Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Virol ; 98(6): e0164123, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38690874

RESUMEN

Numerous viruses have been found to exploit glycoconjugates expressed on human cells as their initial attachment factor for viral entry and infection. The virus-cell glycointeractome, when characterized, may serve as a template for antiviral drug design. Heparan sulfate proteoglycans extensively decorate the human cell surface and were previously described as a primary receptor for human metapneumovirus (HMPV). After respiratory syncytial virus, HMPV is the second most prevalent respiratory pathogen causing respiratory tract infection in young children. To date, there is neither vaccine nor drug available to prevent or treat HMPV infection. Using a multidisciplinary approach, we report for the first time the glycointeractome of the HMPV fusion (F) protein, a viral surface glycoprotein that is essential for target-cell recognition, attachment, and entry. Our glycan microarray and surface plasmon resonance results suggest that Galß1-3/4GlcNAc moieties that may be sialylated or fucosylated are readily recognized by HMPV F. The bound motifs are highly similar to the N-linked and O-linked glycans primarily expressed on the human lung epithelium. We demonstrate that the identified glycans have the potential to compete with the cellular receptors used for HMPV entry and consequently block HMPV infection. We found that lacto-N-neotetraose demonstrated the strongest HMPV binding inhibition in a cell infection assay. Our current findings offer an encouraging and novel avenue for the design of anti-HMPV drug candidates using oligosaccharide templates.IMPORTANCEAll cells are decorated with a dense coat of sugars that makes a sugar code. Many respiratory viruses exploit this sugar code by binding to these sugars to cause infection. Human metapneumovirus is a leading cause for acute respiratory tract infections. Despite its medical importance, there is no vaccine or antiviral drug available to prevent or treat human metapneumovirus infection. This study investigates how human metapneumovirus binds to sugars in order to more efficiently infect the human host. We found that human metapneumovirus binds to a diverse range of sugars and demonstrated that these sugars can ultimately block viral infection. Understanding how viruses can take advantage of the sugar code on our cells could identify new intervention and treatment strategies to combat viral disease.


Asunto(s)
Metapneumovirus , Infecciones por Paramyxoviridae , Polisacáridos , Metapneumovirus/metabolismo , Metapneumovirus/fisiología , Humanos , Polisacáridos/metabolismo , Infecciones por Paramyxoviridae/virología , Infecciones por Paramyxoviridae/metabolismo , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Acoplamiento Viral , Unión Proteica , Receptores Virales/metabolismo , Línea Celular
2.
Front Immunol ; 13: 970750, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36045682

RESUMEN

Retinoic acid-inducible gene I (RIG-I) is a receptor that senses viral RNA and interacts with mitochondrial antiviral signaling (MAVS) protein, leading to the production of type I interferons and inflammatory cytokines to establish an antiviral state. This signaling axis is initiated by the K63-linked RIG-I ubiquitination, mediated by E3 ubiquitin ligases such as TRIM25. However, many viruses, including several members of the family Paramyxoviridae and human respiratory syncytial virus (HRSV), a member of the family Pneumoviridae, escape the immune system by targeting RIG-I/TRIM25 signaling. In this study, we screened human metapneumovirus (HMPV) open reading frames (ORFs) for their ability to block RIG-I signaling reconstituted in HEK293T cells by transfection with TRIM25 and RIG-I CARD (an N-terminal CARD domain that is constitutively active in RIG-I signaling). HMPV M2-2 was the most potent inhibitor of RIG-I/TRIM25-mediated interferon (IFN)-ß activation. M2-2 silencing induced the activation of transcription factors (IRF and NF-kB) downstream of RIG-I signaling in A549 cells. Moreover, M2-2 inhibited RIG-I ubiquitination and CARD-dependent interactions with MAVS. Immunoprecipitation revealed that M2-2 forms a stable complex with RIG-I CARD/TRIM25 via direct interaction with the SPRY domain of TRIM25. Similarly, HRSV NS1 also formed a stable complex with RIG-I CARD/TRIM25 and inhibited RIG-I ubiquitination. Notably, the inhibitory actions of HMPV M2-2 and HRSV NS1 are similar to those of V proteins of several members of the Paramyxoviridae family. In this study, we have identified a novel mechanism of immune escape by HMPV, similar to that of Pneumoviridae and Paramyxoviridae family members.


Asunto(s)
Interferón Tipo I , Metapneumovirus , Infecciones por Paramyxoviridae/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Antivirales , Proteína 58 DEAD Box/metabolismo , Células HEK293 , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Interferón beta/metabolismo , Paramyxoviridae , Infecciones por Paramyxoviridae/virología , Receptores Inmunológicos/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
3.
J Clin Invest ; 131(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34609969

RESUMEN

The capacity of respiratory viruses to undergo evolution within the respiratory tract raises the possibility of evolution under the selective pressure of the host environment or drug treatment. Long-term infections in immunocompromised hosts are potential drivers of viral evolution and development of infectious variants. We showed that intrahost evolution in chronic human parainfluenza virus 3 (HPIV3) infection in immunocompromised individuals elicited mutations that favored viral entry and persistence, suggesting that similar processes may operate across enveloped respiratory viruses. We profiled longitudinal HPIV3 infections from 2 immunocompromised individuals that persisted for 278 and 98 days. Mutations accrued in the HPIV3 attachment protein hemagglutinin-neuraminidase (HN), including the first in vivo mutation in HN's receptor binding site responsible for activating the viral fusion process. Fixation of this mutation was associated with exposure to a drug that cleaves host-cell sialic acid moieties. Longitudinal adaptation of HN was associated with features that promote viral entry and persistence in cells, including greater avidity for sialic acid and more active fusion activity in vitro, but not with antibody escape. Long-term infection thus led to mutations promoting viral persistence, suggesting that host-directed therapeutics may support the evolution of viruses that alter their biophysical characteristics to persist in the face of these agents in vivo.


Asunto(s)
Huésped Inmunocomprometido , Enfermedades Pulmonares/virología , Pulmón/virología , Virus de la Parainfluenza 3 Humana/metabolismo , Infecciones por Paramyxoviridae/virología , Adulto , Sitios de Unión , Análisis Mutacional de ADN , Femenino , Frecuencia de los Genes , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Células HEK293 , Humanos , Leucemia Mieloide Aguda , Mutación , Ácido Micofenólico/administración & dosificación , Ácido N-Acetilneuramínico/química , Virus de la Parainfluenza 3 Humana/genética , Infecciones por Paramyxoviridae/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/virología , Receptores Virales/metabolismo , Sirolimus/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Adulto Joven
4.
J Immunother Cancer ; 9(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34172515

RESUMEN

BACKGROUND: There is intense interest in developing novel oncolytic viruses, which can be used in cancer therapies along with immune cells such as natural killer (NK) cells. We have previously developed a particle-based method for in vitro expansion of highly cytotoxic human NK cells (PM21-NK cells). Here, we have tested the hypothesis that oncolytic parainfluenza virus 5 (P/V virus) can combine with PM21-NK cells for targeted killing of lung cancer cells. METHODS: PM21-NK cells were assayed for killing of P/V virus-infected A549, H1299 and Calu-1 lung cancer cells in two-dimensional (2D) and three-dimensional (3D) cultures using flow cytometry, luminescence and kinetic imaging-based methods. Blocking antibodies were used to evaluate NK cell activating receptors involved in PM21-NK cell killing of infected target cells. Media transfer experiments tested soluble factors that increase PM21-NK cell killing of both P/V virus-infected and uninfected tumor cells. RESULTS: In 2D cultures, PM21-NK cells efficiently killed P/V virus-infected cancer cells compared with non-infected cells, through involvement of the viral glycoprotein and NK cell receptors NKp30, NKp46 and NKG2D. In 3D spheroid cultures, P/V virus infection was restricted to the outer layer of the spheroid. However, PM21-NK cells were able to more efficiently kill both the outer layer of infected cells in the spheroid and progressing further to kill the uninfected interior cells. Media transfer experiments demonstrated that P/V virus infection produced both type I and type III interferons, which decreased cell growth, which contributed to a reduction in the overall number of uninfected tumor cells in conjunction with PM21-NK cells. Across five cancer cell lines, the contribution of P/V virus infection on PM21-NK cell killing of target cells correlated with interferon induction. CONCLUSION: Our data support the potential of combining oncolytic parainfluenza virus with PM21-NK cell adoptive therapy against lung cancer.


Asunto(s)
Células Asesinas Naturales/metabolismo , Neoplasias Pulmonares/virología , Virus Oncolíticos/metabolismo , Infecciones por Paramyxoviridae/metabolismo , Esferoides Celulares/metabolismo , Humanos , Imagenología Tridimensional , Interferón Tipo I , Interferones , Neoplasias Pulmonares/inmunología , Transducción de Señal , Interferón lambda
5.
J Virol ; 95(18): e0059321, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34160259

RESUMEN

Human metapneumovirus (hMPV) is an important cause of acute viral respiratory infection. As the only target of neutralizing antibodies, the hMPV fusion (F) protein has been a major focus for vaccine development and targeting by drugs and monoclonal antibodies (MAbs). While X-ray structures of trimeric prefusion and postfusion hMPV F proteins from genotype A, and monomeric prefusion hMPV F protein from genotype B have been determined, structural data for the postfusion conformation for genotype B is lacking. We determined the crystal structure of this protein and compared the structural differences of postfusion hMPV F between hMPV A and B genotypes. We also assessed the receptor binding properties of the hMPV F protein to heparin and heparan sulfate (HS). A library of HS oligomers was used to verify the HS binding activity of hMPV F, and several compounds showed binding to predominantly prefusion hMPV F, but had limited binding to postfusion hMPV F. Furthermore, MAbs to antigenic sites III and the 66-87 intratrimeric epitope block heparin binding. In addition, we evaluated the efficacy of postfusion hMPV B2 F protein as a vaccine candidate in BALB/c mice. Mice immunized with hMPV B2 postfusion F protein showed a balanced Th1/Th2 immune response and generated neutralizing antibodies against both subgroup A2 and B2 hMPV strains, which protected the mice from hMPV challenge. Antibody competition analysis revealed the antibodies generated by immunization target two known antigenic sites (III and IV) on the hMPV F protein. Overall, this study provides new characteristics of the hMPV F protein, which may be informative for vaccine and therapy development. IMPORTANCE Human metapneumovirus (hMPV) is an important cause of viral respiratory disease. In this paper, we report the X-ray crystal structure of the hMPV fusion (F) protein in the postfusion conformation from genotype B. We also assessed binding of the hMPV F protein to heparin and heparan sulfate, a previously reported receptor for the hMPV F protein. Furthermore, we determined the immunogenicity and protective efficacy of postfusion hMPV B2 F protein, which is the first study using a homogenous conformation of the protein. Antibodies generated in response to vaccination give a balanced Th1/Th2 response and target two previously discovered neutralizing epitopes.


Asunto(s)
Anticuerpos Antivirales/inmunología , Epítopos/inmunología , Heparina/metabolismo , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Femenino , Heparina/análogos & derivados , Humanos , Inmunización , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Paramyxoviridae/metabolismo , Infecciones por Paramyxoviridae/virología , Unión Proteica , Conformación Proteica , Proteoglicanos/metabolismo , Células TH1/inmunología , Células Th2/inmunología , Proteínas Virales de Fusión/metabolismo
6.
Mediators Inflamm ; 2019: 4964239, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31686982

RESUMEN

Human metapneumovirus (HMPV) may cause severe respiratory disease. The early innate immune response to viruses like HMPV is characterized by induction of antiviral interferons (IFNs) and proinflammatory immune mediators that are essential in shaping adaptive immune responses. Although innate immune responses to HMPV have been comprehensively studied in mice and murine immune cells, there is less information on these responses in human cells, comparing different cell types infected with the same HMPV strain. The aim of this study was to characterize the HMPV-induced mRNA expression of critical innate immune mediators in human primary cells relevant for airway disease. In particular, we determined type I versus type III IFN expression in human epithelial cells and monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs). In epithelial cells, HMPV induced only low levels of IFN-ß mRNA, while a robust mRNA expression of IFN-λs was found in epithelial cells, MDMs, and MDDCs. In addition, we determined induction of the interferon regulatory factors (IRFs) IRF1, IRF3, and IRF7 and critical inflammatory cytokines (IL-6, IP-10, and IL-1ß). Interestingly, IRF1 mRNA was predominantly induced in MDMs and MDDCs. Overall, our results suggest that for HMPV infection of MDDCs, MDMs, NECs, and A549 cells (the cell types examined), cell type is a strong determinator of the ability of HMPV to induce different innate immune mediators. HMPV induces the transcription of IFN-ß and IRF1 to higher extents in MDMs and MDDCs than in A549s and NECs, whereas the induction of type III IFN-λ and IRF7 is considerable in MDMs, MDDCs, and A549 epithelial cells.


Asunto(s)
Inmunidad Innata/fisiología , Metapneumovirus/patogenicidad , Infecciones por Paramyxoviridae/inmunología , Células A549 , Células Cultivadas , Quimiocina CXCL10/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunidad Innata/genética , Interleucina-6/metabolismo , Macrófagos/metabolismo , Metapneumovirus/inmunología , Microscopía Confocal , Infecciones por Paramyxoviridae/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal
7.
Adv Med Sci ; 64(2): 338-343, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31022559

RESUMEN

PURPOSE: Respiratory viral infection and nonsteroidal anti-inflammatory drugs (NSAIDs) may affect arachidonic acid (AA) metabolism in the airway epithelium, however their joint effect has not been studied. We hypothesized, that alternations of AA metabolism in human airway epithelial cells (ECs) - induced by Parainfluenza virus type 3 (PIV3) - may be modified by concomitant treatment with NSAIDs. MATERIALS AND METHODS: Nasal (RPMI 2650) and bronchial (BEAS-2B) epithelial cells were cultured into confluence and then infected with PIV3. Prostaglandin E2 (PGE2) and 15-hydroxyeicosatetraenoic acid (15-HETE) levels in cell supernatants were measured by ELISA and expression of cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), 5-lipoxygenase (5-LO) and 15-lipoxygenase (15-LO) mRNA in cells was evaluated after reverse transcription with real-time polymerase chain reactions. RESULTS: PGE2 generation was decreased by PIV3 infection in the upper airway epithelial cells, and increased in the lower airway epithelial cells. Both naproxen and celecoxib induced significant reduction in PGE2 release in both infected and non-infected upper and lower airway epithelial cells. However, in PIV3-infected epithelial cells celecoxib inhibited PGE2 release and COX-2 expression to significantly higher degree as compared to non-infected cells. 15-HETE generation or COX-1, 5-LO and 15-LO expression were not affected by the virus infection or by NSAIDs. CONCLUSION: Virus infection in airway epithelial cells enhances inhibitory effect of NSAIDs on prostaglandin E2 generation.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Infecciones por Paramyxoviridae/metabolismo , Celecoxib/farmacología , Línea Celular , Células Epiteliales , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Virology ; 531: 183-191, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30927711

RESUMEN

Human metapneumovirus (hMPV) is an important cause of acute lower respiratory tract infections in infants, elderly and immunocompromised individuals. Ingenuity pathway analysis of microarrays data showed that 20% of genes affected by hMPV infection of airway epithelial cells (AECs) were related to metabolism. We found that levels of the glycolytic pathway enzymes hexokinase 2, pyruvate kinase M2, and lactate dehydrogenase A were significantly upregulated in normal human AECs upon hMPV infection, as well as levels of enzymes belonging to the hexosamine biosynthetic and glycosylation pathways. On the other hand, expression of the majority of the enzymes belonging to the tricarboxylic acid cycle was significantly diminished. Inhibition of hexokinase 2 and of the glycosylating enzyme O-linked N-acetylglucosamine transferase led to a significant reduction in hMPV titer, indicating that metabolic changes induced by hMPV infection play a major role during the virus life cycle, and could be explored as potential antiviral targets.


Asunto(s)
Células Epiteliales/metabolismo , Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/metabolismo , Mucosa Respiratoria/metabolismo , Línea Celular , Células Epiteliales/virología , Glucólisis , Hexosaminas/biosíntesis , Humanos , Redes y Vías Metabólicas , Metapneumovirus/genética , Fosforilación Oxidativa , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/fisiopatología , Infecciones por Paramyxoviridae/virología , Mucosa Respiratoria/virología , Replicación Viral
9.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29321315

RESUMEN

Paramyxovirus V proteins are known antagonists of the RIG-I-like receptor (RLR)-mediated interferon induction pathway, interacting with and inhibiting the RLR MDA5. We report interactions between the Nipah virus V protein and both RIG-I regulatory protein TRIM25 and RIG-I. We also observed interactions between these host proteins and the V proteins of measles virus, Sendai virus, and parainfluenza virus. These interactions are mediated by the conserved C-terminal domain of the V protein, which binds to the tandem caspase activation and recruitment domains (CARDs) of RIG-I (the region of TRIM25 ubiquitination) and to the SPRY domain of TRIM25, which mediates TRIM25 interaction with the RIG-I CARDs. Furthermore, we show that V interaction with TRIM25 and RIG-I prevents TRIM25-mediated ubiquitination of RIG-I and disrupts downstream RIG-I signaling to the mitochondrial antiviral signaling protein. This is a novel mechanism for innate immune inhibition by paramyxovirus V proteins, distinct from other known V protein functions such as MDA5 and STAT1 antagonism.IMPORTANCE The host RIG-I signaling pathway is a key early obstacle to paramyxovirus infection, as it results in rapid induction of an antiviral response. This study shows that paramyxovirus V proteins interact with and inhibit the activation of RIG-I, thereby interrupting the antiviral signaling pathway and facilitating virus replication.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Infecciones por Paramyxoviridae/metabolismo , Paramyxoviridae/fisiología , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Células A549 , Animales , Proteína 58 DEAD Box/genética , Perros , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Infecciones por Paramyxoviridae/genética , Receptores Inmunológicos , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética , Proteínas Virales/genética
10.
PLoS Pathog ; 11(12): e1005303, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26629703

RESUMEN

Human metapneumovirus (HMPV), a member of the Paramyxoviridae family, is a leading cause of lower respiratory illness. Although receptor binding is thought to initiate fusion at the plasma membrane for paramyxoviruses, the entry mechanism for HMPV is largely uncharacterized. Here we sought to determine whether HMPV initiates fusion at the plasma membrane or following internalization. To study the HMPV entry process in human bronchial epithelial (BEAS-2B) cells, we used fluorescence microscopy, an R18-dequenching fusion assay, and developed a quantitative, fluorescence microscopy assay to follow virus binding, internalization, membrane fusion, and visualize the cellular site of HMPV fusion. We found that HMPV particles are internalized into human bronchial epithelial cells before fusing with endosomes. Using chemical inhibitors and RNA interference, we determined that HMPV particles are internalized via clathrin-mediated endocytosis in a dynamin-dependent manner. HMPV fusion and productive infection are promoted by RGD-binding integrin engagement, internalization, actin polymerization, and dynamin. Further, HMPV fusion is pH-independent, although infection with rare strains is modestly inhibited by RNA interference or chemical inhibition of endosomal acidification. Thus, HMPV can enter via endocytosis, but the viral fusion machinery is not triggered by low pH. Together, our results indicate that HMPV is capable of entering host cells by multiple pathways, including membrane fusion from endosomal compartments.


Asunto(s)
Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/metabolismo , Mucosa Respiratoria/virología , Internalización del Virus , Bronquios/virología , Línea Celular , Endosomas/metabolismo , Citometría de Flujo , Humanos , Microscopía Confocal , ARN Interferente Pequeño , Transfección , Proteínas Virales de Fusión/metabolismo
11.
J Virol ; 89(10): 5557-68, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25740991

RESUMEN

UNLABELLED: Hydrogen sulfide (H2S) is an endogenous gaseous mediator that has gained increasing recognition as an important player in modulating acute and chronic inflammatory diseases. However, its role in virus-induced lung inflammation is currently unknown. Respiratory syncytial virus (RSV) is a major cause of upper and lower respiratory tract infections in children for which no vaccine or effective treatment is available. Using the slow-releasing H2S donor GYY4137 and propargylglycin (PAG), an inhibitor of cystathionine-γ-lyase (CSE), a key enzyme that produces intracellular H2S, we found that RSV infection led to a reduced ability to generate and maintain intracellular H2S levels in airway epithelial cells (AECs). Inhibition of CSE with PAG resulted in increased viral replication and chemokine secretion. On the other hand, treatment of AECs with the H2S donor GYY4137 reduced proinflammatory mediator production and significantly reduced viral replication, even when administered several hours after viral absorption. GYY4137 also significantly reduced replication and inflammatory chemokine production induced by human metapneumovirus (hMPV) and Nipah virus (NiV), suggesting a broad inhibitory effect of H2S on paramyxovirus infections. GYY4137 treatment had no effect on RSV genome replication or viral mRNA and protein synthesis, but it inhibited syncytium formation and virus assembly/release. GYY4137 inhibition of proinflammatory gene expression occurred by modulation of the activation of the key transcription factors nuclear factor κB (NF-κB) and interferon regulatory factor 3 (IRF-3) at a step subsequent to their nuclear translocation. H2S antiviral and immunoregulatory properties could represent a novel treatment strategy for paramyxovirus infections. IMPORTANCE: RSV is a global health concern, causing significant morbidity and economic losses as well as mortality in developing countries. After decades of intensive research, no vaccine or effective treatment, with the exception of immunoprophylaxis, is available for this infection as well as for other important respiratory mucosal viruses. This study identifies hydrogen sulfide as a novel cellular mediator that can modulate viral replication and proinflammatory gene expression, both important determinants of lung injury in respiratory viral infections, with potential for rapid translation of such findings into novel therapeutic approaches for viral bronchiolitis and pneumonia.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Infecciones por Paramyxoviridae/metabolismo , Alquinos/farmacología , Línea Celular , Quimiocinas/biosíntesis , Quimiocinas/genética , Cistationina gamma-Liasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Glicina/análogos & derivados , Glicina/farmacología , Humanos , Mediadores de Inflamación/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Morfolinas/farmacología , FN-kappa B/metabolismo , Compuestos Organotiofosforados/farmacología , Infecciones por Paramyxoviridae/tratamiento farmacológico , Infecciones por Paramyxoviridae/etiología , Regiones Promotoras Genéticas , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/fisiología , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
12.
Eur J Immunol ; 45(6): 1680-95, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25763996

RESUMEN

Human metapneumovirus (hMPV) is a leading cause of acute respiratory tract infections in children and the elderly. The mechanism by which this virus triggers an inflammatory response still remains unknown. Here, we evaluated whether the thymic stromal lymphopoietin (TSLP) pathway contributes to lung inflammation upon hMPV infection. We found that hMPV infection promotes TSLP expression both in human airway epithelial cells and in the mouse lung. hMPV infection induced lung infiltration of OX40L(+) CD11b(+) DCs. Mice lacking the TSLP receptor deficient mice (tslpr(-/-) ) showed reduced lung inflammation and hMPV replication. These mice displayed a decreased number of neutrophils as well a reduction in levels of thymus and activation-regulated chemokine/CCL17, IL-5, IL-13, and TNF-α in the airways upon hMPV infection. Furthermore, a higher frequency of CD4(+) and CD8(+) T cells was found in tslpr(-/-) mice compared to WT mice, which could contribute to controlling viral spread. Depletion of neutrophils in WT and tslpr(-/-) mice decreased inflammation and hMPV replication. Remarkably, blockage of TSLP or OX40L with specific Abs reduced lung inflammation and viral replication following hMPV challenge in mice. Altogether, these results suggest that activation of the TSLP pathway is pivotal in the development of pulmonary pathology and pulmonary hMPV replication.


Asunto(s)
Citocinas/metabolismo , Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/metabolismo , Infecciones por Paramyxoviridae/virología , Neumonía Viral/metabolismo , Neumonía Viral/virología , Transducción de Señal , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Línea Celular , Citocinas/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/virología , Expresión Génica , Humanos , Interleucina-33 , Interleucina-8/genética , Interleucina-8/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Metapneumovirus/efectos de los fármacos , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Ligando OX40/antagonistas & inhibidores , Ligando OX40/genética , Ligando OX40/metabolismo , Infecciones por Paramyxoviridae/tratamiento farmacológico , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/patología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/genética , Neumonía Viral/patología , Receptores de Citocinas/antagonistas & inhibidores , Receptores de Citocinas/deficiencia , Transducción de Señal/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Replicación Viral , Linfopoyetina del Estroma Tímico
13.
Virus Res ; 200: 19-23, 2015 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-25645280

RESUMEN

Human metapneumovirus (hMPV) is a major cause of respiratory tract infections in children, elderly and immunocompromised hosts, for which no vaccine or treatment are currently available. Oxidative stress and inflammatory responses represent important pathogenic mechanism(s) of hMPV infection. Here, we explored the potential protective role of dietary antioxidants in hMPV infection. Treatment of airway epithelial cells with resveratrol and quercetin during hMPV infection significantly reduced cellular oxidative damage, inflammatory mediator secretion and viral replication, without affecting viral gene transcription and protein synthesis, indicating that inhibition of viral replication occurred at the level of viral assembly and/or release. Modulation of proinflammatory mediator expression occurred through the inhibition of transcription factor nuclear factor (NF)-κB and interferon regulatory factor (IRF)-3 binding to their cognate site of endogenous gene promoters. Our results indicate the use of dietary antioxidants as an effective treatment approach for modulating hMPV induced lung oxidative damage and inflammation.


Asunto(s)
Antioxidantes/farmacología , Suplementos Dietéticos/análisis , Metapneumovirus/efectos de los fármacos , Infecciones por Paramyxoviridae/virología , Línea Celular , Citocinas/genética , Citocinas/inmunología , Humanos , Metapneumovirus/genética , Metapneumovirus/fisiología , Estrés Oxidativo/efectos de los fármacos , Infecciones por Paramyxoviridae/tratamiento farmacológico , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/metabolismo , Quercetina/farmacología , Resveratrol , Estilbenos/farmacología , Replicación Viral/efectos de los fármacos
14.
Am J Respir Cell Mol Biol ; 51(4): 494-501, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24735073

RESUMEN

Respiratory viral infections are associated with the majority of asthma attacks. Inhibitory M2 receptors on parasympathetic nerves, which normally limit acetylcholine (ACh) release, are dysfunctional after respiratory viral infection. Because IL-1ß is up-regulated during respiratory viral infections, we investigated whether IL-1ß mediates M2 receptor dysfunction during parainfluenza virus infection. Virus-infected guinea pigs were pretreated with the IL-1ß antagonist anakinra. In the absence of anakinra, viral infection increased bronchoconstriction in response to vagal stimulation but not to intravenous ACh, and neuronal M2 muscarinic receptors were dysfunctional. Pretreatment with anakinra prevented virus-induced increased bronchoconstriction and M2 receptor dysfunction. Anakinra did not change smooth muscle M3 muscarinic receptor response to ACh, lung viral loads, or blood and bronchoalveolar lavage leukocyte populations. Respiratory virus infection decreased M2 receptor mRNA expression in parasympathetic ganglia extracted from infected animals, and this was prevented by blocking IL-1ß or TNF-α. Treatment of SK-N-SH neuroblastoma cells or primary cultures of guinea pig parasympathetic neurons with IL-1ß directly decreased M2 receptor mRNA, and this was not synergistic with TNF-α treatment. Treating guinea pig trachea segment with TNF-α or IL-1ß in vitro increased tracheal contractions in response to activation of airway nerves by electrical field stimulation. Blocking IL-1ß during TNF-α treatment prevented this hyperresponsiveness. These data show that virus-induced hyperreactivity and M2 dysfunction involves IL-1ß and TNF-α, likely in sequence with TNF-α causing production of IL-1ß.


Asunto(s)
Hiperreactividad Bronquial/metabolismo , Broncoconstricción , Interleucina-1beta/metabolismo , Pulmón/metabolismo , Infecciones por Paramyxoviridae/metabolismo , Paramyxoviridae/patogenicidad , Receptor Muscarínico M2/metabolismo , Infecciones del Sistema Respiratorio/metabolismo , Animales , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/fisiopatología , Hiperreactividad Bronquial/prevención & control , Hiperreactividad Bronquial/virología , Pruebas de Provocación Bronquial , Broncoconstricción/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Cobayas , Interacciones Huésped-Patógeno , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/inervación , Pulmón/fisiopatología , Pulmón/virología , Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/fisiopatología , Infecciones por Paramyxoviridae/virología , Sistema Nervioso Parasimpático/inmunología , Sistema Nervioso Parasimpático/metabolismo , Sistema Nervioso Parasimpático/fisiopatología , Sistema Nervioso Parasimpático/virología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/fisiopatología , Infecciones del Sistema Respiratorio/virología , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
15.
Curr Opin Virol ; 5: 24-33, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24530984

RESUMEN

The paramyxoviruses represent a diverse virus family responsible for a wide range of human and animal diseases. In contrast to other viruses, such as HIV and influenza virus, which use a single glycoprotein to mediate host receptor binding and virus entry, the paramyxoviruses require two distinct proteins. One of these is an attachment glycoprotein that binds receptor, while the second is a fusion glycoprotein, which undergoes conformational changes that drive virus-cell membrane fusion and virus entry. The details of how receptor binding by one protein activates the second to undergo conformational changes have been poorly understood until recently. Over the past couple of years, structural and functional data have accumulated on representative members of this family, including parainfluenza virus 5, Newcastle disease virus, measles virus, Nipah virus and others, which suggest a mechanistic convergence of activation models. Here we review the data indicating that paramyxovirus attachment glycoproteins shield activating residues within their N-terminal stalk domains, which are then exposed upon receptor binding, leading to the activation of the fusion protein by a 'provocateur' mechanism.


Asunto(s)
Infecciones por Paramyxoviridae/metabolismo , Infecciones por Paramyxoviridae/virología , Paramyxoviridae/metabolismo , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Animales , Humanos , Fusión de Membrana , Paramyxoviridae/química , Paramyxoviridae/genética , Infecciones por Paramyxoviridae/genética , Receptores Virales/química , Receptores Virales/genética , Receptores Virales/metabolismo , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética
16.
J Virol ; 88(8): 4338-52, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24478423

RESUMEN

UNLABELLED: Human metapneumovirus (hMPV) is a relatively recently identified paramyxovirus that causes acute upper and lower respiratory tract infection. Entry of hMPV is unusual among the paramyxoviruses, in that fusion is accomplished by the fusion (F) protein without the attachment glycoprotein (G protein). It has been suggested that hMPV F protein utilizes integrin αvß1 as a cellular receptor. Consistent with this, the F proteins of all known hMPV strains possess an integrin-binding motif ((329)RGD(331)). The role of this motif in viral entry, infectivity, and pathogenesis is poorly understood. Here, we show that α5ß1 and αv integrins are essential for cell-cell fusion and hMPV infection. Mutational analysis found that residues R329 and G330 in the (329)RGD(331) motif are essential for cell-cell fusion, whereas mutations at D331 did not significantly impact fusion activity. Furthermore, fusion-defective RGD mutations were either lethal to the virus or resulted in recombinant hMPVs that had defects in viral replication in cell culture. In cotton rats, recombinant hMPV with the R329K mutation in the F protein (rhMPV-R329K) and rhMPV-D331A exhibited significant defects in viral replication in nasal turbinates and lungs. Importantly, inoculation of cotton rats with these mutants triggered a high level of neutralizing antibodies and protected against hMPV challenge. Taken together, our data indicate that (i) α5ß1 and αv integrins are essential for cell-cell fusion and viral replication, (ii) the first two residues in the RGD motif are essential for fusion activity, and (iii) inhibition of the interaction of the integrin-RGD motif may serve as a new target to rationally attenuate hMPV for the development of live attenuated vaccines. IMPORTANCE: Human metapneumovirus (hMPV) is one of the major causative agents of acute respiratory disease in humans. Currently, there is no vaccine or antiviral drug for hMPV. hMPV enters host cells via a unique mechanism, in that viral fusion (F) protein mediates both attachment and fusion activity. Recently, it was suggested that hMPV F protein utilizes integrins as receptors for entry via a poorly understood mechanism. Here, we show that α5ß1 and αv integrins are essential for hMPV infectivity and F protein-mediated cell-cell fusion and that the integrin-binding motif in the F protein plays a crucial role in these functions. Our results also identify the integrin-binding motif to be a new, attenuating target for the development of a live vaccine for hMPV. These findings not only will facilitate the development of antiviral drugs targeting viral entry steps but also will lead to the development new live attenuated vaccine candidates for hMPV.


Asunto(s)
Integrina alfa5beta1/metabolismo , Integrina alfaV/metabolismo , Metapneumovirus/fisiología , Metapneumovirus/patogenicidad , Infecciones por Paramyxoviridae/metabolismo , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Animales , Femenino , Humanos , Integrina alfa5beta1/genética , Integrina alfaV/genética , Metapneumovirus/genética , Mutación Missense , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Unión Proteica , Ratas , Sigmodontinae , Proteínas Virales de Fusión/genética , Virulencia
17.
J Gen Virol ; 95(Pt 1): 26-37, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24114793

RESUMEN

The host cytokine IL-6 plays an important role in host defence and prevention of lung injury from various pathogens, making IL-6 an important mediator in the host's susceptibility to respiratory infections. The cellular response to IL-6 is mediated through a Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) signal transduction pathway. Human metapneumovirus (hMPV) is an important causative agent of viral respiratory infections known to inhibit the IFN-mediated activation of STAT1. However, little is known about the interactions between this virus and other STAT signalling cascades. Herein, we showed that hMPV can attenuate the IL-6-mediated JAK/STAT3 signalling cascade in lung epithelial cells. HMPV inhibited a key event in this pathway by impeding the phosphorylation and nuclear translocation of STAT3 in A549 cells and in primary normal human bronchial epithelial cells. Further studies established that hMPV interrupted the IL-6-induced JAK/STAT pathway early in the signal transduction pathway by blocking the phosphorylation of JAK2. By antagonizing the IL-6-mediated JAK/STAT3 pathway, hMPV perturbed the expression of IL-6-inducible genes important for apoptosis, cell differentiation and growth. Infection with hMPV also differentially regulated the effects of IL-6 on apoptosis. Thus, hMPV regulation of these genes could usurp the protective roles of IL-6, and these data provide insight into an important element of viral pathogenesis.


Asunto(s)
Células Epiteliales/virología , Interleucina-6/metabolismo , Janus Quinasa 2/metabolismo , Pulmón/metabolismo , Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulación hacia Abajo , Células Epiteliales/metabolismo , Interacciones Huésped-Patógeno , Humanos , Interleucina-6/genética , Janus Quinasa 2/genética , Pulmón/citología , Pulmón/virología , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Factor de Transcripción STAT3/genética
18.
J Clin Invest ; 122(8): 2967-82, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22797302

RESUMEN

Viruses are leading causes of severe acute lower respiratory infections (LRIs). These infections evoke incomplete immunity, as individuals can be repeatedly reinfected throughout life. We report that acute viral LRI causes rapid pulmonary CD8+ cytotoxic T lymphocyte (TCD8) functional impairment via programmed death-1/programmed death ligand-1 (PD-1/PD-L1) signaling, a pathway previously associated with prolonged antigenic stimulation during chronic infections and cancer. PD-1-mediated TCD8 impairment occurred acutely in mice following infection with human metapneumovirus or influenza virus. Viral antigen was sufficient for PD-1 upregulation, but induction of PD-L1 was required for impairment. During secondary viral infection or epitope-only challenge, memory TCD8 rapidly reexpressed PD-1 and exhibited severe functional impairment. Inhibition of PD-1 signaling using monoclonal antibody blockade prevented TCD8 impairment, reduced viral titers during primary infection, and enhanced protection of immunized mice against challenge infection. Additionally, PD-1 and PD-L1 were upregulated in the lungs of patients with 2009 H1N1 influenza virus, respiratory syncytial virus, or parainfluenza virus infection. These results indicate that PD-1 mediates TCD8 functional impairment during acute viral infection and may contribute to recurrent viral LRIs. Therefore, the PD-1/PD-L1 pathway may represent a therapeutic target in the treatment of respiratory viruses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/metabolismo , Virosis/inmunología , Virosis/metabolismo , Enfermedad Aguda , Animales , Antígenos Virales , Antígeno HLA-B7/genética , Humanos , Imidazoles , Memoria Inmunológica , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/inmunología , Gripe Humana/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Metapneumovirus , Ratones , Ratones Transgénicos , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Piridinas , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitial Respiratorio Humano , Transducción de Señal , Regulación hacia Arriba
19.
J Virol ; 86(10): 5730-41, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22438532

RESUMEN

The hemagglutinin-neuraminidase (HN) protein of paramyxoviruses carries out three distinct activities contributing to the ability of HN to promote viral fusion and entry: receptor binding, receptor cleavage (neuraminidase), and activation of the fusion protein. The relationship between receptor binding and fusion triggering functions of HN are not fully understood. For Newcastle disease virus (NDV), one bifunctional site (site I) on HN's globular head can mediate both receptor binding and neuraminidase activities, and a second site (site II) in the globular head is also capable of mediating receptor binding. The receptor analog, zanamivir, blocks receptor binding and cleavage activities of NDV HN's site I while activating receptor binding by site II. Comparison of chimeric proteins in which the globular head of NDV HN is connected to the stalk region of either human parainfluenza virus type 3 (HPIV3) or Nipah virus receptor binding proteins indicates that receptor binding to NDV HN site II not only can activate its own fusion (F) protein but can also activate the heterotypic fusion proteins. We suggest a general model for paramyxovirus fusion activation in which receptor engagement at site II plays an active role in F activation.


Asunto(s)
Proteína HN/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Infecciones por Paramyxoviridae/metabolismo , Paramyxovirinae/metabolismo , Receptores Virales/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , Sitios de Unión , Proteínas Portadoras , Línea Celular , Proteína HN/química , Proteína HN/genética , Humanos , Virus de la Enfermedad de Newcastle/química , Virus de la Enfermedad de Newcastle/enzimología , Virus de la Enfermedad de Newcastle/genética , Infecciones por Paramyxoviridae/virología , Paramyxovirinae/genética , Estructura Terciaria de Proteína , Proteínas Virales de Fusión/metabolismo , Proteínas Virales/química , Proteínas Virales/genética
20.
J Virol ; 86(6): 3230-43, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22238303

RESUMEN

Human metapneumovirus (HMPV) is a major worldwide respiratory pathogen that causes acute upper and lower respiratory tract disease. The mechanism by which this virus recognizes and gains access to its target cell is still largely unknown. In this study, we addressed the initial steps in virus binding and infection and found that the first binding partner for HMPV is heparan sulfate (HS). While wild-type CHO-K1 cells are permissive to HMPV infection, mutant cell lines lacking the ability to synthesize glycosaminoglycans (GAGs), specifically, heparan sulfate proteoglycans (HSPGs), were resistant to binding and infection by HMPV. The permissiveness to HMPV infection was also abolished when CHO-K1 cells were treated with heparinases. Importantly, using recombinant HMPV lacking both the G and small hydrophobic (SH) proteins, we report that this first virus-cell binding interaction is driven primarily by the fusion protein (HMPV F) and that this interaction is needed to establish a productive infection. Finally, HMPV binding to cells did not require ß1 integrin expression, and RGD-mediated interactions were not essential in promoting HMPV F-mediated cell-to-cell membrane fusion. Cells lacking ß1 integrin, however, were less permissive to HMPV infection, indicating that while ß1 integrins play an important role in promoting HMPV infection, the interaction between integrins and HMPV occurs after the initial binding of HMPV F to heparan sulfate proteoglycans.


Asunto(s)
Heparitina Sulfato/metabolismo , Metapneumovirus/metabolismo , Infecciones por Paramyxoviridae/metabolismo , Receptores Virales/metabolismo , Proteínas Virales de Fusión/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Humanos , Metapneumovirus/química , Metapneumovirus/genética , Infecciones por Paramyxoviridae/virología , Unión Proteica , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA