Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Retrovirology ; 3: 54, 2006 Aug 22.
Article in English | MEDLINE | ID: mdl-16925802

ABSTRACT

BACKGROUND: The retroviral restriction factor tripartite motif protein (TRIM)5alpha, is characterized by marked amino acid diversity among primates, including specific clusters of residues under positive selection. The identification of multiple non-synonymous changes in humans suggests that TRIM5alpha variants might be relevant to retroviral pathogenesis. Previous studies have shown that such variants are unlikely to modify susceptibility to HIV-1 infection, or the course of early infection. However, the longterm effect of carrying Trim5alpha variants on disease progression in individuals infected with HIV-1 has not previously been investigated. METHODS: In a cohort of 979 untreated individuals infected with HIV-1 with median follow up 3.2 years and 9,828 CD4 T cell measurements, we analysed common amino acid variations: H43Y, V112F, R136Q, G249D, and H419Y. The rate of CD4 T cell decline before treatment was used as the phenotype. In addition, we extended previous work on the in vitro susceptibility of purified donor CD4 T cells (n = 125) to HIV-1 infection, and on the susceptibility of HeLa cells that were stably transduced with the different TRIM5 variants. Haplotypes were analysed according to the most parsimonious evolutionary structure, where two main human TRIM5alpha groups can be defined according to the residue at amino acid 136. Humans present both Q136 and R136 at similar frequency, and additional TRIM5alpha amino acid variants are almost exclusively derived from R136-carrying haplotypes. RESULTS: We observed modest differences in disease progression for evolutionary branches carrying R136-derived haplotypes, and with the non-synonymous polymorphisms G249D and H419Y. In vitro analysis of susceptibility of donor CD4 T cells, and of the various transduced HeLa cell lines supported the absence of significant differential restriction of HIV-1 infection by the various huTRIM5alpha alleles. CONCLUSION: Common human variants of TRIM5alpha have no effect or modest effect on HIV-1 disease progression. These variants occur at sites conserved throughout evolution, and are remote from clusters of positive selection in the primate lineage. The evolutionary value of the substitutions remains unclear.


Subject(s)
Carrier Proteins/genetics , HIV Infections/genetics , HIV-1 , Adult , Antiviral Restriction Factors , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Cohort Studies , Disease Progression , Female , Genetic Predisposition to Disease , HIV Infections/immunology , HIV Infections/pathology , HeLa Cells , Humans , Male , Polymorphism, Single Nucleotide , Protein Isoforms , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
2.
Retrovirology ; 3: 11, 2006 Feb 07.
Article in English | MEDLINE | ID: mdl-16460575

ABSTRACT

BACKGROUND: Evolutionary analysis may serve as a useful approach to identify and characterize host defense and viral proteins involved in genetic conflicts. We analyzed patterns of coding sequence evolution of genes with known (TRIM5alpha and APOBEC3G) or suspected (TRIM19/PML) roles in virus restriction, or in viral pathogenesis (PPIA, encoding Cyclophilin A), in the same set of human and non-human primate species. RESULTS AND CONCLUSION: This analysis revealed previously unidentified clusters of positively selected sites in APOBEC3G and TRIM5alpha that may delineate new virus-interaction domains. In contrast, our evolutionary analyses suggest that PPIA is not under diversifying selection in primates, consistent with the interaction of Cyclophilin A being limited to the HIV-1M/SIVcpz lineage. The strong sequence conservation of the TRIM19/PML sequences among primates suggests that this gene does not play a role in antiretroviral defense.


Subject(s)
Evolution, Molecular , Proteins/genetics , Retroviridae/pathogenicity , Animals , Genomics , Host-Parasite Interactions/genetics , Primates/genetics , Primates/virology , Retroviridae/genetics , Selection, Genetic , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity
3.
Pharmacogenetics ; 14(4): 255-60, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15083070

ABSTRACT

A recent study with 69 Japanese liver transplants treated with tacrolimus found that the MDR13435 C >T polymorphism, but not the MDR12677 G >T polymorphism, was associated with differences in the intestinal expression level of CYP3A4 mRNA. In the present study, over 6 h, we measured the kinetics of a 75 microg oral dose of midazolam, a CYP3A substrate, in 21 healthy subjects genotyped for the MDR13435 C >T and 2677 G >T polymorphism. No statistically significant differences were found in the calculated pharmacokinetic parameters between the three 3435 C >T genotypes (TT, CT and CC group, respectively: Cmax (mean +/- SD: 0.30 +/- 0.08 ng/ml, 0.31 +/- 0.09 ng/ml and 0.31 +/- 0.11 ng/ml; Apparent clearance: 122 +/- 29 l/h, 156 +/- 92 l/h and 111 +/- 35 l/h; t1/2: 1.9 +/- 1.1 h, 1.6 +/- 0.90 h and 1.7 +/- 0.7 h). In addition, the 30-min 1'OH midazolam to midazolam ratio, a marker of CYP3A activity, determined in 74 HIV-positive patients before the introduction of antiretroviral treatment, was not significantly different between the three 3435 C >T genotypes (mean ratio +/- SD: 3.65 +/- 2.24, 4.22 +/- 3.49 and 4.24 +/- 2.03, in the TT, CT and CC groups, respectively). Similarly, no association was found between the MDR12677 G >T polymorphism and CYP3A activity in the healthy subjects or in the HIV-positive patients. The existence of a strong association between the activity of CYP3A and MDR13435 C >T and 2677 G >T polymorphisms appears unlikely, at least in Caucasian populations and/or in the absence of specific environmental factors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Cytochrome P-450 Enzyme System/metabolism , Midazolam/pharmacology , Polymorphism, Genetic , Adult , Base Sequence , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , DNA Primers , Female , Humans , Male , Midazolam/pharmacokinetics , Middle Aged , RNA, Messenger/genetics
4.
AIDS Res Hum Retroviruses ; 20(4): 399-407, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15157358

ABSTRACT

The Nef protein plays a major role in vivo in promoting HIV and SIV replication and pathogenesis. In vitro, Nef has been shown to down-regulate cell surface molecules, such as CD4 and MHC-I, alter T cell signaling, and enhance virion infectivity. These effects are attributed to interactions of Nef with cellular proteins. In addition, HIV Nef is incorporated into viral particles, mainly localizing in the virion cores. However, no report has been published to date regarding Nef interactions with virion proteins. By immunoprecipitation, Nef was found to bind to viral enzymes. Using yeast two-hybrid and GST pulldown procedures to find out direct potential partners of Nef, Nef was consistently found to interact with viral integrase (IN). The interaction between Nef and IN was stronger when Nef was present as the viral protease-cleaved isoform. We hypothesize that the interaction of Nef with viral integrase or other virion proteins may explain the presence of Nef in viral cores. In addition, this interaction suggests that Nef may accompany the reverse transcription and the preintegration complexes during the early steps of the infection cycle and potentially affect infectivity during these steps.


Subject(s)
Gene Products, nef/metabolism , HIV-1/metabolism , Viral Core Proteins/metabolism , Gene Products, nef/chemistry , HIV Integrase/metabolism , HIV Protease/metabolism , HIV Reverse Transcriptase/metabolism , Protein Binding , Two-Hybrid System Techniques , Viral Core Proteins/chemistry , Virion/metabolism , nef Gene Products, Human Immunodeficiency Virus
5.
J Virol ; 80(14): 6757-63, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809281

ABSTRACT

Tumor susceptibility gene 101 (TSG101) encodes a host cellular protein that is appropriated by human immunodeficiency virus type 1 (HIV-1) in the budding process of viral particles from infected cells. Variation in the coding or noncoding regions of the gene could potentially affect the degree of TSG101-mediated release of viral particles. While the coding regions of the gene were found to lack nonsynonymous variants, two polymorphic sites in the TSG101 5' area were identified that were associated with the rate of AIDS progression among Caucasians. These single-nucleotide polymorphisms (SNPs), located at positions -183 and +181 relative to the translation start, specify three haplotypes termed A, B, and C, which occur at frequencies of 67%, 21%, and 12%, respectively. Haplotype C is associated with relatively rapid AIDS progression, while haplotype B is associated with slower disease progression. Both effects were dominant over the intermediate haplotype A. The haplotypes also demonstrated parallel effects on the rate of CD4 T-cell depletion and viral load increase over time, as well as a possible influence on HIV-1 infection. The data raise the hypothesis that noncoding variation in TSG101 affects the efficiency of TSG101-mediated release of viral particles from infected cells, thereby altering levels of plasma viral load and subsequent disease progression.


Subject(s)
Acquired Immunodeficiency Syndrome/genetics , DNA-Binding Proteins/genetics , Gene Frequency/genetics , HIV-1 , Open Reading Frames/genetics , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Acquired Immunodeficiency Syndrome/blood , CD4-Positive T-Lymphocytes/virology , Cohort Studies , Disease Progression , Endosomal Sorting Complexes Required for Transport , Female , Haplotypes/genetics , Humans , Male , Time Factors , Viral Load , Virus Shedding/genetics
6.
J Virol ; 79(20): 12674-80, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16188970

ABSTRACT

Humans differ substantially with respect to susceptibility to human immunodeficiency virus type 1 (HIV-1). We evaluated variants of nine host genes participating in the viral life cycle for their role in modulating HIV-1 infection. Alleles were assessed ex vivo for their impact on viral replication in purified CD4 T cells from healthy blood donors (n = 128). Thereafter, candidate alleles were assessed in vivo in a cohort of HIV-1-infected individuals (n = 851) not receiving potent antiretroviral therapy. As a benchmark test, we tested 12 previously reported host genetic variants influencing HIV-1 infection as well as single nucleotide polymorphisms in the nine candidate genes. This led to the proposition of three alleles of PML, TSG101, and PPIA as potentially associated with differences in progression of HIV-1 disease. In a model considering the combined effects of new and previously reported gene variants, we estimated that their effect might be responsible for lengthening or shortening by up to 2.8 years the period from 500 CD4 T cells/mul to <200 CD4 T cells/mul.


Subject(s)
HIV Infections/genetics , HIV-1/physiology , Alleles , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Genetic Predisposition to Disease , HIV Infections/virology , Humans , Virus Replication
7.
J Infect Dis ; 192(8): 1381-6, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16170755

ABSTRACT

BACKGROUND: Unconjugated hyperbilirubinemia results from Gilbert syndrome and from antiretroviral therapy (ART) containing protease inhibitors. An understanding of the interaction between genetic predisposition and ART may help to identify individuals at highest risk for developing jaundice. METHODS: We quantified the contribution of UGT1A1*28 and ART to hyperbilirubinemia by longitudinally modeling 1386 total bilirubin levels in 96 human immunodeficiency virus (HIV)-infected individuals during a median of 6 years. RESULTS: The estimated average bilirubin level was 8.8 micromol/L (0.51 mg/dL). Atazanavir increased bilirubin levels by 15 mu mol/L (0.87 mg/dL), and indinavir increased bilirubin levels by 8 micromol/L (0.46 mg/dL). Ritonavir, lopinavir, saquinavir, and nelfinavir had no or minimal effect on bilirubin levels. Homozygous UGT1A1*28 increased bilirubin levels by 5.2 micromol/L (0.3 mg/dL). As a consequence, 67% of individuals homozygous for UGT1A1*28 and receiving atazanavir or indinavir had > or =2 episodes of hyperbilirubinemia in the jaundice range (>43 micromol/L [>2.5 mg/dL]), versus 7% of those with the common allele and not receiving either of those protease inhibitors (P<.001). Efavirenz resulted in decreased bilirubin levels, which is consistent with the induction of UDP-glucuronosyltransferase 1A1. CONCLUSIONS: Genotyping for UGT1A1*28 before initiation of ART would identify HIV-infected individuals at risk for hyperbilirubinemia and decrease episodes of jaundice.


Subject(s)
Antiretroviral Therapy, Highly Active/adverse effects , Gilbert Disease/complications , Glucuronosyltransferase/genetics , Hyperbilirubinemia/chemically induced , Adult , Cohort Studies , Female , Humans , Hyperbilirubinemia/complications , Male , Middle Aged , Treatment Outcome
8.
Pharmacogenet Genomics ; 15(9): 599-608, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16041239

ABSTRACT

OBJECTIVES: The human immunodeficiency virus protease inhibitor nelfinavir is substrate of polyspecific drug transporters encoded by ABCB1 (P-glycoprotein), ABCC1 (MRP1) and ABCC2 (MRP2), and an inhibitor of BCRP, encoded by ABCG2. Genetic polymorphism in these genes may be associated with changes in transport function. METHODS: A comprehensive evaluation of single nucleotide polymorphisms (39 SNPs in ABCB1, 7 in ABCC1, 27 in ABCC2, and 16 in ABCG2), and inferred haplotypes was done to assess possible associations of genetic variants with cellular exposure of nelfinavir in vivo. Analysis used peripheral mononuclear cells from individuals receiving nelfinavir (n=28). Key results were re-examined in a larger sample size (n=129) contributing data on plasma drug levels. RESULTS AND CONCLUSIONS: There was no significant association between cellular nelfinavir area under the curve (AUC) and SNPs or haplotypes at ABCC1, ABCC2, ABCG2. There was an association with cellular exposure for two loci in strong linkage disequilibrium: ABCB1 3435C>T; AUCTT>AUCCT>AUCCC (ratio 2.1, 1.4, 1, Ptrend=0.01), and intron 26 +80T>C; AUCCC> AUCCT > AUCTT (ratio 2.4, 1.3, 1, Ptrend=0.006). Haplotypic analysis using tagging SNPs did not improve the single SNP association values.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/genetics , HIV Protease Inhibitors/pharmacology , Mitochondrial Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Nelfinavir/pharmacology , Neoplasm Proteins/genetics , Pharmacogenetics/methods , Polymorphism, Genetic , Ribosomal Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Area Under Curve , Cohort Studies , Genetic Variation , Genotype , HIV Seropositivity/drug therapy , HIV Seropositivity/genetics , Haplotypes , Humans , Leukocytes, Mononuclear/cytology , Multidrug Resistance-Associated Protein 2 , Phenotype
9.
Pharmacogenet Genomics ; 15(1): 1-5, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15864119

ABSTRACT

BACKGROUND: Efavirenz (EFV) and nevirapine (NVP) are metabolized by cytochrome P450 2B6 (CYP2B6). Allele 516 G>T (Gln172His) is associated with diminished activity of this isoenzyme, and may lead to differences in drug exposure. METHODS: We evaluated this allele as a pharmacogenetic marker of EFV and NVP pharmacokinetics and EFV toxicity in 167 participants receiving EFV and 59 receiving NVP recruited within the genetics project of the Swiss HIV Cohort Study. Drug concentrations were measured in plasma and in peripheral blood mononuclear cells (PBMCs) from the same sample. Neuropsychological toxicity of EFV (sleep disorders, mood disorders, fatigue) was assessed using a standardized questionnaire. RESULTS AND CONCLUSIONS: CYP2B6 516TT was associated with greater plasma and intracellular exposure to EFV, and greater plasma exposure to NVP. Intracellular drug concentration, and CYP2B6 genotype were predictors of EFV neuropsychological toxicity. CYP2B6 genotyping may be useful to complement an individualization strategy based on plasma drug determinations to increase the safety and tolerability of EFV.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , HIV Infections/drug therapy , HIV Infections/genetics , Nevirapine/pharmacology , Oxazines/pharmacology , Oxidoreductases, N-Demethylating/genetics , Polymorphism, Genetic , Adult , Alkynes , Alleles , Anti-HIV Agents/pharmacology , Benzoxazines , Cyclopropanes , Cytochrome P-450 CYP2B6 , Female , Genotype , HIV Seropositivity , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Odds Ratio , Pharmacogenetics , Protein Isoforms , Regression Analysis , Time Factors
10.
J Infect Dis ; 191(9): 1419-26, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15809899

ABSTRACT

BACKGROUND: Single-nucleotide polymorphisms in genes involved in lipoprotein and adipocyte metabolism may explain why dyslipidemia and lipoatrophy occur in some but not all antiretroviral therapy (ART)-treated individuals. METHODS: We evaluated the contribution of APOC3 -482C-->T, -455T-->C, and 3238C-->G; epsilon 2 and epsilon 4 alleles of APOE; and TNF -238G-->A to dyslipidemia and lipoatrophy by longitudinally modeling >2600 lipid determinations and 2328 lipoatrophy assessments in 329 ART-treated patients during a median follow-up period of 3.4 years. RESULTS: In human immunodeficiency virus (HIV)-infected individuals, the effects of variant alleles of APOE on plasma cholesterol and triglyceride levels and of APOC3 on plasma triglyceride levels were comparable to those reported in the general population. However, when treated with ritonavir, individuals with unfavorable genotypes of APOC3 and [corrected] APOE were at risk of extreme hypertriglyceridemia. They had median plasma triglyceride levels of 7.33 mmol/L, compared with 3.08 mmol/L in the absence of ART. The net effect of the APOE*APOC3*ritonavir interaction was an increase in plasma triglyceride levels of 2.23 mmol/L. No association between TNF -238G-->A and lipoatrophy was observed. CONCLUSIONS: Variant alleles of APOE and APOC3 contribute to an unfavorable lipid profile in patients with HIV. Interactions between genotypes and ART can lead to severe hyperlipidemia. Genetic analysis may identify patients at high risk for severe ritonavir-associated hypertriglyceridemia.


Subject(s)
Anti-HIV Agents/adverse effects , Apolipoproteins C/genetics , Apolipoproteins E/genetics , HIV Infections/drug therapy , Polymorphism, Single Nucleotide , Tumor Necrosis Factor-alpha/genetics , Alleles , Apolipoprotein C-III , CD4 Lymphocyte Count , Cohort Studies , Female , Genetic Variation , HIV Infections/blood , HIV Infections/genetics , Humans , Hypertriglyceridemia/chemically induced , Hypertriglyceridemia/genetics , Male , Middle Aged , Multivariate Analysis , Risk Assessment , Switzerland
11.
J Infect Dis ; 190(11): 1979-88, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15529263

ABSTRACT

To characterize newly arising replication of human immunodeficiency virus (HIV) type 1 in vivo at the cellular level, distinct viral RNA species in peripheral blood mononuclear cells (PBMCs) from HIV-1-infected patients were monitored during 2 weeks of structured treatment interruption (STI). HIV-1 RNA encoding tat/rev and PBMC-associated virions were almost completely depleted during antiretroviral therapy and emerged simultaneously after 2 weeks of STI, thus specifically reflecting productive viral infection at the cellular level. The magnitude of these correlates of reappearing cellular viral replication was predicted by during-therapy levels of nef transcripts in PBMCs. Significant rebound of plasma viremia, representing the progeny of a broader range of anatomical compartments, preceded and predicted productive infection in PBMCs. Thus, cellular viral rebound in PBMCs likely was primed before STI by the expression of nef in HIV-1-infected PBMCs that lacked virion production and was subsequently triggered by the plasma viremia that preceded the recurrence of productively infected PBMCs.


Subject(s)
Anti-HIV Agents/therapeutic use , Gene Products, nef/biosynthesis , HIV Infections/drug therapy , HIV-1/isolation & purification , Leukocytes, Mononuclear/virology , RNA Splicing , RNA, Viral/biosynthesis , Cells, Cultured , Drug Therapy, Combination , Gene Products, nef/analysis , Gene Products, rev/metabolism , Gene Products, tat/metabolism , HIV Infections/blood , HIV Infections/virology , HIV-1/genetics , Humans , Neutrophils/virology , RNA, Viral/analysis , Recurrence , Virus Replication , nef Gene Products, Human Immunodeficiency Virus , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
12.
J Gen Virol ; 85(Pt 4): 921-927, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15039534

ABSTRACT

The human immunodeficiency virus type 1 p6 region encodes p6(Gag) and the transframe p6(Pol) protein. The Gag frame encodes an N-terminal late assembly L domain and a C-terminal Vpr binding domain. In the Pol frame, substitution at a C-terminal motif decreases protease autocleavage. The role of the highly polymorphic central region of p6, comprising amino acids S14-I31 (p6(Gag)) and R20-D39 (p6(Pol)), is unclear. Analysis of this central region demonstrated that 35 % of p6(Gag) appears to be dispensable for virus propagation in vitro and smaller deletion and insertion polymorphisms can be tolerated in vivo. Extensive Pol deletion (deltaR20-D39, 42 % of p6(Pol)) did not alter protease autocleavage.


Subject(s)
Gene Products, gag/genetics , Gene Products, gag/physiology , HIV-1/genetics , HIV-1/physiology , Amino Acid Sequence , Gene Products, gag/chemistry , Gene Products, pol/chemistry , Gene Products, pol/genetics , Gene Products, pol/physiology , HIV Infections/virology , HIV-1/isolation & purification , HIV-1/pathogenicity , Humans , In Vitro Techniques , Molecular Sequence Data , Phenotype , Polymorphism, Genetic , Recombination, Genetic , Sequence Deletion , Virulence/genetics , Virulence/physiology , Virus Replication/genetics , Virus Replication/physiology
13.
J Infect Dis ; 189(4): 583-6, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14767809

ABSTRACT

Nonphysiological overexpression of the ABC transporter P-glycoprotein (P-gp), which is encoded by MDR1, has been associated with reduced susceptibility to human immunodeficiency virus (HIV) type 1 infection in vitro. We analyzed (1) the expression and genotype of MDR1 and their relationship to HIV-1 permissiveness of CD4+ T cells from 128 healthy blood donors and (2) the role that alleles of MDR1 exons 21 and 26 play in modifying disease progression in 411 HIV-1-infected individuals. Differences in physiological levels of MDR1 expression did not modify HIV-1 infection in vitro, nor did MDR1 alleles and haplotypes significantly influence either permissiveness to infection in vitro or disease progression in vivo before the initiation of treatment.


Subject(s)
Acquired Immunodeficiency Syndrome/genetics , Genes, MDR/genetics , HIV Infections/genetics , Polymorphism, Genetic , Acquired Immunodeficiency Syndrome/physiopathology , Base Sequence , CD4-Positive T-Lymphocytes/immunology , Cohort Studies , DNA Primers , Disease Progression , Genetic Variation , Genotype , HIV Infections/physiopathology , HIV-1 , Humans
14.
J Virol ; 76(15): 7868-73, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12097600

ABSTRACT

Human immunodeficiency virus type 1 uses ribosomal frameshifting for translation of the Gag-Pol polyprotein. Frameshift activities are thought to be tightly regulated. Analysis of gag p1 sequences from 270 plasma virions identified in 64% of the samples the occurrence of polymorphism that could lead to changes in thermodynamic stability of the stem-loop. Expression in Saccharomyces cerevisiae of p1-beta-galactosidase fusion proteins from 10 representative natural stem-loop variants and three laboratory mutant constructs (predicted the thermodynamic stability [Delta G degrees] ranging from -23.0 to -4.3 kcal/mol) identified a reduction in frameshift activity of 13 to 67% compared with constructs with the wild-type stem-loop (Delta G degrees, -23.5 kcal/mol). Viruses carrying stem-loops associated with greater than 60% reductions in frameshift activity presented profound defects in viral replication. In contrast, viruses with stem-loop structures associated with 16 to 42% reductions in frameshift efficiency displayed no significant viral replication deficit.


Subject(s)
Frameshifting, Ribosomal , Genes, gag/genetics , Genes, pol/genetics , Genetic Variation , HIV-1/genetics , Nucleic Acid Conformation , Base Sequence , Genes, pol/physiology , HIV-1/pathogenicity , Humans , Leukocytes, Mononuclear/virology , Molecular Sequence Data , Mutagenesis, Site-Directed , Thermodynamics , Virion/metabolism , Virus Replication
15.
Eur J Clin Pharmacol ; 60(4): 237-46, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15114429

ABSTRACT

OBJECTIVE: We investigated whether the oral administration of a low dose (75 micro g) of midazolam, a CYP3A probe, can be used to measure the in vivo CYP3A activity. METHODS: Plasma concentrations of midazolam, 1'OH-midazolam and 4'OH-midazolam were measured after the oral administration of 7.5 mg and 75 micro g midazolam in 13 healthy subjects without medication, in four subjects pretreated for 2 days with ketoconazole (200 mg b.i.d.), a CYP3A inhibitor, and in four subjects pretreated for 4 days with rifampicin (450 mg q.d.), a CYP3A inducer. RESULTS: After oral administration of 75 micro g midazolam, the 30-min total (unconjugated + conjugated) 1'OH-midazolam/midazolam ratios measured in the groups without co-medication, with ketoconazole and with rifampicin were (mean+/-SD): 6.23+/-2.61, 0.79+/-0.39 and 56.1+/-12.4, respectively. No side effects were reported by the subjects taking this low dose of midazolam. Good correlations were observed between the 30-min total 1'OH-midazolam/midazolam ratio and midazolam clearance in the group without co-medication (r(2)=0.64, P<0.001) and in the three groups taken together (r(2)=0.91, P<0.0001). Good correlations were also observed between midazolam plasma levels and midazolam clearance, measured between 1.5 h and 4 h. CONCLUSION: A low oral dose of midazolam can be used to phenotype CYP3A, either by the determination of total 1'OH-midazolam/midazolam ratios at 30 min or by the determination of midazolam plasma levels between 1.5 h and 4 h after its administration.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Midazolam/analogs & derivatives , Midazolam/administration & dosage , Midazolam/pharmacokinetics , Oxidoreductases, N-Demethylating/metabolism , Adult , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/biosynthesis , Cross-Over Studies , Cytochrome P-450 CYP3A , Dose-Response Relationship, Drug , Enzyme Induction , Female , Humans , Ketoconazole/pharmacology , Male , Midazolam/adverse effects , Midazolam/blood , Middle Aged , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/biosynthesis
16.
Eur J Clin Pharmacol ; 60(4): 231-6, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15114431

ABSTRACT

OBJECTIVE: We investigated whether differences in pharmacokinetics of midazolam, a CYP3A probe, could be demonstrated between subjects with different CYP3A4 and CYP3A5 genotypes. METHODS: Plasma concentrations of midazolam, and of total (conjugated + unconjugated) 1'OH-midazolam, and 4'OH-midazolam were measured after the oral administration of 7.5 mg or of 75 micro g of midazolam in 21 healthy subjects. RESULTS: CYP3A5*7, CYP3A4*1E, CYP3A4*2, CYP3A4*4, CYP3A4*5, CYP3A4*6, CYP3A4*8, CYP3A4*11, CYP3A4*12, CYP3A4*13, CYP3A4*17 and CYP3A4*18 alleles were not identified in the 21 subjects. CYP3A5*3, CYP3A5*6, CYP3A4*1B and CYP3A4*1F alleles were identified in 20, 1, 4 and 2 subjects, respectively. No statistically significant differences were observed for the AUC(inf) values between the different genotypes after the 75- micro g or the 7.5-mg dose. CONCLUSION: Presently, CYP3A4 and CYP3A5 genotyping methods do not sufficiently reflect the inter-individual variability of CYP3A activity.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Midazolam/analogs & derivatives , Midazolam/pharmacokinetics , Area Under Curve , Cytochrome P-450 CYP3A , Female , Genotype , Humans , Male , Midazolam/blood
17.
J Virol ; 78(19): 10747-54, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367641

ABSTRACT

Isolated primary human cells from different donors vary in their permissiveness-the ability of cells to be infected and sustain the replication of human immunodeficiency virus type 1 (HIV-1). We used replicating HIV-1 and single-cycle lentivirus vectors in a population approach to identify polymorphic steps during viral replication. We found that phytohemagglutinin-stimulated CD4(+) CD45RO(+) CD57(-) T cells from healthy blood donors (n = 128) exhibited a 5.2-log-unit range in virus production. For 20 selected donors representing the spectrum of CD4 T-cell permissiveness, we could attribute up to 42% of the total variance in virus production to entry factors and 48% to postentry steps. Efficacy at key intracellular steps of the replicative cycle (reverse transcription, integration, transcription and splicing, translation, and budding and release) varied from 0.71 to 1.45 log units among donors. However, interindividual differences in transcription efficiency alone accounted for 64 to 83% of the total variance in virus production that was attributable to postentry factors. While vesicular stomatitis virus G protein-mediated fusion was more efficacious than CCR5/CD4 entry, the latter resulted in greater transcriptional activity per proviral copy. The phenotype of provirus transcription was stable over time, indicating that it represents a genetic trait.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV-1/growth & development , Transcription, Genetic , Virus Replication , Biological Transport , CD4 Antigens/analysis , CD57 Antigens/analysis , Cells, Cultured , DNA, Viral/analysis , HIV Core Protein p24/analysis , Humans , Leukocyte Common Antigens/analysis , Protein Biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Proviruses/physiology , RNA Splicing , RNA, Viral/analysis , Receptors, CCR5/analysis , Time Factors , Vesicular stomatitis Indiana virus , Virus Integration
18.
J Virol ; 78(20): 11070-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15452227

ABSTRACT

The cytosine deaminase APOBEC3G, in the absence of the human immunodeficiency virus type 1 (HIV-1) accessory gene HIV-1 viral infectivity factor (vif), inhibits viral replication by introducing G-->A hypermutation in the newly synthesized HIV-1 DNA negative strand. We tested the hypothesis that genetic variants of APOBEC3G may modify HIV-1 transmission and disease progression. Single nucleotide polymorphisms were identified in the promoter region (three), introns (two), and exons (two). Genotypes were determined for 3,073 study participants enrolled in six HIV-AIDS prospective cohorts. One codon-changing variant, H186R in exon 4, was polymorphic in African Americans (AA) (f = 37%) and rare in European Americans (f < 3%) or Europeans (f = 5%). For AA, the variant allele 186R was strongly associated with decline in CD4 T cells (CD4 slope on square root scale: -1.86, P = 0.009), The 186R allele was also associated with accelerated progression to AIDS-defining conditions in AA. The in vitro antiviral activity of the 186R enzyme was not inferior to that of the common H186 variant. These studies suggest that there may be a modifying role of variants of APOBEC3G on HIV-1 disease progression that warrants further investigation.


Subject(s)
Genetic Variation , HIV Infections/genetics , HIV Infections/physiopathology , HIV-1/pathogenicity , Proteins/genetics , APOBEC-3G Deaminase , Black People , Cell Line , Cohort Studies , Cytidine Deaminase , Disease Progression , Europe , Haplotypes , Humans , Nucleoside Deaminases , Polymorphism, Single Nucleotide , Repressor Proteins , United States , White People
SELECTION OF CITATIONS
SEARCH DETAIL