Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(8): 104987, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37392846

RESUMEN

Porcine epidemic diarrhea virus (PEDV) causes severe morbidity and mortality among newborn piglets. It significantly threatens the porcine industry in China and around the globe. To accelerate the developmental pace of drugs or vaccines against PEDV, a deeper understanding of the interaction between viral proteins and host factors is crucial. The RNA-binding protein, polypyrimidine tract-binding protein 1 (PTBP1), is crucial for controlling RNA metabolism and biological processes. The present work focused on exploring the effect of PTBP1 on PEDV replication. PTBP1 was upregulated during PEDV infection. The PEDV nucleocapsid (N) protein was degraded through the autophagic and proteasomal degradation pathways. Moreover, PTBP1 recruits MARCH8 (an E3 ubiquitin ligase) and NDP52 (a cargo receptor) for N protein catalysis and degradation through selective autophagy. Furthermore, PTBP1 induces the host innate antiviral response via upregulating the expression of MyD88, which then regulates TNF receptor-associated factor 3/ TNF receptor-associated factor 6 expression and induces the phosphorylation of TBK1 and IFN regulatory factor 3. These processes activate the type Ⅰ IFN signaling pathway to antagonize PEDV replication. Collectively, this work illustrates a new mechanism related to PTBP1-induced viral restriction, where PTBP1 degrades the viral N protein and induces type Ⅰ IFN production to suppress PEDV replication.


Asunto(s)
Infecciones por Coronavirus , Interferón Tipo I , Proteína de Unión al Tracto de Polipirimidina , Virus de la Diarrea Epidémica Porcina , Proteolisis , Enfermedades de los Porcinos , Replicación Viral , Animales , Línea Celular , Chlorocebus aethiops , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/veterinaria , Interferón Tipo I/metabolismo , Virus de la Diarrea Epidémica Porcina/fisiología , Transducción de Señal , Porcinos , Enfermedades de los Porcinos/genética , Enfermedades de los Porcinos/virología , Células Vero , Proteína de Unión al Tracto de Polipirimidina/metabolismo
2.
J Biol Chem ; 298(8): 102190, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35753351

RESUMEN

Porcine epidemic diarrhea virus (PEDV) causes diarrhea and dehydration in pigs and leads to great economic losses in the commercial swine industry. However, the underlying molecular mechanisms of host response to viral infection remain unclear. In the present study, we investigated a novel mechanism by which RALY, a member of the heterogeneous nuclear ribonucleoprotein family, significantly promotes the degradation of the PEDV nucleocapsid (N) protein to inhibit viral replication. Furthermore, we identified an interaction between RALY and the E3 ubiquitin ligase MARCH8 (membrane-associated RING-CH 8), as well as the cargo receptor NDP52 (nuclear dot protein 52 kDa), suggesting that RALY could suppress PEDV replication by degrading the viral N protein through a RALY-MARCH8-NDP52-autophagosome pathway. Collectively, these results suggest a preventive role of RALY against PEDV infection via the autophagy pathway and open up the possibility of inducing RALY in vivo as an effective prophylactic and preventive treatment for PEDV infection.


Asunto(s)
Infecciones por Coronavirus , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Animales , Autofagia , Chlorocebus aethiops , Infecciones por Coronavirus/veterinaria , Proteínas de la Nucleocápside , Virus de la Diarrea Epidémica Porcina/fisiología , Ribonucleoproteínas , Porcinos , Células Vero , Replicación Viral
3.
J Virol ; 96(13): e0061822, 2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35695513

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is the globally distributed alphacoronavirus that can cause lethal watery diarrhea in piglets, causing substantial economic damage. However, the current commercial vaccines cannot effectively the existing diseases. Thus, it is of great necessity to identify the host antiviral factors and the mechanism by which the host immune system responds against PEDV infection required to be explored. The current work demonstrated that the host protein, the far upstream element-binding protein 3 (FUBP3), could be controlled by the transcription factor TCFL5, which could suppress PEDV replication through targeting and degrading the nucleocapsid (N) protein of the virus based on selective autophagy. For the ubiquitination of the N protein, FUBP3 was found to recruit the E3 ubiquitin ligase MARCH8/MARCHF8, which was then identified, transported to, and degraded in autolysosomes via NDP52/CALCOCO2 (cargo receptors), resulting in impaired viral proliferation. Additionally, FUBP3 was found to positively regulate type-I interferon (IFN-I) signaling and activate the IFN-I signaling pathway by interacting and increasing the expression of tumor necrosis factor (TNF) receptor-associated factor 3 (TRAF3). Collectively, this study showed a novel mechanism of FUBP3-mediated virus restriction, where FUBP3 was found to degrade the viral N protein and induce IFN-I production, aiming to hinder the replication of PEDV. IMPORTANCE PEDV refers to the alphacoronavirus that is found globally and has re-emerged recently, causing severe financial losses. In PEDV infection, the host activates various host restriction factors to maintain innate antiviral responses to suppress virus replication. Here, FUBP3 was detected as a new host restriction factor. FUBP3 was found to suppress PEDV replication via the degradation of the PEDV-encoded nucleocapsid (N) protein via E3 ubiquitin ligase MARCH8 as well as the cargo receptor NDP52/CALCOCO2. Additionally, FUBP3 upregulated the IFN-I signaling pathway by interacting with and increasing tumor necrosis factor (TNF) receptor-associated factor 3 (TRAF3) expression. This study further demonstrated that another layer of complexity could be added to the selective autophagy and innate immune response against PEDV infection are complicated.


Asunto(s)
Infecciones por Coronavirus , Interferón Tipo I , Proteínas de la Nucleocápside , Virus de la Diarrea Epidémica Porcina , Factores de Transcripción , Animales , Antivirales , Línea Celular , Chlorocebus aethiops , Infecciones por Coronavirus/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Proteínas de la Nucleocápside/metabolismo , Virus de la Diarrea Epidémica Porcina/fisiología , Porcinos , Factor 3 Asociado a Receptor de TNF , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas , Células Vero
4.
J Virol ; 96(10): e0007022, 2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35499322

RESUMEN

In global infection and serious morbidity and mortality, porcine epidemic diarrhea virus (PEDV) has been regarded as a dreadful porcine pathogen, but the existing commercial vaccines are not enough to fully protect against the epidemic strains. Therefore, it is of great necessity to feature the PEDV-host interaction and develop efficient countermeasures against viral infection. As an RNA/DNA protein, the trans-active response DNA binding protein (TARDBP) plays a variety of functions in generating and processing RNA, including transcription, splicing, transport, and mRNA stability, which have been reported to regulate viral replication. The current work aimed to detect whether and how TARDBP influences PEDV replication. Our data demonstrated that PEDV replication was significantly suppressed by TARDBP, regulated by KLF16, which targeted its promoter. We observed that through the proteasomal and autophagic degradation pathway, TARDBP inhibited PEDV replication via the binding as well as degradation of PEDV-encoded nucleocapsid (N) protein. Moreover, we found that TARDBP promoted autophagic degradation of N protein via interacting with MARCHF8, an E3 ubiquitin ligase, as well as NDP52, a cargo receptor. We also showed that TARDBP promoted host antiviral innate immune response by inducing interferon (IFN) expression through the MyD88-TRAF3-IRF3 pathway during PEDV infection. In conclusion, these data revealed a new antiviral role of TARDBP, effectively suppressing PEDV replication through degrading virus N protein via the proteasomal and autophagic degradation pathway and activating type I IFN signaling via upregulating the expression of MyD88. IMPORTANCE PEDV refers to the highly contagious enteric coronavirus that has quickly spread globally and generated substantial financial damage to the global swine industry. During virus infection, the host regulates the innate immunity and autophagy process to inhibit virus infection. However, the virus has evolved plenty of strategies with the purpose of limiting IFN-I production and autophagy processes. Here, we identified that TARDBP expression was downregulated via the transcription factor KLF16 during PEDV infection. TARDBP could inhibit PEDV replication through the combination as well as degradation of PEDV-encoded nucleocapsid (N) protein via proteasomal and autophagic degradation pathways and promoted host antiviral innate immune response by inducing IFN expression through the MyD88-TRAF3-IRF3 pathway. In sum, our data identify a novel antiviral function of TARDBP and provide a better grasp of the innate immune response and protein degradation pathway against PEDV infection.


Asunto(s)
Infecciones por Coronavirus , Proteínas de Unión al ADN , Interferón Tipo I , Virus de la Diarrea Epidémica Porcina , Replicación Viral , Animales , Infecciones por Coronavirus/veterinaria , Proteínas de Unión al ADN/metabolismo , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas de la Nucleocápside/metabolismo , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/fisiología , ARN/metabolismo , Transducción de Señal , Porcinos , Factor 3 Asociado a Receptor de TNF/metabolismo
5.
J Virol ; 96(22): e0155522, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36317879

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is a re-emerging enteric coronavirus currently spreading in several nations and inflicting substantial financial damages on the swine industry. The currently available coronavirus vaccines do not provide adequate protection against the newly emerging viral strains. It is essential to study the relationship between host antiviral factors and the virus and to investigate the mechanisms underlying host immune response against PEDV infection. This study shows that heterogeneous nuclear ribonucleoprotein K (hnRNP K), the host protein determined by the transcription factor KLF15, inhibits the replication of PEDV by degrading the nucleocapsid (N) protein of PEDV in accordance with selective autophagy. hnRNP K was found to be capable of recruiting the E3 ubiquitin ligase, MARCH8, aiming to ubiquitinate N protein. Then, it was found that the ubiquitinated N protein could be delivered into autolysosomes for degradation by the cargo receptor NDP52, thereby inhibiting PEDV proliferation. Moreover, based on the enhanced MyD88 expression, we found that hnRNP K activated the interferon 1 (IFN-1) signaling pathway. Overall, the data obtained revealed a new mechanism of hnRNP K-mediated virus restriction wherein hnRNP K suppressed PEDV replication by degradation of viral N protein using the autophagic degradation pathway and by induction of IFN-1 production based on upregulation of MyD88 expression. IMPORTANCE The spread of the highly virulent PEDV in many countries is still leading to several epidemic and endemic outbreaks. To elucidate effective antiviral mechanisms, it is important to study the relationship between host antiviral factors and the virus and to investigate the mechanisms underlying host immune response against PEDV infection. In the work, we detected hnRNP K as a new host restriction factor which can hinder PEDV replication through degrading the nucleocapsid protein based on E3 ubiquitin ligase MARCH8 and the cargo receptor NDP52. In addition, via the upregulation of MyD88 expression, hnRNP K could also activate the interferon (IFN) signaling pathway. This study describes a previously unknown antiviral function of hnRNP K and offers a new vision toward host antiviral factors that regulate innate immune response as well as a protein degradation pathway against PEDV infection.


Asunto(s)
Infecciones por Coronavirus , Ribonucleoproteína Heterogénea-Nuclear Grupo K , Interferón Tipo I , Virus de la Diarrea Epidémica Porcina , Replicación Viral , Animales , Antivirales , Chlorocebus aethiops , Infecciones por Coronavirus/veterinaria , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Interferones , Factor 88 de Diferenciación Mieloide , Proteínas de la Nucleocápside/fisiología , Virus de la Diarrea Epidémica Porcina/fisiología , Porcinos , Enfermedades de los Porcinos/virología , Ubiquitina-Proteína Ligasas , Células Vero , Interferón Tipo I/inmunología
6.
Vet Res ; 54(1): 106, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37968713

RESUMEN

African swine fever virus (ASFV) is a highly contagious and deadly virus that leads to high mortality rates in domestic swine populations. Although the envelope protein CD2v of ASFV has been implicated in immunomodulation, the molecular mechanisms underlying CD2v-mediated immunoregulation remain unclear. In this study, we generated a stable CD2v-expressing porcine macrophage (PAM-CD2v) line and investigated the CD2v-dependent transcriptomic landscape using RNA-seq. GO terms enrichment analysis and gene set enrichment analysis revealed that CD2v predominantly affected the organization and assembly process of the extracellular matrix. Wound healing and Transwell assays showed that CD2v inhibited swine macrophage migration. Further investigation revealed a significant decrease in the expression of transcription factor early growth response 1 (EGR1) through inhibiting the activity of extracellular signal-regulated kinase 1 and 2 (ERK1/2). Notably, EGR1 knockout in swine macrophages restricted cell migration, whereas EGR1 overexpression in PAM-CD2v restored the ability of macrophage migration, suggesting that CD2v inhibits swine macrophage motility by downregulating EGR1 expression. Furthermore, we performed chromatin immunoprecipitation and sequencing for EGR1 and the histone mark H3K27 acetylation (H3K27ac), and we found that EGR1 co-localized with the activated histone modification H3K27ac neighboring the transcriptional start sites. Further analysis indicated that EGR1 and H3K27ac co-occupy the promoter regions of cell locomotion-related genes. Finally, by treating various derivatives of swine macrophages with lipopolysaccharides, we showed that depletion of EGR1 decreased the expression of inflammatory cytokines including TNFα, IL1α, IL1ß, IL6, and IL8, which play essential roles in inflammation and host immune response. Collectively, our results provide new insights into the immunomodulatory mechanism of ASFV CD2v.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Enfermedades de los Porcinos , Porcinos , Animales , Virus de la Fiebre Porcina Africana/genética , Citocinas/genética , Citocinas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Virales/metabolismo , Macrófagos , Movimiento Celular
7.
Ecotoxicol Environ Saf ; 249: 114389, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36508791

RESUMEN

Hydroquinone (HQ), a well-known carcinogenic agent, induces oxidative stress, cell cycle arrest, apoptosis, and malignant transformation. As an antioxidant actor, the nuclear factor erythroid 2-related factor 2 (Nrf2) drives adaptive cellular protection in response to oxidative stress. The human lymphoblastoid cell line (TK6 cells) is widely used as a model for leukemia researches. In the present study, we focused on exploring whether Nrf2 regulatory cell cycle in TK6 cells upon HQ treatment and the underlying mechanisms. The results showed that the cell cycle arrest in TK6 cells induced by hydroquinone was accompanied by activation of the Nrf2 signaling pathway. We further clarified that Nrf2 loss accelerated cell cycle progression from G0/G1 to S and G2/M phases and promoted ROS production by downregulating the expression of SOD and GSH. Western blotting analysis indicated that Nrf2 regulated cell cycle progression via p16/pRb signaling pathways. Therefore, we conclude that Nrf2 is engaged in HQ-induced cell cycle arrest as well through p16/pRb and antioxidant enzymes.


Asunto(s)
Puntos de Control del Ciclo Celular , Hidroquinonas , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Humanos , Apoptosis , Puntos de Control del Ciclo Celular/efectos de los fármacos , Hidroquinonas/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal
8.
Environ Toxicol ; 38(10): 2344-2351, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37347496

RESUMEN

Hydroquinone (HQ) is an important metabolites of benzene in the body, and it has been found to result in cellular DNA damage, mutation, cell cycle imbalance, and malignant transformation. The JNK1 signaling pathway plays an important role in DNA damage repair. In this study, we focused on whether the JNK1 signaling pathway is involved in the HQ-induced cell cycle abnormalities and the underlying mechanism. The results showed that HQ induced abnormal progression of the cell cycle and initiated the JNK1 signaling pathway. We further confirmed that JNK1 suppression decelerated the cell cycle progression through inhibiting pRb/E2F1 signaling pathway and triggering p53/p21 pathway. Therefore, we concluded that JNK1 might be involved in HQ-induced malignant transformation associated with activating pRb/E2F1 and inhibiting p53/p21 signaling pathway which resulting in accelerating the cell cycle progression.


Asunto(s)
Hidroquinonas , Proteína p53 Supresora de Tumor , Proteína p53 Supresora de Tumor/metabolismo , Hidroquinonas/toxicidad , División Celular , Transducción de Señal
9.
10.
J Virol ; 95(19): e0064521, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34287043

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is a globally distributed alphacoronavirus that has reemerged lately, resulting in large economic losses. During viral infection, type I interferon (IFN-I) plays a vital role in the antiviral innate immunity. However, PEDV has evolved strategies to limit IFN-I production. To suppress virus replication, the host must activate IFN-stimulated genes and some host restriction factors to circumvent viral replication. This study observed that PEDV infection induced early growth response gene 1 (EGR1) expression in PEDV-permissive cells. EGR1 overexpression remarkably suppressed PEDV replication. In contrast, depletion of EGR1 led to a significant increase in viral replication. EGR1 suppressed PEDV replication by directly binding to the IFN-regulated antiviral (IRAV) promoter and upregulating IRAV expression. A detailed analysis revealed that IRAV interacts and colocalizes with the PEDV nucleocapsid (N) protein, inducing N protein degradation via the E3 ubiquitin ligase MARCH8 to catalyze N protein ubiquitination. Knockdown of endogenous MARCH8 significantly reversed IRAV-mediated N protein degradation. The collective findings demonstrate a new mechanism of EGR1-mediated viral restriction, in which EGR1 upregulates the expression of IRAV to degrade PEDV N protein through MARCH8. IMPORTANCE PEDV is a highly contagious enteric coronavirus that has rapidly emerged worldwide and has caused severe economic losses. No currently available drugs or vaccines can effectively control PEDV. PEDV has evolved many strategies to limit IFN-I production. We identified EGR1 as a novel host restriction factor and demonstrated that EGR1 suppresses PEDV replication by directly binding to the IRAV promoter and upregulating the expression of IRAV, which interacts with and degrades the PEDV N protein via the E3 ubiquitin ligase MARCH8 to catalyze nucleocapsid protein ubiquitination, which adds another layer of complexity to the innate antiviral immunity of this newly identified restriction factor. A better understanding of the innate immune response to PEDV infection will aid the development of novel therapeutic targets and more effective vaccines against virus infection.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/farmacología , Proteínas de la Nucleocápside/metabolismo , Virus de la Diarrea Epidémica Porcina/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Antivirales/metabolismo , Chlorocebus aethiops , Infecciones por Coronavirus , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Nucleocápside/metabolismo , Virus de la Diarrea Epidémica Porcina/genética , Porcinos , Enfermedades de los Porcinos/virología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Células Vero
11.
Arch Virol ; 166(7): 1903-1911, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33900472

RESUMEN

Tripartite motif protein 21 (TRIM21) is an E3 ubiquitin ligase and cytosolic antibody receptor of the TRIM family. Previous reports have indicated that TRIM21 plays an important role during viral infection. This study aimed at examining the role of TRIM21 in the replication of porcine epidemic diarrhea virus (PEDV) and showed that TRIM21 inhibits PEDV proliferation by targeting and degrading the nucleocapsid (N) protein through the proteasomal pathway. Furthermore, the endogenous expression of TRIM21 was found to be downregulated by PEDV infection in Vero and LLC-PK1 cells. Overexpression of TRIM21 inhibited PEDV replication, whereas knockdown of TRIM21 increased viral titers and N protein levels. TRIM21 was found to interact and colocalize with the N protein, and the TRIM21-mediated antiviral effect was dependent on its ubiquitin ligase activity, which engages in polyubiquitination and degradation of the N protein in a proteasome-dependent manner. Taken together, these findings provide information about the role of TRIM21 in PEDV proliferation and increase our understanding of host-virus interactions.


Asunto(s)
Proliferación Celular/fisiología , Infecciones por Coronavirus/metabolismo , Proteínas de la Nucleocápside/metabolismo , Virus de la Diarrea Epidémica Porcina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ribonucleoproteínas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Regulación hacia Abajo/fisiología , Células HEK293 , Células HeLa , Interacciones Microbiota-Huesped/fisiología , Humanos , Proteolisis , Porcinos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Células Vero , Replicación Viral/fisiología
12.
Opt Express ; 28(16): 24069-24078, 2020 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-32752392

RESUMEN

We theoretically investigate an controlled unidirectional reflectionlessness and near perfect absorption by applying external voltage in an electro-optical plasmonic waveguide system based on near-field coupling between two resonators. The system consists of two resonators side coupled to a metal-dielectric-metal plasmonic waveguide. Based on the numerical simulation, when external voltage is U = 7.4 V, the reflections for forward and backward directions are close to 0 and 0.82 at frequency 144.18 THz, while the reflections for forward and backward directions are close to 0.81 and 0 at frequency 150.86 THz when external voltage is U = 1.5 V. And the high absorption for forward (backward) direction is ∼0.97 (∼0.99) at frequency 144.18 THz (150.86 THz).

13.
Opt Express ; 27(21): 30589-30596, 2019 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-31684303

RESUMEN

In this work, we design a structure of metamaterials that consists of double sliver-ring resonators, in which highly-dispersive unidirectional reflectionlessness and absorption are achieved based on high-order plasmon resonance. Reflections of +z and -z directions at 461.34 THz (456.68 THz) are ∼0 (0.82) and ∼0.85 (0) when the distance d=222.9 nm (259.8 nm), respectively. High absorption of ∼0.97 and the quality factor of ∼435 can be obtained in the loss metal structure at room temperature. What's more, unidirectional reflectionlessness is investigated at low temperature.

14.
Biochem Biophys Res Commun ; 504(1): 157-163, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30172377

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) has been a major threat to global industrial pig farming ever since its emergence in the late 1980s. Identification of sustainable and effective control measures against PRRSV transmission is a pressing problem. The nucleocapsid (N) protein of PRRSV is specifically localized in the cytoplasm and nucleus of virus-infected cells which is important for PRRSV replication. In the current study, a new host restricted factor, Moloney leukemia virus 10-like protein (MOV10), was identified as an inhibitor of PRRSV replication. N protein levels and viral replication were significantly reduced in Marc-145 cells stably overexpressing MOV10 compared with those in wild-type Marc-145 cells. Adsorption experiments revealed that MOV10 did not affect the attachment and internalization of PRRSV. Co-immunoprecipitation and immunofluorescence co-localization analyses showed that MOV10 interacted and co-localized with the PRRSV N protein in the cytoplasm. Notably, MOV10 affected the distribution of N protein in the cytoplasm and nucleus, leading to the retention of N protein in the former. Taken together, these findings demonstrate for the first time that MOV10 inhibits PRRSV replication by restricting the nuclear import of N protein. These observations have great implications for the development of anti-PRRSV drugs and provide new insight into the role of N protein in PRRSV biology.


Asunto(s)
Citoplasma/metabolismo , Proteínas de la Nucleocápside/química , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , ARN Helicasas/metabolismo , Replicación Viral , Crianza de Animales Domésticos , Animales , Línea Celular , Chlorocebus aethiops , Replicación del ADN , Células HEK293 , Humanos , Virus de la Leucemia Murina de Moloney/metabolismo , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Unión Proteica , Porcinos , Proteínas no Estructurales Virales/metabolismo
15.
Proteomics ; 17(23-24)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29052333

RESUMEN

Significant differences exist between the highly pathogenic (HP) porcine reproductive and respiratory syndrome virus (PRRSV) and its attenuated pathogenic (AP) strain in the ability to infect host cells. The mechanisms by which different virulent strains invade host cells remain relatively unknown. In this study, pulmonary alveolar macrophages (PAMs) are infected with HP-PRRSV (HuN4) and AP-PRRSV (HuN4-F112) for 24 h, then harvested and subjected to label-free quantitative MS. A total of 2849 proteins are identified, including 95 that are differentially expressed. Among them, 26 proteins are located on the membrane. The most differentially expressed proteins are involved in response to stimulus, metabolic process, and immune system process, which mainly have the function of binding and catalytic activity. Cluster of differentiation CD163, vimentin (VIM), and nmII as well as detected proteins are assessed together by string analysis, which elucidated a potentially different infection mechanism. According to the function annotations, PRRSV with different virulence may mainly differ in immunology, inflammation, immune evasion as well as cell apoptosis. This is the first attempt to explore the differential characteristics between HP-PRRSV and its attenuated PRRSV infected PAMs focusing on membrane proteins which will be of great help to further understand the different infective mechanisms of HP-PRRSV and AP-PRRSV.


Asunto(s)
Macrófagos Alveolares/metabolismo , Proteínas de la Membrana/análisis , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/patogenicidad , Proteoma/análisis , Proteómica/métodos , Alveolos Pulmonares/metabolismo , Animales , Células Cultivadas , Interacciones Huésped-Patógeno , Macrófagos Alveolares/virología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/clasificación , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , Alveolos Pulmonares/virología , Porcinos , Virulencia
16.
Virol J ; 11: 201, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25420583

RESUMEN

BACKGROUND: Porcine reproductive and respiratory syndrome virus (PRRSV) continues to be an important problem for the swine industry. Inactivated vaccines and modified-live virus vaccines are widely used in the field; however, the efficacy of these PRRSV vaccines is suboptimal due to poor immunogenicity. Granulocyte-macrophage colony stimulating factor (GM-CSF) has been extensively used as an effective genetic and protein adjuvant to enhance the efficiencies vaccines expressing tumor or pathogen antigens. The purpose of this study was to determine if GM-CSF could increase the efficiency of PRRSV vaccine. METHODS: The GM-CSF gene was inserted in the HuN4-F112 vaccine strain by overlap PCR. The expression of GM-CSF by the recombinant virus was confirmed with methods of indirect immunofluorescent assay (IFA) and Western blotting. The stability of recombinant virus was assessed by cDNA sequence and IFA after 20 passages. To detect the biological activity of GM-CSF expressed by the recombinant virus, bone marrow-derived dendritic cells (BMDCs) were isolated and co-cultured with the recombinant virus or parental virus and the surface phenotypes of BMDCs were examined by flow cytometric analysis. The cytokines secreted by BMDCs infected with PRRSV, or treated with LPS, GM-CSF or medium alone were evaluated by ProcartaPlexTM Multiplex Immunoassays and qRT-PCR. RESULTS: A novel modified-live PRRSV vaccine strain expressing GM-CSF (rHuN4-GM-CSF) was successfully constructed and rescued. The GM-CSF protein was stable expressed in recombinant virus-infected cells after 20 passages. Analysis of virus replication kinetics showed that the novel vaccine strain expressing GM-CSF had a similar replication rate as the parental virus. In vitro studies showed that infection of porcine BMDCs with rHuN4-GM-CSF resulted in increased surface expression of MHCI+, MHCII + and CD80/86+ that was dependent on virus expressed GM-CSF. The expression of representative cytokines was significantly up-regulated when BMDCs were incubated with the recombinant GM-CSF expressing virus. CONCLUSIONS: Our results indicated that the expression of GM-CSF during infection with a vaccine strain could enhance the activation of BMDCs and increase cytokine response, which is expected to result in higher immune responses and may improve vaccine efficacy against PRRSV infection.


Asunto(s)
Adyuvantes Inmunológicos/genética , Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , Replicación Viral , Animales , Línea Celular , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/virología , Inestabilidad Genómica , Datos de Secuencia Molecular , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , ARN Viral/genética , Análisis de Secuencia de ADN , Cultivo de Virus
17.
Antiviral Res ; 223: 105825, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38311297

RESUMEN

Feline coronavirus (FCoV) is an unsegmented, single-stranded RNA virus belonging to the Alphacoronavirus genus. It can cause fatal feline infectious peritonitis (FIP) in cats of any ages. Currently, there are no effective prevention and control measures to against FCoV. In this study, we developed a recombinant adenovirus vaccine, AD5-N, based on the nucleocapsid(N) protein of FCoV. The immunogenicity of AD5-N was evaluated through intramuscular immunization in 6-week-old Balb/c mice and 9-12 months old cats. Compared to the control group, AD5-N specifically induced a significant increase in IgG and SIgA levels in the vaccinated mice. Furthermore, AD5-N not only effectively promoted strong cellular immune responses in cats but also induced high levels of specific SIgA, effectively helping cats resist FCoV infection. Our findings suggest that adenovirus vector vaccines based on the N gene have the potential to become candidate vaccines for the prevention and control of FCoV infection.


Asunto(s)
Infecciones por Adenoviridae , Vacunas contra el Adenovirus , Infecciones por Coronavirus , Coronavirus Felino , Vacunas , Gatos , Animales , Ratones , Adenoviridae/genética , Coronavirus Felino/genética , Inmunoglobulina A Secretora , Ratones Endogámicos BALB C , Inmunidad
18.
Front Immunol ; 15: 1328266, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38550592

RESUMEN

Background: Porcine deltacoronavirus (PDCoV), a novel swine enteropathogenic coronavirus, challenges the global swine industry. Currently, there are no approaches preventing swine from PDCoV infection. Methods: A new PDCoV strain named JS2211 was isolated. Next, the dimer receptor binding domain of PDCoV spike protein (RBD-dimer) was expressed using the prokaryotic expression system, and a novel nanoparticle containing RBD-dimer and ferritin (SC-Fe) was constructed using the SpyTag/SpyCatcher system. Finally, the immunoprotection of RBD-Fe nanoparticles was evaluated in mice. Results: The novel PDCoV strain was located in the clade of the late Chinese isolate strains and close to the United States strains. The RBD-Fe nanoparticles were successfully established. Immune responses of the homologous prime-boost regime showed that RBD-Fe nanoparticles efficiently elicited specific humoral and cellular immune responses in mice. Notably, high level PDCoV RBD-specific IgG and neutralizing antibody (NA) could be detected, and the histopathological results showed that PDCoV infection was dramatically reduced in mice immunized with RBD-Fe nanoparticles. Conclusion: This study effectively developed a candidate nanoparticle with receptor binding domain of PDCoV spike protein that offers protection against PDCoV infection in mice.


Asunto(s)
Nanovacunas , Glicoproteína de la Espiga del Coronavirus , Porcinos , Animales , Ratones , Deltacoronavirus , Inmunidad , SARS-CoV-2
19.
Toxicol In Vitro ; 100: 105901, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39029599

RESUMEN

Hydroquinone (HQ) is one of benzene metabolites that can cause oxidative stress damage and Homologous recombination repair (HR). A good deal of reactive oxygen species (ROS) generated by oxidative stress can trigger apoptotic signaling pathways. The nuclear factor erythroid 2-related factor 2 (Nrf2) can regulate the cell response to oxidative stress damage. The aim of this study was to explore whether Nrf2 participate in HQ-induced apoptosis and its mechanism. The findings displayed that HQ triggered HR, promoted Nrf2 transfer into the cell nucleus and induced cell apoptosis, while Nrf2 deficient elevated cell apoptosis, attenuated the expression of PARP1 and RAD51. We also observed that Nrf2 deficient triggered Caspase-9. Thus, we speculated that Nrf2 might participate in HQ-induced cell apoptosis through Caspase-9 dependent pathways. Meanwhile, Nrf2 participated in HQ-induced DNA damage repair by regulating the level of PARP1 and RAD51.


Asunto(s)
Apoptosis , Daño del ADN , Hidroquinonas , Factor 2 Relacionado con NF-E2 , Poli(ADP-Ribosa) Polimerasa-1 , Recombinasa Rad51 , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Hidroquinonas/toxicidad , Apoptosis/efectos de los fármacos , Humanos , Línea Celular , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/genética , Recombinasa Rad51/metabolismo , Recombinasa Rad51/genética , Caspasa 9/metabolismo , Reparación del ADN/efectos de los fármacos
20.
Front Plant Sci ; 14: 1169898, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37600201

RESUMEN

The Heilongjiang-Amur River Basin is one of the largest and most complex aquatic systems in Asia, comprising diverse wetland resources. The wetland vegetation in mid-high latitude areas has high natural value and is sensitive to climate changes. In this study, we investigated the wetland vegetation cover changes and associated responses to climate change in the Heilongjiang-Amur River Basin from 2000 to 2018 based on the growing season (May to September) climate and LAI data. Our results indicated that the wetland LAI increased at 0.014 m2·m-2/yr across Heilongjiang-Amur River Basin with the regional climate showed wetting and warming trends. On a regional scale, wetland vegetation in China and Russia had positive partial correlation with solar radiation and minimum air temperature, with precipitation showing a slight lag effect. In contrast, wetland vegetation in Mongolia had positive partial correlation with precipitation. These correlations were further investigated at different climate intervals. We found the precipitation is positively correlated with LAI in the warm regions while is negatively correlated with LAI in the wet regions, indicating an increase in precipitation is beneficial for the growth of wetland vegetation in heat sufficient areas, and when precipitation exceeds a certain threshold, it will hinder the growth of wetland vegetation. In the cold regions, we found solar radiation and minimum air temperature are positively correlated with LAI, suggesting SR and minimum air temperature instead of mean air temperature and maximum air temperature play more important roles in affecting the wetland vegetation growth in the heat limited areas. The LAI was found to be negatively correlated with maximum air temperature in the arid areas, indicating excessive temperature would inhibit the wetland vegetation growth when the water is limited. Our investigation can provide a scientific foundation for the trilateral region in wetland ecosystem protection and is beneficial for a more comprehensive understanding of the responses of wetlands in the middle and high latitudes to climate change.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA