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










Base de datos
Intervalo de año de publicación
1.
Retrovirology ; 15(1): 38, 2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29769087

RESUMEN

BACKGROUND: Hosts are able to restrict viral replication to contain virus spread before adaptive immunity is fully initiated. Many viruses have acquired genes directly counteracting intrinsic restriction mechanisms. This phenomenon has led to a co-evolutionary signature for both the virus and host which often provides a barrier against interspecies transmission events. Through different mechanisms of action, but with similar consequences, spumaviral feline foamy virus (FFV) Bet and lentiviral feline immunodeficiency virus (FIV) Vif counteract feline APOBEC3 (feA3) restriction factors that lead to hypermutation and degradation of retroviral DNA genomes. Here we examine the capacity of vif to substitute for bet function in a chimeric FFV to assess the transferability of anti-feA3 factors to allow viral replication. RESULTS: We show that vif can replace bet to yield replication-competent chimeric foamy viruses. An in vitro selection screen revealed that an engineered Bet-Vif fusion protein yields suboptimal protection against feA3. After multiple passages through feA3-expressing cells, however, variants with optimized replication competence emerged. In these variants, Vif was expressed independently from an N-terminal Bet moiety and was stably maintained. Experimental infection of immunocompetent domestic cats with one of the functional chimeras resulted in seroconversion against the FFV backbone and the heterologous FIV Vif protein, but virus could not be detected unambiguously by PCR. Inoculation with chimeric virus followed by wild-type FFV revealed that repeated administration of FVs allowed superinfections with enhanced antiviral antibody production and detection of low level viral genomes, indicating that chimeric virus did not induce protective immunity against wild-type FFV. CONCLUSIONS: Unrelated viral antagonists of feA3 cellular restriction factors can be exchanged in FFV, resulting in replication competence in vitro that was attenuated in vivo. Bet therefore may have additional functions other than A3 antagonism that are essential for successful in vivo replication. Immune reactivity was mounted against the heterologous Vif protein. We conclude that Vif-expressing FV vaccine vectors may be an attractive tool to prevent or modulate lentivirus infections with the potential option to induce immunity against additional lentivirus antigens.


Asunto(s)
Productos del Gen vif/genética , Virus de la Inmunodeficiencia Felina/genética , Proteínas de los Retroviridae/genética , Spumavirus/genética , Vacunas Virales/genética , Replicación Viral , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Gatos , Línea Celular , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Orden Génico , Productos del Gen gag/metabolismo , Genoma Viral , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Virus de la Inmunodeficiencia Felina/inmunología , Recombinación Genética , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/virología , Spumavirus/inmunología , Carga Viral , Vacunas Virales/inmunología
2.
MAbs ; 10(2): 290-303, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29173063

RESUMEN

Novel biotherapeutic glycoproteins, like recombinant monoclonal antibodies (mAbs) are widely used for the treatment of numerous diseases. The N-glycans attached to the constant region of an antibody have been demonstrated to be crucial for the biological efficacy. Even minor modifications of the N-glycan structure can dictate the potency of IgG effector functions such as the antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Here, we present the development of a glycoengineered CHO-K1 host cell line (HCL), stably expressing ß1,4-N-Acetylglucoseaminyltransferase III (GnT-III) and α-mannosidase II (Man-II), for the expression of a-fucosylated antibodies with enhanced Fc-mediated effector function. Glycoengineered HCLs were generated in a two-step strategy, starting with generating parental HCLs by stable transfection of CHO-K1 cells with GnT-III and Man-II. In a second step, parental HCLs were stably transfected a second time with these two transgenes to increase their copy number in the genetic background. Generated glycoengineered CHO-K1 cell lines expressing two different mAbs deliver antibody products with a content of more than 60% a-fucosylated glycans. In-depth analysis of the N-glycan structure revealed that the majority of the Fc-attached glycans of the obtained mAbs were of complex bisected type. Furthermore, we showed the efficient use of FcγRIIIa affinity chromatography as a novel method for the fast assessment of the mAbs a-fucosylation level. By testing different cultivation conditions for the pre-glycoengineered recombinant CHO-K1 clones, we identified key components essential for the production of a-fucosylated mAbs. The prevalent effect could be attributed to the trace element manganese, which leads to a strong increase of a-fucosylated complex- and hybrid-type glycans. In conclusion, the novel pre-glycoengineered CHO-K1 HCL can be used for the production of antibodies with high ratios of a-fucosylated Fc-attached N-glycans. Application of our newly developed FcγRIIIa affinity chromatography method during cell line development and use of optimized cultivation conditions can ultimately support the efficient development of a-fucosylated mAbs.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Células CHO , Ingeniería Genética/métodos , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Cricetulus , Humanos , N-Acetilglucosaminiltransferasas/inmunología , Ingeniería de Proteínas/métodos , Ratas , Receptores de IgG/inmunología , Transfección , alfa-Manosidasa/inmunología
3.
J Virol ; 90(22): 10193-10208, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27581978

RESUMEN

Lentiviruses have evolved the Vif protein to counteract APOBEC3 (A3) restriction factors by targeting them for proteasomal degradation. Previous studies have identified important residues in the interface of human immunodeficiency virus type 1 (HIV-1) Vif and human APOBEC3C (hA3C) or human APOBEC3F (hA3F). However, the interaction between primate A3C proteins and HIV-1 Vif or natural HIV-1 Vif variants is still poorly understood. Here, we report that HIV-1 Vif is inactive against A3Cs of rhesus macaques (rhA3C), sooty mangabey monkeys (smmA3C), and African green monkeys (agmA3C), while HIV-2, African green monkey simian immunodeficiency virus (SIVagm), and SIVmac Vif proteins efficiently mediate the depletion of all tested A3Cs. We identified that residues N/H130 and Q133 in rhA3C and smmA3C are determinants for this HIV-1 Vif-triggered counteraction. We also found that the HIV-1 Vif interaction sites in helix 4 of hA3C and hA3F differ. Vif alleles from diverse HIV-1 subtypes were tested for degradation activities related to hA3C. The subtype F-1 Vif was identified to be inactive for degradation of hA3C and hA3F. The residues that determined F-1 Vif inactivity in the degradation of A3C/A3F were located in the C-terminal region (K167 and D182). Structural analysis of F-1 Vif revealed that impairing the internal salt bridge of E171-K167 restored reduction capacities to A3C/A3F. Furthermore, we found that D101 could also form an internal interaction with K167. Replacing D101 with glycine and R167 with lysine in NL4-3 Vif impaired its counteractivity to A3F and A3C. This finding indicates that internal interactions outside the A3 binding region in HIV-1 Vif influence the capacity to induce degradation of A3C/A3F. IMPORTANCE: The APOBEC3 restriction factors can serve as potential barriers to lentiviral cross-species transmissions. Vif proteins from lentiviruses counteract APOBEC3 by proteasomal degradation. In this study, we found that monkey-derived A3C, rhA3C and smmA3C, were resistant to HIV-1 Vif. This was determined by A3C residues N/H130 and Q133. However, HIV-2, SIVagm, and SIVmac Vif proteins were found to be able to mediate the depletion of all tested primate A3C proteins. In addition, we identified a natural HIV-1 Vif (F-1 Vif) that was inactive in the degradation of hA3C/hA3F. Here, we provide for the first time a model that explains how an internal salt bridge of E171-K167-D101 influences Vif-mediated degradation of hA3C/hA3F. This finding provides a novel way to develop HIV-1 inhibitors by targeting the internal interactions of the Vif protein.


Asunto(s)
Citidina Desaminasa/metabolismo , Productos del Gen vif/metabolismo , VIH-1/metabolismo , Virus de la Inmunodeficiencia de los Simios/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Sitios de Unión , Línea Celular , Células HEK293 , Infecciones por VIH/virología , VIH-2/metabolismo , Humanos , Lentivirus/metabolismo , Macaca mulatta , Unión Proteica/fisiología
4.
Clin Cancer Res ; 22(17): 4417-27, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27117182

RESUMEN

PURPOSE: CEA TCB (RG7802, RO6958688) is a novel T-cell bispecific antibody, engaging CD3ε upon binding to carcinoembryonic antigen (CEA) on tumor cells. Containing an engineered Fc region, conferring an extended blood half-life while preventing side effects due to activation of innate effector cells, CEA TCB potently induces tumor lysis in mouse tumors. Here we aimed to characterize the pharmacokinetic profile, the biodistribution, and the mode of action of CEA TCB by combining in vitro and in vivo fluorescence imaging readouts. EXPERIMENTAL DESIGN: CEA-expressing tumor cells (LS174T) and human peripheral blood mononuclear cells (PBMC) were cocultured in vitro or cografted into immunocompromised mice. Fluorescence reflectance imaging and intravital 2-photon (2P) microscopy were employed to analyze in vivo tumor targeting while in vitro confocal and intravital time-lapse imaging were used to assess the mode of action of CEA TCB. RESULTS: Fluorescence reflectance imaging revealed increased ratios of extravascular to vascular fluorescence signals in tumors after treatment with CEA TCB compared with control antibody, suggesting specific targeting, which was confirmed by intravital microscopy. Confocal and intravital 2P microscopy showed CEA TCB to accelerate T-cell-dependent tumor cell lysis by inducing a local increase of effector to tumor cell ratios and stable crosslinking of multiple T cells to individual tumor cells. CONCLUSIONS: Using optical imaging, we demonstrate specific tumor targeting and characterize the mode of CEA TCB-mediated target cell lysis in a mouse tumor model, which supports further clinical evaluation of CEA TCB. Clin Cancer Res; 22(17); 4417-27. ©2016 AACRSee related commentary by Teijeira et al., p. 4277.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antígeno Carcinoembrionario/inmunología , Citotoxicidad Inmunológica , Imagen Molecular , Neoplasias/diagnóstico por imagen , Neoplasias/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Biespecíficos/farmacología , Especificidad de Anticuerpos/inmunología , Antineoplásicos Inmunológicos/metabolismo , Antineoplásicos Inmunológicos/farmacología , Biomarcadores , Comunicación Celular/inmunología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Microscopía Confocal , Imagen Molecular/métodos , Neoplasias/metabolismo , Neoplasias/terapia , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Factores de Tiempo , Distribución Tisular
5.
Viruses ; 4(11): 3132-61, 2012 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-23202519

RESUMEN

Human immunodeficiency virus-1 (HIV-1) dynamics reflect an intricate balance within the viruses’ host. The virus relies on host replication factors, but must escape or counter its host’s antiviral restriction factors. The interaction between the HIV-1 protein Vif and many cellular restriction factors from the APOBEC3 protein family is a prominent example of this evolutionary arms race. The viral infectivity factor (Vif) protein largely neutralizes APOBEC3 proteins, which can induce in vivo hypermutations in HIV-1 to the extent of lethal mutagenesis, and ensures the production of viable virus particles. HIV-1 also uses the APOBEC3-Vif interaction to modulate its own mutation rate in harsh or variable environments, and it is a model of adaptation in a coevolutionary setting. Both experimental evidence and the substantiation of the underlying dynamics through coevolutionary models are presented as complementary views of a coevolutionary arms race.


Asunto(s)
Citosina Desaminasa/metabolismo , VIH-1/genética , VIH-1/metabolismo , Mutagénesis , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Desaminasas APOBEC , Evolución Biológica , Citidina Desaminasa , Farmacorresistencia Viral , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , Interacciones Huésped-Patógeno , Humanos , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética
6.
Viruses ; 3(10): 1986-2005, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22069525

RESUMEN

Lentiviruses are known for their narrow cell- and species-tropisms, which are determined by cellular proteins whose absence or presence either support viral replication (dependency factors, cofactors) or inhibit viral replication (restriction factors). Similar to Human immunodeficiency virus type 1 (HIV-1), the cat lentivirus Feline immunodeficiency virus (FIV) is sensitive to recently discovered cellular restriction factors from non-host species that are able to stop viruses from replicating. Of particular importance are the cellular proteins APOBEC3, TRIM5α and tetherin/BST-2. In general, lentiviruses counteract or escape their species' own variant of the restriction factor, but are targeted by the orthologous proteins of distantly related species. Most of the knowledge regarding lentiviral restriction factors has been obtained in the HIV-1 system; however, much less is known about their effects on other lentiviruses. We describe here the molecular mechanisms that explain how FIV maintains its replication in feline cells, but is largely prevented from cross-species infections by cellular restriction factors.


Asunto(s)
Virus de la Inmunodeficiencia Felina/patogenicidad , Infecciones por Lentivirus/inmunología , Replicación Viral/inmunología , Animales , Gatos , Línea Celular , Especificidad del Huésped , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Virus de la Inmunodeficiencia Felina/inmunología , Virus de la Inmunodeficiencia Felina/fisiología , Infecciones por Lentivirus/virología
7.
J Virol ; 84(14): 7312-24, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20444897

RESUMEN

To get more insight into the role of APOBEC3 (A3) cytidine deaminases in the species-specific restriction of feline immunodeficiency virus (FIV) of the domestic cat, we tested the A3 proteins present in big cats (puma, lion, tiger, and lynx). These A3 proteins were analyzed for expression and sensitivity to the Vif protein of FIV. While A3Z3s and A3Z2-Z3s inhibited Deltavif FIV, felid A3Z2s did not show any antiviral activity against Deltavif FIV or wild-type (wt) FIV. All felid A3Z3s and A3Z2-Z3s were sensitive to Vif of the domestic cat FIV. Vif also induced depletion of felid A3Z2s. Tiger A3s showed a moderate degree of resistance against the Vif-mediated counter defense. These findings may imply that the A3 restriction system does not play a major role to prevent domestic cat FIV transmission to other Felidae. In contrast to the sensitive felid A3s, many nonfelid A3s actively restricted wt FIV replication. To test whether Vif(FIV) can protect also the distantly related human immunodeficiency virus type 1 (HIV-1), a chimeric HIV-1.Vif(FIV) was constructed. This HIV-1.Vif(FIV) was replication competent in nonpermissive feline cells expressing human CD4/CCR5 that did not support the replication of wt HIV-1. We conclude that the replication of HIV-1 in some feline cells is inhibited only by feline A3 restriction factors and the absence of the appropriate receptor or coreceptor.


Asunto(s)
Citosina Desaminasa/metabolismo , Felidae/metabolismo , Felidae/virología , Síndrome de Inmunodeficiencia Adquirida del Felino/virología , Productos del Gen vif/metabolismo , Virus de la Inmunodeficiencia Felina/metabolismo , Isoenzimas/metabolismo , Animales , Gatos , Línea Celular , Citosina Desaminasa/genética , Felidae/genética , Productos del Gen vif/genética , Humanos , Virus de la Inmunodeficiencia Felina/genética , Isoenzimas/genética , Empalme del ARN , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
8.
J Virol ; 83(15): 7547-59, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19458006

RESUMEN

The mammalian APOBEC3 (A3) proteins comprise a multigene family of cytidine deaminases that act as potent inhibitors of retroviruses and retrotransposons. The A3 locus on the chromosome 28 of the horse genome contains multiple A3 genes: two copies of A3Z1, five copies of A3Z2, and a single copy of A3Z3, indicating a complex evolution of multiple gene duplications. We have cloned and analyzed for expression the different equine A3 genes and examined as well the subcellular distribution of the corresponding proteins. Additionally, we have tested the functional antiretroviral activity of the equine and of several of the human and nonprimate A3 proteins against the Equine infectious anemia virus (EIAV), the Simian immunodeficiency virus (SIV), and the Adeno-associated virus type 2 (AAV-2). Hematopoietic cells of horses express at least five different A3s: A3Z1b, A3Z2a-Z2b, A3Z2c-Z2d, A3Z2e, and A3Z3, whereas circulating macrophages, the natural target of EIAV, express only part of the A3 repertoire. The five A3Z2 tandem copies arose after three consecutive, recent duplication events in the horse lineage, after the split between Equidae and Carnivora. The duplicated genes show different antiviral activities against different viruses: equine A3Z3 and A3Z2c-Z2d are potent inhibitors of EIAV while equine A3Z1b, A3Z2a-Z2b, A3Z2e showed only weak anti-EIAV activity. Equine A3Z1b and A3Z3 restricted AAV and all equine A3s, except A3Z1b, inhibited SIV. We hypothesize that the horse A3 genes are undergoing a process of subfunctionalization in their respective viral specificities, which might provide the evolutionary advantage for keeping five copies of the original gene.


Asunto(s)
Citidina Desaminasa/metabolismo , Anemia Infecciosa Equina/enzimología , Virus de la Anemia Infecciosa Equina/fisiología , Familia de Multigenes , Desaminasas APOBEC , Animales , Línea Celular , Citidina Desaminasa/genética , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Anemia Infecciosa Equina/virología , Expresión Génica , Células HeLa , Caballos , Humanos , Virus de la Anemia Infecciosa Equina/genética , Datos de Secuencia Molecular
9.
J Biol Chem ; 284(9): 5819-26, 2009 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-19074429

RESUMEN

The APOBEC3 cytidine deaminases are part of the intrinsic defense of cells against retroviruses. Lentiviruses and spumaviruses have evolved essential accessory proteins, Vif and Bet, respectively, which counteract the APOBEC3 proteins. We show here that Bet of the Prototype foamy virus inhibits the antiviral APOBEC3C activity by a mechanism distinct to Vif: Bet forms a complex with APOBEC3C without inducing its degradation. Bet abolished APOBEC3C dimerization as shown by coimmunoprecipitation and cross-linking experiments. These findings implicate a physical interaction between Bet and the APOBEC3C. Subsequently, we identified the Bet interaction domain in human APOBEC3C in the predicted APOBEC3C dimerization site. Taken together, these data support the hypothesis that Bet inhibits incorporation of APOBEC3Cs into retroviral particles. Bet likely achieves this by trapping APOBEC3C protein in complexes rendering them unavailable for newly generated viruses due to direct immobilization.


Asunto(s)
Citidina Desaminasa/antagonistas & inhibidores , Citidina Desaminasa/metabolismo , Provirus/genética , Proteínas de los Retroviridae/fisiología , Desaminasa APOBEC-3G , Animales , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Reactivos de Enlaces Cruzados , Dimerización , Productos del Gen vif/fisiología , Humanos , Immunoblotting , Inmunoprecipitación , Riñón/citología , Riñón/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Macaca mulatta , Macaca nemestrina , Pan troglodytes , Virus Espumoso de los Simios , Transfección , Ensamble de Virus , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
10.
Genome Biol ; 9(3): R48, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18315870

RESUMEN

BACKGROUND: Over the past years a variety of host restriction genes have been identified in human and mammals that modulate retrovirus infectivity, replication, assembly, and/or cross-species transmission. Among these host-encoded restriction factors, the APOBEC3 (A3; apolipoprotein B mRNA-editing catalytic polypeptide 3) proteins are potent inhibitors of retroviruses and retrotransposons. While primates encode seven of these genes (A3A to A3H), rodents carry only a single A3 gene. RESULTS: Here we identified and characterized several A3 genes in the genome of domestic cat (Felis catus) by analyzing the genomic A3 locus. The cat genome presents one A3H gene and three very similar A3C genes (a-c), probably generated after two consecutive gene duplications. In addition to these four one-domain A3 proteins, a fifth A3, designated A3CH, is expressed by read-through alternative splicing. Specific feline A3 proteins selectively inactivated only defined genera of feline retroviruses: Bet-deficient feline foamy virus was mainly inactivated by feA3Ca, feA3Cb, and feA3Cc, while feA3H and feA3CH were only weakly active. The infectivity of Vif-deficient feline immunodeficiency virus and feline leukemia virus was reduced only by feA3H and feA3CH, but not by any of the feA3Cs. Within Felidae, A3C sequences show significant adaptive selection, but unexpectedly, the A3H sequences present more sites that are under purifying selection. CONCLUSION: Our data support a complex evolutionary history of expansion, divergence, selection and individual extinction of antiviral A3 genes that parallels the early evolution of Placentalia, becoming more intricate in taxa in which the arms race between host and retroviruses is harsher.


Asunto(s)
Empalme Alternativo , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Evolución Molecular , Variación Genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Gatos/metabolismo , Gatos/virología , Citosina Desaminasa/química , Amplificación de Genes , Expresión Génica , Orden Génico , Genoma , Datos de Secuencia Molecular , Filogenia , Retroviridae/fisiología , Replicación Viral
11.
J Virol ; 81(13): 7048-60, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17459941

RESUMEN

The productive replication of human immunodeficiency virus type 1 (HIV-1) occurs exclusively in defined cells of human or chimpanzee origin, explaining why heterologous animal models for HIV replication, pathogenesis, vaccination, and therapy are not available. This lack of an animal model for HIV-1 studies prompted us to examine the susceptibility of feline cells in order to evaluate the cat (Felis catus) as an animal model for studying HIV-1. Here, we report that feline cell lines harbor multiple restrictions with respect to HIV-1 replication. The feline CD4 receptor does not permit virus infection. Feline T-cell lines MYA-1 and FeT-1C showed postentry restrictions resulting in low HIV-1 luciferase reporter activity and low expression of viral Gag-Pol proteins when pseudotyped vectors were used. Feline fibroblastic CrFK and KE-R cells, expressing human CD4 and CCR5, were very permissive for viral entry and HIV-long terminal repeat-driven expression but failed to support spreading infection. KE-R cells displayed a profound block with respect to release of HIV-1 particles. In contrast, CrFK cells allowed very efficient particle production; however, the CrFK cell-derived HIV-1 particles had low specific infectivity. We subsequently identified feline apolipoprotein B-editing catalytic polypeptide 3 (feAPOBEC3) proteins as active inhibitors of HIV-1 particle infectivity. CrFK cells express at least three different APOBEC3s: APOBEC3C, APOBEC3H, and APOBEC3CH. While the feAPOBEC3C did not significantly inhibit HIV-1, the feAPOBEC3H and feAPOBEC3CH induced G to A hypermutations of the viral cDNA and reduced the infectivity approximately 10- to approximately 40-fold.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Replicación Viral , Animales , Secuencia de Bases , Antígenos CD4/metabolismo , Gatos , Línea Celular , Citosina Desaminasa/metabolismo , Estudios de Evaluación como Asunto , Proteínas de Fusión gag-pol/biosíntesis , Humanos , Datos de Secuencia Molecular , Pan troglodytes , Receptores CCR5/metabolismo , Especificidad de la Especie , Secuencias Repetidas Terminales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...