Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Poult Sci ; 103(7): 103835, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38772092

ABSTRACT

Avian leukemia virus subgroup J (ALV-J) and chicken infectious anemia virus (CIAV) can be vertically transmitted; however, the pathogenicity of vertically transmitted coinfection with these 2 pathogens has not been studied. In this study, we created a model of chick morbidity in which chicks carried either ALV-J, CIAV, or both viruses via embryo inoculation. Thereafter, we analyzed the effects of vertically transmitted coinfection with CIAV and ALV-J on the pathogenicity of ALV-J and performed a purification assay based on hatching, mortality viremia positivity, and detection of fecal ALV-p27 antigen rates, and body weight. The hatching rate of the ALV-J+CIAV group was 68.57%, lower than those of the single infection and control groups. The survival curve showed that the mortality rates of the CIAV and ALV-J coinfection groups were higher than those of the single infection and control groups. Body weight statistics showed that coinfection aggravated the 7-d growth inhibition effect. The results of ALV-p27 antigen detection in cell culture supernatants showed that the positivity rates of the ALV-J and ALV-J+CIAV groups were 100% at all ages and 0% in the control group. The results of ALV-p27 antigen detection by anal swabs showed that the positivity rates of the ALV-J group were 92.86, 90.90, 88.89, and 93.33% at all ages, and that the ALV-J p27 positivity detection rate of anal swabs was lower than that of plasma virus isolation. The immune organ index of the ALV-J+CIAV group was significantly or very significantly lower than those of the single infection and control groups. The immune organ viral load showed that coinfection with CIAV and ALV-J promoted the proliferation of ALV-J and CIAV in immune organs. Coinfection with ALV-J and CIAV reduced chicken embryo hatchability and increased chick mortality and growth inhibition relative to their respective single infections. Additionally, coinfection with ALV-J + CIAV was even more detrimental in inducing immune organ atrophy (e.g., the thymus, spleen, and bursa), and promoted individual virus replication during coinfection.


Subject(s)
Avian Leukosis Virus , Avian Leukosis , Chicken anemia virus , Chickens , Circoviridae Infections , Coinfection , Infectious Disease Transmission, Vertical , Poultry Diseases , Animals , Avian Leukosis Virus/physiology , Avian Leukosis Virus/pathogenicity , Chickens/virology , Avian Leukosis/virology , Coinfection/veterinary , Coinfection/virology , Poultry Diseases/virology , Chicken anemia virus/physiology , Chicken anemia virus/pathogenicity , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Infectious Disease Transmission, Vertical/veterinary , Virulence , Chick Embryo
2.
Viruses ; 13(10)2021 10 02.
Article in English | MEDLINE | ID: mdl-34696415

ABSTRACT

Newcastle disease virus (NDV) strain R2B, with an altered fusion protein cleavage site, was used as a viral vector to deliver the immunogenic genes VP2 and VP1 of chicken infectious anaemia virus (CIAV) to generate a bivalent vaccine candidate against these diseases in chickens. The immunogenic genes of CIAV were expressed as a single transcriptional unit from the NDV backbone and the two CIA viral proteins were obtained as separate entities using a self-cleaving foot-and-mouth disease virus 2A protease sequence between them. The recombinant virus (rR2B-FPCS-CAV) had similar growth kinetics as that of the parent recombinant virus (rR2B-FPCS) in vitro with similar pathogenicity characteristics. The bivalent vaccine candidate when given in specific pathogen-free chickens as primary and booster doses was able to elicit robust humoral and cell-mediated immune (CMI) responses obtained in a vaccination study that was conducted over a period of 15 weeks. In an NDV and CIAV ELISA trial, there was a significant difference in the titres of antibody between vaccinated and control groups which showed slight reduction in antibody titre by 56 days of age. Hence, a second booster was administered and the antibody titres were maintained until 84 days of age. Similar trends were noticed in CMI response carried out by lymphocyte transformation test, CD4+ and CD8+ response by flow cytometry analysis and response of real time PCR analysis of cytokine genes. Birds were challenged with virulent NDV and CIAV at 84 days and there was significant reduction in the NDV shed on the 2nd and 4th days post challenge in vaccinated birds as compared to unvaccinated controls. Haematological parameters comprising PCV, TLC, PLC and PHC were estimated in birds that were challenged with CIAV that indicated a significant reduction in the blood parameters of controls. Our findings support the development and assessment of a bivalent vaccine candidate against NDV and CIAV in chickens.


Subject(s)
Chicken anemia virus/immunology , Chickens/immunology , Newcastle disease virus/genetics , Animals , Antibodies, Viral/blood , Chicken anemia virus/pathogenicity , Chickens/virology , Genetic Vectors , Immunity/immunology , Immunity, Cellular , Newcastle Disease/virology , Newcastle disease virus/immunology , Newcastle disease virus/pathogenicity , Poultry Diseases/virology , Vaccination/methods , Viral Vaccines/immunology
3.
Vet Microbiol ; 260: 109182, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34315003

ABSTRACT

Immunosuppression can increase the susceptibility of chickens to other disease-causing pathogens and interfere with the efficacy of vaccination against those pathogens. Chicken anaemia virus (CAV) and infectious bursal disease virus (IBDV) are common causes of immunosuppression in chickens. Immunosuppression was induced by experimental infection with either CAV or IBDV to assess the effect of immunosuppression on the efficacy of vaccination with Mycoplasma gallisepticum strain ts-304 against infection with virulent M. gallisepticum, a common bacterial pathogen of chickens worldwide. Birds were experimentally infected with either CAV or IBDV at 1 week of age, before vaccination and challenge with M. gallisepticum to examine the effect of immunosuppression at the time of vaccination, or at 6 weeks of age, after vaccination against M. gallisepticum but before challenge with virulent M. gallisepticum, to investigate the effect of immunosuppression at the time of challenge. All birds were vaccinated with a single dose of the ts-304 vaccine at 3 weeks of age and experimentally challenged with the virulent M. gallisepticum strain Ap3AS at 8 weeks of age. In immunosuppressed chickens there was a reduction in protection offered by the ts-304 vaccine at two weeks after challenge, as measured by tracheal mucosal thicknesses, serum antibody levels against M. gallisepticum, air sac lesion scores and virulent M. gallisepticum load in the trachea. Immunosuppressed birds with detectable serum antibodies against M. gallisepticum were less likely to have tracheal lesions. This study has shown that immunosuppression caused by infection with CAV or IBDV can interfere with vaccination against mycoplasmosis in chickens.


Subject(s)
Birnaviridae Infections/veterinary , Chicken anemia virus/immunology , Chickens/immunology , Circoviridae Infections/veterinary , Infectious bursal disease virus/immunology , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/immunology , Poultry Diseases/prevention & control , Air Sacs/virology , Animals , Birnaviridae Infections/prevention & control , Birnaviridae Infections/virology , Chicken anemia virus/pathogenicity , Chickens/microbiology , Circoviridae Infections/prevention & control , Circoviridae Infections/virology , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Immunosuppression Therapy/veterinary , Infectious bursal disease virus/pathogenicity , Mucous Membrane/virology , Mycoplasma Infections/microbiology , Mycoplasma Infections/prevention & control , Mycoplasma gallisepticum/pathogenicity , Poultry Diseases/microbiology , Trachea/virology
4.
Arch Virol ; 166(2): 439-449, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33389105

ABSTRACT

Chicken infectious anemia (CIA), caused by chicken anemia virus (CAV), is an important immunosuppressive disease that seriously threatens the global poultry industry. Here, we isolated and identified 30 new CAV strains from CAV-positive flocks. The VP1 genes of these strains were sequenced and analyzed at the nucleotide and amino acid levels and were found to have very similar nucleotide sequences (> 97% identity); however, they showed 93.9-100.0% sequence identity to the VP1 genes of 55 reference strains. Furthermore, alignment of the deduced amino acid sequences revealed some unique mutations. Phylogenetic analysis indicated the division of VP1 amino acid sequences into two groups (A and B) and four subgroups (A1, A2, A3 and A4). Interestingly, 22 of the newly isolated strains and some Asian reference strains belonged to the A1 group, whereas the remaining eight new isolates belonged to the A3 group. To evaluate the pathogenicity of the epidemic CAV strains from China, the representative strains CAV-JL16/8901 and CAV-HeN19/3001 and the reference strain Cux-1 were selected for animal experiments. Chickens infected with the isolates and reference strain all showed thymus atrophy and bone marrow yellowing. The mortality rates for CAV-JL16/8901, CAV-HeN19/3001, and the reference strain was 30%, 20%, and 0%, respectively, indicating that the epidemic strains pose a more serious threat to chickens. We not only analyzed the molecular evolution of the epidemic strains but also showed for the first time that the epidemic strains in China are more pathogenic than reference strain Cux-1. Effective measures should be established to prevent the spread of CIA in China.


Subject(s)
Chicken anemia virus/genetics , Chicken anemia virus/pathogenicity , Chickens/virology , Animals , China , Circoviridae Infections/virology , DNA, Viral/genetics , Evolution, Molecular , Genotype , Molecular Epidemiology/methods , Phylogeny , Poultry Diseases/virology , Sequence Analysis, DNA/methods , Virulence/genetics
5.
PLoS One ; 14(1): e0210696, 2019.
Article in English | MEDLINE | ID: mdl-30657774

ABSTRACT

Chicken anaemia virus (CAV), a member of the genus Gyrovirus, is the etiological agent of chicken infectious anaemia. CAV infects bone marrow-derived cells, resulting in severe anaemia and immunosuppression in young chickens and a compromised immune response in older birds. We investigated the molecular epidemiology of CAV in sick chickens in China from 2014 to 2015 and showed that the CAV-positive rate was 13.30%, in which mixed infection (55.56%) was the main type of infection. We isolated and identified 15 new CAV strains using different methods including indirect immunofluorescence assay and Western Blotting. We used overlapping polymerase chain reaction to map the whole genome of the strains. Phylogenetic analyses of the obtained sequences and related sequences available in GenBank generated four distinct groups (A-D). We built phylogenetic trees using predicted viral protein (VP) sequences. Unlike CAV VP2s and VP3s that were well conserved, the diversity of VP1s indicated that the new strains were virulent. Our epidemiological study provided new insights into the prevalence of CAV in clinical settings in recent years in China.


Subject(s)
Chicken anemia virus/pathogenicity , Poultry Diseases/epidemiology , Poultry Diseases/virology , Animals , Chicken anemia virus/genetics , Chickens , China/epidemiology , Circoviridae Infections/epidemiology , Circoviridae Infections/virology , Molecular Epidemiology , Polymerase Chain Reaction , Viral Proteins/genetics
6.
Poult Sci ; 98(5): 1985-1992, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30566627

ABSTRACT

Newcastle disease virus (NDV)-attenuated vaccine has been widely used to prevent ND in poultry flocks, while many reports also mentioned the exogenous virus contamination in attenuated vaccines, which might be the reason for the widespread of some contagious diseases. Recently, the chicken infectious anemia virus (CIAV) contamination in the NDV-attenuated vaccine was also found in China, though no systemic study has studied the pathogenicity or infection mechanism of this special transmission route. Accordingly, simulation experiments were launched using CIAV isolated from a contaminated NDV-attenuated vaccine. Results showed that using NDV-attenuated vaccine contaminated with CIAV could cause CIA in chickens with obvious symptoms, including anemia, hemorrhage, lymphoatrophy, and growth retardation, while the synergistic reaction of CIAV and LaSota prompted their multiplication in vivo and disturbed the production of antibodies against each other. And CIAV could significantly reduce the NDV antibody titers and decrease the protective effectiveness. This study showed the synergetic pathogenicity of CIAV and LaSota strain after using contaminated NDV-attenuated vaccine, helping us to understand how the CIAV causes infection and induces severe diseases with a relatively low dose through the mouth, as well as reminding us that the damage of an attenuated vaccine contaminated with CIAV even in extremely low dose is not insignificant.


Subject(s)
Chicken anemia virus/pathogenicity , Chickens , Newcastle disease virus/pathogenicity , Viral Vaccines/analysis , Animals , Antibodies, Viral/analysis , Chicken anemia virus/immunology , China , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Newcastle Disease/virology , Newcastle disease virus/immunology , Poultry Diseases/virology , Vaccines, Attenuated/analysis , Virulence
7.
Vet Res ; 49(1): 80, 2018 Aug 06.
Article in English | MEDLINE | ID: mdl-30081944

ABSTRACT

Newcastle disease virus (NDV)-attenuated vaccine has been widely used since the 1950s and made great progress in preventing and controlling Newcastle disease. However, many reports mention exogenous virus contamination in attenuated vaccines, while co-contamination with fowl adenovirus (FAdV) and chicken infectious anaemia virus (CIAV) in the NDV-attenuated vaccine also emerged in China recently, which proved to be an important reason for the outbreaks of inclusion body hepatitis-hydropericardium syndrome in some flocks. It is amazing that exogenous virus contamination at extremely low doses still infected chickens and induced severe disease; thus, we speculated that there must be some interaction between the NDV-attenuated vaccine and the contaminated exogenous viruses within. Accordingly, simulation experiments were launched using FAdV and CIAV isolated from the abovementioned vaccine. The results showed that the pathogenicity of FAdV and CIAV co-infection through the contaminated vaccine was significantly higher than that of direct oral infection, while the synergistic reaction of these viruses and LaSota prompted their multiplication in vivo and disturbed the production of antibodies against each other. This study showed the interactions of FAdV, CIAV and LaSota after using contaminated NDV-attenuated vaccine, helping us to understand how the contaminated exogenous viruses cause infection and induce severe disease at a relatively low dose through the oral route.


Subject(s)
Adenoviridae Infections/veterinary , Circoviridae Infections/veterinary , Newcastle Disease/prevention & control , Newcastle disease virus/immunology , Poultry Diseases/immunology , Viral Vaccines/immunology , Adenoviridae Infections/immunology , Adenoviridae Infections/prevention & control , Animals , Aviadenovirus/immunology , Aviadenovirus/pathogenicity , Chicken anemia virus/immunology , Chicken anemia virus/pathogenicity , Circoviridae Infections/immunology , Circoviridae Infections/prevention & control , Newcastle Disease/immunology , Newcastle disease virus/pathogenicity , Poultry Diseases/prevention & control , Random Allocation , Specific Pathogen-Free Organisms , Viral Vaccines/administration & dosage , Virulence
8.
Biomed Res Int ; 2017: 6707868, 2017.
Article in English | MEDLINE | ID: mdl-28326326

ABSTRACT

Chicken infectious anemia virus (CIAV) causes acute viral infection in chickens worldwide. It can infect chickens of all ages, but the disease is seen only in young chickens and is characterized by hemorrhagic lesions in the muscles, atrophic changes in the lymphoid organs, aplastic bone marrow, and immunosuppression causing increased mortality. Previous studies have demonstrated that CIAV can be isolated from blood specimens of humans and fecal samples of stray cats. In the present study, two variants of CIAV were isolated from fecal samples of mice (CIAV-Mouse) and stray dogs (CIAV-Dog), respectively. The genome of the two CIAV variants was sequenced and the results of the recombination detection program suggested that the CIAV-Dog strain could be a recombinant viral strain generated from parental CIAV strains, AB119448 and GD-1-12, with high confidence. Particularly, these findings were obtained from the comparison of genetic diversity and the relationship of CIAV between different hosts. This is the first report indicating that there is a significant difference in the number of transcription factor binding sites in CIAV noncoding regions from different hosts. Further studies are required to investigate the large geographic distribution of CIAV and monitor the variants, host range, and associated diseases.


Subject(s)
Chicken anemia virus/genetics , Genetic Variation , Host-Pathogen Interactions/genetics , Poultry Diseases/genetics , Animals , Cats , Chicken anemia virus/isolation & purification , Chicken anemia virus/pathogenicity , Chickens , Dogs , Genome, Viral , Humans , Mice , Poultry Diseases/epidemiology , Poultry Diseases/virology
9.
Virol J ; 14(1): 38, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28222746

ABSTRACT

BACKGROUND: Chicken anemia virus (CAV) causes anemia and immune suppression, which are important diseases in the poultry industry. CAV VP3, also referred as 'apoptin', has been shown to selectively kill tumor cells, raising great hopes for its utilization as an anticancer therapy. The ability of apoptin to induce apoptosis is closely related to its nuclear localization. The C-terminal region of apoptin contains a bipartite nuclear localization signals (NLS), and a nuclear export signal (NES) is located between the arms of the NLS. Most previous studies have expressed apoptin of different lengths in vitro to understand the relationship between its localization and its induction of apoptosis. METHODS: In this study, we investigated the replication of CAV and its induction of apoptosis in vitro and in vivo with VP3-truncated infectious virus. Quantitative PCR was used to detect viral replication in MDCC-MSB1 cells, and the viral localization was observed by confocal microscopy. Flow cytometry was uesed to analyze virus-induced apoptosis in MDCC-MSB1 cells. Additionally, chickens infected with the rescued viruses compared with the parental virus rM9905 to evaluate the viral replication in vivo and virulence. RESULTS: Based on the infectious clone, we rescued two viruses in which were deleted NES-NLS2 (rCAV-VP3N88) or NLS1-NES-NLS2 (rCAV-VP3N80) in the C-terminal region of apoptin. The viral load of rCAV-VP3N88 decreased significantly between 60 and 108 hpi, and was always 10-100-fold lower than that of the parental virus rM9905. The levels of rCAV-VP3N80 were also 10-100-fold lower than that of rM9905 and declined significantly at three time points. There was almost no difference in the viral loads of rCAV-VP3N88 and rCAV-VP3N80. Additionally, rM9905 induced 85.39 ± 2.18% apoptosis at 96 hpi, whereas rCAV-VP3N88 and rCAV-VP3N80 induced 63.08 ± 4.78% and 62.56 ± 7.35% apoptosis, respectively, which were significantly (about 20%) lower than that induced by the parental virus. The rescued viruses altered the nuclear localization in MDCC-MSB1 cells. Moreover, deletion of C-terminal region of apoptin impaired viral replication in vivo and reduced the virulence of CAV in chickens. CONCLUSIONS: In summary, we have demonstrated that the C-terminal deletion of apoptin in infectious CAV affected the replication of the virus. The deletion of the C-terminal region of apoptin not only significantly reduced viral replication in vitro but also reduced its induction of apoptosis, which correlated with the loss of its nuclear localization. The deletion of the C-terminal region of apoptin also impaired the replication of CAV and attenuated its virulence in chickens.


Subject(s)
Apoptosis , Capsid Proteins/genetics , Chicken anemia virus/physiology , Chicken anemia virus/pathogenicity , Virulence Factors/genetics , Virus Replication , Active Transport, Cell Nucleus , Animals , Capsid Proteins/metabolism , Cell Line , Chickens , DNA Mutational Analysis , Flow Cytometry , Microscopy, Confocal , Real-Time Polymerase Chain Reaction , Virulence , Virulence Factors/metabolism
10.
Virus Genes ; 53(2): 312-316, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27933433

ABSTRACT

An Iranian field strain of chicken anemia virus (CAV), designated IR CAV, was isolated in the Marek's disease virus-transformed lymphoblastoid cell line MDCC-MSB1 (MSB1) culture for the first time. The full-length CAV DNA of this strain was cloned in the bacterial plasmid pTZ57R/T to create the molecular clone pTZ-CAV. The nucleotide and deduced amino acid sequences of viral proteins of IR CAV were compared with those of representative CAV sequences including reference and commercial vaccine strains. IR CAV was not related to vaccine strains and also found to have glutamine at positions 139 and 144 confirming previous studies in which such mutations were associated with a slow rate of virus spread in cell culture. pTZ-CAV was digested with PstI to release IR CAV DNA and then transfected into MSB1 cell by electroporation. The transfected cells showed cytopathic effect similar to virion-initiated infection. One-day old specific pathogen-free chicks were inoculated with the regenerated virus, which had been obtained from transfected MSB1 cells, and compared with the chicks inoculated with IR CAV. Gross lesions in the birds inoculated with the regenerated virus illustrated the infectious nature of the regenerated virus from the cloned IR CAV DNA.


Subject(s)
Chicken anemia virus/genetics , Marek Disease/genetics , Poultry Diseases/genetics , Viral Proteins/genetics , Amino Acid Sequence , Animals , Chicken anemia virus/pathogenicity , Chickens/virology , Cloning, Molecular , DNA, Viral/genetics , Iran , Marek Disease/virology , Poultry Diseases/virology
11.
Biomed Res Int ; 2016: 4275718, 2016.
Article in English | MEDLINE | ID: mdl-27298822

ABSTRACT

The antibody to chicken infectious anemia virus (CIAV) was positive in a specific pathogen-free (SPF) chicken population by ELISA test in our previous inspection, indicating a possible infection with CIAV. In this study, blood samples collected from the SPF chickens were used to isolate CIAV by inoculating into MSB1 cells and PCR amplification. A CIAV strain (SD1403) was isolated and successfully identified. Three overlapping genomic fragments were obtained by PCR amplification and sequencing. The full genome sequence of the SD1403 strain was obtained by aligning the sequences. The genome of the SD1403 strain was 2293 bp with a nucleotide identity of 94.8% to 98.5% when compared with 30 referred CIAV strains. The viral proteins VP2 and VP3 were highly conserved, but VP1 was not relatively conserved. Both amino acids 139 and 144 of VP1 were glutamine, which was in accord with the low pathogenic characteristics. In this study, we first reported that CIAV exists in Chinese SPF chicken populations and may be an important reason why attenuated vaccine can be contaminated with CIAV.


Subject(s)
Chicken anemia virus/genetics , Chickens/genetics , Circoviridae Infections/genetics , Animals , Antibodies, Viral/blood , Antibodies, Viral/genetics , Chicken anemia virus/isolation & purification , Chicken anemia virus/pathogenicity , Chickens/blood , Chickens/virology , China , Circoviridae Infections/blood , Circoviridae Infections/pathology , Circoviridae Infections/virology , Enzyme-Linked Immunosorbent Assay , Genome
12.
Viral Immunol ; 24(1): 3-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21319974

ABSTRACT

Chicken anemia virus (CAV) is an economically important virus affecting the chicken meat and egg industry. CAV is characterized by anemia, lymphoid depletion, and immunosuppression. Microarrays were used to investigate the response of MDCC-MSB1 cells (MSB1) to infection with CAV at 24 and 48 h post-infection (hpi). The major genes responding to CAV infection include genes involved in inflammation, apoptosis, and antiviral activity. Several cytokines were differentially regulated at either 24 or 48 hpi, including interleukin 2 (IL-2), interleukin receptors IL-1R, IL-22R, IL-18R, and IL-7R, and interferon-α (IFN-α). While there were many genes differentially regulated in this experiment, only two genes were common to both time points, suggesting a dramatic change in gene expression over the two time points studied. The present study is the first microarray experiment to investigate CAV, and we identified a number of key pathways involved in viral infection. Overall, there were more genes upregulated at 24 hpi than at 48 hpi, including genes involved in cytokine signaling, apoptosis, and antiviral activity. The two time points were vastly different in their gene expression patterns, in that at 24 hpi there were many genes involved in the response to infection, whereas at 48 hpi there were many genes associated with apoptosis and immunosuppression.


Subject(s)
Chicken anemia virus/pathogenicity , Gene Expression Profiling , Host-Pathogen Interactions , Animals , Cell Line , Chickens , Cytokines/biosynthesis , Microarray Analysis , Time Factors
13.
Afr J Med Med Sci ; 39 Suppl: 21-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-22416640

ABSTRACT

Chicken anaemia virus (CAV) DNA was extracted from thymus, liver and bone marrow samples obtained from broiler and pullet chicken flocks in southwestern Nigeria, which presented with clinical signs and lesions suggestive of both infectious bursal disease and chicken infectious anaemia. While CAV was successfully isolated in MDCC-MSB1 cells from four of the pooled tissue samples, the remaining two samples failed to grow in cells. Monoclonal antibody (MAb) characterization using four MAbs produced against the reference Cuxhaven-1 (Cux-1) CAV isolate showed that Nigerian CAV isolates are antigenically related to each other and to the Cux-1 virus. Pathogenicity studies with the Cux-1 virus and one of the Nigerian isolates (NGR-1) revealed that NGR-1 was more pathogenic that the former. We conclude that although Nigerian CAV isolates are antigenically related to each other, they differ in terms of cell culture growth characteristics and probably pathogenicity. These findings further confirm that CAV exists and can no longer be ignored in poultry disease diagnosis in Nigeria. Cases hitherto diagnosed as IBD may actually be CIA or a co-infection of the two.


Subject(s)
Chicken anemia virus/genetics , Chicken anemia virus/isolation & purification , Chickens/virology , Circoviridae Infections/veterinary , Poultry Diseases/virology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Antigens, Viral/metabolism , Bone Marrow/virology , Cell Line , Chicken anemia virus/immunology , Chicken anemia virus/pathogenicity , Circoviridae , Circoviridae Infections/virology , DNA, Viral/genetics , Fluorescent Antibody Technique, Indirect/veterinary , Liver/virology , Molecular Sequence Data , Nigeria , Polymerase Chain Reaction/veterinary , Poultry Diseases/genetics , Thymus Gland/virology
14.
Biochim Biophys Acta ; 1793(8): 1335-42, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19374922

ABSTRACT

Apoptin, a small protein from chicken anemia virus, has attracted great attention, because it specifically kills tumor cells while leaving normal cells unharmed. The subcellular localization of apoptin appears to be crucial for this tumor-selective activity. In normal cells, apoptin resides in the cytoplasm, whereas in cancerous cells it translocates into the nucleus. The nuclear translocation of apoptin is largely controlled by its phosphorylation. In tumor cells, apoptin causes the nuclear accumulation of survival kinases including Akt and is phosphorylated by CDK2. Thereby, apoptin redirects survival signals into cell death responses. Apoptin also binds as a multimeric complex to DNA and interacts with several nuclear targets, such as the anaphase-promoting complex, resulting in a G2/M phase arrest. The proapoptotic signal of apoptin is then transduced from the nucleus to cytoplasm by Nur77, which triggers a p53-independent mitochondrial death pathway. In this review, we summarize recent discoveries of apoptin's mechanism of action that might provide intriguing insights for the development of novel tumor-selective anticancer drugs.


Subject(s)
Capsid Proteins/physiology , Chicken anemia virus/physiology , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Apoptosis/physiology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Chicken anemia virus/chemistry , Chicken anemia virus/genetics , Chicken anemia virus/pathogenicity , Chickens , Circoviridae Infections/pathology , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Genes, Viral , Models, Biological , Molecular Sequence Data , Phosphorylation , Poultry Diseases/pathology , Poultry Diseases/virology , Sarcoma, Avian/pathology
15.
Virus Genes ; 38(2): 259-62, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19125320

ABSTRACT

Chicken anemia virus (CAV) is the pathogen of chicken infectious anemia. To clarify the driving force in CAV evolution, we have detected positive selection in the structural protein gene VP1 by using maximum-likelihood models. Strong evidence was found that VP1 proteins were subject to the high rates of positive selection, and eight sites were identified to be under positive selection using the Bayes Empirical Bayesian method. Interestingly, four selected sites (amino acids 75, 125, 141, and 144) might be responsible for the attenuation exhibited. One selected site (amino acid 287) was connected with the virulence of CAV. This study provided some implication for the evolution of CAV, development of vaccines, and investigation into the structural and functional profiles of the VP1 protein.


Subject(s)
Capsid Proteins/genetics , Chicken anemia virus/genetics , Circoviridae Infections/veterinary , Poultry Diseases/virology , Selection, Genetic , Animals , Chicken anemia virus/isolation & purification , Chicken anemia virus/pathogenicity , Chickens , Circoviridae Infections/virology , Cluster Analysis , Evolution, Molecular , Models, Statistical , Polymorphism, Genetic , Sequence Homology , Virulence
16.
Virus Res ; 137(1): 1-15, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18656506

ABSTRACT

Current clinical studies on human annelloviruses infections are directed towards finding an associated disease. In this review we have emphasized the many similarities between human anellovirus and avian circoviruses and the cell and tissue types infected by these pathogens. We have done this in order to explore whether knowledge acquired from natural and experimental avian infections could reflect and be extrapolated to the less well-characterized human annellovirus infections. The knowledge gained from the avian system may provide suggestions for decoding the enigmatic human anellovirus infections, and finding the specific disease or diseases caused by these human anellovirus infections. Each additional parallelism between chicken anemia virus (CAV) and Torque teno virus (TTV) further strengthens this premise. As we have seen information from human infections can also be used to better understand avian infections as well. Increased attention must be focused on the "hidden" or unrecognized, seemingly asymptomatic effects of circovirus and anellovirus infections. Understanding the facilitating effect of these infections on disease progression caused by other pathogens may help to explain differences in outcome of complicated poultry and human diseases. The final course of a pathogenic infection is determined by variations in the state of health of the host before, during and after contact with a pathogen, in addition to the phenotype of the pathogen and host. The health burden of circoviridae and anellovirus infections may be underestimated, due to lack of awareness of the need to search past the predominant clinical effect of identified pathogens and look for modulation of cellular-based immunity caused by co-infecting circoviruses, and by analogy, human anneloviruses.


Subject(s)
Anelloviridae/physiology , Chicken anemia virus/physiology , Circoviridae Infections/veterinary , DNA Virus Infections/virology , Poultry Diseases/virology , Anelloviridae/pathogenicity , Animals , Chicken anemia virus/immunology , Chicken anemia virus/pathogenicity , Circoviridae Infections/epidemiology , Circoviridae Infections/immunology , Circoviridae Infections/transmission , DNA Virus Infections/complications , DNA Virus Infections/epidemiology , DNA Virus Infections/transmission , HIV Infections/complications , Humans , Neoplasms/complications , Poultry , Poultry Diseases/epidemiology , Poultry Diseases/immunology , Poultry Diseases/transmission
17.
Virus Res ; 132(1-2): 152-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18177972

ABSTRACT

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.


Subject(s)
Chicken anemia virus/isolation & purification , Circoviridae Infections/veterinary , Feathers/virology , Poultry Diseases/transmission , Animals , Antibodies, Viral/blood , Chicken anemia virus/genetics , Chicken anemia virus/immunology , Chicken anemia virus/pathogenicity , Chickens/growth & development , Chickens/virology , Circoviridae Infections/immunology , Circoviridae Infections/transmission , Circoviridae Infections/virology , Epithelium/pathology , Epithelium/virology , Feathers/chemistry , Feathers/pathology , Histology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Mardivirus/genetics , Mardivirus/immunology , Mardivirus/isolation & purification , Marek Disease/immunology , Marek Disease/virology , Polymerase Chain Reaction , Poultry Diseases/immunology , Poultry Diseases/virology
18.
Wei Sheng Wu Xue Bao ; 47(5): 894-8, 2007 Oct.
Article in Chinese | MEDLINE | ID: mdl-18062269

ABSTRACT

A field strain C14 of chicken infectious anemia virus (CAV) was isolated from a 14 day-old broiler flock with growth runting syndromes. Antibody reactions to inactivated vaccines to avian influenza viruses (AIV) were suppressed in SPF chickens inoculated with C14 strain CAV at 1 day-old. Also C14 strain CAV and reticuloendotheliosis Virus demonstrated a synergism in immunosuppression when chickens were infected with both virus. The viral genomic DNA was amplified by PCR in 3 overlapped fragments and PCR products were cloned into T-vector plasmid for sequencing. The sequencing results indicated that the total genome of C14 strain CAV was 2298bp, it contained 3 overlapped ORF and 1 non-coding regulation fragment. Its whole genome had 97.2% - 99.2% of homogeneity to other several published CAV reference strains. Sequence data indicated that there are many motifs in the non-coding area of about 400bp as the binding sites for transcriptional factors. All these motifs were very conservative. There were some mutations in 3 genes VP1, VP2 and VP3. Relatively, VP1 was less conservative than VP2 and VP3. Among different strains, mutations in these 3 genes were not correlated.


Subject(s)
Chicken anemia virus/genetics , Chicken anemia virus/pathogenicity , Chickens/virology , Genome, Viral , Amino Acid Sequence , Animals , Capsid Proteins/chemistry , Capsid Proteins/genetics , Sequence Analysis, DNA
19.
J Gen Virol ; 88(Pt 8): 2168-2175, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17622619

ABSTRACT

Chicken anemia virus (CAV) is a significant immunosuppressive pathogen of chickens, but relatively little is known about the effect of specific mutations on its virulence. In order to study the virulence of CAV, an infection model was developed in embryos. Significant growth depression, measured as a reduction in mean body weight, was found for wild-type CAV infection. Infection with wild-type CAV resulted in a significant reduction in thymic and splenic weights and consistently produced severe lesions in the thymus, spleen and bone marrow, as well as haemorrhages. CAVs mutated in the VP2 gene were infectious for embryos, but were highly attenuated with respect to growth depression and CAV-specific pathology. Relative to wild-type infection, viruses Mut C86R, Mut R101G, Mut H103Y, Mut R129G, Mut Q131P, Mut R/K/K150/151/152G/A/A, Mut D/E161/162G/G and Mut E186G were highly attenuated, and viruses Mut L163P and Mut D169G were moderately attenuated. Attenuation of the ability to produce lesions was found consistently for the thymus, spleen and bone marrow, thymic and splenic weights, and for CAV-induced haemorrhage. There was no growth depression associated with infection by the group of highly attenuated mutant viruses and a moderate reduction in mean body weight was only found for virus Mut L163P. These findings show that mutations in the VP2 gene can reduce the virulence of CAV and these mutant viruses may have value as vaccine candidates.


Subject(s)
Capsid Proteins/genetics , Chicken anemia virus , Circoviridae Infections/veterinary , Poultry Diseases/pathology , Animals , Bursa of Fabricius/pathology , Chick Embryo , Chicken anemia virus/genetics , Chicken anemia virus/pathogenicity , Chickens , Circoviridae Infections/pathology , Mutagenesis, Site-Directed , Organ Size , Spleen/pathology , Thymus Gland/pathology , Virulence
20.
Avian Dis ; 51(1): 66-77, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17461269

ABSTRACT

Our previous genetic characterization of chicken anemia virus (CAV) in commercial broiler chickens in Alabama revealed a previously undetected polymorphism: a glutamine codon at VP1 position 22, in 7 of the 14 sequences. The novel glutamine codon was always found in association with a VP1 "hypervariable region" identical to CAV field isolates that replicate poorly in culture. The complete genome of CAV73, representative of the sequences with the novel polymorphism, was generated from cloned polymerase chain reaction (PCR) fragments amplified directly from naturally infected tissues. CAV73 had been detected in 31-day-old broilers submitted for examination for reasons unrelated to anemia. After electroporation of the cloned genomes into MDCC-CU147 lymphoblastoid cells, the regenerated CAV caused the culture to fail within 9 days, and the medium contained 5 X 10(6) TCID50 CAV/ml. Use of MDCC-CU147 cells was essential, as identical electroporation of MDCC-MSB1 cells failed to generate CAV able to destroy the culture within 8 wk. Regenerated CAV73 produced anemia and severe lymphocytic depletion of the thymus when inoculated into susceptible 3-day-old chickens and was reisolated from these chickens. Furthermore, it replicated in low- and high-passage MDCC-MSB1 cells similarly to a low-passage CAV field isolate that contains a different VP 1 "hypervariable region." The regeneration of CAV from PCR products directly from naturally infected carcasses, as performed in this study, provides a tool for the evaluation of distinct genetic polymorphisms that may be detected in specimens where infective virions are no longer available. Our results also provide some insight into the differential susceptibility of cell lines for low-passage CAV field isolates.


Subject(s)
Chicken anemia virus/genetics , Chicken anemia virus/isolation & purification , Chickens/virology , Circoviridae Infections/veterinary , Polymerase Chain Reaction/veterinary , Poultry Diseases/virology , Animals , Base Sequence , Cell Line , Chicken anemia virus/pathogenicity , Circoviridae Infections/virology , DNA, Viral/chemistry , Liver/virology , Thymus Gland/virology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL