Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vet Res Commun ; 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38528300

RESUMO

Equine influenza (EI) is a highly contagious acute respiratory disease of equines caused by the H3N8 subtype of Influenza A virus i.e. equine influenza virus (EIV). Vaccination is an important and effective tool for the control of EI in equines. Most of the commercial influenza vaccines are produced in embryonated hen's eggs which has several inherent disadvantages. Hence, subunit vaccine based on recombinant haemagglutinin (HA) antigen, being the most important envelope glycoprotein has been extensively exploited for generating protective immune responses, against influenza A and B viruses. We hypothesized that novel vaccine formulation using baculovirus expressed recombinant HA1 (rHA1) protein coupled with bacteriophage will generate strong protective immune response against EIV. In the present study, the recombinant HA1 protein was produced in insect cells using recombinant baculovirus having cloned HA gene of EIV (Florida clade 2 sublineage) and the purified rHA1 was chemically coupled with bacteriophage using a crosslinker to produce rHA1-phage vaccine candidate. The protective efficacy of vaccine preparations of rHA1-phage conjugate and only rHA1 proteins were evaluated in mouse model through assessing serology, cytokine profiling, clinical signs, gross and histopathological changes, immunohistochemistry, and virus quantification. Immunization of vaccine preparations have stimulated moderate antibody response (ELISA titres-5760 ± 640 and 11,520 ± 1280 for rHA1 and rHA1-phage, respectively at 42 dpi) and elicited strong interferon (IFN)-γ expression levels after three immunizations of vaccine candidates. The immunized BALB/c mice were protected against challenge with wild EIV and resulted in reduced clinical signs and body weight loss, reduced pathological changes, decreased EIV antigen distribution, and restricted EIV replication in lungs and nasopharynx. In conclusion, the immune responses with moderate antibody titer and significantly higher cytokine responses generated by the rHA1-phage vaccine preparation without any adjuvant could be a novel vaccine candidate for quick vaccine preparation through further trials of vaccine in the natural host.

2.
Acta Virol ; 67(1): 79-90, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36950888

RESUMO

Equine herpesvirus 1 (EHV1) infection is a global health problem in equines and the virus is responsible for abortions, respiratory disease and myeloencephalitis in horses. Disease management requires proper biosecurity and immunoprophylactic measures. Vaccines strengthening both arms of immunity are essential for proper control and there has been a continuous focus in this area for generation of better vaccines. Here we report construction of bacterial artificial chromosome (BAC) clone of EHV-1 strain Tohana for mutagenesis of the virus and generation of gE gene deletion mutant EHV1. The BAC clone was generated by inserting the mini-F plasmid replacing ORF71 of EHV1 and transforming into E. coli for generation of EHV1-BAC. The infectious virus was regenerated from EHV-1 BAC DNA in RK13 cells. To check utility of EHV1-BAC, we have generated mutant EHV1 by deleting the virulence-associated gE gene. The mutant virus (vToHΔgE) showed significantly reduced plaque size without affecting replication efficiency. Pathological evaluation of lesions in BALB/c mice infected with vToHΔgE revealed reduction in clinical signs and pathology in comparison to the wild-type virus. Generation of infectious BAC of EHV1 and its usage in construction of attenuated viruses shows potential of the technology for development of indigenous modified live vaccine for EHV1. Keywords: quine herpesvirus 1; bacterial artificial chromosome (BAC); mutation; glycoprotein E; vaccine.


Assuntos
Infecções por Herpesviridae , Herpesvirus Equídeo 1 , Doenças dos Cavalos , Gravidez , Feminino , Animais , Cavalos , Camundongos , Herpesvirus Equídeo 1/genética , Escherichia coli/genética , Modelos Animais de Doenças , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/genética , Doenças dos Cavalos/prevenção & controle , Deleção de Genes
3.
Trop Anim Health Prod ; 54(4): 240, 2022 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-35869164

RESUMO

High cytotoxicity and increasing resistance reports of existing chemotherapeutic agents against T. evansi have raised the demand for novel, potent, and high therapeutic index molecules for the treatment of surra in animals. In this regard, repurposing approach of drug discovery has provided an opportunity to explore the therapeutic potential of existing drugs against new organism. With this objective, the macrocyclic lactone representative, ivermectin, has been investigated for the efficacy against T. evansi in the axenic culture medium. To elucidate the potential target of ivermectin in T. evansi, mRNA expression profile of 13 important drug target genes has been studied at 12, 24, and 48 h interval. In the in vitro growth inhibition assay, ivermectin inhibited T. evansi growth and multiplication significantly (p < 0.001) with IC50 values of 13.82 µM, indicating potent trypanocidal activity. Cytotoxicity assays on equine peripheral blood mononuclear cells (PBMCs) and Vero cell line showed that ivermectin affected the viability of cells with a half-maximal cytotoxic concentration (CC50) at 17.48 and 22.05 µM, respectively. Data generated showed there was significant down-regulation of hexokinase (p < 0.001), ESAG8 (p < 0.001), aurora kinase (p < 0.001), casein kinase 1 (p < 0.001), topoisomerase II (p < 0.001), calcium ATPase 1 (p < 0.001), ribonucleotide reductase I (p < 0.05), and ornithine decarboxylase (p < 0.01). The mRNA expression of oligopeptidase B remains refractory to the exposure of the ivermectin. The arginine kinase 1 and ribonucleotide reductase II showed up-regulation on treatment with ivermectin. The ivermectin was found to affect glycolytic pathways, ATP-dependent calcium ATPase, cellular kinases, and other pathway involved in proliferation and maintenance of internal homeostasis of T. evansi. These data imply that intervention with alternate strategies like nano-formulation, nano-carriers, and nano-delivery or identification of ivermectin homologs with low cytotoxicity and high bioavailability can be explored in the future as an alternate treatment for surra in animals.


Assuntos
Doenças dos Cavalos , Ribonucleotídeo Redutases , Trypanosoma , Tripanossomíase , Animais , Cavalos , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Leucócitos Mononucleares/metabolismo , Redes e Vias Metabólicas , RNA Mensageiro/metabolismo , Ribonucleotídeo Redutases/metabolismo , Ribonucleotídeo Redutases/farmacologia , Tripanossomíase/tratamento farmacológico , Tripanossomíase/veterinária
4.
Transbound Emerg Dis ; 67(3): 1062-1067, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31880100

RESUMO

Porcine circovirus type 3 (PCV3), a novel circovirus, has been reported recently from major swine growing countries globally, and the virus is associated with diseases like porcine dermatitis, nephropathy syndrome and reproductive failure. This report describes the identification of PCV3 associated with reproductive failure in sows and piglet mortality and circulation of the virus in healthy pigs in India. The pathological changes in various tissues from stillborn piglet and characterization of the virus genomes were reported. The genome sequences of Indian PCV3 strains showed 91.4%-99.8% nucleotide identity with other sequences of PCV3 strains circulating worldwide. The phylogenetic analysis showed clustering of Indian strains into a separate group with the isolate from USA (MN/2016) under PCV3a genotype. The results confirmed the circulation of PCV3 in Indian pigs and its association with clinical cases. This study speculates emergence of PCV3 as an important pig pathogen in the country, which warrants the thorough investigation on PCV3 epidemiology, pathogenesis and to implement the control measures.


Assuntos
Infecções por Circoviridae/veterinária , Circovirus/genética , Genoma Viral/genética , Reprodução , Doenças dos Suínos/virologia , Animais , Infecções por Circoviridae/epidemiologia , Infecções por Circoviridae/mortalidade , Infecções por Circoviridae/virologia , Circovirus/isolamento & purificação , Feminino , Genótipo , Índia/epidemiologia , Filogenia , Natimorto/veterinária , Suínos , Doenças dos Suínos/epidemiologia , Doenças dos Suínos/mortalidade
5.
J Vet Med Sci ; 81(12): 1753-1762, 2019 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-31656240

RESUMO

Equine influenza is a leading cause for respiratory illness in equines. Major control measures involve vaccination which requires continuous harmonization owing to antigenic drift. The present study focused on assessing the protective efficacy of an inactivated recombinant equine influenza virus (rgEIV) vaccine candidate adjuvanted with MontanideTM Pet Gel in murine model. The rgEIV was generated using reverse genetics by incorporating HA and NA segments from EIV/H3N8, clade 2-Florida sublineage in an A/WSN/33 /H1N1 backbone and inactivated by formalin. The vaccine was prepared by mixing inactivated rgEIV with MontanideTM Pet Gel adjuvant followed by intranasal inoculation into BALB/c mice intranasally. The immune responses and protective efficacy of the vaccine was evaluated by measurement of antibody titer, immunoglobulin subtyping, cytokines, clinical signs and pathological lesions after immunization and challenge with wild EIV. Serology and cytokine expression pattern indicated that the vaccine activated mixed Th1- and Th2-like responses of vaccine. Booster immunization stimulated strong antibody responses (HAI titre: 192 ± 28.6) at 42 days post immunization and the predominant antibody subtype was IgG1. Upregulation of interferon (IFN)-gamma, interleukin (IL)-12 and IL-2 levels indicates effective induction of Th1 type response. We found that vaccination has protected mice against equine influenza virus challenge as adjudged through a lack of nonappearance of visible clinical signs of disease, no loss of body weight loss, reduced pathology in the lungs and markedly reduced virus shedding from the respiratory tract. Therefore, we conclude that recombinant EIV vaccine candidate adjuvanted with MontanideTM Pet Gel could aid in quick harmonization of the vaccines through replacement of HA and NA genes for control of EIV outbreaks.


Assuntos
Vírus da Influenza A Subtipo H3N8/imunologia , Vacinas contra Influenza/imunologia , Adjuvantes Imunológicos , Animais , Citocinas/genética , Feminino , Géis , Imunidade Humoral/imunologia , Imunização Secundária/veterinária , Isotipos de Imunoglobulinas/classificação , Pulmão/patologia , Manitol/análogos & derivados , Manitol/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ácidos Oleicos/imunologia , RNA Mensageiro/análise , Traqueia/patologia , Conchas Nasais/patologia , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas/imunologia
6.
J Equine Sci ; 29(1): 25-31, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29593446

RESUMO

Bordetella bronchiseptica is a well-known Gram-negative bacterial pathogen causing a plethora of diseases in different animals. Although its infection has been reported from pigs and dogs in India, no report of B. bronchiseptica from horses is described. We report for the first time, isolation, identification and characterization of strains of B. bronchiseptica from respiratory infection in horses from different states in India. The antimicrobial susceptibility testing showed resistance to penicillins, ceftazidime, and chloramphanicol. The virulence capability of the strains was confirmed by sequencing genes such as adenylate cyclase toxin (cyaA), bordetella virulence gene (bvgA) and by PCR detection of flagellin gene (fla). We demonstrate the involvement of B. bronchiseptica strains in respiratory tract infection in horses in India.

7.
Vet Microbiol ; 210: 188-196, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29103691

RESUMO

Equine influenza viruses (EIVs) are responsible for acute contagious respiratory infection in equines and the disease remains a major threat for equine population throughout the world despite vaccination strategies in place. The present study was aimed to assess the suitability of BALB/c mice as a potential small animal model for preliminary screening of EI vaccine candidates. For this, we evaluated the immunogenicity and protective efficacy of an inactivated EIV (H3N8) vaccine in BALB/c mouse model after challenge with homologous H3N8 virus (Clade 2 virus, Florida sublineage) through serology, clinical signs, gross and histopathology lesions with grading, immunohistochemistry and virus quantification. Serological responses in immunized mice were evaluated by haemagglutination inhibition assay (HAI) and antibodies were subtyped by ELISA. The vaccine induced optimum protective antibody titre on 49 dpi along with balanced Th1/Th2 responses. Immunized mice were well protected against EIV challenge as evident by significant rise in serum antibody titre which concurred with mild clinical signs, early recovery, lower gross and histopathological lesions score, less severe intensity of viral antigen distribution, restricted virus replication in respiratory tract and less virus detection in nasal washes for short duration. The duration of the viral load was also lower and only for brief period as compared to unvaccinated challenged mice. In conclusion, induction of H3N8 specific antibody response and protection against H3N8 challenge proves that egg grown inactivated H3N8 whole virus vaccine would provide an effective intercession against H3N8 virus. In addition, BALB/c mouse can serve as an attractive tool for adjudging protective efficacy of vaccine candidates prior to final testing in equines.


Assuntos
Anticorpos Antivirais/sangue , Doenças dos Cavalos/prevenção & controle , Vírus da Influenza A Subtipo H3N8/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Vacinação/veterinária , Animais , Formação de Anticorpos , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/veterinária , Feminino , Doenças dos Cavalos/virologia , Cavalos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/virologia , Vacinas de Produtos Inativados/imunologia , Carga Viral
8.
Artigo em Inglês | MEDLINE | ID: mdl-27260812

RESUMO

Swinepox virus (SWPV), a member of the genus Suipoxvirus causes generalized pock-like lesions on the body of domestic and wild pigs. Although outbreak has been reported in India since 1987, virus isolation and genetic characterization remained elusive. In September 2013, an outbreak of acute skin infection occurred in piglets in a commercial piggery unit at Rohtak district in Haryana, India. The presence of SWPV in scab samples collected from piglets succumbed to infection was confirmed by virus isolation, PCR amplification of SWPV-specific gene segments and nucleotide sequencing. Phylogenetic analysis of host-range genes of the SWPV revealed that the Indian isolate is genetically closely related to reference isolate SWPV/pig/U.S.A/1999/Nebraska. To the best of our knowledge this is the first report on isolation and genetic characterization of SWPV from pigs in India.


Assuntos
Especificidade de Hospedeiro , Infecções por Poxviridae/veterinária , Suipoxvirus/genética , Suipoxvirus/isolamento & purificação , Doenças dos Suínos/virologia , Animais , Repetição de Anquirina/genética , Sequência de Bases , Surtos de Doenças , Índia/epidemiologia , Filogenia , Infecções por Poxviridae/epidemiologia , Infecções por Poxviridae/virologia , Suínos/virologia
9.
PLoS One ; 11(4): e0154376, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27119730

RESUMO

Equine influenza viruses (EIVs) of H3N8 subtype are culprits of severe acute respiratory infections in horses, and are still responsible for significant outbreaks worldwide. Adaptability of influenza viruses to a particular host is significantly influenced by their codon usage preference, due to an absolute dependence on the host cellular machinery for their replication. In the present study, we analyzed genome-wide codon usage patterns in 92 EIV strains, including both H3N8 and H7N7 subtypes by computing several codon usage indices and applying multivariate statistical methods. Relative synonymous codon usage (RSCU) analysis disclosed bias of preferred synonymous codons towards A/U-ended codons. The overall codon usage bias in EIVs was slightly lower, and mainly affected by the nucleotide compositional constraints as inferred from the RSCU and effective number of codon (ENc) analysis. Our data suggested that codon usage pattern in EIVs is governed by the interplay of mutation pressure, natural selection from its hosts and undefined factors. The H7N7 subtype was found less fit to its host (horse) in comparison to H3N8, by possessing higher codon bias, lower mutation pressure and much less adaptation to tRNA pool of equine cells. To the best of our knowledge, this is the first report describing the codon usage analysis of the complete genomes of EIVs. The outcome of our study is likely to enhance our understanding of factors involved in viral adaptation, evolution, and fitness towards their hosts.


Assuntos
Códon , Regulação Viral da Expressão Gênica , Genoma Viral , Doenças dos Cavalos/virologia , Vírus da Influenza A Subtipo H3N8/genética , Vírus da Influenza A Subtipo H7N7/genética , Infecções por Orthomyxoviridae/veterinária , Adaptação Fisiológica/genética , Animais , Evolução Biológica , Código Genético , Cavalos , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H3N8/metabolismo , Vírus da Influenza A Subtipo H7N7/metabolismo , Modelos Estatísticos , Taxa de Mutação , Infecções por Orthomyxoviridae/virologia , RNA de Transferência/genética , RNA de Transferência/metabolismo , Especificidade da Espécie , Replicação Viral
10.
PLoS One ; 10(11): e0143094, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26587990

RESUMO

Equine influenza viruses (EIV)-H3N8 continue to circulate in equine population throughout the world. They evolve by the process of antigenic drift that leads to substantial change in the antigenicity of the virus, thereby necessitating substitution of virus strain in the vaccines. This requires frequent testing of the new vaccines in the in vivo system; however, lack of an appropriate laboratory animal challenge model for testing protective efficacy of equine influenza vaccine candidates hinders the screening of new vaccines and other therapeutic approaches. In the present investigation, BALB/c mouse were explored for suitability for conducting pathogenecity studies for EIV. The BALB/c mice were inoculated intranasally @ 2×106.24 EID50 with EIV (H3N8) belonging to Clade 2 of Florida sublineage and monitored for setting up of infection and associated parameters. All mice inoculated with EIV exhibited clinical signs viz. loss in body weights, lethargy, dyspnea, etc, between 3 and 5 days which commensurate with lesions observed in the respiratory tract including rhinitis, tracheitis, bronchitis, bronchiolitis, alveolitis and diffuse interstitial pneumonia. Transmission electron microscopy, immunohistochemistry, virus quantification through titration and qRT-PCR demonstrated active viral infection in the upper and lower respiratory tract. Serology revealed rise in serum lactate dehydrogenase levels along with sero-conversion. The pattern of disease progression, pathological lesions and virus recovery from nasal washings and lungs in the present investigations in mice were comparable to natural and experimental EIV infection in equines. The findings establish BALB/c mice as small animal model for studying EIV (H3N8) infection and will have immense potential for dissecting viral pathogenesis, vaccine efficacy studies, preliminary screening of vaccine candidates and antiviral therapeutics against EIV.


Assuntos
Doenças dos Cavalos/virologia , Vírus da Influenza A Subtipo H3N8/patogenicidade , Infecções por Orthomyxoviridae/virologia , Animais , Modelos Animais de Doenças , Doenças dos Cavalos/patologia , Cavalos/virologia , Vírus da Influenza A Subtipo H3N8/imunologia , Camundongos , Infecções por Orthomyxoviridae/patologia
11.
Int J Dermatol ; 52(10): 1224-30, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23834521

RESUMO

BACKGROUND: Despite repeated outbreaks of poxvirus infections unique to the Indian subcontinent region and veterinary research work in this field, much less diagnostic awareness with resultant treatment protocols have been formulated in the human medical field. AIMS: With this objective in mind, a combined human medical and veterinary study was conducted on a recent outbreak of buffalopox infection in a village in northern India. METHODS: A team of doctors did the clinical examination and collected swab and serum samples from both cattle and humans, and these were subjected to viral isolation, cell culture, plaque reduction neutralization test, polymerase chain reaction, and partial genome sequencing. RESULTS: A clustered foci of 12 human patients aged 11-60 years, 12 buffaloes, and 10 cows were found to be affected with buffalopox infection with some atypical features. CONCLUSION: Awareness, diagnosis, education, early intervention, and formulation of disaster guidelines are needed in view of the potential epidemiologic outbreak, if this happens in the future.


Assuntos
Doenças dos Bovinos/diagnóstico , Doenças dos Bovinos/epidemiologia , Surtos de Doenças/estatística & dados numéricos , Vírus Vaccinia/isolamento & purificação , Vaccinia , Adolescente , Adulto , Animais , Búfalos , Bovinos , Criança , Chlorocebus aethiops , Feminino , Dermatoses da Mão/diagnóstico , Dermatoses da Mão/epidemiologia , Humanos , Índia/epidemiologia , Masculino , Glândulas Mamárias Animais , Pessoa de Meia-Idade , Filogenia , Vaccinia/diagnóstico , Vaccinia/epidemiologia , Vaccinia/veterinária , Vírus Vaccinia/genética , Vírus Vaccinia/crescimento & desenvolvimento , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...