Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.596
Filtrar
1.
ACS Chem Neurosci ; 15(8): 1712-1727, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38581382

RESUMEN

Short-chain fatty acids (SCFAs) are gut microbial metabolic derivatives produced during the fermentation of ingested complex carbohydrates. SCFAs have been widely regarded to have a potent anti-inflammatory and neuro-protective role and have implications in several disease conditions, such as, inflammatory bowel disease, type-2 diabetes, and neurodegenerative disorders. Japanese encephalitis virus (JEV), a neurotropic flavivirus, is associated with life threatening neuro-inflammation and neurological sequelae in infected hosts. In this study, we hypothesize that SCFAs have potential in mitigating JEV pathogenesis. Postnatal day 10 BALB/c mice were intraperitoneally injected with either a SCFA mixture (acetate, propionate, and butyrate) or PBS for a period of 7 days, followed by JEV infection. All mice were observed for onset and progression of symptoms. The brain tissue was collected upon reaching terminal illness for further analysis. SCFA-supplemented JEV-infected mice (SCFA + JEV) showed a delayed onset of symptoms, lower hindlimb clasping score, and decreased weight loss and increased survival by 3 days (p < 0.0001) upon infection as opposed to the PBS-treated JEV-infected animals (JEV). Significant downregulation of inflammatory cytokines TNF-α, MCP-1, IL-6, and IFN-Υ in the SCFA + JEV group relative to the JEV-infected control group was observed. Inflammatory mediators, phospho-NF-kB (P-NF-kB) and iba1, showed 2.08 ± 0.1 and 3.132 ± 0.43-fold upregulation in JEV versus 1.19 ± 0.11 and 1.31 ± 0.11-fold in the SCFA + JEV group, respectively. Tissue section analysis exhibited reduced glial activation (JEV group─42 ± 2.15 microglia/ROI; SCFA + JEV group─27.07 ± 1.8 microglia/ROI) in animals that received SCFA supplementation prior to infection as seen from the astrocytic and microglial morphometric analysis. Caspase-3 immunoblotting showed 4.08 ± 1.3-fold upregulation in JEV as compared to 1.03 ± 0.14-fold in the SCFA + JEV group and TUNEL assay showed a reduced cellular death post-JEV infection (JEV-6.4 ± 1.5 cells/ROI and SCFA + JEV-3.7 ± 0.73 cells/ROI). Our study critically contributes to the increasing evidence in support of SCFAs as an anti-inflammatory and neuro-protective agent, we further expand its scope as a potential supplementary intervention in JEV-mediated neuroinflammation.


Asunto(s)
Encefalitis Japonesa , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Enfermedades Neuroinflamatorias , Microbioma Gastrointestinal/fisiología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/microbiología , Microglía/efectos de los fármacos , Microglía/inmunología , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/microbiología , Encefalitis Japonesa/prevención & control , Encefalitis Japonesa/virología , Ácidos Grasos Volátiles/farmacología , Ácidos Grasos Volátiles/uso terapéutico , Virus de la Encefalitis Japonesa (Subgrupo)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Subgrupo)/inmunología , Virus de la Encefalitis Japonesa (Subgrupo)/patogenicidad , Análisis de Supervivencia , Quimiocinas/inmunología , Quimiocinas/metabolismo , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/metabolismo , Síndrome de Liberación de Citoquinas/prevención & control , Humanos , Femenino , Animales , Ratones , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/virología , Carga Viral/efectos de los fármacos , Factores de Tiempo
2.
Actual. Sida Infectol. (En linea) ; 32(114): 63-78, 20240000. fig, graf
Artículo en Español | LILACS, BINACIS | ID: biblio-1552316

RESUMEN

La encefalitis equina del oeste (WEEV, por su sigla en inglés, Western Equine Encephalitis) es una enfermedad reemergente en Argentina a partir del año 2023. La co-municación inicial fue en 1933, las últimas epizootias ocurrieron en 1983 y el último caso humano se registró en 1996. Se revisan las características del agente causal, la ecología con especial referencia a los vectores iden-tificados en el país, su competencia en la transmisión y el ciclo así como los factores de riesgo para adquirir la enfermedad. La situación epidemiológica en equinos y humanos desde noviembre 2023 hasta marzo 2024 es analizada. Se describen las formas clínicas de presen-tación de la enfermedad humana, las posibilidades evo-lutivas, los datos disponibles en los casos confirmados y el tratamiento. La metodología y algoritmo empleados para el diagnóstico etiológico en el Centro Nacional de Referencia son detallados. Las estrategias para la pre-vención y el control se basan en la vacunación de los equinos, el saneamiento ambiental y el control del foco ante la presentación de la enfermedad animal (vigilancia epidemiológica activa)


Western equine encephalitis (WEE) is a re-emerging dis-ease in Argentina starting in 2023. Since the initial notifi-cation in 1933, the last epizootics occurred in 1983, and the last human case was recorded in 1996.The charac-teristics of the causative agent, the ecology with special reference to vectors identified in the country, their compe-tence in transmission, and the cycle as well as the risks factors for acquiring the disease, are reviewed.The epidemiological situation in horses and humans from November 2023 to March 2024 is analyzed. The clinical presentation of the human disease, its evolutionary po-tential, available data in confirmed cases, and the treat-ment are described.The methodology and algorithm used for the etiological diagnosis at the National Reference Center are detailed. Strategies for prevention and control are based on vaccination of horses, environmental sani-tation and outbreak control in the presence of the animal disease (active epidemiological surveillance)


Asunto(s)
Humanos , Animales , Masculino , Femenino , Saneamiento/legislación & jurisprudencia , Factores de Riesgo , Encefalomielitis Equina del Oeste/epidemiología , Virus de la Encefalitis Equina del Oeste/inmunología , Monitoreo Epidemiológico/veterinaria
3.
PLoS One ; 19(3): e0297143, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38427645

RESUMEN

The sequential pathology of Kyasanur forest disease (KFD) in mouse brain was assessed in this study. Kyasanur forest disease virus (KFDV) strain P9605 used in this study was confirmed by real-time reverse transcriptase-polymerase chain reaction targeting the NS5 gene. Mouse Lethal Dose 50 (MLD50) of the virus was determined by in-vivo mice inoculation test. One MLD50 of the KFDV was inoculated intra-cerebrally into 36 mice aged 2-3 weeks. Another group of 36 age-matched mice that served as control group were inoculated with plain media. Six mice each from infected and control groups were euthanized every 24 hrs intervals for six days. Brain tissues were collected in 10% NBF. The collected brain tissues were processed and subjected to histopathological studies by Hematoxylin and Eosin staining. Grossly, the infected mice showed symptoms of dullness, hunched back appearance, weakness, sluggish movements with indication of hind quarter paralysis on day four post-infection. These symptoms got aggravated with complete paralysis of the hind quarters, inability to move and death on 5th and 6th day post-infection. Microscopically, the brain showed apoptosis of neurons, perivascular cuffing, gliosis, congestion, neuropil vacuolation, meningitis, degeneration, and necrotic neurons. The real-time RT-PCR on hippocampus of the KFDV-infected mouse brain showed three-fold higher expression levels of Caspase 3, a crucial mediator of apoptosis. The cerebral cortex, cerebellum and hippocampus that control the motor neuron activities and muscle tone were primarily affected, possibly correlating with the gross symptoms of hind quarter paralysis, ataxia, and other motor neuron dysfunctions noticed. Taken together, these findings reveal that KFDV induces apoptosis of neurons in the cerebrum and hippocampus of KFDV infected mice. Further studies are needed to confirm if the lesions noticed in mice brain simulate the brain lesions in humans since gross motor-neuron symptoms are similar in mice as well as humans.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Enfermedad del Bosque de Kyasanur , Humanos , Animales , Ratones , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Encéfalo/patología , Corteza Cerebral/patología , Hipocampo/patología , Apoptosis , Neuronas Motoras/patología , Parálisis
4.
Vector Borne Zoonotic Dis ; 24(4): 226-236, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38436222

RESUMEN

Introduction: Lyme disease (LD) affects ∼476,000 people each year in the United States. Symptoms are variable and include rash and flu-like symptoms. Reasons for the wide variation in disease outcomes are unknown. Powassan virus (POWV) is a tick-borne flavivirus that causes disease ranging from asymptomatic infection to encephalitis, neurologic damage, and death. POWV and LD geographic case distributions overlap, with Ixodes species ticks as the common vectors. Clinical ramifications of coinfection or sequential infection are unknown. Objectives: This study's primary objective was to determine the prevalence of POWV-reactive antibodies in sera samples collected from previously studied cohorts of individuals with self-reported LD history residing in the Northeastern United States. As a secondary objective, we studied clinical differences between people with self-reported LD history and low versus high POWV antibody levels. Methods: We used an enzyme-linked immunosorbent assay (ELISA) to quantify IgG directed at the POWV envelope (E) protein domain III in 538 samples from individuals with self-reported LD history and 16 community controls. The samples were also tested with an ELISA assay to quantify IgG directed at the POWV NS1 protein. Results: The percentage of individuals with LD history and possible evidence of POWV exposure varied depending on the assay utilized. We found no significant difference in clinical symptoms between those with low or high POWV IgG levels in the in-house assay. Congruence of the EDIII and NS1 assays was low with only 12% of those positive in the in-house EDIII ELISA testing positive in the POWV NS1 ELISA. Conclusions: The results highlight the difficulty in flavivirus diagnostic testing, particularly in the retrospective detection of flavivirus exposure. The findings suggest that a prospective study with symptomatic patients using approved clinical testing is necessary to address the incidence and clinical implications of LD and POWV co-infection or sequential infection.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Ixodes , Enfermedad de Lyme , Animales , Humanos , Estados Unidos/epidemiología , Prevalencia , Estudios Retrospectivos , Estudios Prospectivos , Encefalitis Transmitida por Garrapatas/veterinaria , Enfermedad de Lyme/epidemiología , Enfermedad de Lyme/veterinaria , New England/epidemiología , Anticuerpos Antivirales , Inmunoglobulina G
5.
Vaccine ; 42(10): 2695-2706, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38494412

RESUMEN

BACKGROUND: Three encephalitic alphaviruses-western, eastern, and Venezuelan equine encephalitis virus (WEEV, EEEV and VEEV)-can cause severe disease and have the potential to be used as biological weapons. There are no approved vaccines for human use. A novel multivalent MVA-BN-WEV vaccine encodes the envelope surface proteins of the 3 viruses and is thereby potentially able to protect against them all, as previously demonstrated in animal models. This first-in-human study assessed the safety, tolerability, and immunogenicity of MVA-BN-WEV vaccine in healthy adult participants. METHODS: Forty-five participants were enrolled into 3 dose groups (1 × 10E7 Inf.U, 1 × 10E8 Inf.U, and 2 × 10E8 Inf.U), received 2 doses 4 weeks apart, and were then monitored for 6 months. RESULTS: The safety profile of MVA-BN-WEV was acceptable at all administered doses, with incidence of local solicited AEs increased with increasing dose and no other clinically meaningful differences between dose groups. One SAE (Grade 2 pleural effusion) was reported in the lowest dose group and assessed as possibly related. No AEs resulted in death or led to withdrawal from the second vaccination or from the trial. The most common local solicited AE was injection site pain, and general solicited AEs were headache, fatigue, and myalgia. MVA-BN-WEV induced humoral immune responses; WEEV-, EEEV- and VEEV-specific neutralizing antibody responses peaked 2 weeks following the second vaccination, and the magnitude of these responses increased with dose escalation. The highest dose resulted in seroconversion of all (100 %) participants for WEEV and VEEV and 92.9 % for EEEV, 2 weeks following second vaccination, and durability was observed for 6 months. MVA-BN-WEV induced cellular immune responses to VEEV E1 and E2 (EEEV and WEEV not tested) and a dose effect for peptide pool E2. CONCLUSION: The study demonstrated that MVA-BN-WEV is well tolerated, induces immune responses, and is suitable for further development. CLINICAL TRIAL REGISTRY NUMBER: NCT04131595.


Asunto(s)
Alphavirus , Virus de la Encefalitis Equina Venezolana , Encefalomielitis Equina , Animales , Caballos , Humanos , Anticuerpos Antivirales , Encefalomielitis Equina/prevención & control , Anticuerpos Neutralizantes , Virus Vaccinia , Inmunogenicidad Vacunal
6.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(3): 235-243, 2024 Mar.
Artículo en Chino | MEDLINE | ID: mdl-38512034

RESUMEN

Objective To investigate the effect of 3-deazaadenosine (3-DAA), an N6-methyladenosine (m6A) methylation modification inhibitor, on the replication of the Japanese encephalitis virus (JEV). Methods Neuro2a mouse neuroblastoma cells, N9 mouse microglial cells, and BHK baby hamster kidney cells were exposed to JEV and then treated with 3-DAA. JEV was also injected into the footpad of adult C57BL/6 mice, which were then administered 3-DAA intraperitoneally. Real-time quantitative PCR was utilized to measure mRNA expression levels of JEV, interleukin 1ß (IL-1ß), IL-6, tumor necrosis factor α (TNF-α), monocyte chemoattractant protein 1 (MCP-1), inducible nitric oxide synthase (iNOS), arginase 1 (Arg1), interferon (IFN)-α, IFN-ß, IFN-γ, and C-X-C motif chemokine ligand 10 (CXCL10) in the cells and mouse brain tissues. Western blot analysis was used to detect JEV protein expression in the cells and mouse brain tissues. Furthermore, the survival of the mice was monitored and pathological changes in mouse brains were observed via hematoxylin and eosin (HE) staining. Results 3-DAA had a dose-dependent effect on the replication of RNA and protein expression of JEV in both BHK, N9, Neuro 2α cells and mouse brain tissues, which resulted in rapid progression of JEV infection in mice and a decrease in their survival rate. Furthermore, 3-DAA suppressed the expression of inflammatory factors such as IL-6, TNF-α, CXCL10, IL-1ß and iNOS, thus weakening the immune response. Conclusion 3-DAA promotes JEV infection and hastens death of infected cells and mice, indicating that m6A modification may negatively regulate JEV replication.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie) , Tubercidina , Cricetinae , Animales , Ratones , Ratones Endogámicos C57BL , Antivirales/farmacología , Interleucina-6 , Factor de Necrosis Tumoral alfa/genética , Interferón-alfa , Interleucina-1beta/genética
7.
Antiviral Res ; 224: 105837, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38387750

RESUMEN

The COVID-19 pandemic has shown the need to develop effective therapeutics in preparedness for further epidemics of virus infections that pose a significant threat to human health. As a natural compound antiviral candidate, we focused on α-dystroglycan, a highly glycosylated basement membrane protein that links the extracellular matrix to the intracellular cytoskeleton. Here we show that the N-terminal fragment of α-dystroglycan (α-DGN), as produced in E. coli in the absence of post-translational modifications, blocks infection of SARS-CoV-2 in cell culture, human primary gut organoids and the lungs of transgenic mice expressing the human receptor angiotensin I-converting enzyme 2 (hACE2). Prophylactic and therapeutic administration of α-DGN reduced SARS-CoV-2 lung titres and protected the mice from respiratory symptoms and death. Recombinant α-DGN also blocked infection of a wide range of enveloped viruses including the four Dengue virus serotypes, influenza A virus, respiratory syncytial virus, tick-borne encephalitis virus, but not human adenovirus, a non-enveloped virus in vitro. This study establishes soluble recombinant α-DGN as a broad-band, natural compound candidate therapeutic against enveloped viruses.


Asunto(s)
COVID-19 , SARS-CoV-2 , Ratones , Animales , Humanos , Distroglicanos , Pandemias , Escherichia coli , Ratones Transgénicos , Antivirales/farmacología
8.
Front Immunol ; 15: 1352720, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38318179

RESUMEN

Tick-borne Encephalitis (TBE) is a severe disease of the Central Nervous System (CNS) caused by the tick-borne encephalitis virus (TBEV). The generation of protective immunity after TBEV infection or TBE vaccination relies on the integrated responses of many distinct cell types at distinct physical locations. While long-lasting memory immune responses, in particular, form the basis for the correlates of protection against many diseases, these correlates of protection have not yet been clearly defined for TBE. This review addresses the immune control of TBEV infection and responses to TBE vaccination. Potential correlates of protection and the durability of protection against disease are discussed, along with outstanding questions in the field and possible areas for future research.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Humanos , Sistema Nervioso Central , Vacunación
9.
Vaccine ; 42(4): 745-752, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38242736

RESUMEN

Inactivated vaccines, such as tick-borne-encephalitis-virus-(TBEV) vaccine, have been discussed as less immunogenic in elderly and in immunocompromised patients. In this controlled cross-sectional cohort study, the antibody and cellular responses after TBEV-vaccination were investigated in 36 rheumatoid arthritis (RA) patients and 112 healthy controls (HC) by evaluating IgG-anti-TBEV concentration, neutralization and relative avidity index (RAI). Cellular reactivity was assessed by IFNgamma-producing spot-forming-units (SFU) by ELISPOT assay and flow cytometry. RA patients showed lower IgG-anti-TBEV compared to HC, which were influenced by age at and time since last TBEV vaccination and disease duration. High-responders regarding cellular immunity and avidity were less frequent in RA compared to HC. RA patients who had received booster vaccinations were more likely to demonstrate higher IgG-anti-TBEV responses compared to those who had not. In conclusion, RA patients showed a negative effect of age on anti-TBEV-IgG and immunological benefits of timely booster vaccination are suggested.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Garrapatas , Humanos , Anciano , Animales , Anticuerpos Antivirales , Estudios Transversales , Vacunación , Inmunidad Celular , Inmunoglobulina G , Encefalitis Transmitida por Garrapatas/prevención & control
10.
Expert Rev Vaccines ; 23(1): 226-236, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38288983

RESUMEN

INTRODUCTION: Tick-borne encephalitis (TBE) is rapidly spreading to new areas in many parts of Europe. While vaccination remains the most effective method of protection against the disease, vaccine uptake is low in many endemic countries. AREAS COVERED: We conducted a literature search of the MEDLINE database to identify articles published from 2018 to 2023 that evaluated the immunogenicity and effectiveness of TBE vaccines, particularly Encepur, when booster doses were administered up to 10 years apart. We searched PubMed with the MeSH terms 'Encephalitis, Tick-Borne/prevention and control' and 'Vaccination' for articles published in the English language. EXPERT OPINION: Long-term immunogenicity data for Encepur and real-world data on vaccine effectiveness and breakthrough infections following the two European TBE vaccines, Encepur and FSME-Immun, have shown that extending the booster interval from 3-5 years to 10 years does not negatively impact protection against TBE, regardless of age. Such extension not only streamlines the vaccination schedules but may also increase vaccine uptake and compliance among those living in endemic regions.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Vacunas Virales , Humanos , Anticuerpos Antivirales , Vacunación/métodos , Europa (Continente) , Encefalitis Transmitida por Garrapatas/prevención & control
12.
Euro Surveill ; 29(2)2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38214080

RESUMEN

BackgroundIn Sweden, information on seroprevalence of tick-borne encephalitis virus (TBEV) in the population, including vaccination coverage and infection, is scattered. This is largely due to the absence of a national tick-borne encephalitis (TBE) vaccination registry, scarcity of previous serological studies and use of serological methods not distinguishing between antibodies induced by vaccination and infection. Furthermore, the number of notified TBE cases in Sweden has continued to increase in recent years despite increased vaccination.AimThe aim was to estimate the TBEV seroprevalence in Sweden.MethodsIn 2018 and 2019, 2,700 serum samples from blood donors in nine Swedish regions were analysed using a serological method that can distinguish antibodies induced by vaccination from antibodies elicited by infection. The regions were chosen to reflect differences in notified TBE incidence.ResultsThe overall seroprevalence varied from 9.7% (95% confidence interval (CI): 6.6-13.6%) to 64.0% (95% CI: 58.3-69.4%) between regions. The proportion of vaccinated individuals ranged from 8.7% (95% CI: 5.8-12.6) to 57.0% (95% CI: 51.2-62.6) and of infected from 1.0% (95% CI: 0.2-3.0) to 7.0% (95% CI: 4.5-10.7). Thus, more than 160,000 and 1,600,000 individuals could have been infected by TBEV and vaccinated against TBE, respectively. The mean manifestation index was 3.1%.ConclusionA difference was observed between low- and high-incidence TBE regions, on the overall TBEV seroprevalence and when separated into vaccinated and infected individuals. The estimated incidence and manifestation index argue that a large proportion of TBEV infections are not diagnosed.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Infecciones por Flavivirus , Humanos , Encefalitis Transmitida por Garrapatas/epidemiología , Encefalitis Transmitida por Garrapatas/prevención & control , Suecia/epidemiología , Cobertura de Vacunación , Estudios Seroepidemiológicos , Vacunación , Anticuerpos Antivirales
13.
EMBO Mol Med ; 16(1): 185-217, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177535

RESUMEN

Japanese encephalitis virus (JEV) pathogenesis is driven by a combination of neuronal death and neuroinflammation. We tested 42 FDA-approved drugs that were shown to induce autophagy for antiviral effects. Four drugs were tested in the JE mouse model based on in vitro protective effects on neuronal cell death, inhibition of viral replication, and anti-inflammatory effects. The antipsychotic phenothiazines Methotrimeprazine (MTP) & Trifluoperazine showed a significant survival benefit with reduced virus titers in the brain, prevention of BBB breach, and inhibition of neuroinflammation. Both drugs were potent mTOR-independent autophagy flux inducers. MTP inhibited SERCA channel functioning, and induced an adaptive ER stress response in diverse cell types. Pharmacological rescue of ER stress blocked autophagy and antiviral effect. MTP did not alter translation of viral RNA, but exerted autophagy-dependent antiviral effect by inhibiting JEV replication complexes. Drug-induced autophagy resulted in reduced NLRP3 protein levels, and attenuation of inflammatory cytokine/chemokine release from infected microglial cells. Our study suggests that MTP exerts a combined antiviral and anti-inflammatory effect in JEV infection, and has therapeutic potential for JE treatment.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , Animales , Ratones , Virus de la Encefalitis Japonesa (Especie)/fisiología , Metotrimeprazina/farmacología , Metotrimeprazina/uso terapéutico , Enfermedades Neuroinflamatorias , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/patología , Antivirales/farmacología , Antivirales/uso terapéutico , Autofagia , Antiinflamatorios/uso terapéutico
14.
Vector Borne Zoonotic Dis ; 24(2): 118-121, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37870590

RESUMEN

Background: Eastern equine encephalitis virus (EEEV) is a rare mosquito-borne illness exhibiting rapid neurological deterioration and permanent damage. Despite its >30% mortality and >60% long-term neurological damage, EEEV has no approved antiviral medication or vaccination. This report uniquely aims to describe a rare case of EEEV and provide a current literature review of therapeutic and preventative options from the clinical perspective to guide clinicians and public health workers, along with informing them about its impact and current knowledge gaps. Methods: A retrospective chart review of the electronic medical record was performed for a patient's 10-day hospital admission in July 2021. In addition, PubMed was searched using relevant keywords for a literature review of EEEV. Results: A 61-year-old woman presented with dysarthria and right-sided facial droop. Acute ischemic stroke was ruled out, and empiric intravenous (IV) antibiotics were initiated for possible infectious etiology. The patient developed worsening mental status and fever and was intubated, with antibiotics broadened with concern for meningitis along with tick-borne illness. The patient remained encephalopathic and febrile, and lumbar serologies were consistent with viral meningoencephalitis or acute disseminated encephalomyelitis. Several days after collection, quantitative antibody testing returned positive for EEEV. The patient was pronounced dead on hospital day 10. On review of the literature regarding EEEV, supportive care and prevention remain the cornerstone of management. Although early IV immunoglobulin and high-dose steroids have shown potential as treatments to reduce morbidity and mortality, no vaccines have been approved to date. Conclusion: Prospective trials and further investigations into treatment and preventative options may be useful in reducing the morbidity and mortality associated with EEEV.


Asunto(s)
Virus de la Encefalitis Equina del Este , Encefalomielitis Equina , Accidente Cerebrovascular Isquémico , Humanos , Femenino , Caballos , Animales , Persona de Mediana Edad , Accidente Cerebrovascular Isquémico/veterinaria , Estudios Retrospectivos , Estudios Prospectivos , Encefalomielitis Equina/veterinaria , Antibacterianos
15.
Pediatr Res ; 95(2): 464-479, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37880334

RESUMEN

This review utilizes quatitative methods and bibliometric data to analyse the trends of emerging and re-emerging vector-borne diseases, with a focus on their impact on pediatric population. To conduct this analysis, a systematic search of PubMed articles from the past two decades was performed, specifically looking at 26 different vector-borne viruses listed in WHO and CDC list of vector-borne viruses. The review found that diseases like Dengue, Zika, West Nile, and Chikungunya were frequently discussed in the literature. On the other hand, diseases such as Tick-borne encephalitis, Rift Valley fever, Venezuelan equine encephalitis, Sindbis fever, Venezuelan equine encephalitis, Ross River virus, and Eastern equine encephalitis showed an upward trend in publications, indicating potential resurgence. In addition to discussing trends and patterns, the review delves into the clinical manifestations and long-term effects of the top 10 viruses in children. It highlights various factors including deforestation, urbanization, global travel, and immunosuppression that contribute to disease emergence and resurgence. To effectively combat these vector-borne diseases, continuous surveillance is crucial. The review also emphasizes the importance of increased vaccination efforts and targeted research to address the health challenges they pose. IMPACT: This review employs quantitative analysis of publications to elucidate trends in emerging pediatric vector-borne viral diseases over two decades. Dengue, the most prevalent of these diseases, has spread to new regions. New strains of Japanese Encephalitis have caused outbreaks. Resurgence of Tick-borne Encephalitis, West Nile, and Yellow Fever due to vaccine hesitancy has also transpired. Continuous global surveillance, increased vaccination, and research into novel therapeutics are imperative to combat the substantial morbidity and mortality burden these diseases pose for children worldwide.


Asunto(s)
Dengue , Encefalitis Transmitida por Garrapatas , Encefalomielitis Equina Venezolana , Virosis , Virus , Infección por el Virus Zika , Virus Zika , Animales , Caballos , Niño , Humanos , Dengue/epidemiología
16.
Bull Exp Biol Med ; 176(1): 72-76, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38091143

RESUMEN

A promising approach to the development of new means for preventing infection caused by tick-borne encephalitis virus can be DNA vaccines encoding polyepitope T-cell immunogens. A DNA vaccine pVAX-AG4-ub encoding an artificial polyepitope immunogen that includes cytotoxic and T-helper epitopes from the NS1, NS3, NS5, and E proteins of the tick-borne encephalitis virus has been obtained. The developed construct ensured the synthesis of the corresponding mRNAs in transfected eukaryotic cells. Immunization of mice with pVAX-AG4-ub induced the formation of a virus-specific T-cell response providing 50% protection from lethal infection with the virus.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Vacunas de ADN , Vacunas Virales , Animales , Ratones , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Vacunas de ADN/genética , Vacunas Virales/genética , Linfocitos T , Inmunización
17.
MMWR Recomm Rep ; 72(5): 1-29, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37943707

RESUMEN

Tick-borne encephalitis (TBE) virus is focally endemic in parts of Europe and Asia. The virus is primarily transmitted to humans by the bites of infected: Ixodes species ticks but can also be acquired less frequently by alimentary transmission. Other rare modes of transmission include through breastfeeding, blood transfusion, solid organ transplantation, and slaughtering of viremic animals. TBE virus can cause acute neurologic disease, which usually results in hospitalization, often permanent neurologic or cognitive sequelae, and sometimes death. TBE virus infection is a risk for certain travelers and for laboratory workers who work with the virus. In August 2021, the Food and Drug Administration approved Ticovac TBE vaccine for use among persons aged ≥1 year. This report summarizes the epidemiology of and risks for infection with TBE virus, provides information on the immunogenicity and safety of TBE vaccine, and summarizes the recommendations of the Advisory Committee on Immunization Practices (ACIP) for use of TBE vaccine among U.S. travelers and laboratory workers.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Ixodes , Vacunas , Humanos , Animales , Estados Unidos/epidemiología , Encefalitis Transmitida por Garrapatas/epidemiología , Encefalitis Transmitida por Garrapatas/prevención & control , Comités Consultivos , Vacunación
18.
J Med Virol ; 95(11): e29245, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-38009693

RESUMEN

Arthropod-borne flaviviruses include a number of medically relevant human pathogens such as the mosquito-borne dengue (DEN), Zika, and yellow fever (YF) viruses as well as tick-borne encephalitis virus (TBEV). All flaviviruses are antigenically related and anamnestic responses due to prior immunity can modulate antibody specificities in subsequent infections or vaccinations. In our study, we analyzed the induction of broadly flavivirus cross-reactive antibodies in tick-borne encephalitis (TBE) and DEN patients without or with prior flavivirus exposure through TBE and/or YF vaccination, and determined the contribution of these antibodies to TBE and dengue virus (DENV) neutralization. In addition, we investigated the formation of cross-reactive antibodies in TBE-vaccination breakthroughs (VBTs). A TBEV infection without prior YF or TBE vaccination induced predominantly type-specific antibodies. In contrast, high levels of broadly cross-reactive antibodies were found in samples from TBE patients prevaccinated against YF as well as in DEN patients prevaccinated against TBE and/or YF. While these cross-reactive antibodies did not neutralize TBEV, they were effective in neutralizing DENV. This discrepancy points to structural differences between the two viruses and indicates that broadly cross-reactive epitopes are less accessible in TBEV than in DENV. In TBE VBT infections, type-specific antibodies dominated the antibody response, thus revealing no difference from that of unvaccinated TBE patients. Our results emphasize significant differences in the structural properties of different flaviviruses that have an impact on the induction of broadly cross-reactive antibodies and their functional activities in virus neutralization.


Asunto(s)
Dengue , Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Infecciones por Flavivirus , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Encefalitis Transmitida por Garrapatas/prevención & control , Formación de Anticuerpos , Anticuerpos Antivirales , Infecciones por Flavivirus/prevención & control , Vacunación , Dengue/prevención & control
19.
PLoS Pathog ; 19(11): e1011813, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38011306

RESUMEN

Innate immune signaling in the central nervous system (CNS) exhibits many remarkable specializations that vary across cell types and CNS regions. In the setting of neuroinvasive flavivirus infection, neurons employ the immunologic kinase receptor-interacting kinase 3 (RIPK3) to promote an antiviral transcriptional program, independently of the traditional function of this enzyme in promoting necroptotic cell death. However, while recent work has established roles for neuronal RIPK3 signaling in controlling mosquito-borne flavivirus infections, including West Nile virus and Zika virus, functions for RIPK3 signaling in the CNS during tick-borne flavivirus infection have not yet been explored. Here, we use a model of Langat virus (LGTV) encephalitis to show that RIPK3 signaling is specifically required in neurons of the cerebellum to control LGTV replication and restrict disease pathogenesis. This effect did not require the necroptotic executioner molecule mixed lineage kinase domain like protein (MLKL), a finding similar to previous observations in models of mosquito-borne flavivirus infection. However, control of LGTV infection required a unique, region-specific dependence on RIPK3 to promote expression of key antiviral interferon-stimulated genes (ISG) in the cerebellum. This RIPK3-mediated potentiation of ISG expression was associated with robust cell-intrinsic restriction of LGTV replication in cerebellar granule cell neurons. These findings further illuminate the complex roles of RIPK3 signaling in the coordination of neuroimmune responses to viral infection, as well as provide new insight into the mechanisms of region-specific innate immune signaling in the CNS.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Enfermedades por Picaduras de Garrapatas , Garrapatas , Animales , Encéfalo/patología , Virus de la Encefalitis Transmitidos por Garrapatas/fisiología , Encefalitis Transmitida por Garrapatas/patología , Interferones/metabolismo , Enfermedades por Picaduras de Garrapatas/patología , Replicación Viral/genética , Ratones
20.
J Virol ; 97(12): e0118323, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37991381

RESUMEN

IMPORTANCE: Central nervous system infection by flaviviruses such as Japanese encephalitis virus, Dengue virus, and West Nile virus results in neuroinflammation and neuronal damage. However, little is known about the role of long non-coding RNAs (lncRNAs) in flavivirus-induced neuroinflammation and neuronal cell death. Here, we characterized the role of a flavivirus-induced lncRNA named JINR1 during the infection of neuronal cells. Depletion of JINR1 during virus infection reduces viral replication and cell death. An increase in GRP78 expression by JINR1 is responsible for promoting virus replication. Flavivirus infection induces the expression of a cellular protein RBM10, which interacts with JINR1. RBM10 and JINR1 promote the proinflammatory transcription factor NF-κB activity, which is detrimental to cell survival.


Asunto(s)
Muerte Celular , Virus de la Encefalitis Japonesa (Especie) , FN-kappa B , Neuronas , ARN Largo no Codificante , Proteínas de Unión al ARN , Humanos , Virus de la Encefalitis Japonesa (Especie)/crecimiento & desarrollo , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/virología , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/metabolismo , Neuronas/patología , Neuronas/virología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...