistic link between neuro-inflammation and neurodegeneration. Moreover, therapeutics directed at sphingolipid metabolism or the sphingolipid-regulated NOX2 have the potential to alleviate neurodegeneration arising out of neuro-inflammation.


Asunto(s)
NADPH Oxidasa 2/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Esfingolípidos/fisiología , Complejo SIDA Demencia/metabolismo , Animales , Productos Biológicos/uso terapéutico , Encefalopatías Metabólicas Innatas/genética , Encefalopatías Metabólicas Innatas/metabolismo , Encefalopatías Metabólicas Innatas/terapia , Descubrimiento de Drogas , Encefalitis Viral/metabolismo , Activación Enzimática , Terapia de Reemplazo Enzimático , Humanos , Inflamación , Naftalenos/uso terapéutico , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/terapia , Neuronas/metabolismo , Oxidación-Reducción , Pirimidinonas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Infección por el Virus Zika/metabolismo
10.
Ann Clin Transl Neurol ; 8(3): 558-570, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33465303

RESUMEN

OBJECTIVE: Rasmussen's encephalitis (RE) is a rare and severe progressive epileptic syndrome with unknown etiology. Infection by viruses such as human cytomegalovirus (HCMV) has been hypothesized to be a potential trigger for RE. Interferon-induced transmembrane protein-3 (IFITM3) single-nucleotide polymorphism (SNP) rs12252 is associated with the severity of viral infection disease. This study aimed to address the possibility that HCMV infection and IFITM3 rs12252 might be associated with RE disease progression. METHODS: The expression of HCMV and IFITM3 was detected with immunohistochemical staining, in situ hybridization and immunofluorescence double staining. The genotype of IFITM3 rs12252 was detected using the Sanger sequencing method. A genetic association analysis was carried out for this SNP and HCMV antigen expression. The relationship between this SNP and the clinical characteristics of these patients was further analyzed. In in vitro study, HCMV replication in SH-SY5Y cells with overexpressed IFITM3 variant was detected by immunofluorescence and real-time RT-PCR. RESULTS: Elevated expression of HCMV and IFITM3 was observed in the brain tissue of RE patients. Moreover, the IFITM3 polymorphism rs12252-C was found to associate with HCMV high detection and rapid disease progression in RE patients with the IFITM3 rs12252-CC genotype. In vitro study showed the overexpressed IFITM3 variant was associated with HCMV high infection level. CONCLUSION: These results suggest that the IFITM3 rs12252-C is associated with the disease progression of RE patients via facilitating persistent HCMV infection in brain tissue and provides new insight into understanding the pathogenesis of RE.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Progresión de la Enfermedad , Encefalitis , Proteínas de la Membrana/genética , Proteínas de Unión al ARN/genética , Células Cultivadas , Citomegalovirus/aislamiento & purificación , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Encefalitis/genética , Encefalitis/metabolismo , Encefalitis/virología , Encefalitis Viral/genética , Encefalitis Viral/metabolismo , Encefalitis Viral/virología , Genotipo , Humanos , Polimorfismo de Nucleótido Simple
11.
Pathog Dis ; 79(1)2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33238302

RESUMEN

BACKGROUND: biomarkers can be helpful in identifying patients who may profit by explicit treatments or evaluating the reaction to the treatment of specific disease. Finding unique biomarkers in the process of disease could help clinicians in identifying serious disease in the early stage, so as to improve prognosis. OBJECTIVE: these investigations, nonetheless, have made constrained progress. Numerous infections are known to cause intense viral encephalitis (VE) in people which can cause a variable level of meningeal just as parenchymal aggravation. Initial clinical manifestations in most encephalitis are nonspecific, resembling a viral-like illness. However, with disease progression, symptoms can become quite severe and fatal, including prominent cranial hypertension, cognitive problems, cerebral hernia and respiratory failure. Forwards: the clinical and research center discoveries in huge numbers of those viral issues are to a great extent comparable and in this way increasingly explicit biomarkers for indicative and prognostic intentions are justified. These biomarkers are progressively significant in the acknowledgment and treatment of the viral central nervous system (CNS) issue. CONCLUSION: Clinical manifestations have been the indicative approaches for analysis of viral encephalitis. Lots of studies have been endeavored to distinguish progressively objective laboratory-based quantitative CSF biomarkers for VE.


Asunto(s)
Antivirales/uso terapéutico , Encefalitis Viral/tratamiento farmacológico , Encefalitis Viral/metabolismo , Encefalitis Viral/virología , Vacunas/uso terapéutico , Biomarcadores , Infecciones por Coronaviridae/virología , Encefalitis Japonesa/virología , Infecciones por VIH/virología , Herpes Simple/virología , Humanos , Gripe Humana/virología , Pronóstico , Fiebre del Nilo Occidental/virología , Infección por el Virus Zika/virología
12.
J Stroke Cerebrovasc Dis ; 29(8): 104941, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32689643

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global health threat. Some COVID-19 patients have exhibited widespread neurological manifestations including stroke. Acute ischemic stroke, intracerebral hemorrhage, and cerebral venous sinus thrombosis have been reported in patients with COVID-19. COVID-19-associated coagulopathy is increasingly recognized as a result of acute infection and is likely caused by inflammation, including inflammatory cytokine storm. Recent studies suggest that axonal transport of SARS-CoV-2 to the brain can occur via the cribriform plate adjacent to the olfactory bulb that may lead to symptomatic anosmia. The internalization of SARS-CoV-2 is mediated by the binding of the spike glycoprotein of the virus to the angiotensin-converting enzyme 2 (ACE2) on cellular membranes. ACE2 is expressed in several tissues including lung alveolar cells, gastrointestinal tissue, and brain. The aim of this review is to provide insights into the clinical manifestations and pathophysiological mechanisms of stroke in COVID-19 patients. SARS-CoV-2 can down-regulate ACE2 and, in turn, overactivate the classical renin-angiotensin system (RAS) axis and decrease the activation of the alternative RAS pathway in the brain. The consequent imbalance in vasodilation, neuroinflammation, oxidative stress, and thrombotic response may contribute to the pathophysiology of stroke during SARS-CoV-2 infection.


Asunto(s)
Betacoronavirus/patogenicidad , Encéfalo/fisiopatología , Infecciones por Coronavirus/fisiopatología , Encefalitis Viral/fisiopatología , Neumonía Viral/fisiopatología , Accidente Cerebrovascular/fisiopatología , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/metabolismo , Coagulación Sanguínea , Encéfalo/metabolismo , Encéfalo/virología , COVID-19 , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Encefalitis Viral/epidemiología , Encefalitis Viral/metabolismo , Encefalitis Viral/virología , Interacciones Microbiota-Huesped , Humanos , Mediadores de Inflamación/metabolismo , Estrés Oxidativo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/epidemiología , Neumonía Viral/metabolismo , Neumonía Viral/virología , Sistema Renina-Angiotensina , SARS-CoV-2 , Transducción de Señal , Glicoproteína de la Espiga del Coronavirus/metabolismo , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/virología , Vasodilatación , Virulencia
13.
PLoS One ; 15(6): e0232381, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32584818

RESUMEN

Alphaviruses such as Venezuelan equine encephalitis virus (VEEV) and Eastern equine encephalitis virus (EEEV) are arboviruses that can cause severe zoonotic disease in humans. Both VEEV and EEEV are highly infectious when aerosolized and can be used as biological weapons. Vaccines and therapeutics are urgently needed, but efficacy determination requires animal models. The cynomolgus macaque (Macaca fascicularis) provides a relevant model of human disease, but questions remain whether vaccines or therapeutics can mitigate CNS infection or disease in this model. The documentation of alphavirus encephalitis in animals relies on traditional physiological biomarkers and behavioral/neurological observations by veterinary staff; quantitative measurements such as electroencephalography (EEG) and intracranial pressure (ICP) can recapitulate underlying encephalitic processes. We detail a telemetry implantation method suitable for continuous monitoring of both EEG and ICP in awake macaques, as well as methods for collection and analysis of such data. We sought to evaluate whether changes in EEG/ICP suggestive of CNS penetration by virus would be seen after aerosol exposure of naïve macaques to VEEV IC INH9813 or EEEV V105 strains compared to mock-infection in a cohort of twelve adult cynomolgus macaques. Data collection ran continuously from at least four days preceding aerosol exposure and up to 50 days thereafter. EEG signals were processed into frequency spectrum bands (delta: [0.4 - 4Hz); theta: [4 - 8Hz); alpha: [8-12Hz); beta: [12-30] Hz) and assessed for viral encephalitis-associated changes against robust background circadian variation while ICP data was assessed for signal fidelity, circadian variability, and for meaningful differences during encephalitis. Results indicated differences in delta, alpha, and beta band magnitude in infected macaques, disrupted circadian rhythm, and proportional increases in ICP in response to alphavirus infection. This novel enhancement of the cynomolgus macaque model offers utility for timely determination of onset, severity, and resolution of encephalitic disease and for the evaluation of vaccine and therapeutic candidates.


Asunto(s)
Infecciones por Alphavirus/patología , Encéfalo/fisiología , Encefalitis Viral/patología , Presión Intracraneal/fisiología , Alphavirus/aislamiento & purificación , Alphavirus/patogenicidad , Infecciones por Alphavirus/metabolismo , Animales , Biomarcadores/metabolismo , Ritmo Circadiano , Modelos Animales de Enfermedad , Electroencefalografía/métodos , Encefalitis Viral/metabolismo , Femenino , Macaca , Masculino , Índice de Severidad de la Enfermedad , Telemetría
14.
J Neuroimmunol ; 344: 577241, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32334204

RESUMEN

BACKGROUND: The cerebral innate immune system has a critical role in control processes of viral replication in the brain after primary infactivo and immunologic disregulation and inflammation has been reported as a primary determinant of pathogenesis and prognosis of subsequent HSV-1 related encephalitis (HSE). Interaction linking LTR3-activated DCs is also represented by the killer Ig-like receptor (KIR) + pathways on NK cells. Only a few studies analyzed the role of of MMP-9 activity regulating genetic polymorphism on clinical outcome of viral infections. Susceptibility to symptomatic encephalitis depends on SNC viral invasion and BBB disruption. We hypothesize that certain KIR genes and MMP allele may help to characterize a risk profile of developing an acute encephalitis due to HSV 1. AIM OF THE STUDY: Analyze the frequency of KIR genes and the C(-1562)T MMP-9 allels in subjects with HSV-1 encephalitis and to analyze their interaction with regard of the risk of occurrence of a symptomatic encephalitis. MATERIALS AND METHODS: Between November 2014 and January 2019, all consecutive patients with symptomatic acute encephalitis were recruited from three wards (Internal Medicine, Neurology, and Infectious Diseases) of "P. Giaccone" University Hospital, Palermo. RESULTS: Patients with acute viral encephalitis in comparison to controls showed a higher frequency AA KIR haplotype, HLA-C2 and of HLA-A-Bw4 alleles. With regard of HLA allele frequency patients with acute viral encephalitis In comparison to controls also showed a higher frequency of HLA-C2 and of HLA-A-Bw4 alleles. With regard of MMP-9 alleles, subjects with acute viral encephalitis were more likely to have the TT genotype. The multiple logistic regression analysis considering variables predictive of the occurrence of acute viral encephalitis showed the detrimental effect of AA KIR, HLAC1, HLA-A-BW4 and HLA-B-BW4T and of TT aplotype of MMP-9 genotype. CONCLUSIONS: Our study shows that in immunocompetent adult subjects there is an association between some KIR genes, MMP-9 alleles and HLA-ligand alleles and susceptibility to develop a symptomatic acute viral encephalitis. Definition of the genetic and immunological background of acute viral encephalitis can play a key role to determine personalized medicine.


Asunto(s)
Encefalitis Viral/genética , Antígenos HLA/genética , Herpes Simple/genética , Herpesvirus Humano 1/genética , Metaloproteinasa 9 de la Matriz/genética , Receptores KIR/genética , Anciano , Estudios de Cohortes , Encefalitis Viral/diagnóstico , Encefalitis Viral/metabolismo , Femenino , Antígenos HLA/metabolismo , Herpes Simple/diagnóstico , Herpes Simple/metabolismo , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteasas/genética , Metaloproteasas/metabolismo , Persona de Mediana Edad , Receptores KIR/metabolismo
15.
Int J Mol Sci ; 21(2)2020 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-31941046

RESUMEN

The etiology of viral encephalitis in cattle often remains unresolved, posing a potential risk for animal and human health. In metagenomics studies of cattle with bovine non-suppurative encephalitis, parainfluenza virus 5 (PIV5) was identified in three brain samples. Interestingly, in two of these animals, bovine herpesvirus 6 and bovine astrovirus CH13 were additionally found. We investigated the role of PIV5 in bovine non-suppurative encephalitis and further characterized the three cases. With traditional sequencing methods, we completed the three PIV5 genomes, which were compared to one another. However, in comparison to already described PIV5 strains, unique features were revealed, like an 81 nucleotide longer open reading frame encoding the small hydrophobic (SH) protein. With in situ techniques, we demonstrated PIV5 antigen and RNA in one animal and found a broad cell tropism of PIV5 in the brain. Comparative quantitative analyses revealed a high viral load of PIV5 in the in situ positive animal and therefore, we propose that PIV5 was probably the cause of the disease. With this study, we clearly show that PIV5 is capable of naturally infecting different brain cell types in cattle in vivo and therefore it is a probable cause of encephalitis and neurological disease in cattle.


Asunto(s)
Antígenos Virales , Enfermedades de los Bovinos , Encefalitis Viral , Genoma Viral , Virus de la Parainfluenza 5 , ARN Viral , Infecciones por Rubulavirus , Animales , Antígenos Virales/genética , Antígenos Virales/metabolismo , Bovinos , Enfermedades de los Bovinos/genética , Enfermedades de los Bovinos/metabolismo , Enfermedades de los Bovinos/virología , Encefalitis Viral/genética , Encefalitis Viral/metabolismo , Encefalitis Viral/virología , Virus de la Parainfluenza 5/genética , Virus de la Parainfluenza 5/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Infecciones por Rubulavirus/genética , Infecciones por Rubulavirus/metabolismo
16.
mBio ; 10(4)2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31387911

RESUMEN

Blood-brain barrier (BBB) breakdown is a hallmark of many diseases of the central nervous system (CNS). Loss of BBB integrity in CNS diseases such as viral encephalitis results in the loss of nutrient/oxygen delivery, rapid infiltration of immune cells, and brain swelling that can exacerbate neuronal injury. Despite this, the cellular and molecular mechanisms that underlie BBB breakdown in viral encephalitis are incompletely understood. We undertook a comprehensive analysis of the cellular and molecular signaling events that induce BBB breakdown in an experimental model of virus-induced encephalitis in which neonatal mice are infected with reovirus (serotype 3 strain Abney). We show that BBB leakage during reovirus infection correlates with morphological changes in the vasculature, reductions in pericytes (BBB supporting cells), and disorganization of vascular junctions. Pathway analysis on RNA sequencing from brain endothelial cells identified the activation of interferon (IFN) signaling within the brain vasculature following reovirus infection. Our in vitro and in vivo studies show that type II IFN mediated by IFN-γ, a well known antiviral signal, is a major contributor to BBB leakage during reovirus infection. We show that IFN-γ reduces barrier properties in cultured brain endothelial cells through Rho kinase (ROCK)-mediated cytoskeletal contractions, resulting in junctional disorganization and cell-cell separations. In vivo neutralization of IFN-γ during reovirus infection significantly improved BBB integrity, pericyte coverage, attenuated vascular ROCK activity, and junctional disorganization. Our work supports a model in which IFN-γ acts directly on the brain endothelium to induce BBB breakdown through a mechanism involving ROCK-induced junctional disorganization.IMPORTANCE In an experimental viral encephalitis mouse model in which mice are infected with reovirus, we show that IFN-γ induces blood-brain barrier leakage. We show that IFN-γ promotes Rho kinase activity, resulting in actin cytoskeletal contractions in the brain endothelium that lead to vascular junctional disorganization and cell-cell separations. These studies now provide insight into a previously unknown mechanism for how blood-brain barrier breakdown occurs in viral encephalitis and implicates IFN-γ-Rho kinase activity as major contributor to this phenomenon. By identifying this mechanism of blood-brain barrier breakdown, we now provide potential therapeutic targets in treating patients with viral causes of encephalitis with the hope of limiting damage to the central nervous system.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalitis Viral/metabolismo , Interferón gamma/metabolismo , Orthoreovirus Mamífero 3/fisiología , Infecciones por Reoviridae/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Barrera Hematoencefálica/virología , Encéfalo/enzimología , Encéfalo/metabolismo , Encéfalo/virología , Modelos Animales de Enfermedad , Encefalitis Viral/genética , Encefalitis Viral/virología , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Células Endoteliales/virología , Femenino , Humanos , Interferón gamma/genética , Masculino , Ratones , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/virología , Quinasas Asociadas a rho/genética
17.
Int J Mol Sci ; 20(13)2019 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-31262031

RESUMEN

(1) Background: Canine distemper virus (CDV)-induced demyelinating leukoencephalitis (CDV-DL) in dogs and Theiler's murine encephalomyelitis (TME) virus (TMEV)-induced demyelinating leukomyelitis (TMEV-DL) are virus-induced demyelinating conditions mimicking Multiple Sclerosis (MS). Reactive oxygen species (ROS) can induce the degradation of lipids and nucleic acids to characteristic metabolites such as oxidized lipids, malondialdehyde, and 8-hydroxyguanosine. The hypothesis of this study is that ROS are key effector molecules in the pathogenesis of myelin membrane breakdown in CDV-DL and TMEV-DL. (2) Methods: ROS metabolites and antioxidative enzymes were assessed using immunofluorescence in cerebellar lesions of naturally CDV-infected dogs and spinal cord tissue of TMEV-infected mice. The transcription of selected genes involved in ROS generation and detoxification was analyzed using gene-expression microarrays in CDV-DL and TMEV-DL. (3) Results: Immunofluorescence revealed increased amounts of oxidized lipids, malondialdehyde, and 8-hydroxyguanosine in CDV-DL while TMEV-infected mice did not reveal marked changes. In contrast, microarray-analysis showed an upregulated gene expression associated with ROS generation in both diseases. (4) Conclusion: In summary, the present study demonstrates a similar upregulation of gene-expression of ROS generation in CDV-DL and TMEV-DL. However, immunofluorescence revealed increased accumulation of ROS metabolites exclusively in CDV-DL. These results suggest differences in the pathogenesis of demyelination in these two animal models.


Asunto(s)
Moquillo/metabolismo , Encefalitis Viral/metabolismo , Vaina de Mielina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Catalasa/metabolismo , Moquillo/patología , Perros , Encefalitis Viral/patología , Encefalitis Viral/virología , Femenino , Masculino , Ratones , Vaina de Mielina/patología , Médula Espinal/metabolismo , Médula Espinal/patología , Superóxido Dismutasa/metabolismo , Theilovirus/patogenicidad
18.
Neurology ; 92(21): e2406-e2420, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31028126

RESUMEN

OBJECTIVE: To characterize the full spectrum, relative frequency, and prognosis of the neurologic manifestations in Zika virus (ZIKV) postnatal infection. METHODS: We conducted an observational study in consecutive ZIKV-infected patients presenting with neurologic manifestations during the French West Indies 2016 outbreak. RESULTS: Eighty-seven patients, including 6 children, were enrolled. Ninety-five percent of all cases required hospitalization. Guillain-Barré syndrome was the most frequent manifestation (46.0%) followed by encephalitis or encephalomyelitis (20.7%), isolated single or multiple cranial nerve palsies (9.2%), other peripheral manifestations (6.9%), and stroke (1.1%). Fourteen patients (16.1%), including one child, developed a mixed disorder involving both the central and peripheral nervous system. Mechanical ventilation was required in 21 cases, all of whom had ZIKV RNA in at least one biological fluid. Two adult patients died due to neuroZika. Clinical follow-up (median 14 months; interquartile range, 13-17 months) was available for 76 patients. Residual disability (modified Rankin Scale score ≥2) was identified in 19 (25.0%) patients; in 6 cases (7.9%), disability was severe (modified Rankin Scale score ≥4). Among patients with ZIKV RNA detected in one biological fluid, the risk of residual disability or death was higher (odds ratio 9.19; confidence interval 1.12-75.22; p = 0.039). CONCLUSIONS: NeuroZika spectrum represents a heterogeneous group of clinical neurologic manifestations. During an outbreak, clinicians should consider neuroZika in patients presenting with cranial nerve palsies and a mixed neurologic disorder. Long-term sequelae are frequent in NeuroZika. ZIKV reverse-transcription PCR status at admission can inform prognosis and should therefore be taken into consideration in the management of hospitalized patients.


Asunto(s)
Enfermedades de los Nervios Craneales/terapia , Encefalitis Viral/terapia , Encefalomielitis/terapia , Síndrome de Guillain-Barré/fisiopatología , Infección por el Virus Zika/terapia , Adolescente , Adulto , Anciano , Niño , Preescolar , Enfermedades de los Nervios Craneales/metabolismo , Enfermedades de los Nervios Craneales/fisiopatología , Encefalitis Viral/metabolismo , Encefalitis Viral/fisiopatología , Encefalomielitis/metabolismo , Encefalomielitis/fisiopatología , Femenino , Hospitalización , Humanos , Lactante , Masculino , Persona de Mediana Edad , Pronóstico , ARN Viral/sangre , ARN Viral/líquido cefalorraquídeo , ARN Viral/orina , Respiración Artificial , Resultado del Tratamiento , Indias Occidentales , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/fisiopatología
19.
J Virol ; 93(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30728259

RESUMEN

A clinical isolate of measles virus (MeV) bearing a single amino acid alteration in the viral fusion protein (F; L454W) was previously identified in two patients with lethal sequelae of MeV central nervous system (CNS) infection. The mutation dysregulated the viral fusion machinery so that the mutated F protein mediated cell fusion in the absence of known MeV cellular receptors. While this virus could feasibly have arisen via intrahost evolution of the wild-type (wt) virus, it was recently shown that the same mutation emerged under the selective pressure of small-molecule antiviral treatment. Under these conditions, a potentially neuropathogenic variant emerged outside the CNS. While CNS adaptation of MeV was thought to generate viruses that are less fit for interhost spread, we show that two animal models can be readily infected with CNS-adapted MeV via the respiratory route. Despite bearing a fusion protein that is less stable at 37°C than the wt MeV F, this virus infects and replicates in cotton rat lung tissue more efficiently than the wt virus and is lethal in a suckling mouse model of MeV encephalitis even with a lower inoculum. Thus, either during lethal MeV CNS infection or during antiviral treatment in vitro, neuropathogenic MeV can emerge, can infect new hosts via the respiratory route, and is more pathogenic (at least in these animal models) than wt MeV.IMPORTANCE Measles virus (MeV) infection can be severe in immunocompromised individuals and lead to complications, including measles inclusion body encephalitis (MIBE). In some cases, MeV persistence and subacute sclerosing panencephalitis (SSPE) occur even in the face of an intact immune response. While they are relatively rare complications of MeV infection, MIBE and SSPE are lethal. This work addresses the hypothesis that despite a dysregulated viral fusion complex, central nervous system (CNS)-adapted measles virus can spread outside the CNS within an infected host.


Asunto(s)
Sistema Nervioso Central/virología , Encefalitis Viral , Cuerpos de Inclusión Viral , Pulmón/virología , Virus del Sarampión/fisiología , Sarampión , Mutación Missense , Proteínas Virales de Fusión , Replicación Viral , Sustitución de Aminoácidos , Animales , Sistema Nervioso Central/metabolismo , Chlorocebus aethiops , Modelos Animales de Enfermedad , Encefalitis Viral/genética , Encefalitis Viral/metabolismo , Encefalitis Viral/transmisión , Humanos , Cuerpos de Inclusión Viral/genética , Cuerpos de Inclusión Viral/metabolismo , Pulmón/metabolismo , Sarampión/metabolismo , Sarampión/transmisión , Ratones , Ratones Transgénicos , Sigmodontinae , Células Vero , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo
20.
Neuron ; 99(1): 56-63.e3, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-30001512

RESUMEN

Amyloid-ß peptide (Aß) fibrilization and deposition as ß-amyloid are hallmarks of Alzheimer's disease (AD) pathology. We recently reported Aß is an innate immune protein that protects against fungal and bacterial infections. Fibrilization pathways mediate Aß antimicrobial activities. Thus, infection can seed and dramatically accelerate ß-amyloid deposition. Here, we show Aß oligomers bind herpesvirus surface glycoproteins, accelerating ß-amyloid deposition and leading to protective viral entrapment activity in 5XFAD mouse and 3D human neural cell culture infection models against neurotropic herpes simplex virus 1 (HSV1) and human herpesvirus 6A and B. Herpesviridae are linked to AD, but it has been unclear how viruses may induce ß-amyloidosis in brain. These data support the notion that Aß might play a protective role in CNS innate immunity, and suggest an AD etiological mechanism in which herpesviridae infection may directly promote Aß amyloidosis.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Amiloidosis/metabolismo , Encéfalo/metabolismo , Encefalitis Viral/metabolismo , Herpesviridae , Enfermedad de Alzheimer/virología , Amiloidosis/virología , Animales , Encéfalo/virología , Células Cultivadas , Modelos Animales de Enfermedad , Encefalitis por Herpes Simple/metabolismo , Encefalitis por Herpes Simple/virología , Encefalitis Viral/virología , Herpesvirus Humano 1 , Herpesvirus Humano 6 , Humanos , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Neuronas , Placa Amiloide/metabolismo , Infecciones por Roseolovirus/metabolismo , Infecciones por Roseolovirus/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA