Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Radiat Prot Dosimetry ; 199(15-16): 1824-1828, 2023 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-37819323

RESUMO

Currently, the most used methods of plastic scintillator (PS) manufacturing are cell casting and bulk polymerisation, extrusion, injection molding, whereas digital light processing (DLP) 3D printing technique has been recently introduced. For our research, we measured blue-emitting EJ-200, EJ-208, green-emitting EJ-260, EJ-262 cell cast and two types of blue-emitting DLP-printed PSs. The light output of the samples, with the same dimension of 10 mm × 10 mm × 10 mm, was compared. The light output of the samples, relative to the reference EJ-200 cell-cast scintillator, equals about 40-49 and 70-73% for two types of 3D-printed, and two green-emitting cell-casted PSs, respectively. Performance of the investigated scintillators is sufficient to use them in a plastic scintillation dosemeter operating in high fluence gamma radiation fields.


Assuntos
Radiometria , Contagem de Cintilação , Contagem de Cintilação/métodos , Impressão Tridimensional
2.
Phys Med ; 47: 80-85, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29609822

RESUMO

PURPOSE: To develop an assumption-free methodology for aligning the geometry of on-board imagers with the geometry of medical linear accelerators applied in image-guided radiotherapy (IGRT). MATERIAL: Alignment of the on-board imaging (OBI) system with respect to the accelerator system is achieved using a multi-modular phantom described elsewhere (Tabor et al., 2017), enabling the geometry of the linear accelerator to be specified without any pre-assumptions. METHODS: The placement of two isocentres (of the on-board imager and of the therapeutic system) and of three frames of reference (those of the on-board imager, of the therapeutic system, and of the treatment table) is formulated as an optimization problem. It is solved by analysing the images of fiducial points positioned in 3D space in phantom modules attached to the treatment table and to the collimator of the accelerator. Fiducials are projected onto an imaging plane of unknown characteristics from a virtual source of unknown coordinates. CONCLUSIONS: An analytical framework exploiting projection images of the proposed multi-modular phantom has been developed, enabling precise alignment of the reference frame related to the on-board imager with the reference frame related to the therapeutic system. Within the proposed framework, the necessary corrections of treatment table positioning prior to patient irradiation, are delivered in the treatment table coordinates.


Assuntos
Diagnóstico por Imagem/instrumentação , Aceleradores de Partículas , Algoritmos , Calibragem , Processamento de Imagem Assistida por Computador , Radioterapia de Intensidade Modulada
3.
Curr Top Microbiol Immunol ; 281: 29-106, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12932075

RESUMO

Evidence obtained during the last few years has greatly extended our understanding of the cell surface receptors that mediate infections of retroviruses and has provided many surprising insights. In contrast to other cell surface components such as lectins or proteoglycans that influence infections indirectly by enhancing virus adsorption onto specific cells, the true receptors induce conformational changes in the viral envelope glycoproteins that are essential for infection. One surprise is that all of the cell surface receptors for gamma-retroviruses are proteins that have multiple transmembrane (TM) sequences, compatible with their identification in known instances as transporters for important solutes. In striking contrast, almost all other animal viruses use receptors that exclusively have single TM sequences, with the sole proven exception we know of being the coreceptors used by lentiviruses. This evidence strongly suggests that virus genera have been prevented because of their previous evolutionary adaptations from switching their specificities between single-TM and multi-TM receptors. This evidence also implies that gamma-retroviruses formed by divergent evolution from a common origin millions of years ago and that individual viruses have occasionally jumped between species (zoonoses) while retaining their commitment to using the orthologous receptor of the new host. Another surprise is that many gamma-retroviruses use not just one receptor but pairs of closely related receptors as alternatives. This appears to have enhanced viral survival by severely limiting the likelihood of host escape mutations. All of the receptors used by gamma-retroviruses contain hypervariable regions that are often heavily glycosylated and that control the viral host range properties, consistent with the idea that these sequences are battlegrounds of virus-host coevolution. However, in contrast to previous assumptions, we propose that gamma-retroviruses have become adapted to recognize conserved sites that are important for the receptor's natural function and that the hypervariable sequences have been elaborated by the hosts as defense bulwarks that surround the conserved viral attachment sites. Previously, it was believed that binding to receptors directly triggers a series of conformational changes in the viral envelope glycoproteins that culminate in fusion of the viral and cellular membranes. However, new evidence suggests that gamma-retroviral association with receptors triggers an obligatory interaction or cross-talk between envelope glycoproteins on the viral surface. If this intermediate step is prevented, infection fails. Conversely, in several circumstances this cross-talk can be induced in the absence of a cell surface receptor for the virus, in which case infection can proceed efficiently. This new evidence strongly implies that the role of cell surface receptors in infections of gamma-retroviruses (and perhaps of other enveloped animal viruses) is more complex and interesting than was previously imagined. Recently, another gammaretroviral receptor with multiple transmembrane sequences was cloned. See Prassolov, Y., Zhang, D., Ivanov, D., Lohler, J., Ross, S.R., and Stocking, C. Sodium-dependent myo-inositol transporter 1 is a receptor for Mus cervicolor M813 murine leukemia virus.


Assuntos
Gammaretrovirus/fisiologia , Receptores de Superfície Celular/fisiologia , Infecções por Retroviridae/metabolismo , Infecções Tumorais por Vírus/metabolismo , Animais , Sítios de Ligação/fisiologia , Evolução Molecular , Humanos , Glicoproteínas de Membrana/fisiologia , Receptores de HIV/fisiologia , Receptores Virais/fisiologia , Proteínas do Envelope Viral/metabolismo
4.
J Virol ; 75(24): 12266-78, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11711617

RESUMO

To identify sites in gp120 that interact with the CCR5 coreceptor and to analyze the mechanisms of infection, we selected variants of the CCR5-dependent JRCSF molecular clone of human immunodeficiency virus type 1 (HIV-1) that adapted to replicate in HeLa-CD4 cells that express the mutant coreceptor CCR5(Y14N) or CCR5(G163R), which were previously shown to bind purified gp120-CD4 complexes only weakly. Correspondingly, these mutant CCR5s mediate infections of wild-type virus only at relatively high cell surface concentrations, demonstrating a concentration-dependent assembly requirement for infection. The plots of viral infectivity versus concentration of coreceptors had sigmoidal shapes, implying involvement of multiple coreceptors, with an estimated stoichiometry of four to six CCR5s in the active complexes. All of the adapted viruses had mutations in the V3 loops of their gp120s. The titers of recombinant HIV-1 virions with these V3 mutations were determined in previously described panels of HeLa-CD4 cell clones that express discrete amounts of CCR5(Y14N) or CCR5(G163R). The V3 loop mutations did not alter viral utilization of wild-type CCR5, but they specifically enhanced utilization of the mutant CCR5s by two distinct mechanisms. Several mutant envelope glycoproteins were highly fusogenic in syncytium assays, and these all increased the efficiency of infection of the CCR5(Y14N) or CCR5(G163R) clonal panels without enhancing virus adsorption onto the cells or viral affinity for the coreceptor. In contrast, V3 loop mutation N300Y was selected during virus replication in cells that contained only a trace of CCR5(Y14N) and this mutation increased the apparent affinity of the virus for this coreceptor, as indicated by a shift in the sigmoid-shaped infectivity curve toward lower concentrations. Surprisingly, N300Y increased viral affinity for the second extracellular loop of CCR5(Y14N) rather than for the mutated amino terminus. Indeed, the resulting virus was able to use a mutant CCR5 that lacks 16 amino acids at its amino terminus, a region previously considered essential for CCR5 coreceptor function. Our results demonstrate that the role of CCR5 in infection involves at least two steps that can be strongly and differentially altered by mutations in either CCR5 or the V3 loop of gp120: a concentration-dependent binding step that assembles a critical multivalent virus-coreceptor complex and a postassembly step that likely involves a structural rearrangement of the complex. The postassembly step can severely limit HIV-1 infections and is not an automatic consequence of virus-coreceptor binding, as was previously assumed. These results have important implications for our understanding of the mechanism of HIV-1 infection and the factors that may select for fusogenic gp120 variants during AIDS progression.


Assuntos
Proteína gp120 do Envelope de HIV/química , HIV-1/fisiologia , Fusão de Membrana , Receptores CCR5/química , Sequência de Aminoácidos , Animais , Cricetinae , Proteína gp120 do Envelope de HIV/fisiologia , Células HeLa , Humanos , Dados de Sequência Molecular , Receptores CCR5/fisiologia , Sulfatos/química
5.
J Virol ; 75(18): 8449-60, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11507190

RESUMO

In contrast to humans, several primate species are believed to have harbored simian immunodeficiency viruses (SIVs) since ancient times. In particular, the geographically dispersed species of African green monkeys (AGMs) are all infected with highly diversified SIVagm viruses at high prevalences (greater than 50% of sexually mature individuals) without evident diseases, implying that the progenitor monkeys were infected prior to their dispersal. If this is correct, AGMs would be expected to have accumulated frequent resistance-conferring polymorphisms in host genes that are important for SIV replication. Accordingly, we analyzed the coding sequences of the CCR5 coreceptors from 26 AGMs (52 alleles) in distinct populations of the four species. These samples contained 29 nonsynonymous coding changes and only 15 synonymous nucleotide substitutions, implying intense functional selection. Moreover, 24 of the resulting amino acid substitutions were tightly clustered in the CCR5 amino terminus (D13N in the vervets and Y14N in the tantalus species) or in the first extracellular loop (Q93R and Q93K in all species). The Y14N substitution was extremely frequent in the 12 wild-born African tantalus, with 7 monkeys being homozygous for this substitution and 4 being heterozygous. Although two of these heterozygotes and the only wild-type homozygote were naturally infected with SIVagm, none of the Y14N homozygotes were naturally infected. A focal infectivity assay for SIVagm indicated that all five tested SIVagms efficiently use CCR5 as a coreceptor and that they also use CXCR6 (STRL33/Bonzo) and GPR15 (BOB) with lower efficiencies but not CXCR4. Interestingly, the D13N, Y14N, Q93R, and Q93K substitutions in AGM CCR5 all strongly inhibited infections by the SIVagm isolates in vitro. The Y14N substitution eliminates a tyrosine sulfation site that is important for infections and results in partial N-linked glycosylation (i.e., 60% efficiency) at this position. Nevertheless, the CCR5(Y14N) component that lacks an N-linked oligosaccharide binds the chemokine MIP-lbeta with a normal affinity and is fully active in signal transduction. Similarly, D13N and Q93R substitutions did not interfere with signal transduction. Thus, the common substitution polymorphisms in AGM CCR5 strongly inhibit SIVagm infections while substantially preserving chemokine signaling. In contrast, polymorphisms of human CCR5 are relatively infrequent, and the amino acid substitutions are randomly situated and generally without effects on coreceptor function. These results support an ancient coevolution of AGMs and SIVagm viruses and establish AGMs as a highly informative model for learning about host proteins that play critical roles in immunodeficiency virus infections.


Assuntos
Evolução Molecular , Família Multigênica , Polimorfismo Genético , Receptores CCR5/genética , Vírus da Imunodeficiência Símia/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Chlorocebus aethiops , Células HeLa , Humanos , Dados de Sequência Molecular , Mutagênese , Receptores CCR5/metabolismo , Vírus da Imunodeficiência Símia/metabolismo , Células Vero , Xenopus laevis
6.
J Biol Chem ; 276(29): 27221-30, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11350958

RESUMO

The sodium-dependent neutral amino acid transporter type 2 (ASCT2) was recently identified as a cell surface receptor for endogenously inherited retroviruses of cats, baboons, and humans as well as for horizontally transmitted type-D simian retroviruses. By functional cloning, we obtained 10 full-length 2.9-kilobase pair (kbp) cDNAs and two smaller identical 2.1-kbp cDNAs that conferred susceptibility to these viruses. Compared with the 2.9-kbp cDNA, the 2.1-kbp cDNA contains exonic deletions in its 3' noncoding region and a 627-bp 5' truncation that eliminates sequences encoding the amino-terminal portion of the full-length ASCT2 protein. Although expression of the truncated mRNA caused enhanced amino acid transport and viral receptor activities, the AUG codon nearest to its 5' end is flanked by nucleotides that are incompatible with translational initiation and the next in-frame AUG codon is far downstream toward the end of the protein coding sequence. Interestingly, the 5' region of the truncated ASCT2 mRNA contains a closely linked series of CUG(Leu) and GUG(Val) codons in optimal consensus contexts for translational initiation. By deletion and site-directed mutagenesis, cell-free translation, and analyses of epitope-tagged ASCT2 proteins synthesized intracellularly, we determined that the truncated mRNA encodes multiple ASCT2 isoforms with distinct amino termini that are translationally initiated by a leaky scanning mechanism at these CUG and GUG codons. Although the full-length ASCT2 mRNA contains a 5'-situated AUG initiation codon, a significant degree of leaky scanning also occurred in its translation. ASCT2 isoforms with relatively short truncations were active in both amino acid transport and viral reception, whereas an isoform with a 79-amino acid truncation that lacked the first transmembrane sequence was active only in viral reception. We conclude that ASCT2 isoforms with truncated amino termini are synthesized in mammalian cells by a leaky scanning mechanism that employs multiple alternative CUG and GUG initiation codons.


Assuntos
Proteínas de Transporte/metabolismo , Códon , Biossíntese de Proteínas , Receptores Virais/metabolismo , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Primers do DNA , DNA Complementar , Humanos , Camundongos , Dados de Sequência Molecular , Conformação Proteica , RNA Mensageiro/genética , Receptores Virais/química , Receptores Virais/genética , Retroviridae/metabolismo
7.
Phys Rev Lett ; 86(8): 1426-9, 2001 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-11290159

RESUMO

We develop an approximation scheme for the quantum mechanics of N D0-branes at finite temperature in the 't Hooft large- N limit. The entropy of the quantum mechanics calculated using this approximation agrees well with the Bekenstein-Hawking entropy of a ten-dimensional nonextremal black hole with 0-brane charge. This result is in accordance with the duality conjectured by Itzhaki, Maldacena, Sonnenschein, and Yankielowicz [Phys. Rev. D 58, 046004 (1998)]. Our approximation scheme provides a model for the density matrix which describes a black hole in the strongly coupled quantum mechanics.


Assuntos
Astronomia , Gravitação , Fenômenos Astronômicos , Teoria Quântica , Sistema Solar , Termodinâmica
8.
J Virol ; 74(20): 9797-801, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11000257

RESUMO

Chinese hamster ovary (CHO) cells are resistant to infections by gibbon ape leukemia virus (GALV) and amphotropic murine leukemia virus (A-MLV) unless they are pretreated with tunicamycin, an inhibitor of N-linked glycosylation. These viruses use the related sodium-phosphate symporters Pit1 and Pit2, respectively, as receptors in nonhamster cells, and evidence has suggested that the corresponding transporters of CHO cells may be masked by tunicamycin-sensitive secreted inhibitors. Although the E36 line of Chinese hamster cells was reported to secrete the putative Pit2 inhibitor and to be sensitive to the inhibitory CHO factors, E36 cells are highly susceptible to both GALV and A-MLV in the absence of tunicamycin. Moreover, expression of E36 Pit2 in CHO cells conferred tunicamycin-independent susceptibilities to both viruses. Based on the latter results, it was suggested that E36 Pit2 must functionally differ from the endogenous Pit2 of CHO cells. To test these ideas, we analyzed the receptor properties of CHO Pit1 and Pit2 in CHO cells. Surprisingly, and counterintuitively, transfection of a CHO Pit2 expression vector into CHO cells conferred strong susceptibility to both GALV and A-MLV, and similar overexpression of CHO Pit1 conferred susceptibility to GALV. Thus, CHO Pit2 is a promiscuous functional receptor for both viruses, and CHO Pit1 is a functional receptor for GALV. Similarly, we found that the natural resistance of Mus dunni tail fibroblasts to subgroup C feline leukemia viruses (FeLV-C) was eliminated simply by overexpression of the endogenous FeLV-C receptor homologue. These results demonstrate a novel and simple method to unmask latent retroviral receptor activities that occur in some cells. Specifically, resistances to retroviruses that are caused by subthreshold levels of receptor expression or by stoichiometrically limited masking or interference mechanisms can be efficiently overcome simply by overexpressing the endogenous receptors in the same cells.


Assuntos
Vírus da Leucemia Felina/fisiologia , Vírus da Leucemia do Macaco Gibão/fisiologia , Receptores Virais/análise , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Camundongos , Dados de Sequência Molecular , Receptores Virais/fisiologia , Tunicamicina/farmacologia
9.
J Virol ; 74(17): 8085-93, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10933718

RESUMO

The baboon endogenous retrovirus (BaEV) belongs to a large, widely dispersed interference group that includes the RD114 feline endogenous virus and primate type D retroviruses. Recently, we and another laboratory independently cloned a human receptor for these viruses and identified it as the human sodium-dependent neutral amino acid transporter type 2 (hASCT2). Interestingly, mouse and rat cells are efficiently infected by BaEV but only become susceptible to RD114 and type D retroviruses if the cells are pretreated with tunicamycin, an inhibitor of protein N-linked glycosylation. To investigate this host range difference, we cloned and analyzed NIH Swiss mouse ASCT2 (mASCT2). Surprisingly, mASCT2 did not mediate BaEV infection, which implied that mouse cells might have an alternative receptor for this virus. In addition, elimination of the two N-linked oligosaccharides from mASCT2 by mutagenesis, as substantiated by protein N-glycosidase F digestions and Western immunoblotting, did not enable it to function as a receptor for RD114 or type D retroviruses. Based on these results, we found that the related ASCT1 transporters of humans and mice are efficient receptors for BaEV but are relatively inactive for RD114 and type D retroviruses. Furthermore, elimination of the two N-linked oligosaccharides from extracellular loop 2 of mASCT1 by mutagenesis enabled it to function as an efficient receptor for RD114 and type D retroviruses. Thus, we infer that the tunicamycin-dependent infection of mouse cells by RD114 and type D retroviruses is caused by deglycosylation of mASCT1, which unmasks previously buried sites for viral interactions. In contrast, BaEV efficiently employs the glycosylated forms of mASCT1 that occur normally in untreated mouse cells.


Assuntos
Proteínas de Transporte/genética , Retrovirus Endógenos/genética , Receptores Virais/genética , Sódio/metabolismo , Células 3T3 , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos , Animais , Proteínas de Transporte/metabolismo , Gatos , Retrovirus Endógenos/metabolismo , Células HeLa , Humanos , Immunoblotting , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligossacarídeos/genética , Papio , Ratos , Receptores Virais/metabolismo , Alinhamento de Sequência
10.
J Virol ; 74(15): 7005-15, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10888639

RESUMO

In addition to the primary cell surface receptor CD4, CCR5 or another coreceptor is necessary for infections by human immunodeficiency virus type 1 (HIV-1), yet the mechanisms of coreceptor function and their stoichiometries in the infection pathway remain substantially unknown. To address these issues, we studied the effects of CCR5 concentrations on HIV-1 infections using wild-type CCR5 and two attenuated mutant CCR5s, one with the mutation Y14N at a critical tyrosine sulfation site in the amino terminus and one with the mutation G163R in extracellular loop 2. The Y14N mutation converted a YYT sequence at positions 14 to 16 to an NYT consensus site for N-linked glycosylation, and the mutant protein was shown to be glycosylated at that position. The relationships between HIV-1 infectivity values and CCR5 concentrations took the form of sigmoidal (S-shaped) curves, which were dramatically altered in different ways by these mutations. Both mutations shifted the curves by factors of approximately 30- to 150-fold along the CCR5 concentration axis, consistent with evidence that they reduce affinities of virus for the coreceptor. In addition, the Y14N mutation specifically reduced the maximum efficiencies of infection that could be obtained at saturating CCR5 concentrations. The sigmoidal curves for all R5 HIV-1 isolates were quantitatively consistent with a simple mathematical model, implying that CCR5s reversibly associate with cell surface HIV-1 in a concentration-dependent manner, that approximately four to six CCR5s assemble around the virus to form a complex needed for infection, and that both mutations inhibit assembly of this complex but only the Y14N mutation also significantly reduces its ability to successfully mediate HIV-1 infections. Although several alternative models would be compatible with our data, a common feature of these alternatives is the cooperation of multiple CCR5s in the HIV-1 infection pathway. This cooperativity will need to be considered in future studies to address in detail the mechanism of CCR5-mediated HIV-1 membrane fusion.


Assuntos
HIV-1/patogenicidade , Receptores CCR5/metabolismo , Antígenos CD4/metabolismo , Quimiocina CCL4 , Citometria de Fluxo/métodos , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/metabolismo , Células HeLa , Humanos , Proteínas Inflamatórias de Macrófagos/metabolismo , Modelos Biológicos , Mutação , Radioimunoensaio , Receptores CCR5/genética
11.
J Virol ; 74(13): 5982-7, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10846079

RESUMO

The vif gene of human immunodeficiency virus type 1 (HIV-1) greatly enhances the infectivity of HIV-1 virions that are released from cells classified as nonpermissive (e.g., lymphocytes, macrophages, and H9 leukemic T cells) but is irrelevant in permissive cells (e.g., HeLa or COS cells). Recently, it was reported that vif expression in nonpermissive cells dramatically increases infectivity not only of HIV-1 but also of other enveloped viruses, including murine leukemia viruses (MLVs). This was surprising in part because MLVs and other murine retroviruses lack vif genes yet replicate efficiently in T lymphocytes. To investigate these issues, we first developed improved methods for producing substantial quantities of HIV-1 virions with vif deletions from healthy H9 cells. These virions had approximately the same amounts of major core proteins and envelope glycoproteins as the control wild-type virions but were only approximately 1% as infectious. We then produced H9 cells that contained wild-type or vif deletion HIV-gpt proviruses, which lack a functional env gene. After superinfection with either xenotropic or amphotropic MLVs, these cells released HIV-gpt virions pseudotyped with an MLV envelope plus replication-competent MLV. Interestingly, the pseudotyped HIV-gpt (vif deletion) virions were noninfectious, whereas the MLV virions simultaneously released from the same H9 cells were fully infectious. These results strongly suggest that the Vif protein functions in a manner that is both cell specific and at least substantially specific for HIV-1 and related lentiviruses. In addition, these results confirm that vif deletion HIV-1 virions from nonpermissive cells are blocked at a postpenetration stage of the infection pathway.


Assuntos
Produtos do Gene vif/metabolismo , HIV-1/metabolismo , HIV-1/fisiologia , Vírus da Leucemia Murina/fisiologia , Animais , Células COS , Células HeLa , Humanos , Células Tumorais Cultivadas , Vírion , Produtos do Gene vif do Vírus da Imunodeficiência Humana
12.
AIDS Res Hum Retroviruses ; 16(9): 871-82, 2000 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-10875613

RESUMO

Strains of human immunodeficiency virus type 1 (HIV-1) that use the coreceptor CXCR4 (X4 strains) become laboratory adapted (LA) when selected for ability to replicate in leukemic T cell lines such as H9. Compared with patient X4 viruses, the gp120-gp41 complexes of LA viruses have a constellation of common properties including enhanced affinities for CD4, greater sensitivities to inactivations by diverse antibodies and by soluble CD4, increased shedding of gp120, and improved abilities to infect HeLa-CD4 cell clones that contain only trace quantities of CD4. These common characteristics, which may result from a concerted structural rearrangement of the gp120-gp41 complexes, have made it difficult to identify a specific feature that is critical for laboratory adaptation. To test the hypothesis that replication of patient X4 HIV-1 is limited by the low CD4 concentration in H9 cells (7.0 x 10(3) CD4/cell), we constructed H9 derivatives that express at least 10 times more of this receptor. Interestingly, most patient X4 isolates readily grew in these derivative cells, and the resulting virus preparations retained the characteristics of primary viruses throughout multiple passages. In contrast, selection of the same viruses in the parental H9 cells resulted in outgrowth of LA derivatives. We conclude that a weak interaction of patient X4 HIV-1 isolates with CD4 is the primary factor that limits their replication in leukemic T cell lines.


Assuntos
Antígenos CD4/fisiologia , HIV-1/fisiologia , HIV-1/patogenicidade , Adaptação Fisiológica , Linhagem Celular , Variação Genética , Proteína gp120 do Envelope de HIV/fisiologia , Proteína gp41 do Envelope de HIV/fisiologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/isolamento & purificação , Células HeLa , Humanos , Linfócitos T/imunologia , Linfócitos T/virologia , Replicação Viral
13.
J Virol ; 74(1): 237-44, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10590111

RESUMO

Because mutations in envelope glycoproteins of retroviruses or in their cell surface receptors can eliminate function by multiple mechanisms, it has been difficult to unambiguously identify sites for their interactions by site-directed mutagenesis. Recently, we developed a gain-of-function approach to overcome this problem. Our strategy relies on the fact that feline leukemia virus subgroup B (FeLV-B) and amphotropic murine leukemia virus (A-MLV) have closely related gp70 surface envelope glycoproteins and use related Na(+)-dependent phosphate symporters, Pit1 and Pit2, respectively, as their receptors. We previously observed that FeLV-B/A-MLV envelope glycoprotein chimeras spliced between the variable regions VRA and VRB were unable to use Pit1 or Pit2 as a receptor but could efficiently use specific Pit1/Pit2 chimeras. The latter study suggested that the VRA of A-MLV and FeLV-B functionally interact with the presumptive extracellular loops 4 and 5 (ECL4 and -5) of their respective receptors, whereas VRB interacts with ECL2. We also found that FeLV-B gp70 residues F60 and P61 and A-MLV residues Y60 and V61 in the first disulfide-bonded loop of VRA were important for functional interaction with the receptor's ECL4 or -5. We have now extended this approach to identify additional VRA and VRB residues that are involved in receptor recognition. Our studies imply that FeLV-B VRA residues F60 and P61 interact with the Pit1 ECL5 region, whereas VRA residues 66 to 78 interact with Pit1 ECL4. Correspondingly, A-MLV VRA residues Y60 and V61 interact with the Pit2 ECL5 region, whereas residues 66 to 78 interact with Pit2 ECL4. Similar studies that focused on the gp70 VRB implicated residues 129 to 139 as contributing to specific interactions with the receptor ECL2. These results identify three regions of gp70 that interact in a specific manner with distinct portions of their receptors, thereby providing a map of the functionally interacting surfaces.


Assuntos
Vírus da Leucemia Felina/fisiologia , Vírus da Leucemia Murina/fisiologia , Receptores Virais/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Quimera , Primers do DNA , Vírus da Leucemia Felina/química , Vírus da Leucemia Felina/genética , Vírus da Leucemia Murina/química , Vírus da Leucemia Murina/genética , Camundongos , Dados de Sequência Molecular , Mutagênese , Homologia de Sequência de Aminoácidos
14.
J Virol ; 73(11): 9362-8, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10516044

RESUMO

The differential susceptibilities of mouse strains to xenotropic and polytropic murine leukemia viruses (X-MLVs and P-MLVs, respectively) are poorly understood but may involve multiple mechanisms. Recent evidence has demonstrated that these viruses use a common cell surface receptor (the X-receptor) for infection of human cells. We describe the properties of X-receptor cDNAs with distinct sequences cloned from five laboratory and wild strains of mice and from hamsters and minks. Expression of these cDNAs in resistant cells conferred susceptibilities to the same viruses that naturally infect the animals from which the cDNAs were derived. Thus, a laboratory mouse (NIH Swiss) X-receptor conferred susceptibility to P-MLVs but not to X-MLVs, whereas those from humans, minks, and several wild mice (Mus dunni, SC-1 cells, and Mus spretus) mediated infections by both X-MLVs and P-MLVs. In contrast, X-receptors from the resistant mouse strain Mus castaneus and from hamsters were inactive as viral receptors. These results suggest that X-receptor polymorphisms are a primary cause of resistances of mice to members of the X-MLV/P-MLV family of retroviruses and are responsible for the xenotropism of X-MLVs in laboratory mice. By site-directed mutagenesis, we substituted sequences between the X-receptors of M. dunni and NIH Swiss mice. The NIH Swiss protein contains two key differences (K500E in presumptive extracellular loop 3 [ECL 3] and a T582 deletion in ECL 4) that are both required to block X-MLV infections. Accordingly, a single inverse mutation in the NIH Swiss protein conferred X-MLV susceptibility. Furthermore, expression of an X-MLV envelope glycoprotein in Chinese hamster ovary cells interfered efficiently with X-MLV and P-MLV infections mediated by X-receptors that contained K500 and/or T582 but had no effect on P-MLV infections mediated by X-receptors that lacked these amino acids. In contrast, moderate expression of a P-MLV (MCF247) envelope glycoprotein did not cause substantial interference, suggesting that X-MLV and P-MLV glycoproteins interfere nonreciprocally with X-receptor-mediated infections. We conclude that P-MLVs have become adapted to utilize X-receptors that lack K500 and T582. A penalty for this adaptation is a reduced ability to interfere with superinfection. Because failure of interference is a hallmark of several exceptionally pathogenic retroviruses, we propose that it contributes to P-MLV-induced diseases.


Assuntos
Vírus da Leucemia Murina/metabolismo , Leucemia Experimental/virologia , Polimorfismo Genético , Receptores Virais/genética , Infecções por Retroviridae/virologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Clonagem Molecular , Cricetinae , DNA Complementar/genética , Suscetibilidade a Doenças , Humanos , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/imunologia , Leucemia Experimental/imunologia , Leucemia Experimental/metabolismo , Camundongos , Dados de Sequência Molecular , Muridae , Mutagênese Sítio-Dirigida , Receptores Acoplados a Proteínas G , Receptores Virais/química , Receptores Virais/metabolismo , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/metabolismo , Transfecção , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/metabolismo , Infecções Tumorais por Vírus/virologia , Receptor do Retrovírus Politrópico e Xenotrópico
15.
J Biol Chem ; 274(33): 23499-507, 1999 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-10438529

RESUMO

Infections by human immunodeficiency virus type 1 (HIV-1) involve interactions of the viral envelope glycoprotein gp120 with CD4 and then with a coreceptor. R5 isolates of HIV-1 use CCR5 as a coreceptor, whereas X4 isolates use CXCR4. It is not known whether coreceptors merely trigger fusion of the viral and cellular membranes or whether they also influence the energetics of virus adsorption, the placement of the membrane fusion reaction, and the metabolism of adsorbed gp120. Surprisingly, the pathway for metabolism of adsorbed gp120 has not been investigated thoroughly in any cells. To address these issues, we used purified (125)I-gp120s derived from the R5 isolate BaL and from the X4 isolate IIIB as ligands for binding onto human cells that expressed CD4 alone or CD4 with a coreceptor. The gp120 preparations were active in forming ternary complexes with CD4 and the appropriate coreceptor. Moreover, the cellular quantities of CD4 and coreceptors were sufficient for efficient infections by the corresponding HIV-1 isolates. In these conditions, the kinetics and affinities of (125)I-gp120 adsorptions and their subsequent metabolisms were strongly dependent on CD4 but were not significantly influenced by CCR5 or CXCR4. After binding to CD4, the (125)I-gp120s slowly became resistant to extraction from the cell monolayers by pH 3.0 buffer, suggesting that they were endocytosed with half-times of 1-2 h. Within 20-30 min of endocytosis, the (125)I-gp120s were proteolytically degraded to small products that were shed into the media. The weak base chloroquine strongly inhibited (125)I-gp120 proteolysis and caused its intracellular accumulation, suggesting involvement of a low pH organelle. Results supporting these methods and conclusions were obtained by confocal immunofluorescence microscopy. We conclude that the energetics, kinetics, and pathways of (125)I-gp120 binding, endocytosis, and proteolysis are determined principally by CD4 rather than by coreceptors in cells that contain sufficient coreceptors for efficient infections. Therefore, the role of coreceptors in HIV-1 infections probably does not include steerage or subcellular localization of adsorbed virus.


Assuntos
Antígenos CD4/fisiologia , Proteína gp120 do Envelope de HIV/metabolismo , Receptores de HIV/fisiologia , Antígenos CD4/metabolismo , Endocitose , HIV-1/fisiologia , Células HeLa , Humanos , Hidrólise , Imuno-Histoquímica , Cinética , Fusão de Membrana , Microscopia Confocal , Ligação Proteica , Receptores CCR5/metabolismo , Receptores CCR5/fisiologia , Receptores CXCR4/metabolismo , Receptores CXCR4/fisiologia , Receptores de HIV/metabolismo
16.
J Virol ; 73(8): 6500-5, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10400745

RESUMO

Domestic cats infected with the horizontally transmitted feline leukemia virus subgroup A (FeLV-A) often produce mutants (termed FeLV-C) that bind to a distinct cell surface receptor and cause severe aplastic anemia in vivo and erythroblast destruction in bone marrow cultures. The major determinant for FeLV-C-induced anemia has been mapped to a small region of the surface envelope glycoprotein that is responsible for its receptor binding specificity. Thus, erythroblast destruction may directly or indirectly result from FeLV-C binding to its receptor. To address these issues, we functionally cloned a putative cell surface receptor for FeLV-C (FLVCR) by using a human T-lymphocyte cDNA library in a retroviral vector. Expression of the 2.0-kbp FLVCR cDNA in naturally resistant Swiss mouse fibroblasts and Chinese hamster ovary cells caused substantial susceptibility to FeLV-C but no change in susceptibilities to FeLV-B and other retroviruses. The predicted FLVCR protein contains 555 amino acids and 12 hydrophobic potential membrane-spanning sequences. Database searches indicated that FLVCR is a member of the major-facilitator superfamily of transporters and implied that it may transport an organic anion. RNA blot analyses showed that FLVCR mRNA is expressed in multiple hematopoietic lineages rather than specifically in erythroblasts. These results suggest that the targeted destruction of erythroblasts by FeLV-C may derive from their greater sensitivity to this virus rather than from a preferential susceptibility to infection.


Assuntos
Anemia Aplástica/virologia , Proteínas de Transporte/classificação , Vírus da Leucemia Felina/metabolismo , Receptores Virais/classificação , Células 3T3 , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Caenorhabditis elegans , Proteínas de Transporte/química , Proteínas de Transporte/genética , Gatos , Cricetinae , DNA Complementar , Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Receptores Virais/química , Receptores Virais/genética , Homologia de Sequência de Aminoácidos , Distribuição Tecidual
17.
J Virol ; 73(5): 4470-4, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10196349

RESUMO

The type D simian retroviruses cause immunosuppression in macaques and have been reported as a presumptive opportunistic infection in a patient with AIDS. Previous evidence based on viral interference has strongly suggested that the type D simian viruses share a common but unknown cell surface receptor with three type C viruses: feline endogenous virus (RD114), baboon endogenous virus, and avian reticuloendotheliosis virus. Furthermore, the receptor gene for these viruses has been mapped to human chromosome 19q13.1-13.2. We now report the isolation and characterization of a cell surface receptor for this group of retroviruses by using a human T-lymphocyte cDNA library in a retroviral vector. Swiss mouse fibroblasts (NIH 3T3), which are naturally resistant to RD114, were transduced with the retroviral library and then challenged with an RD114-pseudotyped virus containing a dominant selectable gene for puromycin resistance. Puromycin selection yielded 12 cellular clones that were highly susceptible to a beta-galactosidase-encoding lacZ(RD114) pseudotype virus. Using PCR primers specific for vector sequences, we amplified a common 2.9-kb product from 10 positive clones. Expression of the 2.9-kb cDNA in Chinese hamster ovary cells conferred susceptibility to RD114, baboon endogenous virus, and the type D simian retroviruses. The 2.9-kb cDNA predicted a protein of 541 amino acids that had 98% identity with the previously cloned human Na+-dependent neutral-amino-acid transporter Bo. Accordingly, expression of the RD114 receptor in NIH 3T3 cells resulted in enhanced cellular uptake of L-[3H]alanine and L-[3H]glutamine. RNA blot (Northern) analysis suggested that the RD114 receptor is widely expressed in human tissues and cell lines, including hematopoietic cells. The human Bo transporter gene has been previously mapped to 19q13.3, which is closely linked to the gene locus of the RD114 receptor.


Assuntos
Sistema ASC de Transporte de Aminoácidos , Proteínas de Transporte/metabolismo , Retrovirus Endógenos/metabolismo , Receptores Virais/metabolismo , Retrovirus dos Símios/metabolismo , Sódio/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Proteínas de Transporte/genética , Gatos , Clonagem Molecular , Cricetinae , DNA Complementar , Retrovirus Endógenos/fisiologia , Expressão Gênica , Humanos , Camundongos , Antígenos de Histocompatibilidade Menor , Dados de Sequência Molecular , Papio , Ratos , Receptores Virais/genética , Retrovirus dos Símios/fisiologia , Distribuição Tecidual
18.
Proc Natl Acad Sci U S A ; 96(3): 927-32, 1999 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-9927670

RESUMO

Xenotropic and polytropic murine leukemia viruses (X-MLVs and P-MLVs) cross-interfere to various extents in non-mouse species and in wild Asian mice, suggesting that they might use a common receptor for infection. Consistent with this hypothesis, the susceptibility of some wild mice to X-MLVs has been mapped to the P-MLV receptor locus at the distal end of mouse chromosome 1. In this study, we report the isolation and characterization of a cDNA for the human X-MLV cell surface receptor (X-receptor) by using a human T lymphocyte cDNA library in a retroviral vector. The predicted X-receptor contains 696 amino acids with multiple hydrophobic potential membrane-spanning sequences and with weak homologies to the yeast proteins SYG1, of unknown function, and PHO81, which has been implicated in a system that regulates transport of inorganic phosphate. Expression of the X-receptor in Chinese hamster ovary cells, which are substantially resistant to P-MLVs and to X-MLVs, made them susceptible to both of these virus groups. The mouse homologue of the X-receptor was mapped by hybridization to the distal end of chromosome 1 at the same position as the P-MLV receptor gene Rmc1. These results strongly support the hypothesis that a common gene encodes the receptors for X-MLVs and P-MLVs, with the human X-receptor preferentially mediating X-MLV infections and the homologous protein of inbred mice mediating only P-MLV infections. We propose that X-MLVs and P-MLVs comprise a single family of retroviruses that have coevolved in response to diversification in X-receptor genes of the host.


Assuntos
Vírus da Leucemia Murina/fisiologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiologia , Receptores Virais/química , Receptores Virais/fisiologia , Sequência de Aminoácidos , Animais , Ásia , Células CHO , Linhagem Celular , Mapeamento Cromossômico , Clonagem Molecular , Cricetinae , Humanos , Glicoproteínas de Membrana/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Muridae , Receptores Acoplados a Proteínas G , Receptores Virais/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/fisiologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Linfócitos T/virologia , Receptor do Retrovírus Politrópico e Xenotrópico
19.
J Biol Chem ; 274(4): 1905-13, 1999 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-9890944

RESUMO

Like the CCR5 chemokine receptors of humans and rhesus macaques, the very homologous (approximately 98-99% identical) CCR5 of African green monkeys (AGMs) avidly binds beta-chemokines and functions as a coreceptor for simian immunodeficiency viruses. However, AGM CCR5 is a weak coreceptor for tested macrophage-tropic (R5) isolates of human immunodeficiency virus type 1 (HIV-1). Correspondingly, gp120 envelope glycoproteins derived from R5 isolates of HIV-1 bind poorly to AGM CCR5. We focused on a unique extracellular amino acid substitution at the juncture of transmembrane helix 4 (TM4) and extracellular loop 2 (ECL2) (Arg for Gly at amino acid 163 (G163R)) as the likely source of the weak R5 gp120 binding and HIV-1 coreceptor properties of AGM CCR5. Accordingly, a G163R mutant of human CCR5 was severely attenuated in its ability to bind R5 gp120s and to mediate infection by R5 HIV-1 isolates. Conversely, the R163G mutant of AGM CCR5 was substantially strengthened as a coreceptor for HIV-1 and had improved R5 gp120 binding affinity relative to the wild-type AGM CCR5. These substitutions at amino acid position 163 had no effect on chemokine binding or signal transduction, suggesting the absence of structural alterations. The 2D7 monoclonal antibody has been reported to bind to ECL2 and to block HIV-1 binding and infection. Whereas 2D7 antibody binding to CCR5 was unaffected by the G163R mutation, it was prevented by a conservative ECL2 substitution (K171R), shared between rhesus and AGM CCR5s. Thus, it appears that the 2D7 antibody binds to an epitope that includes Lys-171 and may block HIV-1 infection mediated by CCR5 by occluding an HIV-1-binding site in the vicinity of Gly-163. In summary, our results identify a site for gp120 interaction that is critical for R5 isolates of HIV-1 in the central core of human CCR5, and we propose that this site collaborates with a previously identified region in the CCR5 amino terminus to enable gp120 binding and HIV-1 infections.


Assuntos
Glicina/metabolismo , HIV-1/fisiologia , Fusão de Membrana/fisiologia , Receptores CCR5/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Sítios de Ligação de Anticorpos , Ligação Competitiva , Linhagem Celular , Chlorocebus aethiops , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/patogenicidade , Humanos , Macaca mulatta , Dados de Sequência Molecular , Receptores CCR5/química , Receptores CCR5/imunologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Especificidade da Espécie
20.
J Virol ; 72(12): 10251-5, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9811770

RESUMO

The vif gene of human immunodeficiency virus type 1 (HIV-1) encodes a basic Mr 23,000 protein that is necessary for production of infectious virions by nonpermissive cells (human lymphocytes and macrophages) but not by permissive cells such as HeLa-CD4. It had been proposed that permissive cells may contain an unidentified factor that functions like the viral Vif protein. To test this hypothesis, we produced pseudotyped wild-type and vif-deleted HIV gpt virions (which contain the HIV-1 genome with the bacterial mycophenolic acid resistance gene gpt in place of the viral env gene) in permissive cells, and we used them to generate nonpermissive H9 leukemic T cells that express these proviruses. We then fused these H9 cells with permissive HeLa cells that express the HIV-1 envelope glycoprotein gp120-gp41, and we asked whether the heterokaryons would release infectious HIV gpt virions. The results clearly showed that the vif-deleted virions released by the heterokaryons were noninfectious whereas the wild-type virions were highly infectious. This strongly suggests that nonpermissive cells, the natural targets of HIV-1, contain a potent endogenous inhibitor of HIV-1 replication that is overcome by Vif.


Assuntos
Antivirais/antagonistas & inibidores , Antivirais/fisiologia , Produtos do Gene vif/fisiologia , HIV-1/patogenicidade , Linfócitos/imunologia , Linfócitos/virologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Células COS , Linhagem Celular , Efeito Citopatogênico Viral , Deleção de Genes , Produtos do Gene vif/genética , Genes vif , Teste de Complementação Genética , HIV-1/genética , HIV-1/fisiologia , Células HeLa , Humanos , Virulência/genética , Virulência/fisiologia , Produtos do Gene vif do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...