Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
PLoS Pathog ; 18(4): e1010469, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35486576

RESUMEN

Dengue virus (DENV) which infects about 390 million people per year in tropical and subtropical areas manifests various disease symptoms, ranging from fever to life-threatening hemorrhage and even shock. To date, there is still no effective treatment for DENV disease, but only supportive care. DENV nonstructural protein 1 (NS1) has been shown to play a key role in disease pathogenesis. Recent studies have shown that anti-DENV NS1 antibody can provide disease protection by blocking the DENV-induced disruption of endothelial integrity. We previously demonstrated that anti-NS1 monoclonal antibody (mAb) protected mice from all four serotypes of DENV challenge. Here, we generated humanized anti-NS1 mAbs and transferred them to mice after DENV infection. The results showed that DENV-induced prolonged bleeding time and skin hemorrhage were reduced, even several days after DENV challenge. Mechanistic studies showed the ability of humanized anti-NS1 mAbs to inhibit NS1-induced vascular hyperpermeability and to elicit Fcγ-dependent complement-mediated cytolysis as well as antibody-dependent cellular cytotoxicity of cells infected with four serotypes of DENV. These results highlight humanized anti-NS1 mAb as a potential therapeutic agent in DENV infection.


Asunto(s)
Virus del Dengue , Dengue , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Dengue/prevención & control , Modelos Animales de Enfermedad , Hemorragia/etiología , Humanos , Ratones , Proteínas no Estructurales Virales/metabolismo
2.
Int J Mol Sci ; 21(24)2020 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-33352639

RESUMEN

Dengue virus (DENV) infection is a significant public health threat in tropical and subtropical regions; however, there is no specific antiviral drug. Accumulated studies have revealed that DENV infection induces several cellular responses, including autophagy and apoptosis. The crosstalk between autophagy and apoptosis is associated with the interactions among components of these two pathways, such as apoptotic caspase-mediated cleavage of autophagy-related proteins. Here, we show that DENV-induced autophagy inhibits early cell apoptosis and hence enhances DENV replication. Later, the apoptotic activities are elevated to suppress autophagy through cleavage of Beclin-1, an essential autophagy-related protein. Inhibition of cleavage of Beclin-1 by a pan-caspase inhibitor, Z-VAD, increases both autophagy and viral replication. Regarding the mechanism, we further found that DENV nonstructural protein 1 (NS1) is able to interact with Beclin-1 during DENV infection. The interaction between Beclin-1 and NS1 attenuates Beclin-1 cleavage and facilitates autophagy to prevent cell apoptosis. Our study suggests a novel mechanism whereby NS1 preserves Beclin-1 for maintaining autophagy to antagonize early cell apoptosis; however, elevated caspases trigger apoptosis by degrading Beclin-1 in the late stage of infection. These findings suggest implications for anti-DENV drug design.


Asunto(s)
Beclina-1/metabolismo , Caspasas/metabolismo , Virus del Dengue/aislamiento & purificación , Dengue/patología , Dengue/virología , Proteínas no Estructurales Virales/metabolismo , Células A549 , Aedes , Animales , Autofagia , Dengue/metabolismo , Humanos
3.
Artículo en Inglés | MEDLINE | ID: mdl-29581121

RESUMEN

Group A Streptococcus (GAS) is an important human pathogen that causes a wide spectrum of diseases, including necrotizing fasciitis and streptococcal toxic shock syndrome. Dextromethorphan (DM), an antitussive drug, has been demonstrated to efficiently reduce inflammatory responses, thereby contributing to an increased survival rate of GAS-infected mice. However, the anti-inflammatory mechanisms underlying DM treatment in GAS infection remain unclear. DM is known to exert neuroprotective effects through an NADPH oxidase-dependent regulated process. In the present study, membrane translocation of NADPH oxidase subunit p47phox and subsequent reactive oxygen species (ROS) generation induced by GAS infection were significantly inhibited via DM treatment in RAW264.7 murine macrophage cells. Further determination of proinflammatory mediators revealed that DM effectively suppressed inducible nitric oxide synthase (iNOS) expression and NO, tumor necrosis factor alpha, and interleukin-6 generation in GAS-infected RAW264.7 cells as well as in air-pouch-infiltrating cells from GAS/DM-treated mice. GAS infection caused AKT dephosphorylation, glycogen synthase kinase-3ß (GSK-3ß) activation, and subsequent NF-κB nuclear translocation, which were also markedly inhibited by treatment with DM and an NADPH oxidase inhibitor, diphenylene iodonium. These results suggest that DM attenuates GAS infection-induced overactive inflammation by inhibiting NADPH oxidase-mediated ROS production that leads to downregulation of the GSK-3ß/NF-κB/NO signaling pathway.


Asunto(s)
Dextrometorfano/uso terapéutico , Óxido Nítrico Sintasa de Tipo II/metabolismo , Infecciones Estreptocócicas/tratamiento farmacológico , Infecciones Estreptocócicas/enzimología , Animales , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Compuestos Onio/farmacología , Oxidación-Reducción/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Infecciones Estreptocócicas/metabolismo , Células THP-1
4.
Mediators Inflamm ; 2013: 720689, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23533310

RESUMEN

Group A streptococcus (GAS) imposes a great burden on humans. Efforts to minimize the associated morbidity and mortality represent a critical issue. Glycogen synthase kinase-3 ß (GSK-3 ß) is known to regulate inflammatory response in infectious diseases. However, the regulation of GSK-3 ß in GAS infection is still unknown. The present study investigates the interaction between GSK-3 ß , NF- κ B, and possible related inflammatory mediators in vitro and in a mouse model. The results revealed that GAS could activate NF- κ B, followed by an increased expression of inducible nitric oxide synthase (iNOS) and NO production in a murine macrophage cell line. Activation of GSK-3 ß occurred after GAS infection, and inhibition of GSK-3 ß reduced iNOS expression and NO production. Furthermore, GSK-3 ß inhibitors reduced NF- κ B activation and subsequent TNF- α production, which indicates that GSK-3 ß acts upstream of NF- κ B in GAS-infected macrophages. Similar to the in vitro findings, administration of GSK-3 ß inhibitor in an air pouch GAS infection mouse model significantly reduced the level of serum TNF- α and improved the survival rate. The inhibition of GSK-3 ß to moderate the inflammatory effect might be an alternative therapeutic strategy against GAS infection.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Western Blotting , Línea Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Ensayo de Inmunoadsorción Enzimática , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Inmunohistoquímica , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo II , Infecciones Estreptocócicas
5.
Front Immunol ; 10: 2147, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31620121

RESUMEN

Thioredoxin-interacting protein (Txnip) inhibits the activity of thioredoxin (Trx) to modulate inflammatory responses. The burden of inflammation caused by microbial infection is strongly associated with disease severity; however, the role of Txnip in bacterial infection remains unclear. In Group A Streptococcus (GAS)-infected macrophages, Txnip was degraded independent of glucose consumption and streptococcal cysteine protease expression. Treatment with proteasome inhibitors reversed GAS-induced Txnip degradation. The activation of Toll-like receptor 2 (TLR2) initiated Txnip degradation, while no further Txnip degradation was observed in TLR2-deficient bone marrow-derived macrophages. NADPH oxidase-regulated NF-κB activation and pro-inflammatory activation were induced and accompanied by Txnip degradation during GAS infection. Silencing Txnip prompted TLR2-mediated inducible nitric oxide synthase (iNOS)/NO, TNF-α, and IL-6 production whereas the blockage of Txnip degradation by pharmacologically inhibiting the HECT E3 ubiquitin ligase with heclin and AMP-dependent protein kinase with dorsomorphin effectively reduced such effects. Our findings reveal that TLR2/NADPH oxidase-mediated Txnip proteasomal degradation facilitates pro-inflammatory cytokine production during GAS infection.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamación/metabolismo , Infecciones Estreptocócicas/metabolismo , Tiorredoxinas/metabolismo , Receptor Toll-Like 2/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas Portadoras/inmunología , Inflamación/inmunología , Ratones , Células RAW 264.7 , Infecciones Estreptocócicas/inmunología , Tiorredoxinas/inmunología , Ubiquitina-Proteína Ligasas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA