Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 482
Filtrar
1.
Vet Microbiol ; 295: 110107, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38838382

RESUMEN

Pseudorabies virus (PRV), an alphaherpesvirus, is a neglected zoonotic pathogen. Dectin-1 sensing of ß-glucan (BG) induces trained immunity, which can possibly form a new strategy for the prevention of viral infection. However, alphaherpesvirus including PRV have received little to no investigation in the context of trained immunity. Here, we found that BG pretreatment improved the survival rate, weight loss outcomes, alleviated histological injury and decreased PRV copy number of tissues in PRV-infected mice. Type I interferons (IFNs) including IFN-α/ß levels in serum were significantly increased by BG. However, these effects were abrogated in the presence of Dectin-1 antagonist. Dectin-1-mediated effect of BG was also confirmed in porcine and murine macrophages. These results suggested that BG have effects on type I IFNs with antiviral property involved in Dectin-1. In piglets, oral or injected immunization with BG and PRV vaccine could significantly elevated the level of PRV-specific IgG and type I IFNs. And it also increased the antibody levels of porcine reproductive and respiratory syndrome virus vaccine and classical swine fever vaccine that were later immunized, indicating a broad-spectrum effect on improving vaccine immunity. On the premise that the cost was greatly reducing, the immunological effect of oral was better than injection administration. Our findings highlighted that BG induced type I IFNs related antiviral effect against PRV involved in Dectin-1 and potential application value as a feed additive to help control the spread of PRV and future emerging viruses.


Asunto(s)
Herpesvirus Suido 1 , Interferón Tipo I , Lectinas Tipo C , Seudorrabia , beta-Glucanos , Animales , beta-Glucanos/farmacología , beta-Glucanos/administración & dosificación , Ratones , Porcinos , Lectinas Tipo C/inmunología , Seudorrabia/inmunología , Seudorrabia/prevención & control , Interferón Tipo I/inmunología , Herpesvirus Suido 1/inmunología , Herpesvirus Suido 1/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Antivirales/farmacología , Vacunas Virales/inmunología , Femenino
2.
Viruses ; 16(5)2024 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-38793591

RESUMEN

In recent years, pseudorabies virus (PRV) variants have resulted in an epidemic in swine herds and huge economic losses in China. Therefore, it is essential to develop an efficacious vaccine against the spread of PRV variants. Here, the triple-gene-deletion virus and the triple-gene-deletion plus gC virus were constructed by homologous recombination (HR). And then, their growth capacity, proliferation ability, and immune efficacy were evaluated. The results showed that the growth kinetics of the recombinant viruses were similar to those of the parental strain PRV-AH. Compared with the triple-gene-deletion virus group, the more dominant level of neutralizing antibody (NA) can be induced in the triple-gene-deletion plus gC virus group with the same 106.0 TCID50 dose after 4 and 6 weeks post-initial immunization (PII) (p < 0.0001). In addition, the antibody titers in mice immunized with the triple-gene-deletion plus gC virus were significantly higher than those immunized with triple-gene deletion virus with the same 105.0 TCID50 dose after 6 weeks PII (p < 0.001). More importantly, in the triple-gene-deletion plus gC virus group with 105.0 TCID50, the level of NA was close to that in the triple-gene deletion virus group with 106.0 TCID50 at 6 weeks PII. Meanwhile, the cytokines IL-4 and IFN-γ in sera were tested by enzyme-linked immunosorbent assay (ELISA) in each group. The highest level of IL-4 or IFN-γ was also elicited in the triple-gene deletion plus gC virus group at a dose of 106.0 TCID50. After challenge with PRV-AH, the survival rates of the triple-gene deletion plus gC virus immunized groups were higher than those of other groups. In immunized groups with 105.0 TCID50, the survival rate shows a significant difference between the triple-gene deletion plus gC virus group (75%, 6/8) and the triple-gene deletion virus group (12.5%, 1/8). In general, the immune efficacy of the PRV TK/gI/gE-deleted virus can be increased with additional gC insertion in mice, which has potential for developing an attenuated vaccine candidate for PRV control.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Eliminación de Gen , Herpesvirus Suido 1 , Vacunas contra la Seudorrabia , Seudorrabia , Animales , Herpesvirus Suido 1/genética , Herpesvirus Suido 1/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Ratones , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Seudorrabia/prevención & control , Seudorrabia/inmunología , Seudorrabia/virología , Vacunas contra la Seudorrabia/inmunología , Vacunas contra la Seudorrabia/genética , Vacunas contra la Seudorrabia/administración & dosificación , Ratones Endogámicos BALB C , Porcinos , Femenino , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Recombinación Homóloga , Citocinas/metabolismo , China
3.
Int J Biol Macromol ; 269(Pt 2): 132172, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38719009

RESUMEN

Adjuvants including aluminum adjuvant (Alum) and oil-water emulsion have been widely used in inactivated pseudorabies virus (PRV) vaccines to improve their performance, however, they are not sufficient to protect from PRV infection because of the weak immune response and poor Th1-type immune response. Divalent manganese ion (Mn2+) has been reported to increase the cellular immune response significantly. In this work, a xanthan gum and carbomer-dispersed Mn2+-loaded tannic acid-polyethylene glycol (TPMnXC) nanoparticle colloid is developed and used as an adjuvant to improve the performance of the inactivated PRV vaccine. The good in vitro and in vivo biocompatibility of the developed TPMnXC colloid has been confirmed by the cell viability assay, erythrocyte hemolysis, blood routine analysis, and histological analysis of mouse organs and injection site. The TPMnXC-adjuvanted inactivated PRV vaccine (TPMnXC@PRV) significantly promotes higher and more balanced immune responses indicating with an increased specific total IgG antibody and IgG2a/IgG1 ratio, efficient splenocytes proliferation, and elevated Th1- and Th2-type cytokine secretion than those of control groups. Wild PRV challenge experiment is performed using mice as a model animal, achieving a protection rate of up to 86.67 %, which is much higher than those observed from the commercial Alum. This work not only demonstrates the high potentiality of TPMnXC in practical applications but also provides a new way to develop the Mn2+-loaded nanoadjuvant for veterinary vaccines.


Asunto(s)
Adyuvantes Inmunológicos , Herpesvirus Suido 1 , Inmunidad Celular , Inmunidad Humoral , Manganeso , Nanopartículas , Polisacáridos Bacterianos , Taninos , Animales , Ratones , Adyuvantes Inmunológicos/farmacología , Nanopartículas/química , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Taninos/química , Taninos/farmacología , Manganeso/química , Polisacáridos Bacterianos/química , Polisacáridos Bacterianos/farmacología , Polisacáridos Bacterianos/inmunología , Herpesvirus Suido 1/inmunología , Vacunas contra la Seudorrabia/inmunología , Vacunas de Productos Inactivados/inmunología , Seudorrabia/prevención & control , Seudorrabia/inmunología , Femenino , Citocinas/metabolismo , Ratones Endogámicos BALB C , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Polifenoles
4.
PLoS Pathog ; 20(4): e1012139, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38578790

RESUMEN

Alpha herpesviruses naturally infect the peripheral nervous system, and can spread to the central nervous system, causing severe debilitating or deadly disease. Because alpha herpesviruses spread along synaptic circuits, and infected neurons exhibit altered electrophysiology and increased spontaneous activity, we hypothesized that alpha herpesviruses use activity-dependent synaptic vesicle-like regulated secretory mechanisms for egress and spread from neurons. Using live-cell fluorescence microscopy, we show that Pseudorabies Virus (PRV) particles use the constitutive Rab6 post-Golgi secretory pathway to exit from the cell body of primary neurons, independent of local calcium signaling. Some PRV particles colocalize with Rab6 in the proximal axon, but we did not detect colocalization/co-transport in the distal axon. Thus, the specific secretory mechanisms used for viral egress from axons remains unclear. To address the role of neuronal activity more generally, we used a compartmentalized neuron culture system to measure the egress and spread of PRV from axons, and pharmacological and optogenetics approaches to modulate neuronal activity. Using tetrodotoxin to silence neuronal activity, we observed no inhibition, and using potassium chloride or optogenetics to elevate neuronal activity, we also show no increase in virus spread from axons. We conclude that PRV egress from neurons uses constitutive secretory mechanisms: generally, activity-independent mechanisms in axons, and specifically, the constitutive Rab6 post-Golgi secretory pathway in cell bodies.


Asunto(s)
Alphaherpesvirinae , Herpesvirus Suido 1 , Seudorrabia , Animales , Cuerpo Celular/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Axones , Alphaherpesvirinae/metabolismo , Neuronas , Herpesvirus Suido 1/metabolismo , Seudorrabia/metabolismo , Exocitosis
5.
Vet Microbiol ; 290: 110011, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38310713

RESUMEN

Senecavirus A (SVA)-associated porcine idiopathic vesicular disease (PIVD) and Pseudorabies (PR) are highly contagious swine disease that pose a significant threat to the global pig industry. In the absence of an effective commercial vaccine, outbreaks caused by SVA have occurred in many parts of the world. In this study, the PRV variant strain PRV-XJ was used as the parental strain to construct a recombinant PRV strain with the TK/gE/gI proteins deletion and the VP3 protein co-expression, named rPRV-XJ-ΔTK/gE/gI-VP3. The results revealed that PRV is a suitable viral live vector for VP3 protein expressing. As a vaccine, rPRV-XJ-ΔTK/gE/gI-VP3 is safe for mice, vaccination with it did not cause any clinical symptoms of PRV. Intranasal immunization with rPRV-XJ-ΔTK/gE/gI-VP3 induced strong cellular immune response and high levels of specific antibody against VP3 and gB and neutralizing antibodies against both PRV and SVA in mice. It provided 100% protection to mice against the challenge of virulent strain PRV-XJ, and alleviated the pathological lesion of heart and liver tissue in SVA infected mice. rPRV-XJ-ΔTK/gE/gI-VP3 appears to be a promising vaccine candidate against PRV and SVA for the control of the PRV variant and SVA.


Asunto(s)
Herpesvirus Suido 1 , Picornaviridae , Seudorrabia , Enfermedades de los Roedores , Enfermedades de los Porcinos , Vacunas Virales , Porcinos , Animales , Ratones , Proteínas del Envoltorio Viral , Anticuerpos Antivirales , Vacunas contra la Seudorrabia
6.
PLoS Pathog ; 20(1): e1011956, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38295116

RESUMEN

Viral infection is a significant risk factor for fertility issues. Here, we demonstrated that infection by neurotropic alphaherpesviruses, such as pseudorabies virus (PRV), could impair female fertility by disrupting the hypothalamus-pituitary-ovary axis (HPOA), reducing progesterone (P4) levels, and consequently lowering pregnancy rates. Our study revealed that PRV exploited the transient receptor potential mucolipin 1 (TRPML1) and its lipid activator, phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), to facilitate viral entry through lysosomal cholesterol and Ca2+. P4 antagonized this process by inducing lysosomal storage disorders and promoting the proteasomal degradation of TRPML1 via murine double minute 2 (MDM2)-mediated polyubiquitination. Overall, the study identifies a novel mechanism by which PRV hijacks the lysosomal pathway to evade P4-mediated antiviral defense and impair female fertility. This mechanism may be common among alphaherpesviruses and could contribute significantly to their impact on female reproductive health, providing new insights for the development of antiviral therapies.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Femenino , Ratones , Animales , Herpesvirus Suido 1/fisiología , Progesterona/farmacología , Progesterona/metabolismo , Internalización del Virus , Lisosomas/metabolismo , Antivirales/metabolismo , Seudorrabia/metabolismo
7.
Vet Microbiol ; 290: 109974, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38262115

RESUMEN

Pseudorabies virus (PRV) is a neurotropic virus, which infects a wide range of mammals. The activity of PRV is gradually suppressed in hosts that have tolerated the primary infection. Increased glucocorticoid levels resulting from stressful stimuli overcome repression of PRV activity. However, the host cell mechanism involved in the activation processes under stressful conditions remains unclear. In this study, infection of rat PC-12 pheochromocytoma cells with neuronal properties using PRV at a multiplicity of infection (MOI) = 1 for 24 h made the activity of PRV be the relatively repressed state, and then incubation with 0.5 µM of the corticosteroid dexamethasone (DEX) for 4 h overcomes the relative repression of PRV activity. RNA-seq deep sequencing and bioinformatics analyses revealed different microRNA and mRNA profiles of PC-12 cells with/without PRV and/or DEX treatment. qRT-PCR and western blot analyses confirmed the negative regulatory relationship of miRNA-194-5p and its target heparin-binding EGF-like growth factor (Hbegf); a dual-luciferase reporter assay revealed that Hbegf is directly targeted by miRNA-194-5p. Further, miRNA-194-5p mock transfection contributed to PRV activation, Hbegf was downregulated in DEX-treated PRV infection cells, and Hbegf overexpression contributed to returning activated PRV to the repression state. Moreover, miRNA-194-5p overexpression resulted in reduced levels of HBEGF, c-JUN, and p-EGFR, whereas Hbegf overexpression suppressed the reduction caused by miRNA-194-5p overexpression. Overall, this study is the first to report that changes in the miR-194-5p-HBEGF/EGFR pathway in neurons are involved in DEX-induced activation of PRV, laying a foundation for the clinical prevention of stress-induced PRV activation.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales , Herpesvirus Suido 1 , MicroARNs , Feocromocitoma , Seudorrabia , Enfermedades de los Roedores , Ratas , Animales , Herpesvirus Suido 1/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Feocromocitoma/veterinaria , MicroARNs/genética , MicroARNs/metabolismo , Receptores ErbB/metabolismo , Neoplasias de las Glándulas Suprarrenales/veterinaria , Dexametasona/farmacología , Mamíferos
8.
Vet Microbiol ; 288: 109931, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38056181

RESUMEN

Since late 2011, the PRV variants have emerged in China, characterized by the increased virulence. The traditional attenuated vaccines have proven insufficient in providing complete protection, resulting in substantial economic losses to swine industry. In this study, a vaccine candidate strain, ZJ01-ΔgI/gE/TK/UL21, carrying the quadruple gene deletion was derived from the previously generated three gene-deleted virus ZJ01-ΔgI/gE/TK. As anticipated, piglets inoculated with ZJ01-ΔgI/gE/TK/UL21 exhibited normal body temperatures and showed no viral shedding, consistent with the observations from piglets treated with ZJ01-ΔgI/gE/TK. Importantly, a significant higher level of interferon induction was observed among piglets in the ZJ01-ΔgI/gE/TK/UL21 group compared to those in the ZJ01-ΔgI/gE/TK group. Upon challenge with the PRV variant ZJ01, piglets immunized with ZJ01-ΔgI/gE/TK/UL21 exhibited reduced viral shedding compared to the ZJ01-ΔgI/gE/TK group. Furthermore, piglets vaccinated with ZJ01-ΔgI/gE/TK/UL21 exhibited minimal pathological lesions in brain tissues, similar to those in the ZJ01-ΔgI/gE/TK group. These results underscore the potential of ZJ01-ΔgI/gE/TK/UL21 as a promising vaccine for controlling PRV infection.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Enfermedades de los Porcinos , Porcinos , Animales , Virulencia , Proteínas del Envoltorio Viral/genética , Vacunas Atenuadas , Vacunas contra la Seudorrabia
9.
J Vet Med Sci ; 86(1): 120-127, 2024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38030279

RESUMEN

An approach to genetically engineered resistance to pseudorabies virus (PRV) infection was examined by using a mouse model with defined point mutation in primary receptor for alphaherpesviruses, nectin-1, by the CRISPR/Cas9 system. It has become clear that phenylalanine at position 129 of nectin-1 is important for binding to viral glycoprotein D (gD), and mutation of phenylalanine 129 to alanine (F129A) prevents nectin-1 binding to gD and virus entry in vitro. Here, to assess the antiviral potential of the single amino acid mutation of nectin-1, F129A, in vivo, we generated genome-edited mutant mouse lines; F129A and 135 knockout (KO). The latter, 135 KO used as a nectin-1 knockout line for comparison, expresses a carboxy-terminal deleted polypeptide consisting of 135 amino acids without phenylalanine 129. In the challenge with 10 LD50 PRV via intranasal route, perfect protection of disease onset was induced by expression of the mutation of nectin-1, F129A (survival rate: 100% in F129A and 135 KO versus 0% in wild type mice). Neither viral DNA/antigens nor pathological changes were detected in F129A, suggesting that viral entry was prevented at the primary site in natural infection. In the challenge with 50 LD50 PRV, lower but still strong protective effect against disease onset was observed (survival rate: 57% in F129A and 75% in 135 KO versus 0% in wild type mice). The present results indicate that single amino acid mutation of nectin-1 F129A provides significant resistance against lethal pseudorabies.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Animales , Ratones , Aminoácidos/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Mutación , Nectinas/genética , Nectinas/metabolismo , Fenilalanina/genética , Fenilalanina/metabolismo , Seudorrabia/prevención & control , Proteínas del Envoltorio Viral/genética
10.
Virol J ; 20(1): 303, 2023 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-38115115

RESUMEN

BACKGROUND: Pseudorabies virus (PRV) causes substantial losses in the swine industry worldwide. Attenuated PRV strains with deletions of immunomodulatory genes glycoprotein E (gE), glycoprotein I (gI) and thymidine kinase (TK) are candidate vaccines. However, the effects of gE/gI/TK deletions on PRV-host interactions are not well understood. METHODS: To characterize the impact of gE/gI/TK deletions on host cells, we analyzed and compared the transcriptomes of PK15 cells infected with wild-type PRV (SD2017), PRV with gE/gI/TK deletions (SD2017gE/gI/TK) using RNA-sequencing. RESULTS: The attenuated SD2017gE/gI/TK strain showed increased expression of inflammatory cytokines and pathways related to immunity compared to wild-type PRV. Cell cycle regulation and metabolic pathways were also perturbed. CONCLUSIONS: Deletion of immunomodulatory genes altered PRV interactions with host cells and immune responses. This study provides insights into PRV vaccine design.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Enfermedades de los Porcinos , Porcinos , Animales , Herpesvirus Suido 1/genética , Timidina Quinasa/genética , Proteínas del Envoltorio Viral/genética , Glicoproteínas/genética , Perfilación de la Expresión Génica
11.
J Mater Chem B ; 12(1): 122-130, 2023 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-37997769

RESUMEN

With the urgent need for antiviral agents, antiviral materials with high biocompatibility and antiviral effects have attracted a lot of attention. In this study, gallic acid, a natural polyphenolic compound, was transformed into biocompatible graphene quantum dots (GAGQDs) which exhibit enhanced antiviral activity against pseudorabies virus (PRV). The as-prepared GAGQDs inhibit PRV proliferation with a 104-fold reduction in viral titers. Investigation of the antiviral mechanism revealed that GAGQDs inhibit the adsorption, invasion and replication of PRV infection. Treatment with GAGQDs regulates the expression levels of interferon-related antiviral proteins, including mitochondrial antiviral-signaling protein (MAVS), signal transducer and activator of transcription 1 (STAT1) and 2',5'-oligoadenylate synthetase 1 (OAS1), suggesting that GAGQDs can stimulate innate antiviral immune responses, resulting in enhanced antiviral effects. More importantly, GAGQD treatments alleviate clinical symptoms and reduce mortality in PRV-infected mice. Our results reveal the enhanced therapeutic effects of GAGQDs against PRV infection in vitro and in vivo, suggesting the potential of GAGQDs as a promising novel antiviral agent.


Asunto(s)
Grafito , Herpesvirus Suido 1 , Seudorrabia , Puntos Cuánticos , Ratones , Animales , Herpesvirus Suido 1/fisiología , Interferones/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Grafito/farmacología , Grafito/uso terapéutico , Seudorrabia/tratamiento farmacológico , Inmunidad Innata
12.
J Biol Chem ; 299(11): 105347, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37838171

RESUMEN

The pseudorabies virus (PRV) TJ strain, a variant of PRV, induces more severe neurological symptoms and higher mortality in piglets and mice than the PRV SC strain isolated in 1980. However, the mechanism underlying responsible for the discrepancy in virulence between these strains remains unclear. Our study investigated the differences in neurotropism between PRV TJ and PRV SC using both in vitro and in vivo models. We discovered that PRV TJ enters neural cells more efficiently than PRV SC. Furthermore, we found that PRV TJ has indistinguishable genomic DNA replication capability and axonal retrograde transport dynamics compared to the PRV SC. To gain deeper insights into the mechanisms underlying these differences, we constructed gene-interchanged chimeric virus constructs and assessed the affinity between envelope glycoprotein B, C, and D (gD) and corresponding receptors. Our findings confirmed that mutations in these envelope proteins, particularly gD, significantly contributed to the heightened attachment and penetration capabilities of PRV TJ. Our study revealed the critical importance of the gDΔR278/P279 and gDV338A in facilitating viral invasion. Furthermore, our observations indicated that mutations in envelope proteins have a more significant impact on viral invasion than on virulence in the mouse model. Our findings provide valuable insights into the roles of natural mutations on the PRV envelope glycoproteins in cell tropism, which sheds light on the relationship between cell tropism and clinical symptoms and offers clues about viral evolution.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Proteínas del Envoltorio Viral , Tropismo Viral , Animales , Ratones , Genómica , Herpesvirus Suido 1/genética , Mutagénesis , Mutación , Seudorrabia/genética , Porcinos , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
13.
Comp Immunol Microbiol Infect Dis ; 101: 102054, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37651789

RESUMEN

Porcine circovirus type 2 (PCV2) plays a key role in the etiology of PCV2-associated disease (PCVAD), and its predominant strain is PCV2d which is not completely controlled by most commercially available vaccines against PCV2a strains. Pseudorabies (PR) caused by pseudorabies virus (PRV) variants re-emerged in Bartha-K61 vaccine-immunized swine herds in late 2011, which brought considerable losses to the global pig husbandry. Therefore, it is significantly important to develop a safe and effective vaccine against both PCV2d and PRV infection. In the present study, the PCV2d ORF2 gene was amplified by PCR, and cloned into the BamHI site of PRV transfer plasmid pG vector to obtain the recombinant transfer plasmid pG-PCV2dCap-EGFP. Subsequently, it was transfected into ST cells infected with the three gene deleted PRV variant strain NY-gE-/gI-/TK- to generate a recombinant virus rPRV NY-gE-/gI-/TK-/PCV2dCap+/EGFP+, and then the EGFP gene was knocked out to harvest the rPRV NY-gE-/gI-/TK-/PCV2dCap+ using gene-editing technology termed CRISPR/Cas9 system. The recombinant virus rPRV NY-gE-/gI-/TK-/PCV2dCap+ had similar genetic stability and proliferation characteristics to the parental PRV as indicated by PCR and one-step growth curve test, and the expression of Cap was validated by Western blot. In animal experiment, higher PCV2-specific ELISA antibodies and detectable PCV2-specific neutralizing antibodies could be elicited in mice immunized with rPRV NY-gE-/gI-/TK-/PCV2dCap+ compared to commercial PCV2 inactivated vaccine. Moreover, the recombinant virus rPRV NY-gE-/gI-/TK-/PCV2dCap+ significantly reduced the viral loads in the hearts, livers, spleens, lungs, and kidneys in mice following a virulent PCV2d challenge. Mice immunized with rPRV NY-gE-/gI-/TK-/PCV2dCap+ developed comparable PRV-specific humoral immune responses and provided complete protection against a lethal PRV challenge. Together, the rPRV NY-gE-/gI-/TK-/PCV2dCap+ recombinant strain has strong immunogenicity.


Asunto(s)
Circovirus , Herpesvirus Suido 1 , Seudorrabia , Enfermedades de los Porcinos , Vacunas Virales , Porcinos , Animales , Ratones , Herpesvirus Suido 1/genética , Circovirus/genética , Seudorrabia/prevención & control , Proteínas del Envoltorio Viral/genética , Vacunas Virales/genética , Anticuerpos Antivirales
14.
Biologicals ; 83: 101692, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37442044

RESUMEN

Productivity and stability of Pseudorabies virus (PRV) are critical for the manufacture and storage of live attenuated pseudorabies vaccine. Trehalose is commonly used as a cryoprotectant to stabilize organisms during freezing and lyophilization. Trehalose transporter 1 (Tret1), derived from Polypedilum vanderplanki, can deliver trehalose with a reversible transporting direction. In this study, we demonstrated that productivity and stability of PRV proliferated in recombinant ST cells with stable expression of Tret1 were enhanced. As a result, a five-fold increase of intracellular trehalose amount was observed, and the significant increase of progeny viral titer was achieved in recombinant cells with the addition of 20 mM trehalose. Particularly, after storage for 8 weeks at 20 °C, the loss of viral titer was 0.8 and 1.7 lgTCID50/mL lower than the control group with or without the addition of trehalose. Additionally, the freeze-thaw resistance at -20 °C and -70 °C of PRV was significantly enhanced. Furthermore, according to standard international protocols, a series of tests, including karyotype analysis, tumorigenicity, and the ability of proliferation PRV, were conducted. Our results demonstrated that the recombinant ST cell with Tret1 is a promising cell substrate and has a high potential for producing more stable PRV for the live attenuated vaccine.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Enfermedades de los Porcinos , Animales , Porcinos , Herpesvirus Suido 1/metabolismo , Trehalosa/metabolismo , Seudorrabia/prevención & control , Congelación , Enfermedades de los Porcinos/prevención & control
15.
Virus Res ; 331: 199111, 2023 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-37062496

RESUMEN

Pseudorabies (PR) and classical swine fever (CSF) are economically important infectious diseases in pigs. Most pig farms in China are vaccinated against these two diseases. Gene-deleted pseudorabies virus (PRV) can be used to develop promising and economical multivalent live attenuated viral vector vaccines. It has been reported that recombinant PRV can express a truncated E2 protein (1-338 aa), but it has not been reported that recombinant PRV can express a full-length E2 protein. We constructed nine groups of E2 proteins with different expression forms and found that the E2 protein could be expressed in vitro only when the transmembrane region of E2 was removed and the signal peptide was added. Analysis of the transmembrane region of E2 revealed that the high hydrophobicity of the E2 transmembrane region was the main reason for its inability to express. By mutating an amino acid to reduce the hydrophobicity of the transmembrane region, it was found that the full-length mutant of E2 (E2FL-muta3 or E2FL-muta4) could be expressed. The expressed full-length mutant E2 could also localize to the cell membrane. Mice immunized with a PRV vector vaccine expressing E2FL-muta3 or E2FL-muta4 developed specific cellular immunity to the E2 protein and stimulated higher levels of E2 antibody than mice immunized with a PRV vector expressing truncated E2. After immunizing the rabbits, the lethal challenge by PRV-ZJ2013 and the febrile response elicited by CSFV were simultaneously prevented. These results suggest that rPRV-dTK/gE-E2FL-muta4 is a promising bivalent vaccine against CSFV and PRV infections.


Asunto(s)
Virus de la Fiebre Porcina Clásica , Peste Porcina Clásica , Herpesvirus Suido 1 , Seudorrabia , Enfermedades de los Porcinos , Vacunas Virales , Animales , Porcinos , Ratones , Conejos , Herpesvirus Suido 1/genética , Virus de la Fiebre Porcina Clásica/genética , Aminoácidos/genética , Vacunas Virales/genética , Anticuerpos Antivirales , Inmunización , Seudorrabia/prevención & control , Mutación , Proteínas del Envoltorio Viral/genética
16.
J Biol Chem ; 299(4): 104605, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36918100

RESUMEN

Pseudorabies virus (PRV) has become a "new life-threatening zoonosis" since the human-originated PRV strain was first isolated in 2020. To identify novel anti-PRV agents, we screened a total of 107 ß-carboline derivatives and found 20 compounds displaying antiviral activity against PRV. Among them, 14 compounds showed better antiviral activity than acyclovir. We found that compound 45 exhibited the strongest anti-PRV activity with an IC50 value of less than 40 nM. Our in vivo studies showed that treatment with 45 significantly reduced the viral loads and protected mice challenged with PRV. To clarify the mode of action of 45, we conducted a time of addition assay, an adsorption assay, and an entry assay. Our results indicated that 45 neither had a virucidal effect nor affected viral adsorption while significantly inhibiting PRV entry. Using the FITC-dextran uptake assay, we determined that 45 inhibits macropinocytosis. The actin-dependent plasma membrane protrusion, which is important for macropinocytosis, was also suppressed by 45. Furthermore, the kinase DYRK1A (dual-specificity tyrosine phosphorylation-regulated kinase 1A) was predicted to be a potential target for 45. The binding of 45 to DYRK1A was confirmed by drug affinity responsive target stability and cellular thermal shift assay. Further analysis revealed that knockdown of DYRK1A by siRNA suppressed PRV macropinocytosis and the tumor necrosis factor alpha-TNF-induced formation of protrusions. These results suggested that 45 could restrain PRV macropinocytosis by targeting DYRK1A. Together, these findings reveal a unique mechanism through which ß-carboline derivatives restrain PRV infection, pointing to their potential value in the development of anti-PRV agents.


Asunto(s)
Antivirales , Carbolinas , Herpesvirus Suido 1 , Animales , Humanos , Ratones , Aciclovir/farmacología , Aciclovir/toxicidad , Antivirales/química , Antivirales/farmacología , Antivirales/uso terapéutico , Carbolinas/química , Carbolinas/farmacología , Carbolinas/uso terapéutico , Técnicas de Silenciamiento del Gen , Herpesvirus Suido 1/efectos de los fármacos , Concentración 50 Inhibidora , Pinocitosis/efectos de los fármacos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Seudorrabia/tratamiento farmacológico , Seudorrabia/prevención & control , Seudorrabia/virología , Internalización del Virus/efectos de los fármacos , Células HeLa , Modelos Químicos , Quinasas DyrK
17.
J Virol ; 97(2): e0187122, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36648234

RESUMEN

Pseudorabies virus (PRV) is a neurotropic virus causing obvious neurological disorders and reproductive failure in pigs. PRV entry into target cells is a complex multistep process initiated by interacting viral envelope glycoproteins with cellular receptors. In the current study, we found that thrombospondin 3 (THBS3) plays an important role in PRV entry into target cells, indicating that THBS3 is a new PRV coreceptor. To confirm this hypothesis, the knockdown of THBS3 in several permissive cells inhibited PRV primary infection, and overexpression of THBS3 in PK15 cells promoted PRV infection. CRISPR-Cas9 knockout markedly reduced PRV infection in PK15 cells. Antibodies against THBS3 blocked PRV infection in naturally permissive target cells. Moreover, soluble THBS3 protein neutralized the infectivity of PRV. Mechanistically, THBS3 interacted with the PRV gD via its N and C termini to facilitate PRV binding in permissive and nonpermissive cells. Also, in the absence of Nectin-1, THBS3 promoted cell-to-cell fusion mediated by virus glycoproteins. While THBS3 alone could not increase virus entry, overexpression of it in the presence of Nectin-1 promoted virus entry into CHO-K1 cells. Our results have identified THBS3 as a critical player in PRV binding and subsequent membrane fusion and entry. IMPORTANCE Herpesvirus entry occurs through a cascade of virus-cell interactions, and multiple surface glycoproteins play a role in virus binding and entry during the virus invasion process. Early studies showed that attachment to cells by PRV, as well as other alphaherpesviruses, is mediated by interactions between the viral glycoprotein gC and cell membrane proteoglycans carrying heparan sulfate chains (HSPGs). However, gD may also be involved in virus binding in an HSPG-independent manner. To date, the respective cellular receptors are still unknown. In this report, we identified a host molecule, THBS3, involved in gD-mediated PRV binding and subsequent membrane fusion and entry, which increases our understanding of the initial events in alpha herpesvirus infections.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Acoplamiento Viral , Internalización del Virus , Animales , Cricetinae , Células CHO , Herpesvirus Suido 1/metabolismo , Herpesvirus Suido 1/patogenicidad , Nectinas/genética , Nectinas/metabolismo , Porcinos , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Técnicas de Silenciamiento del Gen
18.
Antiviral Res ; 211: 105548, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36702445

RESUMEN

Pseudorabies (PR) and classical swine fever (CSF) are economically important infectious diseases of pigs. Most pig farms in China are immunized against these two diseases. Here, we describe a stabilized E2 protein as an immunogen inserted into the PRV genome as a bivalent live virus-vectored vaccine. The E2 protein has 48 variant sites, there are 2-5 candidate amino acids per variant site, and the relative energy contribution of each amino acid to E2 energy was calculated. Combined substitutions of amino acids at the neighbor variant site (neighbor substitution) were performed to obtain the E2 protein sequence with the lowest energy (stabilized E2). Multiple amino acid substitutions at 48 variant sites were performed, and the results were consistent with neighbor substitutions. The stabilized E2 sequence was obtained, and its energy decreased by 22 Rosetta Energy Units (REUs) compared with the original sequence. After the recombinant PRV expressing stabilized E2 of CSFV was constructed, the secretion efficiency of stabilized E2 was increased by 2.97 times, and the thermal stability was increased by 10.5 times. Immunization of mice resulted in a 2-fold increase in antibody production, and a balanced antibody level against subtype 1.1 and subtype 2.1d E2 was achieved. In rabbits immunized, the lethal challenge of PRV-ZJ and the fever response induced by CSFV could be prevented simultaneously. These findings suggest that rPRV-muta/287aaE2 is a promising bivalent vaccine against CSFV and PRV infections.


Asunto(s)
Virus de la Fiebre Porcina Clásica , Peste Porcina Clásica , Herpesvirus Suido 1 , Seudorrabia , Vacunas Virales , Conejos , Animales , Porcinos , Ratones , Virus de la Fiebre Porcina Clásica/genética , Herpesvirus Suido 1/genética , Seudorrabia/prevención & control , Aminoácidos , Proteínas del Envoltorio Viral/genética , Anticuerpos Antivirales
19.
J Med Virol ; 95(1): e28228, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36251622

RESUMEN

Pseudorabies virus (PRV), as a neuroherpes virus, leads to heavy economic losses in the pig industry worldwide. This study was designed to establish recombinant PRV glycoprotein B (gB), C, and D proteins as PRV diagnostic antigens. The gB/C, gC/D, and gB/C/D fusion sequences were synthesized and inserted into pET-28a+ vector to generate the recombinant plasmids. The identified positive recombinant plasmids were transformed into BL21 Escherichia coli. The results of the polymerase chain reaction and enzyme digestion showed that the gB/C, gC/D, and gB/C/D fusion proteins were successfully expressed. An indirect sandwich ELISA was developed with the gB/C, gC/D, and gB/C/D as coating antigens. The results of indirect enzyme-linked immunosorbent assay (ELISA) analysis of 184 PRV-positive porcine sera showed that the positive coincidence rates of three recombinant proteins ELISAs relative to IDEXX kit were 98.25%, 95.32%, and 98.83%, and the negative coincidence rates were 85.71%, 75% and 100%, respectively. The inter and intra batch repeatability tests showed that the coefficient of variations of our kits were all less than 5%. Especially, the gB/C/D-ELISA has the highest specificity and sensitivity among the ELISA methods developed in this study. We established a series expression system of gB/C, gC/D, and gB/C/D antigen epitope genes and Recombinant protein-based indirect ELISA, providing new ideas for PV diagnosis and vaccine development.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Animales , Porcinos , Seudorrabia/diagnóstico , Seudorrabia/prevención & control , Proteínas Recombinantes , Proteínas del Envoltorio Viral , Ensayo de Inmunoadsorción Enzimática/métodos , Herpesvirus Suido 1/genética , Epítopos/metabolismo , Anticuerpos Antivirales
20.
Int J Mol Sci ; 23(19)2022 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-36232898

RESUMEN

Pseudorabies (also called Aujeszky's disease) is a highly infectious viral disease caused by the pseudorabies virus (PRV, or Suid herpesvirus 1). Although the disease has been controlled by immunization with the PRV-attenuated vaccine, the emerging PRV variants can escape the immune surveillance in the vaccinated pig, resulting in recent outbreaks. Furthermore, the virus has been detected in other animals and humans, indicating cross-transmission of PRV. However, the mechanism of PRV cross-species transmission needs further study. In this study, we compared the amino acid sequences of glycoproteins (gD), gL, and thymidine kinase (TK) of PRV strains, human PRV hSD-1 2019 strain, and the attenuated strain Bartha-K61, followed by predication of their spatial conformation. In addition, the interactions between the viral gD protein and host nectin-1, nectin-2, and HS were also evaluated via molecular docking. The results showed that the amino acid sequence homology of the gD, gL, and TK proteins of hSD-1 2019 and JL-CC was 97.5%, 94.4%, and 99.1%, respectively. Moreover, there were mutations in the amino acid sequences of gD, gL, and TK proteins of hSD-1 2019 and JL-CC compared with the corresponding reference sequences of the Bartha strain. The mutations of gD, gL, and TK might not affect the spatial conformation of the protein domain but may affect the recognition of antibodies and antigen epitopes. Moreover, the gD protein of JL-CC, isolated previously, can bind to human nectin-1, nectin-2, and HS, suggesting the virus may be highly infectious and pathogenic to human beings.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Enfermedades de los Porcinos , Animales , Epítopos/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Herpesvirus Suido 1/genética , Humanos , Simulación del Acoplamiento Molecular , Mutación , Nectinas/metabolismo , Porcinos , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Vacunas Atenuadas , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA