Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 97(7): e0053223, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37367226

RESUMEN

During viral infection, host defensive proteins either enhance the host immune response or antagonize viral components directly. In this study, we report on the following two mechanisms employed by zebrafish mitogen-activated protein kinase kinase 7 (MAP2K7) to protect the host during spring viremia of carp virus (SVCV) infection: stabilization of host IRF7 and degradation of SVCV P protein. In vivo, map2k7+/- (map2k7-/- is a lethal mutation) zebrafish showed a higher lethality, more pronounced tissue damage, and more viral proteins in major immune organs than the controls. At the cellular level, overexpression of map2k7 significantly enhanced host cell antiviral capacity, and viral replication and proliferation were significantly suppressed. Additionally, MAP2K7 interacted with the C terminus of IRF7 and stabilized IRF7 by increasing K63-linked polyubiquitination. On the other hand, during MAP2K7 overexpression, SVCV P proteins were significantly decreased. Further analysis demonstrated that SVCV P protein was degraded by the ubiquitin-proteasome pathway, as the attenuation of K63-linked polyubiquitination was mediated by MAP2K7. Furthermore, the deubiquitinase USP7 was indispensable in P protein degradation. These results confirm the dual functions of MAP2K7 during viral infection. IMPORTANCE Normally, during viral infection, host antiviral factors individually modulate the host immune response or antagonize viral components to defense infection. In the present study, we report that zebrafish MAP2K7 plays a crucial positive role in the host antiviral process. According to the weaker antiviral capacity of map2k7+/- zebrafish than that of the control, we find that MAP2K7 reduces host lethality through two pathways, as follows: enhancing K63-linked polyubiquitination to promote host IRF7 stability and attenuating K63-mediated polyubiquitination to degrade the SVCV P protein. These two mechanisms of MAP2K7 reveal a special antiviral response in lower vertebrates.


Asunto(s)
Enfermedades de los Peces , Factores Reguladores del Interferón , Proteínas Quinasas Activadas por Mitógenos , Infecciones por Rhabdoviridae , Ubiquitinación , Proteínas Estructurales Virales , Animales , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Rhabdoviridae/genética , Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/virología , Pez Cebra/genética , Pez Cebra/inmunología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Estabilidad Proteica , Proteolisis , Proteínas Estructurales Virales/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Regulación hacia Arriba
2.
J Immunol ; 208(5): 1189-1203, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35101889

RESUMEN

The small HERC family currently comprises four members (HERC3-6) involved in the regulation of various physiological activities. Little is known about the role of HERCs in IFN response. In this study, we identify a novel fish HERC member, named crucian carp HERC7, as a negative regulator of fish IFN response. Genome-wide search of homologs and comprehensive phylogenetic analyses reveal that the small HERC family, apart from HERC3-6 that have been well-characterized in mammals, contains a novel HERC7 subfamily exclusively in nonmammalian vertebrates. Lineage-specific and even species-specific expansion of HERC7 subfamily in fish indicates that crucian carp HERC7 might be species-specific. In virally infected fish cells, HERC7 is induced by IFN and selectively targets three retinoic acid-inducible gene-I-like receptor signaling factors for degradation to attenuate IFN response by two distinct strategies. Mechanistically, HERC7 delivers mediator of IFN regulatory factor 3 activator and mitochondrial antiviral signaling protein for proteasome-dependent degradation at the protein level and facilitates IFN regulatory factor 7 transcript decay at the mRNA level, thus abrogating cellular IFN induction to promote virus replication. Whereas HERC7 is a putative E3 ligase, the E3 ligase activity is not required for its negative regulatory function. These results demonstrate that the ongoing expansion of the small HERC family generates a novel HERC7 to fine-tune fish IFN antiviral response.


Asunto(s)
Factor 7 Regulador del Interferón/metabolismo , Interferones/inmunología , Reoviridae/inmunología , Rhabdoviridae/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Carpas , Línea Celular , Proteínas de Peces/genética , Células HEK293 , Humanos , Factor 7 Regulador del Interferón/genética , Proteínas de la Membrana/metabolismo , Estabilidad del ARN/genética , ARN Mensajero/genética , Transducción de Señal/inmunología , Transactivadores/genética
3.
J Immunol ; 208(5): 1099-1114, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35101892

