Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
Avian Dis ; 65(2): 227-236, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34412452

RESUMO

The history of pullorum disease is closely intertwined with the history of avian health research and that of the poultry industry. The seriousness of the disease galvanized the attention and brought together, for the first time, the pioneers of poultry health research to work cooperatively on different aspects of the disease. Control of the disease made it possible for intensive poultry production to develop as the basis for the modern poultry industry. During the early 1900s, bacillary white diarrhea (BWD) was a devastating disease of young chickens threatening the developing poultry industry. Dr. Leo F. Rettger isolated and described the bacterial pathogen, Salmonella enterica serotype Pullorum, for the first time in 1900. BWD was renamed pullorum disease in 1929. In subsequent years, Rettger and coworkers were able to reproduce the disease and fulfill Koch's postulates. Rettger et al. also showed that Salmonella Pullorum was vertically transmitted, which was the first time that a pathogen was shown to be vertically transmitted. The development of serologic tests was of crucial importance because it led to the development of effective eradication methods to identify carrier birds and to exclude these birds from the breeder flocks. The negative impact of pullorum disease on the poultry industry ultimately was one of the major reasons that the National Poultry Improvement Plan (NPIP) was developed by scientists, the poultry industry, and the United States Department of Agriculture (USDA). Needless to say, the work of the pioneering researchers formed the basis for the control of the disease. The NPIP started in 1935, with 34 states participating in testing 4 million birds representing 58.2% of the birds hatched. The program rapidly expanded to 47 states by 1948 and tested more than 30 million birds. In 1967, all commercial chicken hatcheries participating in the NPIP were 100% free of pullorum and typhoid disease caused by Salmonella enterica serotype Gallinarum. This historical overview of pullorum disease describes in some detail the progress made, especially during the early years, toward controlling this disease using methodologies that were often very basic but nonetheless effective. One has to admire the ingenuity and persistence of the early researchers leading to their achievements considering the research tools that were available at the time.


Artículo histórico­Pulorosis: Evolución de las estrategias de erradicación La historia de la pulorosis está estrechamente relacionada con la historia de la investigación en salud aviar y de la industria avícola. La severidad de la enfermedad despertó la atención y reunió, por primera vez a los pioneros de la investigación en salud avícola para trabajar de manera cooperativa en diferentes aspectos de la enfermedad. El control de la enfermedad hizo posible que la producción avícola intensiva se desarrollara como base de la industria avícola moderna. A principios de la década de los 1900, la diarrea blanca bacilar (con las siglas en inglés BWD) era una enfermedad devastadora de pollos jóvenes que amenazaba la industria avícola en desarrollo. El Dr. Leo F. Rettger aisló y describió el patógeno bacteriano, Salmonella enterica serotipo Pullorum, por primera vez en 1900. La diarrea blanca bacilar pasó a llamarse pulorosis (pullorum disease) en 1929. En los años siguientes, Rettger y sus colaboradores pudieron reproducir la enfermedad y cumplir los postulados de Koch. Rettger y col. también mostraron que Salmonella Pullorum se transmitía verticalmente, y fue la primera vez que se demostró que un patógeno se transmitía verticalmente. El desarrollo de pruebas serológicas fue de crucial importancia porque condujo al desarrollo de métodos de erradicación efectivos para identificar aves portadoras y eliminar a estas aves de las parvadas reproductoras. El impacto negativo de la pulorosis en la industria avícola fue, en última instancia, una de las principales razones por las que los científicos, la industria avícola y el Departamento de Agricultura de los Estados Unidos (USDA) desarrollaron el Plan Nacional de Mejoramiento Avícola (NPIP). Es importante decir que el trabajo de los investigadores pioneros formó la base para el control de la enfermedad. El Plan Nacional de Mejoramiento Avícola comenzó en año 1935, con 34 estados participando en el análisis de 4 millones de aves que representaban el 58.2% de las aves producidas. El programa se expandió rápidamente a 47 estados en 1948 y evaluó a más de 30 millones de aves. En 1967, todas las plantas incubadoras de pollos comerciales que participaban en el Plan Nacional de Mejoramiento Avícola estaban 100% libres de pulorosis y tifoidea aviar causada por Salmonella enterica serotipo Gallinarum. Esta reseña histórica de la pulorosis describe con cierto detalle el progreso realizado, especialmente durante los primeros años, hacia el control de esta enfermedad utilizando metodologías que a menudo eran muy básicas no obstante efectivas. Es admirable el ingenio y la persistencia de los primeros investigadores que los llevaron a sus logros considerando las herramientas de investigación que estaban disponibles en ese momento.


Assuntos
Galinhas , Doenças das Aves Domésticas/prevenção & controle , Salmonelose Animal/prevenção & controle , Salmonella/classificação , Fatores Etários , Animais , História do Século XX , Transmissão Vertical de Doenças Infecciosas/história , Transmissão Vertical de Doenças Infecciosas/veterinária , Doenças das Aves Domésticas/diagnóstico , Doenças das Aves Domésticas/história , Doenças das Aves Domésticas/transmissão , Salmonella/patogenicidade , Salmonelose Animal/história , Salmonelose Animal/microbiologia , Salmonelose Animal/transmissão
2.
Avian Dis ; 62(3): 272-285, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30339511

RESUMO

A healthy immune system is a cornerstone for poultry production. Any factor diminishing the immune responses will affect production parameters and increase cost. There are numerous factors, infectious and noninfectious, causing immunosuppression (IS) in chickens. This paper reviews the three viral diseases that most commonly induce IS or subclinical IS in chickens: Marek's disease virus (MDV), chicken infectious anemia virus (CIAV), and infectious bursal disease virus (IBDV), as well as the interactions among them. MDV-induced IS (MDV-IS) affects both humoral and cellular immune responses. It is very complex, poorly understood, and in many cases underdiagnosed. Vaccination protects against some but not all aspects of MDV-IS. CIAV induces apoptosis of the hemocytoblasts resulting in anemia, hemorrhages, and increased susceptibility to bacterial infections. It also causes apoptosis of thymocytes and dividing T lymphocytes, affecting T helper functions, which are essential for antibody production and cytotoxic T lymphocyte (CTL) functions. Control of CIAV is based on vaccination of breeders and maternal antibodies (MAbs). However, subclinical IS can occur after MAbs wane. IBDV infection affects the innate immune responses during virus replication and humoral immune responses as a consequence of the destruction of B-cell populations. Vaccines with various levels of attenuation are used to control IBDV. Interactions with MAbs and residual virulence of the vaccines need to be considered when designing vaccination plans. The interaction between IBDV, CIAV, and MDV is critical although underestimated in many cases. A proper control of IBDV is a must to have proper humoral immune responses needed to control CIAV. Equally, long-term control of MDV is not possible if chickens are coinfected with CIAV, as CIAV jeopardizes CTL functions critical for MDV control.


Assuntos
Vírus da Anemia da Galinha/imunologia , Galinhas , Herpesvirus Galináceo 2/imunologia , Terapia de Imunossupressão/veterinária , Vírus da Doença Infecciosa da Bursa/imunologia , Doenças das Aves Domésticas/imunologia , Vacinas Virais/imunologia , Animais , Infecções por Birnaviridae/imunologia , Infecções por Birnaviridae/veterinária , Infecções por Circoviridae/imunologia , Infecções por Circoviridae/veterinária , Imunidade Celular , Imunidade Humoral , Doença de Marek/imunologia , Vacinação/veterinária
3.
Avian Pathol ; 42(5): 464-73, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24015918

RESUMO

Several outbreaks of gout were reported in commercial broilers in India during 2011 and 2012, causing up to 40% mortality in the birds. Gross and histopathological observations confirmed gout. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) analysis from kidney samples of gout-affected birds indicated the presence of chicken astrovirus (CAstV) in 41.7% of cases and a mixed infection of CAstV and avian nephritis virus (ANV) in 36.4% of cases. CAstV isolated from gout-affected kidneys by inoculating embryonated specific pathogen free (SPF) eggs showed dwarfing in embryos and a cytopathic effect in chicken embryo kidney cells. Inoculation of 1-day-old SPF and broiler chicks with CAstVs caused gout and mortality between 4 and 10 days post inoculation. Virus isolation and qRT-PCR analysis showed the presence of only CAstV in inoculated chicks. Sequence analysis of capsid genes indicated a major group of Indian CAstVs that displayed 92.0 to 99.2% intergroup amino acid identity and 83.9 to 90.4% identity with subgroup Bi CAstVs of UK origin. We designated this group Indian Bi. Analysis of the partial polymerase amino acid sequences of our isolates indicated two groups of CAstVs (Indian 1 and 2) that displayed 90.2 to 95.5% amino acid identity between them. We thus report for the first time that, in addition to infectious bronchitis virus and ANV, CAstVs are a causative agent of gout.


Assuntos
Infecções por Astroviridae/veterinária , Avastrovirus/isolamento & purificação , Galinhas/virologia , Surtos de Doenças/veterinária , Gota/veterinária , Doenças das Aves Domésticas/epidemiologia , Animais , Infecções por Astroviridae/epidemiologia , Infecções por Astroviridae/patologia , Infecções por Astroviridae/virologia , Avastrovirus/genética , Avastrovirus/patogenicidade , Sequência de Bases , Proteínas do Capsídeo/genética , Embrião de Galinha , Primers do DNA/genética , Gota/epidemiologia , Gota/patologia , Gota/virologia , Índia/epidemiologia , Rim/patologia , Rim/virologia , Dados de Sequência Molecular , Filogenia , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , RNA Viral/química , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Análise de Sequência de RNA , Organismos Livres de Patógenos Específicos
4.
Vet Pathol ; 48(5): 985-92, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21311069

RESUMO

Systemic isosporosis, also known as atoxoplasmosis, is a common parasitic disease of passerines. Infection is thought to be endemic in wild birds with fulminant, fatal disease occurring under the influence of stress, concurrent infections, or immunosuppression. Here, we describe the histologic and immunohistochemical characteristics of the cellular infiltrate occurring in captive colonies of American goldfinches and house sparrows. Necropsies were performed on 9 birds, and histologic examination was performed on the intestines of 7 additional birds. Lesions were most severe in the proximal small intestines. Histologically, the changes ranged from variably intense infiltrates of lymphocytes that filled the lamina propria to sheets of large, atypical cells that expanded and obliterated normal mucosal epithelium and invaded through the wall of the intestine and into the ceolomic cavity. Both the smaller lymphocytes and large atypical cells were immunoreactive for CD3. Intracellular parasites consistent with Isospora were detected in the large atypical cells, but they were more easily detectable in the more differentiated lymphocytes. Polymerase chain reaction and virus isolation performed on tissues from 7 birds were negative for retroviruses and herpesvirus. The immunohistochemical results of this study and the destructive nature of the cellular infiltrate suggest that the lesion represents T-cell lymphoma. In birds, lymphomas are most often associated with herpes and retroviruses; the absence of these viruses suggests that the parasite initiated neoplastic transformation. Though much work needs to be done to prove the transformative nature of the lesions, these preliminary results suggest that passerine birds may be susceptible to parasite-associated lymphomas.


Assuntos
Doenças das Aves/parasitologia , Surtos de Doenças/veterinária , Enteropatias Parasitárias/veterinária , Isospora/isolamento & purificação , Isosporíase/veterinária , Linfoma de Células T/veterinária , Aves Canoras , Animais , Doenças das Aves/patologia , Imuno-Histoquímica/veterinária , Enteropatias Parasitárias/parasitologia , Enteropatias Parasitárias/patologia , Isospora/genética , Isosporíase/parasitologia , Isosporíase/patologia , Linfoma de Células T/parasitologia , Linfoma de Células T/patologia
5.
J Evol Biol ; 23(8): 1680-8, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20561136

RESUMO

Host genetic diversity can mediate pathogen resistance within and among populations. Here we test whether the lower prevalence of Mycoplasmal conjunctivitis in native North American house finch populations results from greater resistance to the causative agent, Mycoplasma gallisepticum (MG), than introduced, recently-bottlenecked populations that lack genetic diversity. In a common garden experiment, we challenged wild-caught western (native) and eastern (introduced) North American finches with a representative eastern or western MG isolate. Although introduced finches in our study had lower neutral genetic diversity than native finches, we found no support for a population-level genetic diversity effect on host resistance. Instead we detected strong support for isolate differences: the MG isolate circulating in western house finch populations produced lower virulence, but higher pathogen loads, in both native and introduced hosts. Our results indicate that contemporary differences in host genetic diversity likely do not explain the lower conjunctivitis prevalence in native house finches, but isolate-level differences in virulence may play an important role.


Assuntos
Doenças das Aves/microbiologia , Tentilhões/genética , Interações Hospedeiro-Patógeno/genética , Infecções por Mycoplasma/veterinária , Mycoplasma gallisepticum/patogenicidade , Animais , Doenças das Aves/epidemiologia , Tentilhões/imunologia , Variação Genética , Imunocompetência/imunologia , Repetições de Microssatélites/genética , Infecções por Mycoplasma/epidemiologia , Infecções por Mycoplasma/microbiologia , Mycoplasma gallisepticum/isolamento & purificação , Prevalência , Fatores de Tempo
6.
Avian Dis ; 54(1): 46-52, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20408398

RESUMO

Cornell University maintains two genetic lines of specific-pathogen-free chickens in a filtered-air, positive-pressure house as a closed colony. Offspring from each generation are maintained in the same house as the parents without clean-out between successive generations. The two lines have been persistently infected with chicken infectious anemia virus (CIAV) since the mid-1990s. All flocks were monitored from 1999 to 2008 for the presence of CIAV antibodies two to four times over the 65-wk life span of each flock, starting at approximately 15 wk of age. The serologic data were modeled using the logistic mixed model for seroprevalence and the Poisson generalized linear mixed model for seroconversion. We defined seroprevalence as the percentage of seropositive birds on a sampling date; seroconversion was defined as the difference in the percentage of seropositive birds between two subsequent bleeding dates. Seroprevalence varied between flocks from 1% to 95% but was never zero. Strain and gender in general did not influence seroprevalence or seroconversion rates, but sires of the P2a line had a significantly higher seroprevalence than all other groups. There are at least two different explanations possible for the extreme variation in seroconversion. The first one is that a low level of continuous horizontal infection from seropositive to seronegative birds occurs in the facility. The second explanation is based on the concept of latency of infection, with reactivation occurring during and after sexual maturity. Latency may occur in both seropositive and seronegative chickens. Our data are compatible with reactivation from latency, perhaps followed by limited horizontal spread as well as with a low level of continuous horizontal transmission. Although the fitted Poisson model supports both options, we propose that the reactivation from latency is the likely explanation for the observed data.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Anemia da Galinha/imunologia , Galinhas/genética , Infecções por Circoviridae/veterinária , Doenças das Aves Domésticas/transmissão , Animais , Anticorpos Antivirais/isolamento & purificação , Infecções por Circoviridae/sangue , Infecções por Circoviridae/transmissão , Infecções por Circoviridae/virologia , Feminino , Estudos Longitudinais , Masculino , Doenças das Aves Domésticas/virologia , Estudos Soroepidemiológicos , Organismos Livres de Patógenos Específicos , Fatores de Tempo , Latência Viral
7.
Curr Top Microbiol Immunol ; 331: 151-83, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19230563

RESUMO

Chicken anemia virus (CAV), the only member of the genus Gyrovirus of the Circoviridae, is a ubiquitous pathogen of chickens and has a worldwide distribution. CAV shares some similarities with Torque teno virus (TTV) and Torque teno mini virus (TTMV) such as coding for a protein inducing apoptosis and a protein with a dual-specificity phosphatase. In contrast to TTV, the genome of CAV is highly conserved. Another important difference is that CAV can be isolated in cell culture. CAV produces a single polycistronic messenger RNA (mRNA), which is translated into three proteins. The promoter-enhancer region has four direct repeats resembling estrogen response elements. Transcription is enhanced by estrogen and repressed by at least two other transcription factors, one of which is COUP-TF1. A remarkable feature of CAV is that the virus can remain latent in gonadal tissues in the presence or absence of virus-neutralizing antibodies. In contrast to TTV, CAV can cause clinical disease and subclinical immunosuppression especially affecting CD8+ T lymphocytes. Clinical disease is associated with infection in newly hatched chicks lacking maternal antibodies or older chickens with a compromised humoral immune response.


Assuntos
Vírus da Anemia da Galinha/fisiologia , Galinhas , Infecções por Circoviridae/veterinária , Doenças das Aves Domésticas/virologia , Animais , Infecções por Circoviridae/virologia
8.
Virus Res ; 132(1-2): 152-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18177972

RESUMO

Chicken anemia virus (CAV) spreads vertically and horizontally, however, the process is mostly still obscure. To further clarify the horizontal CAV spread, we examined the contribution of feathers. We demonstrated that CAV could be amplified from DNA purified from feather shafts of experimentally infected chicks, and the process efficacy was evaluated by comparing the amplification of DNA purified from feather shafts and lymphoid organs of CAV-experimentally infected chicks. DNA from feathers was found as an efficient source for CAV detection. Further, to substantiate whether CAV reaches the feather shafts passively via the blood, or intrinsically, causing histopathological changes, the feather follicle tissues were examined for CAV-induced lesions. Specific histological changes were found, however, immunohistochemistry failed to detect viral proteins. To determine whether the feather shafts are a source of infective virus, they were homogenized and used to infect 1-day-old chicks via the mucosal entries (eyes, nose and oropharynx). That infection mode simulates the natural route of horizontal infection in commercial poultry houses. We demonstrated the CAV-infection by serology, virology and pathology, showing that feather shafts carry infectious CAV either on their surface or within their feather pulp, and concluded that feathers contribute to the horizontal CAV dissemination.


Assuntos
Vírus da Anemia da Galinha/isolamento & purificação , Infecções por Circoviridae/veterinária , Plumas/virologia , Doenças das Aves Domésticas/transmissão , Animais , Anticorpos Antivirais/sangue , Vírus da Anemia da Galinha/genética , Vírus da Anemia da Galinha/imunologia , Vírus da Anemia da Galinha/patogenicidade , Galinhas/crescimento & desenvolvimento , Galinhas/virologia , Infecções por Circoviridae/imunologia , Infecções por Circoviridae/transmissão , Infecções por Circoviridae/virologia , Epitélio/patologia , Epitélio/virologia , Plumas/química , Plumas/patologia , Histologia , Tecido Linfoide/patologia , Tecido Linfoide/virologia , Mardivirus/genética , Mardivirus/imunologia , Mardivirus/isolamento & purificação , Doença de Marek/imunologia , Doença de Marek/virologia , Reação em Cadeia da Polimerase , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia
9.
Rev Sci Tech ; 26(2): 327-38, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17892155

RESUMO

Despite reducing disease, vaccination rarely protects against infection and many pathogens persist within vaccinated animal populations. Circulation of viral pathogens within vaccinated populations may favour the development of vaccine resistance with implications for the evolution of virus pathogenicity and the emergence of variant viruses. The high rate of mutations during replication of ribonucleic acid (RNA) viruses is conducive to the development of escape mutants. In vaccinated cattle, unusual mutations have been found in the major antigenic site of foot and mouth disease virus, which is also involved in receptor recognition. Likewise, atypical changes have been detected in the immunodominant region of bovine respiratory syncytial virus. Large deoxyribonucleic acid (DNA) viruses are able to recombine, generating new genotypes, as shown by the potential of glycoprotein E-negative vaccine strains of bovine herpesvirus-1 to recombine with wild-type strains. Marek's disease virus is often quoted as an example of vaccine-induced change in pathogenicity. The reasons for this increase in virulence have not been elucidated and possible explanations are discussed.


Assuntos
Vacinas Virais/imunologia , Viroses/veterinária , Vírus/patogenicidade , Animais , Mutação , Resultado do Tratamento , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos , Virulência/fisiologia , Viroses/imunologia , Viroses/prevenção & controle , Viroses/virologia
10.
Avian Pathol ; 34(2): 75-90, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16191686

RESUMO

The concept of pathotype in Marek's disease (MD) probably dates from the recognition of a more virulent form of the disease in the late 1950s (Benton & Cover, 1957). Distinctions between MD virus strains were further expanded with the description of the vv pathotype in the early 1980s and of the vv+ pathotype in the 1990s. Pathotype designations reflect important biological properties that correlate with the break-through of vaccinal immunity in the field. However, pathotyping methods applied by various laboratories have not been uniform, preventing critical comparison of results. Better uniformity of pathotyping procedures is desirable.The Avian Disease and Oncology Laboratory (ADOL) method is based on induction of lymphoproliferative lesions in vaccinated chickens. This method has been used to pathotype more than 45 isolates and is the basis for the current pathotype classification of MD virus strains. Its limitations include requirements for a specific type of chickens (15x7 ab+), large numbers of animals, and a statistical method to compare lesion responses to those of JM/102W and Md5 control strains. Because of these limitations, it has not been and is not likely to be used in other laboratories. Comparability in pathotyping can be improved by the comparison of field isolates with standard prototype strains such as JM/102W, Md5 and 648A (American Type Culture Collection) or their equivalents. Data may be generated by different in vivo procedures that measure tumour induction, neurological disease (both neoplastic and non-neoplastic lesions), or solely non-neoplastic criteria (such as lymphoid organ weights or virus replication). Methods based on neoplastic criteria, especially when generated in MD-immunized chickens, will probably correlate most closely with that of the ADOL method and be most relevant to evolution of MD virus in the field. Based on data from several trials, a modification of the ADOL method that utilizes fewer chickens and can be conducted with commercial specific pathogen free strains is proposed. The modified method is based on "best fit" comparisons with prototype strains, and is expected to provide results generally comparable with the original method. A variety of other alternative criteria (see earlier) are also evaluated both for primary pathotyping and as adjuncts to other pathotyping methods. Advantages and disadvantages of alternative methods are presented.


Assuntos
Mardivirus/classificação , Mardivirus/patogenicidade , Doença de Marek/virologia , Animais , Galinhas/genética , Galinhas/virologia , Predisposição Genética para Doença , Vacinas contra Doença de Marek , Virulência
11.
Avian Pathol ; 34(2): 91-5, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16191687

RESUMO

Splenocytes from chickens infected with low-passage stocks of Marek's disease virus (MDV) RB-1B, a very virulent (vv) strain and vv+ RK-1 were used to compare the efficacy of chick kidney cells (CKC), chicken embryo fibroblasts (CEF) and chicken embryo kidney cells (CEKC) for virus isolation. CKC were superior to CEF and CEKC. MDV foci were present at 4 days post infection in CKC but not until 6 days post infection in CEF or CEKC. Virus yield was higher in CKC than in CEF or CEKC at 6 days post infection. Passage of RB-1B in CKC yielded a significantly higher virus increase than with CEF or CEKC. The same was true for RK-1 comparing CKC with CEKC. Interestingly, RK-1-infected CEF were negative or had very low number of foci in passage 1, but virus yield increased 500-fold to 600-fold on passage in CKC, CEF, and CEKC. Recommendations on procedures for successful virus isolation are provided.


Assuntos
Herpesvirus Galináceo 2/classificação , Herpesvirus Galináceo 2/isolamento & purificação , Animais , Células Cultivadas , Embrião de Galinha , Galinhas , Fibroblastos , Rim/citologia , Baço/citologia , Virulência , Cultura de Vírus/métodos , Cultura de Vírus/veterinária , Replicação Viral/fisiologia
12.
Vet Microbiol ; 105(1): 65-72, 2005 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-15607085

RESUMO

Previous evidence for the presence of chicken anemia virus (CAV) in the gonads of immune specific-pathogen-free chickens raised the question whether this occurs also in commercial breeders. The presence of CAV was investigated by nested PCR in the gonads and spleens of hens from two 55- and 59-week-old, CAV-vaccinated (flocks 2 and 3), and two 48- and 31-week-old non-vaccinated broiler breeder flocks (flocks 1 and 4). In addition, lymphoid tissues of 20-day-old embryos from these hens were also investigated for the presence of CAV. CAV was detected in the gonads and of 5/6 and 11/22 of the vaccinated hens and in some hens also in the spleen alone. Embryos from 7/8 and 5/18 of these hens were positive. In the non-vaccinated flocks, CAV was detected in the gonads of 11/34 and 10/10 hens in flocks 1 and 4, respectively. In addition, 11 birds in flock 1 had positive spleens. CAV DNA was detected in 3/11 and 2/10 of their embryos. CAV-positive gonads and embryos were detected in samples from hens with moderate as well as high VN antibody titers. Vaccinated chickens positive for CAV in the gonads and in their embryos had VN titers ranging from >1:512 to <1:2048. In non-vaccinated chickens, the VN titers of CAV positive chickens ranged from 1:128 to 1:4096. These results demonstrate that CAV genome can remain present in the gonads of hens in commercial broiler breeder flocks even in the presence of high neutralizing antibody titers that have been associated with protection against CAV vertical transmission. It also suggests that transmission to the progeny may occur irrespectively of the level of the humoral immune response in the hens.


Assuntos
Embrião de Galinha/virologia , Vírus da Anemia da Galinha/isolamento & purificação , Galinhas , Infecções por Circoviridae/veterinária , Gônadas/virologia , Transmissão Vertical de Doenças Infecciosas/veterinária , Doenças das Aves Domésticas/virologia , Animais , Anticorpos Antivirais/sangue , Vírus da Anemia da Galinha/genética , Infecções por Circoviridae/transmissão , Infecções por Circoviridae/virologia , DNA Viral/química , DNA Viral/genética , Ensaio de Imunoadsorção Enzimática/veterinária , Feminino , Masculino , Testes de Neutralização/veterinária , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/transmissão , Baço/virologia
13.
Avian Dis ; 48(4): 734-45, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15666854

RESUMO

Chicken infectious anemia virus (CIAV) is a resistant and ubiquitous virus of chickens causing disease in young chickens and immunosuppression in all birds. This paper reviews the current knowledge of CIAV with a focus on new findings indicating that immunosuppressive effects have not been fully appreciated, especially as they relate to the development of antigen-specific cytotoxic T cells. A more complete understanding of the immunosuppressive effects of CIAV emphasizes the need for better vaccines, especially for the broiler industry. In addition, a new model is proposed for the control of viral replication in the reproductive tract of specific-pathogen-free chickens, which may be latently infected. This model suggests that virus transcription is controlled by viral enhancer and repressor elements, which are regulated by different hormones. As a consequence, CIAV has a well-adapted relationship with its host, avoiding immune detection, ensuring passage of virus to the next generation, and eliciting limited pathology to the host.


Assuntos
Vírus da Anemia da Galinha/patogenicidade , Galinhas/virologia , Infecções por Circoviridae/veterinária , Doenças das Aves Domésticas/virologia , Animais , Galinhas/imunologia , Infecções por Circoviridae/imunologia , Infecções por Circoviridae/virologia , Surtos de Doenças/veterinária , Tolerância Imunológica , Doenças das Aves Domésticas/imunologia , Latência Viral
14.
Poult Sci ; 81(11): 1661-7, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12455593

RESUMO

An avian reovirus, ARV-CU98, has recently been isolated from poults experiencing poult enteritis and mortality syndrome (PEMS). To further understand ARV-CU98 and its role in PEMS, the current study investigates interactions of ARV-CU98 with various cell types in vitro. When macrophages, B cells, T cells, and liver cells of chicken or turkey origin were co-incubated with ARV-CU98, only cells of liver origin demonstrated cytopathic effects, the presence of viral antigen, and reduced metabolic activity over time. Furthermore, distinctive pockets of viral particles were evident in electron microscopic examination of a chicken hepatocellular carcinoma (LMH) cell line, but not in a chicken macrophage cell line (MQ-NCSU) co-incubated with virus. Additional evidence of viral replication in LMH, cells but not MQ-NCSU cells was demonstrated by the presence of two viral bands (43 and 145 kD size) in cell lysates from LMH cells exposed to ARV-CU98. Although not capable of being infected by ARV-CU98, MQ-NCSU cells do appear to be activated by the virus since IL-1 mRNA expression is increased in MQ-NCSU cells 2 h after addition of the virus. LMH cells exposed to the virus demonstrate a decrease in IL-1 mRNA expression by 8 to 10 h after addition of the virus, perhaps corresponding to the initiation of infection by the virus. In conclusion, this study demonstrates that ARV-CU98 actively infects and replicates in LMH cells, but not in lymphocytes or macrophages, suggesting that the liver may be a target and site of replication of ARV-CU98 in poults experiencing PEMS.


Assuntos
Galinhas , Orthoreovirus Aviário/patogenicidade , Síndrome de Mortalidade do Peruzinho por Enterite/virologia , Infecções por Reoviridae/veterinária , Perus , Animais , Antígenos Virais/análise , Linfócitos B/metabolismo , Linfócitos B/virologia , Linhagem Celular , Células Cultivadas , Efeito Citopatogênico Viral , Interleucina-1/genética , Interleucina-1/metabolismo , Fígado/citologia , Fígado/virologia , Macrófagos/metabolismo , Macrófagos/virologia , Microscopia Eletrônica/veterinária , Orthoreovirus Aviário/crescimento & desenvolvimento , Orthoreovirus Aviário/imunologia , Síndrome de Mortalidade do Peruzinho por Enterite/metabolismo , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/virologia , RNA Mensageiro/metabolismo , Infecções por Reoviridae/virologia , Organismos Livres de Patógenos Específicos , Linfócitos T/metabolismo , Linfócitos T/virologia , Células Tumorais Cultivadas , Replicação Viral
15.
Avian Dis ; 46(3): 636-49, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12243528

RESUMO

Nitric oxide (NO), a free radical produced by the enzyme NO synthase (NOS), is a potent antiviral agent in addition to having immune regulating functions. Recently, it was reported that chickens resistant (N2a, MHC: B21B21) to the development of Marek's disease (MD) had a greater potential to produce NO than MD-susceptible chickens (P2a, MHC: B19B19). This difference was shown by measuring NO levels in chick embryo fibroblast cultures obtained from these chickens after treatment with lipopolysaccharide and recombinant chicken interferon-gamma (IFN-gamma). To extend these results, the levels of NO in blood plasma from N2a and P2a chickens inoculated with the nonattenuated JM-16 strain of MD virus (MDV) were examined. In four out of five experiments, N2a chickens had increased NO levels at 7 days postinoculation (DPI). In contrast, P2a chickens challenged with JM-16 had a significant increase in NO in only one of four experiments, and in that experiment the increase was delayed (10 DPI) compared with N2a chickens. Attenuation abrogated MDV-induced NO in chickens. Inoculation with MDV strains ranging from mild to very virulent plus showed that the more virulent strains induced the highest level of NO in blood plasma, suggesting a role of NO in the pathogenesis of MD with more virulent strains. On the basis of quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR) assays for analysis of mRNA expression, IFN-gamma does not appear to be the primary inducer of inducible (i)NOS gene expression during MDV infection. iNOS gene expression and NO production are mediated during the cytolytic phase of MDV infection on the basis of real-time RT-PCR assays with primers specific for glycoprotein B, a late gene expressed only during the cytolytic phase of MDV infection. These findings implicate NO as a factor potentially involved in increasing virulence of MDV, possibly through immune suppression.


Assuntos
Galinhas , Herpesvirus Galináceo 2/patogenicidade , Imunidade Inata/genética , Doença de Marek/imunologia , Óxido Nítrico/biossíntese , Animais , Células Cultivadas , Embrião de Galinha , DNA Complementar/genética , Suscetibilidade a Doenças/imunologia , Fibroblastos/imunologia , Regulação Enzimológica da Expressão Gênica , Herpesvirus Galináceo 2/classificação , Herpesvirus Galináceo 2/imunologia , Imunidade Inata/imunologia , Interferon gama/biossíntese , Interferon gama/genética , Doença de Marek/genética , Óxido Nítrico/sangue , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Análise de Sequência de DNA , Organismos Livres de Patógenos Específicos , Baço/imunologia , Virulência
16.
Avian Dis ; 46(1): 32-47, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11922348

RESUMO

Poult enteritis and mortality syndrome (PEMS) is an acute, infectious intestinal disease of turkey poults, characterized by high mortality and 100% morbidity, that decimated the turkey industry in the mid-1990s. The etiology of PEMS is not completely understood. This report describes the testing of various filtrates of fecal material from control and PEMS-affected poults by oral inoculation into poults under experimental conditions, the subsequent isolation of a reovirus, ARV-CU98, from one of the PEMS fecal filtrates, and in vivo and in vitro studies conducted to determine the pathogenicity of ARV-CU98 in turkey poults. In order to identify a filtrate fraction of fecal material containing a putative etiologic agent, poults were challenged in two independent experiments with 220- and 100-nm filtrates of fecal material from PEMS-negative and PEMS-positive poults. The 100-nm filtrate was chosen for further evaluation because poults inoculated with this filtrate exhibited mortality and significantly lower (P < or = 0.05) body weight and relative bursa weight, three clinical signs associated with PEMS. These results were confirmed in a third experiment with 100-nm fecal filtrates from a separate batch of PEMS fecal material. In Experiment 3, body weight and relative bursa and thymus weights were significantly lower (P < or = 0.05) in poults inoculated with 100-nm filtrate of PEMS fecal material as compared with poults inoculated with 100-nm filtrate of control fecal material. Subsequently, a virus was isolated from the 100-nm PEMS fecal filtrate and propagated in liver cells. This virus was identified as a reovirus on the basis of cross-reaction with antisera against avian reovirus (FDO strain) as well as by electrophoretic analysis and was designated ARV-CU98. When inoculated orally into poults reared under controlled environmental conditions in isolators, ARV-CU98 was associated with a higher incidence of thymic hemorrhaging and gaseous intestines. In addition, relative bursa and liver weights were significantly lower (P < or = 0.05) in virus-inoculated poults as compared with controls. Virus was successfully reisolated from virus-challenged poults but not from control birds. Furthermore, viral antigen was detected by immunofluorescence in liver sections from virus-challenged poults at 3 and 6 days postinfection and virus was isolated from liver at 6 days postinfection, suggesting that ARV-CU98 replicates in the liver. In addition to a decrease in liver weight, there was a functional degeneration as indicated by altered plasma alanine aminotransferase and aspartate aminotransferase activities in virus poults as compared with controls. Although this reovirus does not induce fulminating PEMS, our results demonstrated that ARV-CU98 does cause some of the clinical signs in PEMS, including intestinal alterations and significantly lower relative bursa and liver weights. ARV-CU98 may contribute directly to PEMS by affecting the intestine, bursa, and liver and may contribute indirectly by increasing susceptibility to opportunistic pathogens that facilitate development of clinical PEMS.


Assuntos
Fezes/virologia , Orthoreovirus Aviário/isolamento & purificação , Síndrome de Mortalidade do Peruzinho por Enterite/virologia , Infecções por Reoviridae/veterinária , Animais , Peso Corporal , Feminino , Imunofluorescência/veterinária , Tamanho do Órgão , Orthoreovirus Aviário/classificação , Orthoreovirus Aviário/patogenicidade , Síndrome de Mortalidade do Peruzinho por Enterite/imunologia , Síndrome de Mortalidade do Peruzinho por Enterite/patologia , Infecções por Reoviridae/etiologia , Infecções por Reoviridae/virologia , Perus
17.
J Interferon Cytokine Res ; 21(5): 287-96, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11429159

RESUMO

Mammalian type I interferons (IFN-alpha/beta) are potent mediators of innate antiviral immune responses, in particular through enhancement of natural killer (NK) cell cytotoxicity. Recently, chicken IFN-alpha (ChIFN-alpha) has been identified and shown to ameliorate Newcastle disease virus (NDV) infection when given to chickens at relatively high concentrations in the drinking water. In this report, the effect of recombinant ChIFN-alpha (rChIFN-alpha) on NK cell cytotoxicity was examined using (51)Cr-release assays. NK cell cytotoxic activity was also analyzed following inoculation with attenuated Marek's disease virus (MDV) serotype 1 strain R2/23 and a recombinant MDV (parent strain R2/23)-expressing ChIFN-alpha [rMDV(IFN-alpha)]. Treatment of chickens with high doses of rChIFN-alpha in the drinking water significantly decreased NK cell cytotoxicity compared with untreated chickens over a 7-day period. Inoculation of chickens with R2/23 significantly decreased NK cell cytotoxicity as well, whereas the rMDV(IFN-alpha) had no effect on NK cell cytotoxicity. Treatment of chicken embryo cell cultures with rChIFN-alpha inhibited replication of the very virulent MDV RB-1B strain in vitro, and oral treatment of chickens with rChIFN-alpha reduced MDV R2/23 replication in vivo.


Assuntos
Herpesvirus Galináceo 2/imunologia , Interferon Tipo I/administração & dosagem , Interferon Tipo I/biossíntese , Interferon-alfa/administração & dosagem , Interferon-alfa/biossíntese , Doença de Marek/imunologia , Administração Oral , Animais , Antivirais/administração & dosagem , Antivirais/metabolismo , Galinhas , Clonagem Molecular , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/genética , Ingestão de Líquidos , Herpesvirus Galináceo 2/efeitos dos fármacos , Herpesvirus Galináceo 2/genética , Células Matadoras Naturais/imunologia , Proteínas Recombinantes , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/biossíntese , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/biossíntese , Vacinas Virais/imunologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética , Replicação Viral/imunologia
19.
J Virol ; 74(21): 10176-86, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11024146

RESUMO

The QT35 cell line was established from a methylcholanthrene-induced tumor in Japanese quail (Coturnix coturnix japonica) (C. Moscovici, M. G. Moscovici, H. Jimenez, M. M. Lai, M. J. Hayman, and P. K. Vogt, Cell 11:95-103, 1977). Two independently maintained sublines of QT35 were found to be positive for Marek's disease virus (MDV)-like genes by Southern blotting and PCR assays. Sequence analysis of fragments of the ICP4, ICP22, ICP27, VP16, meq, pp14, pp38, open reading frame (ORF) L1, and glycoprotein B (gB) genes showed a strong homology with the corresponding fragments of MDV genes. Subsequently, a serotype 1 MDV-like herpesvirus, tentatively name QMDV, was rescued from QT35 cells in chicken kidney cell (CKC) cultures established from 6- to 9-day-old chicks inoculated at 8 days of embryonation with QT35 cells. Transmission electron microscopy failed to show herpesvirus particles in QT35 cells, but typical intranuclear herpesvirus particles were detected in CKCs. Reverse transcription-PCR analysis showed that the following QMDV transcripts were present in QT35 cells: sense and antisense meq, ORF L1, ICP4, and latency-associated transcripts, which are antisense to ICP4. A transcript of approximately 4.5 kb was detected by Northern blotting using total RNA from QT35 cells. Inoculation of QT35 cells with herpesvirus of turkeys (HVT)-infected chicken embryo fibroblasts (CEF) but not with uninfected CEF resulted in the activation of ICP22, ICP27, VP16, pp38, and gB. In addition, the level of ICP4 mRNA was increased compared to that in QT35 cells. The activation by HVT resulted in the production of pp38 protein. It was not possible to detect if the other activated genes were translated due to the lack of serotype 1-specific monoclonal antibodies.


Assuntos
Fibroblastos/virologia , Herpesviridae/genética , Herpesvirus Galináceo 2/fisiologia , Ativação Transcricional , Latência Viral , Animais , Antígenos Virais/metabolismo , Southern Blotting , Células Cultivadas , Coturnix , Herpesviridae/fisiologia , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/isolamento & purificação , Dados de Sequência Molecular , Fosfoproteínas/metabolismo , Reação em Cadeia da Polimerase , Codorniz , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Perus/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
20.
Avian Dis ; 44(3): 661-7, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11007016

RESUMO

This is a comparative study on seroconversion to chicken infectious anemia virus (CIAV) in a closed flock of specific-pathogen-free chickens undergoing a natural outbreak and after vaccination of some of these flocks with a commercial, live vaccine. The N2a strain (B21B21 haplotype) had the highest seroconversion after natural infection (94%) or vaccination (100%), followed by the P2a strain (B19B19) at 75%-82% seroconversion after natural infection and 85% seroconversion after vaccination. The S13 (B13B13) chickens were 26% seropositive after natural infection and 75% seropositive after vaccination. N2a chickens with polymerase chain reaction (PCR)-positive tissues were 97% seropositive compared to 80%-83% PCR-positive and seropositive for the P2a chickens and only 8% seropositive and PCR-positive for the S13 chickens. Seroconversion occurred at or near sexual maturity after natural infection in seven flocks studied.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Anemia da Galinha/imunologia , Galinhas/imunologia , Infecções por Circoviridae/veterinária , Doenças das Aves Domésticas/imunologia , Vacinas Virais , Criação de Animais Domésticos , Animais , Formação de Anticorpos , Vírus da Anemia da Galinha/isolamento & purificação , Infecções por Circoviridae/epidemiologia , Infecções por Circoviridae/imunologia , Infecções por Circoviridae/prevenção & controle , Surtos de Doenças/veterinária , Reação em Cadeia da Polimerase , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/prevenção & controle , Especificidade da Espécie , Organismos Livres de Patógenos Específicos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA