Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
PLoS Pathog ; 20(5): e1012209, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38709723

RESUMEN

[This corrects the article DOI: 10.1371/journal.ppat.1003231.].

2.
PLoS Pathog ; 18(5): e1010505, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35503798

RESUMEN

The Hippo signaling pathway, which is historically considered as a dominator of organ development and homeostasis has recently been implicated as an immune regulator. However, its role in host defense against influenza A virus (IAV) has not been widely investigated. Here, we found that IAV could activate the Hippo effectors Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) through physical binding of the IAV non-structural protein 1 (NS1) with C-terminal domain of YAP/TAZ, facilitating their nuclear location. Meanwhile, YAP/TAZ downregulated the expression of pro-inflammatory and anti-viral cytokines against IAV infection, therefore benefiting virus replication and host cell apoptosis. A mouse model of IAV infection further demonstrated Yap deficiency protected mice against IAV infection, relieving lung injury. Mechanistically, YAP/TAZ blocked anti-viral innate immune signaling via downregulation of Toll-like receptor 3 (TLR3) expression. YAP directly bound to the putative TEADs binding site on the promoter region of TLR3. The elimination of acetylated histone H3 occupancy in the TLR3 promoter resulted in its transcriptional silence. Moreover, treatment of Trichostatin A, a histone deacetylases (HDACs) inhibitor or disruption of HDAC4/6 reversed the inhibition of TLR3 expression by YAP/TAZ, suggesting HDAC4/6 mediated the suppression function of YAP/TAZ. Taken together, we uncovered a novel immunomodulatory mechanism employed by IAV, where YAP/TAZ antagonize TLR3-mediated innate immunity.


Asunto(s)
Virus de la Influenza A , Receptor Toll-Like 3 , Proteínas no Estructurales Virales/metabolismo , Animales , Inmunidad Innata , Virus de la Influenza A/metabolismo , Ratones , Transducción de Señal , Factores de Transcripción/metabolismo
3.
J Biol Chem ; 298(5): 101850, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35339492

RESUMEN

Pyroptosis is an inflammatory form of programmed cell death that is executed by the gasdermin (GSDM)-N domain of GSDM family proteins, which form pores in the plasma membrane. Although pyroptosis acts as a host defense against invasive pathogen infection, its role in the pathogenesis of enterovirus 71 (EV71) infection is unclear. In the current study, we found that EV71 infection induces cleavage of GSDM E (GSDME) by using western blotting analysis, an essential step in the switch from caspase-3-mediated apoptosis to pyroptosis. We show that this cleavage is independent of the 3C and 2A proteases of EV71. However, caspase-3 activation is essential for this cleavage, as GSDME could not be cleaved in caspase-3-KO cells upon EV71 infection. Further analyses showed that EV71 infection induced pyroptosis in WT cells but not in caspase-3/GSDME double-KO cells. Importantly, GSDME is required to induce severe disease during EV71 infection, as GSDME deficiency in mice was shown to alleviate pathological symptoms. In conclusion, our results reveal that GSDME is important for the pathogenesis of EV71 via mediating initiation of pyroptosis.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Proteínas Citotóxicas Formadoras de Poros , Piroptosis , Animales , Apoptosis , Caspasa 3/genética , Caspasa 3/metabolismo , Muerte Celular , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/metabolismo , Humanos , Ratones , Proteínas Citotóxicas Formadoras de Poros/metabolismo
4.
Environ Res ; 214(Pt 3): 113964, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35944621

RESUMEN

In this study, activation of peroxymonosulfate (PMS) by amorphous FeOOH to degrade sulfamethoxazole (SMX) was investigated. The amorphous FeOOH showed a better performance in the decomposition of PMS and the degradation of SMX than the crystallized α-FeOOH and ß-FeOOH. The quenching experiments and EPR measurements suggested that the mechanism of PMS activation by amorphous FeOOH was mainly the surface-bound radicals (●OH and SO4●-). Basically, the surface-bound ●OH radicals were the dominate reactive oxide species in this system, which were mainly generated via the decomposition of amorphous FeOOH-PMS complexes. The degradation of SMX was significantly inhibited with the presence of H2PO4-, and this adverse impact was negligibly affected by the increase of H2PO4- concentration, implying that the inhibition of SMX degradation was caused by competitive adsorption. Consequently, the Fe-OH bonds on the amorphous FeOOH were proposed as the reactive sites for forming amorphous FeOOH-PMS complexes. Besides, the amorphous FeOOH showed a better performance in the degradation of SMX in the acid conditions than that in the base conditions due to the surface charge of amorphous FeOOH. More importantly, the reduction efficiency of Fe(III) was significantly enhanced due to the excellent conductivity of amorphous FeOOH.


Asunto(s)
Sulfametoxazol , Contaminantes Químicos del Agua , Electrones , Compuestos Férricos , Radical Hidroxilo/química , Peróxidos , Contaminantes Químicos del Agua/química
5.
Chemistry ; 27(18): 5761-5768, 2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33469957

RESUMEN

Nanoscale composites for high-performance electrodes employed in flexible, all-solid-state supercapacitors are being developed. A series of binder-free composites, each consisting of a transition bimetal oxide, a metal oxide, and a metal nitride grown on N-doped reduced graphene oxide (rGO)-wrapped nickel foam are obtained by using a universal strategy. Three different transition metals, Co, Mo, and Fe, are separately compounded with nickel ions, which originate from the nickel foam, to form three composites, NiCoO2 @Co3 O4 @Co2 N, NiMoO4 @MoO3 @Mo2 N, and NiFe2 O4 @Fe3 O4 @Fe2 N, respectively. These as-prepared active materials have similar regular variation patterns in their properties, including better conductivity and battery-mimicking pseudocapacitance, which result in their high whole-electrode capacitance performance [2598.3 F g-1 (39.85 F cm-2 ), 3472.6 F g-1 (41.43 F cm-2 ) and 1907.5 F g-1 (3.41 F cm-2 ) for the composites incorporating Co, Mo, and Fe, respectively]. The as-assembled flexible, all-solid-state NiCoO2 @Co3 O4 @Co2 N//KOH/PVA//NiCoO2 @Co3 O4 @Co2 N device can be easily bent and exhibits high energy density and power density of 92.8 Wh kg-1 and 1670.4 W kg-1 , respectively. The universality of this design strategy could allow it to be employed in producing hybrid materials for high-performance energy-storage devices.

6.
Environ Res ; 199: 111223, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33991571

RESUMEN

Eutrophication is generally caused by excess nitrogen and phosphorus being released into surface waters by runoff. Developing adsorbents for adsorbing phosphate within soil buffer zones and/or water treatment columns may be effective methods to mitigate this problem. In this study, an amorphous FeOOH (AF) and a well-crystallized α-FeOOH (CF) was formulated to compare phosphate adsorption behavior. The physicochemical properties between these species showed significant differences in morphology, crystallization, zeta potential, and specific surface area. The AF exhibited higher phosphate uptake than CF. X-ray photoelectron spectroscopy (XPS) verified that the hydroxyl groups within AF were 13.28% higher than that in CF. The triply coordinated hydroxyl groups (µ3-OH) associated with AF and CF appeared at different positions as shown in the diffuse reflectance infrared Fourier transform spectroscopy (DRIFTS) analyses, confirming that AF contains more adsorption reactive sites (µ3-OH). Mechanisms for monodentate formations and a stable six-member ring structure were proposed. The X-ray absorption near the edge structure (XANES) and XPS results suggested that the iron valence in AF was dominated by Fe (III). XANES also demonstrated that the amorphous structure found in the AF was caused by the disordered tetrahedron and octahedron alignments, leading to a higher phosphate adsorption.


Asunto(s)
Compuestos de Hierro , Fosfatos , Adsorción , Minerales
7.
Am J Respir Crit Care Med ; 202(5): 717-729, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32543879

RESUMEN

Rationale: Respiratory syncytial virus (RSV) is the leading cause of childhood respiratory infections worldwide; however, no vaccine is available, and treatment options are limited. Identification of host factors pivotal to viral replication may inform the development of novel therapies, prophylaxes, or diagnoses.Objectives: To identify host factors involved in RSV replication and to evaluate their potential for disease management.Methods: A gain-of-function screening was performed on the basis of a genome-wide human complementary DNA library screen for host factors involved in RSV replication. The antiviral mechanism of CXCL4 (chemokine [C-X-C motif] ligand 4) was analyzed. Its clinical role was evaluated via nasopharyngeal aspirates and plasma samples from patients with RSV infection and different disease severities.Measurements and Main Results: Forty-nine host factors restricting RSV replication were identified by gain-of-function screening, with CXCL4 showing the strongest antiviral effect, which was secretion dependent. CXCL4 blocked viral attachment through binding to the RSV main receptor heparan sulfate, instead of through interacting with RSV surface proteins. Intranasal pretreatment with CXCL4 alleviated inflammation in RSV-infected mice, as shown by decreased concentrations of tumor necrosis factor and viral load in BAL fluid samples as well as by viral nucleocapsid protein histological staining in lungs. Compared with non-RSV infections, RSV infections induced elevated CXCL4 concentrations both in plasma and airway samples from mice and pediatric patients. The airway CXCL4 concentration was correlated with viral load and disease severity in patients (P < 0.001).Conclusions: Our results suggest that CXCL4 is an RSV restriction factor that can block viral entry and serve as an indicator of clinical severity in RSV infections.


Asunto(s)
Antivirales/uso terapéutico , Quimiocinas CXC/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitial Respiratorio Humano/genética , Biomarcadores/metabolismo , Preescolar , ADN Viral/análisis , Femenino , Humanos , Lactante , Recién Nacido , Ligandos , Masculino , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Infecciones por Virus Sincitial Respiratorio/virología , Índice de Severidad de la Enfermedad
8.
PLoS Pathog ; 13(3): e1006243, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28253362

RESUMEN

[This corrects the article DOI: 10.1371/journal.ppat.1003231.].

9.
J Virol ; 91(18)2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28679757

RESUMEN

Enterovirus 71 (EV71) can cause hand-foot-and-mouth disease (HFMD) in young children. Severe infection with EV71 can lead to neurological complications and even death. However, the molecular basis of viral pathogenesis remains poorly understood. Here, we report that EV71 induces degradation of gasdermin D (GSDMD), an essential component of pyroptosis. Remarkably, the viral protease 3C directly targets GSDMD and induces its cleavage, which is dependent on the protease activity. Further analyses show that the Q193-G194 pair within GSDMD is the cleavage site of 3C. This cleavage produces a shorter N-terminal fragment spanning amino acids 1 to 193 (GSDMD1-193). However, unlike the N-terminal fragment produced by caspase-1 cleavage, this fragment fails to trigger cell death or inhibit EV71 replication. Importantly, a T239D or F240D substitution abrogates the activity of GSDMD consisting of amino acids 1 to 275 (GSDMD1-275). This is correlated with the lack of pyroptosis or inhibition of viral replication. These results reveal a previously unrecognized strategy for EV71 to evade the antiviral response.IMPORTANCE Recently, it has been reported that GSDMD plays a critical role in regulating lipopolysaccharide and NLRP3-mediated interleukin-1ß (IL-1ß) secretion. In this process, the N-terminal domain of p30 released from GSDMD acts as an effector in cell pyroptosis. We show that EV71 infection downregulates GSDMD. EV71 3C cleaves GSDMD at the Q193-G194 pair, resulting in a truncated N-terminal fragment disrupted for inducing cell pyroptosis. Notably, GSDMD1-275 (p30) inhibits EV71 replication whereas GSDMD1-193 does not. These results reveal a new strategy for EV71 to evade the antiviral response.


Asunto(s)
Enterovirus Humano A/fisiología , Interacciones Huésped-Patógeno , Proteínas de Neoplasias/metabolismo , Piroptosis , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Unión a Fosfato , Unión Proteica , Proteolisis
10.
J Virol ; 91(19)2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28701404

RESUMEN

Like other enteroviruses, enterovirus 71 (EV71) relies on phosphatidylinositol 4-kinase IIIß (PI4KB) for genome RNA replication. However, how PI4KB is recruited to the genome replication sites of EV71 remains elusive. Recently, we reported that a host factor, ACBD3, is needed for EV71 replication by interacting with viral 3A protein. Here, we show that ACBD3 is required for the recruitment of PI4KB to RNA replication sites. Overexpression of viral 3A or EV71 infection stimulates the interaction of PI4KB and ACBD3. Consistently, EV71 infection induces the production of phosphatidylinositol-4-phosphate (PI4P). Furthermore, PI4KB, ACBD3, and 3A are all localized to the viral-RNA replication sites. Accordingly, PI4KB or ACBD3 depletion by small interfering RNA (siRNA) leads to a reduction in PI4P production after EV71 infection. I44A or H54Y substitution in 3A interrupts the stimulation of PI4KB and ACBD3. Further analysis suggests that stimulation of ACBD3-PI4KB interaction is also important for the replication of enterovirus 68 but disadvantageous to human rhinovirus 16. These results reveal a mechanism of enterovirus replication that involves a selective strategy for recruitment of PI4KB to the RNA replication sites.IMPORTANCE Enterovirus 71, like other human enteroviruses, replicates its genome within host cells, where viral proteins efficiently utilize cellular machineries. While multiple factors are involved, it is largely unclear how viral replication is controlled. We show that the 3A protein of enterovirus 71 recruits an enzyme, phosphatidylinositol 4-kinase IIIß, by interacting with ACBD3, which alters cellular membranes through the production of a lipid, PI4P. Consequently, the viral and host proteins form a large complex that is necessary for RNA synthesis at replication sites. Notably, PI4KB-ACBD3 interaction also differentially mediates the replication of enterovirus 68 and rhinovirus 16. These results provide new insight into the molecular network of enterovirus replication.

11.
J Virol ; 90(3): 1613-21, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26608321

RESUMEN

UNLABELLED: Human enterovirus 68 (EV-D68) is a member of the EV-D species, which belongs to the EV genus of the Picornaviridae family. Over the past several years, clusters of EV-D68 infections have occurred worldwide. A recent outbreak in the United States is the largest one associated with severe respiratory illness and neurological complication. Although clinical symptoms are recognized, the virus remains poorly understood. Here we report that EV-D68 inhibits innate antiviral immunity by downregulation of interferon regulatory factor 7 (IRF7), an immune factor with a pivotal role in viral pathogenesis. This process depends on 3C(pro), an EV-D68-encoded protease, to mediate IRF7 cleavage. When expressed in host cells, 3C(pro) targets Q167 and Q189 within the constitutive activation domain, resulting in cleavage of IRF7. Accordingly, wild-type IRF7 is fully active. However, IRF7 cleavage abrogated its capacity to activate type I interferon expression and limit replication of EV-D68. Notably, IRF7 cleavage strictly requires the protease activity of 3C(pro). Together, these results suggest that a dynamic interplay between 3C(pro) and IRF7 may determine the outcome of EV-D68 infection. IMPORTANCE: EV-D68 is a globally emerging pathogen, but the molecular basis of EV-D68 pathogenesis is unclear. Here we report that EV-D68 inhibits innate immune responses by targeting an immune factor, IRF7. This involves the 3C protease encoded by EV-D68, which mediates the cleavage of IRF7. These observations suggest that the 3C(pro)-IRF7 interaction may represent an interface that dictates EV-D68 infection.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Enterovirus Humano D/enzimología , Enterovirus Humano D/inmunología , Interacciones Huésped-Patógeno , Evasión Inmune , Factor 7 Regulador del Interferón/metabolismo , Proteínas Virales/metabolismo , Proteasas Virales 3C , Línea Celular , Humanos , Proteolisis
12.
J Virol ; 88(17): 9830-41, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24942571

RESUMEN

UNLABELLED: Enterovirus 71 (EV71) causes hand, foot, and mouth disease in young children and infants. Severe infection with EV71 can lead to various neurological complications or fatal diseases. However, the mechanism of EV71 pathogenesis is poorly understood. Emerging evidence suggests that EV71 modulates type I interferon (IFN) and cytokine responses. Here, we show that EV71 disables components of the TAB2 complex through the 3C protein. When expressed in mammalian cells, EV71 3C interacts with TAB2 and TAK1, which inhibits NF-κB activation. Furthermore, 3C mediates cleavage of TAB2 and its partners, which requires the protease activity. H40D or C147S substitution in the 3C active sites abolishes its activity, whereas R84Q or V154S substitution in the RNA binding domain has no effect. The 3C protein targets TAB2 at Q113-S114, TAK1 at Q360-S361, TAB1 both at Q414-G415 and Q451-S452, and TAB3 at Q173-G174 and Q343-G344. Importantly, overexpression of TAB2 inhibits EV71 replication, whereas addition of cleaved fragments has no effect. Thus, an equilibrium between the TAB2 complex and EV71 3C represents a control point of viral infection. These results suggest that TAK1/TAB1/TAB2/TAB3 cleavage mediated by EV71 may be a mechanism to interfere with inflammatory responses. IMPORTANCE: The TAK1 complex plays a critical role in the activation of NF-κB and cytokine production. However, little is known about its connection to enterovirus 71 (EV71). We demonstrate that EV71 3C suppresses cytokine expression via cleavage of the TAK1 complex proteins. EV71 3C interacts with TAB2 and TAK1. Furthermore, overexpression of TAB2 inhibits EV71 replication, whereas addition of cleaved fragment has no effect. These results suggest that the interplay of EV71 and the TAK1 complex influences the outcome of viral infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cisteína Endopeptidasas/metabolismo , Citocinas/antagonistas & inhibidores , Enterovirus Humano A/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Virales/metabolismo , Proteasas Virales 3C , Sustitución de Aminoácidos , Línea Celular , Cisteína Endopeptidasas/genética , Enterovirus Humano A/genética , Enterovirus Humano A/inmunología , Enterovirus Humano A/patogenicidad , Humanos , Hidrólisis , Evasión Inmune , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Virales/genética
13.
J Virol ; 88(12): 6650-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24672048

RESUMEN

UNLABELLED: Human enterovirus 68 (EV68) is a member of the EV-D species, which belongs to the EV genus of the Picornaviridae family. Over the past several years, there have been increasingly documented outbreaks of respiratory disease associated with EV68. As a globally emerging pathogen, EV68 infects both adults and children. However, the molecular basis of EV68 pathogenesis is unknown. Here we report that EV68 inhibits Toll-like receptor 3 (TLR3)-mediated innate immune responses by targeting the TIR domain-containing adaptor inducing beta interferon (TRIF). In infected HeLa cells, EV68 inhibits poly(I·C)-induced interferon regulatory factor 3 (IRF3) activation and beta interferon (IFN-ß) expression. Further investigations revealed that TRIF, a critical adaptor downstream of TLR3, is targeted by EV68. When expressed alone, 3C(pro), an EV68-encoded protease, cleaves TRIF. 3C(pro) mediates TRIF cleavage at Q312 and Q653, which are sites in the amino- and carboxyl-terminal domains, respectively. This cleavage relies on 3C(pro)'s cysteine protease activity. Cleavage of TRIF abolishes the capacity of TRIF to activate NF-κB and IFN-ß signaling. These results suggest that control of TRIF by 3C(pro) may be a mechanism by which EV68 subverts host innate immune responses. IMPORTANCE: EV68 is a globally emerging pathogen, but the molecular basis of EV68 pathogenesis is unclear. Here we report that EV68 inhibits TLR3-mediated innate immune responses by targeting TRIF. Further investigations revealed that TRIF is cleaved by 3C(pro). These results suggest that control of TRIF by 3C(pro) may be a mechanism by which EV68 impairs type I IFN production in response to TLR3 activation.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Cisteína Endopeptidasas/metabolismo , Enterovirus Humano D/enzimología , Infecciones por Enterovirus/inmunología , Receptor Toll-Like 3/inmunología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Proteínas Adaptadoras del Transporte Vesicular/genética , Cisteína Endopeptidasas/genética , Enterovirus Humano D/genética , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/virología , Interacciones Huésped-Patógeno , Humanos , Interferón beta/genética , Interferón beta/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Proteolisis , Receptor Toll-Like 3/genética , Proteínas Virales/genética
14.
PLoS Pathog ; 9(3): e1003231, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23555247

RESUMEN

Enterovirus 71 (EV71) is the major causative pathogen of hand, foot, and mouth disease (HFMD). Its pathogenicity is not fully understood, but innate immune evasion is likely a key factor. Strategies to circumvent the initiation and effector phases of anti-viral innate immunity are well known; less well known is whether EV71 evades the signal transduction phase regulated by a sophisticated interplay of cellular and viral proteins. Here, we show that EV71 inhibits anti-viral type I interferon (IFN) responses by targeting the mitochondrial anti-viral signaling (MAVS) protein--a unique adaptor molecule activated upon retinoic acid induced gene-I (RIG-I) and melanoma differentiation associated gene (MDA-5) viral recognition receptor signaling--upstream of type I interferon production. MAVS was cleaved and released from mitochondria during EV71 infection. An in vitro cleavage assay demonstrated that the viral 2A protease (2A(pro)), but not the mutant 2A(pro) (2A(pro)-110) containing an inactivated catalytic site, cleaved MAVS. The Protease-Glo assay revealed that MAVS was cleaved at 3 residues between the proline-rich and transmembrane domains, and the resulting fragmentation effectively inactivated downstream signaling. In addition to MAVS cleavage, we found that EV71 infection also induced morphologic and functional changes to the mitochondria. The EV71 structural protein VP1 was detected on purified mitochondria, suggesting not only a novel role for mitochondria in the EV71 replication cycle but also an explanation of how EV71-derived 2A(pro) could approach MAVS. Taken together, our findings reveal a novel strategy employed by EV71 to escape host anti-viral innate immunity that complements the known EV71-mediated immune-evasion mechanisms.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antivirales/farmacología , Cisteína Endopeptidasas/metabolismo , Enterovirus Humano A/enzimología , Interferón Tipo I/farmacología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Enterovirus Humano A/efectos de los fármacos , Femenino , Células HeLa , Humanos , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Inhibidores de Proteasas/farmacología , Infecciones por Virus ARN , Rabdomiosarcoma , Transducción de Señal
15.
J Virol ; 87(3): 1690-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23175366

RESUMEN

Enterovirus 71 (EV71) is a positive-stranded RNA virus which is capable of inhibiting innate immunity. Among virus-encoded proteins, the 3C protein compromises the type I interferon (IFN-I) response mediated by retinoid acid-inducible gene-I (RIG-I) or Toll-like receptor 3 that activates interferon regulatory 3 (IRF3) and IRF7. In the present study, we report that enterovirus 71 downregulates IRF7 through the 3C protein, which inhibits the function of IRF7. When expressed in mammalian cells, the 3C protein mediates cleavage of IRF7 rather than that of IRF3. This process is insensitive to inhibitors of caspase, proteasome, lysosome, and autophagy. H40D substitution in the 3C active site abolishes its activity, whereas R84Q or V154S substitution in the RNA binding motif has no effect. Furthermore, 3C-mediated cleavage occurs at the Q189-S190 junction within the constitutive activation domain of IRF7, resulting in two cleaved IRF7 fragments that are incapable of activating IFN expression. Ectopic expression of wild-type IRF7 limits EV71 replication. On the other hand, expression of the amino-terminal domain of IRF7 enhances EV71 infection, which correlates with its ability to interact with and inhibit IRF3. These results suggest that control of IRF7 by the 3C protein may represent a viral mechanism to escape cellular responses.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Enterovirus Humano A/patogenicidad , Evasión Inmune , Factor 7 Regulador del Interferón/antagonistas & inhibidores , Factor 7 Regulador del Interferón/metabolismo , Proteínas Virales/metabolismo , Proteasas Virales 3C , Sustitución de Aminoácidos , Dominio Catalítico , Línea Celular , Cisteína Endopeptidasas/genética , Enterovirus Humano A/enzimología , Humanos , Hidrólisis , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Proteínas Virales/genética
16.
Adv Sci (Weinh) ; 11(13): e2305750, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38342599

RESUMEN

Deciphering cellular interactions is essential to both understand the mechanisms underlying a broad range of human diseases, but also to manipulate therapies targeting these diseases. Here, the formation of cell doublets resulting from specific membrane ligand-receptor interactions is discovered. Based on this phenomenon, the study developed DoubletSeeker, a novel high-throughput method for the reliable identification of ligand-receptor interactions. The study shows that DoubletSeeker can accurately identify T cell receptor (TCR)-antigen interactions with high sensitivity and specificity. Notably, DoubletSeeker effectively captured paired TCR-peptide major histocompatibility complex (pMHC) information during a highly complex library-on-library screening and successfully identified three mutant TCRs that specifically recognize the MART-1 epitope. In turn, DoubletSeeker can act as an antigen discovery platform that allows for the development of novel immunotherapy targets, making it valuable for investigating fundamental tumor immunology.


Asunto(s)
Antígenos , Receptores de Antígenos de Linfocitos T , Humanos , Ligandos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Péptidos , Complejo Mayor de Histocompatibilidad
17.
J Biol Chem ; 287(2): 1054-65, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22065578

RESUMEN

Overwhelming evidence indicates that Bax and Bak are indispensable for mediating cytochrome c release from mitochondria during apoptosis. Here we report a Bax/Bak-independent mechanism of cytochrome c release and apoptosis. We identified a natural diterpenoid compound that induced apoptosis in bax/bak double knock-out murine embryonic fibroblasts and substantially reduced the tumor growth from these cells implanted in mice. Treatment with the compound significantly increased expression of Bim, which migrated to mitochondria, altering the conformation of and forming oligomers with resident Bcl-2 to induce cytochrome c release and caspase activation. Importantly, purified Bim and Bcl-2 proteins cooperated to permeabilize a model mitochondrial outer membrane; this was accompanied by oligomerization of these proteins and deep embedding of Bcl-2 in the membrane. Therefore, the diterpenoid compound induces a structural and functional conversion of Bcl-2 through Bim to permeabilize the mitochondrial outer membrane, thereby inducing apoptosis independently of Bax and Bak. Because Bcl-2 family proteins play important roles in cancer development and relapse, this novel cell death mechanism can be explored for developing more effective anticancer therapeutics.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Apoptosis/efectos de los fármacos , Diterpenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de la Membrana/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Proteína X Asociada a bcl-2/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Línea Celular Transformada , Citocromos c/genética , Citocromos c/metabolismo , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Membranas Mitocondriales/metabolismo , Permeabilidad/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína X Asociada a bcl-2/genética
18.
Antiviral Res ; 220: 105761, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37992763

RESUMEN

Hand, foot, and mouth disease (HFMD) is a common infectious disease in infants and children, especially those under five years of age. EV-A71 is a common pathogen that causes HFMD and the primary pathogen leading to severe or fatal HFMD, which is characterized by neurological complications. However, the underlying mechanisms of EV-A71 pathogenesis remain largely unknown. In this report, we used proteomic and phosphorylated proteomic methods to characterize the proteome and phosphoproteome profiles of EV-A71-infected human neuroblastoma SK-N-SH cells. More than 7744 host proteins and 10069 phosphorylation modification sites were successfully quantified. Among them, 974 proteins and 3648 phosphorylation modification sites were regulated significantly during EV-A71 infection. KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis revealed that EV-A71 altered cell biological processes, including protein synthesis, RNA splicing and metabolism in SK-N-SH cells. Notably, based on the prediction of upregulated kinases during EV-A71 infection, we identified specific kinase inhibitors approved by the FDA, with ceralasertib, bosutinib, flavin mononucleotide, minocycline, pimasertib and acetylcysteine inhibiting EV-A71 infection. Finally, EV-A71 proteins were found to be phosphorylated during infection, with one site (S184 on 3D polymerase) observed to be crucial for viral replication because a S184A mutation knocked out viral replication. The results improve our understanding of the host response to EV-A71 infection of neuroblastoma cells and provide potential targets for developing anti-EV-A71 strategies.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Enfermedad de Boca, Mano y Pie , Neuroblastoma , Niño , Lactante , Humanos , Proteómica , Enterovirus Humano A/fisiología , Replicación Viral , Proteoma/farmacología , Antivirales/farmacología
19.
Nat Commun ; 14(1): 3715, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37349329

RESUMEN

Viral RNA-host protein interactions are indispensable during RNA virus transcription and replication, but their detailed structural and dynamical features remain largely elusive. Here, we characterize the binding interface for the SARS-CoV-2 stem-loop 3 (SL3) cis-acting element to human TIA1 protein with a combined theoretical and experimental approaches. The highly structured SARS-CoV-2 SL3 has a high binding affinity to TIA1 protein, in which the aromatic stacking, hydrogen bonds, and hydrophobic interactions collectively direct this specific binding. Further mutagenesis studies validate our proposed 3D binding model and reveal two SL3 variants have enhanced binding affinities to TIA1. And disruptions of the identified RNA-protein interactions with designed antisense oligonucleotides dramatically reduce SARS-CoV-2 infection in cells. Finally, TIA1 protein could interact with conserved SL3 RNA elements within other betacoronavirus lineages. These findings open an avenue to explore the viral RNA-host protein interactions and provide a pioneering structural basis for RNA-targeting antiviral drug design.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , ARN Viral/metabolismo , Unión Proteica , COVID-19/genética , Mutagénesis , Antígeno Intracelular 1 de las Células T/metabolismo
20.
Microbiol Spectr ; 11(4): e0080123, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37409968

RESUMEN

Enterovirus D68 (EV-D68) is a member of the species Enterovirus D in the genus Enterovirus of the family Picornaviridae. As an emerging non-polio enterovirus, EV-D68 is widely spread all over the world and causes severe neurological and respiratory illnesses. Although the intrinsic restriction factors in the cell provide a frontline defense, the molecular nature of virus-host interactions remains elusive. Here, we provide evidence that the major histocompatibility complex class II chaperone, CD74, inhibits EV-D68 replication in infected cells by interacting with the second hydrophobic region of 2B protein, while EV-D68 attenuates the antiviral role of CD74 through 3Cpro cleavage. 3Cpro cleaves CD74 at Gln-125. The equilibrium between CD74 and EV-D68 3Cpro determines the outcome of viral infection. IMPORTANCE As an emerging non-polio enterovirus, EV-D68 is widely spread all over the world and causes severe neurological and respiratory illnesses. Here, we report that CD74 inhibits viral replication in infected cells by targeting 2B protein of EV-D68, while EV-D68 attenuates the antiviral role of CD74 through 3Cpro cleavage. The equilibrium between CD74 and EV-D68 3Cpro determines the outcome of viral infection.


Asunto(s)
Enterovirus Humano D , Infecciones por Enterovirus , Enterovirus , Humanos , Antígenos Virales , Antivirales/farmacología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA