Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Acta Trop ; 242: 106922, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37031926

RESUMO

Countries in the Indian subcontinent are currently facing a deadly epidemic of lumpy skin disease (LSD).  LSD is primarily a disease of cattle. Buffaloes may sometimes develop mild illness, however, other domestic animals are considered resistant to LSD. We confirmed the LSDV infection in camels as evidenced by skin nodules on the body surface of the affected camels, isolation of LSD virus (LSDV) and amplification of LSDV-specific gene segments from the skin nodules (PCR), nucleotide sequencing of the viral genome and, demonstration of anti-LSDV antibodies in serum. Phylogenetic analysis based on nucleotide sequencing of ORF011, ORF012 and ORF036 revealed that the virus (LSDV/Camel/India/2022/Bikaner) is related to the historical NI-2490/Kenya/KSGP-like field strains which are predominantly circulating in the Indian subcontinent. This is the first report wherein LSDV has been to infect camels.


Assuntos
Doença Nodular Cutânea , Vírus da Doença Nodular Cutânea , Animais , Bovinos , Vírus da Doença Nodular Cutânea/genética , Doença Nodular Cutânea/epidemiologia , Camelus , Filogenia , Búfalos , Nucleotídeos , Surtos de Doenças/veterinária
2.
Virulence ; 14(1): 2190647, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-36919498

RESUMO

Lumpy skin disease (LSD) was reported for the first time in India in 2019 and since then, it has become endemic. Since a homologous (LSD-virus based) vaccine was not available in the country, goatpox virus (GPV)-based heterologous vaccine was authorized for mass immunization to induce protection against LSD in cattle. This study describes the evaluation of safety, immunogenicity and efficacy of a new live-attenuated LSD vaccine developed by using an Indian field strain, isolated in 2019 from cattle. The virus was attenuated by continuous passage (P = 50) in Vero cells. The vaccine (50th LSDV passage in Vero cells, named as Lumpi-ProVacInd) did not induce any local or systemic reaction upon its experimental inoculation in calves (n = 10). At day 30 post-vaccination (pv), the vaccinated animals were shown to develop antibody- and cell-mediated immune responses and exhibited complete protection upon virulent LSDV challenge. A minimum Neethling response (0.018% animals; 5 out of 26,940 animals) of the vaccine was observed in the field trials conducted in 26,940 animals. There was no significant reduction in the milk yield in lactating animals (n = 10108), besides there was no abortion or any other reproductive disorder in the pregnant animals (n = 2889). Sero-conversion was observed in 85.18% animals in the field by day 30 pv.


Assuntos
Doença Nodular Cutânea , Vírus da Doença Nodular Cutânea , Vacinas Virais , Animais , Bovinos , Feminino , Chlorocebus aethiops , Doença Nodular Cutânea/prevenção & controle , Doença Nodular Cutânea/epidemiologia , Vírus da Doença Nodular Cutânea/genética , Vacinas Atenuadas/efeitos adversos , Células Vero , Vacinas Virais/administração & dosagem
3.
Antiviral Res ; 189: 105056, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33711336

RESUMO

Emetine is a FDA-approved drug for the treatment of amebiasis. Previously we demonstrated the antiviral efficacy of emetine against some RNA and DNA viruses. In this study, we evaluated the in vitro antiviral efficacy of emetine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and found it to be a low nanomolar (nM) inhibitor. Interestingly, emetine exhibited protective efficacy against lethal challenge with infectious bronchitis virus (IBV; a chicken coronavirus) in the embryonated chicken egg infection model. Emetine treatment led to a decrease in viral RNA and protein synthesis without affecting other steps of viral life cycle such as attachment, entry and budding. In a chromatin immunoprecipitation (CHIP) assay, emetine was shown to disrupt the binding of SARS-CoV-2 mRNA with eIF4E (eukaryotic translation initiation factor 4E, a cellular cap-binding protein required for initiation of protein translation). Further, molecular docking and molecular dynamics simulation studies suggested that emetine may bind to the cap-binding pocket of eIF4E, in a similar conformation as m7-GTP binds. Additionally, SARS-CoV-2 was shown to exploit ERK/MNK1/eIF4E signalling pathway for its effective replication in the target cells. Collectively our results suggest that further detailed evaluation of emetine as a potential treatment for COVID-19 may be warranted.


Assuntos
Antivirais , Emetina , Vírus da Bronquite Infecciosa/efeitos dos fármacos , RNA Viral/metabolismo , SARS-CoV-2/efeitos dos fármacos , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Embrião de Galinha , Chlorocebus aethiops , Infecções por Coronavirus/tratamento farmacológico , Emetina/farmacologia , Emetina/uso terapêutico , Fator de Iniciação 4E em Eucariotos/metabolismo , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transdução de Sinais , Células Vero
4.
PLoS One ; 16(1): e0241022, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33428633

RESUMO

Lumpy skin disease (LSD) has devastating economic impact. During the last decade, LSD had spread to climatically new and previously disease-free countries, which also includes its recent emergence in the Indian subcontinent (2019). This study deals with the LSD outbreak(s) from cattle in Ranchi (India). Virus was isolated from the scabs (skin lesions) in the primary goat kidney cells. Phylogenetic analysis based on nucleotide sequencing of LSD virus (LSDV) ORF011, ORF012 and ORF036 suggested that the isolated virus (LSDV/Bos taurus-tc/India/2019/Ranchi) is closely related to Kenyan LSDV strains. Further, we adapted the isolated virus in Vero cells. Infection of the isolated LSDV to Vero cells did not produce cytopathic effect (CPE) until the 4th blind passage, but upon adaptation, it produced high viral titres in the cultured cells. The kinetics of viral DNA synthesis and one-step growth curve analysis suggested that Vero cell-adapted LSDV initiates synthesizing its genome at ~24 hours post-infection (hpi) with a peak level at ~96 hpi whereas evidence of progeny virus particles was observed at 36-48 hours (h) with a peak titre at ~120 h. To the best of our knowledge, this study describes the first successful isolation of LSDV in India, besides providing insights into the life cycle Vero cell-adapted LSDV.


Assuntos
Genoma Viral , Doença Nodular Cutânea/genética , Vírus da Doença Nodular Cutânea/genética , Fases de Leitura Aberta , Filogenia , Animais , Bovinos , Chlorocebus aethiops , Surtos de Doenças , Índia/epidemiologia , Doença Nodular Cutânea/epidemiologia , Vírus da Doença Nodular Cutânea/isolamento & purificação , Vírus da Doença Nodular Cutânea/metabolismo , Células Vero
5.
Antiviral Res ; 181: 104870, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32707051

RESUMO

We describe herein that Apigenin, which is a dietary flavonoid, exerts a strong in vitro and in ovo antiviral efficacy against buffalopox virus (BPXV). Apigenin treatment was shown to inhibit synthesis of viral DNA, mRNA and proteins, without affecting other steps of viral life cycle such as attachment, entry and budding. Although the major mode of antiviral action of Apigenin was shown to be mediated via targeting certain cellular factors, a modest inhibitory effect of Apigenin was also observed directly on viral polymerase. We also evaluated the selection of drug-resistant virus variants under long-term selection pressure of Apigenin. Wherein Apigenin-resistant mutants were not observed up to ~ P20 (passage 20), a significant resistance was observed to the antiviral action of Apigenin at ~ P30. However, a high degree resistance could not be observed even up to P60. To the best of our knowledge, this is the first report describing in vitro and in ovo antiviral efficacy of Apigenin against poxvirus infection. The study also provides mechanistic insights on the antiviral activity of Apigenin and selection of potential Apigenin-resistant mutants upon long-term culture.


Assuntos
Antivirais/farmacologia , Apigenina/farmacologia , Farmacorresistência Viral , Vírus Vaccinia/efeitos dos fármacos , Animais , Embrião de Galinha/virologia , Galinhas , Chlorocebus aethiops , DNA Viral/genética , DNA Polimerase Dirigida por DNA , Humanos , Vírus Vaccinia/enzimologia , Células Vero , Replicação Viral/efeitos dos fármacos
6.
PLoS One ; 15(4): e0232093, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32330151

RESUMO

Bovine herpesvirus 1 (BoHV1) and 5 (BoHV5) are genetically and antigenically related alphaherpesviruses. Infection with one virus induces protective immunity against the other. However, disease associated with BoHV1 and BoHV5 varies significantly; whereas BoHV1 infection is usually associated with rhinotracheitis and abortion, BoHV5 causes encephalitis in cattle. BoHV5 outbreaks are sporadic and mainly restricted to the South American countries. We report BoHV5 infection for the first time from aborted cattle in India. Based on the characteristic cytopathic effects in MDBK cells, amplification of the viral genome by PCR, differential PCR for BoHV1/BoHV5, nucleotide sequencing and restriction endonuclease patterns, identity of the virus was confirmed as BoHV5 subtype A. Serum samples from the aborted cattle strongly neutralized both BoHV1 and BoHV5 suggesting an active viral infection in the herd. Upon UL27, UL44 and UL54 gene-based sequence and phylogenetic analysis, the isolated virus clustered with BoHV5 strains and showed highest similarity with the Brazilian BoHV5 strains.


Assuntos
Herpesvirus Bovino 5/genética , Herpesvirus Bovino 5/isolamento & purificação , Herpesvirus Bovino 5/metabolismo , Alphaherpesvirinae/genética , Animais , Anticorpos Antivirais/sangue , Bovinos , Doenças dos Bovinos/epidemiologia , Surtos de Doenças/veterinária , Genoma Viral/genética , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/metabolismo , Índia , Filogenia
7.
Front Microbiol ; 10: 209, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30814986

RESUMO

Sarco/endoplasmic reticulum calcium-ATPase (SERCA) is a membrane-bound cytosolic enzyme which is known to regulate the uptake of calcium into the sarco/endoplasmic reticulum. Herein, we demonstrate for the first time that SERCA can also regulate virus replication. Treatment of Vero cells with SERCA-specific inhibitor (Thapsigargin) at a concentration that is nontoxic to the cells significantly reduced Peste des petits ruminants virus (PPRV) and Newcastle disease virus (NDV) replication. Conversely, overexpression of SERCA rescued the inhibitory effect of Thapsigargin on virus replication. PPRV and NDV infection induced SERCA expression in Vero cells, which could be blocked by Thapsigargin. Besides inducing enhanced formation of cytoplasmic foci, Thapsigargin was shown to block viral entry into the target cells as well as synthesis of viral proteins. Furthermore, NDV was shown to acquire significant resistance to Thapsigargin upon long-term passage (P) in Vero cells. As compared to the P0 and P70-Control, the fusion (F) protein of P70-Thapsigargin virus exhibited a unique mutation at amino acid residue 104 (E104K), whereas no Thapsigargin-associated mutations were observed in HN gene. To the best of our knowledge, this is the first report describing the virus-supportive role of SERCA and a rare report suggesting that viruses may acquire resistance even in the presence of an inhibitor that targets a cellular factor.

8.
Virusdisease ; 30(4): 551-561, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31897418

RESUMO

Chicken astroviruses (CAstVs) infect young chicks and are associated with gastroenteritis, stunted growth or visceral gout (gout). True incidence and distribution of CAstVs as well as virus variants circulating in India is not well understood. In this study, 80 gout-affected broiler chicken flocks from Haryana, a north-western state of India, were tested for the presence of astroviruses by targeting the polymerase gene of both CAstV and avian nephritis virus (ANV) and capsid gene of CAstV. Of these, 22 (27.5%) flocks were found positive for CAstV, 7(8.75%) for ANV and 2 (2.5%) for both CAstV and ANV genome by reverse-transcription-polymerase chain reaction. CAstV was isolated by inoculating tissue (kidney) homogenate from gout-affected birds into specific-pathogen free embryonated chicken eggs where the infected embryos showed stunted growth with necrosis of liver and enlarged kidney with urate deposits. Capsid gene-based phylogenetic analysis revealed the clustering of CAstV strains from this study with Indian strains of serogroup Biii suggesting their antigenic relatedness. Thus the present study reveals the presence of chicken astroviruses in broiler chickens affected with gout.

9.
Antiviral Res ; 160: 126-136, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30393013

RESUMO

A small molecule chemical inhibitor CGP57380 that blocks activation of MAPK interacting kinase 1 (MNK1) was found to significantly suppress buffalopox virus (BPXV) replication. BPXV infection was shown to induce MNK1 activation. Depletion of MNK1 by small interfering RNA (siRNA), blocking activation of extracellular regulated kinase (ERK, an upstream activator of MNK1) and disruption of eIF4E/eIF4G interaction (downstream substrate of MNK1 which plays a central role in cap-dependent translation initiation), resulted in reduced BPXV replication, suggesting that ERK/MNK1/eIF4E signaling is a prerequisite for BPXV replication. With the help of time-of-addition and virus step-specific assays, CGP57380 treatment was shown to decrease synthesis of viral genome (DNA). Disruption of ERK/MNK1/eIF4E signaling resulted in reduced synthesis of viral proteins, suggesting that BPXV utilizes cap-dependent mechanism of translation initiation. Therefore, we concluded that decreased synthesis of viral genome in presence of MNK1 inhibitor is the result of reduced synthesis of viral proteins. Furthermore, BPXV was sequentially passaged (P = 40) in presence of CGP57380 or vehicle control (DMSO). As compared to P0 and P40-control viruses, P40-CGP57380 virus replicated at significantly higher (∼10-fold) titers in presence of CGP57380, although a complete resistance could not be achieved. In a BPXV egg infection model, CGP57380 was found to prevent development of pock lesions on chorioallantoic membrane (CAM) as well as associated mortality of the embryonated chicken eggs. We for the first time demonstrated in vitro and in ovo antiviral efficacy of CGP57380 against BPXV and identified that ERK/MNK1 signaling is a prerequisite for synthesis of viral proteins. Our study also describes a rare report about generation of drug-resistant viral variants against a host-targeting antiviral agent.


Assuntos
Compostos de Anilina/farmacologia , Antivirais/farmacologia , DNA Viral/biossíntese , Inibidores de Proteases/farmacologia , Purinas/farmacologia , Vírus Vaccinia/efeitos dos fármacos , Proteínas Virais/biossíntese , Replicação Viral/efeitos dos fármacos , Animais , Embrião de Galinha , Galinhas , Membrana Corioalantoide/patologia , Farmacorresistência Viral , Óvulo/virologia , Inoculações Seriadas , Vírus Vaccinia/fisiologia , Carga Viral
10.
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.

11.
Antiviral Res ; 144: 196-204, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28624461

RESUMO

At a noncytotoxic concentration, emetine was found to inhibit replication of DNA viruses [buffalopoxvirus (BPXV) and bovine herpesvirus 1 (BHV-1)] as well as RNA viruses [peste des petits ruminants virus (PPRV) and Newcastle disease virus (NDV)]. Using the time-of-addition and virus step-specific assays, we showed that emetine treatment resulted in reduced synthesis of viral RNA (PPRV and NDV) and DNA (BPXV and BHV-1) as well as inhibiting viral entry (NDV and BHV-1). In addition, emetine treatment also resulted in decreased synthesis of viral proteins. In a cell free endogenous viral polymerase assay, emetine was found to significantly inhibit replication of NDV, but not BPXV genome, suggesting that besides directly inhibiting specific viral polymerases, emetine may also target other factors essentially required for efficient replication of the viral genome. Moreover, emetine was found to significantly inhibit BPXV-induced pock lesions on chorioallantoic membrane (CAM) along with associated mortality of embryonated chicken eggs. At a lethal dose 50 (LD50) of 126.49 ng/egg and at an effective concentration 50 (EC50) of 3.03 ng/egg, the therapeutic index of the emetine against BPXV was determined to be 41.74. Emetine was also found to significantly delay NDV-induced mortality in chicken embryos associated with reduced viral titers. Further, emetine-resistant mutants were not observed upon long-term (P = 25) sequential passage of BPXV and NDV in cell culture. Collectively, we have extended the effective antiviral activity of emetine against diverse groups of DNA and RNA viruses and propose that emetine could provide significant therapeutic value against some of these viruses without inducing an antiviral drug-resistant phenotype.


Assuntos
Antivirais/farmacologia , Vírus de DNA/efeitos dos fármacos , Emetina/farmacologia , Vírus de RNA/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Bovinos , Embrião de Galinha , Chlorocebus aethiops , Vírus de DNA/fisiologia , Farmacorresistência Viral , Vírus de RNA/fisiologia
12.
Vet Microbiol ; 206: 91-101, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28161212

RESUMO

Peste des petits ruminants (PPR) is a highly contagious disease of small ruminants that leads to high morbidity and mortality thereby results in devastating economic consequences to the livestock industry. PPR is currently endemic across most parts of Asia and Africa, the two regions with the highest concentration of poor people in the world. Sheep and goats in particularly contribute significantly towards the upliftment of livelihood of the poor and marginal farmers in these regions. In this context, PPR directly affecting the viability of sheep and goat husbandry has emerged as a major hurdle in the development of these regions. The control of PPR in these regions could significantly contribute to poverty alleviation, therefore, the Office International des Epizooties (OIE) and Food and Agricultural Organization (FAO) have targeted the control and eradication of PPR by 2030 a priority. In order to achieve this goal, a potent, safe and efficacious live-attenuated PPR vaccine with long-lasting immunity is available for immunoprophylaxis. However, the live-attenuated PPR vaccine is thermolabile and needs maintenance of an effective cold chain to deliver into the field. In addition, the infected animals cannot be differentiated from vaccinated animals. To overcome these limitations, some recombinant vaccines have been developed. This review comprehensively describes about the latest developments in PPR vaccines.


Assuntos
Doenças das Cabras/prevenção & controle , Peste dos Pequenos Ruminantes/prevenção & controle , Vírus da Peste dos Pequenos Ruminantes/imunologia , Doenças dos Ovinos/prevenção & controle , Vacinas Virais/imunologia , Animais , Doenças das Cabras/epidemiologia , Doenças das Cabras/virologia , Cabras , Peste dos Pequenos Ruminantes/epidemiologia , Peste dos Pequenos Ruminantes/virologia , Ovinos , Doenças dos Ovinos/epidemiologia , Doenças dos Ovinos/virologia , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/imunologia
13.
Arch Virol ; 162(1): 157-163, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27699511

RESUMO

Equine herpesvirus 1 (EHV1) is a common pathogen of horses that causes upper respiratory tract disease, abortion, neonatal death and neurological disease. The neurological form of disease is called equine herpesvirus myeloencephalopathy (EHM). During the past decade, the incidence of EHM has been on the rise in Europe, North America, Australia and Asia. Some EHV1 isolates causing EHM exhibit a single-nucleotide polymorphism (SNP) in the DNA polymerase gene (ORF30) at position 2254 (A2254 to G2254). Further, based on polymorphism in the ORF68, EHV1 isolates have been classified into different groups. The aim of the present study was to estimate the genetic diversity of EHV1 and to determine the prevalence of the neuropathogenic genotype of EHV1 in India. Out of 133 clinical specimens from abortion cases in northern India, 56 were positive for EHV1 infection. Analysis of the A/G SNP by real-time PCR and sequence analysis revealed that 54 of 56 samples (96.43 %) were of the non-neuropathogenic genotype (A2254), while two (3.57 %) had the neuropathogenic marker (G2254). Sequence analysis of the polymorphic region of ORF68 of EHV1 isolates (n = 9) from India indicated that the Delhi/1998, Tohana-2/2013, Hisar-2/2014 and Hisar-15/1990 isolates belonged to group 4, while the Jind/1996, Rajasthan/1998, Delhi-3/2007 and Tohana-5/1996 isolates clustered within group 5. One isolate (Hisar-7/1990) exhibited SNPs at positions C710 and C713, forming a separate group. Here, we report for the first time the detection of neuropathogenic genotypes of EHV1 in India and show that Indian EHV1 isolates cluster within groups 4 and 5.


Assuntos
Aborto Animal/epidemiologia , Surtos de Doenças , Encefalomielite/veterinária , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/isolamento & purificação , Doenças dos Cavalos/epidemiologia , Aborto Animal/virologia , Animais , Análise por Conglomerados , Encefalomielite/complicações , Encefalomielite/epidemiologia , Variação Genética , Genótipo , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/epidemiologia , Herpesvirus Equídeo 1/classificação , Herpesvirus Equídeo 1/genética , Doenças dos Cavalos/virologia , Cavalos , Índia/epidemiologia , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único , Prevalência , Análise de Sequência de DNA
14.
J Gen Virol ; 97(12): 3458-3466, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27902329

RESUMO

The ecosystem is continuously exposed to a wide variety of antimicrobials through waste effluents, agricultural run-offs and animal-related and anthropogenic activities, which contribute to the spread of antibiotic resistance genes (ARGs). The contamination of ecosystems with ARGs may create increased opportunities for their transfer to naive microbes and eventually lead to entry into the human food chain. Transduction is a significant mechanism of horizontal gene transfer in natural environments, which has traditionally been underestimated as compared to transformation. We explored the presence of ARGs in environmental bacteriophages in order to recognize their contribution in the spread of ARGs in environmental settings. Bacteriophages were isolated against environmental bacterial isolates, purified and bulk cultured. They were characterized, and detection of ARG and intI genes including blaTEM, blaOXA-2, intI1, intI2, intI3, tetA and tetW was carried out by PCR. This study revealed the presence of various genes [tetA (12.7 %), intI1 (10.9 %), intI2 (10.9 %), intI3 (9.1 %), tetW (9.1 %) and blaOXA-2 (3.6 %)] and blaTEM in a significantly higher proportion (30.9 %). blaSHV, blaOXA-1, tetO, tetB, tetG, tetM and tetS were not detected in any of the phages. Soil phages were the most versatile in terms of ARG carriage. Also, the relative abundance of tetA differed significantly vis-à-vis source. The phages from organized farms showed varied ARGs as compared to the unorganized sector, although blaTEM ARG incidences did not differ significantly. The study reflects on the role of phages in dissemination of ARGs in environmental reservoirs, which may provide an early warning system for future clinically relevant resistance mechanisms.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/virologia , Bacteriófagos/genética , Farmacorresistência Bacteriana , Proteínas Virais/genética , Bactérias/genética , Bactérias/metabolismo , Bacteriófagos/isolamento & purificação , Bacteriófagos/metabolismo , Microbiologia Ambiental , Transferência Genética Horizontal , Proteínas Virais/metabolismo
15.
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
16.
PLoS One ; 11(5): e0156110, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27227480

RESUMO

Successful purification of multiple viruses from mixed infections remains a challenge. In this study, we investigated peste des petits ruminants virus (PPRV) and foot-and-mouth disease virus (FMDV) mixed infection in goats. Rather than in a single cell type, cytopathic effect (CPE) of the virus was observed in cocultured Vero/BHK-21 cells at 6th blind passage (BP). PPRV, but not FMDV could be purified from the virus mixture by plaque assay. Viral RNA (mixture) transfection in BHK-21 cells produced FMDV but not PPRV virions, a strategy which we have successfully employed for the first time to eliminate the negative-stranded RNA virus from the virus mixture. FMDV phenotypes, such as replication competent but noncytolytic, cytolytic but defective in plaque formation and, cytolytic but defective in both plaque formation and standard FMDV genome were observed respectively, at passage level BP8, BP15 and BP19 and hence complicated virus isolation in the cell culture system. Mixed infection was not found to induce any significant antigenic and genetic diversity in both PPRV and FMDV. Further, we for the first time demonstrated the viral interference between PPRV and FMDV. Prior transfection of PPRV RNA, but not Newcastle disease virus (NDV) and rotavirus RNA resulted in reduced FMDV replication in BHK-21 cells suggesting that the PPRV RNA-induced interference was specifically directed against FMDV. On long-term coinfection of some acute pathogenic viruses (all possible combinations of PPRV, FMDV, NDV and buffalopox virus) in Vero cells, in most cases, one of the coinfecting viruses was excluded at passage level 5 suggesting that the long-term coinfection may modify viral persistence. To the best of our knowledge, this is the first documented evidence describing a natural mixed infection of FMDV and PPRV. The study not only provides simple and reliable methodologies for isolation and purification of two epidemiologically and economically important groups of viruses, but could also help in establishing better guidelines for trading animals that could transmit further infections and epidemics in disease free nations.


Assuntos
Vírus da Febre Aftosa/isolamento & purificação , Febre Aftosa/epidemiologia , Doenças das Cabras/epidemiologia , Cabras/virologia , Peste dos Pequenos Ruminantes/epidemiologia , Vírus da Peste dos Pequenos Ruminantes/isolamento & purificação , Animais , Chlorocebus aethiops , Efeito Citopatogênico Viral , Febre Aftosa/virologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Doenças das Cabras/virologia , Cabras/genética , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/crescimento & desenvolvimento , RNA Viral/genética
17.
Genome Announc ; 2(4)2014 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-25081265

RESUMO

The Pasteurella multocida subsp. multocida B:2 serotype causes hemorrhagic septicemia in bubalines with high morbidity and mortality in the Indian subcontinent. We report the draft genome sequence of Pasteurella multocida strain VTCCBAA264 isolated from the small-intestine of a buffalo calf that died of high fever.

19.
Vet Ital ; 48(2): 167-78, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22718333

RESUMO

Glanders, a bacterial disease of equines caused by Burkholderia mallei, is a fatal infectious disease of equines and has zoonotic significance. The disease has been eradicated from many countries by statutory testing, elimination of infected animals and import restrictions. However, it is still endemic in parts of Africa, Asia, the Middle East and Central and South America. In India, major glanders outbreaks were reported from different parts of the country between 1976 and 1982. Later, sporadic cases of the disease were reported in 1988, 1990 and 1998. The country remained free of glanders for about eight years until the recent outbreaks occurred in eight States from 2006 to 2007. Recurrent episodes have occurred in Himachal Pradesh and Uttar Pradesh, whereas fresh outbreaks occurred in Chhattisgarh from 2009 to 2010. A total of 164 equines were declared positive; a majority of the positive cases (n=77) were from Uttar Pradesh, followed by Maharashtra (n=23), Uttarakhand (n=21) and Andhra Pradesh (n=16). Under the provision of Prevention and Control of Infectious and Contagious Disease in Animals Act, 2009, all the infected animals were euthanised and bio-security measures were implemented to curb the further spread of the disease.


Assuntos
Surtos de Doenças , Mormo/epidemiologia , Animais , Mormo/diagnóstico , Cavalos , Índia/epidemiologia , Fatores de Tempo
20.
Virol Sin ; 26(5): 324-37, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21979572

RESUMO

In this study, thermo-adapted (Ta) PPR vaccines were assessed for their stability at 25, 37, 40, 42 and 45°C in lyophilized form using two extrinsic stabilizers {lactalbumin hydrolysate-sucrose (LS) and stabilizer E} and in reconstituted form with the diluents (1 mol/L MgSO(4) or 0.85% NaCl). The lyophilized vaccines showed an expiry period of 24-26 days at 25°C, 7-8 days at 37°C and 3-4 days at 40°C. LS stabilizer was superior at 42°C with a shelf-life of 44 h, whereas in stabilizer E, a 40 h shelf-life with a comparable half-life was observed. At 45°C, the half-life in stabilizer E was better than LS and lasted for 1 day. Furthermore, the reconstituted vaccine maintained the titre for 48 h both at 4°C and 25°C and for 24-30 h at 37°C. As both the stabilizers performed equally well with regard to shelf-life and half-life, the present study suggests LS as stabilizer as a choice for lyophilization with 0.85% NaCl diluent, because it has better performance at higher temperature. These Ta vaccines can be used as alternatives to existing vaccines for the control of the disease in tropical countries as they are effective in avoiding vaccination failure due to the breakdown in cold-chain maintenance, as this vaccine is considerably more stable at ambient temperatures.


Assuntos
Excipientes/farmacologia , Peste dos Pequenos Ruminantes/prevenção & controle , Vacinas Virais/imunologia , Animais , Estabilidade de Medicamentos , Armazenamento de Medicamentos , Excipientes/química , Liofilização , Temperatura , Vacinas Virais/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...