Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
1.
Fish Shellfish Immunol ; 149: 109553, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615704

ABSTRACT

Viral diseases have caused great economic losses to the aquaculture industry. However, there are currently no specific drugs to treat these diseases. Herein, we utilized Siniperca chuatsi as an experimental model, and successfully extracted two tissue factor pathway inhibitors (TFPIs) that were highly distributed in different tissues. We then designed four novel peptides based on the TFPIs, named TS20, TS25, TS16, and TS30. Among them, TS25 and TS30 showed good biosafety and high antiviral activity. Further studies showed that TS25 and TS30 exerted their antiviral functions by preventing viruses from invading Chinese perch brain (CPB) cells and disrupting Siniperca chuatsi rhabdovirus (SCRV)/Siniperca chuatsi ranairidovirus (SCRIV) viral structures. Additionally, compared with the control group, TS25 and TS30 could significantly reduce the mortality of Siniperca chuatsi, the relative protection rates of TS25 against SCRV and SCRIV were 71.25 % and 53.85 % respectively, and the relative protection rate of TS30 against SCRIV was 69.23 %, indicating that they also had significant antiviral activity in vivo. This study provided an approach for designing peptides with biosafety and antiviral activity based on host proteins, which had potential applications in the prevention and treatment of viral diseases.


Subject(s)
Fish Diseases , Rhabdoviridae Infections , Rhabdoviridae , Animals , Fish Diseases/virology , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/prevention & control , Rhabdoviridae/physiology , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Perches , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Peptides/pharmacology , Peptides/chemistry , RNA Virus Infections/veterinary , RNA Virus Infections/immunology , RNA Virus Infections/prevention & control
2.
Jpn J Infect Dis ; 77(3): 169-173, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38171846

ABSTRACT

Some lyssaviruses, including the rabies virus (RABV), cause lethal neurological symptoms in humans. However, the efficacy of commercial vaccines has only been evaluated against RABV. To assess cross-reactivity among lyssaviruses, including RABV, sera from rabbits inoculated with human and animal RABV vaccines and polyclonal antibodies from rabbits immunized with expression plasmids of the glycoproteins of all 18 lyssaviruses were prepared, and cross-reactivity was evaluated via virus-neutralization tests using Duvenhage lyssavirus (DUVV), European bat lyssavirus-1 (EBLV-1), Mokola lyssavirus (MOKV), Lagos bat lyssavirus (LBV), and RABV. The sera from rabbits inoculated with RABV vaccines showed cross-reactivity with EBLV-1 and DUVV, both belonging to phylogroup I. However, reactivity with MOKV and LBV in phylogroup II was notably limited or below the detection level. Next, we compared the cross-reactivity of the polyclonal antibodies against all lyssavirus glycoproteins. Polyclonal antibodies had high virus-neutralization titers against the same phylogroup but not different phylogroups. Our findings indicate that a new vaccine should be developed for pre- and post-exposure prophylaxis against lyssaviral infections.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Cross Reactions , Glycoproteins , Lyssavirus , Neutralization Tests , Animals , Lyssavirus/immunology , Rabbits , Antibodies, Viral/immunology , Antibodies, Viral/blood , Glycoproteins/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Humans , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/prevention & control
3.
Inn Med (Heidelb) ; 65(6): 608-611, 2024 Jun.
Article in German | MEDLINE | ID: mdl-38100072

ABSTRACT

Germany has been considered free of terrestrial rabies since 2008 as a result of intensive vaccination and surveillance efforts but reservoirs of the lyssaviruses EBLV­1 and EBLV­2 persist in bat colonies and thus pose a potential risk of infection. We report on a patient who suffered a bat bite in an urban setting in which European bat lyssavirus 1 (EBLV-1) was detected in the euthanized bat. We performed active and passive postexposure prophylaxis (PEP). This case study illustrates the ongoing risk of rabies infection due to close bat contacts in Germany and is intended to sensitize primary care physicians to take such exposure events seriously and to perform a regular PEP including administration of rabies immunoglobulin.


Subject(s)
Bites and Stings , Chiroptera , Lyssavirus , Post-Exposure Prophylaxis , Rabies , Chiroptera/virology , Humans , Animals , Lyssavirus/immunology , Bites and Stings/virology , Rabies/prevention & control , Rabies/transmission , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/transmission , Rhabdoviridae Infections/virology , Male , Germany , Rabies Vaccines/administration & dosage
4.
EMBO Mol Med ; 15(10): e16394, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37767784

ABSTRACT

Infections with rabies virus (RABV) and related lyssaviruses are uniformly fatal once virus accesses the central nervous system (CNS) and causes disease signs. Current immunotherapies are thus focused on the early, pre-symptomatic stage of disease, with the goal of peripheral neutralization of virus to prevent CNS infection. Here, we evaluated the therapeutic efficacy of F11, an anti-lyssavirus human monoclonal antibody (mAb), on established lyssavirus infections. We show that a single dose of F11 limits viral load in the brain and reverses disease signs following infection with a lethal dose of lyssavirus, even when administered after initiation of robust virus replication in the CNS. Importantly, we found that F11-dependent neutralization is not sufficient to protect animals from mortality, and a CD4 T cell-dependent adaptive immune response is required for successful control of infection. F11 significantly changes the spectrum of leukocyte populations in the brain, and the FcRγ-binding function of F11 contributes to therapeutic efficacy. Thus, mAb therapy can drive potent neutralization-independent T cell-mediated effects, even against an established CNS infection by a lethal neurotropic virus.


Subject(s)
Central Nervous System Infections , Chiroptera , Lyssavirus , Rabies virus , Rabies , Rhabdoviridae Infections , Animals , Humans , Rhabdoviridae Infections/drug therapy , Rhabdoviridae Infections/prevention & control , CD4-Positive T-Lymphocytes , Immunotherapy , Antibodies, Monoclonal/therapeutic use , Rabies/prevention & control
5.
Aust Vet J ; 101(11): 430-439, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37661860

ABSTRACT

Australian bat lyssavirus (ABLV) is a member of the Lyssavirus genus of the Rhabdoviridae family and is found in Australian bat species. It is of public health concern because of the rabies-like syndrome it causes in humans, resulting in government health and wildlife agencies using varied communication approaches to inform targeted audiences about zoonotic risks associated with handling bats. Despite these warnings, the number of reports of human-bat interactions remains high. This paper details a survey conducted to analyse the approaches utilised by a range of stakeholders to educate and communicate warnings to their target audiences. The survey focused on identifying the target audiences, communication methods used, along with the message frequency, content, and perceived effectiveness. Analysis of the top three messages delivered by stakeholders revealed that over half were information-focused messages and over a third, instruction-focused. Stakeholders identified the need to balance messaging about bat handling risks with information regarding the vulnerable status of bats and their environmental significance. Whilst the most common and (perceived) effective method of communication was one-on-one discussions, it was also identified to be ineffective for targeting mass audiences leading stakeholders to recognise the need to adapt to more efficient means of communication. The outcomes of this study may be useful to improve risk communication strategies regarding ABLV in Australia.


Subject(s)
Chiroptera , Lyssavirus , Rhabdoviridae Infections , Humans , Animals , Australia , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Public Health
6.
Vaccine ; 41(38): 5580-5586, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37517909

ABSTRACT

Vaccination procedures can be stressful for fish and can bring severe side effects. Therefore, vaccines that can minimize the number of administrations and maximize cross-protection against multiple serotypes, genotypes, or even different species would be highly advantageous. In the present study, we investigated the cross-protective ability of two types of vaccines - viral hemorrhagic septicemia virus (VHSV) G protein-expressing DNA vaccine and G gene-deleted single-cycle VHSV genotype IVa (rVHSV-ΔG) vaccine - against both VHSV genotype Ia and infectious hematopoietic necrosis virus (IHNV) in rainbow trout (Oncorhynchus mykiss). The results showed that rainbow trout immunized with VHSV genotype Ia G gene- or IVa G gene-expressing DNA vaccine were significantly protected against VHSV genotype Ia, but were not protected against IHNV. In contrast to the DNA vaccine, the single-cycle VHSV IVa vaccine induced significant protection against not only VHSV Ia but also IHNV. Considering no significant increase in ELISA titer and serum neutralization activity against IHNV in fish immunized with single-cycle VHSV IVa, the protection might be independent of humoral adaptive immunity. The scarcity of cytotoxic T cell epitopes between VHSV and IHNV suggested that the possibility of involvement of cytotoxic T cell-mediated cellular adaptive immunity would be low. The role of trained immunity (innate immune memory) in cross-protection should be further investigated.


Subject(s)
Fish Diseases , Hemorrhagic Septicemia, Viral , Infectious hematopoietic necrosis virus , Novirhabdovirus , Oncorhynchus mykiss , Rhabdoviridae Infections , Vaccines, DNA , Viral Vaccines , Animals , Infectious hematopoietic necrosis virus/genetics , Novirhabdovirus/genetics , Immunization , Hemorrhagic Septicemia, Viral/prevention & control , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary
7.
Fish Shellfish Immunol ; 137: 108782, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37141957

ABSTRACT

Herbal immunomodulators are an important part of prevention and control on viral diseases in aquaculture because of their propensity to improve immunity in fish. The present study was conducted to evaluate the immunomodulatory effect and antiviral activity of a synthesized derivative (serial number: LML1022) against spring viremia of carp virus (SVCV) infection in vitro and in vivo. The antiviral data suggested that LML1022 at 100 µM significantly inhibited the virus replication in epithelioma papulosum cyprini (EPC) cells, and may completely inhibit the infectivity of SVCV virion particles to fish cells by affecting the viral internalization. The results in the related stability of water environments also demonstrated that LML1022 had an inhibitory half-life of 2.3 d at 15 °C, which would facilitate rapid degradation of LML1022 in aquaculture application. For in vivo study, the survival rate of SVCV-infected common carp was increased 30% at least under continuous oral injection of LML1022 at 2.0 mg/kg for 7 d treatment. Additionally, pretreatment of LML1022 on fish prior to SVCV infection also obviously reduced the viral loads in vivo as well as an improved survival rate, showing that LML1022 was potential as an immunomodulator. As an immune response, LML1022 significantly upregulated the immune-related gene expression including IFN-γ2b, IFN-I, ISG15 and Mx1, indicating that its dietary administration may improve the resistance of common carp against SVCV infection.


Subject(s)
Carps , Fish Diseases , Rhabdoviridae Infections , Rhabdoviridae , Animals , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/drug therapy , Rhabdoviridae/physiology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Immunologic Factors/pharmacology , Adjuvants, Immunologic/pharmacology , Viremia/drug therapy
8.
J Fish Dis ; 46(2): 127-135, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36321410

ABSTRACT

Siniperca chuatsi rhabdovirus (SCRV) is an important pathogen that infects mandarin fish. A reverse genetics system is an important technical platform for virus research. In this study, the minigenome in which the enhanced green fluorescent protein gene is flanked by the viral genomic ends of SCRV and transcribed using a T7 promoter-terminator cassette was constructed. Co-transfection of the minigenome construct with SCRV-supporting plasmids of N, P, and L in BSRT7 cells resulted in the expression of the reporter gene. Transcription of a positive-strand RNA copy from cDNA of the SCRV genome along with the viral N, P, and L proteins resulted in the recovery of infectious SCRV in cells. Viral titre up to 108 PFU/ml was achieved. Recombinant SCRV was verified by the detection of a unique restriction site engineered into the SCRV genome. The phenotypes of the recombinant SCRV and the parental virus were evaluated by plaque size, replication kinetics in vitro, and pathogenicity in vivo. The recovered SCRV from cDNA showed similar phenotypes compared to the parental virus. The established reverse genetics system is of great significance and value for the functional genome study of SCRV and for laying a foundation for the development of the viral vector and SCRV vaccine.


Subject(s)
Fish Diseases , Rhabdoviridae Infections , Rhabdoviridae , Animals , DNA, Complementary/genetics , Rhabdoviridae/genetics , Fishes/genetics , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Genome, Viral
9.
Fish Shellfish Immunol ; 132: 108457, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36455780

ABSTRACT

Infectious hematopoietic necrosis virus (IHNV) and infectious pancreatic necrosis virus (IPNV) are typical pathogens of rainbow trout Oncorhynchus mykiss, and the concurrent infection of the two viruses is very common among modern trout hatcheries, which has caused huge economic losses to the rainbow trout farming industry. To prevent and control the spread of IHNV and IPNV in juvenile trout simultaneously, in this study a bivalent recombinant adenovirus vaccine with IHNV Glycoprotein (G) and IPNV VP2 genes was developed. After immunizing juvenile trout with this bivalent vaccine via the immersion route, the expression levels of IHNV G and IPNV VP2 and the representative immune genes in vaccinated and control rainbow trout were tested to evaluate the correlation of immune responses with the expression of viral genes. The neutralizing antibody level induced by this bivalent vaccine as well as the protection efficacy of the vaccine against IHNV and IPNV was also evaluated. The results showed that IHNV G and IPNV VP2 were successfully expressed in juvenile trout, and all the innate and adaptive immune genes were up-regulated. This indicated that the level of the innate and adaptive immune responses were significantly increased, which might be induced by the high expression of the two viral proteins. Compared with the controls, high levels of neutralizing antibodies against IHNV and IPNV were induced in the vaccinated trout. Besides, the bivalent recombinant adenovirus vaccine showed high protection rate against IHNV, with the relative percent survival (RPS) of 81.25%, as well as against IPNV, with the RPS of 78.95%. Taken together, our findings clearly demonstrated that replication-defective adenovirus can be developed as a qualified vector for fish vaccines and IHNV G and IPNV VP2 were two suitable antigenic genes that could induce effective immune protection against these two pathogens. This study provided new insights into developing bivalent vectored vaccines and controlling the spread of IHNV and IPNV simultaneously in juvenile trout.


Subject(s)
Adenovirus Vaccines , Birnaviridae Infections , Fish Diseases , Infectious hematopoietic necrosis virus , Infectious pancreatic necrosis virus , Oncorhynchus mykiss , Rhabdoviridae Infections , Viral Vaccines , Animals , Infectious pancreatic necrosis virus/physiology , Infectious hematopoietic necrosis virus/physiology , Vaccines, Synthetic , Adenoviridae/genetics , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Birnaviridae Infections/prevention & control , Birnaviridae Infections/veterinary
10.
Viruses ; 14(12)2022 12 02.
Article in English | MEDLINE | ID: mdl-36560709

ABSTRACT

Infectious hematopoietic necrosis virus (IHNV) is the most important pathogen threatening the aquaculture of salmonid fish in China. In addition to the common genogroup J IHNV, genogroup U has been newly discovered in China. However, there is no effective DNA vaccine to fight against this emerging genogroup U IHNV in China. In this study, DNA vaccines encoding the IHNV viral glycoprotein (G) gene of the GS2014 (genogroup J) and BjLL (genogroup U) strains isolated from northern China were successfully developed, which were identified by restriction analysis and IFA. The expression of the Mx-1 gene and G gene in the spleens and muscles of the injection site as well as the titers of the serum antibodies were measured to evaluate the vaccine efficacy by RT-qPCR and ELISA. We found that DNA vaccine immunization could activate Mx1 gene expression and upregulate G gene expression, and the mRNA levels of the Mx1 gene in the muscles were significantly higher than those in the spleens. Notably, DNA vaccine immunization might not promote the serum antibody in fish at the early stage of immunization. Furthermore, the efficacy of the constructed vaccines was tested in intra- and cross-genogroup challenges by a viral challenge in vivo. It seemed that the DNA vaccines were able to provide great immune protection against IHNV infection. In addition, the genogroup J IHNV-G DNA vaccine showed better immune efficacy than the genogroup U IHNV-G or divalent vaccine, which could provide cross-immune protection against the genogroup U IHNV challenge. Therefore, this is the first study to construct an IHNV DNA vaccine using the G gene from an emerging genogroup U IHNV strain in China. The results provide great insight into the advances of new prophylactic strategies to fight both the genogroup J and U IHNV in China.


Subject(s)
Fish Diseases , Infectious hematopoietic necrosis virus , Oncorhynchus mykiss , Rhabdoviridae Infections , Vaccines, DNA , Viral Vaccines , Animals , Vaccines, DNA/genetics , Infectious hematopoietic necrosis virus/genetics , Genotype , China/epidemiology , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Viral Vaccines/genetics
11.
Viruses ; 14(12)2022 12 09.
Article in English | MEDLINE | ID: mdl-36560754

ABSTRACT

Rabies is a neglected tropical disease. The prototype virus, the rabies virus, still causes tens of thousands of human fatalities annually. Rabies is one member of the genus Lyssavirus. The burden of other lyssaviruses is unclear. The continued emergence of novel lyssaviruses means that assessment of vaccine efficacy against these viruses is critical, as standard rabies vaccines are not efficacious against all lyssaviruses. Taiwan bat lyssavirus (TWBLV) was first reported in 2018 following isolation from Japanese house bats. Since the initial detection and genetic characterisation, no attempts have been made to antigenically define this virus. Due to the inaccessibility of the wildtype isolate, the successful generation of a live recombinant virus, cSN-TWBLV, is described, where the full-length genome clone of the RABV vaccine strain, SAD-B19, was constructed with the glycoprotein of TWBLV. In vitro and in vivo characterization of cSN-TWBLV was undertaken and demonstrated evidence for cross-neutralisation of cSN-TWBLV with phylogroup I -specific sera and rabies virus standard sera. For neutralisation equivalent to 0.5 IU/mL of WHO and World Organisation of Animal Health (WOAH) sera against CVS, 0.5 IU/mL of WOAH sera and 2.5 IU/mL of WHO sera were required to neutralise cSN-TWBLV. In addition, specific sera for ARAV and EBLV-1 exhibited the highest neutralising antibody titres against cSN-TWBLV, compared to other phylogroup I-specific sera.


Subject(s)
Chiroptera , Lyssavirus , Rabies Vaccines , Rabies virus , Rabies , Rhabdoviridae Infections , Animals , Humans , Rabies/prevention & control , Rabies/veterinary , Taiwan , Antibodies, Viral , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Rabies virus/genetics
12.
Microbiol Spectr ; 10(6): e0324522, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36409094

ABSTRACT

Previous inactivated vaccines against infectious hematopoietic necrosis (IHN) usually had a strong early immune protective effect but failed to provide long-term protection in rainbow trout (Oncorhynchus mykiss). To find a method for stabilizing the desired protective effect of IHN vaccines, we assessed the immune enhancement effect of four adjuvants on formaldehyde inactivated vaccine for IHN at 60 days postvaccination (dpv). The efficacy of a two-dose vaccination with the candidate adjuvant-formaldehyde inactivated vaccine for IHN was evaluated in terms of early protection and long-term protection (30 to 285 dpv). Neutralizing antibody titers were also measured at each time point. The Montanide GEL 02 PR (Gel 02) adjuvant significantly enhanced the immune protection provided by the IHN inactivated vaccine, whereas the immune-boosting effect of the other tested adjuvants lacked statistical significance. Both tested Gel 02-adjuvanted IHN inactivated vaccine dosages had a strong immune protection effect within 2 months postvaccination, with a relative percent of survival (RPS) of 89.01% to 100%, and the higher dosage provided complete protection at 204 dpv and a RPS of 60.79% on 285 dpv by reducing viral titers in rainbow trout. The neutralizing antibodies were observed only in vaccinated fish on 30 and 60 dpv. Through compatibility with an appropriate adjuvant, the highly immune protective effect of an IHN inactivated vaccine was prolonged from 60 dpv to at least 284 dpv; this novel adjuvant-IHN inactivated vaccine has promise as a commercial vaccine that provides the best available and longest duration of protection against IHN to rainbow trout. IMPORTANCE Infectious hematopoietic necrosis virus (IHNV) is one of the most serious pathogens threatening the global salmon and trout industry. However, there is currently only one commercialized infectious hematopoietic necrosis (IHN) vaccine, and it is inadequate for solving the global IHN problem. In this study, a promising adjuvanted inactivated vaccine with long-term protection was developed and comprehensively studied. We confirmed the presence of a late antiviral response stage in vaccinated rainbow trout that lacked detectable neutralizing antibodies, which are commonly recognized to be responsible for long-term specific protection in mammals. These findings further our understanding of unique features of fish immune systems and could lead to improved prevention and control of fish diseases.


Subject(s)
Fish Diseases , Infectious hematopoietic necrosis virus , Oncorhynchus mykiss , Rhabdoviridae Infections , Viral Vaccines , Animals , Vaccines, Inactivated , Antibodies, Neutralizing , Formaldehyde , Fish Diseases/prevention & control , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Necrosis/drug therapy , Mammals
13.
Fish Shellfish Immunol ; 131: 1051-1062, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36371050

ABSTRACT

Spring viremia of carp (SVC) remains as a vaccine orphan disease mostly affecting juvenile specimens. Young fish are especially difficult to vaccinate and oral administration of vaccine combined with food would be the election system to minimise stress and the vaccination costs associated to injection. However, administration of prophylactics with food pellets faces off several drawbacks mainly related with vaccine degradation and weak protection correlates of oral vaccines. Here we present a platform based on recombinant proteins (subunit vaccines) manufactured as highly resistant nanostructured materials, and providing excellent levels of protection against SVC virus in a preliminary i.p injection challenge. The G3 domain of SVCV glycoprotein G was overexpressed in E. coli together with IFNγ and the modular protein was purified from bacterial aggregates (inclusion bodies) as highly organised nanostructured biomaterial (nanopellets, NP). These SVCV-IFNNP were taken up by zebrafish cells leading to the enhanced expression of different antiviral and IFN markers (e.g vig1, mx, lmp2 or ifngr1 among others) in zebrafish liver cells (ZFL). To monitor if SVCVNP and SVCV-IFNNP can be taken up by intestinal epithelia and can induce antiviral response we performed experiments with SVCVNP and SVCV-IFNNP in 3 days post fertilization (dpf) zebrafish larvae. Both, SVCVNP and SVCV-IFNNP were taken up and accumulated in the intestine without signs of toxicity. The antiviral response in larvae showed a different induction pattern: SVCV-IFNNP did not induce an antiviral response while SVCVNP showed a good antiviral induction. Interestingly ZF4, an embryonic derived cell line, showed an antiviral response like ZFL cells, although the lmp2 and ifngr1 (markers of the IFNγ response) were not overexpressed. Experiments with adult zebrafish indicated an excellent level of protection against a SVCV model infection where SVCV-IFNNP vaccinated fish reached 20% cumulative mortality while control fish reached over 80% cumulative mortality.


Subject(s)
Carps , Fish Diseases , Nanoparticles , Rhabdoviridae Infections , Rhabdoviridae , Animals , Zebrafish , Viremia , Antiviral Agents/therapeutic use , Escherichia coli , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/drug therapy , Vaccines, Subunit
14.
Microbiol Spectr ; 10(5): e0308522, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36073822

ABSTRACT

Spring viremia of carp virus (SVCV) is highly contagious and lethal to most cyprinid fish, causing serious economic losses to the carp aquaculture industry. Although DNA vaccines can generate long-term humoral and cellular immune responses, which provide protective immunity against SVCV, the major drawback of DNA vaccines is their low immunogenicity in clinical tests. Here, we construct a dual-targeted polymer DNA vaccine delivery platform (MCS-PCHG) by using mannosylated chitosan to encapsulate the poly(d,l-lactide-co-glycolide)-loaded DNA vaccine containing the heavy-chain CH3 region (CH3) of common carp IgM and the antigenic domain (G131c). The developed nanovaccine delivery platform showed good biocompatibility in vivo and in vitro. With the modification of the mannose moiety and the modification of CH3, the constructed MCS-PCHG could efficiently activate the maturation of antigen-presenting cells. Moreover, we observe significantly high level of immune-related genes expression, serum antigen-specific IgM, SVCV-neutralizing antibody titers in fish vaccinated with MCS-PCHG. Next, the protective efficacy of MCS-PCHG was further evaluated by challenge test. The highest survival rate (ca. 84%) was observed in fish vaccinated with MCS-PCHG after challenging with SVCV. This study presents a novel design for smart, dual-targeted polymer nanoparticles, which are inherently biocompatible, promising for targeted vaccine delivery. IMPORTANCE Spring viremia of carp virus (SVCV) affects global cyprinid fish farming industry, with no available commercial vaccine. Herein, we developed a dual-targeting polymer nanovaccine (MCS-PCHG) by using mannose and common carp IgM heavy chain CH3 region (CH3) as antigen presenting cell (APCs) recognition moiety, attaining the effective delivery of antigen. This dual-targeting polymer vaccine can efficiently activate the APCs, and further induce robust and durable adaptive immune response with good protection against SVCV infection. Our study provides valuable theoretical basis for developing efficient vaccine against infectious diseases in aquaculture.


Subject(s)
Carps , Chitosan , Fish Diseases , Nanoparticles , Rhabdoviridae Infections , Vaccines, DNA , Animals , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Viremia/prevention & control , Viremia/veterinary , Mannose , Polymers , Polylactic Acid-Polyglycolic Acid Copolymer , Fish Diseases/prevention & control , Immunoglobulin M , Antibodies, Neutralizing
15.
J Fish Dis ; 45(6): 895-905, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35445749

ABSTRACT

Micropterus salmoides rhabdovirus (MSRV) is an significant pathogen that causes high mortality and related economic losses in bass aquaculture. There is no effective or approved therapy to date. In this study, we evaluated the anti-MSRV effects of 22 quinoline derivatives in grass carp ovary (GCO) cells. Among these compounds, 8-hydroxyquinoline exhibited valid inhibition in decreasing MSRV nucleoprotein gene expression levels of 99.3% with a half-maximal inhibitory concentrations (IC50 ) value of 4.66 µM at 48 h. Moreover, 8-hydroxyquinoline significantly enhanced a protective effect in GCO cells by reducing the cytopathic effect (CPE). By comparing the anti-MSRV activity of 22 quinoline derivatives, we found that 8-hydroxyquinoline possessed the efficient active site of 8-hydroxyl and inhibited MSRV infection in vitro. For in vivo studies, 8-hydroxyquinoline via intraperitoneal injection exhibited an antiviral effect in MSRV-infected largemouth bass by substantially enhancing the survival rate by 15.0%. Importantly, the viral loads in the infected largemouth bass notably reduced in the spleen on the third days post-infection. Overall, 8-hydroxyquinoline was considered to be an efficient agent against MSRV in aquaculture.


Subject(s)
Bass , Carps , Fish Diseases , Quinolines , Rhabdoviridae Infections , Rhabdoviridae , Animals , Catalytic Domain , Female , Oxyquinoline/pharmacology , Quinolines/pharmacology , Rhabdoviridae/genetics , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary
16.
J Fish Dis ; 44(12): 1925-1936, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34383969

ABSTRACT

Immersion vaccination of single-walled carbon nanotubes loaded with mannose-modified glycoprotein (SWCNTs-MG) vaccine has been proved to be effective in preventing spring viraemia of carp virus (SVCV). Immunization procedure has immense consequence on the immune effect of the immersion vaccine. However, immunization procedure optimization for SWCNTs-MG vaccine against SVCV has not been reported. In this study, accordingly, a full-factor experiment was designed to optimize the immunization procedure of SWCNTs-MG vaccine by three aspects of vaccine dose (30 mg/L, 40 mg/L and 50 mg/L), immunization density (8 fish L-1 , 24 fish L-1 and 48 fish L-1 ) and immunization time (6, 12 and 24 hr). Furthermore, we used the immunization group (A1B2C1, 30 mg/L, 24 fish L-1 and 6 hr) in the previous study as a positive control (PC) to evaluate the immunization effect optimized conditions from the expression of immune-related genes and relative percentage survival (RPS). At 28 days post-vaccination (DPV), common carps were intraperitoneal injected SVCV challenged test indicated that the A1B2C2 group (30 mg/L, 24 fish L-1 , 12 hr) displayed superiority of protective efficacy compare with other groups and the RPS with 77.9%, which was 15.6% higher than the PC group of RPS with 62.3%. Moreover, the expression of immune-related genes such as IL-10, CD4 and MHC-II was also significantly higher than PC group. The specific experimental flow chart is shown in Figure 1. Conclusively, these results demonstrated that vaccine dose, immunization density and immunization time are 30 mg/L, 24 fish L-1 and 12 hr, which is the more appropriate immunization programme with juvenile carp for SWCNTs-MG vaccine. This study provides a profitable reference for improving the immune efficiency of aquatic immersion vaccine. [Figure: see text].


Subject(s)
Fish Diseases/virology , Immunization/veterinary , Rhabdoviridae Infections/veterinary , Viral Vaccines/administration & dosage , Animals , Aquaculture , Carps , Fish Diseases/prevention & control , Immersion , Immunization/methods , Mannose , Nanotubes, Carbon , Rhabdoviridae , Rhabdoviridae Infections/prevention & control , Vaccines, Subunit/administration & dosage
17.
J Fish Dis ; 44(12): 2021-2029, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34431113

ABSTRACT

The interactive applications of immunization route, vaccine type and delivery vectors are emerging as a key area of research within the field of mass immunization in fishery production. In an effort to improve DNA vaccine's immune efficiency in large-scale immunization, a promising bacterial ghost-loaded DNA vaccine was constructed based on Escherichia coli DH5α. In common carp was investigated the immune response to immersion immunization via related indicator analysis, and the challenge test of spring viraemia of carp virus (SVCV) was carried out. The result indicated that BG-loaded DNA vaccine induced higher serum antibody level than naked pEG-G. Simultaneously, the immunophysiological indicators and genes change at the more advanced levels in the BG/pEG-G immune group. At the treatment concentration of 20 mg/L of the BG/pEG-G group, IgM and IgZ expressions in vivo were markedly increased by 21.62 times and 6.91 times, respectively, and the relative percentage survival reached the peak of 59.57%. This study paves the way for future aquatic animal vaccine research, which aimed to develop the highly effective immersion vaccine system by delivery vectors, with the ultimate aim to prevent and restrict SVCV in actual production.


Subject(s)
Fish Diseases/immunology , Rhabdoviridae Infections/veterinary , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Aquaculture , Carps , Escherichia coli , Fish Diseases/virology , Immersion , Immunization/veterinary , Rhabdoviridae , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/virology , Viral Vaccines/administration & dosage
18.
Commun Biol ; 4(1): 921, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326461

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) senses viral RNA and instigates an innate immune signaling cascade to induce type I interferon expression. Currently, the regulatory mechanisms controlling RIG-I activation remain to be fully elucidated. Here we show that the FAK family kinase-interacting protein of 200 kDa (FIP200) facilitates RIG-I activation. FIP200 deficiency impaired RIG-I signaling and increased host susceptibility to RNA virus infection. In vivo studies further demonstrated FIP200 knockout mice were more susceptible to RNA virus infection due to the reduced innate immune response. Mechanistic studies revealed that FIP200 competed with the helicase domain of RIG-I for interaction with the two tandem caspase activation and recruitment domains (2CARD), thereby facilitating the release of 2CARD from the suppression status. Furthermore, FIP200 formed a dimer and facilitated 2CARD oligomerization, thereby promoting RIG-I activation. Taken together, our study defines FIP200 as an innate immune signaling molecule that positively regulates RIG-I activation.


Subject(s)
Autophagy-Related Proteins/genetics , Common Cold/prevention & control , Coronavirus OC43, Human/physiology , DEAD Box Protein 58/genetics , Rhabdoviridae Infections/prevention & control , Vesicular stomatitis Indiana virus/physiology , A549 Cells , Animals , Autophagy-Related Proteins/metabolism , Chlorocebus aethiops , Common Cold/metabolism , Coronavirus Infections/metabolism , Coronavirus Infections/prevention & control , DEAD Box Protein 58/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , RAW 264.7 Cells , Rhabdoviridae Infections/metabolism , Vero Cells , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/prevention & control
19.
J Fish Dis ; 44(10): 1587-1594, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34165796

ABSTRACT

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.


Subject(s)
Carps , Fish Diseases/prevention & control , Immunization/veterinary , Rhabdoviridae Infections/veterinary , Rhabdoviridae/immunology , Vaccines, Subunit/immunology , Viral Vaccines/immunology , Animals , Epitopes/immunology , Fish Diseases/virology , Immunity , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/virology , Vaccines, Subunit/administration & dosage , Viral Vaccines/administration & dosage
20.
Front Immunol ; 12: 667478, 2021.
Article in English | MEDLINE | ID: mdl-34025669

ABSTRACT

Viral encephalitis is the most common cause of encephalitis. It is responsible for high morbidity rates, permanent neurological sequelae, and even high mortality rates. The host immune response plays a critical role in preventing or clearing invading pathogens, especially when effective antiviral treatment is lacking. However, due to blockade of the blood-brain barrier, it remains unclear how peripheral immune cells contribute to the fight against intracerebral viruses. Here, we report that peripheral injection of an antibody against human Tim-3, an immune checkpoint inhibitor widely expressed on immune cells, markedly attenuated vesicular stomatitis virus (VSV) encephalitis, marked by decreased mortality and improved neuroethology in mice. Peripheral injection of Tim-3 antibody enhanced the recruitment of immune cells to the brain, increased the expression of major histocompatibility complex-I (MHC-I) on macrophages, and as a result, promoted the activation of VSV-specific CD8+ T cells. Depletion of macrophages abolished the peripheral injection-mediated protection against VSV encephalitis. Notably, for the first time, we found a novel post-translational modification of MHC-I by Tim-3, wherein, by enhancing the expression of MARCH9, Tim-3 promoted the proteasome-dependent degradation of MHC-I via K48-linked ubiquitination in macrophages. These results provide insights into the immune response against intracranial infections; thus, manipulating the peripheral immune cells with Tim-3 antibody to fight viruses in the brain may have potential applications for combating viral encephalitis.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antigen-Presenting Cells/drug effects , Brain/drug effects , Encephalitis, Viral/prevention & control , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Macrophages/drug effects , Rhabdoviridae Infections/prevention & control , Vesiculovirus/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , Brain/immunology , Brain/metabolism , Brain/virology , Chlorocebus aethiops , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/metabolism , Encephalitis, Viral/virology , HEK293 Cells , Hepatitis A Virus Cellular Receptor 2/immunology , Histocompatibility Antigens Class I/metabolism , Host-Pathogen Interactions , Humans , Injections, Intraperitoneal , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RAW 264.7 Cells , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/metabolism , Rhabdoviridae Infections/virology , Ubiquitination , Vero Cells , Vesiculovirus/pathogenicity , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...