Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 905
Filter
1.
Fish Shellfish Immunol ; 151: 109748, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964434

ABSTRACT

The high mortality rate of Singapore grouper iridovirus (SGIV) posing a serious threat to the grouper aquaculture industry and causing significant economic losses. Therefore, finding effective drugs against SGIV is of great significance. Eugenol (C10H12O2) is a phenolic aromatic compound, has been widely studied for its anti-inflammatory, antioxidant and antiviral capacity. In this study, we explored the effect of eugenol on SGIV infection and its possible mechanisms using grouper spleen cells (GS) as an in vitro model. We found that treatment of GS cells with 100 µM eugenol for 4 h exhibited the optimal inhibitory effect on SGIV. Eugenol was able to reduce the expression level of inflammatory factors by inhibiting the activation of MAPK pathway and also inhibited the activity of NF-κB and AP-1 promoter. On the other hand, eugenol attenuated cellular oxidative stress by reducing intracellular ROS and promoted the expression of interferon-related genes. Therefore, we conclude that eugenol inhibits SGIV infection by enhancing cellular immunity through its anti-inflammatory and antioxidant functions.


Subject(s)
Antiviral Agents , Bass , DNA Virus Infections , Eugenol , Fish Diseases , Ranavirus , Animals , Eugenol/pharmacology , Fish Diseases/immunology , Fish Diseases/virology , Antiviral Agents/pharmacology , Bass/immunology , DNA Virus Infections/veterinary , DNA Virus Infections/immunology , DNA Virus Infections/drug therapy , Ranavirus/physiology , Spleen/immunology , Spleen/drug effects , Spleen/cytology , Cells, Cultured
2.
Fish Shellfish Immunol ; 152: 109784, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067495

ABSTRACT

Exocyst, a protein complex, plays a crucial role in various cellular functions, including cell polarization, migration, invasion, cytokinesis, and autophagy. Sec3, known as Exoc1, is a key subunit of the Exocyst complex and can be involved in cell survival and apoptosis. In this study, two subtypes of Sec3 were isolated from Epinephelus coioides, an important marine fish in China. The role of E. coioides Sec3 was explored during Singapore grouper iridovirus (SGIV) infection, an important pathogen of marine fish which could induce 90 % mortality. E. coioides Sec3 sequences showed a high similarity with that from other species, indicating the presence of a conserved Sec3 superfamily domain. E. coioides Sec3 mRNA could be detected in all examined tissues, albeit at varying expression levels. SGIV infection could upregulate E. coioides Sec3 mRNA. Upregulated Sec3 significantly promoted SGIV-induced CPE, and the expressions of viral key genes. E. coioides Sec3 could inhibit the activation of NF-κB and AP-1, as well as SGIV-induced cell apoptosis. The results illustrated that E. coioides Sec3 promotes SGIV infection by regulating the innate immune response.


Subject(s)
Bass , DNA Virus Infections , Fish Diseases , Fish Proteins , Immunity, Innate , Phylogeny , Ranavirus , Animals , Fish Diseases/immunology , Fish Diseases/virology , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Immunity, Innate/genetics , Bass/immunology , Ranavirus/physiology , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Gene Expression Regulation/immunology , Sequence Alignment/veterinary , Amino Acid Sequence , Gene Expression Profiling/veterinary
3.
Vet Res ; 55(1): 88, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39010235

ABSTRACT

Each year, due to climate change, an increasing number of new pathogens are being discovered and studied, leading to an increase in the number of known diseases affecting various fish species in different regions of the world. Viruses from the family Iridoviridae, which consist of the genera Megalocytivirus, Lymphocystivirus, and Ranavirus, cause epizootic outbreaks in farmed and wild, marine, and freshwater fish species (including ornamental fish). Diseases caused by fish viruses of the family Iridoviridae have a significant economic impact, especially in the aquaculture sector. Consequently, vaccines have been developed in recent decades, and their administration methods have improved. To date, various types of vaccines are available to control and prevent Iridoviridae infections in fish populations. Notably, two vaccines, specifically targeting Red Sea bream iridoviral disease and iridoviruses (formalin-killed vaccine and AQUAVAC® IridoV, respectively), are commercially available. In addition to exploring these themes, this review examines the immune responses in fish following viral infections or vaccination procedures. In general, the evasion mechanisms observed in iridovirus infections are characterised by a systemic absence of inflammatory responses and a reduction in the expression of genes associated with the adaptive immune response. Finally, this review also explores prophylactic procedure trends in fish vaccination strategies, focusing on future advances in the field.


Subject(s)
DNA Virus Infections , Fish Diseases , Fishes , Iridoviridae , Vaccination , Viral Vaccines , Animals , Fish Diseases/virology , Fish Diseases/prevention & control , Fish Diseases/immunology , DNA Virus Infections/veterinary , DNA Virus Infections/immunology , DNA Virus Infections/virology , DNA Virus Infections/prevention & control , Iridoviridae/physiology , Viral Vaccines/immunology , Fishes/virology , Fishes/immunology , Vaccination/veterinary
4.
PeerJ ; 12: e17605, 2024.
Article in English | MEDLINE | ID: mdl-39011377

ABSTRACT

Viral outbreaks are a constant threat to aquaculture, limiting production for better global food security. A lack of diagnostic testing and monitoring in resource-limited areas hinders the capacity to respond rapidly to disease outbreaks and to prevent viral pathogens becoming endemic in fisheries productive waters. Recent developments in diagnostic testing for emerging viruses, however, offers a solution for rapid in situ monitoring of viral outbreaks. Genomic epidemiology has furthermore proven highly effective in detecting viral mutations involved in pathogenesis and assisting in resolving chains of transmission. Here, we demonstrate the application of an in-field epidemiological tool kit to track viral outbreaks in aquaculture on farms with reduced access to diagnostic labs, and with non-destructive sampling. Inspired by the "lab in a suitcase" approach used for genomic surveillance of human viral pathogens and wastewater monitoring of COVID19, we evaluated the feasibility of real-time genome sequencing surveillance of the fish pathogen, Infectious spleen and kidney necrosis virus (ISKNV) in Lake Volta. Viral fractions from water samples collected from cages holding Nile tilapia (Oreochromis niloticus) with suspected ongoing ISKNV infections were concentrated and used as a template for whole genome sequencing, using a previously developed tiled PCR method for ISKNV. Mutations in ISKNV in samples collected from the water surrounding the cages matched those collected from infected caged fish, illustrating that water samples can be used for detecting predominant ISKNV variants in an ongoing outbreak. This approach allows for the detection of ISKNV and tracking of the dynamics of variant frequencies, and may thus assist in guiding control measures for the rapid isolation and quarantine of infected farms and facilities.


Subject(s)
Aquaculture , Fish Diseases , Iridoviridae , Animals , Fish Diseases/virology , Fish Diseases/epidemiology , Fish Diseases/diagnosis , Iridoviridae/genetics , Iridoviridae/isolation & purification , Ghana/epidemiology , Lakes/virology , DNA Virus Infections/virology , DNA Virus Infections/epidemiology , DNA Virus Infections/veterinary , DNA Virus Infections/transmission , Genome, Viral/genetics , Tilapia/virology , Disease Outbreaks/veterinary , Disease Outbreaks/prevention & control , Whole Genome Sequencing/methods , Cichlids/virology
5.
Fish Shellfish Immunol ; 152: 109770, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39025166

ABSTRACT

Prohibitin 1 (PHB1) is ubiquitously expressed in multiple compartments within cells and is involved in the cell cycle, cell signaling, apoptosis, transcriptional regulation, and mitochondrial biogenesis at the cellular level and in the inflammation-associated and immunological functions of B and T lymphocytes. PHB1 is an important protein that performs antioxidant regulation and immune functions inside and outside cells but has not been sufficiently studied in teleost fish. Our study aimed to elucidate the functional properties and gain new insights into the biological processes and immune system of red seabream (Pagrus major), a commercially important fish cultured in South Korea and East Asia. PHB1 mRNA was most abundantly expressed in the head kidney of healthy red seabream, and significant changes in its expression were observed after artificial infection with bacteria and viruses. On analysis, reporter gene was also significantly upregulated by polyinosinic-polycytidylic acid, lipopolysaccharides, and hydrogen peroxide. Consequent to the functional characterization of PHB1 in cells via recombinant protein preparation, the activity of leukocytes was enhanced and the reactive oxygen species-induced stress in red blood cells was reduced. The results reveal the functional characteristics of PHB1 and provide new insights into the biological processes and immune system of P. major, with beneficial implications in the study of stress responses.


Subject(s)
Fish Diseases , Fish Proteins , Gene Expression Profiling , Gene Expression Regulation , Immunity, Innate , Prohibitins , Repressor Proteins , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/immunology , Fish Diseases/immunology , Immunity, Innate/genetics , Gene Expression Regulation/immunology , Gene Expression Profiling/veterinary , Poly I-C/pharmacology , Phylogeny , Sea Bream/immunology , Sea Bream/genetics , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Amino Acid Sequence , Sequence Alignment/veterinary , Lipopolysaccharides/pharmacology , Perciformes/immunology , Perciformes/genetics , Iridoviridae/physiology , Vibrio/physiology
6.
Fish Shellfish Immunol ; 152: 109774, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39019127

ABSTRACT

Singapore grouper iridovirus (SGIV) belongs to the family Iridoviridae and the genus Ranavirus, which is a large cytoplasmic DNA virus. Infection of grouper with SGIV can cause hemorrhage and swelling of the spleen of the fish. Previous work on genome annotation demonstrated that SGIV contained numerous uncharacterized or hypothetical open reading frames (ORFs), whose functions remained largely unknown. In the present study, the protein encoded by SGIV ORF128 (VP128) was identified. VP128 is predominantly localized within the endoplasmic reticulum (ER). Overexpression of VP128 significantly promoted SGIV replication. VP128 inhibited the interferon (IFN)-3 promoter activity and mRNA level of IFN-related genes induced by poly(I:C), Epinephelus coioides cyclic GMP/AMP synthase (EccGAS)/stimulator of IFN genes (EcSTING), and TANK-binding kinase 1 (EcTBK1). Moreover, VP128 interacted with EcSTING and EcTBK1. The interaction between VP128 and EcSTING was independent of any specific structural domain of EcSTING. Together, our results demonstrated that SGIV VP128 negatively regulated the IFN response by inhibiting EcSTING-EcTBK1 signaling for viral evasion.


Subject(s)
DNA Virus Infections , Fish Diseases , Fish Proteins , Immunity, Innate , Ranavirus , Signal Transduction , Animals , Fish Diseases/immunology , Fish Diseases/virology , Ranavirus/physiology , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Fish Proteins/genetics , Fish Proteins/immunology , Signal Transduction/immunology , Immunity, Innate/genetics , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , Immune Evasion , Bass/immunology , Gene Expression Regulation/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence
7.
Arch Virol ; 169(7): 148, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38888759

ABSTRACT

The inflammasome is a multimeric protein complex that plays a vital role in the defence against pathogens and is therefore considered an essential component of the innate immune system. In this study, the expression patterns of inflammasome genes (NLRC3, ASC, and CAS-1), antiviral genes (IFNγ and MX), and immune genes (IL-1ß and IL-18) were analysed in Oreochromis niloticus liver (ONIL) cells following stimulation with the bacterial ligands peptidoglycan (PGN) and lipopolysaccharide (LPS) and infection with TiLV. The cells were stimulated with PGN and LPS at concentrations of 10, 25, and 50 µg/ml. For viral infection, 106 TCID50 of TiLV per ml was used. After LPS stimulation, all seven genes were found to be expressed at specific time points at each of the three doses tested. However, at even higher doses of LPS, NLRC3 levels decreased. Following TiLV infection, all of the genes showed significant upregulation, especially at early time points. However, the gene expression pattern was found to be unique in PGN-treated cells. For instance, NLRC3 and ASC did not show any response to PGN stimulation, and the expression of IFNγ was downregulated at 25 and 50 µg of PGN per ml. CAS-1 and IL-18 expression was downregulated at 25 µg of PGN per ml. At a higher dose (50 µg/ml), IL-1ß showed downregulation. Overall, our results indicate that these genes are involved in the immune response to viral and bacterial infection and that the degree of response is ligand- and dose-dependent.


Subject(s)
Cichlids , Fish Diseases , Inflammasomes , Animals , Cichlids/immunology , Cichlids/genetics , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Fish Diseases/immunology , Fish Diseases/virology , Fish Diseases/microbiology , Fish Diseases/genetics , Cell Line , Peptidoglycan/pharmacology , Liver/virology , Liver/immunology , Lipopolysaccharides/pharmacology , Immunity, Innate , Fish Proteins/genetics , Interleukin-18/genetics , Interleukin-18/metabolism , Ligands , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , DNA Virus Infections/virology , DNA Virus Infections/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/immunology
8.
BMC Vet Res ; 20(1): 267, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902724

ABSTRACT

BACKGROUND: Scale drop disease virus (SDDV) threatens Asian seabass (Lates calcarifer) aquaculture production by causing scale drop disease (SDD) in Asian seabass. Research on the development of SDDV vaccines is missing an in-depth examination of long-term immunity and the immune reactions it provokes. This study investigated the long-term immune protection and responses elicited by an SDDV vaccine. The research evaluated the effectiveness of a formalin-inactivated SDDV vaccine (SDDV-FIV) using both prime and prime-booster vaccination strategies in Asian seabass. Three groups were used: control (unvaccinated), single-vaccination (prime only), and booster (prime and booster). SDDV-FIV was administered via intraperitoneal route, with a booster dose given 28 days post-initial vaccination. RESULTS: The immune responses in vaccinated fish (single and booster groups) showed that SDDV-FIV triggered both SDDV-specific IgM and total IgM production. SDDV-specific IgM levels were evident until 28 days post-vaccination (dpv) in the single vaccination group, while an elevated antibody response was maintained in the booster group until 70 dpv. The expression of immune-related genes (dcst, mhc2a1, cd4, ighm, cd8, il8, ifng, and mx) in the head kidney and peripheral blood lymphocytes (PBLs) of vaccinated and challenged fish were significantly upregulated within 1-3 dpv and post-SDDV challenge. Fish were challenged with SDDV at 42 dpv (challenge 1) and 70 dpv (challenge 2). In the first challenge, the group that received booster vaccinations demonstrated notably higher survival rates than the control group (60% versus 20%, P < 0.05). However, in the second challenge, while there was an observable trend towards improved survival rates for the booster group compared to controls (42% versus 25%), these differences did not reach statistical significance (P > 0.05). These findings suggest that the SDDV-FIV vaccine effectively stimulates both humoral and cellular immune responses against SDDV. Booster vaccination enhances this response and improves survival rates up to 42 dpv. CONCLUSIONS: This research provides valuable insights into the development of efficient SDDV vaccines and aids in advancing strategies for immune modulation to enhance disease management in the aquaculture of Asian seabass.


Subject(s)
Fish Diseases , Immunization, Secondary , Vaccines, Inactivated , Viral Vaccines , Animals , Fish Diseases/prevention & control , Fish Diseases/immunology , Fish Diseases/virology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Immunization, Secondary/veterinary , Iridoviridae/immunology , DNA Virus Infections/veterinary , DNA Virus Infections/prevention & control , DNA Virus Infections/immunology , Formaldehyde , Antibodies, Viral/blood , Vaccination/veterinary , Immunoglobulin M/blood , Perciformes/immunology , Bass/immunology
9.
Environ Pollut ; 356: 124340, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38851377

ABSTRACT

Small plastic debris (0.1 µm-5 mm) or microplastics (MPs) have become major pollutants of aquatic ecosystems worldwide and studies suggest that MPs exposure can pose serious threats to human and wildlife health. However, to date the potential biological impacts of MPs accumulating in low amount in tissues during early life remains unclear. Here, for a more realistic assessment, we have used environmentally representative, mildly weathered, polyethylene terephthalate microplastics (PET MPs), cryomilled (1-100 µm) and fluorescently labelled. We leveraged the amphibian Xenopus laevis tadpoles as an animal model to define the biodistribution of PET MPs and determine whether exposure to PET MPs induce perturbations of antiviral immunity. Exposure to PET MPs for 1-14 days resulted in detectable PET MPs biodistribution in intestine, gills, liver, and kidney as determined by fluorescence microscopy on whole mount tissues. PET MPs accumulation rate in tissues was further evaluated via a novel in situ enzymatic digestion and subsequent filtration using silicon nanomembranes, which shows that PET MPs rapidly accumulate in tadpole intestine, liver and kidneys and persist over a week. Longer exposure (1 month) of tadpoles to relatively low concentration of PET MPs (25 µg/ml) significantly increased susceptibility to viral infection and altered innate antiviral immunity without inducing overt inflammation. This study provides evidence that exposure to MPs negatively impact immune defenses of aquatic vertebrates.


Subject(s)
Larva , Microplastics , Polyethylene Terephthalates , Ranavirus , Water Pollutants, Chemical , Xenopus laevis , Animals , Microplastics/toxicity , Ranavirus/physiology , Water Pollutants, Chemical/toxicity , DNA Virus Infections/veterinary , DNA Virus Infections/immunology
10.
Arch Virol ; 169(7): 136, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847927

ABSTRACT

Here, we report the first detection of lymphocystis disease virus (LCDV) in Indian glass fish in the Andaman Islands, India. Microscopic examination revealed the presence of whitish clusters of nodules on the fish's skin, fins, and eyes. The histopathology of the nodules revealed typical hypertrophied fibroblasts. Molecular characterization of the major capsid protein (MCP) gene of the virus showed a significant resemblance to known LCDV sequences from Korea and Iran, with 98.92% and 97.85% sequence identity, respectively. Phylogenetic analysis confirmed that the MCP gene sequence of the virus belonged to genotype V. This study represents the first documented case of LCDV in finfish from the Andaman Islands, emphasizing the necessity for continued monitoring and research on the health of aquatic species in this fragile ecosystem.


Subject(s)
Capsid Proteins , DNA Virus Infections , Fish Diseases , Iridoviridae , Phylogeny , Animals , Fish Diseases/virology , India , Iridoviridae/genetics , Iridoviridae/isolation & purification , Iridoviridae/classification , DNA Virus Infections/virology , DNA Virus Infections/veterinary , Capsid Proteins/genetics , Fishes/virology , Genotype , Islands
11.
J Wildl Dis ; 60(3): 683-690, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38741369

ABSTRACT

Ranaviruses are pathogens of ectothermic vertebrates (fish, amphibians, and reptiles). Turtles are the most common group of reptiles reported with ranaviral infections. However, there have been no surveys for wild ranaviral infection in any turtles from the suborder Pleurodira, despite ranaviral distributions and experimentally susceptible pleurodiran turtle populations overlapping in several areas, including Australia. We assayed 397 pooled blood samples from six Australian freshwater turtle species collected from five different sites in northern Australia between 2014 and 2019. Historical serologic surveys in the area had found antiranaviral antibodies; however, we did not detect any ranaviral DNA in our samples. Discrepancies between historical serologic and our molecular results may be explained by low viral prevalence during the years that these samples were collected, survivorship bias, or possibly an age class bias in sampling.


Subject(s)
DNA Virus Infections , Ranavirus , Turtles , Animals , Turtles/virology , Turtles/blood , DNA Virus Infections/veterinary , DNA Virus Infections/epidemiology , DNA Virus Infections/virology , DNA Virus Infections/blood , Australia/epidemiology , DNA, Viral/blood
12.
Fish Shellfish Immunol ; 150: 109611, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734119

ABSTRACT

During virus-host co-evolution, viruses have developed multiple strategies to dampen IFN response and prevent its antiviral activity in host cells. To date, the interactions between host IFN response and the immune evasion strategies exploited by fish iridoviruses still remain largely uncertain. Here, a potential immune evasion protein candidate of Singapore grouper iridovirus (SGIV), VP82 (encoded by SGIV ORF82) was screened and its roles during viral replication were investigated in detail. Firstly, VP82 overexpression dramatically decreased IFN or ISRE promoter activity and the transcription levels of IFN stimulated genes (ISGs) stimulated by grouper cyclic GMP-AMP synthase (EccGAS)/stimulator of interferon genes (EcSTING), TANK-binding kinase 1 (EcTBK1), IFN regulatory factor 3 (EcIRF3)and EcIRF7. Secondly, Co-IP assays indicated that VP82 interacted with EcIRF3 and EcIRF7, but not EcSTING and EcTBK1, which was consistent with the co-localization between VP82 and EcIRF3 or EcIRF7. Furthermore, VP82 promoted the degradation of EcIRF3 and EcIRF7 in a dose-dependent manner via the autophagy pathway. Finally, VP82 overexpression accelerated SGIV replication, evidenced by the increased transcriptions of viral core genes and viral production. Moreover, the antiviral action of EcIRF3 or EcIRF7 was significantly depressed in VP82 overexpressed cells. Together, VP82 was speculated to exert crucial roles for SGIV replication by inhibiting the IFN response via the degradation of IRF3 and IRF7. Our findings provided new insights into understanding the immune evasion strategies utilized by fish iridovirus through IFN regulation.


Subject(s)
DNA Virus Infections , Fish Diseases , Fish Proteins , Interferon Regulatory Factor-3 , Interferon Regulatory Factor-7 , Ranavirus , Viral Proteins , Animals , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Fish Diseases/immunology , Fish Diseases/virology , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Ranavirus/physiology , Viral Proteins/genetics , Viral Proteins/metabolism , Immunity, Innate/genetics , Interferons/genetics , Interferons/immunology , Interferons/metabolism , Immune Evasion , Bass/immunology , Bass/genetics , Virus Replication , Zebrafish Proteins , Interferon Regulatory Factors
13.
Fish Shellfish Immunol ; 150: 109598, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38697375

ABSTRACT

In mammals, IL-22 is considered as a critical cytokine regulating of immunity and homeostasis at barrier surfaces. Although IL-22 have been functional characterization in different species of fish, the studies about distinct responses of IL-22 in different organs/tissues/cell types is rather limited. Here, we identified and cloned IL-22 gene (named as Ec-IL-22) from grouper (Epinephelus coioides). Ec-IL-22 gene was detected in all orangs/tissues examined, and was induced in intestine, gill, spleen, head kidney, and primary head kidney/intestine leukocytes following the stimulation of LPS and poly (I:C), as well as Vibrio harveyi and Singapore grouper iridovirus infection (SGIV). In addition, the stimulation of DSS could induce the expression of Ec-IL-22 in intestine and primary leukocytes from intestine. Importantly, the treatment of recombinant Ec-IL-22 induced the mRNA level of proinflammatory cytokines in primary intestine/head kidney leukocytes. The present results improve the understanding of expression patterns and functional characteristics of fish IL-22 in different organs/tissues/cell types.


Subject(s)
Bass , DNA Virus Infections , Fish Diseases , Fish Proteins , Gene Expression Regulation , Interleukin-22 , Interleukins , Vibrio Infections , Vibrio , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Fish Diseases/immunology , Interleukins/genetics , Interleukins/immunology , Bass/immunology , Bass/genetics , Vibrio/physiology , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Gene Expression Regulation/immunology , Gene Expression Regulation/drug effects , Vibrio Infections/immunology , Vibrio Infections/veterinary , Amino Acid Sequence , Gene Expression Profiling/veterinary , Phylogeny , Sequence Alignment/veterinary , Immunity, Innate/genetics , Poly I-C/pharmacology , Lipopolysaccharides/pharmacology , Ranavirus/physiology
14.
Fish Shellfish Immunol ; 149: 109614, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38710342

ABSTRACT

Chemokines are critical molecules involved in immune reaction and immune system homeostasis, and some chemokines play a role in antiviral immunity. It is not known if the C-C motif chemokine ligand 3 (CCL3), a member of the CC chemokine family, possesses antiviral properties in fish. In this study, a ccl3 was cloned from the mandarin fish (Siniperca chuatsi), and it has an open reading frame (ORF) of 276 base pairs, which are predicted to encode a 91-amino acid peptide. Mandarin fish CCL3 revealed conserved sequence features with four cysteine residues and closely relationships with the CCL3s from other vertebrates based on the sequence alignment and phylogenetic analysis. The transcripts of ccl3 were notably enriched in immune-related organs, such as spleen and gills in healthy mandarin fish, and the ccl3 was induced in the isolated mandarin fish brain (MFB) cells following infection with infectious spleen and kidney necrosis virus (ISKNV). Moreover, in MFB cells, overexpression of CCL3 induced immune factors, such as IL1ß, TNFα, MX, IRF1 and IFNh, and exhibited antiviral activity against ISKNV. This study sheds light on the immune role of CCL3 in immune response of mandarin fish, and its antiviral defense mechanism is of interest for further investigation.


Subject(s)
Amino Acid Sequence , DNA Virus Infections , Fish Diseases , Fish Proteins , Immunity, Innate , Iridoviridae , Perciformes , Phylogeny , Sequence Alignment , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Fish Diseases/immunology , Fish Diseases/virology , Perciformes/immunology , Perciformes/genetics , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Iridoviridae/physiology , Sequence Alignment/veterinary , Immunity, Innate/genetics , Gene Expression Regulation/immunology , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Cloning, Molecular , Gene Expression Profiling/veterinary , Base Sequence
15.
Fish Shellfish Immunol ; 150: 109643, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763177

ABSTRACT

The lymphocystis disease (LCD), caused by Lymphocystis disease virus (LCDV), is a benign and self-limiting disease described in a many freshwater and marine fish species. Hypertrophic fibroblasts and extensive aggregation of inflammatory cells are characteristics of LCD. In the present study, small animal imaging and ultrastructural investigations were carried out on the lymphocystis nodules of black rockfish (Sebastes schlegelii) naturally infected with lymphocystis iridovirus, to assess pathology, and the exudate with particular attention to the formation of extracellular traps (ETs) in vivo. Ex vivo were examined by nodules sections and primary cells stimulation. By histopathological analysis, the nodules contained infiltrated inflammatory cells and extensive basophilic fibrillar filaments at the periphery of the hypertrophied fibroblasts. ETs were assessed in nodules samples using indirect immunofluorescence to detect DNA and myeloperoxidase. Moreover, LCDV was able to infect peritoneal cells of black rockfish in vitro and induce the formation of ETs within 4 h. In summary, this study proved that ETs are involved in the response to LCDV infection and may be involved in formation of lymphoid nodules. Taken together, the findings provide a new perspective to determine the impact factors on the growth of nodules.


Subject(s)
DNA Virus Infections , Extracellular Traps , Fish Diseases , Iridoviridae , Perciformes , Animals , Fish Diseases/virology , Fish Diseases/immunology , DNA Virus Infections/veterinary , DNA Virus Infections/immunology , DNA Virus Infections/virology , Extracellular Traps/immunology , Iridoviridae/physiology , Perciformes/immunology , Skin/virology , Skin/pathology , Fishes/immunology , Fishes/virology
16.
Viruses ; 16(5)2024 04 25.
Article in English | MEDLINE | ID: mdl-38793557

ABSTRACT

Frog virus 3 (FV3) in the genus Ranavirus of the family Iridoviridae causes mass mortality in both anurans and urodeles worldwide; however, the phylogenetic origin of FV3-like ranaviruses is not well established. In Asia, three FV3-like ranaviruses have been reported in farmed populations of amphibians and reptiles. Here, we report the first case of endemic FV3-like ranavirus infections in the Korean clawed salamander Onychodactylus koreanus, caught in wild mountain streams in the Republic of Korea (ROK), through whole-genome sequencing and phylogenetic analysis. Two isolated FV3-like ranaviruses (Onychodactylus koreanus ranavirus, OKRV1 and 2) showed high similarity with the Rana grylio virus (RGV, 91.5%) and Rana nigromaculata ranavirus (RNRV, 92.2%) but relatively low similarity with the soft-shelled turtle iridovirus (STIV, 84.2%) in open reading frame (ORF) comparisons. OKRV1 and 2 formed a monophyletic clade with previously known Asian FV3-like ranaviruses, a sister group of the New World FV3-like ranavirus clade. Our results suggest that OKRV1 and 2 are FV3-like ranaviruses endemic to the ROK, and RGV and RNRV might also be endemic strains in China, unlike previous speculation. Our data have great implications for the study of the phylogeny and spreading routes of FV3-like ranaviruses and suggest the need for additional detection and analysis of FV3-like ranaviruses in wild populations in Asian countries.


Subject(s)
DNA Virus Infections , Genome, Viral , Phylogeny , Ranavirus , Urodela , Animals , Ranavirus/genetics , Ranavirus/isolation & purification , Ranavirus/classification , Urodela/virology , Republic of Korea/epidemiology , DNA Virus Infections/veterinary , DNA Virus Infections/virology , DNA Virus Infections/epidemiology , Open Reading Frames , Whole Genome Sequencing
17.
Viruses ; 16(5)2024 04 30.
Article in English | MEDLINE | ID: mdl-38793597

ABSTRACT

In September 2021, 14 smallmouth bass (SMB; Micropterus dolomieu) with skin lesions were collected from Green Bay waters of Lake Michigan and submitted for diagnostic evaluation. All the skin samples tested positive for largemouth bass virus (LMBV) by conventional PCR. The complete genome of the LMBV (99,328 bp) isolated from a homogenized skin sample was determined using an Illumina MiSeq sequencer. A maximum likelihood (ML) phylogenetic analysis based on the 21 core iridovirus genes supported the LMBV isolated from SMB (LMBV-WVL21117) as a member of the species Santee-Cooper ranavirus. Pairwise nucleotide comparison of the major capsid protein (MCP) gene showed that LMBV-WVL21117 is identical to other LMBV reported from the United States and nearly identical to doctor fish virus and guppy virus 6 (99.2%) from Southeast Asia, as well as LMBV isolates from China and Thailand (99.1%). In addition, ML phylogenetic analysis based on the MCP gene suggests three genotypes of LMBV separated by region: genotype one from the United States, genotype two from Southeast Asia, and genotype three from China and Thailand. Additional research is needed to understand the prevalence and genetic diversity of LMBV strains circulating in wild and managed fish populations from different regions.


Subject(s)
Bass , DNA Virus Infections , Fish Diseases , Genome, Viral , Phylogeny , Ranavirus , Animals , Ranavirus/genetics , Ranavirus/isolation & purification , Ranavirus/classification , Bass/virology , DNA Virus Infections/virology , DNA Virus Infections/veterinary , Fish Diseases/virology , Capsid Proteins/genetics , Genotype , Lakes/virology
18.
Zool Res ; 45(3): 520-534, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38682434

ABSTRACT

Iridovirus poses a substantial threat to global aquaculture due to its high mortality rate; however, the molecular mechanisms underpinning its pathogenesis are not well elucidated. Here, a multi-omics approach was applied to groupers infected with Singapore grouper iridovirus (SGIV), focusing on the roles of key metabolites. Results showed that SGIV induced obvious histopathological damage and changes in metabolic enzymes within the liver. Furthermore, SGIV significantly reduced the contents of lipid droplets, triglycerides, cholesterol, and lipoproteins. Metabolomic analysis indicated that the altered metabolites were enriched in 19 pathways, with a notable down-regulation of lipid metabolites such as glycerophosphates and alpha-linolenic acid (ALA), consistent with disturbed lipid homeostasis in the liver. Integration of transcriptomic and metabolomic data revealed that the top enriched pathways were related to cell growth and death and nucleotide, carbohydrate, amino acid, and lipid metabolism, supporting the conclusion that SGIV infection induced liver metabolic reprogramming. Further integrative transcriptomic and proteomic analysis indicated that SGIV infection activated crucial molecular events in a phagosome-immune depression-metabolism dysregulation-necrosis signaling cascade. Of note, integrative multi-omics analysis demonstrated the consumption of ALA and linoleic acid (LA) metabolites, and the accumulation of L-glutamic acid (GA), accompanied by alterations in immune, inflammation, and cell death-related genes. Further experimental data showed that ALA, but not GA, suppressed SGIV replication by activating antioxidant and anti-inflammatory responses in the host. Collectively, these findings provide a comprehensive resource for understanding host response dynamics during fish iridovirus infection and highlight the antiviral potential of ALA in the prevention and treatment of iridoviral diseases.


Subject(s)
Fish Diseases , Iridovirus , Liver , alpha-Linolenic Acid , Animals , alpha-Linolenic Acid/metabolism , Fish Diseases/virology , Fish Diseases/metabolism , Liver/metabolism , Liver/virology , Iridovirus/physiology , DNA Virus Infections/veterinary , DNA Virus Infections/virology , Metabolomics , Antiviral Agents/pharmacology , Transcriptome , Metabolic Reprogramming , Multiomics
19.
Dis Aquat Organ ; 158: 65-74, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38661138

ABSTRACT

Red sea bream iridovirus (RSIV) causes substantial economic damage to aquaculture. In the present study, RSIV in wild fish near aquaculture installations was surveyed to evaluate the risk of wild fish being an infection source for RSIV outbreaks in cultured fish. In total, 1102 wild fish, consisting of 44 species, were captured from 2 aquaculture areas in western Japan using fishing, gill nets, and fishing baskets between 2019 and 2022. Eleven fish from 7 species were confirmed to harbor the RSIV genome using a probe-based real-time PCR assay. The mean viral load of the RSIV-positive wild fish was 101.1 ± 0.4 copies mg-1 DNA, which was significantly lower than that of seemingly healthy red sea bream Pagrus major in a net pen during an RSIV outbreak (103.3 ± 1.5 copies mg-1 DNA) that occurred in 2021. Sequencing analysis of a partial region of the major capsid protein gene demonstrated that the RSIV genome detected in the wild fish was identical to that of the diseased fish in a fish farm located in the same area in which the wild fish were captured. Based on the diagnostic records of RSIV in the sampled area, the RSIV-infected wild fish appeared during or after the RSIV outbreak in cultured fish, suggesting that RSIV detected in wild fish was derived from the RSIV outbreak in cultured fish. Therefore, wild fish populations near aquaculture installations may not be a significant risk factor for RSIV outbreaks in cultured fish.


Subject(s)
Aquaculture , DNA Virus Infections , Disease Outbreaks , Fish Diseases , Iridovirus , Animals , Fish Diseases/virology , Fish Diseases/epidemiology , DNA Virus Infections/veterinary , DNA Virus Infections/epidemiology , DNA Virus Infections/virology , Disease Outbreaks/veterinary , Iridovirus/genetics , Sea Bream/virology , Fishes , Risk Assessment , Japan/epidemiology , Animals, Wild
20.
Fish Shellfish Immunol ; 149: 109528, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38570119

ABSTRACT

Stimulator of interferon genes (STING) has been demonstrated as a critical mediator in the innate immune response to cytosolic DNA and RNA derived from different pathogens. While the role of Micropterus salmoides STING (MsSTING) in largemouth bass virus is still unknown. In this study, RT-qPCR assay and Western-blot assay showed that the expression levels of MsSTING and its downstream genes were up-regulated after LMBV infection. Pull down experiment proved that a small peptide called Fusion peptide (FP) that previously reported to target to marine and human STING as a selective inhibitor also interacted with MsSTING in vitro. Comparing with the RNA-seq of Largemouth bass infected with LMBV singly, 326 genes were significantly up-regulated and 379 genes were significantly down-regulated in the FP plus LMBV group in which Largemouth bass was treatment with FP before LMBV-challenged. KEGG analysis indicated that the differentially expressed genes (DEGs) were mainly related to signaling transduction, infectious disease viral, immune system and endocrine system. Besides, the survival rate of LMBV-infected largemouth bass was highly decreased following FP treatment. Taken together, our study showed that MsSTING played an important role in immune response against LMBV infection.


Subject(s)
Bass , Fish Diseases , Fish Proteins , Immunity, Innate , Animals , Fish Diseases/immunology , Fish Diseases/virology , Bass/immunology , Bass/genetics , Fish Proteins/genetics , Fish Proteins/immunology , Immunity, Innate/genetics , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Gene Expression Regulation/immunology , Gene Expression Regulation/drug effects , Ranavirus/physiology , Membrane Proteins/genetics , Membrane Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL