Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3738, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702297

RESUMEN

Whole virus-based inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide have been critical to the COVID-19 pandemic response. Although these vaccines are protective against homologous coronavirus infection, the emergence of novel variants and the presence of large zoonotic reservoirs harboring novel heterologous coronaviruses provide significant opportunities for vaccine breakthrough, which raises the risk of adverse outcomes like vaccine-associated enhanced respiratory disease. Here, we use a female mouse model of coronavirus disease to evaluate inactivated vaccine performance against either homologous challenge with SARS-CoV-2 or heterologous challenge with a bat-derived coronavirus that represents a potential emerging disease threat. We show that inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide can cause enhanced respiratory disease during heterologous infection, while use of an alternative adjuvant does not drive disease and promotes heterologous viral clearance. In this work, we highlight the impact of adjuvant selection on inactivated vaccine safety and efficacy against heterologous coronavirus infection.


Asunto(s)
Hidróxido de Aluminio , Vacunas contra la COVID-19 , COVID-19 , SARS-CoV-2 , Vacunas de Productos Inactivados , Animales , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Femenino , COVID-19/prevención & control , COVID-19/inmunología , COVID-19/virología , Ratones , Vacunas de Productos Inactivados/inmunología , SARS-CoV-2/inmunología , Hidróxido de Aluminio/administración & dosificación , Modelos Animales de Enfermedad , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes de Vacunas , Anticuerpos Antivirales/inmunología , Ratones Endogámicos BALB C , Humanos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología
2.
Orthop J Sports Med ; 12(3): 23259671231219023, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38435717

RESUMEN

Background: One proposed mechanism of rotator cuff disease is scapular motion impairments contributing to rotator cuff compression and subsequent degeneration. Purpose: To model the effects of scapular angular deviations on rotator cuff tendon proximity for subacromial and internal mechanical impingement risk during scapular plane abduction. Study Design: Descriptive laboratory study. Methods: Three-dimensional bone models were reconstructed from computed tomography scans obtained from 10 asymptomatic subjects and 9 symptomatic subjects with a clinical presentation of impingement syndrome. Models were rotated to average scapular orientations from a healthy dataset at higher (120°) and lower (subject-specific) humeral elevation angles to investigate internal and subacromial impingement risks, respectively. Incremental deviations in scapular upward/downward rotation, internal/external rotation, and anterior/posterior tilt were imposed on the models to simulate scapular movement impairments. The minimum distance between the rotator cuff insertions and potential impinging structures (eg, glenoid, acromion) was calculated. Two-way mixed-model analyses of variance assessed for effects of scapular deviation and group. Results: At 120° of humerothoracic elevation, minimum distances from the supraspinatus and infraspinatus insertions to the glenoid increased with ≥5° changes in upward rotation (1.6-9.8 mm, P < .001) or external rotation (0.9-5.0 mm, P≤ .048), or with ≥10° changes in anterior tilt (1.1-3.2 mm, P < .001). At lower angles, ≥20° changes in most scapular orientations significantly increased the distance between the supraspinatus and infraspinatus insertions and the acromion or coracoacromial ligament. Conclusion: A reduction in scapular upward rotation decreases the distance between the rotator cuff tendon insertions and glenoid at 120° humerothoracic elevation. Interpretation is complicated for lower angles because the humeral elevation angle was defined by the minimum distance. Clinical Relevance: These results may assist clinical decision making regarding the effects of scapular movement deviations in patients with rotator cuff pathology and scapular dyskinesia and may help inform the selection of clinical interventions.

3.
Cell ; 187(6): 1363-1373.e12, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38366591

RESUMEN

In response to the 2022 outbreak of mpox driven by unprecedented human-to-human monkeypox virus (MPXV) transmission, we designed BNT166, aiming to create a highly immunogenic, safe, accessible, and scalable next-generation vaccine against MPXV and related orthopoxviruses. To address the multiple viral forms and increase the breadth of immune response, two candidate multivalent mRNA vaccines were evaluated pre-clinically: a quadrivalent vaccine (BNT166a; encoding the MPXV antigens A35, B6, M1, H3) and a trivalent vaccine (BNT166c; without H3). Both candidates induced robust T cell responses and IgG antibodies in mice, including neutralizing antibodies to both MPXV and vaccinia virus. In challenge studies, BNT166a and BNT166c provided complete protection from vaccinia, clade I, and clade IIb MPXV. Furthermore, immunization with BNT166a was 100% effective at preventing death and at suppressing lesions in a lethal clade I MPXV challenge in cynomolgus macaques. These findings support the clinical evaluation of BNT166, now underway (NCT05988203).


Asunto(s)
Monkeypox virus , Mpox , Vacuna contra Viruela , Animales , Humanos , Ratones , Macaca fascicularis , Monkeypox virus/genética , Mpox/inmunología , Mpox/prevención & control , Vacunas Combinadas , Virus Vaccinia/genética
4.
Vaccines (Basel) ; 12(1)2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38276675

RESUMEN

The COVID-19 pandemic led to the rapid and worldwide development of highly effective vaccines against SARS-CoV-2. However, there is significant individual-to-individual variation in vaccine efficacy due to factors including viral variants, host age, immune status, environmental and host genetic factors. Understanding those determinants driving this variation may inform the development of more broadly protective vaccine strategies. While host genetic factors are known to impact vaccine efficacy for respiratory pathogens such as influenza and tuberculosis, the impact of host genetic variation on vaccine efficacy against COVID-19 is not well understood. To model the impact of host genetic variation on SARS-CoV-2 vaccine efficacy, while controlling for the impact of non-genetic factors, we used the Diversity Outbred (DO) mouse model. We found that DO mice immunized against SARS-CoV-2 exhibited high levels of variation in vaccine-induced neutralizing antibody responses. While the majority of the vaccinated mice were protected from virus-induced disease, similar to human populations, we observed vaccine breakthrough in a subset of mice. Importantly, we found that this variation in neutralizing antibody, virus-induced disease, and viral titer is heritable, indicating that the DO serves as a useful model system for studying the contribution of genetic variation of both vaccines and disease outcomes.

5.
Res Sq ; 2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37961507

RESUMEN

Inactivated whole virus SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide (Alum) are among the most widely used COVID-19 vaccines globally and have been critical to the COVID-19 pandemic response. Although these vaccines are protective against homologous virus infection in healthy recipients, the emergence of novel SARS-CoV-2 variants and the presence of large zoonotic reservoirs provide significant opportunities for vaccine breakthrough, which raises the risk of adverse outcomes including vaccine-associated enhanced respiratory disease (VAERD). To evaluate this possibility, we tested the performance of an inactivated SARS-CoV-2 vaccine (iCoV2) in combination with Alum against either homologous or heterologous coronavirus challenge in a mouse model of coronavirus-induced pulmonary disease. Consistent with human results, iCoV2 + Alum protected against homologous challenge. However, challenge with a heterologous SARS-related coronavirus, Rs-SHC014-CoV (SHC014), up to at least 10 months post-vaccination, resulted in VAERD in iCoV2 + Alum-vaccinated animals, characterized by pulmonary eosinophilic infiltrates, enhanced pulmonary pathology, delayed viral clearance, and decreased pulmonary function. In contrast, vaccination with iCoV2 in combination with an alternative adjuvant (RIBI) did not induce VAERD and promoted enhanced SHC014 clearance. Further characterization of iCoV2 + Alum-induced immunity suggested that CD4+ T cells were a major driver of VAERD, and these responses were partially reversed by re-boosting with recombinant Spike protein + RIBI adjuvant. These results highlight potential risks associated with vaccine breakthrough in recipients of Alum-adjuvanted inactivated vaccines and provide important insights into factors affecting both the safety and efficacy of coronavirus vaccines in the face of heterologous virus infections.

6.
Cell Rep ; 42(4): 112326, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37000623

RESUMEN

Group 2B ß-coronaviruses (sarbecoviruses) have caused regional and global epidemics in modern history. Here, we evaluate the mechanisms of cross-sarbecovirus protective immunity, currently less clear yet important for pan-sarbecovirus vaccine development, using a panel of alphavirus-vectored vaccines covering bat to human strains. While vaccination does not prevent virus replication, it protects against lethal heterologous disease outcomes in both severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and clade 2 bat sarbecovirus challenge models. The spike vaccines tested primarily elicit a highly S1-specific homologous neutralizing antibody response with no detectable cross-virus neutralization. Rather, non-neutralizing antibody functions, mechanistically linked to FcgR4 and spike S2, mediate cross-protection in wild-type mice. Protection is lost in FcR knockout mice, further supporting a model for non-neutralizing, protective antibodies. These data highlight the importance of FcR-mediated cross-protective immune responses in universal pan-sarbecovirus vaccine designs.


Asunto(s)
Alphavirus , COVID-19 , Quirópteros , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Vacunas Virales , Humanos , Animales , Ratones , Anticuerpos Antivirales , SARS-CoV-2 , COVID-19/prevención & control , Anticuerpos Neutralizantes , Vacunación
7.
bioRxiv ; 2022 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-36482964

RESUMEN

Two group 2B ß-coronaviruses (sarbecoviruses) have caused regional and global epidemics in modern history. The mechanisms of cross protection driven by the sarbecovirus spike, a dominant immunogen, are less clear yet critically important for pan-sarbecovirus vaccine development. We evaluated the mechanisms of cross-sarbecovirus protective immunity using a panel of alphavirus-vectored vaccines covering bat to human strains. While vaccination did not prevent virus replication, it protected against lethal heterologous disease outcomes in both SARS-CoV-2 and clade 2 bat sarbecovirus HKU3-SRBD challenge models. The spike vaccines tested primarily elicited a highly S1-specific homologous neutralizing antibody response with no detectable cross-virus neutralization. We found non-neutralizing antibody functions that mediated cross protection in wild-type mice were mechanistically linked to FcgR4 and spike S2-binding antibodies. Protection was lost in FcR knockout mice, further supporting a model for non-neutralizing, protective antibodies. These data highlight the importance of FcR-mediated cross-protective immune responses in universal pan-sarbecovirus vaccine designs.

8.
Biochem Biophys Res Commun ; 592: 38-43, 2022 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-35026603

RESUMEN

Ovarian cancer is the deadliest gynecological cancer which rarely causes symptoms, and goes undetected until reaching the advanced stage of drug-resistant metastases. The cationic porphyrin meso-tetra(4-N-methylpyridyl)porphine (TMPyP) is a well-known photosensitizer (PS) used in photodyamic therapy (PDT) for curing cancer due to its strong affinity for DNA and high yield of reactive oxygen species (ROS) upon light activation. The practicality to irradiate tumor cells alone in the physiological system being slim (due to the close proximity of healthy cells and tumors), we looked for a variation in the PDT using a mixture of TMPyP with 1,5-dihydroxynapthalene (DHN) and Fe(III) ions at a mole ratio of 1:20:17 (drug combo) respectively in aqueous solution. The drug combo needs no photoactivation in H2O2 rich environment (mimicking the microenvironment of cancer/tumor), where it generates È®H and juglone, the latter being a known potent anticancer agent. In vitro studies of the drug combo in drug resistant and sensitive ovarian cancer cell lines showed drastic growth inhibition and cell death compared to normal epithelial cells. The drug combo provides an effective and non-invasive alternative to conventional PDT, exploiting the cytosolic carcinogenic H2O2 to produce an efficient anticancer treatment. The unique action of cancer-specific cytotoxicity arises from the redox chemistry involving activation of Fe(III) as the oxidizing agent to generate juglone, which utilizes the cytosolic ROS in cancer cells against itself.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Ácidos/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones , Naftoles/farmacología , Naftoquinonas/farmacología , Oxidación-Reducción , Porfirinas/farmacología , Espectrometría de Fluorescencia
9.
Health Sci Rep ; 4(3): e345, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34386613

RESUMEN

BACKGROUND AND AIMS: According to the World Health Organization (WHO), more than 75.7 million confirmed cases of coronavirus disease 2019 (COVID-19), a global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), have been reported so far. Researchers are working relentlessly to find effective solutions to this catastrophe, using genomic sequence-based investigation, immunological analysis, and more. The role of health disparity has also emerged as an intriguing factor that made a huge impact on the lives of people. METHODS: We analyzed various factors that triggered the health disparity in the United States of America along with the rate of COVID-19 morbidity and mortality. Furthermore, we have also focused on the State of Mississippi, which is suffering from an extreme health disparity. Data have been obtained from publicly available data sources including, Center for Disease Control and Prevention and Mississippi State Department of Health. Correlation analysis of the dataset has been performed using R software. RESULTS: Our analysis suggested that the COVID-19 infection rate per 100 000 people is directly correlated with the increasing number of the African American population in the United States. We have found a strong correlation between the obesity and the COVID-19 cases as well. All the counties in Mississippi demonstrate a strong correlation between a higher number of African American population to COVID-19 cases and obesity. Our data also indicate that a higher number of African American populations are facing socioeconomic disadvantages, which enhance their chances of becoming vulnerable to pre-existing ailments such as obesity, type-2 diabetes, and cardiovascular diseases. CONCLUSION: We proposed a possible explanation of increased COVID-19 infectivity in the African American population in the United States. This work has highlighted the intriguing factors that increased the health disparity at the time of the COVID-19 pandemic.

10.
J Infect Dis ; 224(3): 415-419, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-33961695

RESUMEN

Mutagenic ribonucleosides can act as broad-based antiviral agents. They are metabolized to the active ribonucleoside triphosphate form and concentrate in genomes of RNA viruses during viral replication. ß-d-N4-hydroxycytidine (NHC, initial metabolite of molnupiravir) is >100-fold more active than ribavirin or favipiravir against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with antiviral activity correlated to the level of mutagenesis in virion RNA. However, NHC also displays host mutational activity in an animal cell culture assay, consistent with RNA and DNA precursors sharing a common intermediate of a ribonucleoside diphosphate. These results indicate highly active mutagenic ribonucleosides may hold risk for the host.


Asunto(s)
Antivirales/farmacología , Citidina/análogos & derivados , Mutágenos/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/efectos adversos , Células CHO/efectos de los fármacos , Células Cultivadas , Cricetulus , Citidina/efectos adversos , Citidina/farmacología , Relación Dosis-Respuesta a Droga , Mutagénesis/efectos de los fármacos , Mutágenos/efectos adversos , SARS-CoV-2/genética , Replicación Viral/efectos de los fármacos
11.
J Med Chem ; 64(8): 4762-4786, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33835811

RESUMEN

A benzo[6]annulene, 4-(tert-butyl)-N-(3-methoxy-5,6,7,8-tetrahydronaphthalen-2-yl) benzamide (1a), was identified as an inhibitor against Chikungunya virus (CHIKV) with antiviral activity EC90 = 1.45 µM and viral titer reduction (VTR) of 2.5 log at 10 µM with no observed cytotoxicity (CC50 = 169 µM) in normal human dermal fibroblast cells. Chemistry efforts to improve potency, efficacy, and drug-like properties of 1a resulted in a novel lead compound 8q, which possessed excellent cellular antiviral activity (EC90 = 270 nM and VTR of 4.5 log at 10 µM) and improved liver microsomal stability. CHIKV resistance to an analog of 1a, compound 1c, tracked to a mutation in the nsP3 macrodomain. Further mechanism of action studies showed compounds working through inhibition of human dihydroorotate dehydrogenase in addition to CHIKV nsP3 macrodomain. Moderate efficacy was observed in an in vivo CHIKV challenge mouse model for compound 8q as viral replication was rescued from the pyrimidine salvage pathway.


Asunto(s)
Antivirales/farmacología , Derivados del Benceno/química , Virus Chikungunya/fisiología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Antivirales/farmacocinética , Antivirales/uso terapéutico , Derivados del Benceno/metabolismo , Derivados del Benceno/farmacología , Derivados del Benceno/uso terapéutico , Sitios de Unión , Línea Celular , Supervivencia Celular/efectos de los fármacos , Fiebre Chikungunya/tratamiento farmacológico , Dihidroorotato Deshidrogenasa , Modelos Animales de Enfermedad , Femenino , Semivida , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Relación Estructura-Actividad
12.
J Virol ; 94(24)2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32999019

RESUMEN

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus associated with debilitating arthralgia in humans. RNA secondary structure in the viral genome plays an important role in the lifecycle of alphaviruses; however, the specific role of RNA structure in regulating CHIKV replication is poorly understood. Our previous studies found little conservation in RNA secondary structure between alphaviruses, and this structural divergence creates unique functional structures in specific alphavirus genomes. Therefore, to understand the impact of RNA structure on CHIKV biology, we used SHAPE-MaP to inform the modeling of RNA secondary structure throughout the genome of a CHIKV isolate from the 2013 Caribbean outbreak. We then analyzed regions of the genome with high levels of structural specificity to identify potentially functional RNA secondary structures and identified 23 regions within the CHIKV genome with higher than average structural stability, including four previously identified, functionally important CHIKV RNA structures. We also analyzed the RNA flexibility and secondary structures of multiple 3'UTR variants of CHIKV that are known to affect virus replication in mosquito cells. This analysis found several novel RNA structures within these 3'UTR variants. A duplication in the 3'UTR that enhances viral replication in mosquito cells led to an overall increase in the amount of unstructured RNA in the 3'UTR. This analysis demonstrates that the CHIKV genome contains a number of unique, specific RNA secondary structures and provides a strategy for testing these secondary structures for functional importance in CHIKV replication and pathogenesis.IMPORTANCE Chikungunya virus (CHIKV) is a mosquito-borne RNA virus that causes febrile illness and debilitating arthralgia in humans. CHIKV causes explosive outbreaks but there are no approved therapies to treat or prevent CHIKV infection. The CHIKV genome contains functional RNA secondary structures that are essential for proper virus replication. Since RNA secondary structures have only been defined for a small portion of the CHIKV genome, we used a chemical probing method to define the RNA secondary structures of CHIKV genomic RNA. We identified 23 highly specific structured regions of the genome, and confirmed the functional importance of one structure using mutagenesis. Furthermore, we defined the RNA secondary structure of three CHIKV 3'UTR variants that differ in their ability to replicate in mosquito cells. Our study highlights the complexity of the CHIKV genome and describes new systems for designing compensatory mutations to test the functional relevance of viral RNA secondary structures.


Asunto(s)
Regiones no Traducidas 3'/genética , Virus Chikungunya/genética , ARN Viral/química , ARN Viral/genética , Animales , Línea Celular , Fiebre Chikungunya/virología , Chlorocebus aethiops , Culicidae , Efecto Citopatogénico Viral , Genoma Viral , Mutación , Conformación de Ácido Nucleico , Análisis de Secuencia , Células Vero , Replicación Viral/genética
13.
Clin Ther ; 41(10): 1912-1922, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31540729

RESUMEN

Mouse models are important tools both for studying the pathogenesis of infectious diseases and for the preclinical evaluation of vaccines and therapies against a wide variety of human pathogens. The use of genetically defined inbred mouse strains, humanized mice, and gene knockout mice has allowed the research community to explore how pathogens cause disease, define the role of specific host genes in either controlling or promoting disease, and identify potential targets for the prevention or treatment of a wide range of infectious agents. This review discusses several of the most commonly used mouse model systems, as well as new resources such as the Collaborative Cross as models for studying infectious diseases.


Asunto(s)
Enfermedades Transmisibles , Modelos Animales de Enfermedad , Animales , Humanos , Ratones , Ratones Endogámicos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Virol ; 93(23)2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31511388

RESUMEN

Equid herpesvirus 1 (EHV-1) is a viral pathogen of horse populations worldwide spread by the respiratory route and is known for causing outbreaks of neurologic syndromes and abortion storms. Previously, we demonstrated that an EHV-1 strain of the neuropathogenic genotype, T953, downregulates the beta interferon (IFN-ß) response in vitro in equine endothelial cells (EECs) at 12 h postinfection (hpi). In the present study, we explored the molecular correlates of this inhibition as clues toward an understanding of the mechanism. Data from our study revealed that EHV-1 infection of EECs significantly reduced both Toll-like receptor 3 (TLR3) and TLR4 mRNA expression at 6 hpi and 12 hpi. While EHV-1 was able to significantly reduce IRF9 mRNA at both 6 hpi and 12 hpi, the virus significantly reduced IFN regulatory factor 7 (IRF7) mRNA only at 12 hpi. EHV-1 did not alter the cellular level of Janus-activated kinase 1 (JAK1) at any time point. However, EHV-1 reduced the cellular level of expression of tyrosine kinase 2 (TYK2) at 12 hpi. Downstream of JAK1-TYK2 signaling, EHV-1 blocked the phosphorylation and activation of signal transducer and activator of transcription 2 (STAT2) when coincubated with exogenous IFN, at 12 hpi, although not at 3 or 6 hpi. Immunofluorescence staining revealed that the virus prevented the nuclear translocation of STAT2 molecules, confirming the virus-mediated inhibition of STAT2 activation. The pattern of suppression of phosphorylation of STAT2 by EHV-1 implicated viral late gene expression. These data help illuminate how EHV-1 strategically inhibits the host innate immune defense by limiting steps required for type I IFN sensitization and induction.IMPORTANCE To date, no commercial vaccine label has a claim to be fully protective against the diseases caused by equid herpesvirus 1 (EHV-1), especially the neurologic form. The interferon (IFN) system, of which type I IFN is of great importance, still remains a viable immunotherapeutic option against EHV-1 infection. The type I IFN system has been exploited successfully to treat other viral infections, such as chronic hepatitis B and C in humans. The current state of research on how EHV-1 interferes with the protective effect of type I IFN has indicated transient induction of type I IFN production followed by a rapid shutdown in vitro in equine endothelial cells (EECs). The significance of our study is the identification of certain steps in the type I IFN signaling pathway targeted for inhibition by EHV-1. Understanding this pathogen-host relationship is essential for the long-term goal of developing effective immunotherapy against EHV-1.


Asunto(s)
Células Endoteliales/metabolismo , Células Endoteliales/virología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/metabolismo , Herpesvirus Équido 1/inmunología , Interferón Tipo I/metabolismo , Animales , Regulación de la Expresión Génica , Hepatitis B Crónica , Infecciones por Herpesviridae/virología , Herpesvirus Équido 1/genética , Enfermedades de los Caballos/virología , Caballos , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Janus Quinasa 1/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal , TYK2 Quinasa/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo
15.
ACS Infect Dis ; 5(12): 2014-2028, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31257853

RESUMEN

Alphaviruses are arthropod-transmitted members of the Togaviridae family that can cause severe disease in humans, including debilitating arthralgia and severe neurological complications. Currently, there are no approved vaccines or antiviral therapies directed against the alphaviruses, and care is limited to treating disease symptoms. A phenotypic cell-based high-throughput screen was performed to identify small molecules that inhibit the replication of Venezuelan Equine Encephalitis Virus (VEEV). The compound, 1-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-N-(3-fluoro-4-methoxybenzyl)ethan-1-amine (1), was identified as a highly active, potent inhibitor of VEEV with an effective concentration for 90% inhibition of virus (EC90) of 0.89 µM and 7.49 log reduction in virus titers at 10 µM concentration. These data suggest that further investigation of compound 1 as an antiviral therapeutic against VEEV, and perhaps other alphaviruses, is warranted. Experiments suggested that the antiviral activity of compound 1 is directed at an early step in the VEEV replication cycle by blocking viral RNA and protein synthesis.


Asunto(s)
Antivirales/farmacología , Bencilaminas/farmacología , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Encefalomielitis Equina Venezolana/virología , Animales , Antivirales/química , Bencilaminas/química , Línea Celular , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Encefalomielitis Equina Venezolana/tratamiento farmacológico , Ensayos Analíticos de Alto Rendimiento , Humanos , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Células Vero , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
Artículo en Inglés | MEDLINE | ID: mdl-30917980

RESUMEN

Alphaviruses are arthropod-transmitted RNA viruses that can cause arthralgia, myalgia, and encephalitis in humans. Since the role of cellular kinases in alphavirus replication is unknown, we profiled kinetic changes in host kinase abundance and phosphorylation following chikungunya virus (CHIKV) infection of fibroblasts. Based upon the results of this study, we treated CHIKV-infected cells with kinase inhibitors targeting the Src family kinase (SFK)-phosphatidylinositol 3-kinase (PI3K)-AKT-mTORC signaling pathways. Treatment of cells with SFK inhibitors blocked the replication of CHIKV as well as multiple other alphaviruses, including Mayaro virus, O'nyong-nyong virus, Ross River virus, and Venezuelan equine encephalitis virus. Dissecting the effect of SFK inhibition on alphavirus replication, we found that viral structural protein levels were significantly reduced, but synthesis of viral genomic and subgenomic RNAs was unaffected. By measuring the association of viral RNA with polyribosomes, we found that the SFK inhibitor dasatinib blocks alphavirus subgenomic RNA translation. Our results demonstrate a role for SFK signaling in alphavirus subgenomic RNA translation and replication. Targeting host factors involved in alphavirus replication represents an innovative, perhaps paradigm-shifting, strategy for exploring the replication of CHIKV and other alphaviruses while promoting antiviral therapeutic development.


Asunto(s)
Infecciones por Alphavirus/tratamiento farmacológico , Alphavirus/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , Familia-src Quinasas/genética , Alphavirus/genética , Infecciones por Alphavirus/virología , Animales , Antivirales/farmacología , Línea Celular , Chlorocebus aethiops , Genoma Viral/efectos de los fármacos , Genoma Viral/genética , Humanos , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , ARN Viral/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células Vero , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
17.
Vet Immunol Immunopathol ; 197: 24-30, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29475503

RESUMEN

Equine herpesvirus-1 (EHV-1) infection is an important and highly prevalent disease in equine populations worldwide. Previously we have demonstrated that a neuropathogenic strain of EHV-1, T953, suppresses the host cell's antiviral type-I interferon (IFN) response in vitro. Whether or not this is unique to EHV-1 strains possessing the neuropathogenic genotype has been undetermined. Here, we examined whether there is any direct relationship between neuropathogenic genotype and the induced IFN-ß response in equine endothelial cells (EECs) infected with 10 different strains of EHV-1. The extent of virus cell-to-cell spread following infection in EECs was also compared between the neuropathogenic and the non-neuropathogenic genotype of EHV-1. We then compared IFN-ß and the total type-I IFN protein suppression between T953, an EHV-1 strain that is neuropathogenic and T445, an EHV-4 strain mainly associated only with respiratory disease. Data from our study revealed no relationship between the neuropathogenic genotype of EHV-1 and the induced IFN-ß mRNA by the host cell. Results also indicate no statistically significant difference in plaque sizes of both genotypes of EHV-1 produced in EECs. However, while the T953 strain of EHV-1 was able to suppress IFN-ß mRNA and type-I IFN biological activity at 12 h post-infection (hpi), EHV-4 weakly induces both IFN-ß mRNA and type-I IFN biological activity. This finding correlated with a statistically significant difference in the mean plaque sizes produced by the two EHV subtypes in EECs. Our data help illuminate how EHV-1, irrespective of its genotype, evades the host cell's innate immune response thereby enabling viral spread to susceptible cells.


Asunto(s)
Infecciones por Herpesviridae/veterinaria , Herpesvirus Équido 1/patogenicidad , Enfermedades de los Caballos/inmunología , Interferón beta/inmunología , Animales , Células Endoteliales/inmunología , Células Endoteliales/virología , Genotipo , Infecciones por Herpesviridae/inmunología , Herpesvirus Équido 1/clasificación , Enfermedades de los Caballos/virología , Caballos/inmunología , Evasión Inmune , Inmunidad Innata , Interferón Tipo I/inmunología
18.
Virology ; 515: 250-260, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29324290

RESUMEN

Mannose binding lectin (MBL) generally plays a protective role during viral infection, yet MBL-mediated complement activation promotes Ross River virus (RRV)-induced inflammatory tissue destruction, contributing to arthritis and myositis. As MBL binds to carbohydrates, we hypothesized that N-linked glycans on the RRV envelope glycoproteins act as ligands for MBL. Using a panel of RRV mutants lacking the envelope N-linked glycans, we found that MBL deposition onto infected cells was dependent on the E2 glycans. Moreover, the glycan-deficient viruses exhibited reduced disease and tissue damage in a mouse model of RRV-induced myositis compared to wild-type RRV, despite similar viral load and inflammatory infiltrates within the skeletal muscle. Instead, the reduced disease induced by glycan-deficient viruses was linked to decreased MBL deposition and complement activation within inflamed tissues. These results demonstrate that the viral N-linked glycans promote MBL deposition and complement activation onto RRV-infected cells, contributing to the development of RRV-induced myositis.


Asunto(s)
Infecciones por Alphavirus/inmunología , Proteínas del Sistema Complemento/inmunología , Polisacáridos/inmunología , Virus del Río Ross/inmunología , Proteínas del Envoltorio Viral/inmunología , Infecciones por Alphavirus/virología , Animales , Activación de Complemento , Modelos Animales de Enfermedad , Humanos , Lectina de Unión a Manosa/inmunología , Ratones Endogámicos C57BL , Polisacáridos/química , Virus del Río Ross/genética , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
19.
Zootaxa ; 4236(1): zootaxa.4236.1.10, 2017 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-28264345

RESUMEN

Two new genera and two new species of eriophyoid mites viz., Propeaciota genusetosis n. gen. and n. sp. infesting Acer sp. (Aceraceae) and Spinaephyes alnus n. gen. and n. sp. infesting Alnus nepalensis D. Don (Betulaceae) are described in the tribe Tegonotini (Eriophyidae: Phyllocoptinae) from North Bengal, India. Aciota secundum Flechtmann et al.1995 is re-assigned (n. comb.) to Propeaciota. Relationships of the new genera with other eriophyoid genera are discussed.


Asunto(s)
Ácaros , Animales , India
20.
PLoS Genet ; 12(12): e1006467, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27930647

RESUMEN

Equine arteritis virus (EAV) is the causative agent of equine viral arteritis (EVA), a respiratory, systemic, and reproductive disease of horses and other equid species. Following natural infection, 10-70% of the infected stallions can become persistently infected and continue to shed EAV in their semen for periods ranging from several months to life. Recently, we reported that some stallions possess a subpopulation(s) of CD3+ T lymphocytes that are susceptible to in vitro EAV infection and that this phenotypic trait is associated with long-term carrier status following exposure to the virus. In contrast, stallions not possessing the CD3+ T lymphocyte susceptible phenotype are at less risk of becoming long-term virus carriers. A genome wide association study (GWAS) using the Illumina Equine SNP50 chip revealed that the ability of EAV to infect CD3+ T lymphocytes and establish long-term carrier status in stallions correlated with a region within equine chromosome 11. Here we identified the gene and mutations responsible for these phenotypes. Specifically, the work implicated three allelic variants of the equine orthologue of CXCL16 (EqCXCL16) that differ by four non-synonymous nucleotide substitutions (XM_00154756; c.715 A → T, c.801 G → C, c.804 T → A/G, c.810 G → A) within exon 1. This resulted in four amino acid changes with EqCXCL16S (XP_001504806.1) having Phe, His, Ile and Lys as compared to EqCXL16R having Tyr, Asp, Phe, and Glu at 40, 49, 50, and 52, respectively. Two alleles (EqCXCL16Sa, EqCXCL16Sb) encoded identical protein products that correlated strongly with long-term EAV persistence in stallions (P<0.000001) and are required for in vitro CD3+ T lymphocyte susceptibility to EAV infection. The third (EqCXCL16R) was associated with in vitro CD3+ T lymphocyte resistance to EAV infection and a significantly lower probability for establishment of the long-term carrier state (viral persistence) in the male reproductive tract. EqCXCL16Sa and EqCXCL16Sb exert a dominant mode of inheritance. Most importantly, the protein isoform EqCXCL16S but not EqCXCL16R can function as an EAV cellular receptor. Although both molecules have equal chemoattractant potential, EqCXCL16S has significantly higher scavenger receptor and adhesion properties compared to EqCXCL16R.


Asunto(s)
Infecciones por Arterivirus/genética , Quimiocinas CXC/genética , Equartevirus/genética , Enfermedades de los Caballos/genética , Alelos , Secuencia de Aminoácidos/genética , Animales , Infecciones por Arterivirus/veterinaria , Infecciones por Arterivirus/virología , Complejo CD3/genética , Complejo CD3/inmunología , Equartevirus/patogenicidad , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Enfermedades de los Caballos/virología , Caballos/genética , Caballos/virología , Masculino , Filogenia , Semen/metabolismo , Linfocitos T/inmunología , Linfocitos T/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...