Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 32(5): 1510-1525, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38454605

RESUMEN

The acute respiratory virus infection can induce uncontrolled inflammatory responses, such as cytokine storm and viral pneumonia, which are the major causes of death in clinical cases. Cyclophilin A (CypA) is mainly distributed in the cytoplasm of resting cells and released into the extracellular space in response to inflammatory stimuli. Extracellular CypA (eCypA) is upregulated and promotes inflammatory response in severe COVID-19 patients. However, how eCypA promotes virus-induced inflammatory response remains elusive. Here, we observe that eCypA is induced by influenza A and B viruses and SARS-CoV-2 in cells, mice, or patients. Anti-CypA mAb reduces pro-inflammatory cytokines production, leukocytes infiltration, and lung injury in virus-infected mice. Mechanistically, eCypA binding to integrin ß2 triggers integrin activation, thereby facilitating leukocyte trafficking and cytokines production via the focal adhesion kinase (FAK)/GTPase and FAK/ERK/P65 pathways, respectively. These functions are suppressed by the anti-CypA mAb that specifically blocks eCypA-integrin ß2 interaction. Overall, our findings reveal that eCypA-integrin ß2 signaling mediates virus-induced inflammatory response, indicating that eCypA is a potential target for antibody therapy against viral pneumonia.


Asunto(s)
COVID-19 , Ciclofilina A , Ciclofilina A/metabolismo , Animales , Humanos , Ratones , COVID-19/metabolismo , COVID-19/virología , COVID-19/inmunología , Antígenos CD18/metabolismo , SARS-CoV-2 , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Neumonía Viral/metabolismo , Neumonía Viral/inmunología , Citocinas/metabolismo , Anticuerpos Monoclonales/farmacología , Transducción de Señal , Virus de la Influenza A , Modelos Animales de Enfermedad
2.
Sheng Wu Gong Cheng Xue Bao ; 39(12): 4796-4808, 2023 Dec 25.
Artículo en Chino | MEDLINE | ID: mdl-38147982

RESUMEN

This study aimed to explore the mechanism of how African swine fever virus (ASFV) I226R protein inhibits the cGAS-STING signaling pathway. We observed that I226R protein (pI226R) significantly inhibited the cGAS-STING-mediated type Ⅰ interferons and the interferon-stimulated genes production by dual-luciferase reporter assay system and real-time quantitative PCR. The results of co-immunoprecipitation assay and confocal microscopy showed that pI226R interacted with cGAS. Furthermore, pI226R promoted cGAS degradation through autophagy-lysosome pathway. Moreover, we found that pI226R decreased the binding of cGAS to E3 ligase tripartite motif protein 56 (TRIM56), resulting in the weakened monoubiquitination of cGAS, thus inhibiting the activation of cGAS and cGAS-STING signaling. In conclusion, ASFV pI226R suppresses the antiviral innate immune response by antagonizing cGAS, which contributes to an in-depth understanding of the immune escape mechanism of ASFV and provides a theoretical basis for the development of vaccines.


Asunto(s)
Virus de la Fiebre Porcina Africana , Animales , Porcinos , Virus de la Fiebre Porcina Africana/genética , Virus de la Fiebre Porcina Africana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Inmunidad Innata , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Transducción de Señal/genética
3.
Sheng Wu Gong Cheng Xue Bao ; 39(12): 4809-4823, 2023 Dec 25.
Artículo en Chino | MEDLINE | ID: mdl-38147983

RESUMEN

In order to understand the prevalence and evolution of porcine reproductive and respiratory syndrome virus (PRRSV) in China and to develop subunit vaccine against the epidemic lineage, the genetic evolution analysis of PRRSV strains isolated in China from 2001 to 2021 was performed. The representative strains of the dominant epidemic lineage were selected to optimize the membrane protein GP5 and M nucleotide sequences, which were used, with the interferon and the Fc region of immunoglobulin, to construct the eukaryotic expression plasmids pCDNA3.4-IFNα-GP5-Fc and pCDNA3.4-IFNα-M-Fc. Subsequently, the recombinant proteins IFNα-GP5-Fc and IFNα-M-Fc were expressed by HEK293T eukaryotic expression system. The two recombinant proteins were mixed with ISA206VG adjuvant to immunize weaned piglets. The humoral immunity level was evaluated by ELISA and neutralization test, and the cellular immunity level was detected by ELISPOT test. The results showed that the NADC30-like lineage was the main epidemic lineage in China in recent years, and the combination of IFNα-GP5-Fc and IFNα-M-Fc could induce high levels of antibody and cellular immunity in piglets. This study may facilitate the preparation of a safer and more effective new PRRSV subunit vaccine.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Vacunas Virales , Humanos , Animales , Porcinos , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Síndrome Respiratorio y de la Reproducción Porcina/prevención & control , Células HEK293 , Proteínas del Envoltorio Viral/genética , Anticuerpos Antivirales , Vacunas Virales/genética , Proteínas Recombinantes , Vacunas de Subunidad
4.
Autophagy ; 19(12): 3113-3131, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37482689

RESUMEN

ABBREVIATIONS: aa: amino acid; ATF6: activating transcription factor 6; ATG5: autophagy related 5; CCPG1: cell cycle progression 1; CFTR: CF transmembrane conductance regulator; cGAMP: cyclic GMP-AMP; CGAS: cyclic GMP-AMP synthase; CHX: cycloheximide; Co-IP: co-immunoprecipitation; CQ: chloroquine; EIF2A/eIF2α: eukaryotic translation initiation factor 2A; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; ER: endoplasmic reticulum; ERN1/IRE1: endoplasmic reticulum to nucleus signaling 1; GFP: green fluorescent protein; HSPA5/GRP78: heat shock protein family A (Hsp70) member 5; HSV-1: herpes simplex virus type 1; IFIT1: interferon induced protein with tetratricopeptide repeats 1; IFNB1/IFN-ß: interferon beta 1; IRF3: interferon regulatory factor 3; ISG15: ISG15 ubiquitin like modifier; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAP3K7/TAK1: mitogen-activated protein kinase kinase kinase 7; MAVS: mitochondrial antiviral signaling protein; MOI: multiplicity of infection; NFKB/NF-κB: nuclear factor kappa B; NSP6: non-structural protein 6; Δ106-108: deletion of amino acids 106-108 in NSP6 of SARS-CoV-2; Δ105-107: deletion of amino acids 105-107 in NSP6 of SARS-CoV-2; RETREG1/FAM134B: reticulophagy regulator 1; RIGI/DDX58: RNA sensor RIG-I; SQSTM1/p62: sequestosome 1; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Autofagia/fisiología , Estrés del Retículo Endoplásmico/fisiología , Chaperón BiP del Retículo Endoplásmico , Interferones , Aminoácidos
5.
Antiviral Res ; 215: 105644, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37244381

RESUMEN

African swine fever (ASF) is a highly contagious and deadly disease that affects domestic and wild pigs. No commercial vaccine or antiviral is currently available against ASF. The control of ASF primarily relies on implementing effective biosecurity measures during the breeding process. Here, we evaluated the preventive and therapeutic potential of the interferon (IFN) cocktail (a mixture of recombinant porcine IFN α and γ) on ASF. The IFN cocktail treatment delayed the onset of ASF symptoms and ASF virus (ASFV) replication for approximately one week. However, IFN cocktail treatment could not prevent the death of the pigs. Further analysis showed that IFN cocktail treatment increased the expression of multiple IFN-stimulated genes (ISGs) in porcine peripheral blood mononuclear cells in vivo and in vitro. Additionally, IFN cocktail modulated the expression of pro- and anti-inflammatory cytokines and reduced tissue injury in the ASFV-infected pigs. Collectively, the results suggest that the IFN cocktail restricts the progression of acute ASF by inducing high levels of ISGs, contributing to the pre-establishment of antiviral status, and modulating the balance of pro- and anti-inflammatory mediators to lessen cytokine storm-mediated tissue damage.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Porcinos , Animales , Fiebre Porcina Africana/tratamiento farmacológico , Fiebre Porcina Africana/prevención & control , Leucocitos Mononucleares , Interferón-alfa/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico
6.
Sheng Wu Gong Cheng Xue Bao ; 38(9): 3329-3343, 2022 Sep 25.
Artículo en Chino | MEDLINE | ID: mdl-36151803

RESUMEN

This paper aims to explore the effects of chicken interferon-γ (ChIFN-γ) and interleukin-2 (ChIL-2) on type 1 helper (Th1) T lymphocyte differentiation. To be specific, ChIFN-γ and ChIL-2 were first expressed in Escherichia coli competent cells and then purified by Ni-NTA affinity chromatography. Different concentration of ChIFN-γ and ChIL-2 were employed to stimulate the lymphocytes in chicken peripheral blood which had been activated by concanavalin A (Con A), and the mRNA levels of cytokines related to Th1 cell differentiation were detected by real-time quantitative PCR (RT-qPCR). The results showed that both ChIFN-γ and ChIL-2 can significantly up-regulate mRNA levels of cytokines related to Th1 cell differentiation and the optimal concentration was 12.5 µg/mL and 25.0 µg/mL, respectively. In addition, specific-pathogen-free (SPF) chickens were immunized with ChIL-2 or ChIFN-γ together with H9N2 vaccine, or H9N2 vaccine alone by oral administration or intramuscular injection, respectively. The mRNA levels of cytokines related to Th1 cell differentiation were detected after immunization. The results showed that ChIFN-γ and ChIL-2 significantly up-regulated the mRNA levels of cytokines related to Th1 cell differentiation induced by H9N2 vaccine compared with H9N2 vaccine alone, and that the intramuscular injection was better than oral administration. In this study, we verified that ChIFN-γ and ChIL-2 can significantly enhance mRNA levels of cytokines related to Th1 cell differentiation induced by ConA or H9N2 vaccine in vitro and in vivo. The results of this study can lay a theoretical basis for using ChIFN-γ and ChIL-2 as vaccine adjuvants.


Asunto(s)
Pollos , Subtipo H9N2 del Virus de la Influenza A , Animales , Diferenciación Celular , Concanavalina A , Citocinas/genética , Subtipo H9N2 del Virus de la Influenza A/genética , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-2/genética , ARN Mensajero
7.
Sheng Wu Gong Cheng Xue Bao ; 38(9): 3390-3405, 2022 Sep 25.
Artículo en Chino | MEDLINE | ID: mdl-36151808

RESUMEN

Influenza B virus (IBV) is more likely to cause complications than influenza A virus (IAV) and even causes higher disease burden than IAV in a certain season, but IBV has received less attention. In order to analyze the genetic evolution characteristics of the clinical strain IBV (B/Guangxi-Jiangzhou/1352/2018), we constructed genetic evolution trees and analyzed the homology and different amino acids of hemagglutinin and neuraminidase referring to the vaccine strains recommended by World Health Organization (WHO). We found that strain B/Guangxi-Jiangzhou/1352/2018 was free of interlineage reassortment and poorly matched with the vaccine strain B/Colorado/06/2017 of the same year. We also determined the median lethal dose (LD50) and the pathogenicity of strain B/Guangxi-Jiangzhou/1352/2018 in mice. The results showed that the LD50 was 105.9 TCID50 (median tissue culture infective dose), the IBV titer in the lungs reached peak 1 d post infection and the mRNA level of the most of inflammatory cytokines in the lungs reached peak 12 h post infection. The alveoli in the lungs were severely damaged and a large number of inflammatory cells were infiltrated post infection. The study demonstrated that the clinical strain IBV (B/Guangxi-Jiangzhou/1352/2018) could infect mice and induce typical lung inflammation. This will facilitate the research on the pathogenesis and transmission mechanism of IBV, and provide an ideal animal model for evaluation of new vaccines, antiviral and anti-inflammatory drug.


Asunto(s)
Virus de la Influenza B , Gripe Humana , Aminoácidos/genética , Animales , Antivirales/farmacología , China , Citocinas/metabolismo , Hemaglutininas/metabolismo , Humanos , Virus de la Influenza B/genética , Virus de la Influenza B/patogenicidad , Gripe Humana/inmunología , Gripe Humana/virología , Ratones , Neuraminidasa/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Filogenia , ARN Mensajero/metabolismo , Virulencia/genética
8.
Cell Commun Signal ; 20(1): 103, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35820905

RESUMEN

BACKGROUND: Cytosolic RNA sensing can elicit immune responses against viral pathogens. However, antiviral responses must be tightly regulated to avoid the uncontrolled production of type I interferons (IFN) that might have deleterious effects on the host. Upon bacterial infection, the germinal center kinase MST4 can directly phosphorylate the adaptor TRAF6 to limit the inflammatory responses, thereby avoiding the damage caused by excessive immune activation. However, the molecular mechanism of how MST4 regulates virus-mediated type I IFN production remains unknown. METHODS: The expression levels of IFN-ß, IFIT1, and IFIT2 mRNA were determined by RT-PCR. The expression levels of p-IRF3, IRF3, RIG-I, MAVS, and MST4 proteins were determined by Western blot. The effect of secreted level of IFN-ß was measured by ELISA. The relationship between MST4 and MAVS was investigated by immunofluorescence staining and coimmunoprecipitation. RESULTS: In this study, we reported that MST4 can act as a negative regulator of type I IFN production. Ectopic expression of MST4 suppressed the Poly (I:C) (polyino-sinic-polycytidylic acid)- and Sendai virus (SeV)-triggered production of type I IFN, while the knockdown of MST4 enhanced the production of type I IFN. Mechanistically, upon SeV infection, the MST4 competed with TRAF3 to bind to the 360-540 domain of MAVS, thereby inhibiting the TRAF3/MAVS association. Additionally, MST4 facilitated the interaction between the E3 ubiquitin ligase Smurf1 and MAVS. This promoted the K48-linked ubiquitination of MAVS, thereby accelerating the ubiquitin-mediated proteasome degradation of MAVS. CONCLUSIONS: Our findings showed that MST4 acted as a crucial negative regulator of RLR-mediated type I IFN production. Video Abstract.


Asunto(s)
Interferón Tipo I , Factor 3 Asociado a Receptor de TNF , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Ubiquitinación
10.
Transbound Emerg Dis ; 69(4): e204-e215, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34369669

RESUMEN

African swine fever is an acute, haemorrhagic fever and contagious disease of pigs caused by African swine fever virus (ASFV), which has a great impact on the pig farming industry and related international trade. Understanding the response processes of various tissues in pigs after ASFV infection may help to address current major concerns, such as the exploration of key genes for vaccine development, the cooperative mechanism of the host response and the possibility of establishing active herd immunity. ASFV is able to infect core tissues and is associated with acute death. RNA and protein samples were obtained and verified from five tissues, including the lung, spleen, liver, kidney and lymph nodes. Multiple duplicate samples were quantitatively analyzed by corresponding transcriptomic and proteomic comparison. The results showed that different tissues cooperated in responses to ASFV infection and coordinated the defence against ASFV in the form of an inflammatory cytokine storm and interferon activation. The lung and spleen were mainly involved (dominant) in the innate immune response pathway; the liver and kidney were involved in the metabolic regulatory pathway and the inflammatory response; and the lymph nodes cooperated with the liver to complete energy metabolism regulation. The key pathways and responsive genes in each tissue of the contracted pigs were comprehensively mapped by infectomics, providing further evidence to investigate the complicated tie between ASFV and host cells.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Enfermedades de los Porcinos , Virus de la Fiebre Porcina Africana/fisiología , Animales , Comercio , Inmunidad Innata , Internacionalidad , Proteómica , Porcinos , Proteínas Virales/genética
12.
Front Microbiol ; 11: 1203, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32655518

RESUMEN

Interferons (IFNs) are proteins produced by a variety of cells during the process of virus infection. It can activate the transcription of multiple functional genes in cells, regulate the synergistic effect of multiple signaling pathways, and mediate a variety of biological functions such as antiviral activity and immune regulation. The symptoms of hosts infected with African swine fever virus (ASFV) depend on the combined interaction between viruses and the host. However, it is unclear whether IFNs can be used as an emergency preventive treatment for ASFV. This study focused on the use of recombinant porcine IFNs, produced by Escherichia coli, to inhibit the replication of ASFV. The activity of IFN against ASFV was detected using primary alveolar macrophages at different doses through immunofluorescence assays and quantitative real-time PCR. We found that both 1000 and 100 U/mL doses significantly inhibited the replication of ASFV. Meanwhile, we found that IFNs could significantly trigger the production of a variety of IFN-induced genes (IFIT1, IFITM3, Mx-1, OASL, ISG15, PKR, GBP1, Viperin, BST2, IRF-1, and CXCL10) and MHC molecules, which play key roles in resistance to virus infection. Peripheral blood samples were also obtained from surviving pigs treated with IFNs, and the viral load was determined. Consistent with in vitro tests, low-dose (105 U/kg) recombinant porcine IFNs (PoIFN-α and PoIFN-γ) significantly reduced viral load compared to that with high-dose (106 U/kg) treatment. Our results suggest that recombinant porcine IFNs have high antiviral activity against ASFV, providing a new strategy for the prevention of African swine fever.

13.
PLoS One ; 15(4): e0231039, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32267861

RESUMEN

Influenza B virus (IBV) belongs to the Orthomyxoviridae family and generally causes sporadic epidemics but is occasionally deadly to individuals. The current research mainly focuses on clinical and pathological characteristics of IBV. However, to better prevent or treat the disease, one must determine the strategies developed by IBV to invade and disrupt cellular proteins and approach to replicate itself, to suppress antiviral innate immunity, and understand how the host responds to IBV infection. The B/Shanghai/PD114/2018 virus was able to infect alveolar epithelial cells (A549) cells, with good potential for replication. To identify host cellular responses against IBV infection, differentially expressed genes (DEGs) were obtained using RNA sequencing. The GO and KEGG pathway term enrichment analyses with the DEGs were performed, and we found that the DEGs were primary involved in metabolic processes and cellular function, which may be related to the host response, including the innate immune response against the virus. Our transcriptome analysis results demonstrated robust induction of interferon and interferon-stimulated gene expression by IBV in human cells during the early stages of infection, providing a foundation for further studies focused on antiviral drug development and interactions between the virus and host.


Asunto(s)
Virus de la Influenza B/metabolismo , Gripe Humana/metabolismo , Interferones/metabolismo , Células A549/metabolismo , Células A549/virología , Western Blotting , Técnica del Anticuerpo Fluorescente , Regulación Viral de la Expresión Génica , Humanos , Virus de la Influenza B/genética , Gripe Humana/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , Ensayo de Placa Viral , Replicación Viral
14.
Sheng Wu Gong Cheng Xue Bao ; 36(1): 109-121, 2020 Jan 25.
Artículo en Chino | MEDLINE | ID: mdl-32072786

RESUMEN

The CRISPR/Cas9 gene editing technology directs Cas9 protein to recognize, bind and cleave the target site specifically by using artificial single-guide RNA (sgRNA), through non-homologous end joining or homologous end-recombinant repair mechanisms of cells, which can be engineered to knockout or knock-in of genomes. RIG-I is a pattern recognition receptor that recognizes the 5'-triphosphate-containing RNA in the cytoplasm and activates IRF3/7 and NF-κB by interacting with the downstream signaling molecule MAVS, thus initiating the expression of type I interferons and inflammatory factors. Previous studies found that influenza B virus (IBV) can up-regulate the expression of RIG-I. In the present study, to explore whether RIG-I is the major receptor for IBV to active the antiviral innate immune response and its effect on IBV replication, RIG-I gene in 293T cells was knocked out by CRISPR-Cas9 system, and a stable RIG-I knockout 293T (RIG-I(-/-) 293T) cell line was screened by puromycin pressure. The results of Western blotting showed that RIG-I was not expressed in this cell line after IBV or Sendai virus (SeV) infection, indicating that the RIG-I(-/-) 293T cell line was successfully constructed. The transcription levels of interferons, inflammatory factors and interferon-stimulated genes in RIG-I(-/-) 293T cells which were infected by IBV decreased significantly compared with those in wild-type 293T cells. Moreover, the phosphorylation of p65 and IRF3 were not detected in IBV or SeV infected RIG-I(-/-) 293T cells. It is indicated that the expression of cytokines mainly depends on the RIG-I-mediated signaling pathway at the early stage of IBV infection. Furthermore, the multi-step growth curves of IBV in the wild type and RIG-I(-/-) 293T cells showed that RIG-I inhibited the replication of IBV. Collectively, the RIG-I knockout 293T cell line was successfully constructed. We found that RIG-I is the main receptor for IBV to active the antiviral innate immune response and is critical for inhibiting IBV replication, which lays the foundation for further study of IBV infection mechanism.


Asunto(s)
Virus de la Influenza B , Proteína 58 DEAD Box , Células HEK293 , Humanos , Inmunidad Innata , Interferones , Replicación Viral
15.
Vet Microbiol ; 239: 108458, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31767074

RESUMEN

The aim of this study was to determine the antigenic relatedness of Infectious Bursal Disease Viruses (IBDVs) in the field in southern China during the period 2000-2017, as well as the antigenic relationship between the field strains and the most commonly used vaccine strains by using a virus neutralization (VN) test in vitro. The antigenic relatedness (R) value and the difference in VN titers were analyzed, and the antigenic index based on the sequences of the hypervariable region of VP2 (vVP2) of the strains was further evaluated. As a result, the R value of representative field strains showed that there were three subtypes present in the field strains examined, with 7 strains belonging to subtype 1, while strains BH11 and JS7 belonged to subtype 2 and subtype 3, respectively. The commonly used vaccine strains B87 and FW2512 belonged to subtype 1. The analysis of the VN titer differences revealed that all the 136 field strains were classified into subtype 1, except BH11 and JS7. All the field strains in subtype 1 have been divided into at least 5 subgroups, suggesting the antigenic diversity among these strains. The antigenic index based on IBDV-VP2 sequences further confirmed the antigenic differences between the three subtype strains and also the antigenic diversity among the subtype 1. The results demonstrated the antigenic diversity of field IBDVs in southern China during the years 2000-2017 and the antigenic differences between the field strains and the commonly used vaccine strains. This would indicate that the commonly used vaccines are only partially effective. These results enhance our understanding of IBDV genetic evolution and should help to develop more effective vaccines for the control of this disease in the future.


Asunto(s)
Antígenos Virales/inmunología , Infecciones por Birnaviridae/veterinaria , Virus de la Enfermedad Infecciosa de la Bolsa/fisiología , Enfermedades de las Aves de Corral/virología , Animales , Infecciones por Birnaviridae/epidemiología , Infecciones por Birnaviridae/virología , Pollos/virología , China/epidemiología , Evolución Molecular , Virus de la Enfermedad Infecciosa de la Bolsa/clasificación , Virus de la Enfermedad Infecciosa de la Bolsa/genética , Virus de la Enfermedad Infecciosa de la Bolsa/inmunología , Tipificación Molecular , Enfermedades de las Aves de Corral/epidemiología , Vacunas Virales/inmunología
16.
Cell Rep ; 27(6): 1875-1885.e5, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31067470

RESUMEN

Naproxen is a non-steroidal anti-inflammatory drug that has previously been shown to exert antiviral activity against influenza A virus by inhibiting nucleoprotein (NP) binding to RNA. Here, we show that naproxen is a potential broad, multi-mechanistic anti-influenza virus therapeutic, as it inhibits influenza B virus replication both in vivo and in vitro. The anti-influenza B virus activity of naproxen is more efficient than that of the commonly used neuraminidase inhibitor oseltamivir in mice. Furthermore, the NP of influenza B virus (BNP) has a higher binding affinity to naproxen than influenza A virus NP (ANP). Specifically, naproxen targets the NP at residues F209 (BNP) and Y148 (ANP). This interaction antagonizes the nuclear export of NP normally mediated by the host export protein CRM1. This study reveals a crucial mechanism of broad-spectrum anti-influenza virus activity of naproxen, suggesting that the existing drug naproxen may be used as an anti-influenza drug.


Asunto(s)
Antivirales/farmacología , Núcleo Celular/metabolismo , Virus de la Influenza B/efectos de los fármacos , Naproxeno/farmacología , Nucleoproteínas/metabolismo , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Línea Celular , Núcleo Celular/efectos de los fármacos , Pollos , Perros , Femenino , Humanos , Carioferinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Fenilalanina/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Replicación Viral/efectos de los fármacos , Proteína Exportina 1
17.
Virol Sin ; 33(5): 440-448, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30328013

RESUMEN

Cyclophilin A (CypA) is a peptidyl-prolyl cis/trans isomerase that interacts with the matrix protein (M1) of influenza A virus (IAV) and restricts virus replication by regulating the ubiquitin-proteasome-mediated degradation of M1. However, the mechanism by which CypA regulates M1 ubiquitination remains unknown. In this study, we reported that E3 ubiquitin ligase AIP4 promoted K48-linked ubiquitination of M1 at K102 and K104, and accelerated ubiquitin-proteasome-mediated degradation of M1. The recombinant IAV with mutant M1 (K102R/K104R) could not be rescued, suggesting that the ubiquitination of M1 at K102/K104 was essential for IAV replication. Furthermore, CypA inhibited AIP4-mediated M1 ubiquitination by impairing the interaction between AIP4 and M1. More importantly, both the mutations of M1 (K102R/K104R) and CypA inhibited the nuclear export of M1, indicating that CypA regulates the cellular localization of M1 via inhibition of AIP4-mediated M1 ubiquitination at K102 and K104, which results in the reduced replication of IAV. Collectively, our findings reveal a novel ubiquitination-based mechanism by which CypA regulates the replication of IAV.


Asunto(s)
Ciclofilina A/metabolismo , Virus de la Influenza A/fisiología , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas de la Matriz Viral/metabolismo , Ciclofilina A/genética , Regulación de la Expresión Génica , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Proteínas Represoras/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas de la Matriz Viral/genética , Replicación Viral
18.
Sheng Wu Gong Cheng Xue Bao ; 34(6): 821-831, 2018 Jun 25.
Artículo en Chino | MEDLINE | ID: mdl-29943528

RESUMEN

Influenza B virus (IBV) is a segmented negative-strand RNA virus, which often causes local outbreak or seasonal epidemic along with influenza A virus (IAV) in the world. It is pathogenic to children, teenagers and elderly people and has a higher mortality rate in children and adolescents, so it poses a serious threat to public health and health. IBV is more likely to cause complications than IAV and the disease burden of IBV even exceeds IAV in the epidemic season. Recently, especially after winter of 2017, IBV has become the dominant strain in many areas of our country and seriously affects people's health. In view of this, this article reviews the structure, epidemiology, immunology and prevention of IBV, aiming at enhancing public's perceptions of the virus and providing reference for making strategies for prevention and control of influenza B.


Asunto(s)
Virus de la Influenza B , Gripe Humana/epidemiología , Gripe Humana/prevención & control , Adolescente , Anciano , Niño , Brotes de Enfermedades/prevención & control , Humanos , Virus de la Influenza A , Gripe Humana/mortalidad , Estaciones del Año
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...