Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 10(3): e0121059, 2015.
Article in English | MEDLINE | ID: mdl-25803873

ABSTRACT

Cell fate is determined by the balance of conserved molecular mechanisms regulating death (apoptosis) and survival (autophagy). Autophagy is a process by which cells recycle their organelles and macromolecules through degradation within the vacuole in yeast and plants, and lysosome in metazoa. In the yeast Schizosaccharomyces pombe, autophagy is strongly induced under nitrogen starvation and in aging cells. Previously, we demonstrated that calnexin (Cnx1p), a highly conserved transmembrane chaperone of the endoplasmic reticulum (ER), regulates apoptosis under ER stress or inositol starvation. Moreover, we showed that in stationary phase, Cnx1p is cleaved into two moieties, L_Cnx1p and S_Cnx1p. Here, we show that the processing of Cnx1p is regulated by autophagy, induced by nitrogen starvation or cell aging. The cleavage of Cnx1p involves two vacuolar proteases: Isp6, which is essential for autophagy, and its paralogue Psp3. Blocking autophagy through the knockout of autophagy-related genes (atg) results in inhibition of both, the cleavage and the trafficking of Cnx1p from the ER to the vacuole. We demonstrate that Cnx1p is required for cell survival under nitrogen-starvation and in chronological aging cultures. The death of the mini_cnx1 mutant (overlapping S_cnx1p) cells is accompanied by accumulation of high levels of reactive-oxygen species (ROS), a slowdown in endocytosis and severe cell-wall defects. Moreover, mutant cells expressing only S_Cnx1p showed cell wall defects. Co-expressing mutant overlapping the L_Cnx1p and S_Cnx1p cleavage products reverses the death, ROS phenotype and cell wall defect to wild-type levels. As it is involved in both apoptosis and autophagy, Cnx1p could be a nexus for the crosstalk between these pro-death and pro-survival mechanisms. Ours, and observations in mammalian systems, suggest that the multiple roles of calnexin depend on its sub-cellular localization and on its cleavage. The use of S. pombe should assist in further shedding light on the multiple roles of calnexin.


Subject(s)
Autophagy/physiology , Calnexin/metabolism , Nitrogen/deficiency , Schizosaccharomyces/physiology , Calnexin/genetics , Cell Wall/pathology , Genetic Vectors/genetics , Immunoblotting , Membrane Proteins/metabolism , Microscopy, Fluorescence , Oligonucleotides/genetics , Reactive Oxygen Species/metabolism , Schizosaccharomyces pombe Proteins/metabolism
3.
Biochem Pharmacol ; 78(11): 1382-90, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19631193

ABSTRACT

CXCR4, a G-protein-coupled receptor, which binds the chemokine stromal cell-derived factor 1 alpha (SDF-1alpha, CXCL12), is one of two co-receptors most frequently used by HIV-1 to infect CD4+ lymphocytes. The SDF-1alpha/CXCR4 axis is also involved in angiogenesis, in stem cell homing to bone marrow, in rheumatoid arthritis and in cancer. Here, we directly determined the binding site of the inverse agonist T140 on CXCR4 using photoaffinity labeling. Two T140 photoanalogs were synthesized containing the photoreactive amino acid p-benzoyl-l-phenylalanine (Bpa) in positions 5 or 10, yielding [Bpa(5)]T140 and [Bpa(10)]T140. Binding experiments on HEK293 cells stably expressing the wild-type CXCR4 receptor using 125I-SDF-1alpha demonstrated that T140 and both photoanalogs had affinities in the nanomolar range, similar to SDF-1alpha. Photolabeling led to the formation of specific, covalent 42 kDa T140-CXCR4 complexes. V8 protease digestion of both CXCR4/125I-[Bpa(5)]T140 and CXCR4/125I-[Bpa(10)]T140 adducts generated a fragment of 6kDa suggesting that the T140 photoanalogs labeled a fragment corresponding to Lys(154)-Glu(179) of the receptor's 4th transmembrane domain. Further digestion of this 6kDa fragment with endo Asp-N led to the generation of a shorter fragment validating the photolabeled region. Our results demonstrate that T140 interacts with residues of the fourth transmembrane domain of the CXCR4 receptor and provide new structural constraints enabling us to model the complex between T140 and CXCR4.


Subject(s)
Benzophenones/chemistry , Oligopeptides/metabolism , Phenylalanine/analogs & derivatives , Photoaffinity Labels , Receptors, CXCR4/metabolism , Amino Acid Sequence , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Binding Sites , Binding, Competitive , Cell Line , DNA, Viral/genetics , HIV-1/genetics , Humans , Iodine Radioisotopes , Models, Molecular , Molecular Sequence Data , Oligopeptides/chemistry , Oligopeptides/pharmacology , Phenylalanine/chemistry , Protein Structure, Tertiary , Proviruses/genetics , Receptors, CXCR4/chemistry , Structure-Activity Relationship
4.
RNA ; 14(5): 981-91, 2008 May.
Article in English | MEDLINE | ID: mdl-18367719

ABSTRACT

The human immunodeficiency virus of type 1 (HIV-1) uses a programmed -1 ribosomal frameshift to produce the precursor of its enzymes, and changes in frameshift efficiency reduce replicative fitness of the virus. We used a fluorescent two-reporter system to screen for peptides that reduce HIV-1 frameshift in bacteria, knowing that the frameshift can be reproduced in Escherichia coli. Expression of one reporter, the green fluorescent protein (GFP), requires the HIV-1 frameshift, whereas the second reporter, the red fluorescent protein (RFP), is used to assess normal translation. A peptide library biased for RNA binding was inserted into the sequence of the protein thioredoxin and expressed in reporter-containing bacteria, which were then screened by fluorescence-activated cell sorting (FACS). We identified peptide sequences that reduce frameshift efficiency by over 50% without altering normal translation. The identified sequences are also active against different frameshift stimulatory signals, suggesting that they bind a target important for frameshifting in general, probably the ribosome. Successful transfer of active sequences to a different scaffold in a eukaryotic test system demonstrates that the anti-frameshift activity of the peptides is neither due to scaffold-dependent conformation nor effects of the scaffold protein itself on frameshifting. The method we describe identifies peptides that will provide useful tools to further study the mechanism of frameshift and may permit the development of lead compounds of therapeutic interest.


Subject(s)
Frameshifting, Ribosomal/drug effects , HIV-1/drug effects , HIV-1/metabolism , Peptides/pharmacology , Amino Acid Sequence , Base Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HIV-1/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Nucleic Acid Conformation , Peptide Library , Peptides/genetics , Plasmids/genetics , Protein Biosynthesis/drug effects , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Red Fluorescent Protein
5.
Nucleic Acids Res ; 36(1): 30-40, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17984074

ABSTRACT

HIV-1 uses a programmed -1 ribosomal frameshift to synthesize the precursor of its enzymes, Gag-Pol. The frameshift efficiency that is critical for the virus replication, is controlled by an interaction between the ribosome and a specific structure on the viral mRNA, the frameshift stimulatory signal. The rate of cap-dependent translation initiation is known to be altered by the TAR RNA structure, present at the 5' and 3' end of all HIV-1 mRNAs. Depending upon its concentration, TAR activates or inhibits the double-stranded RNA-dependent protein kinase (PKR). We investigated here whether changes in translation initiation caused by TAR affect HIV-1 frameshift efficiency. CD4+ T cells and 293T cells were transfected with a dual-luciferase construct where the firefly luciferase expression depends upon the HIV-1 frameshift. Translation initiation was altered by adding TAR in cis or trans of the reporter mRNA. We show that HIV-1 frameshift efficiency correlates negatively with changes in the rate of translation initiation caused by TAR and mediated by PKR. A model is presented where changes in the rate of initiation affect the probability of frameshifting by altering the distance between elongating ribosomes on the mRNA, which influences the frequency of encounter between these ribosomes and the frameshift stimulatory signal.


Subject(s)
Frameshifting, Ribosomal , Gene Expression Regulation, Viral , HIV Long Terminal Repeat , HIV-1/genetics , Peptide Chain Initiation, Translational , Cell Line , Frameshifting, Ribosomal/drug effects , HIV-1/drug effects , Humans , Jurkat Cells , Models, Genetic , Peptide Chain Initiation, Translational/genetics , RNA Caps/metabolism , RNA, Messenger/chemistry , RNA, Viral/chemistry , eIF-2 Kinase/antagonists & inhibitors
6.
Nucleic Acids Res ; 35(16): 5581-92, 2007.
Article in English | MEDLINE | ID: mdl-17704133

ABSTRACT

The -1 programmed ribosomal frameshifts (PRF), which are used by many viruses, occur at a heptanucleotide slippery sequence and are currently thought to involve the tRNAs interacting with the ribosomal P- and A-site codons. We investigated here whether the tRNA occupying the ribosomal E site that precedes a slippery site influences -1 PRF. Using the human immunodeficiency virus type 1 (HIV-1) frameshift region, we found that mutating the E-site codon altered the -1 PRF efficiency. When the HIV-1 slippery sequence was replaced with other viral slippery sequences, mutating the E-site codon also altered the -1 PRF efficiency. Because HIV-1 -1 PRF can be recapitulated in bacteria, we used a bacterial ribosome system to select, by random mutagenesis, 16S ribosomal RNA (rRNA) mutations that modify the expression of a reporter requiring HIV-1 -1 PRF. Three mutants were isolated, which are located in helices 21 and 22 of 16S rRNA, a region involved in translocation and E-site tRNA binding. We propose a novel model where -1 PRF is triggered by an incomplete translocation and depends not only on the tRNAs interacting with the P- and A-site codons, but also on the tRNA occupying the E site.


Subject(s)
Frameshifting, Ribosomal , HIV-1/genetics , Models, Genetic , RNA, Transfer/metabolism , RNA, Viral/chemistry , Ribosomes/chemistry , Cell Line , Codon/chemistry , Genes, Reporter , Humans , Mutation , Nucleotides/chemistry , RNA, Messenger/chemistry , RNA, Ribosomal, 16S/chemistry , Ribosomes/metabolism
7.
PLoS Med ; 4(1): e36, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17227139

ABSTRACT

BACKGROUND: HIV protease inhibitor (PI) therapy results in the rapid selection of drug resistant viral variants harbouring one or two substitutions in the viral protease. To combat PI resistance development, two approaches have been developed. The first is to increase the level of PI in the plasma of the patient, and the second is to develop novel PI with high potency against the known PI-resistant HIV protease variants. Both approaches share the requirement for a considerable increase in the number of protease mutations to lead to clinical resistance, thereby increasing the genetic barrier. We investigated whether HIV could yet again find a way to become less susceptible to these novel inhibitors. METHODS AND FINDINGS: We have performed in vitro selection experiments using a novel PI with an increased genetic barrier (RO033-4649) and demonstrated selection of three viruses 4- to 8-fold resistant to all PI compared to wild type. These PI-resistant viruses did not have a single substitution in the viral protease. Full genomic sequencing revealed the presence of NC/p1 cleavage site substitutions in the viral Gag polyprotein (K436E and/or I437T/V) in all three resistant viruses. These changes, when introduced in a reference strain, conferred PI resistance. The mechanism leading to PI resistance is enhancement of the processing efficiency of the altered substrate by wild-type protease. Analysis of genotypic and phenotypic resistance profiles of 28,000 clinical isolates demonstrated the presence of these NC/p1 cleavage site mutations in some clinical samples (codon 431 substitutions in 13%, codon 436 substitutions in 8%, and codon 437 substitutions in 10%). Moreover, these cleavage site substitutions were highly significantly associated with reduced susceptibility to PI in clinical isolates lacking primary protease mutations. Furthermore, we used data from a clinical trial (NARVAL, ANRS 088) to demonstrate that these NC/p1 cleavage site changes are associated with virological failure during PI therapy. CONCLUSIONS: HIV can use an alternative mechanism to become resistant to PI by changing the substrate instead of the protease. Further studies are required to determine to what extent cleavage site mutations may explain virological failure during PI therapy.


Subject(s)
Drug Resistance, Viral/genetics , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/genetics , Mutation , Amino Acid Sequence , Base Sequence , Blotting, Western , Cell Line , Codon/genetics , Frameshift Mutation , Gene Products, gag/genetics , Gene Products, gag/metabolism , Genome, Viral , HIV Protease/genetics , HIV Protease/metabolism , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Humans , Immunoblotting , Molecular Sequence Data , RNA, Viral/genetics , Ritonavir/pharmacology , Substrate Specificity , Transfection
8.
Med Sci (Paris) ; 22(11): 969-72, 2006 Nov.
Article in French | MEDLINE | ID: mdl-17101099

ABSTRACT

Ribosomal frameshift is used by HIV-1 to synthesize the precursor of its enzymes. The frameshift stimulator is a peculiar structure in the viral messenger RNA coding for this precursor, which increases the probability that this frameshift occurs. It was proposed to be either a triplex structure or an irregular stem-loop. Recently, two NMR groups independently showed that the frameshift stimulatory signal of HIV-1 is an extended stem-loop, with an internal three-purine bulge separating two helical regions. However, it remains unclear how such a structure promotes frameshifting. It is proposed that frameshifting results from a specific interaction between the stimulatory signal and either a hypothetical protein factor or the ribosome. The characterization of the structure of the frameshift stimulatory signal paves the way to the rational design of novel antiviral drugs, which, by binding to this signal, could interfere with frameshifting and viral replication.


Subject(s)
Frameshift Mutation , HIV Infections/therapy , HIV-1/genetics , RNA, Ribosomal/genetics , RNA, Viral/genetics , Genetic Therapy , HIV-1/pathogenicity , Humans
9.
Virology ; 345(1): 127-36, 2006 Feb 05.
Article in English | MEDLINE | ID: mdl-16256163

ABSTRACT

The Gag-Pol polyprotein of the human immunodeficiency virus type 1 (HIV-1) is the precursor of the virus enzymatic activities and is produced via a programmed -1 translational frameshift. In this study, we altered the frameshift efficiency by introducing mutations within the slippery sequence and the frameshift stimulatory signal, the two elements that control the frameshift. These mutations decreased the frameshift efficiency to different degrees, ranging from approximately 0.3% to 70% of the wild-type efficiency. These values were mirrored by a reduced incorporation of Gag-Pol into virus-like particles, as assessed by a decrease in the reverse transcriptase activity associated to these particles. Analysis of Gag processing in infectious mutant virions revealed processing defects to various extents, with no clear correlation with frameshift decrease. Nevertheless, the observed frameshift reductions translated into equivalently reduced viral infectivity and replication kinetics. Our results show that even moderate variations in frameshift efficiency, as obtained with mutations in the frameshift stimulatory signal, reduce viral replication. Therapeutic targeting of this structure may therefore result in the attenuation of virus replication and in clinical benefit.


Subject(s)
Frameshifting, Ribosomal , HIV-1/physiology , RNA, Viral/genetics , Virus Replication/physiology , Amino Acid Sequence , Blotting, Western , Fusion Proteins, gag-pol/metabolism , Genes, Reporter , HIV Core Protein p24/analysis , HIV-1/genetics , Luciferases/analysis , Luciferases/genetics , Molecular Sequence Data , Point Mutation , Protein Processing, Post-Translational , RNA, Viral/physiology , RNA-Directed DNA Polymerase/analysis , Viral Proteins/analysis , Virosomes/metabolism , Virus Replication/genetics
10.
Virology ; 334(2): 342-52, 2005 Apr 10.
Article in English | MEDLINE | ID: mdl-15780884

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) requires a programmed -1 translational frameshift event to synthesize the precursor of its enzymes, Gag-Pol, when ribosomes from the infected cells translate the full-length viral messenger RNA. Translation of the same RNA according to conventional translational rules produces Gag, the precursor of the structural proteins of the virus. The efficiency of the frameshift controls the ratio of Gag-Pol to Gag, which is critical for viral infectivity. The Moloney murine leukemia virus (MoMuLV) uses a different strategy, the programmed readthrough of a stop codon, to synthesize Gag-Pol. In this study, we investigated whether different forms of the HIV-1 frameshift region can functionally replace the readthrough signal in MoMuLV. Chimeric proviral DNAs were obtained by inserting into the MoMuLV genome the HIV-1 frameshift region encompassing the slippery sequence where the frameshift occurs, followed by the frameshift stimulatory signal. The inserted signal was either a simple stem-loop, previously considered as the stimulatory signal, or a longer bulged helix, now shown to be the complete stimulatory signal, or a mutated version of the complete signal with a three-nucleotide deletion. Although the three chimeric viruses can propagate essentially as the wild-type virus in NIH 3T3 cells, single-round infectivity assays revealed that the infectivity of the chimeric virions is about three to fivefold lower than that of the wild-type virions, depending upon the nature of the frameshift signal. It was also observed that the Gag-Pol to Gag ratio was decreased about two to threefold in chimeric virions. Comparison of the readthrough efficiency of MoMuLV to the HIV-1 frameshift efficiency, by monitoring the expression of a luciferase reporter in cultured cells, revealed that the frameshift efficiencies were only 30-60% of the readthrough efficiency. Altogether, these observations indicate that replacement of the readthrough region of MoMuLV with the frameshift region of HIV-1 results in virions that are replication competent, although less infectious than wild-type MoMuLV. This type of chimera could provide an interesting tool for in vivo studies of novel drugs targeted against the HIV-1 frameshift event.


Subject(s)
Frameshifting, Ribosomal , Fusion Proteins, gag-pol/metabolism , HIV-1/genetics , Moloney murine leukemia virus/metabolism , Recombinant Fusion Proteins/metabolism , Virion/metabolism , Animals , Base Sequence , Cell Line , Gene Expression Regulation, Viral , Genes, gag , Genes, pol , HIV-1/chemistry , HIV-1/metabolism , Humans , Mice , Molecular Sequence Data , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/pathogenicity , NIH 3T3 Cells , Virion/pathogenicity , Virus Replication
11.
RNA ; 9(10): 1246-53, 2003 Oct.
Article in English | MEDLINE | ID: mdl-13130138

ABSTRACT

The synthesis of the Gag-Pol polyprotein, the precursor of the enzymes of the human immunodeficiency virus type 1 (HIV-1), requires a programmed -1 ribosomal frameshift. This frameshift has been investigated so far only for subtype B of HIV-1 group M. In this subtype, the frameshift stimulatory signal was found to be a two-stem helix, in which a three-purine bulge interrupts the two stems. In this study, using a luciferase reporter system, we compare, for the first time, the frameshift efficiency of all the subtypes of group M. Mutants of subtype B, including a natural variant were also investigated. Our results with mutants of subtype B confirm that the bulge and the lower stem of the frameshift stimulatory signal contribute to the frameshift in addition to the upper stem-loop considered previously as the sole participant. Our results also show that the frameshift stimulatory signal of all of the other subtypes of group M can be folded into the same structure as in subtype B, despite sequence variations. Moreover, the frameshift efficiency of these subtypes, when assessed in cultured cells, falls within a narrow window (the maximal deviation from the mean value calculated from the experimental values of all the subtypes being approximately 35%), although the predicted thermodynamic stability of the frameshift stimulatory signal differs between the subtypes (from -17.2 kcal/mole to -26.2 kcal/mole). The fact that the frameshift efficiencies fall within a narrow range for all of the subtypes of HIV-1 group M stresses the potential of the frameshift event as an antiviral target.


Subject(s)
Frameshifting, Ribosomal , Fusion Proteins, gag-pol/genetics , Gene Expression Regulation, Viral , HIV-1/genetics , RNA, Viral/genetics , Virus Replication , Base Sequence , Conserved Sequence , Genes, Reporter , HIV-1/isolation & purification , Humans , Luciferases/analysis , Luciferases/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Viral/chemistry , Sequence Homology, Nucleic Acid , Transcription, Genetic
12.
J Mol Biol ; 331(3): 571-83, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12899829

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) requires a programmed -1 ribosomal frameshift to produce Gag-Pol, the precursor of its enzymatic activities. This frameshift occurs at a slippery sequence on the viral messenger RNA and is stimulated by a specific structure, downstream of the shift site. While in group M, the most abundant HIV-1 group, the frameshift stimulatory signal is an extended bulged stem-loop, we show here, using a combination of mutagenesis and probing studies, that it is a pseudoknot in group O. The mutagenesis and probing studies coupled to an in silico analysis show that group O pseudoknot is a hairpin-type pseudoknot with two coaxially stacked stems of eight base-pairs (stem 1 and stem 2), connected by single-stranded loops of 2nt (loop 1) and 20nt (loop 2). Mutations impairing formation of stem 1 or stem 2 of the pseudoknot reduce frameshift efficiency, whereas compensatory changes that allow re-formation of these stems restore the frameshift efficiency to near wild-type level. The difference between the frameshift stimulatory signal of group O and group M supports the hypothesis that these groups originate from a different monkey to human transmission.


Subject(s)
Frameshifting, Ribosomal/genetics , HIV-1/genetics , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/genetics , Regulatory Sequences, Ribonucleic Acid/genetics , Base Sequence , Cell Line , Computer Simulation , Humans , Models, Molecular , Molecular Sequence Data , Sequence Analysis, DNA
13.
Nucleic Acids Res ; 30(23): 5094-102, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12466532

ABSTRACT

Synthesis of the Gag-Pol protein of the human immunodeficiency virus type 1 (HIV-1) requires a programmed -1 ribosomal frameshifting when ribosomes translate the unspliced viral messenger RNA. This frameshift occurs at a slippery sequence followed by an RNA structure motif that stimulates frameshifting. This motif is commonly assumed to be a simple stem-loop for HIV-1. In this study, we show that the frameshift stimulatory signal is more complex than believed and consists of a two-stem helix. The upper stem-loop corresponds to the classic stem-loop, and the lower stem is formed by pairing the spacer region following the slippery sequence and preceding this classic stem-loop with a segment downstream of this stem-loop. A three-purine bulge interrupts the two stems. This structure was suggested by enzymatic probing with nuclease V1 of an RNA fragment corresponding to the gag/pol frameshift region of HIV-1. The involvement of the novel lower stem in frameshifting was supported by site-directed mutagenesis. A fragment encompassing the gag/pol frameshift region of HIV-1 was inserted in the beginning of the coding sequence of a reporter gene coding for the firefly luciferase, such that expression of luciferase requires a -1 frameshift. When the reporter was expressed in COS cells, mutations that disrupt the capacity to form the lower stem reduced frameshifting, whereas compensatory changes that allow re-formation of this stem restored the frameshift efficiency near wild-type level. The two-stem structure that we propose for the frameshift stimulatory signal of HIV-1 differs from the RNA triple helix structure recently proposed.


Subject(s)
Frameshifting, Ribosomal , Fusion Proteins, gag-pol/genetics , Gene Expression Regulation, Viral , HIV-1/genetics , Animals , Base Sequence , COS Cells , Conserved Sequence , Genes, Reporter , HIV-1/isolation & purification , Humans , Luciferases/analysis , Luciferases/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...