RESUMEN

Circular RNA (circRNA) is produced by splicing head to tail and is widely distributed in multicellular organisms, and circRNA reportedly can participate in various cell biological processes. In this study, we discovered a novel exon-intron circRNA derived from probable E3 ubiquitin-protein ligase RNF217 (RNF217) gene, namely, circRNF217, which was related to the antibacterial responses in teleost fish. Results indicated that circRNF217 played essential roles in host antibacterial immunity and inhibited the Vibrio anguillarum invasion into cells. Our study also found a microRNA miR-130-3p, which could inhibit antibacterial immune response and promote V. anguillarum invasion into cells by targeting NOD1. Moreover, we also found that the antibacterial effect inhibited by miR-130-3p could be reversed with circRNF217. In mechanism, our data revealed that circRNF217 was a competing endogenous RNA of NOD1 by sponging miR-130-3p, leading to activation of the NF-κB pathway and then enhancing the innate antibacterial responses. In addition, we also found that circRNF217 can promote the antiviral response caused by Siniperca chuatsi rhabdovirus through targeting NOD1. Our study provides new insights for understanding the impact of circRNA on host-pathogen interactions and formulating fish disease prevention to resist the severely harmful V. anguillarum infection.


Asunto(s)
Enfermedades de los Peces/inmunología , Inmunidad Innata/inmunología , MicroARNs/genética , Percas/inmunología , ARN Circular/genética , Vibrio/inmunología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/prevención & control , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Percas/virología , Rhabdoviridae/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
PLoS Pathog ; 17(3): e1009438, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33735323

RESUMEN

Circular RNAs (circRNAs) represent a class of widespread and diverse covalently closed circular endogenous RNAs that exert crucial functions in regulating gene expression in mammals. However, the function and regulation mechanism of circRNAs in lower vertebrates are still unknown. Here, we discovered a novel circRNA derived from Deltex E3 ubiquitin ligase 1 (Dtx1) gene, namely, circDtx1, which was related to the antiviral responses in teleost fish. Results indicated that circDtx1 played essential roles in host antiviral immunity and inhibition of SCRV replication. Our study also found a microRNA miR-15a-5p, which could inhibit antiviral immune response and promote viral replication by targeting TRIF. Moreover, we also found that the antiviral effect inhibited by miR-15a-5p could be reversed with the circDtx1. In mechanism, our data revealed that circDtx1 was a competing endogenous RNA (ceRNA) of TRIF by sponging miR-15a-5p, leading to activation of the NF-κB/IRF3 pathway, and then enhancing the innate antiviral responses. Our results indicated that circRNAs played a regulatory role in immune responses in teleost fish.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/biosíntesis , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/genética , Factor 3 Regulador del Interferón/inmunología , MicroARNs/inmunología , ARN Circular/inmunología , Animales , Regulación hacia Abajo , Inmunidad Innata/inmunología , Perciformes , Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/inmunología
5.
J Immunol ; 207(10): 2570-2580, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34654690

RESUMEN

TNFR-associated factor 6 (TRAF6) not only recruits TBK1/IKKε to MAVS upon virus infection but also catalyzes K63-linked polyubiquitination on substrate or itself, which is critical for NEMO-dependent and -independent TBK1/IKKε activation, leading to the production of type I IFNs. The regulation at the TRAF6 level could affect the activation of antiviral innate immunity. In this study, we demonstrate that zebrafish prmt2, a type I arginine methyltransferase, attenuates traf6-mediated antiviral response. Prmt2 binds to the C terminus of traf6 to catalyze arginine asymmetric dimethylation of traf6 at arginine 100, preventing its K63-linked autoubiquitination, which results in the suppression of traf6 activation. In addition, it seems that the N terminus of prmt2 competes with mavs for traf6 binding and prevents the recruitment of tbk1/ikkε to mavs. By zebrafish model, we show that loss of prmt2 promotes the survival ratio of zebrafish larvae after challenge with spring viremia of carp virus. Therefore, we reveal, to our knowledge, a novel function of prmt2 in the negative regulation of antiviral innate immunity by targeting traf6.


Asunto(s)
Inmunidad Innata/inmunología , Proteína-Arginina N-Metiltransferasas/inmunología , Infecciones por Rhabdoviridae/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología , Animales , Rhabdoviridae/inmunología , Pez Cebra
6.
J Immunol ; 207(2): 371-375, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34233911

RESUMEN

The repertoire of Abs is generated by genomic rearrangements during B cell differentiation. Although V(D)J rearrangements lead to repertoires mostly different between individuals, recent studies have shown that they contain a substantial fraction of overrepresented and shared "public" clones. We previously reported a strong public IgHµ clonotypic response against the rhabdovirus viral hemorrhagic septicemia virus in a teleost fish. In this study, we identified an IgL chain associated with this public response that allowed us to characterize its functionality. We show that this public Ab response has a potent neutralizing capacity that is typically associated with host protection during rhabdovirus infections. We also demonstrate that the public response is not restricted to a particular trout isogenic line but expressed in multiple genetic backgrounds and may be used as a marker of successful vaccination. Our work reveals that public B cell responses producing generic Abs constitute a mechanism of protection against infection conserved across vertebrates.


Asunto(s)
Formación de Anticuerpos/inmunología , Peces/inmunología , Mamíferos/inmunología , Animales , Linfocitos B/inmunología , Células Clonales/inmunología , Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/inmunología , Recombinación V(D)J/inmunología , Vacunación/métodos
7.
PLoS Pathog ; 16(7): e1008670, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32678830

RESUMEN

Increasing evidence suggests important roles for long noncoding RNAs (lncRNAs) as new gene modulators involved in various biological processes. However, the function roles of lncRNAs in lower vertebrates are still unknown. Here, we firstly identify a lncRNA, named MAVS antiviral-related lncRNA (MARL), as a key regulator for antiviral immunity in teleost fish. The results indicate that fish MAVS play essential roles in host antiviral responses and inhibition of Siniperca chuatsi rhabdovirus (SCRV) replication. miR-122 reduces MAVS expression and suppress MAVS-mediated antiviral responses, which may help viruses evade host antiviral responses. Further, MARL functions as a competing endogenous RNA (ceRNA) for miR-122 to control protein abundance of MAVS, thereby inhibiting SCRV replication and promoting antiviral responses. Our data not only shed new light on understanding the function role of lncRNA in biological processes in lower vertebrates, but confirmed the hypothesis that ceRNA regulatory networks exist widely in vertebrates.


Asunto(s)
MicroARNs/metabolismo , Perciformes/inmunología , ARN Largo no Codificante/inmunología , Infecciones por Rhabdoviridae/inmunología , Animales , Regulación hacia Abajo , Perciformes/virología , Rhabdoviridae/inmunología
8.
J Immunol ; 205(1): 237-250, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32471880

RESUMEN

In mammals, transcription factors of IFN-regulatory factors (IRFs) family translate viral recognition into IFN antiviral responses through translocating to nucleus and subsequently binding to the promoters of IFN and IFN-stimulated genes (ISGs). In addition to IRF1-9 conserved across vertebrates and IRF10 in teleost fish and bird, teleost fish has another novel member, IRF11; however, little is known about its role in IFN response. In this study, we provide evidence that IRF11 is present only in Osteichthyes (bony fish) but lost in tetrapods and subsequently characterize the stimulatory potential of zebrafish IRF11 to IFN antiviral response relevant to its subcellular localization and promoter binding. Overexpression of zebrafish IRF11 restricts virus replication through induction of IFN and ISGs. Zebrafish IRF11 is constitutively localized to nucleus, which is driven by a tripartite NLS motif, consisting of three interdependent basic clusters, two in DNA binding domain (DBD) and one in the region immediately C-terminal to DBD. Nuclear IRF11 binds to the IRF-binding element/IFN-stimulated response element motifs of zebrafish IFN promoters depending on the two conserved amino acids (K78, R82) within DBD helix α3. K78 and R82 also benefit zebrafish IRF11 nuclear import as two key residues positioned at the first basic cluster of the tripartite NLS motif. Such features enable zebrafish IRF11 to function as a positive transcription factor for fish IFN antiviral response. Our results identify a unique tripartite NLS motif that integrates DNA-binding activity and nuclear import ability, allowing zebrafish IRF11 to initiate IFN and ISG expression.


Asunto(s)
Factor 1 Regulador del Interferón/metabolismo , Interferones/genética , Infecciones por Rhabdoviridae/veterinaria , Factores de Transcripción TFII/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/inmunología , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos/genética , Animales , Línea Celular , Núcleo Celular/metabolismo , Secuencia Conservada/genética , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Factor 1 Regulador del Interferón/genética , Interferones/metabolismo , Regiones Promotoras Genéticas/genética , Dominios Proteicos/genética , Elementos de Respuesta , Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/virología , Transducción de Señal/genética , Factores de Transcripción TFII/genética , Replicación Viral/inmunología , Pez Cebra/genética , Pez Cebra/metabolismo , Pez Cebra/virología
9.
FASEB J ; 34(8): 10212-10227, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32643209

RESUMEN

Arginine methylation catalyzed by protein arginine methyltransferases (PRMT) is a common post-translational modification in histone and nonhistone proteins, which regulates many cellular functions. Protein arginine methyltransferase 3 (prmt3), a type I arginine methyltransferase, has been shown to carry out the formation of stable monomethylarginine as an intermediate before the establishment of asymmetric dimethylarginine. To date, however, the role of PRMT3 in antiviral innate immunity has not been elucidated. This study showed that zebrafish prmt3 was upregulated by virus infection and that the overexpression of prmt3 suppressed cellular antiviral response. The PRMT3 inhibitor, SGC707, enhanced antiviral capability. Consistently, prmt3-null zebrafish were more resistant to Spring Viremia of Carp Virus (SVCV) and Grass Carp Reovirus (GCRV) infection. Further assays showed that the overexpression of prmt3 diminished the phosphorylation of irf3 and prmt3 interacted with rig-i. In addition, both zinc-finger domain and catalytic domain of prmt3 were required for the suppressive function of prmt3 on IFN activation. Our findings suggested that zebrafish prmt3 negatively regulated the antiviral responses, implicating the vital role of prmt3-or even arginine methylation-in antiviral innate immunity.


Asunto(s)
Antivirales/inmunología , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/inmunología , Pez Cebra/genética , Pez Cebra/inmunología , Animales , Células Cultivadas , Histonas/genética , Histonas/inmunología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Isoquinolinas/inmunología , Metilación , Fosforilación/genética , Fosforilación/inmunología , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/inmunología , Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/genética , Infecciones por Rhabdoviridae/inmunología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Virosis/genética , Virosis/inmunología , Virosis/virología , Pez Cebra/virología , Dedos de Zinc/genética , Dedos de Zinc/inmunología
10.
J Fish Dis ; 44(10): 1587-1594, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34165796

RESUMEN

Spring viraemia of carp (SVC) caused by spring viraemia of carp virus (SVCV) can infect almost all fish of cyprinids, which bring huge economic losses to aquaculture. Glycoprotein (G), as the most important antigenic determinant protein of SVCV, is widely considered as an effective method against SVCV. In our previous study, we found that G3 (131 aa) is the potential dominant antigen epitope that induces strong immune responses similar to G protein (510 aa). Here, in order to further improve the immune effect, we reported a subunit vaccine (PEG-G3) constructed by PEG-modified dominant epitope protein (G3). The results of serum antibody production, enzyme activities and immune-related genes expression showed that PEG-G3 induces significantly stronger immune protective responses against SVCV than G3. PEG modification significantly increased the serum antibody level of the vaccine, which increased significantly after immunization and reached the peak at 21 day post-vaccination. T-AOC and AKP activities in the lowest concentration group (5 µg) of PEG-G3 were significantly higher than those in the highest concentration group (20 µg) of G3. In PEG-G3 group, the expression of almost all genes increased at least 4 times compared with the control group. After 14-day challenge, the RPS (relative percentage survival) of the highest concentration of PEG-G3 group was 53.6%, while that of G3 group is 38.9%. Therefore, this work shows that PEG modification and dominant epitope screening may be effective methods to improve the immune protective effect of vaccines and to resist the infection of aquatic animal viral diseases.


Asunto(s)
Carpas , Enfermedades de los Peces/prevención & control , Inmunización/veterinaria , Infecciones por Rhabdoviridae/veterinaria , Rhabdoviridae/inmunología , Vacunas de Subunidad/inmunología , Vacunas Virales/inmunología , Animales , Epítopos/inmunología , Enfermedades de los Peces/virología , Inmunidad , Infecciones por Rhabdoviridae/prevención & control , Infecciones por Rhabdoviridae/virología , Vacunas de Subunidad/administración & dosificación , Vacunas Virales/administración & dosificación
11.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31413136

RESUMEN

Interferon (IFN) production activated by phosphorylated interferon regulatory factor 7 (IRF7) is a pivotal process during host antiviral infection. For viruses, suppressing the host IFN response is beneficial for viral proliferation; in such cases, evoking host-derived IFN negative regulators would be very useful for viruses. Here, we report that the zebrafish rapunzel 5 (RPZ5) protein which activated by virus degraded phosphorylated IRF7 is activated by TANK-binding kinase 1 (TBK1), leading to a reduction in IFN production. Upon viral infection, zebrafish rpz5 was significantly upregulated, as was ifn, in response to the stimulation. Overexpression of RPZ5 blunted the IFN expression induced by both viral and retinoic acid-inducible gene I (RIG-I) like-receptor (RLR) factors. Subsequently, RPZ5 interacted with RLRs but did not affect the stabilization of the proteins in the normal state. Interestingly, RPZ5 degraded the phosphorylated IRF7 under TBK1 activation through K48-linked ubiquitination. Finally, the overexpression of RPZ5 remarkably reduced the host cell antiviral capacity. These findings suggest that zebrafish RPZ5 is a negative regulator of phosphorylated IRF7 and attenuates IFN expression during viral infection, providing insight into the IFN balance mechanism in fish.IMPORTANCE The phosphorylation of IRF7 is helpful for host IFN production to defend against viral infection; thus, it is a potential target for viruses to mitigate the antiviral response. We report that the fish RPZ5 is an IFN negative regulator induced by fish viruses and degrades the phosphorylated IRF7 activated by TBK1, leading to IFN suppression and promotion of viral proliferation. These findings reveal a novel mechanism for interactions between the host cell and viruses in the lower vertebrate.


Asunto(s)
Enfermedades de los Peces/virología , Inmunidad Innata/inmunología , Interferones/metabolismo , Infecciones por Rhabdoviridae/veterinaria , Rhabdoviridae/inmunología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/virología , Animales , Antivirales/inmunología , Antivirales/metabolismo , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/metabolismo , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferones/inmunología , Fosforilación , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/metabolismo , Infecciones por Rhabdoviridae/virología , Ubiquitinación , Replicación Viral , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética
12.
Fish Shellfish Immunol ; 105: 327-329, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32721570

RESUMEN

Spring viremia of carp (SVC) is highly contagious and lethal disease in cyprinid fish, in particular common carps (Cyprinus carpio), causing numerous economic losses to the aquaculture industry. SVC is presently endemic disease in Europe, America, and several countries in Asia and its causative agent is spring viremia of carp virus (SVCV). In this study, a chitosan-alginate microcapsule probiotic vaccine expressing G protein of SVCV was prepared, and the immunogenicity in carps of orally administrated with the microcapsule probiotic vaccine was evaluated. Our results showed that the microcapsule probiotic vaccine can induce potent antigen-specific immune responses in carps via oral vaccination, and provide effective anti-SVCV protection for carps. Significantly, the microcapsule probiotic vaccine is suitable for mass fish immunization, suggesting a promising vaccine strategy for fish.


Asunto(s)
Alginatos/administración & dosificación , Carpas/inmunología , Quitosano/administración & dosificación , Inmunización/veterinaria , Probióticos/administración & dosificación , Infecciones por Rhabdoviridae/veterinaria , Vacunas Virales/inmunología , Administración Oral , Animales , Cápsulas , Enfermedades de los Peces/prevención & control , Rhabdoviridae/genética , Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/prevención & control , Infecciones por Rhabdoviridae/virología , Proteínas Virales/química , Vacunas Virales/administración & dosificación
13.
Fish Shellfish Immunol ; 98: 429-437, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31988017

RESUMEN

Oxyeleotris marmoratus iridovirus (OMIV) and Oxyeleotris marmoratus rhabdovirus (OMRV) are the two major causative agents of disease leading to massive mortality and severe economic losses in marbled sleepy goby (Oxyeleotris marmoratus) industry. It's urgent to develop an effective vaccine against these fatal diseases. In this study, we developed bivalent inactivated vaccine against OMIV and OMRV and evaluated its protective effect in Oxyeleotris marmoratus. The intraperitoneally vaccinated fish were protected against challenge with OMIV and OMRV with both relative percent survival (RPS) of 100%. In addition, deep RNA sequencing was used to analyze the transcriptomic profiles of the spleen tissues at progressive time points post-vaccination with bivalent inactivated vaccine and challenge with OMIV and OMRV infection. Results showed that adaptive immune response was induced in Oxyeleotris marmoratus injected with bivalent inactivated vaccine. Furthermore, robust adaptive immune responses were also detected in vaccinated fish at 7 d and 2 d post-challenge with OMIV and OMRV. Taken together, these results indicated that bivalent inactivated vaccine activated adaptive immune responses in Oxyeleotris marmoratus, and provided protection against OMIV and OMRV lethal challenge.


Asunto(s)
Infecciones por Virus ADN/veterinaria , Enfermedades de los Peces/prevención & control , Iridovirus/inmunología , Perciformes , Rhabdoviridae/inmunología , Vacunas Virales/inmunología , Inmunidad Adaptativa , Animales , Infecciones por Virus ADN/prevención & control , Enfermedades de los Peces/virología , Infecciones por Rhabdoviridae/prevención & control , Infecciones por Rhabdoviridae/veterinaria , Infecciones por Rhabdoviridae/virología , Transcriptoma/inmunología , Vacunas de Productos Inactivados/inmunología
14.
Fish Shellfish Immunol ; 104: 262-268, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32534229

RESUMEN

Spring viremia of carp virus (SVCV) causes devastating disease in aquaculture, resulting in significant economic impact. To develop an effective means against SVCV infection, a Lactococcus lactis (L.lactis) based subunit vaccine (pNZ-UGA) was developed based on surface displaying of SVCV glycoprotein using anchoring motif of the cA (C terminus of the peptidoglyvsn-binding) domains of AcmA, a major autolysin from L.lactis. The surface expression of SVCV glycoprotein was verified by indirect immunofluorescence assay. The efficacy of the constructed vaccine was further evaluated in common carp. The results showed that the higher levels of specific IgM could be detected in fish vaccinated with pNZ-UGA, compared with that in PBS and L.lactis groups. Immune-related genes including TNF-α, IL-6b, IL-1ß, Cxcr 1, Cxca, IFNg2b, I-IFN, and IgM expression in pNZ-UGA group were strongly up-regulated, revealing that robust innate immune response was induced. Notably, the lowest cumulative mortality (13.46%) was observed in fish vaccinated with pNZ-UGA vaccine after SVCV challenge, whereas the cumulative mortality were 100.00% and 92.31% in PBS and L.lactis groups, respectively. This study suggests the potential use of the recombinant L.lactis with surface displaying antigen proteins as effective vaccines against SVCV and other fish virus infection.


Asunto(s)
Carpas/inmunología , Enfermedades de los Peces/prevención & control , Glicoproteínas/inmunología , Lactococcus lactis/inmunología , Infecciones por Rhabdoviridae/prevención & control , Rhabdoviridae/inmunología , Vacunas de Subunidad/administración & dosificación , Proteínas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Carpas/sangre , Carpas/virología , Citocinas/genética , Citocinas/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Lactococcus lactis/genética , Infecciones por Rhabdoviridae/veterinaria
15.
J Immunol ; 201(3): 982-994, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29967101

RESUMEN

Viral infection induces type I IFN production, which plays critical roles in orchestrating the antiviral defense by inducing direct antiviral activities. To establish a persistent infection, viruses have evolved numerous strategies to specifically interfere with IFN production or its downstream mediators, thereby evading the immune responses. MicroRNAs (miRNAs) are a family of small noncoding RNAs that posttranscriptionally regulate the expressions of specific target genes. Although accumulating evidence demonstrates that miRNAs play vital roles in regulating viral infection, miRNAs that target intracellular sensors and adaptors of innate immunity have not been fully uncovered. In this paper, we identify fish miR-210 as a robust regulator involved in regulating virus-host interactions. We found that rhabdovirus significantly upregulated the expression of fish miR-210. Inducible miR-210 modulates virus-triggered type I IFN and inflammatory cytokine production by targeting stimulator of IFN genes (STING), thereby promoting viral replication. Furthermore, we demonstrated that miR-210 regulates innate immune response through NF-κB, IFN regulatory factor 3, and JAK/STAT signaling pathways. The collective findings indicate that inducible miR-210 plays a regulatory role in virus-host interactions through STING-mediated singling pathway by targeting STING.


Asunto(s)
Peces/inmunología , Inmunidad Innata/inmunología , Proteínas de la Membrana/metabolismo , MicroARNs/inmunología , Rhabdoviridae/inmunología , Transducción de Señal/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Marcación de Gen/métodos , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/inmunología , Humanos , Regulación hacia Arriba/inmunología , Replicación Viral/inmunología
16.
J Virol ; 92(2)2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29093090

RESUMEN

Effectively recognizing invading viruses and subsequently inducing innate antiviral immunity are essential for host antiviral defense. Although these processes are closely regulated by the host to maintain immune balance, viruses have evolved the ability to downregulate or upregulate these processes for their survival. MicroRNAs (miRNAs) are a family of small noncoding RNAs that play vital roles in modulating host immune response. Accumulating evidence demonstrates that host miRNAs as mediators are involved in regulating viral replication and host antiviral immunity in mammals. However, the underlying regulatory mechanisms in fish species are still poorly understood. Here, we found that rhabdovirus infection significantly upregulated host miR-3570 expression in miiuy croaker macrophages. Induced miR-3570 negatively modulated RNA virus-triggered type I interferon (IFN) and antiviral gene production, thus facilitating viral replication. Furthermore, miR-3570 was found to target and posttranscriptionally downregulate mitochondrial antiviral signaling protein (MAVS), which functions as a platform for innate antiviral signal transduction. Moreover, we demonstrated that miR-3570 suppressed the expression of MAVS, thereby inhibiting MAVS-mediated NF-κB and IRF3 signaling. The collective results demonstrated a novel regulation mechanism of MAVS-mediated immunity during RNA viral infection by miRNA.IMPORTANCE RNA viral infection could upregulate host miR-3570 expression in miiuy croaker macrophages. Induced miR-3570 negatively modulates RNA virus-triggered type I IFN and antiviral gene production, thus facilitating viral replication. Remarkably, miR-3570 could target and inhibit MAVS expression, which thus modulates MAVS-mediated NF-κB and IRF3 signaling. The collective results of this study suggest a novel regulation mechanism of MAVS-mediated immunity during RNA viral infection by miR-3570. Thus, a novel mechanism for virus evasion in fish is proposed.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/genética , MicroARNs/genética , Interferencia de ARN , Rhabdoviridae/inmunología , Regiones no Traducidas 3' , Animales , Enfermedades de los Peces/genética , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Procesamiento Postranscripcional del ARN , Infecciones por Rhabdoviridae/veterinaria , Transducción de Señal , Replicación Viral
17.
Fish Shellfish Immunol ; 94: 833-841, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31580936

RESUMEN

Glycoprotein (G) is the most common gene used in SVCV vaccine constructions. To identify the major immunogenicity determinant region of SVCV G gene, herein we truncated G gene to 4 parts (G-1, G-2, G-3 and G-4). Bioinformatics and the enzyme linked immunosorbent assay (ELISA) were used to identify the antigenicity of these 4 truncated G proteins. Immunological assays (serum antibody production, enzyme activity, immune genes expression and challenge test) were carried out to further identify the immunogenicity of the screened G protein in common carp. Moreover, to further verify the immune response of the screened G protein-based subunit vaccine, its protective effects on common carp against SVCV infection using single-walled carbon nanotubes (SWCNTs) as a carrier were evaluated. Results showed that G-3 protein could induce higher antibody titer than other truncated G proteins. Furthermore, carps vaccinated with G-3 and G (positive control) showed significant enhancement of immune response (serum antibody production, enzyme activity and immune related genes expression) when compared with control groups. Meanwhile, as a promising vaccine carrier, SWCNTs could significantly enhance the immune effect of naked subunit vaccine (G-3 and G). Notably, after SVCV challenge, there was no significant difference in immune protection between G-3 and G, nor between SWCNTs-G-3 and SWCNTs-G. These results so far suggest G-3 might be the potential antigen epitope of SVCV. This study lays a foundation for developing vaccine and immunodiagnostic techniques.


Asunto(s)
Carpas/inmunología , Inmunogenicidad Vacunal/inmunología , Nanotubos de Carbono/análisis , Rhabdoviridae/inmunología , Animales , Epítopos/inmunología
18.
Fish Shellfish Immunol ; 88: 403-406, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30862516

RESUMEN

IFN-γ is a pleiotropic cytokine with significant roles in antiviral, antitumor and immune regulation. It could be used as an immuno-enhancer to improve fish protectiveness against pathogens. In this study, the prokaryotic expression plasmid pTwin1-N-IFN-γ was constructed to express Cyprinus carpio (common carp) IFN-γ fused with a chitin binding domain (CBD) and a self-cleavable intein-tag, Synechocystis sp DnaB. The recombinant protein CBD-DnaB-IFN-γ with the molecular weight of 44.25 kD was successfully expressed in soluble form, and the rIFN-γ (approximate 18.61 kD) was further cleaved and eluted under pH = 7.0 at 25 °C. rIFN-γ could be recognized by western blotting with rabbit anti-grass carp IFN-γ polyclonal antibody. Cytotoxicity studies on EPC cells showed that only 500 ng/ml rIFN-γ had a subtle effect on cells growth and its proliferation rate was reduced to 76.2%. EPC cells incubated with 100 ng/ml rIFN-γ showed significantly higher resistance against SVCV, reducing the TCID50/ml by more than 800-fold. In vivo studies suggested that intraperitoneal injection of rIFN-γ significantly improved the survival rate of common carps compared with SVCV challenge alone. These results implied that rIFN-γ would act as an immuno-enhancer in carp aquaculture.


Asunto(s)
Carpas/virología , Enfermedades de los Peces/prevención & control , Inteínas/genética , Interferón gamma/inmunología , Infecciones por Rhabdoviridae/veterinaria , Animales , Acuicultura , Carpas/inmunología , Línea Celular Tumoral , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Inmunidad Innata , Interferón gamma/genética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/prevención & control , Viremia/inmunología
19.
Fish Shellfish Immunol ; 85: 66-77, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29567136

RESUMEN

We recently reported on a successful vaccine for carp against SVCV based on the intramuscular injection of a DNA plasmid encoding the SVCV glycoprotein (SVCV-G). This shows that the intramuscular (i.m.) route of vaccination is suitable to trigger protective responses against SVCV, and that the SVCV G-protein is a suitable vaccine antigen. Yet, despite the general success of DNA vaccines, especially against fish rhabdoviruses, their practical implementation still faces legislative as well as consumer's acceptance concerns. Furthermore, the i.m. route of plasmid administration is not easily combined with most of the current vaccination regimes largely based on intraperitoneal or immersion vaccination. For this reason, in the current study we evaluated possible alternatives to a DNA-based i.m. injectable vaccine using the SVCV-G protein as the vaccine antigen. To this end, we tested two parallel approaches: the first based on the optimization of an alginate encapsulation method for oral delivery of DNA and protein antigens; the second based on the baculovirus recombinant expression of transmembrane SVCV-G protein in insect cells, administered as whole-cell subunit vaccine through the oral and injection route. In addition, in the case of the oral DNA vaccine, we also investigated the potential benefits of the mucosal adjuvants Escherichia coli lymphotoxin subunit B (LTB). Despite the use of various vaccine types, doses, regimes, and administration routes, no protection was observed, contrary to the full protection obtained with our reference i.m. DNA vaccine. The limited protection observed under the various conditions used in this study, the nature of the host, of the pathogen, the type of vaccine and encapsulation method, will therefore be discussed in details to provide an outlook for future vaccination strategies against SVCV.


Asunto(s)
Carpas , Enfermedades de los Peces/prevención & control , Infecciones por Rhabdoviridae/veterinaria , Rhabdoviridae/inmunología , Vacunación/veterinaria , Vacunas Virales/farmacología , Animales , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/prevención & control , Infecciones por Rhabdoviridae/virología , Células Sf9 , Spodoptera , Vacunas de ADN/administración & dosificación , Vacunas de ADN/clasificación , Vacunas de ADN/farmacología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/clasificación , Vacunas de Subunidad/farmacología , Vacunas Virales/administración & dosificación , Vacunas Virales/clasificación
20.
PLoS Pathog ; 12(6): e1005699, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27351838

RESUMEN

TNFα is a pleiotropic pro-inflammatory cytokine with a key role in the activation of the immune system to fight viral infections. Despite its antiviral role, a few viruses might utilize the host produced TNFα to their benefit. Some recent reports have shown that anti-TNFα therapies could be utilized to treat certain viral infections. However, the underlying mechanisms by which TNFα can favor virus replication have not been identified. Here, a rhabdoviral infection model in zebrafish allowed us to identify the mechanism of action by which Tnfa has a deleterious role for the host to combat certain viral infections. Our results demonstrate that Tnfa signals through its receptor Tnfr2 to enhance viral replication. Mechanistically, Tnfa does not affect viral adhesion and delivery from endosomes to the cytosol. In addition, the host interferon response was also unaffected by Tnfa levels. However, Tnfa blocks the host autophagic response, which is required for viral clearance. This mechanism of action provides new therapeutic targets for the treatment of SVCV-infected fish, and advances our understanding of the previously enigmatic deleterious role of TNFα in certain viral infections.


Asunto(s)
Interacciones Huésped-Parásitos/inmunología , Infecciones por Rhabdoviridae/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Autofagia/fisiología , Western Blotting , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Organismos Modificados Genéticamente , Reacción en Cadena de la Polimerasa , Rhabdoviridae/inmunología , Replicación Viral/fisiología , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA