Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Fish Dis ; 47(4): e13915, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38191774

RESUMO

Micropterus salmoides rhabdovirus (MSRV) is a formidable pathogen, presenting a grave menace to juvenile largemouth bass. This viral infection frequently leads to epidemic outbreaks, resulting in substantial economic losses within the aquaculture industry. Unfortunately, at present, there are no commercially available vaccines or pharmaceutical treatments to combat this threat. In order to address the urgent need for therapeutic strategy to resist MSRV infection, the antiviral activity of natural product honokiol against MSRV was explored in this study. Firstly, cellular morphology was directly observed in an inverted microscope when treated with honokiol after MSRV infection. The results clarified that honokiol significantly lessened cytopathic effect (CPE) induced by MSRV and protected the integrity of GCO cells. Furthermore, the viral nucleic acid expression (G gene) was detected by reverse transcription real-time quantitative PCR (RT-qPCR) and the results indicated that honokiol significantly decreased the viral loads of MSRV in a concentration-dependent manner, and honokiol showed a high antiviral activity with IC50 of 2.92 µM. Besides, honokiol significantly decreased the viral titre and suppressed apoptosis caused by MSRV. Mechanistically, honokiol primarily inhibited the initial replication of MSRV and discharge of progeny virus to exert anti-MSRV activity. More importantly, in vivo experiments suggested that honokiol (40 mg/kg) expressed a fine antiviral activity against MSRV when administrated with intraperitoneal injection, which led to a notable 40% improvement in the survival rate among infected largemouth bass. In addition, it also resulted in significant reduction in the viral nucleic acid expression within liver, spleen and kidney at 2, 4 and 6 days following infection. What is more, 100 mg/kg honokiol with oral administration also showed certain antiviral efficacy in MSRV-infected largemouth bass via improving the survival rate by 10.0%, and decreasing significantly the viral nucleic acid expression in liver, spleen and kidney of largemouth bass on day 2. In summary, natural product honokiol is a good candidate to resist MSRV infection and has promising application prospects in aquaculture.


Assuntos
Compostos Alílicos , Bass , Produtos Biológicos , Compostos de Bifenilo , Doenças dos Peixes , Ácidos Nucleicos , Fenóis , Infecções por Rhabdoviridae , Rhabdoviridae , Animais , Doenças dos Peixes/epidemiologia , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/veterinária , Antivirais/farmacologia , Antivirais/uso terapêutico
2.
Mar Biotechnol (NY) ; 25(6): 1076-1084, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37861943

RESUMO

Spring viremia of carp virus (SVCV) is a highly lethal virus in common carp (Cyprinus carpio) and other cyprinid fish species. The aim of the present study was to develop an in vivo therapeutic measure against SVCV using artificial microRNA (AmiRNA) targeting the SVCV P gene transcript. Three candidates of AmiRNAs (AmiR-P1, -P2, and -P3) were selected, and their ability to downregulate SVCV P gene transcript was analyzed by both synthesized AmiRNA mimics and AmiRNA-expressing vector system, in which AmiR-P3 showed the strongest inhibitory activity among the three candidates. To overcome in vivo limitation of miRNA mimics or plasmid-based miRNA expression systems, we rescued recombinant snakehead rhabdoviruses (SHRVs) expressing SVCV P gene-targeting AmiRNA (rSHRV-AmiR-P3) or control AmiRNA (rSHRV-AmiR-C) using reverse genetic technology. The successful expression of AmiR-P3 and AmiR-C in cells infected with the rescued viruses was verified by quantitative PCR. To evaluate the availability of rSHRV-AmiR-P3 for in vivo control of SVCV, zebrafish (Danio rerio) were (i) infected with either rSHRV-AmiR-C or rSHRV-AmiR-P3 followed by SVCV infection or (ii) infected with SVCV followed by either rSHRV-AmiR-C or rSHRV-AmiR-P3 infection. Fish infected with rSHRVs before and after SVCV infection showed significantly higher survival rates than fish infected with SVCV alone. There was no significant difference in survival rates between groups of fish infected with rSHRV-AmiR-C and rSHRV-AmiR-P3 before SVCV infection; however, fish infected with SVCV followed by infection with rSHRV-AmiR-P3 showed significantly higher survival rates than fish infected with rSHRV-AmiR-C. These results suggest that rSHRV-AmiR-P3 has therapeutic potential against SVCV in fish when administered after SVCV infection, and rSHRVs expressing artificial microRNAs targeting SVCV transcripts could be used as a tool to control SVCV infection in fish for a therapeutic purpose.


Assuntos
Carpas , MicroRNAs , Infecções por Rhabdoviridae , Rhabdoviridae , Animais , Infecções por Rhabdoviridae/veterinária , Infecções por Rhabdoviridae/tratamento farmacológico , Peixe-Zebra/genética , Viremia , MicroRNAs/genética , Rhabdoviridae/genética
3.
EMBO Mol Med ; 15(10): e16394, 2023 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-37767784

RESUMO

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.


Assuntos
Infecções do Sistema Nervoso Central , Quirópteros , Lyssavirus , Vírus da Raiva , Raiva , Infecções por Rhabdoviridae , Animais , Humanos , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/prevenção & controle , Linfócitos T CD4-Positivos , Imunoterapia , Anticorpos Monoclonais/uso terapêutico , Raiva/prevenção & controle
4.
Fish Shellfish Immunol ; 137: 108782, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37141957

RESUMO

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.


Assuntos
Carpas , Doenças dos Peixes , Infecções por Rhabdoviridae , Rhabdoviridae , Animais , Infecções por Rhabdoviridae/prevenção & controle , Infecções por Rhabdoviridae/veterinária , Infecções por Rhabdoviridae/tratamento farmacológico , Rhabdoviridae/fisiologia , Antivirais/farmacologia , Antivirais/uso terapêutico , Fatores Imunológicos/farmacologia , Adjuvantes Imunológicos/farmacologia , Viremia/tratamento farmacológico
5.
Fish Shellfish Immunol ; 131: 1051-1062, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36371050

RESUMO

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.


Assuntos
Carpas , Doenças dos Peixes , Nanopartículas , Infecções por Rhabdoviridae , Rhabdoviridae , Animais , Peixe-Zebra , Viremia , Antivirais/uso terapêutico , Escherichia coli , Infecções por Rhabdoviridae/prevenção & controle , Infecções por Rhabdoviridae/veterinária , Infecções por Rhabdoviridae/tratamento farmacológico , Vacinas de Subunidades
6.
Zool Res ; 43(6): 966-976, 2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36257828

RESUMO

Spring viremia of carp virus (SVCV) is globally widespread and poses a serious threat to aquatic ecology and aquaculture due to its broad host range. To develop effective agents to control SVCV infection, we selected 16 naturally active small molecules to assess their anti-SVCV activity. Notably, dihydroartemisinin (DHA) (100 µmol/L) and (S, S)-(+)-tetrandrine (TET) (16 µmol/L) exhibited high antiviral effects in epithelioma papulosum cyprinid (EPC) cells, with inhibitory rates of 70.11% and 73.54%, respectively. The possible antiviral mechanisms were determined as follows: 1. Pre-incubation with DHA and TET decreased viral particle infectivity in fish cells, suggesting that horizontal transmission of SVCV in the aquatic environment was disrupted; 2. Although neither had an effect on viral adhesion, TET (but not DHA) interfered with SVCV entry into host cells (>80%), suggesting that TET may have an antiviral function in early viral replication. For in vivo study, both agents enhanced the survival rate of SVCV-infected zebrafish by 53.3%, significantly decreased viral load, and modulated the expression of antiviral-related genes, indicating that DHA and TET may stimulate the host innate immune response to prevent viral infection. Overall, our findings indicated that DHA and TET had positive effects on suppressing SVCV infection by affecting early-stage viral replication, thus holding great potential as immunostimulants to reduce the risk of aquatic rhabdovirus disease outbreaks.


Assuntos
Carpas , Doenças dos Peixes , Infecções por Rhabdoviridae , Rhabdoviridae , Animais , Infecções por Rhabdoviridae/veterinária , Infecções por Rhabdoviridae/tratamento farmacológico , Antivirais/farmacologia , Peixe-Zebra , Replicação Viral , Viremia/veterinária , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/uso terapêutico
7.
Virus Res ; 316: 198798, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35562080

RESUMO

Spring viraemia of carp virus (SVCV) poses a serious threat to aquaculture industry due to the lack of approved antiviral treatments. Therefore, a novel arctigenin derivative, 4-(2-methylimidazole) octanoxy-arctigenin (MON), was synthesized to assess the antiviral activity against SVCV in vitro and in vivo. The results indicated MON decreased the SVCV glycoprotein (G) gene expression in vitro by a maximum inhibitory rate of > 99% at 3.5 µM. Furthermore, MON showed the protective effect on epithelioma papulosum cyprinid (EPC) cells and considerably decreased the cytopathic effect (CPE). More importantly, MON inhibited SVCV G gene expression levels in vitro at the half-maximal activity (IC50) of 0.18 µM at 48 h. For in vivo studies, MON demonstrated anti-SVCV activity by enhancing the survival rate of zebrafish (Danio rerio) after infection via pelvic fin base injection. These results tended to be consistent with MON decreasing the SVCV titer of infected zebrafish. During this time, viral loads of the spleen and kidney have declined in SVSV-infected zebrafish. Based on the histopathological assay, MON exhibited the high protective effect in the spleen and kidney of SVCV-infected fish. Combined, MON is on track to become a novel agent to address SVCV infection in aquaculture.


Assuntos
Carpas , Doenças dos Peixes , Infecções por Rhabdoviridae , Rhabdoviridae , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Doenças dos Peixes/tratamento farmacológico , Furanos , Lignanas , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/veterinária , Peixe-Zebra
8.
Virol Sin ; 37(2): 277-283, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35249853

RESUMO

Since the anti-inflammatory effect of hydrogen has been widely known, it was supposed that hydrogen could suppress tissue damage by inhibiting virus-related inflammatory reactions. However, hydrogen is slightly soluble in water, which leads to poor effect of oral hydrogen-rich water therapy. In this study, the nano-bubble hydrogen water (nano-HW) (about 0.7 â€‹ppm) was prepared and its therapeutic effect against viral infection was investigated by utilizing spring viraemia of carp virus (SVCV)-infected zebrafish as model. Three-month-old zebrafish were divided into nano-HW treatment-treated group and aquaculture water treated group (control group). The results revealed that the cumulative mortality rate of SVCV-infected zebrafish was reduced by 40% after treatment with nano-bubble hydrogen water, and qRT-PCR results showed that SVCV replication was significantly inhibited. Histopathological examination staining showed that SVCV infection caused tissue damage was greatly alleviated after treatment with nano-bubble hydrogen water. Futhermore, SVCV infection caused reactive oxygen species (ROS) accumulation was significantly reduced upon nano-HW treatment. The level of proinflammatory cytokines IL-1ß, IL-8, and TNF-α was remarkably reduced in the nano-HW-treated group in vivo and in vitro. Taken together, our data demonstrated for the first time that nano-HW could inhibit the inflammatory response caused by viral infection in zebrafish, which suggests that nano-HW can be applied to antiviral research,and provides a novel therapeutic strategy for virus-caused inflammation related disease.


Assuntos
Carpas , Doenças dos Peixes , Infecções por Rhabdoviridae , Animais , Doenças dos Peixes/tratamento farmacológico , Hidrogênio/farmacologia , Hidrogênio/uso terapêutico , Inflamação/tratamento farmacológico , Rhabdoviridae , Infecções por Rhabdoviridae/tratamento farmacológico , Água , Peixe-Zebra
9.
Antiviral Res ; 195: 105192, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34687821

RESUMO

Due to the lack of relevant therapies for infectious haematopoietic necrosis virus (IHNV) infection, the viral outbreak invariably causes serious economic losses in salmonid species. In this study, we evaluated the anti-IHNV effects of 7-(6-benzimidazole) coumarin (C10) and 4-phenyl-2-thioxo-1,2,3,4-tetrahydro-5H-chromeno[4,3-d]pyrimidin-5-one (S5) in vitro and in vivo. The results revealed that C10 at 12.5 mg/L and S5 at 25 mg/L significantly inhibited IHNV replication in epithelioma papulosum cyprini (EPC) cells with a maximum inhibitory rate >90%, showing that IHNV-induced cytopathic effect (CPE) was alleviated by C10 and S5. There are two complementary effects on antiviral mechanism: 1. C10 completely inhibited IHNV infectivity when the virus was preincubated with C10 at 12.5 mg/L, determining that C10 may have a negative impact on IHNV binding to the cell; 2. C10 also up-regulated the gene expression of extracellular proto type galectin-1 (Gal1-L2) and a chimera galectin-3 (Gal3-L1) of EPC cells to inhibit IHNV adhesion. For the in vivo study, injection and immersion of the coumarins enhanced the survival rate of rainbow trout (Oncorhynchus mykiss) juveniles by 25% (at least) at 12 dpi. IHNV loads in the kidney and spleen were also obviously decreased at 96 h, and thus we considered that they had a delaying effect on IHNV replication in vivo. Meanwhile, C10 with a high stability in aquacultural water in immersion suppressed IHNV horizontal transmission by decreasing the viral loads in recipient fish. Overall, our data suggest that there is a positive effect of C10 and S5 against IHNV infection in aquaculture, and C10 had the potential to be a broad-spectrum antiviral against fish rhabdoviruses.


Assuntos
Antivirais/farmacologia , Cumarínicos/farmacologia , Vírus da Necrose Hematopoética Infecciosa/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Aquicultura , Linhagem Celular , Doenças dos Peixes/tratamento farmacológico , Doenças dos Peixes/mortalidade , Doenças dos Peixes/patologia , Oncorhynchus mykiss/virologia , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/mortalidade , Infecções por Rhabdoviridae/patologia , Taxa de Sobrevida , Carga Viral/efeitos dos fármacos , Proteínas Virais/genética , Proteínas Virais/metabolismo
10.
Eur J Med Chem ; 223: 113739, 2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34375787

RESUMO

Diseases caused by rhabdoviruses have had a huge impact on the productive lives of the entire human population. The main problem is the lack of drugs for the treatment of this family of viruses. Infectious hematopoietic necrosis virus (IHNV), the causative agent of IHN, is a typical rhabdovirus which has caused huge losses to the salmonid industry. Therefore, in this study, IHNV was studied as a model to evaluate the antiviral activity of 35 novel coumarin derivatives. Coumarin A9 was specifically selected for further validation studies upon comparing the half maximum inhibitory concentration (IC50) of four screened candidate derivatives in epithelioma papulosum cyprinid (EPC) cells, as it exhibited an IC50 value of 2.96 µM against IHNV. The data revealed that A9 treatment significantly suppressed the virus-induced cytopathic effect (CPE) in EPC cells. In addition, A9 showed IC50 values of 1.68 and 2.12 µM for two other rhabdoviruses, spring viremia of carp virus and micropterus salmoides rhabdovirus, respectively. Furthermore, our results suggest that A9 exerts antiviral activity, but not by destroying the virus particles and interfering with the adsorption of IHNV. Moreover, we found that A9 had an inhibitory effect on IHNV-induced apoptosis in EPC cells, as reflected by the protection against cell swelling, formation of apoptotic bodies, and loss of cell morphology and nuclear division. There was a 19.05 % reduction in the number of apoptotic cells in the A9 treatment group compared with that in the IHNV group. In addition, enzyme activity assays proved that A9 suppressed the expression of caspase 3, 8 and 9. These results suggested that A9 inhibit viral replication, to some extent, by blocking IHNV-induced apoptosis. In an in vivo study, A9 exhibited an anti-rhabdovirus effect in virus-infected fish by substantially enhancing the survival rate. Consistent with the above results, A9 repressed IHNV gene expression in virus-sensitive tissues (brain, kidney and spleen) in the early stages of virus infection. Importantly, the data showed that horizontal transmission of IHNV was reduced by A9 in a static cohabitation challenge model, especially in fish that underwent bath treatment, suggesting that A9 might be a suitable therapeutic agent for IHNV in aquaculture. Therefore, coumarin derivatives can be developed as antiviral agents against rhabdoviruses.


Assuntos
Antivirais/síntese química , Cumarínicos/química , Rhabdoviridae/efeitos dos fármacos , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular , Cumarínicos/farmacologia , Cumarínicos/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Doenças dos Peixes/tratamento farmacológico , Doenças dos Peixes/mortalidade , Doenças dos Peixes/patologia , Humanos , Oncorhynchus mykiss/metabolismo , Oncorhynchus mykiss/virologia , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/mortalidade , Infecções por Rhabdoviridae/patologia , Relação Estrutura-Atividade , Taxa de Sobrevida , Proteínas Virais/genética , Proteínas Virais/metabolismo , Internalização do Vírus/efeitos dos fármacos
11.
J Fish Biol ; 98(1): 208-218, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33000466

RESUMO

Selenium, as an essential trace element, interferes through selenoproteins in many physiological processes of plants and mammals. Its antiviral activity has recently attracted much attention because selenium improves the antiviral capacity of animal cells against a few viruses relevant to human diseases. In this study, the red elemental selenium was purified from the fermentative culture of Herbaspirillum camelliae WT00C and then used to culture epithelioma papulosum cyprinid (EPC) cells or feed crucian carp and zebrafish. Finally, its antiviral effects were investigated at the cell level and living fishes after spring viraemia of carp virus infection. At the cell level, 5, 10 and 20 µg ml-1 red elemental selenium significantly induced the expression of interferon (IFN) and ISG15 genes in EPC cells. The viral TCID50 (50% tissue culture infective dose) values in the EPC cells incubated with 5, 10 and 20 µg ml-1 red elemental selenium were significantly less than those of the control. More expression of IFN and ISG15 genes and less TCID50 values indicate that red elemental selenium indeed improves the antiviral capability of EPC cells. In the crucian carp fed with the food containing 5 and 10 µg g-1 red elemental selenium, IFN expressions showed 13- and 39-fold increases at the 16th day of post-injection, and its expression was dependent on selenium concentrations. Meanwhile, no fish death occurred in all the experimental groups. In the zebrafish fed with the red worm containing 5 µg g-1 red elemental selenium, IFN and Mx expressions and survival rate were significantly higher than those of the control. The results of this study show that red elemental selenium indeed improves the antiviral activity of fish. The antiviral effects of selenium mainly come from its immune regulation through its incorporation into selenoproteins. The optimum level of selenium contributes to improving fish immunity, whereas excess selenium causes excessive immune and inflammatory responses.


Assuntos
Carpas/imunologia , Doenças dos Peixes/tratamento farmacológico , Doenças dos Peixes/imunologia , Infecções por Rhabdoviridae/veterinária , Selênio/farmacologia , Viremia/veterinária , Peixe-Zebra/imunologia , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/uso terapêutico , Animais , Antivirais/farmacologia , Carcinoma , Carpas/virologia , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Interferons/genética , Rhabdoviridae , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/imunologia , Selênio/uso terapêutico , Viremia/tratamento farmacológico , Viremia/imunologia , Peixe-Zebra/virologia
12.
Virus Res ; 291: 198221, 2021 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-33152382

RESUMO

Phenylpropanoids, common natural compounds, possess many different biological activities such as antioxidant, anti-inflammatory and antiviral. Spring viraemia of carp virus (SVCV) can cause a high mortality in common carp (Cyprinus carpio). However, there are currently no licenced drugs that effectively cure this disease. In this study, we designed and synthesized a phenylpropanoid derivative 4-(4-methoxyphenyl)-3,4-dihydro-2H-chromeno[4,3-d]pyrimidine-2,5(1 H)-dione (E2), and explored the antiviral effect against SVCV in vitro and in vivo. Up to 25 mg/L of E2 significantly inhibited the expression levels of SVCV protein genes in the epithelioma papulosum cyprini (EPC) cell line by a maximum inhibitory rate of >90%. As expected, E2 remarkably declined the apoptotic of SVCV-infected cells and suppressed potential enhancement of the mitochondrial membrane potential (ΔΨm), these data implied that E2 could protect mitochondria from structural damage in response to SVCV. Meanwhile, E2 was added to EPC cells under four different conditions: time-of-addition, time-of-removal, pre-treatment of viruses and pre-treatment of cells indicated that E2 may block the post-entry transport process of the virus. Additionally, the up-regulation of six interferon (IFN)-related genes also demonstrated that E2 indirectly activated IFNs for the clearance of SVCV in common carp. Drug cure effect showed that treatment with E2 at 0.5 d post infection (dpi) is more effective than at 0, 1 or 2 dpi. Most importantly, intraperitoneal therapy of E2 markedly improved common carp survival rate and reduced virus copies in body. Therefore, the E2 has potential to be developed into a novel anti-SVCV agent.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Carpas/virologia , Doenças dos Peixes/tratamento farmacológico , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/veterinária , Rhabdoviridae/efeitos dos fármacos , Viremia/tratamento farmacológico , Animais , Antivirais/síntese química , Linhagem Celular , Doenças dos Peixes/virologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Interferons/genética , Interferons/imunologia , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
13.
Zool Res ; 41(4): 395-409, 2020 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-32390373

RESUMO

Spring viremia of carp virus (SVCV) causes devastating losses in aquaculture. Coumarin has an advantageous structure for the design of novel antiviral agents with high affinity and specificity. In this study, we evaluated a hydroxycoumarin medicine, i.e., 7-(6-benzimidazole) coumarin (C10), regarding its anti-SVCV effects in vitro and in vivo. Results showed that up to 12.5 mg/L C10 significantly inhibited SVCV replication in the epithelioma papulosum cyprini (EPC) cell line, with a maximum inhibitory rate of >97%. Furthermore, C10 significantly reduced cell death and relieved cellular morphological damage in SVCV-infected cells. Decreased mitochondrial membrane potential (ΔΨm) also suggested that C10 not only protected mitochondria, but also reduced apoptosis in SVCV-infected cells. For in vivo studies, intraperitoneal injection of C10 resulted in an anti-SVCV effect and substantially enhanced the survival rate of virus-infected zebrafish. Furthermore, C10 significantly enhanced antioxidant enzyme activities and decreased reactive oxygen species (ROS) to maintain antioxidant-oxidant balance within the host, thereby contributing to inhibition of SVCV replication. The up-regulation of six interferon (IFN)-related genes also demonstrated that C10 indirectly activated IFNs for the clearance of SVCV in zebrafish. This was beneficial for the continuous maintenance of antiviral effects because of the low viral loads in fish. Thus, C10 is suggested as a therapeutic agent with great potential against SVCV infection in aquaculture.


Assuntos
Antivirais/farmacologia , Carpas , Cumarínicos/farmacologia , Doenças dos Peixes/tratamento farmacológico , Infecções por Rhabdoviridae/veterinária , Rhabdoviridae/efeitos dos fármacos , Animais , Linhagem Celular , Doenças dos Peixes/virologia , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/virologia , Carga Viral/efeitos dos fármacos , Carga Viral/veterinária
14.
Virus Res ; 285: 198019, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32417180

RESUMO

Rhabdoviruses cause devastating diseases in aquaculture, resulting in enormous economic losses. Our previous studies indicated that imidazole arctigenin derivatives possessed antiviral activities against aquatic rhabdoviruses. Based on the data of structure-activity relationship, a new imidazole arctigenin derivative, 4-(8-(2-bromoimidazole)octyloxy)-arctigenin (BOA), was designed and synthesized. And its antiviral activities against aquatic rhabdoviruses (SVCV, IHNV and MSRV) were evaluated in vitro. By comparing inhibitory concentration at half-maximal activity (IC 50), we found that BOA (IC50 = 1.11 µM) possessed a higher anti-IHNV activity than the antiviral imidazole arctigenin derivatives which were found in our previous study. Besides, BOA could cause profound inhibition of SVCV and MSRV replication. By the reduction assays on cytopathic effect, BOA exhibited a protective effect on two host cell lines. As a typical rhabdovirus, SVCV was chosen as a model to illuminate the anti-rhabdovirus mechanism of BOA. BOA was discovered to not impact directly on viral particles or interfere with SVCV adsorption. And it worked within the 2-6 h of the early phase of virus replication. In addition, after repression of cell cycle S phase and recovery of caspase-3/8/9 activities activated by SVCV, BOA inhibited SVCV-induced apoptosis and then reduced the release of viral particles at the late stage of virus replication. Altogether, BOA was expected to be a highly efficient antiviral agent against multiple rhabdoviruses in the field of aquaculture.


Assuntos
Antivirais/farmacologia , Doenças dos Peixes/tratamento farmacológico , Furanos/farmacologia , Lignanas/farmacologia , Infecções por Rhabdoviridae , Rhabdoviridae/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Aquicultura , Linhagem Celular , Peixes , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/veterinária
15.
Antiviral Res ; 174: 104672, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31825851

RESUMO

Spring viremia of carp virus (SVCV) is one of the most serious pathogens in aquaculture, resulting in devastating damage in cyprinid. In this study, we designed and synthesized a novel coumarin derivative (C3007) for evaluating its in vitro and in vivo anti-SVCV effects. Here, we determined that up to 25 mg/L C3007 significantly decreased SVCV protein gene expression levels in EPC cells by a maximum inhibitory rate of >95%. When C3007 was preincubated with SVCV, infectivity was significantly inhibited in vitro in a time-dependent manner, with complete inhibition at 25 mg/L. For in vivo studies, C3007 exhibited an anti-SVCV effect by substantially enhancing the survival rate of virus-infected fish via intraperitoneal injection. Although the horizontal transmission of SVCV was hindered by C3007 in a static cohabitation challenge model, it was not completely blocked, showing that the viral loads in recipient fish were obviously reduced. Thus, C3007 could potentially be used as a therapeutic agent with great potential in aquatic systems and may also be suitable for applications in pond aquaculture settings against viral transmission. Additionally, the C3007-preincubated virus induced an antiviral immune response with high levels of IFN expression, suggesting that C3007 pre-treatment could be used in vaccine development.


Assuntos
Antivirais/uso terapêutico , Carpas/virologia , Cumarínicos/uso terapêutico , Transmissão de Doença Infecciosa/veterinária , Doenças dos Peixes/tratamento farmacológico , Infecções por Rhabdoviridae/veterinária , Animais , Aquicultura , Cumarínicos/síntese química , Transmissão de Doença Infecciosa/prevenção & controle , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Interferons/imunologia , Rhabdoviridae/efeitos dos fármacos , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/imunologia , Replicação Viral/efeitos dos fármacos
16.
J Fish Dis ; 43(2): 185-195, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31762053

RESUMO

In a search for alternative, environmentally friendly and effective disinfecting agents, a commercially available protease-Neutrase® -was tested in this work for inactivation of koi herpesvirus (KHV) and of viral haemorrhagic septicaemia virus (VHSV). For comparison, the stability of these viral pathogens in similar configurations at various pH values and concentrations of peracetic acid or quicklime, typically used for disinfection, was tested. Therefore, virus suspensions were incubated with various concentrations of different agents for 24 hr and the titre of the remaining infectious particles was determined by virus titration. Furthermore, the treatment of both viruses, with the agents at concentrations that were previously appointed as effective, was also examined in the presence of solid material (quartz sand). All procedures investigated in this study, including the protease treatment, were able to reduce the titre of KHV and VHSV below the detection limit of the titration. Although further studies are necessary, this is the first report of the application of a protease for the inactivation of the selected fish pathogens, demonstrating the great potential of the latter for disinfection.


Assuntos
Antivirais/farmacologia , Herpesviridae/efeitos dos fármacos , Novirhabdovirus/efeitos dos fármacos , Animais , Doenças dos Peixes/tratamento farmacológico , Doenças dos Peixes/virologia , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Peptídeo Hidrolases/farmacologia , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/veterinária , Infecções por Rhabdoviridae/virologia , Carga Viral/veterinária
17.
Virus Res ; 273: 197741, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31494148

RESUMO

Infectious hematopoietic necrosis virus (IHNV) is a fish viral pathogen that causes severe disease and huge economic losses in the salmonid aquaculture industry. However, anti-IHNV drugs currently are scarce. For the purpose of seeking out anti-IHNV drugs, the anti-IHNV activities of 32 medicinal plants were investigated by using epithelioma papulosum cyprini (EPC) cells. Among these plants, Prunella vulgaris L. (PVL) showed the strongest inhibition on IHNV replication with an inhibitory percentage of 99.3% at the concentration 100 mg/L. Further studies demonstrated that ursolic acid (UA), a major constituent of PVL, also showed a highly effective anti-IHNV activity. The half-maximal inhibitory concentration (IC50) at 72 h of UA on IHNV was 8.0 µM. Besides, UA could significantly decrease cytopathic effect (CPE) and the viral titer induced by IHNV in EPC cells. More importantly, UA also showed a strong anti-IHNV activity in vivo, as indicated by increasing the survival rate of rainbow trout and inhibiting viral gene expression. Intraperitoneal injection of UA increased the relative percentage of survival of rainbow trout by 18.9% and inhibited IHNV glycoprotein mRNA expression by > 90.0% in the spleen at the 1st-day post-infection. Altogether, UA was expected to be a therapeutic agent against IHNV infection in aquaculture.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Vírus da Necrose Hematopoética Infecciosa/efeitos dos fármacos , Prunella/química , Infecções por Rhabdoviridae/veterinária , Triterpenos/farmacologia , Triterpenos/uso terapêutico , Animais , Aquicultura , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Doenças dos Peixes/tratamento farmacológico , Doenças dos Peixes/virologia , Vírus da Necrose Hematopoética Infecciosa/fisiologia , Concentração Inibidora 50 , Oncorhynchus mykiss/virologia , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Infecções por Rhabdoviridae/tratamento farmacológico , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
18.
Virus Res ; 268: 11-17, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31095989

RESUMO

Spring viraemia of carp (SVC) in aquaculture is challenging because there are few preventative measures and/or treatments. The previous study demonstrated that an antiviral coumarin derivative, 7-(4-(4-methyl-imidazole))-coumarin (C2), inhibits spring viremia of carp virus (SVCV) infection by targeting Nrf2-ARE signaling pathway in fish cells. Thus, we hypothesized whether C2 may be used as a potential therapeutic agent for controlling SVCV infection in aquaculture. In this study, SVCV infectivity was significantly inhibited in vitro in a dose-dependent manner by preincubation with C2. C2 was verified against SVCV in zebrafish, in which the mortality and viral titer in fish body were decreased. Like other coumarins, C2 was stable with a prolonged inhibitory half-life (3.5 days) at 15 °C in the early stage of SVCV infection. The results show that horizontal transmission of SVCV was reduced by C2 in a static cohabitation challenge model, especially for recipient fish in injection treatment, which suggested that C2 may be suitable as a possible therapeutic agent for SVCV in aquaculture. Overall, this study provides the new insight that a small molecule antiviral drug can be used to control rhabdovirus infection in fish aquacultures.


Assuntos
Antivirais/uso terapêutico , Cumarínicos/uso terapêutico , Doenças dos Peixes/tratamento farmacológico , Infecções por Rhabdoviridae/veterinária , Rhabdoviridae/efeitos dos fármacos , Animais , Antivirais/química , Aquicultura , Cumarínicos/química , Doenças dos Peixes/virologia , Infecções por Rhabdoviridae/tratamento farmacológico , Carga Viral/efeitos dos fármacos , Peixe-Zebra
19.
Virus Res ; 263: 112-118, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30658072

RESUMO

As an efficient pathogen resulting in economic impact in aquaculture, spring viremia of carp virus (SVCV) causes devastating disease in cyprinids. Based on the previous study that 7-(6-(2-methyl-imidazole))-coumarin (D5) exhibited anti-SVCV activity in fish cells, we hypothesized that D5 may be useful as a potential therapeutic agent for controlling SVCV infection in vivo. In this study, we verified that D5 inhibited SVCV replication in zebrafish, with reducing 22.5% mortality of SVCV-infected fish. Further data suggested that coumarin D5 was more stable with a prolonged inhibitory half-life in the early stage of virus infection (1-4 days). Consistent with above results, D5 decreased the viral titer in fish body and repressed SVCV glycoprotein gene expression in virus sensitive tissues (kidney and spleen) in the early stage of virus infection. In addition, the results replied that D5 elicited an innate immune response in non-viral infected zebrafish by up-regulating the expression of interferon genes (IFNγ, IFNφ1, IFNφ2 and RIG-1). D5 also enhanced the levels of antioxidant-related gene transcription and enzyme activities in SVCV-infected zebrafish, suggesting that D5 exhibited an antioxidant protection on fish by keeping the balance of redox state. Therefore, D5 is a potential therapeutic agent for the devastating fish rhabdovirus infections.


Assuntos
Antivirais/administração & dosagem , Cumarínicos/administração & dosagem , Imidazóis/administração & dosagem , Fatores Imunológicos/administração & dosagem , Infecções por Rhabdoviridae/tratamento farmacológico , Rhabdoviridae/isolamento & purificação , Animais , Antivirais/química , Antivirais/farmacocinética , Cumarínicos/química , Cumarínicos/farmacocinética , Modelos Animais de Doenças , Meia-Vida , Imidazóis/química , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/química , Fatores Imunológicos/farmacocinética , Infecções por Rhabdoviridae/virologia , Análise de Sobrevida , Resultado do Tratamento , Carga Viral , Peixe-Zebra
20.
Eur J Med Chem ; 163: 183-194, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30508667

RESUMO

Infectious hematopoietic necrosis virus (IHNV) is a common pathogen that causes severe disease and huge economic losses in the salmonid aquaculture industry. Herein, a series of arctigenin derivatives are synthesized to evaluate their antiviral activity against IHNV. The results indicate that the length of linker and imidazole substituent groups play an important role in decreasing IHNV replication. In this study, the arctigenin-imidazole hybrid derivative 15 with an eight carbon atoms length of the linker reduces IHNV replication with an IC50 value of 1.3 µM. In addition, derivative 15 significantly inhibits apoptosis and cellular morphological damage induced by IHNV. Mechanistically, derivative 15 can not damage the viral particle directly. While time-of-addition and viral binding assays reveal that derivative 15 mainly affect the early replication of IHNV but do not interfere with IHNV adsorption. Overall, derivative 15 could be considered to develop as a promising agent to treat IHNV infection.


Assuntos
Antivirais/síntese química , Furanos/uso terapêutico , Vírus da Necrose Hematopoética Infecciosa/efeitos dos fármacos , Lignanas/uso terapêutico , Infecções por Rhabdoviridae/tratamento farmacológico , Animais , Antivirais/farmacologia , Furanos/síntese química , Imidazóis/química , Imidazóis/farmacologia , Lignanas/síntese química , Salmão/virologia , Relação Estrutura-Atividade , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...