Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 280: 116541, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38848637

RESUMEN

Although accumulating evidence indicates that endangered animals suffer from plastic pollution, this has been largely overlooked. Here, we explored the bacteria and eukaryotes living in the plastics gathered from the natural habitat of the highly endangered crocodile lizard. The results demonstrated that the bacterial and eukaryotic communities on plastics formed a unique ecosystem that exhibited lower diversity than those in the surrounding water and soil. However, microbes displayed a more complex and stable network on plastic than that in water or soil, implying unique mechanisms of stabilization. These mechanisms enhanced their resilience and contributed to the provision of stable ecological services. Eukaryotes formed a simpler and smaller network than bacteria, indicating different survival strategies. The bacteria residing on the plastics played a significant role in carbon transformation and sequestration, which likely impacted carbon cycling in the habitat. Furthermore, microbial exchange between plastics and the crocodile lizard was observed, suggesting that plastisphere serves as a mobile gene bank for the exchange of information, including potentially harmful substances. Overall, microbes on plastic appear to significantly impact the crocodile lizard and its natural habitat via various pathways. These results provided novel insights into risks evaluation of plastic pollution and valuable guidance for government efforts in plastic pollutant control in nature reserves.

2.
Sci Total Environ ; 900: 165807, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37506917

RESUMEN

Plastisphere is a new niche for microorganisms that complicate the ecological effects of plastics, and may profoundly influence biodiversity and habitat conservation. The DaGuishan National Nature Reserve, one of the largest habitats of the highly endangered crocodile lizard (Shinisaurus crocodilurus), is experiencing plastic pollution without sufficient attention. Here, plastisphere collected from captive tanks of crocodile lizards in this nature reserve was characterized for the first time. Three types of plastic (PE-PP, PE1, and PE2) together with the surrounding water and soil samples, were collected, and 16S rRNA sequencing technology was used to characterize the bacterial composition. The results demonstrated that plastisphere was driven by stochastic process and had a distinct bacterial community with higher diversity than that in surrounding water (p < 0.05). Bacteria related to nitrogen and carbon cycles (Pseudomonas psychrotolerans, Methylobacterium-Methylorubrum) were more abundant in plastisphere than in water or soil (p < 0.05). More importantly, plastics recruited pathogens and those bacteria function in antibiotic resistant genes (ARGs) coding. Bacteria related to polymer degradation also proliferated in plastisphere, especially Bacillus subtilis with a fold change of 42.01. The PE2 plastisphere, which had the lowest diversity and was dominated by Methylobacterium-Methylorubrum differed from PE 1 and PE-PP plastispheres totally. Plastics' morphology and aquatic nutrient levels contributed to the heterogeneity of different plastispheres. Overall, this study demonstrated that plastispheres diversify in composition and function, affecting ecosystem services directly or indirectly. Pathogens and bacteria related to ARGs expression enriched in the plastisphere should not be ignored because they may threaten the health of crocodile lizards by increasing the risk of infection. Plastic pollution control should be included in conservation efforts for crocodile lizards. This study provides new insights into the potential impacts of plastisphere, which is important for ecological risk assessments of plastic pollution in the habitats of endangered species.


Asunto(s)
Ecosistema , Lagartos , Animales , ARN Ribosómico 16S , Bacterias , Plásticos , Agua , Lagartos/genética , Antibacterianos
3.
Mar Biotechnol (NY) ; 24(6): 1084-1093, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36227511

RESUMEN

The nervous necrosis virus (NNV) causes the viral nervous necrosis (VNN) disease in aquatic animals and has been a major threat in aquaculture. Thus, it is essential for the development of a prevention method to minimize economic losses caused by NNV such as the identification of NNV resistance genes and application of these genes in molecular breeding to increase disease resistance. gab3 is an important NNV resistance gene in Asian seabass. However, the mechanism of gab3 in NNV resistance has not been elucidated. In this study, knockdown of gab3 in NNV-infected Asian seabass cells resulted in a significant decrease in viral RNA and virus titers. Knockout of gab3 in zebrafish led to an increased survival rate and resistant time after NNV infection. Cellular localization of the GAB3 and NNV by immunofluorescence staining showed that the GAB3 was translocated from the nucleus to the cytoplasm, and finally reached the cell membrane of SB cells after 48 h post NNV infection. Our study suggests that gab3 plays an important role in NNV replication and silencing gab3 can inhibit virus replication.


Asunto(s)
Lubina , Enfermedades de los Peces , Nodaviridae , Perciformes , Infecciones por Virus ARN , Animales , Infecciones por Virus ARN/genética , Pez Cebra , Nodaviridae/fisiología , Replicación Viral , Necrosis , Lubina/genética
4.
Front Microbiol ; 13: 988730, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36118205

RESUMEN

Pangolins are endangered animals and are listed in the CITES Appendix I of the Convention International Trade Endangered Species of Wild Fauna and Flora as well as being the national first-level protected wild animal in China. Based on a few reports on pangolins infected with pestiviruses of the Flaviviridae family, Pestivirus infections in pangolins have attracted increasing attention. Pangolin pestivirus is a pathogen that may cause diseases such as acute diarrhea and acute hemorrhagic syndrome. To better understand the epidemiology and genomic characterization of pestiviruses carried by pangolins, we detected pestiviruses in dead Malayan pangolin using metavirome sequencing technology and obtained a Pestivirus sequence of 12,333 nucleotides (named Guangdong pangolin Pestivirus, GDPV). Phylogenetic tree analysis based on the entire coding sequence, NS3 gene or RdRp gene sequences, showed that GDPV was closely related to previously reported pangolin-derived Pestivirus and clustered into a separate branch. Molecular epidemiological investigation revealed that 15 Pestivirus-positive tissues from two pangolins individuals with a positivity rate of 5.56%, and six Amblyomma javanense carried pestiviruses with a positivity rate of 19.35%. Moreover, the RdRp gene of the Pestivirus carried by A. javanense showed a high similarity to that carried by pangolins (93-100%), indicating A. javanense is likely to represent the vector of Pestivirus transmission. This study expands the diversity of viruses carried by pangolins and provides an important reference value for interrupting the transmission route of the virus and protecting the health of pangolins.

5.
mBio ; 13(3): e0046322, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35467426

RESUMEN

Severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) and SARS-CoV-2, the causative agents of SARS, which broke out in 2003, and coronavirus disease 2019 (COVID-2019), which broke out in 2019, probably originated in Rhinolophus sinicus and R. affinis, respectively. Rhinolophus bats are important hosts for coronaviruses. Many SARS-related coronaviruses (SARSr-CoVs) have been detected in bats from different areas of China; however, the diversity of bat SARSr-CoVs is increasing, and their transmission mechanisms have attracted much attention. Here, we report the findings of SARSr-CoVs in R. sinicus and R. affinis from South China from 2008 to 2021. The full-length genome sequences of the two novel SARSr-CoVs obtained from Guangdong shared 83 to 88% and 71 to 72% nucleotide identities with human SARS-CoV and SARS-CoV-2, respectively, while sharing high similarity with human SARS-CoV in hypervariable open reading frame 8 (ORF8). Significant recombination occurred between the two novel SARSr-CoVs. Phylogenetic analysis showed that the two novel bat SARSr-CoVs from Guangdong were more distant than the bat SARSr-CoVs from Yunnan to human SARS-CoV. We found that transmission in bats contributes more to virus diversity than time. Although our results of the sequence analysis of the receptor-binding motif (RBM) and the expression pattern of angiotensin-converting enzyme 2 (ACE2) inferred that these viruses could not directly infect humans, risks still exist after some unpredictable mutations. Thus, this study increased our understanding of the genetic diversity and transmission of SARSr-CoVs carried by bats in the field. IMPORTANCE Severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2 probably originated from the SARS-related coronaviruses (SARSr-CoVs) carried by Rhinolophus bats from Yunnan, China. Systematic investigations of the reservoir hosts carrying SARSr-CoVs in Guangdong and the reservoir distribution and transmission are urgently needed to prevent future outbreaks. Here, we detected SARSr-CoV in Rhinolophus bat samples from Guangdong in 2009 and 2021 and found that the transmission of SARSr-CoV from different host populations contributes more to increased virus diversity than time. Bat SARSr-CoVs in Guangdong had genetic diversity, and Guangdong was also the hot spot for SARSr-CoVs. We once again prove that R. sinicus plays an important role in the maintenance of the SARS-CoVs. Besides, the SARSr-CoVs are mainly transmitted through the intestines in bats, and these SARSr-CoVs found in Guangdong could not use human ACE2 (hACE2), but whether they can pass through intermediate hosts or directly infect humans requires further research. Our findings demonstrate the ability of SARSr-CoVs to spread across species.


Asunto(s)
Quirópteros , Coronavirus , Enzima Convertidora de Angiotensina 2 , Animales , China/epidemiología , Quirópteros/virología , Coronavirus/clasificación , Evolución Molecular , Genoma Viral , Genómica , Humanos , Filogenia , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , SARS-CoV-2/genética
6.
J Virol ; 95(19): e0046121, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34287045

RESUMEN

The emergence of the CRISPR/Cas system as a technology has transformed our ability to modify nucleic acids, and the CRISPR/Cas13 system has been used to target RNA. CasRx is a small type VI-D effector (Cas13d) with RNA knockdown efficiency that may have an interference effect on RNA viruses. However, the RNA virus-targeting activity of CasRx still needs to be verified in vivo in vertebrates. In this study, we successfully engineered a highly effective CasRx system for fish virus interference. We designed synthetic mRNA coding for CasRx and used CRISPR RNAs to guide it to target the red-spotted grouper nervous necrosis virus (RGNNV). This technique resulted in significant interference with virus infections both in vitro and in vivo. These results indicate that CRISPR/CasRx can be used to engineer interference against RNA viruses in fish, which provides a potential novel mechanism for RNA-guided immunity against other RNA viruses in vertebrates. IMPORTANCE RNA viruses are important viral pathogens infecting vertebrates and mammals. RNA virus populations are highly dynamic due to short generation times, large population sizes, and high mutation frequencies. Therefore, it is difficult to find widely effective ways to inhibit RNA viruses, and we urgently need to develop effective antiviral methods. CasRx is a small type VI-D effector (Cas13d) with RNA knockdown efficiency that can have an interference effect on RNA viruses. Nervous necrosis virus (NNV), a nonenveloped positive-strand RNA virus, is one of the most serious viral pathogens, infecting more than 40 cultured fish species and resulting in huge economic losses worldwide. Here, we establish a novel effective CasRx system for RNA virus interference using NNV and grouper (Epinephelus coioides) as a model. Our data showed that CasRx was most robust for RNA virus interference applications in fish, and we demonstrate its suitability for studying key questions related to virus biology.


Asunto(s)
Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Enfermedades de los Peces/virología , Nodaviridae/genética , Perciformes/virología , Interferencia de ARN , Infecciones por Virus ARN/veterinaria , Animales , Nodaviridae/fisiología , Infecciones por Virus ARN/virología , ARN Viral/genética
7.
Front Immunol ; 11: 1764, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849631

RESUMEN

Palmitic acid is the most common saturated fatty acid in animals, plants, and microorganisms. Studies highlighted that palmitic acid plays a significant role in diverse cellular processes and viral infections. Accumulation of palmitic acid was observed in fish cells (grouper spleen, GS) infected with Singapore grouper iridovirus (SGIV). The fluctuated content levels after viral infection suggested that palmitic acid was functional in virus-cell interactions. In order to investigate the roles of palmitic acid in SGIV infection, the effects of palmitic acid on SGIV induced cytopathic effect, expression levels of viral genes, viral proteins, as well as virus production were evaluated. The infection and replication of SGIV were increased after exogenous addition of palmitic acid but suppressed after knockdown of fatty acid synthase (FASN), of which the primary function was to catalyze palmitate synthesis. Besides, the promotion of virus replication was associated with the down-regulating of interferon-related molecules, and the reduction of IFN1 and ISRE promotor activities by palmitic acid. We also discovered that palmitic acid restricted TBK1, but not MDA5-induced interferon immune responses. On the other hand, palmitic acid decreased autophagy flux in GS cells via suppressing autophagic degradation, and subsequently enhanced viral replication. Together, our findings indicate that palmitic acid is not only a negative regulator of TBK1-IRF3/7 pathway, but also a suppressor of autophagic flux. Finally, palmitic acid promotes the replication of SGIV in fish cells.


Asunto(s)
Autofagia/efectos de los fármacos , Lubina/virología , Infecciones por Virus ADN/veterinaria , Enfermedades de los Peces/virología , Proteínas de Peces/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Iridovirus/efectos de los fármacos , Ácido Palmítico/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Lubina/genética , Lubina/inmunología , Lubina/metabolismo , Línea Celular , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/metabolismo , Infecciones por Virus ADN/virología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/metabolismo , Proteínas de Peces/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Factor 3 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/genética , Iridovirus/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
8.
Front Microbiol ; 11: 1446, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32676067

RESUMEN

Autophagy is a conserved catabolic process that occurs at basal levels to maintain cellular homeostasis. Most virus infections can alter the autophagy level, which functions as either a pro-viral or antiviral pathway, depending on the virus and host cells. Singapore grouper iridovirus (SGIV) is a novel fish DNA virus that has caused great economic losses for the marine aquaculture industry. In this study, we found that SGIV inhibited autophagy in grouper spleen (GS) cells which was evidenced by the changes of LC3-II, Beclin1 and p-mTOR levels. Further study showed that SGIV developed at least two strategies to inhibit autophagy: (1) increasing the cytoplasmic p53 level; and (2) encoding viral proteins (VP48, VP122, VP132) that competitively bind autophagy related gene 5 and mediately affect LC3 conversion. Moreover, activation of autophagy by rapamycin or overexpressing LC3 decreased SGIV replication. These results provide an antiviral strategy from the perspective of autophagy.

9.
Front Microbiol ; 11: 886, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32508768

RESUMEN

Nervous necrosis virus (NNV) has infected more than 50 fish species worldwide, and has caused serious economic losses in the aquaculture industries. However, there is no effective antiviral therapy. The development of a rapid and accurate point-of-care diagnostic method for the prevention and control of NNV infection is urgently required. Commonly used methods for NNV detection include the cell culture-based assay, antibody-based assay and polymerase chain reaction (PCR)-based assay. However, these methods have disadvantages as they are time-consuming and complex. In the present study, we developed a simple and sensitive aptamer-based lateral flow biosensor (LFB) method for the rapid detection of red-spotted grouper nervous necrosis virus (RGNNV). An aptamer is a single-stranded nucleotide, which can specifically bind to the target and has many advantages. Based on a previously selected aptamer, which specifically bound to the coat protein of RGNNV (RGNNV-CP), two modified aptamers were used in this study. One aptamer was used for magnetic bead enrichment and the other was used for isothermal strand displacement amplification (SDA). After amplification, the product was further tested by the LFB, and the detection results were observed by the naked eye within 5 min with high specificity and sensitivity. The LFB method could detect RGNNV-CP protein as low as 5 ng/mL or 5 × 103 RGNNV-infected GB (grouper brain) cells. Overall, it is the first application of a LFB combined with aptamer in the rapid diagnosis of virus from aquatic animals, which provides a new option for virus detection in aquaculture.

10.
Fish Shellfish Immunol ; 99: 9-18, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32007559

RESUMEN

The Kruppel-like factor 6 (KLF6) is a member of Kruppel-like factor family, which belong to the Zinc finger family of transcription factors that mediates various cellular processes, such as proliferation, differentiation, development, and programmed cell death. Peroxisome proliferator-activated receptors (PPARs) are a family of transcription factors belonging to the nuclear receptor superfamily and they regulate numerous genes through ligand-dependent transcriptional activation and repression. In this study, we focus on the role of KLF6 gene in virus infection and the regulation of KLF6 on PPAR-δ in orange-spotted grouper (Epinephelus coioides). The ORF sequence of EcKLF6 was 846 bp, encoding a polypeptide of 282 amino acids with three conserved Zinc finger (type Cys2-His2) domain in the C-terminal region. Basing on the detection of the mRNA levels of viral genes, western blotting of MCP protein, and morphological CPEs, we found that the overexpression of EcKLF6 suppressed the replication of Singapore grouper iridovirus (SGIV), exerting its antiviral activity against fish virus. Moreover, promoter analysis was performed to investigate whether EcKLF6 was a regulator of EcPPAR-δ. The luciferase reporter assay and real time PCR results indicated a negative regulatory role of EcKLF6 on EcPPAR-δ transcription in grouper. Further experimental analysis shows that the potential EcKLF6 binding sites may locate in the EcPPAR-δ-4-M3 (+133 to +154) and EcPPAR-δ-4-M4 (+354 to +368) region of the EcPPAR-δ promoter. Electrophoretic mobile shift assays (EMSAs) verified that EcKLF6 interacted with the binding site of the EcPPAR-δ-4-M4 promoter region. In addition, we also found that KLF6 promotes inflammatory responses in GS cells. Considering that KLF6 and PPAR-δ play opposite roles in regulating inflammatory responses, we speculated the promoting effect of KLF6 on inflammatory response may be related to its negative regulation on EcPPAR-δ. In conclusion, the present study provides the first evidence of the negative regulation of EcPPAR-δ transcription by EcKLF6 and contributes to a better understanding of the transcriptional mechanisms of EcKLF6 in fish.


Asunto(s)
Lubina/genética , Lubina/virología , Infecciones por Virus ADN/veterinaria , Regulación de la Expresión Génica , Factor 6 Similar a Kruppel/genética , PPAR delta/genética , Animales , Lubina/inmunología , Clonación Molecular , Infecciones por Virus ADN/genética , Infecciones por Virus ADN/inmunología , Enfermedades de los Peces/genética , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Proteínas de Peces/genética , Iridovirus , Ranavirus
11.
Fish Shellfish Immunol ; 98: 908-916, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31770643

RESUMEN

Autophagy is an evolutionarily conserved cellular degradation process that is essential for homeostasis. As a cell steward, autophagy is thought to be a process that may have evolved to combat intracellular pathogens. However, some virus can subvert or utilize autophagy-related membrane structures to increase viral replication. The red-spotted grouper nervous necrosis virus (RGNNV) is a fish pathogen which leads to disastrous viral nervous necrosis in larvae and juvenile groupers and other marine fishes. To better comprehend the pathogenesis and replication mechanism of RGNNV, we investigated the relationship between RGNNV and autophagy. Here, we demonstrated that RGNNV induced autophagy in grouper spleen (GS) cells, as the significant increase in ultrastructural autophagosome-like vesicles, fluorescent punctate pattern of microtubule-associated protein 1 light chain 3 (LC3), and the conversion of LC3-I to LC3-II. Additionally, ultraviolet-inactivated RGNNV and the capsid protein also triggered autophagy. Enhancement of autophagy contributed to RGNNV replication, whereas blocked autophagy decreased RGNNV replication. Moreover, impeded fusion of autophagosomes and lysosomes also reduced RGNNV replication, indicating that RGNNV utilized the different steps of autophagy pathway to facilitate viral replication. The further study showed that RGNNV induced autophagy through activating the phosphorylation of eIF2α and inhibiting the phosphorylation of mTOR. These results will assist the search for novel drugs targets and vaccine design against RGNNV from the perspective of downregulating autophagy.


Asunto(s)
Autofagia , Lubina/inmunología , Enfermedades de los Peces/inmunología , Nodaviridae/fisiología , Infecciones por Virus ARN/veterinaria , Replicación Viral , Animales , Enfermedades de los Peces/virología , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/virología
12.
Sci Rep ; 9(1): 15647, 2019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-31666545

RESUMEN

Clathrins, composed of clathrin heavy chains (CHCs) and clathrin light chains (CLCs), are usually hijacked by viruses for infection. However, the role of CLCs, especially in regulating fish virus infection, remains poorly understood. Here, two isoforms of CLCs were cloned from the red-spotted grouper (Epinephelus akaara) (EaCLCa and EaCLCb). Both EaCLC transcripts were expressed in all examined tissues, and the expression of EaCLCa was much higher than that of EaCLCb. Over-expressing EaCLCa-W119R mutant significantly reduced Singapore grouper iridovirus (SGIV) infectivity. However, no effect of EaCLCb-W122R on SGIV infection was observed. The detailed steps were further studied, mainly including virus attachment, entry and the following transport to early endosomes. EaCLCa-W119R mutant notably inhibited internalization of SGIV particles with no effect on SGIV attachment. Furthermore, EaCLCa-W119R mutant obviously impaired the delivery of SGIV to early endosomes after virus internalization. In addition, the EaCLCa-W119R mutant markedly reduced the colocalization of SGIV and actin. However, EaCLCb is not required for such events during SGIV infection. Taken together, these results demonstrate for the first time that EaCLCa and EaCLCb exerted different impacts on iridovirus infection, providing a better understanding of the mechanisms of SGIV infection and opportunities for the design of new antiviral strategies.


Asunto(s)
Cadenas Ligeras de Clatrina/metabolismo , Iridovirus/enzimología , Iridovirus/fisiología , Perciformes/virología , Secuencia de Aminoácidos , Animales , Cadenas Ligeras de Clatrina/química , Cadenas Ligeras de Clatrina/genética , Endosomas/metabolismo , Regulación Enzimológica de la Expresión Génica , Espacio Intracelular/metabolismo , Iridovirus/genética , Mutación , Transporte de Proteínas , Análisis de Secuencia , Internalización del Virus
13.
Fish Shellfish Immunol ; 94: 38-49, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31470135

RESUMEN

Peroxisome proliferator-activated receptor δ (PPAR-δ), also called PPAR-ß or PPAR-ß/δ, is a member of the peroxisome proliferator-activated receptor (PPAR) family, which belongs to the nuclear steroid receptor superfamily. Activated PPARs participate in the regulation of lipid and glucose metabolism and also affect cellular proliferation, differentiation, and apoptosis, and the immune responses. To investigate the roles of PPAR-δ in Singapore grouper iridovirus (SGIV) infection, we cloned and characterized the gene encoding a PPAR-δ homologue from the orange-spotted grouper, Epinephelus coioides (EcPPAR-δ). EcPPAR-δ encodes a 514-amino-acid polypeptide, with 95.29% and 74.76% homologue to the Seriola dumerili and human proteins, respectively. EcPPAR-δ contains a typical DNA-binding domain and a ligand-binding domain. Its expression was induced by SGIV infection in vitro. A subcellular localization analysis showed that EcPPAR-δ localizes throughout the cytoplasm and nucleus, with a diffuse intracellular expression pattern. SGIV replication was reduced by EcPPAR-δ overexpression, which was evident in the reduced severity of the cytopathic effect, reduced viral gene transcription, and the reduced expression of the viral capsid protein. The replication of SGIV increased with the knockdown of EcPPAR-δ. The overexpression and silencing of EcPPAR-δ in grouper spleen cells showed that EcPPAR-δ plays a positive role in the regulation of the interferon signaling pathway, but has an anti-inflammatory effect on the inflammatory response. The anti-inflammatory effect of EcPPAR-δ may be related to its function in maintaining cell homeostasis. Because the interferon signaling pathway plays an important role in antiviral immune responses, we speculate that the activation of the interferon signaling pathway by EcPPAR-δ overexpression underlies its inhibitory effect on SGIV replication. Together, our data greatly extend our understanding of the roles of the EcPPAR-δ family members in the pathogenesis of fish viruses.


Asunto(s)
Lubina/genética , Lubina/inmunología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , PPAR delta/genética , PPAR delta/inmunología , Secuencia de Aminoácidos , Animales , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/veterinaria , Infecciones por Virus ADN/virología , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , PPAR delta/química , Ranavirus/fisiología , Alineación de Secuencia/veterinaria
14.
Fish Shellfish Immunol ; 93: 702-710, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31421242

RESUMEN

Autophagy is an evolutionarily conserved, multi-step lysosomal degradation process used to maintain cell survival and homeostasis. A series of autophagy-related genes (Atgs) are involved in the autophagic pathway. In mammals, a growing number of studies have attributed functions to some Atgs that are distinct from their classical role in autophagosome biogenesis, such as resistance to pathogens. However, little is known about the functions of fish Atgs. In this study, we cloned and characterized an atg12 homolog from orange spotted grouper (Epinephelus coioides) (Ecatg12). Ecatg12 encodes a 117 amino acid protein that shares 94.0% and 76.8% identity with gourami (Anabas_testudineus) and humans (Homo sapiens), respectively. The transcription level of Ecatg12 was lower in cells infected with Singapore grouper iridovirus (SGIV) than in non-infected cells. Fluorescence microscopy revealed that EcAtg12 localized in the cytoplasm and nucleus in grouper spleen cells. Overexpression of EcAtg12 significantly increased the replication of SGIV, as evidenced by increased severity of the cytopathic effect, transcription levels of viral genes, levels of viral proteins, and progeny virus yield. Further studies showed that EcAtg12 overexpression decreased the expression levels of interferon (IFN) related molecules and pro-inflammatory factors and inhibited the promoter activity of IFN-3, interferon-stimulated response element, and nuclear factor-κB. Together, these results demonstrate that EcAtg12 plays crucial roles in SGIV replication by downregulating antiviral immune responses.


Asunto(s)
Proteína 12 Relacionada con la Autofagia/genética , Proteína 12 Relacionada con la Autofagia/inmunología , Lubina/genética , Lubina/inmunología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , Proteína 12 Relacionada con la Autofagia/química , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/veterinaria , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Filogenia , Ranavirus/fisiología , Alineación de Secuencia/veterinaria
15.
Fish Shellfish Immunol ; 91: 40-49, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31082519

RESUMEN

DEAD (Asp-Glu-Ala-Asp)-box polypeptide 41 (DDX41) is a member of the DEXDc family of helicases, that has recently been identified to be a crucial intracellular DNA sensor that triggers multiple signaling molecules to activate the type I interferon response. However, the precise function of DDX41 in fish during a viral infection remains unknown. In the present study, the DDX41 homolog from orange spotted grouper, Epinephelus coioides (EcDDX41), was cloned and its potential role in the immune response to a fish viral infection were investigated. EcDDX41 encodes a putative protein of 614 amino acid residues that contained two conserved domains: 1) DEADc domain; and 2) HELICc domain. The sequence analysis indicated that EcDDX41 shared 99%, 94%, and 86% identity with Asian seabass (Lates calcarifer), zebrafish (Danio rerio), and humans (Homo sapiens), respectively. EcDDX41 mRNA was present in all of the detected tissues, with the highest level of expression in the gills. The level of EcDDX41 expression was up-regulated following infection with Singapore grouper iridovirus (SGIV) or red-spotted grouper nervous necrosis virus (RGNNV) in grouper spleen (GS) cell cultures, suggesting that EcDDX41 may be involved in fish virus infection. Furthermore, EcDDX41 overexpression in GS cells significantly inhibited SGIV and RGNNV replication. EcDDX41 overexpression significantly increased the expression of antiviral and inflammatory cytokine genes, including interferon regulatory factor genes (e.g., IRF1, IRF2, IRF3, and IRF7), interferon induced genes (e.g., ISG15, ISG56, IFP35, Viperin, and MXI), and pro-inflammatory cytokine genes (e.g., TNFα, IL-1ß, and IL-8). Moreover, EcDDX41 positively regulated the mitochondrial antiviral-signaling protein (MAVS) and TANK-binding kinase 1 (TBK1)-induced interferon immune response, but did mediate IRF3 activation (MITA) to evoke an interferon immune response in unstimulated cells. Together, our results provide novel insight into the role of fish DDX41 in the antiviral innate immune response.


Asunto(s)
Lubina/genética , Lubina/inmunología , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , ARN Helicasas DEAD-box/química , Infecciones por Virus ADN/inmunología , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Nodaviridae/fisiología , Filogenia , Infecciones por Virus ARN/inmunología , Ranavirus/fisiología , Alineación de Secuencia/veterinaria
16.
Dev Comp Immunol ; 96: 37-46, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30822452

RESUMEN

Cystatin C is an endogenous inhibitor of cysteine proteases and widely exist in organisms. Several studies in mammals have showed that Cystatin C plays critical role in the immune defense against microorganisms. It is also well known that some fish Cystatin C have important immune regulation functions in inflammatory responses. However, the function of fish Cystatin C in virus infection as well as its underlying molecular mechanisms remain to be elucidated. In the present study, a Cystatin C gene termed Ec-CysC was identified from orange-spotted grouper, Epinephelus coioides. The full-length of Ec-CysC cDNA was 817 bp with a 387 bp open reading frame (ORF) that encoded a 129-amino acid (aa) protein, including 18-aa signal peptide and 111-aa mature polypeptide. The deduced amino acid of Ec-CysC shared three conserved domains containing Glycine at the N-terminus region, QVVAG motif in the middle and PW motif near the C-terminus region. Transcription analysis of the Ec-CysC gene showed its expression in all twelve examined tissues including liver, spleen, kidney, brain, intestine, heart, skin, muscle, fin, stomach, gill and head kidney. Its expression following stimulation with Singapore grouper iridovirus (SGIV) was further tested in spleen, the relative expression of Ec-CysC was significantly up-regulated at 12 h post-infection. The subcellular localization experiment revealed that Ec-CysC was mainly distributed in the cytoplasm in Grouper Spleen (GS) cells. In vitro, Overexpression of Ec-CysC in GS cells significantly reduced the expression of viral genes, namely, ORF162, ORF049 and ORF072. Meanwhile, we found that overexpression of Ec-CysC resulted in upward trend of expression of inflammatory cytokines TNF-a, IL-1ß and IL8 during SGIV infection. Further, SGIV-inducible apoptosis and Caspase-3 activity were also weakened by overexpression Ec-CysC in fathead minnow (FHM) cells. These results indicated that Ec-CysC might have a deeper involvement in fish immune defense, and played important roles in inflammation and apoptosis induced by SGIV.


Asunto(s)
Lubina/inmunología , Cistatina C/inmunología , Enfermedades de los Peces/inmunología , Proteínas de Peces/inmunología , Iridovirus/inmunología , Animales , Apoptosis/inmunología , Secuencia de Bases , Lubina/genética , Lubina/metabolismo , Línea Celular , Clonación Molecular , Cistatina C/genética , Cistatina C/metabolismo , Enfermedades de los Peces/virología , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Bazo/inmunología , Bazo/metabolismo , Regulación hacia Arriba
17.
Fish Shellfish Immunol ; 89: 677-686, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30905839

RESUMEN

Krϋppel-like factor 9 (KLF9) is a member of the SP/KL family, which are transcription factors implicated in several biological processes, including cell proliferation, differentiation, development and apoptosis. Studies have focused on the function of KLF9 in mammalian disease and the immune system, such as its regulatory role in the growth of tumors and its impact on interferon-related genes and inflammatory cytokines. In fish, little is known about the role of KLF9, especially its regulatory function in the innate antiviral immune response. In this study, we characterized the grouper KLF9 gene (EcKLF9) and investigated its role in viral infection. Amino acid alignment analysis showed that EcKLF9 was approximately 228 amino acids long and contained a typical three-tandem Krϋppel-like zinc fingers. Phylogenetic tree analysis revealed that EcKLF9 clustered with three fish species: Amphiprion ocellaris, Acanthochromis pollyacanthus and Stegastes partitus. Comparison analyses showed that the three Kruppel-like zinc finger domains of KLF9 were highly conserved in different fish species. Tissue expression analysis showed that EcKLF9 was constitutively expressed in all 12 tissues tested, in the healthy grouper, the highest expression being detected in the gonads. The relative expression levels of EcKLF9 in the head kidney, spleen and brain was significantly increased during red-spotted grouper nervous necrosis virus (RGNNV) and Singapore grouper iridovirus (SGIV) infections. Using fluorescence microscopy, EcKLF9 was primarily localized to the nucleus and cytoplasm. The in vitro ectopic expression of EcKLF9 significantly increased the severity of vacuoles induced by RGNNV and the cytopathic effect progression evoked by SGIV infection. Real-time PCR results showed that the transcription levels of viral genes, such as the Singapore grouper iridovirus infection genes, MCP (major capsid protein), LITAF (lipopolysaccharide induced TNF-α factor), VP19 (envelop protein) ICP-18 (infected cell protein-18) and the red-spotted grouper nervous necrosis virus genes, CP (coat protein), RdRp (RNA-dependent RNA polymerase), were all significantly increased in EcKLF9 overexpressing cells, when compared to control cells. Furthermore, western blotting analyses showed that protein levels of the RGNNV gene, CP and the SGIV gene, MCP were also increased in EcKLF9 overexpressing cells, suggesting EcKLF9 may promote viral activity against iridovirus and nodavirus, in vitro. Moreover, the overexpression of EcKLF9 significantly inhibited the expression of several interferon related cytokines and several inflammatory cytokines. Accordingly, we speculate that EcKLF9 may exert stimulatory effects on RGNNV and SGIV replication, through the negative regulation of host immune and inflammation responses.


Asunto(s)
Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/inmunología , Perciformes/genética , Perciformes/inmunología , Secuencia de Aminoácidos , Animales , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/veterinaria , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Factores de Transcripción de Tipo Kruppel/química , Nodaviridae/fisiología , Filogenia , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/veterinaria , Ranavirus/fisiología , Alineación de Secuencia/veterinaria , Especificidad de la Especie
18.
Fish Shellfish Immunol ; 88: 217-224, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30807858

RESUMEN

Chemokine receptors are a superfamily of seven-transmembrane domain G-coupled receptors and have important roles in immune surveillance, inflammation, and development. In previous studies, a series of CXCRs in grouper (Epinephelus coioides) was identified; however, the function of CXCR in viral infection has not been studied. To better understand the effect of the CXCR family on the fish immune response, full-length CXCR1a was cloned, and its immune response to Singapore grouper iridovirus (SGIV) was investigated. Grouper CXCR1a shared a seven-transmembrane (7-TM) region and a G protein-coupled receptor (GPCR) family 1 that contained a triaa stretch (DRY motif). Phylogenetic analysis indicated that CXCR1a showed the nearest relationship to Takifugu rubripes, followed by other fish, bird and mammal species. Fluorescence microscopy revealed that CXCR1a was expressed predominantly in the cytoplasm. Overexpression of CXCR1a in grouper cells significantly inhibited the replication of SGIV, demonstrating that CXCR1a delayed the occurrence of cytopathic effects (CPE) induced by SGIV infection and inhibited viral gene transcription. Furthermore, our results also showed that CXCR1a overexpression significantly increased the expression of interferon-related cytokines and activated ISRE and IFN promoter activities. Taken together, the results demonstrated that CXCR1a might have an antiviral function against SGIV infection.


Asunto(s)
Lubina/genética , Lubina/inmunología , Infecciones por Virus ADN/veterinaria , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Animales , Citocinas/metabolismo , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/virología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Microscopía Fluorescente , Filogenia , Ranavirus/fisiología , Replicación Viral/inmunología
19.
Chemosphere ; 221: 235-245, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30640006

RESUMEN

Bisphenol A (BPA) is an abundant endocrine-disrupting compound that is found in the aquatic environment and has adverse effects on fish reproduction; however, the exact pathway of these impacts is unclear. In this study, the different effects of BPA on ovarian and testis development in goldfish (Carassius auratus) and the different mechanisms underlying these effects were investigated. The gonadosomatic index (GSI) and gonadal histology demonstrated that BPA diminished ovarian maturation in goldfish, which recovered after BPA treatment withdrawal. In males, BPA disrupted testis maturation, but this disruption could not be recovered after BPA treatment withdrawal. The hypothalamic-pituitary-gonad (HPG) axis-related genes sgnrh, fshß and lhß were significantly decreased in BPA-treated female fish, while no changes in sex steroid hormone levels and no TUNEL and PCNA staining were found in the ovary, suggesting that BPA may reduce ovarian maturation through the HPG axis. In male fish, TUNEL staining was found in 1 µg L-1 BPA-exposed germ cells and 50 and 500 µg L-1 BPA-exposed Leydig cells. Decreases in 11-KT levels were also found in 50 and 500 µg L-1 BPA-exposed fish, but BPA did not affect genes associated with the HPG axes. This result shows that BPA disrupts testis maturation through apoptosis of germ cells and Leydig cells, thus inducing decreases in 11-KT levels that disrupt spermatogenesis. Collectively, our findings provide insights into the molecular and cellular mechanisms underlying BPA disturbance of goldfish reproduction.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Carpa Dorada/crecimiento & desarrollo , Gónadas , Fenoles/toxicidad , Reproducción/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Femenino , Células Germinativas/citología , Carpa Dorada/metabolismo , Gónadas/efectos de los fármacos , Células Intersticiales del Testículo/citología , Masculino , Ovario/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad
20.
Dev Comp Immunol ; 90: 10-20, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30165083

RESUMEN

Chemokines comprise a group of small molecular weight (6-14 kDa) cytokines; chemokine receptors are a superfamily of seven transmembrane domain G-coupled receptors. Both chemokines and their receptors have important roles in immune surveillance, inflammation, and development. Recently, 9 CXC chemokine ligands (CXCLs) and 8 CXC chemokine receptors (CXCRs) were identified and cloned from orange-spotted grouper (Epinephelus coioides) and annotated by phylogenetic and syntenic analyses. We detected mRNA transcripts for CXCLs and CXCRs in healthy tissues of E. coioides. Our data show that CXCL genes are highly expressed in the spleen, kidney and liver and that CXCR genes are ubiquitously expressed, rather than being expressed only in immune organs. Analysis of gene expression after Singapore grouper iridovirus infection indicated that CXCL and CXCR genes are regulated in a gene-specific manner. CXCL8 and CXCL12a were significantly upregulated in the spleen, kidney and liver of resistant fish, indicating potential roles in immunity against the pathogen. Additionally, CXCR4a was upregulated in all three organs in resistant fish, suggesting that CXCL8 or CXCL12a may participate in the immune response via interaction with CXCR4a. In addition, the new orange-spotted grouper receptor CXCR1b was found to be upregulated in the spleen and kidney of resistant fish, indicating that this receptor plays an important role in immune responses to viral infection. These results are valuable for comparative immunological studies and provide insight into the roles of these genes in viral infection.


Asunto(s)
Quimiocina CXCL12/genética , Infecciones por Virus ADN/inmunología , Enfermedades de los Peces/inmunología , Proteínas de Peces/genética , Interleucina-8/genética , Iridovirus/fisiología , Perciformes/inmunología , Receptores CXCR4/genética , Receptores de Interleucina-8A/genética , Animales , Quimiocina CXCL12/metabolismo , Clonación Molecular , Proteínas de Peces/metabolismo , Regulación de la Expresión Génica , Inmunidad Innata/genética , Interleucina-8/metabolismo , Filogenia , Receptores CXCR4/metabolismo , Receptores de Interleucina-8A/metabolismo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...