Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Gen Virol ; 105(2)2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38421278

RESUMEN

Background. Chikungunya virus (CHIKV) causes chikungunya fever and has been responsible for major global epidemics of arthritic disease over the past two decades. Multiple CHIKV vaccine candidates are currently undergoing or have undergone human clinical trials, with one vaccine candidate receiving FDA approval. This scoping review was performed to evaluate the 'efficacy', 'safety' and 'duration of protection' provided by CHIKV vaccine candidates in human clinical trials.Methods. This scoping literature review addresses studies involving CHIKV vaccine clinical trials using available literature on the PubMed, Medline Embase, Cochrane Library and Clinicaltrial.gov databases published up to 25 August 2023. Covidence software was used to structure information and review the studies included in this article.Results. A total of 1138 studies were screened and, after removal of duplicate studies, 12 relevant studies were thoroughly reviewed to gather information. This review summarizs that all seven CHIKV vaccine candidates achieved over 90 % seroprotection against CHIKV after one or two doses. All vaccines were able to provide neutralizing antibody protection for at least 28 days.Conclusions. A variety of vaccine technologies have been used to develop CHIKV vaccine candidates. With one vaccine candidate having recently received FDA approval, it is likely that further CHIKV vaccines will be available commercially in the near future.


Asunto(s)
Fiebre Chikungunya , Virus Chikungunya , Vacunas Virales , Humanos , Vacunas Virales/efectos adversos , Fiebre Chikungunya/prevención & control , Anticuerpos Neutralizantes , Bases de Datos Factuales
2.
PLoS Pathog ; 18(2): e1010185, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35143591

RESUMEN

Arthritogenic alphaviruses are mosquito-borne viruses that are a major cause of infectious arthropathies worldwide, and recent outbreaks of chikungunya virus and Ross River virus (RRV) infections highlight the need for robust intervention strategies. Alphaviral arthritis can persist for months after the initial acute disease, and is mediated by cellular immune responses. A common strategy to limit inflammation and pathology is to dampen the overwhelming inflammatory responses by modulating proinflammatory cytokine pathways. Here, we investigate the contribution of interleukin-17 (IL-17), a cytokine involved in arthropathies such as rheumatoid arthritis, in the development RRV-induced arthritis and myositis. IL-17 was quantified in serum from RRV-infected patients, and mice were infected with RRV and joints and muscle tissues collected to analyse cellular infiltrates, tissue mRNA, cytokine expression, and joint and muscle histopathology. IL-17 expression was increased in musculoskeletal tissues and serum of RRV-infected mice and humans, respectively. IL-17-producing T cells and neutrophils contributed to the cellular infiltrate in the joint and muscle tissue during acute RRV disease in mice. Blockade of IL-17A/F using a monoclonal antibody (mAb) reduced disease severity in RRV-infected mice and led to decreased proinflammatory proteins, cellular infiltration in synovial tissues and cartilage damage, without affecting viral titers in inflamed tissues. IL-17A/F blockade triggered a shift in transcriptional profile of both leukocyte infiltrates and musculoskeletal stromal cells by downregulating proinflammatory genes. This study highlights a previously uncharacterized role for an effector cytokine in alphaviral pathology and points towards potential therapeutic benefit in targeting IL-17 to treat patients presenting with RRV-induced arthropathy.


Asunto(s)
Artritis Reumatoide/inmunología , Inmunidad Celular , Inflamación/inmunología , Interleucina-17/inmunología , Miositis/inmunología , Virus del Río Ross/inmunología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Animales , Artritis Reumatoide/virología , Chlorocebus aethiops , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miositis/virología , Células Vero , Carga Viral
3.
Phytopathology ; 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38427607

RESUMEN

The image-based detection and classification of plant diseases has become increasingly important to the development of precision agriculture. We consider the case of tomato, a high-value crop supporting the livelihoods of many farmers around the world. Many biotic and abiotic plant health issues impede the efficient production of this crop, and laboratory-based diagnostics are inaccessible in many remote regions. Early detection of these plant health issues is essential for efficient and accurate response, prompting exploration of alternatives for field detection. Considering the availability of low-cost smartphones, artificial intelligence-based classification facilitated by mobile phone imagery can be a practical option. This study introduces a smartphone-attachable 30x microscopic lens, used to produce the novel tomato microimaging dataset of 8500 images representing 34 tomato plant conditions on the upper and lower sides of leaves as well as on the surface of tomato fruits. We introduce TOMMicroNet, a 14-layer convolutional neural network (CNN) trained to classify amongst biotic and abiotic plant health issues, and we compare it against six existing pre-trained CNN models. We compared two separate pipelines of grouping data for training TOMMicroNet, either presenting all data at once or separating into subsets based on the three parts of the plant. Comparing configurations based on cross-validation and F1 scores, we determined that TOMMicroNet attained the highest performance when trained on the complete dataset, with 95% classification accuracy on both training and external datasets. Given TOMMicroNet's capabilities when presented with unfamiliar data, this approach has the potential for the identification of plant health issues.

4.
J Virol ; 96(17): e0099922, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-36000846

RESUMEN

Arthritogenic alphaviruses are mosquito-borne arboviruses that include several re-emerging human pathogens, including the chikungunya (CHIKV), Ross River (RRV), Mayaro (MAYV), and o'nyong-nyong (ONNV) virus. Arboviruses are transmitted via a mosquito bite to the skin. Herein, we describe intradermal RRV infection in a mouse model that replicates the arthritis and myositis seen in humans with Ross River virus disease (RRVD). We show that skin infection with RRV results in the recruitment of inflammatory monocytes and neutrophils, which together with dendritic cells migrate to draining lymph nodes (LN) of the skin. Neutrophils and monocytes are productively infected and traffic virus from the skin to LN. We show that viral envelope N-linked glycosylation is a key determinant of skin immune responses and disease severity. RRV grown in mammalian cells elicited robust early antiviral responses in the skin, while RRV grown in mosquito cells stimulated poorer early antiviral responses. We used glycan mass spectrometry to characterize the glycan profile of mosquito and mammalian cell-derived RRV, showing deglycosylation of the RRV E2 glycoprotein is associated with curtailed skin immune responses and reduced disease following intradermal infection. Altogether, our findings demonstrate skin infection with an arthritogenic alphavirus leads to musculoskeletal disease and envelope glycoprotein glycosylation shapes disease outcome. IMPORTANCE Arthritogenic alphaviruses are transmitted via mosquito bites through the skin, potentially causing debilitating diseases. Our understanding of how viral infection starts in the skin and how virus systemically disseminates to cause disease remains limited. Intradermal arbovirus infection described herein results in musculoskeletal pathology, which is dependent on viral envelope N-linked glycosylation. As such, intradermal infection route provides new insights into how arboviruses cause disease and could be extended to future investigations of skin immune responses following infection with other re-emerging arboviruses.


Asunto(s)
Infecciones por Alphavirus , Artritis , Miositis , Polisacáridos , Virus del Río Ross , Piel , Infecciones por Alphavirus/complicaciones , Infecciones por Alphavirus/inmunología , Animales , Antivirales/inmunología , Artritis/complicaciones , Artritis/inmunología , Culicidae/virología , Células Dendríticas , Modelos Animales de Enfermedad , Glicosilación , Humanos , Espectrometría de Masas , Ratones , Monocitos , Miositis/complicaciones , Miositis/inmunología , Neutrófilos , Polisacáridos/química , Polisacáridos/inmunología , Virus del Río Ross/inmunología , Piel/inmunología , Piel/virología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología
5.
J Gen Virol ; 102(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33410734

RESUMEN

The complement alternative pathway (AP) is tightly regulated and changes in two important AP components, factor B (FB) and factor H (FH) are linked to severe dengue in humans. Here, a mouse model of dengue was investigated to define the changes in FB and FH and assess the utility of this model to study the role of the AP in severe dengue. Throughout the period of viremia in the AG129 IFN signalling-deficient mouse, an increase in FB and a decrease in FH was observed following dengue virus (DENV) infection, with the former only seen in a model of more severe disease associated with antibody-dependent enhancement (ADE). Terminal disease was associated with a decrease in FB and FH, with greater changes during ADE, and accompanied by increased C3 degradation consistent with complement activation. In silico analysis of NFκΒ, signal transducer and activator of transcription (STAT) and IFN-driven FB and FH promoter elements to reflect the likely impact of the lack of IFN-responses in AG129 mice, demonstrated that these elements differed markedly between human and mouse, notably with mouse FH lacking NFκΒ and key IFN-stimulated response elements (ISRE), and FB with many more NFκΒ and STAT-responsive elements than human FB. Thus, the AG129 mouse offers utility in demonstrating changes in FB and FH that, similar to humans, are associated with severe disease, but lack predicted important human-specific and IFN-dependent responses of FB and FH to DENV-infection that are likely to regulate the subtleties of the overall AP response during dengue disease in humans.


Asunto(s)
Factor B del Complemento/metabolismo , Factor H de Complemento/metabolismo , Vía Alternativa del Complemento , Dengue/inmunología , Dengue Grave/inmunología , Animales , Acrecentamiento Dependiente de Anticuerpo , Factor B del Complemento/genética , Factor H de Complemento/genética , Dengue/virología , Virus del Dengue/inmunología , Virus del Dengue/fisiología , Modelos Animales de Enfermedad , Humanos , Interferones/metabolismo , Ratones , Regiones Promotoras Genéticas , Dengue Grave/virología , Viremia
6.
J Virol ; 94(8)2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-31996431

RESUMEN

Ross River virus (RRV) belongs to the genus Alphavirus and is prevalent in Australia. RRV infection can cause arthritic symptoms in patients and may include rash, fever, arthralgia, and myalgia. Type I interferons (IFN) are the primary antiviral cytokines and trigger activation of the host innate immune system to suppress the replication of invading viruses. Alphaviruses are able to subvert the type I IFN system, but the mechanisms used are ill defined. In this study, seven RRV field strains were analyzed for induction of and sensitivity to type I IFN. The sensitivities of these strains to human IFN-ß varied significantly and were highest for the RRV 2548 strain. Compared to prototype laboratory strain RRV-T48, RRV 2548 also induced higher type I IFN levels both in vitro and in vivo and caused milder disease. To identify the determinants involved in type I IFN modulation, the region encoding the nonstructural proteins (nsPs) of RRV 2548 was sequenced, and 42 amino acid differences from RRV-T48 were identified. Using fragment swapping and site-directed mutagenesis, we discovered that substitutions E402A and R522Q in nsP1 as well as Q619R in nsP2 were responsible for increased sensitivity of RRV 2548 to type I IFN. In contrast, substitutions A31T, N219T, S580L, and Q619R in nsP2 led to induction of higher levels of type I IFN. With exception of E402A, all these variations are common for naturally occurring RRV strains. However, they are different from all known determinants of type I IFN modulation reported previously in nsPs of alphaviruses.IMPORTANCE By identifying natural Ross River virus (RRV) amino acid determinants for type I interferon (IFN) modulation, this study gives further insight into the mechanism of type I IFN modulation by alphaviruses. Here, the crucial role of type I IFN in the early stages of RRV disease pathogenesis is further demonstrated. This study also provides a comparison of the roles of different parts of the RRV nonstructural region in type I IFN modulation, highlighting the importance of nonstructural protein 1 (nsP1) and nsP2 in this process. Three substitutions in nsP1 and nsP2 were found to be independently associated with enhanced type I IFN sensitivity, and four independent substitutions in nsP2 were important in elevated type I IFN induction. Such evidence has clear implications for RRV immunobiology, persistence, and pathology. The identification of viral proteins that modulate type I IFN may also have importance for the pathogenesis of other alphaviruses.


Asunto(s)
Antivirales/farmacología , Interferón Tipo I/inmunología , Interferón Tipo I/farmacología , Virus del Río Ross/efectos de los fármacos , Virus del Río Ross/inmunología , Alphavirus/genética , Alphavirus/inmunología , Infecciones por Alphavirus/virología , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Citocinas , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Virus del Río Ross/genética , Células Vero , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Virulencia , Replicación Viral/efectos de los fármacos
7.
J Gen Virol ; 101(6): 622-634, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375993

RESUMEN

Zika virus (ZIKV) has recently emerged as an important human pathogen due to the strong evidence that it causes disease of the central nervous system, particularly microcephaly and Guillain-Barré syndrome. The pathogenesis of disease, including mechanisms of neuroinvasion, may include both invasion via the blood-brain barrier and via peripheral (including cranial) nerves. Cellular responses to infection are also poorly understood. This study characterizes the in vitro infection of laboratory-adapted ZIKV African MR766 and two Asian strains of (1) brain endothelial cells (hCMEC/D3 cell line) and (2) olfactory ensheathing cells (OECs) (the neuroglia populating cranial nerve I and the olfactory bulb; both human and mouse OEC lines) in comparison to kidney epithelial cells (Vero cells, in which ZIKV infection is well characterized). Readouts included infection kinetics, intracellular virus localization, viral persistence and cytokine responses. Although not as high as in Vero cells, viral titres exceeded 104 plaque-forming units (p.f.u.) ml-1 in the endothelial/neuroglial cell types, except hOECs. Despite these substantial titres, a relatively small proportion of neuroglial cells were primarily infected. Immunolabelling of infected cells revealed localization of the ZIKV envelope and NS3 proteins in the cytoplasm; NS3 staining overlapped with that of dsRNA replication intermediate and the endoplasmic reticulum (ER). Infected OECs and endothelial cells produced high levels of pro-inflammatory chemokines. Nevertheless, ZIKV was also able to establish persistent infection in hOEC and hCMEC/D3 cells. Taken together, these results provide basic insights into ZIKV infection of endothelial and neuroglial cells and will form the basis for further study of ZIKV disease mechanisms.


Asunto(s)
Encéfalo/virología , Células Endoteliales/virología , Neuroglía/virología , Infección por el Virus Zika/virología , Virus Zika/patogenicidad , Animales , Barrera Hematoencefálica/virología , Línea Celular , Chlorocebus aethiops , Retículo Endoplásmico/genética , Humanos , Ratones , Células Vero , Replicación Viral/genética
8.
PLoS Pathog ; 14(1): e1006835, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29377936

RESUMEN

Virus reprogramming of cellular metabolism is recognised as a critical determinant for viral growth. While most viruses appear to activate central energy metabolism, different viruses have been shown to rely on alternative mechanisms of metabolic activation. Whether related viruses exploit conserved mechanisms and induce similar metabolic changes is currently unclear. In this work we investigate how two alphaviruses, Semliki Forest virus and Ross River virus, reprogram host metabolism and define the molecular mechanisms responsible. We demonstrate that in both cases the presence of a YXXM motif in the viral protein nsP3 is necessary for binding to the PI3K regulatory subunit p85 and for activating AKT. This leads to an increase in glucose metabolism towards the synthesis of fatty acids, although additional mechanisms of metabolic activation appear to be involved in Ross River virus infection. Importantly, a Ross River virus mutant that fails to activate AKT has an attenuated phenotype in vivo, suggesting that viral activation of PI3K/AKT contributes to virulence and disease.


Asunto(s)
Infecciones por Alphavirus/metabolismo , Infecciones por Alphavirus/virología , Alphavirus/fisiología , Glucosa/metabolismo , Interacciones Huésped-Patógeno , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Alphavirus/patogenicidad , Animales , Células Cultivadas , Cricetinae , Activación Enzimática , Glucólisis/fisiología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Virus del Río Ross/fisiología , Virus de los Bosques Semliki/fisiología , Virulencia
10.
PLoS Pathog ; 13(12): e1006788, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29281739

RESUMEN

Chikungunya virus (CHIKV) belongs to a group of mosquito-borne alphaviruses associated with acute and chronic arthropathy, with peripheral and limb joints most commonly affected. Using a mouse model of CHIKV infection and arthritic disease, we show that CHIKV replication and the ensuing foot arthropathy were dramatically reduced when mice were housed at 30°C, rather than the conventional 22°C. The effect was not associated with a detectable fever, but was dependent on type I interferon responses. Bioinformatics analyses of RNA-Seq data after injection of poly(I:C)/jetPEI suggested the unfolded protein response and certain type I interferon responses are promoted when feet are slightly warmer. The ambient temperature thus appears able profoundly to effect anti-viral activity in the periphery, with clear consequences for alphaviral replication and the ensuing arthropathy. These observations may provide an explanation for why alphaviral arthropathies are largely restricted to joints of the limbs and the extremities.


Asunto(s)
Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Artritis Experimental/inmunología , Artritis Experimental/virología , Artritis Infecciosa/inmunología , Artritis Infecciosa/virología , Interferón Tipo I/metabolismo , Infecciones por Alphavirus/patología , Animales , Artritis Experimental/patología , Artritis Infecciosa/patología , Fiebre Chikungunya/inmunología , Fiebre Chikungunya/patología , Fiebre Chikungunya/virología , Virus Chikungunya/inmunología , Virus Chikungunya/patogenicidad , Virus Chikungunya/fisiología , Femenino , Pie , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Virus del Río Ross/inmunología , Virus del Río Ross/patogenicidad , Virus del Río Ross/fisiología , Temperatura , Carga Viral , Replicación Viral/inmunología , Replicación Viral/fisiología
11.
PLoS Pathog ; 13(2): e1006155, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28207896

RESUMEN

Chikungunya virus (CHIKV) is an arthritogenic alphavirus causing epidemics of acute and chronic arthritic disease. Herein we describe a comprehensive RNA-Seq analysis of feet and lymph nodes at peak viraemia (day 2 post infection), acute arthritis (day 7) and chronic disease (day 30) in the CHIKV adult wild-type mouse model. Genes previously shown to be up-regulated in CHIKV patients were also up-regulated in the mouse model. CHIKV sequence information was also obtained with up to ≈8% of the reads mapping to the viral genome; however, no adaptive viral genome changes were apparent. Although day 2, 7 and 30 represent distinct stages of infection and disease, there was a pronounced overlap in up-regulated host genes and pathways. Type I interferon response genes (IRGs) represented up to ≈50% of up-regulated genes, even after loss of type I interferon induction on days 7 and 30. Bioinformatic analyses suggested a number of interferon response factors were primarily responsible for maintaining type I IRG induction. A group of genes prominent in the RNA-Seq analysis and hitherto unexplored in viral arthropathies were granzymes A, B and K. Granzyme A-/- and to a lesser extent granzyme K-/-, but not granzyme B-/-, mice showed a pronounced reduction in foot swelling and arthritis, with analysis of granzyme A-/- mice showing no reductions in viral loads but reduced NK and T cell infiltrates post CHIKV infection. Treatment with Serpinb6b, a granzyme A inhibitor, also reduced arthritic inflammation in wild-type mice. In non-human primates circulating granzyme A levels were elevated after CHIKV infection, with the increase correlating with viral load. Elevated granzyme A levels were also seen in a small cohort of human CHIKV patients. Taken together these results suggest granzyme A is an important driver of arthritic inflammation and a potential target for therapy. TRIAL REGISTRATION: ClinicalTrials.gov NCT00281294.


Asunto(s)
Artritis/virología , Fiebre Chikungunya/genética , Fiebre Chikungunya/inmunología , Granzimas/inmunología , Inflamación/virología , Animales , Virus Chikungunya , Modelos Animales de Enfermedad , Granzimas/análisis , Granzimas/biosíntesis , Humanos , Inmunohistoquímica , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/análisis , Transcriptoma
12.
Immunol Rev ; 268(1): 340-64, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26497532

RESUMEN

Sensitization of the humoral immune response to invading viruses and production of antiviral antibodies forms part of the host antiviral repertoire. Paradoxically, for a number of viral pathogens, under certain conditions, antibodies provide an attractive means of enhanced virus entry and replication in a number of cell types. Known as antibody-dependent enhancement (ADE) of infection, the phenomenon occurs when virus-antibody immunocomplexes interact with cells bearing complement or Fc receptors, promoting internalization of the virus and increasing infection. Frequently associated with exacerbation of viral disease, ADE of infection presents a major obstacle to the prevention of viral disease by vaccination and is thought to be partly responsible for the adverse effects of novel antiviral therapeutics such as intravenous immunoglobulins. There is a growing body of work examining the intracellular signaling pathways and epitopes responsible for mediating ADE, with a view to aiding rational design of antiviral strategies. With in vitro studies also confirming ADE as a feature of infection for a growing number of viruses, challenges remain in understanding the multilayered molecular mechanisms of ADE and its effect on viral pathogenesis.


Asunto(s)
Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , Interacciones Huésped-Patógeno , Inmunomodulación , Receptores Fc/metabolismo , Virosis/inmunología , Virosis/metabolismo , Animales , Epítopos/inmunología , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Receptores Fc/antagonistas & inhibidores , Transducción de Señal , Virosis/terapia , Virosis/virología , Replicación Viral/inmunología , Virus/inmunología
13.
J Gen Virol ; 99(4): 525-535, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29517478

RESUMEN

Low-passage clinical isolates of chikungunya virus (CHIKV) were found to be a mixture of large- and small-plaque viruses, with small-plaque viruses being the predominant species. To investigate the contribution of plaque variants to the pathology of the joint, primary human fibroblast-like synoviocytes (HFLS) were used. Large- and small-plaque viruses were purified from two clinical isolates, CHIKV-031C and CHIKV-033C, and were designated CHIKV-031L and CHIKV-031S and CHIKV-033L and CHIKV-033S, respectively. The replication efficiencies of these viruses in HFLSs were compared and it was found that CHIKV-031S and CHIKV-033S replicated with the highest efficiency, while the parental clinical isolates had the lowest efficiency. Interestingly, the cytopathic effects (CPE) induced by these viruses correlated with neither the efficiency of replication nor the plaque size. The small-plaque viruses and the clinical isolates induced cell death rapidly, while large-plaque viruses induced slow CPE in which only 50 % of the cells in infected cultures were rounded up and detached on day 5 of infection. The production of proinflammatory cytokines and chemokines from infected HFLSs was evaluated. The results showed that the large-plaque viruses and the clinical isolates, but not small-plaque variants, were potent inducers of IL-6, IL-8 and MCP-1, and were able to migrate monocytes/macrophages efficiently. Sequencing data revealed a number of differences in amino acid sequences between the small- and large-plaque viruses. The results suggest that it is common for clinical isolates of CHIKV to be heterogeneous, while the variants may have distinct roles in the pathology of the joint.


Asunto(s)
Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Fibroblastos/virología , Sinoviocitos/virología , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Fiebre Chikungunya/genética , Fiebre Chikungunya/inmunología , Virus Chikungunya/genética , Virus Chikungunya/aislamiento & purificación , Efecto Citopatogénico Viral , Fibroblastos/inmunología , Humanos , Interleucina-6/genética , Interleucina-6/inmunología , Interleucina-8/genética , Interleucina-8/inmunología , Sinoviocitos/inmunología
14.
J Infect Dis ; 215(10): 1536-1545, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28379462

RESUMEN

Background: Human metapneumovirus (hMPV) infection is implicated in exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Research into the pathogenesis of infection is restricted to animal models, and information about hMPV replication and inflammatory and immune responses in human disease is limited. Methods: Human primary bronchial epithelial cells (PBECs) from healthy and asthmatic subjects and those with COPD were infected with hMPV, with or without glucocorticosteroid (GCS) exposure. Viral replication, inflammatory and immune responses, and apoptosis were analyzed. We also determined whether adjuvant interferon (IFN) can blunt hMPV infection in vitro and in a murine model. Results: hMPV infected human PBECs and viral replication was enhanced in cells from patients with COPD. The virus induced gene expression of IFN-stimulated gene 56 (ISG56) and IFN-ß, as well as IFN-γ-inducible protein 10 (IP-10) and regulated on activation, normal T cell expressed and secreted (RANTES), and more so in cells from patients with COPD. GCS exposure enhanced hMPV replication despite increased IFN expression. Augmented virus replication associated with GCS was mediated by reduced apoptosis via induction of antiapoptotic genes. Adjuvant IFN treatment suppressed hMPV replication in PBECs and reduced hMPV viral titers and inflammation in vivo. Conclusions: hMPV infects human PBECs, eliciting innate and inflammatory responses. Replication is enhanced by GCS and adjuvant IFN is an effective treatment, restricting virus replication and proinflammatory consequences of hMPV infections.


Asunto(s)
Glucocorticoides/farmacología , Interferón gamma/farmacología , Metapneumovirus , Infecciones por Paramyxoviridae/virología , Enfermedad Pulmonar Obstructiva Crónica/virología , Animales , Apoptosis/efectos de los fármacos , Asma/virología , Bronquios/citología , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Mucosa Respiratoria/citología , Replicación Viral/efectos de los fármacos
15.
J Gen Virol ; 98(10): 2413-2420, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28874226

RESUMEN

Tembusu virus (TMUV, genus Flavivirus, family Flaviviridae) was first isolated in 1955 from Culex tritaeniorhynchus mosquitoes in Kuala Lumpur, Malaysia. In April 2010, duck TMUV was first identified as the causative agent of egg-drop syndrome, characterized by a substantial decrease in egg laying and depression, growth retardation and neurological signs or death in infected egg-laying and breeder ducks, in the People's Republic of China. Since 2010, duck TMUV has spread to most of the duck-producing regions in China, including many of the coastal provinces, neighbouring regions and certain Southeast Asia areas (i.e. Thailand and Malaysia). This review describes the current understanding of the genome characteristics, host range, transmission, epidemiology, phylogenetic and immune evasion of avian-origin TMUV and the innate immune response of the host.


Asunto(s)
Patos/virología , Infecciones por Flavivirus/transmisión , Infecciones por Flavivirus/veterinaria , Flavivirus/genética , Enfermedades de las Aves de Corral/epidemiología , Animales , China/epidemiología , Culex/virología , Patos/inmunología , Flavivirus/inmunología , Flavivirus/aislamiento & purificación , Infecciones por Flavivirus/virología , Genoma Viral/genética , Especificidad del Huésped , Evasión Inmune , Insectos Vectores/virología , Enfermedades de las Aves de Corral/transmisión , Enfermedades de las Aves de Corral/virología
16.
J Gen Virol ; 98(11): 2712-2724, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29022864

RESUMEN

Zika virus (ZIKV, genus Flavivirus) has emerged as a major mosquito-transmitted human pathogen, with recent outbreaks associated with an increased incidence of neurological complications, particularly microcephaly and the Guillain-Barré syndrome. Because the virus has only very recently emerged as an important pathogen, research is being hampered by a lack of reliable molecular tools. Here we report an infectious cDNA (icDNA) clone for ZIKV isolate BeH819015 from Brazil, which was selected as representative of South American ZIKV isolated at early stages of the outbreak. icDNA clones were assembled from synthetic DNA fragments corresponding to the consensus sequence of the BeH819015 isolate. Virus rescued from the icDNA clone had properties identical to a natural ZIKV isolate from South America. Variants of the clone-derived virus, expressing nanoluciferase, enhanced green fluorescent or mCherry marker proteins in both mammalian and insect cells and being genetically stable for multiple in vitro passages, were obtained. A ZIKV subgenomic replicon, lacking a prM- and E glycoprotein encoding region and expressing a Gaussia luciferase marker, was constructed and shown to replicate both in mammalian and insect cells. In the presence of the Semliki Forest virus replicon, expressing ZIKV structural proteins, the ZIKV replicon was packaged into virus-replicon particles. Efficient reverse genetic systems, genetically stable marker viruses and packaged replicons offer significant improvements for biological studies of ZIKV infection and disease, as well as for the development of antiviral approaches.


Asunto(s)
Genética Inversa/métodos , Virus Zika/genética , Brasil , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Genes Reporteros , Luciferasas/genética , Coloración y Etiquetado/métodos , Virus Zika/aislamiento & purificación
17.
J Virol ; 90(8): 4150-4159, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26865723

RESUMEN

UNLABELLED: The alphaviral6kgene region encodes the two structural proteins 6K protein and, due to a ribosomal frameshift event, the transframe protein (TF). Here, we characterized the role of the6kproteins in the arthritogenic alphavirus Ross River virus (RRV) in infected cells and in mice, using a novel6kin-frame deletion mutant. Comprehensive microscopic analysis revealed that the6kproteins were predominantly localized at the endoplasmic reticulum of RRV-infected cells. RRV virions that lack the6kproteins 6K and TF [RRV-(Δ6K)] were more vulnerable to changes in pH, and the corresponding virus had increased sensitivity to a higher temperature. While the6kdeletion did not reduce RRV particle production in BHK-21 cells, it affected virion release from the host cell. Subsequentin vivostudies demonstrated that RRV-(Δ6K) caused a milder disease than wild-type virus, with viral titers being reduced in infected mice. Immunization of mice with RRV-(Δ6K) resulted in a reduced viral load and accelerated viral elimination upon secondary infection with wild-type RRV or another alphavirus, chikungunya virus (CHIKV). Our results show that the6kproteins may contribute to alphaviral disease manifestations and suggest that manipulation of the6kgene may be a potential strategy to facilitate viral vaccine development. IMPORTANCE: Arthritogenic alphaviruses, such as chikungunya virus (CHIKV) and Ross River virus (RRV), cause epidemics of debilitating rheumatic disease in areas where they are endemic and can emerge in new regions worldwide. RRV is of considerable medical significance in Australia, where it is the leading cause of arboviral disease. The mechanisms by which alphaviruses persist and cause disease in the host are ill defined. This paper describes the phenotypic properties of an RRV6kdeletion mutant. The absence of the6kgene reduced virion release from infected cells and also reduced the severity of disease and viral titers in infected mice. Immunization with the mutant virus protected mice against viremia not only upon exposure to RRV but also upon challenge with CHIKV. These findings could lead to the development of safer and more immunogenic alphavirus vectors for vaccine delivery.


Asunto(s)
Infecciones por Alphavirus/virología , Virus del Río Ross/genética , Virus del Río Ross/inmunología , Proteínas Estructurales Virales/genética , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/fisiopatología , Animales , Línea Celular , Línea Celular Tumoral , Virus Chikungunya/inmunología , Chlorocebus aethiops , Cricetinae , Humanos , Concentración de Iones de Hidrógeno , Ratones , Mutación , Sistemas de Lectura , Virus del Río Ross/patogenicidad , Eliminación de Secuencia , Células Vero , Carga Viral , Proteínas Estructurales Virales/análisis , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Vacunas Virales/inmunología , Replicación Viral
18.
PLoS Pathog ; 11(2): e1004649, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25695775

RESUMEN

The rising prevalence of arthritogenic alphavirus infections, including chikungunya virus (CHIKV) and Ross River virus (RRV), and the lack of antiviral treatments highlight the potential threat of a global alphavirus pandemic. The immune responses underlying alphavirus virulence remain enigmatic. We found that pentraxin 3 (PTX3) was highly expressed in CHIKV and RRV patients during acute disease. Overt expression of PTX3 in CHIKV patients was associated with increased viral load and disease severity. PTX3-deficient (PTX3(-/-)) mice acutely infected with RRV exhibited delayed disease progression and rapid recovery through diminished inflammatory responses and viral replication. Furthermore, binding of the N-terminal domain of PTX3 to RRV facilitated viral entry and replication. Thus, our study demonstrates the pivotal role of PTX3 in shaping alphavirus-triggered immunity and disease and provides new insights into alphavirus pathogenesis.


Asunto(s)
Infecciones por Alphavirus/inmunología , Proteína C-Reactiva/inmunología , Proteínas del Tejido Nervioso/inmunología , Componente Amiloide P Sérico/inmunología , Replicación Viral/inmunología , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma , Transfección , Carga Viral/inmunología
19.
Clin Sci (Lond) ; 131(14): 1713-1721, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28667069

RESUMEN

Since its discovery in 2001, human metapneumovirus (hMPV) has been identified as an important cause of respiratory tract infection in young children, second only to the closely related respiratory syncytial virus (RSV). Clinical evidence suggests that hMPV is associated with acute exacerbations of asthma in both children and adults, and may play a role in initiating asthma development in children. Animal models have demonstrated that airway hyperresponsiveness (AHR) and inflammation are triggered following hMPV infection, and hMPV is able to persist in vivo by inhibiting innate immune responses and causing aberrant adaptive responses. In this review, we discuss the prevalence of hMPV infection in pediatric and adult populations and its potential role in asthma exacerbation. We also review recent advances made in animal models to determine immune responses following hMPV infection, and compare to what is known about RSV.


Asunto(s)
Asma/virología , Metapneumovirus , Infecciones por Paramyxoviridae/complicaciones , Infecciones por Virus Sincitial Respiratorio/complicaciones , Virus Sincitial Respiratorio Humano , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Infecciones por Paramyxoviridae/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología
20.
Proc Natl Acad Sci U S A ; 111(16): 6040-5, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24733914

RESUMEN

Arthritogenic alphaviruses including Ross River virus (RRV), Sindbis virus, and chikungunya virus cause worldwide outbreaks of musculoskeletal disease. The ability of alphaviruses to induce bone pathologies remains poorly defined. Here we show that primary human osteoblasts (hOBs) can be productively infected by RRV. RRV-infected hOBs produced high levels of inflammatory cytokine including IL-6. The RANKL/OPG ratio was disrupted in the synovial fluid of RRV patients, and this was accompanied by an increase in serum Tartrate-resistant acid phosphatase 5b (TRAP5b) levels. Infection of bone cells with RRV was validated using an established RRV murine model. In wild-type mice, infectious virus was detected in the femur, tibia, patella, and foot, together with reduced bone volume in the tibial epiphysis and vertebrae detected by microcomputed tomographic (µCT) analysis. The RANKL/OPG ratio was also disrupted in mice infected with RRV; both this effect and the bone loss were blocked by treatment with an IL-6 neutralizing antibody. Collectively, these findings provide previously unidentified evidence that alphavirus infection induces bone loss and that OBs are capable of producing proinflammatory mediators during alphavirus-induced arthralgia. The perturbed RANKL/OPG ratio in RRV-infected OBs may therefore contribute to bone loss in alphavirus infection.


Asunto(s)
Infecciones por Alphavirus/patología , Infecciones por Alphavirus/virología , Artritis/virología , Resorción Ósea/patología , Resorción Ósea/virología , Osteoblastos/patología , Virus del Río Ross/fisiología , Fosfatasa Ácida/sangre , Adulto , Infecciones por Alphavirus/sangre , Animales , Anticuerpos Neutralizantes/farmacología , Artritis/sangre , Artritis/patología , Resorción Ósea/sangre , Huesos/diagnóstico por imagen , Huesos/patología , Huesos/virología , Femenino , Placa de Crecimiento/efectos de los fármacos , Placa de Crecimiento/patología , Placa de Crecimiento/virología , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-6/biosíntesis , Isoenzimas/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Neutralización , Osteoblastos/efectos de los fármacos , Osteoblastos/virología , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteoclastos/virología , Osteogénesis/efectos de los fármacos , Osteoprotegerina/metabolismo , Fenotipo , Ligando RANK/metabolismo , Virus del Río Ross/efectos de los fármacos , Líquido Sinovial/metabolismo , Fosfatasa Ácida Tartratorresistente , Replicación Viral/efectos de los fármacos , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA