Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Gene Ther ; 16(8): 625-37, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19197322

RESUMO

Preclinical biodistribution studies with INGN 007, an oncolytic adenovirus (Ad) vector, supporting an early stage clinical trial were conducted in Syrian hamsters, which are permissive for Ad replication, and mice, which are a standard model for assessing toxicity and biodistribution of replication-defective (RD) Ad vectors. Vector dissemination and pharmacokinetics following intravenous administration were examined by real-time PCR in nine tissues and blood at five time points spanning 1 year. Select organs were also examined for the presence of infectious vector/virus. INGN 007 (VRX-007), wild-type Ad5 and AdCMVpA (an RD vector) were compared in the hamster model, whereas only INGN 007 was examined in mice. DNA of all vectors was widely disseminated early after injection, but decayed rapidly in most organs. In the hamster model, DNA of INGN 007 and Ad5 was more abundant than that of the RD vector AdCMVpA at early times after injection, but similar levels were seen later. An increased level of INGN 007 and Ad5 DNA but not AdCMVpA DNA in certain organs early after injection, and the presence of infectious INGN 007 and Ad5 in lung and liver samples at early times after injection, strongly suggests that replication of INGN 007 and Ad5 occurred in several Syrian hamster organs. There was no evidence of INGN 007 replication in mice. In addition to providing important information about INGN 007, the results underscore the utility of the Syrian hamster as a permissive immunocompetent model for Ad5 pathogenesis and oncolytic Ad vectors.


Assuntos
Adenoviridae/fisiologia , Vetores Genéticos/farmacocinética , Animais , Cricetinae , DNA Viral/isolamento & purificação , Feminino , Terapia Genética , Vetores Genéticos/administração & dosagem , Injeções Intravenosas , Fígado/virologia , Pulmão/virologia , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Neoplasias/terapia , Vírus Oncolíticos , Especificidade da Espécie , Replicação Viral
2.
Cancer Gene Ther ; 16(8): 644-54, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19197324

RESUMO

Oncolytic (replication-competent) adenoviruses as anticancer agents provide new, promising tools to fight cancer. In support of a Phase I clinical trial, here we report safety data with INGN 007 (VRX-007), an oncolytic adenovirus with increased anti-tumor efficacy due to overexpression of the adenovirus-encoded ADP protein. Wild-type adenovirus type 5 (Ad5) and a replication-defective version of Ad5 were also studied as controls. A parallel study investigating the biodistribution of these viruses is described elsewhere in this issue. The toxicology experiments were conducted in two species, the Syrian hamster, which is permissive for INGN 007 and Ad5 replication and the poorly permissive mouse. The studies demonstrated that the safety profile of INGN 007 is similar to Ad5. Both viruses caused transient liver damage upon intravenous injection that resolved by 28 days post-infection. The No-Observable-Adverse-Effect-Level (NOAEL) for INGN 007 in hamsters was 3 x 10(10) viral particles per kg. In hamsters, the replication-defective vector caused less toxicity, indicating that replication of Ad vectors in the host is an important factor in pathogenesis. With mice, INGN 007 and Ad5 caused toxicity comparable to the replication-defective adenovirus vector. Partially based on these results, the FDA granted permission to enter into a Phase I clinical trial with INGN 007.


Assuntos
Adenoviridae/fisiologia , Vetores Genéticos/efeitos adversos , Proteínas E3 de Adenovirus/biossíntese , Animais , Contagem de Células Sanguíneas , Linhagem Celular , Cricetinae , Eritropoese , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intravenosas , Fígado/patologia , Fígado/virologia , Mesocricetus , Camundongos , Camundongos Endogâmicos C57BL , Vírus Oncolíticos , Replicação Viral
3.
J Virol ; 75(19): 8875-87, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11533151

RESUMO

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) induces apoptosis through two receptors, TRAIL-R1 (also known as death receptor 4) and TRAIL-R2 (also known as death receptor 5), that are members of the TNF receptor superfamily of death domain-containing receptors. We show that human adenovirus type 5 encodes three proteins, named RID (previously named E3-10.4K/14.5K), E3-14.7K, and E1B-19K, that independently inhibit TRAIL-induced apoptosis of infected human cells. This conclusion was derived from studies using wild-type adenovirus, adenovirus replication-competent mutants that lack one or more of the RID, E3-14.7K, and E1B-19K genes, and adenovirus E1-minus replication-defective vectors that express all E3 genes, RID plus E3-14.7K only, RID only, or E3-14.7K only. RID inhibits TRAIL-induced apoptosis when cells are sensitized to TRAIL either by adenovirus infection or treatment with cycloheximide. RID induces the internalization of TRAIL-R1 from the cell surface, as shown by flow cytometry and indirect immunofluorescence for TRAIL-R1. TRAIL-R1 was internalized in distinct vesicles which are very likely to be endosomes and lysosomes. TRAIL-R1 is degraded, as indicated by the disappearance of the TRAIL-R1 immunofluorescence signal. Degradation was inhibited by bafilomycin A1, a drug that prevents acidification of vesicles and the sorting of receptors from late endosomes to lysosomes, implying that degradation occurs in lysosomes. RID was also shown previously to internalize and degrade another death domain receptor, Fas, and to prevent apoptosis through Fas and the TNF receptor. RID was shown previously to force the internalization and degradation of the epidermal growth factor receptor. E1B-19K was shown previously to block apoptosis through Fas, and both E1B-19K and E3-14.7K were found to prevent apoptosis through the TNF receptor. These findings suggest that the receptors for TRAIL, Fas ligand, and TNF play a role in limiting virus infections. The ability of adenovirus to inhibit killing through these receptors may prolong acute and persistent infections.


Assuntos
Infecções por Adenoviridae/virologia , Adenoviridae/fisiologia , Proteínas E3 de Adenovirus/fisiologia , Apoptose/fisiologia , Glicoproteínas de Membrana/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Infecções por Adenoviridae/metabolismo , Infecções por Adenoviridae/patologia , Proteínas Reguladoras de Apoptose , Linhagem Celular , Humanos , Ligantes , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Ligante Indutor de Apoptose Relacionado a TNF , Replicação Viral
4.
Virology ; 263(2): 408-17, 1999 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-10544113

RESUMO

The long terminal repeat (LTR) is reported to be one of the most variable portions of the equine infectious anemia virus (EIAV) genome. To date, however, no information is available on the effects of observed sequence variations on viral replication properties, despite a widespread assumption of the biological importance of EIAV LTR variation. EIAV LTR sequence variability is confined mostly to a small portion of the enhancer within the U3 segment of the LTR. Analysis of published EIAV LTR sequences revealed six different types of LTR based on the pattern of putative transcription factor motifs within the variable region of the enhancer. To test directly the significance of LTR variation, the in vitro and in vivo replication properties of two variant LTR species were investigated using two isogenic viruses, EIAV(19-2) and EIAV(19-2-6A), differing only within the enhancer region. The results of these studies demonstrated that the two variants replicated with similar kinetics and to equal levels in cultured equine fibroblasts or in equine macrophage, the natural target cell of EIAV, even after prolonged serial passage in the latter cell type. Furthermore, EIAV(19-2) and EIAV(19-2-6A) variants demonstrated similar replication levels in experimentally infected ponies. However, ponies infected with EIAV(19-2-6A) exhibited a rapid switch in the prevalent LTR type, such that by 112 days postinfection, no original-LTR-type viruses were evident. This specific and rapid shift in LTR quasispecies indicates an in vivo selection that is not reflected in simple in vitro replication rates, suggesting undefined selection pressures in vivo that drive LTR variation during persistent EIAV infection.


Assuntos
Variação Genética/genética , Vírus da Anemia Infecciosa Equina/crescimento & desenvolvimento , Vírus da Anemia Infecciosa Equina/genética , Sequências Repetidas Terminais/genética , Replicação Viral/genética , Animais , Sequência de Bases , Linhagem Celular , Elementos Facilitadores Genéticos/genética , Anemia Infecciosa Equina/sangue , Anemia Infecciosa Equina/virologia , Fibroblastos/virologia , Variação Genética/fisiologia , Cavalos/virologia , Vírus da Anemia Infecciosa Equina/patogenicidade , Macrófagos/virologia , Dados de Sequência Molecular , Mutação/genética , RNA Viral/análise , RNA Viral/genética , Elementos de Resposta/genética , Seleção Genética , Inoculações Seriadas , Sequências Repetidas Terminais/fisiologia , Fatores de Transcrição/metabolismo , Viremia/sangue , Viremia/virologia
5.
Curr Opin Immunol ; 11(4): 380-6, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10448144

RESUMO

Adenoviruses encode proteins that block responses to interferons, intrinsic cellular apoptosis, killing by CD8(+) cytotoxic T lymphocytes and killing by the death ligands TNF, Fas ligand and TRAIL. The viral proteins are believed to prolong acute and persistent adenovirus infections. The proteins may prove useful in protecting adenovirus gene therapy vectors and transplanted cells from the immune system.


Assuntos
Adenoviridae/imunologia , Adenoviridae/genética , Proteínas E1A de Adenovirus/fisiologia , Proteínas E1B de Adenovirus/fisiologia , Animais , Apoptose , Terapia Genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Linfócitos T Citotóxicos/imunologia
6.
Nature ; 392(6677): 726-30, 1998 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-9565035

RESUMO

DNA viruses have evolved elaborate mechanisms to overcome host antiviral defences. In adenovirus-infected cells, programmed cell death (apoptosis) induced by the cytokine tumour necrosis factor (TNF) is inhibited by several adenovirus-encoded proteins. Occupation of the cell-surface receptor Fas, a member of the TNF-receptor superfamily that is expressed on most cell types, triggers apoptosis of that cell. Here we show that the adenovirus RID (for receptor internalization and degradation) protein complex, which is an inhibitor of TNF-induced apoptosis, mediates internalization of cell-surface Fas and its destruction inside lysosomes within the cell. Fas has not previously been shown to be internalized and then degraded. RID also mediates internalization of the receptor for epidermal growth factor, but it does not affect the transferrin receptor or class I antigens of the major histocompatibility complex. Removal of Fas from the surface of adenovirus-infected cells expressing RID may allow infected cells to resist Fas-mediated cell death and thus promote their survival.


Assuntos
Adenoviridae/fisiologia , Apoptose , Macrolídeos , Receptor fas/fisiologia , Proteínas E1B de Adenovirus , Animais , Antibacterianos/farmacologia , Linhagem Celular Transformada , Humanos , Camundongos , Mutação , Proteínas Virais
7.
J Virol ; 72(2): 1383-93, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9445039

RESUMO

An infectious nonpathogenic molecular clone (19-2-6A) of equine infectious anemia virus (EIAV) was modified by substitution of a 3.3-kbp fragment amplified by PCR techniques from a pathogenic variant (EIAV(PV)) of the cell culture-adapted strain of EIAV (EIAV(PR)). This substitution consisted of coding sequences for 77 amino acids at the carboxyl terminus of the integrase, the S1 (encoding the second exon of tat), S2, and S3 (encoding the second exon of rev) open reading frames, the complete env gene (including the first exon of rev), and the 3' long terminal repeat (LTR). Modified 19-2-6A molecular clones were designated EIAV(PV3.3), and infection of a single pony (678) with viruses derived from a mixture of five of these molecular clones induced clinical signs of acute equine infectious anemia (EIA) at 23 days postinfection (dpi). As a consequence of this initial study, a single molecular clone, EIAV(PV3.3#3) (redesignated EIAV(UK)), was selected for further study and inoculated into two ponies (613 and 614) and two horses (700 and 764). Pony 614 and the two horses developed febrile responses by 12 dpi, which was accompanied by a 48 to 64% reduction in platelet number, whereas pony 613 did not develop fever (40.6 degrees C) until 76 dpi. EIAV could be isolated from the plasma of these animals by 5 to 7 dpi, and all became seropositive for antibodies to this virus by 21 dpi. Analysis of the complete nucleotide sequence demonstrated that the 3.3-kbp 3' fragment of EIAV(UK) differed from the consensus sequence of EIAV(PV) by just a single amino acid residue in the second exon of the rev gene. Complete homology with the EIAV(PV) consensus sequence was observed in the hypervariable region of the LTR. However, EIAV(UK) was found to contain an unusual 68-bp nucleotide insertion/duplication in a normally conserved region of the LTR sequence. These results demonstrate that substitution of a 3.3-kbp fragment from the EIAV(PV) strain into the infectious nonpathogenic molecular clone 19-2-6A leads to the production of progeny virus particles with the ability to induce clinical signs of EIA. Therefore, EIAV(UK), which is the first pathogenic, cell culture-adapted molecular clone of EIAV to be described, should be of value in identifying viral determinants of pathogenicity.


Assuntos
Anemia Infecciosa Equina/virologia , Vírus da Anemia Infecciosa Equina/genética , Integrases/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sequência de Bases , Vírus da Anemia Infecciosa Equina/patogenicidade , Dados de Sequência Molecular , Alinhamento de Sequência , Virulência/genética
8.
Am J Vet Res ; 58(12): 1402-7, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9401688

RESUMO

OBJECTIVE: To characterize infection of bone marrow-derived macrophages (BMDM) with equine infectious anemia virus (EIAV) by determining virus production, effects on viability, and induction of cytokines. SAMPLE POPULATION: BMDM obtained from bone marrow of 6 clinically normal adult horses. PROCEDURE: BMDM were infected with EIAV at a multiplicity of infection of 8. Cell viability, percentage of cells with detectable viral protein, reverse transcriptase activity, and concentrations of infective virus (focus-forming units/ml), interleukin 6, and tumor necrosis factor-alpha were measured in culture supernatant samples obtained at various days after infection. RESULTS: Cell viability was decreased on day 4 and was maximally decreased on day 8. The number of cells with detectable viral protein and supernatant reverse transcriptase activity increased significantly on day 4 and increased until day 6. Virus concentration (focus-forming units per milliliter) peaked on day 4 after infection and was constant thereafter. Infection with EIAV caused significant induction of interleukin 6 production by BMDM. The maximal difference was seen on day 4 after infection. Control and infected BMDM produced only negligible amounts of tumor necrosis factor-alpha. CONCLUSIONS: BMDM are useful, as a cell population, to study the effects of infection with EIAV, including cell death and induction of interleukin 6 but not tumor necrosis factor-alpha production.


Assuntos
Medula Óssea/patologia , Vírus da Anemia Infecciosa Equina/fisiologia , Macrófagos/virologia , Análise de Variância , Animais , Medula Óssea/virologia , Sobrevivência Celular , Células Cultivadas , Efeito Citopatogênico Viral , Anemia Infecciosa Equina/imunologia , Anemia Infecciosa Equina/patologia , Cavalos , Interleucina-6/biossíntese , Macrófagos/metabolismo , Macrófagos/patologia , Microscopia Eletrônica/métodos , Microscopia Eletrônica/veterinária , DNA Polimerase Dirigida por RNA/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Proteínas Virais/metabolismo , Vírion/isolamento & purificação
9.
J Virol ; 71(5): 3840-52, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9094660

RESUMO

Equine infectious anemia virus (EIAV) provides a natural model system by which immunological control of lentivirus infections may be studied. To date, no detailed study addressing in parallel both the humoral and cellular immune responses induced in horses upon infection by EIAV has been conducted. Therefore, we initiated the first comprehensive characterization of the cellular and humoral immune responses during clinical progression from chronic disease to inapparent stages of EIAV infection. Using new analyses of antibody avidity and antibody epitope conformation dependence that had not been previously employed in this system, we observed that the humoral immune response to EIAV required a 6- to 8-month period in which to fully mature. During this time frame, EIAV-specific antibody evolved gradually from a population characterized by low-avidity, nonneutralizing, and predominantly linear epitope specificity to an antibody population with an avidity of moderate to high levels, neutralizing activity, and predominantly conformational epitope specificity. Analyses of the cell-mediated immune response to EIAV revealed CD4+ and CD8+ major histocompatibility complex-restricted, EIAV-specific cytotoxic T-lymphocyte (CTL) activity apparent within 3 to 4 weeks postinfection, temporally correlating with the resolution of the primary viremia. After resolution of the initial viremia, EIAV-specific CTL activity differed greatly among the experimentally infected ponies, with some animals having readily detectable CTL activity while others had little measurable CTL activity. Thus, in contrast to the initial viremia, it appeared that no single immune parameter correlated with the resolution of further viremic episodes. Instead, immune control of EIAV infection during the clinically inapparent stage of infection appears to rely on a complex combination of immune system mechanisms to suppress viral replication that effectively functions only after the immune system has evolved to a fully mature state 6 to 8 months postinfection. These findings strongly imply the necessity for candidate EIAV and other lentivirus vaccines to achieve this immune system maturation for efficacious immunological control of lentivirus challenge.


Assuntos
Anticorpos Antivirais/sangue , Anemia Infecciosa Equina/imunologia , Ativação Linfocitária , Animais , Afinidade de Anticorpos , Linfócitos T CD4-Positivos/imunologia , Cavalos , Imunoglobulina G/sangue , Complexo Principal de Histocompatibilidade , Conformação Proteica , Linfócitos T Citotóxicos/imunologia
10.
J Virol ; 70(6): 3346-54, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8648664

RESUMO

Equine infectious anemia virus (EIAV) provides a uniquely dynamic system in which to study the mechanism and role of genomic variation in lentiviral persistence and pathogenesis. We have used a Shetland pony model of infection to investigate the association of specific long terminal repeat (LTR) and env gene genomic sequences with the initiation of infection and the onset of disease. We analyzed viral RNA isolated from a pathogenic stock of virus (EIAV PV) and from plasma taken during the first disease episode from two ponies infected with EIAV PV. Overall sequence variation within gp90 was low in EIAV PV and only slightly higher in plasma virus samples isolated from ponies during the first disease episode. However, a high proportion of mutations were localized to the principal neutralizing domain in EIAV PV and to the principal neutralizing domain and the gp90 hypervariable region in the two pony-derived samples. The rate of fixation of mutations was analyzed and determined to be approximately 4 x 10(-2) mutations per site per year. Sequence diversity within the U3 region of the LTR was extremely low, which suggested that the previously reported hypervariability of this region may be a consequence of selection for replication of EIAV in different host cells. The predominant EIAV PV env and LTR sequences were used to construct chimeric viruses so that the contribution of these sequences to viral pathogenicity could be examined. The chimeras replicated in cultured equine monocytes to the same extent as the parental nonpathogenic virus and did not cause disease in Shetland ponies by 120 days postinfection, suggesting that the EIAV genomic determinants of pathogenesis are complex.


Assuntos
Anemia Infecciosa Equina/virologia , Genoma Viral , Vírus da Anemia Infecciosa Equina/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Cavalos , Dados de Sequência Molecular , Sequências Repetitivas de Ácido Nucleico , Proteínas Virais/química
11.
J Gen Virol ; 77 ( Pt 1): 109-18, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8558117

RESUMO

The attachment protein G of human respiratory syncytial (RS) virus is a type II transmembrane glycoprotein. A secreted from of the G protein is also produced. To examine the two distinct hydrophobic regions in the N-terminal 63 amino acids of G protein for their role(s) in membrane insertion and anchoring, transport to the cell surface, and secretion, G proteins that contained point mutations or deletions were synthesized by cell-free transcription-translation and in cells by expression from recombinant vaccinia virus vectors. A mutant protein lacking the entire major hydrophobic region (amino acids 38-63) was not glycosylated, not expressed on the cell surface, and not secreted, because it was not inserted into membranes. In contrast, deletion of the minor hydrophobic region (amino acids 23-31) had no detectable effect on membrane insertion or anchoring. These data provided direct evidence that amino acids 38-63 were necessary for membrane insertion and contained the signal/anchor domain of RS virus G protein. Mutant proteins that lacked either the N-terminal or the C-terminal half of this 26 residue hydrophobic region were inserted into membranes and processed to maturity, showing that either half of this region was sufficient for membrane insertion. However, these two mutant proteins were secreted more abundantly than wild-type G protein. We propose that their truncated hydrophobic domains interacted with membranes in a way that mimicked the N-terminal signal sequence of naturally secreted proteins, allowing proteolytic cleavage of the mutant proteins.


Assuntos
Proteína HN , Vírus Sincicial Respiratório Humano/fisiologia , Proteínas Virais de Fusão/fisiologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Sistema Livre de Células , Endopeptidases/metabolismo , Vetores Genéticos , Células HeLa , Humanos , Fusão de Membrana , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos , Coelhos , Proteínas Recombinantes , Vírus Sincicial Respiratório Humano/genética , Células Tumorais Cultivadas , Vaccinia virus/genética , Proteínas do Envelope Viral , Proteínas Virais de Fusão/genética , Proteínas Virais/genética
12.
J Virol ; 69(5): 2881-8, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7707512

RESUMO

As an important enzyme in DNA synthesis, dUTPase is present in a wide variety of organisms and viruses and has been identified as a component of the equine infectious anemia virus (EIAV) pol gene. The role of EIAV dUTPase, designated DU, in virus replication in vitro and in vivo was investigated with a recently described infectious molecular clone of EIAV. A deletion mutant that was deficient in dUTPase activity was constructed, and its replication kinetics was examined in fetal equine kidney (FEK) cells and primary equine bone marrow macrophage (EBMM) cells. In FEK cells, which are permissive for EIAV replication, the mutant virus replicated as well as the parental virus. In primary cultures of EBMM cells, which are primary targets of EIAV infection in vivo, the DU mutant showed delayed replication kinetics and replicated to a lower extent than did the parental virus. As the multiplicity of infection decreased, the difference between the parental and mutant viruses increased, such that at the lowest multiplicity of infection tested, there was over a 100-fold difference in virus production. The mutant virus was also much less cytopathic. The role of DU in replication in vivo was examined using a Shetland pony model of EIAV infection. Shetland ponies that were infected with the parental and mutant viruses showed transient virus RNA levels in plasma approximately 5 to 10 days postinfection. The peak virus levels in plasma (as measured by a quantitative reverse transcriptase PCR assay) were 10- to 100-fold lower in the mutant virus-infected animals than in the animals infected with the parental virus. However, ponies infected with the mutant virus mounted similar antibody responses despite the marked differences in virus replication. These studies demonstrate that EIAV DU is important for the efficient replication of the virus in macrophages in vitro and in vivo and suggests that variations in the DU sequence could markedly affect the biological and pathogenic properties of EIAV.


Assuntos
Deleção de Genes , Vírus da Anemia Infecciosa Equina/enzimologia , Vírus da Anemia Infecciosa Equina/genética , Pirofosfatases/genética , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/biossíntese , Sequência de Bases , Linhagem Celular , Efeito Citopatogênico Viral/genética , Efeito Citopatogênico Viral/fisiologia , Primers do DNA/genética , DNA Viral/genética , Anemia Infecciosa Equina/virologia , Genes pol , Cavalos , Vírus da Anemia Infecciosa Equina/fisiologia , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Pirofosfatases/deficiência , RNA Viral/sangue , RNA Viral/genética , Replicação Viral/genética , Replicação Viral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...