Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Oncogene ; 40(22): 3815-3825, 2021 06.
Article in English | MEDLINE | ID: mdl-33958722

ABSTRACT

The integration of viral DNA into the host genome is mediated by viral integrase, resulting in the accumulation of double-strand breaks. Integrase-derived peptides (INS and INR) increase the number of integration events, leading to escalated genomic instability that induces apoptosis. CD24 is a surface protein expressed mostly in cancer cells and is very rarely found in normal cells. Here, we propose a novel targeted cancer therapeutic platform based on the lentiviral integrase, stimulated by integrase-derived peptides, that are specifically delivered to cancerous cells via CD24 antigen-antibody targeting. INS and INR were synthesized and humanized and anti-CD24 antibodies were fused to the lentivirus envelope. The activity, permeability, stability, solubility, and toxicity of these components were analyzed. Cell death was measured by fluorescent microscopy and enzymatic assays and potency were tested in vitro and in vivo. Lentivirus particles, containing non-functional DNA led to massive cell death (40-70%). Raltegravir, an antiretroviral drug, inhibited the induction of apoptosis. In vivo, single and repeated administrations of INS/INR were well tolerated without any adverse effects. Tumor development in nude mice was significantly inhibited (by 50%) as compared to the vehicle arm. In summary, a novel and generic therapeutic platform for selective cancer cell eradication with excellent efficacy and safety are presented.


Subject(s)
CD24 Antigen/biosynthesis , Integrases/pharmacology , Lentivirus/enzymology , Neoplasms/therapy , Oncolytic Virotherapy/methods , Peptide Fragments/pharmacology , Animals , Antibodies, Monoclonal/immunology , Apoptosis/drug effects , CD24 Antigen/immunology , Cell Line, Tumor , Humans , Integrases/chemistry , Lentivirus/genetics , Lentivirus/immunology , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/virology , Peptide Fragments/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays
2.
Virology ; 477: 56-60, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25644513

ABSTRACT

Tomato yellow leaf curl virus (TYLCV), a major tomato pathogen causing extensive crop losses, is a whitefly-transmitted geminivirus. V2 mutants of TYLCV-Is and related viruses tend to induce symptomless infection with attenuated viral DNA levels, while accumulating close to wild-type DNA levels in protoplasts, suggesting V2 as a movement protein. The discovery of plant-silencing mechanisms and viral silencing suppressors, V2 included, led us to reconsider V2׳s involvement in viral movement. We studied two mutant versions of the virus, one impaired in V2 silencing-suppression activity, and another carrying a non-translatable V2. While both mutant viruses spread in the infected plant to newly emerged leaves at the same rate as the wild-type virus, their DNA-accumulation levels were tenfold lower than in the wild-type virus. Thus, we suggest that the setback in virus proliferation, previously ascribed to a movement impediment, is due to lack of silencing-suppression activity.


Subject(s)
Begomovirus/physiology , Plant Viral Movement Proteins/metabolism , Virus Replication , Begomovirus/genetics , Gene Knockout Techniques , Plant Viral Movement Proteins/genetics , Nicotiana/virology
3.
Protein Eng Des Sel ; 27(11): 439-46, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25212215

ABSTRACT

Agrobacterium is a pathogen that genetically transforms plants. The bacterial VirE2 protein envelopes the T-DNA of Agrobacterium and protects it from degradation. Within the transfected cells, VirE2 interacts with the plant VIP1 leading to nuclear transport of the T-DNA complex. Active VirE2 is an oligomer with a tendency to aggregate, hampering its studies at the molecular level. In addition, no structural or quantitative information is available regarding VIP1 or its interactions. The lack of information is mainly because both VIP1 and VirE2 are difficult to express and purify. Here, we present the development of efficient protocols that resulted in pure and stable His-tagged VIP1 and VirE2. Circular dichroism spectroscopy and computational predictions indicated that VIP1 is mostly intrinsically disordered. This may explain the variety of protein-protein interactions it participates in. Size exclusion chromatography revealed that VirE2 exists in a two-state equilibrium between a monomer and an oligomeric form. Using the purified proteins, we performed peptide array screening and revealed the binding sites on both proteins. VirE2 binds the disordered regions of VIP1, while the site in VirE2 that binds VIP1 is different from the VirE2 DNA-binding site. Peptides derived from these sites may be used as lead compounds that block Agrobacterium infection of plants.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA, Plant/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Ion Channels/chemistry , Ion Channels/metabolism , Arabidopsis Proteins/genetics , Bacterial Proteins/genetics , Binding Sites , DNA, Plant/chemistry , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Ion Channels/genetics , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
Angew Chem Int Ed Engl ; 53(36): 9450-5, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24827640

ABSTRACT

We present a new approach for peptide cyclization during solid phase synthesis under highly acidic conditions. Our approach involves simultaneous in situ deprotection, cyclization and trifluoroacetic acid (TFA) cleavage of the peptide, which is achieved by forming an amide bond between a lysine side chain and a succinic acid linker at the peptide N-terminus. The reaction proceeds via a highly active succinimide intermediate, which was isolated and characterized. The structure of a model cyclic peptide was solved by NMR spectroscopy. Theoretical calculations support the proposed mechanism of cyclization. Our new methodology is applicable for the formation of macrocycles in solid-phase synthesis of peptides and organic molecules.


Subject(s)
Peptides, Cyclic/chemical synthesis , Trifluoroacetic Acid/chemistry , Amino Acid Sequence , Catalysis , Cyclization , Lysine/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Peptides, Cyclic/chemistry , Peptidomimetics/chemical synthesis , Solid-Phase Synthesis Techniques
5.
Chem Commun (Camb) ; 49(30): 3113-5, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23463374

ABSTRACT

Integration of the HIV cDNA into the host chromosome is a key event in the viral replication cycle. It is mediated by the viral integrase (IN) enzyme, which is an attractive anti-HIV drug target. Here we present the first AFM imaging of IN-mediated DNA integration products in a two-LTR system.


Subject(s)
HIV Integrase/metabolism , HIV Long Terminal Repeat , DNA, Complementary/genetics , DNA, Complementary/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Enzyme Activation , HIV Integrase/chemistry , HIV Integrase/genetics , HIV Long Terminal Repeat/genetics , Microscopy, Atomic Force
6.
FEBS J ; 279(16): 2795-809, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22742518

ABSTRACT

HIV-1 integrase (IN) is one of the key enzymes in the viral replication cycle. It mediates the integration of viral cDNA into the host cell genome. IN activity requires interactions with several viral and cellular proteins, as well as IN oligomerization. Inhibition of IN is an important target for the development of anti-HIV therapies, but there is currently only one anti-HIV drug used in the clinic that targets IN. Several other small-molecule anti-IN drug leads are either undergoing clinical trials or in earlier stages of development. These molecules specifically inhibit one of the IN-mediated reactions necessary for successful integration. However, small-molecule inhibitors of protein-protein interactions are difficult to develop. In this review, we focus on peptides that inhibit IN. Peptides have advantages over small-molecule inhibitors of protein-protein interactions: they can mimic the structures of the binding domains within proteins, and are large enough to competitively inhibit protein-protein interactions. The development of peptides that bind IN and inhibit its protein-protein interactions will increase our understanding of the IN mode of action, and lead to the development of new drug leads, such as small molecules derived from these peptides, for better anti-HIV therapy.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , Peptides/pharmacology , Amino Acid Sequence , Combinatorial Chemistry Techniques , Drug Evaluation, Preclinical , HIV Reverse Transcriptase/metabolism , Humans , Models, Molecular , Peptide Fragments/pharmacology , Peptide Library , Protein Multimerization/drug effects
7.
Bioorg Med Chem ; 20(10): 3317-22, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22507205

ABSTRACT

Peptide cyclization is an important tool for overcoming the limitations of linear peptides as drugs. Backbone cyclization (BC) has advantages over side chain (SC) cyclization because it combines N-alkylation for extra peptide stability. However, the appropriate building blocks for BC are not yet commercially available. This problem can be overcome by preparing SC cyclic peptide analogs of the most active BC peptide using commercially available building blocks. We have recently developed BC peptides that inhibit the HIV-1 integrase enzyme (IN) activity and HIV-1 replication in infected cells. Here we used this system as a model for systematically comparing the BC and SC cyclization modes using biophysical, biochemical and structural methods. The most potent SC cyclic peptide was active almost as the BC peptide and inhibited IN activity in vitro and blocked IN activity in cells even after 6 days. We conclude that both cyclization types have their respective advantages: The BC peptide is more active and stable, probably due to the N-alkylation, while SC cyclic peptides are easier to synthesize. Due to the high costs and efforts involved in preparing BC peptides, SC may be a more approachable method in many cases. We suggest that both methods are interchangeable.


Subject(s)
HIV Integrase Inhibitors , HIV-1/drug effects , Peptides, Cyclic , Amino Acid Sequence , Enzyme Activation/drug effects , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Peptide Library , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology
8.
Biochem Biophys Res Commun ; 416(3-4): 252-7, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22093836

ABSTRACT

The HIV-1 Rev and integrase (IN) proteins control important functions in the viral life cycle. We have recently discovered that the interaction between these proteins results in inhibition of IN enzymatic activity. Peptides derived from the Rev and IN binding interfaces have a profound effect on IN catalytic activity: Peptides derived from Rev inhibit IN, while peptides derived from IN stimulate IN activity by inhibiting the Rev-IN interaction. This inhibition leads to multi integration, genomic instability and specific death of virus-infected cells. Here we used protein docking combined with refinement and energy function ranking to suggest a structural model for the Rev-IN complex. Our results indicate that a Rev monomer binds IN at two sites that match our experimental binding data: (1) IN residues 66-80 and 118-128; (2) IN residues 174-188. According to our model, IN binds Rev and its cellular cofactor, lens epithelium derived growth factor (LEDGF), through overlapping interfaces. This supports previous observations that IN is regulated by a tight interplay between Rev and LEDGF. Rev may bind either the IN dimer or tetramer. Accordingly, Rev is suggested to inhibit IN by two possible mechanisms: (i) shifting the oligomerization equilibrium of IN from an active dimer to an inactive tetramer; (ii) displacing LEDGF from IN, resulting in inhibition of IN binding to the viral DNA. Our model is expected to contribute to the development of lead compounds that inhibit the Rev-IN interaction and thus lead to multi-integration of viral cDNA and consequently to apoptosis of HIV-1 infected cells.


Subject(s)
HIV Integrase/chemistry , Models, Chemical , rev Gene Products, Human Immunodeficiency Virus/chemistry , Anti-HIV Agents/chemistry , Drug Discovery , Humans , Protein Conformation , rev Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors
9.
Biochim Biophys Acta ; 1813(9): 1646-53, 2011 Sep.
Article in English | MEDLINE | ID: mdl-20719241

ABSTRACT

Nuclear import is a critical step in the life cycle of HIV-1. During the early (preintegration) stages of infection, HIV-1 has to transport its preintegration complex into the nucleus for integration into the host cell chromatin, while at the later (postintegration) stages viral regulatory proteins Tat and Rev need to get into the nucleus to stimulate transcription and regulate splicing and nuclear export of subgenomic and genomic RNAs. Given such important role of nuclear import in HIV-1 life cycle, this step presents an attractive target for antiviral therapeutic intervention. In this review, we describe the current state of our understanding of the interactions regulating nuclear import of the HIV-1 preintegration complex and describe current approaches to inhibit it. This article is part of a Special Issue entitled: Regulation of Signaling and Cellular Fate through Modulation of Nuclear Protein Import.


Subject(s)
Active Transport, Cell Nucleus/drug effects , HIV-1/drug effects , HIV-1/physiology , Human Immunodeficiency Virus Proteins/physiology , Active Transport, Cell Nucleus/physiology , Anti-HIV Agents/pharmacology , HIV-1/growth & development , Humans , Models, Biological , Nuclear Localization Signals/physiology , Peptides/pharmacology , Virus Replication/drug effects , Virus Replication/physiology
10.
FEBS J ; 278(2): 316-30, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21156026

ABSTRACT

Several peptides that specifically bind the HIV-1 integrase (IN) and either inhibit or stimulate its enzymatic activity were developed in our laboratories. Kinetic studies using 3'-end processing and strand-transfer assays were performed to study the mode of action of these peptides. The effects of the various peptides on the interaction between IN and its substrate DNA were also studied by fluorescence anisotropy. On the basis of our results, we divided these IN-interacting peptides into three groups: (a) IN-inhibitory peptides, whose binding to IN decrease its affinity for the substrate DNA - these peptides increased the K(m) of the IN-DNA interaction, and were thus inhibitory; (b) peptides that slightly increased the K(m) of the IN-DNA interaction, but in addition modified the V(max) and K(cat) values of the IN, and thus stimulated or inhibited IN activity, respectively; and (c) peptides that bound IN but had no effect on its enzymatic activity. We elucidated the approximate binding sites of the peptides in the structure of IN, providing structural insights into their mechanism of action. The IN-stimulating peptide bound IN in several specific sites that did not bind any of the inhibitory peptides. This may account for its unique activity.


Subject(s)
Biocatalysis/drug effects , HIV Integrase/metabolism , Peptides/metabolism , Amino Acid Sequence , Binding Sites/physiology , Binding, Competitive/physiology , DNA/chemistry , DNA/metabolism , HIV Integrase/chemistry , HIV Integrase/genetics , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/metabolism , HIV Integrase Inhibitors/pharmacology , Kinetics , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/drug effects , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Protein Conformation/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Bioorg Med Chem ; 18(23): 8388-95, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20974536

ABSTRACT

Restricting linear peptides to their bioactive conformation is an attractive way of improving their stability and activity. We used a cyclic peptide library with conformational diversity for selecting an active and stable peptide that mimics the structure and activity of the HIV-1 integrase (IN) binding loop from its cellular cofactor LEDGF/p75 (residues 361-370). All peptides in the library had the same primary sequence, and differed only in their conformation. Library screening revealed that the ring size and linker structure had a huge effect on the conformation, binding and activity of the peptides. One of the cyclic peptides, c(MZ 4-1), was a potent and stable inhibitor of IN activity in vitro and in cells even after 8 days. The NMR structure of c(MZ 4-1) showed that it obtains a bioactive conformation that is similar to the parent site in LEDGF/p75.


Subject(s)
HIV Integrase Inhibitors/chemistry , HIV Integrase/chemistry , Intercellular Signaling Peptides and Proteins/chemistry , Peptides, Cyclic/chemistry , Amino Acid Sequence , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , HeLa Cells , Humans , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Protein Binding , Protein Structure, Tertiary , Virus Replication/drug effects
12.
Virol J ; 7: 177, 2010 Aug 02.
Article in English | MEDLINE | ID: mdl-20678206

ABSTRACT

BACKGROUND: The presence of the cellular Lens Epithelium Derived Growth Factor p75 (LEDGF/p75) protein is essential for integration of the Human immunodeficiency virus type 1 (HIV-1) cDNA and for efficient virus production. In the absence of LEDGF/p75 very little integration and virus production can be detected, as was demonstrated using LEDGF/p75-knockdown cells. RESULTS: Here we show that the failure to infect LEDGF/p75-knockdown cells has another reason aside from the lack of LEDGF/p75. It is also due to inhibition of the viral integrase (IN) enzymatic activity by an early expressed viral Rev protein. The formation of an inhibitory Rev-IN complex in virus-infected cells can be disrupted by the addition of three IN-derived, cell-permeable peptides, designated INr (IN derived-Rev interacting peptides) and INS (IN derived-integrase stimulatory peptide). The results of the present work confirm previous results showing that HIV-1 fails to infect LEDGF/p75-knockdown cells. However, in the presence of INrs and INS peptides, relatively high levels of viral cDNA integration as well as productive virus infection were obtained following infection by a wild type (WT) HIV-1 of LEDGF/p75-knockdown cells. CONCLUSIONS: It appears that the lack of integration observed in HIV-1 infected LEDGF/p75-knockdown cells is due mainly to the inhibitory effect of Rev following the formation of a Rev-IN complex. Disruption of this inhibitory complex leads to productive infection in those cells.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , DNA, Complementary/genetics , DNA, Viral/genetics , HIV Integrase/metabolism , HIV-1/pathogenicity , Transcription Factors/antagonists & inhibitors , Virus Integration , rev Gene Products, Human Immunodeficiency Virus/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Gene Knockdown Techniques , HIV Integrase/genetics , HIV-1/genetics , Humans , Models, Biological , Protein Binding , Transcription Factors/genetics
13.
AIDS Res Ther ; 7: 31, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20723214

ABSTRACT

A correlation between increase in the integration of Human Immunodeficiency virus-1 (HIV-1) cDNA and cell death was previously established. Here we show that combination of peptides that stimulate integration together with the protease inhibitor Ro 31-8959 caused apoptotic cell death of HIV infected cells with total extermination of the virus. This combination did not have any effect on non-infected cells. Thus it appears that cell death is promoted only in the infected cells. It is our view that the results described in this work suggest a novel approach to specifically promote death of HIV-1 infected cells and thus may eventually be developed into a new and general anti-viral therapy.

14.
Biopolymers ; 93(8): 740-51, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20517955

ABSTRACT

Here we describe the features of a peptide that was selected from the human immunodeficiency virus Type 1 (HIV-1) Integrase (IN) peptide library which interacts with both, the viral Rev and IN proteins. Because of its ability to stimulate the IN enzymatic activity this peptide was designated INS (IN stimulatory). Modification of its amino acid sequence revealed that replacement of its C-terminal lysine by glutamic acid (INS K188E) converts it from a stimulatory peptide to an inhibitory one. Both peptides promoted the dissociation of a previously described complex formed between Rev and IN whose formation results in IN inactivation. INS and INS K188E penetrated HIV-1-infected cells and caused stimulation and inhibition of viral genome integration, respectively. Using cultured cells infected with a DeltaRev HIV revealed that INS can directly activate the viral IN. These results suggest that the stimulatory effect of INS in wild-type virus-infected cells is due to a dual effect: it dissociates the inactive Rev-IN complex and directly activates the free IN.


Subject(s)
HIV Integrase/chemistry , HIV Integrase/metabolism , HIV-1/enzymology , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Cell Line , Cell Membrane Permeability , DNA, Complementary/genetics , DNA, Complementary/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , HIV Integrase/genetics , HIV-1/genetics , HeLa Cells , Humans , In Vitro Techniques , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Library , Protein Binding , Virus Integration , rev Gene Products, Human Immunodeficiency Virus/metabolism
15.
Antiviral Res ; 87(1): 30-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20398703

ABSTRACT

The transactivating factor (Tat) of HIV-1 is involved in AIDS progression and associated pathologies. Tat possesses a basic amino acid sequence implicated in heparan sulfate proteoglycan (HSPG)-mediated internalization, nuclear localization and transactivation by Tat and in the interaction of Tat with integrins and with the vascular endothelial growth factor receptor 2 (KDR) (kinase insert domain receptor). A BSA conjugate bearing an average of four copies of a peptide representing the basic domain/nuclear localization signal of Tat (BSA-Tat-NLS) inhibits transactivation by Tat exogenously added to cells but not by Tat endogenously produced after cell transfection with a tat cDNA, indicating that BSA-Tat-NLS does not interfere with Tat at an intracellular level. Surface plasmon resonance (SPR) experiments revealed that BSA-Tat-NLS binds to the HSPG analogue heparin. Accordingly, BSA-Tat-NLS binds to HSPGs of HL3T1 cell surface and inhibits HSPG-dependent Tat internalization. BSA-Tat-NLS retains its inhibitory potential when pre-incubated with HL3T1 cells before Tat administration, possibly by masking cell-surface HSPGs thus preventing Tat binding and internalization. SPR experiments revealed that BSA-Tat-NLS binds also to integrin alpha(v)beta(3) and KDR. Accordingly, it inhibits pro-angiogenic endothelial cell adhesion to Tat and motogenesis. In conclusion, BSA-Tat-NLS binds/masks three different cell-surface receptors of Tat inhibiting different biological activities. These data point to BSA-Tat-NLS as a prototype for the development of Tat-antagonists endowed with a multitargeted mechanism of action.


Subject(s)
HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Serum Albumin, Bovine/pharmacology , Virus Internalization/drug effects , tat Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , tat Gene Products, Human Immunodeficiency Virus/pharmacology , HIV Fusion Inhibitors/chemistry , HIV-1/physiology , HeLa Cells , Heparin/metabolism , Humans , Protein Binding , Serum Albumin, Bovine/chemistry , Surface Plasmon Resonance , tat Gene Products, Human Immunodeficiency Virus/chemistry
16.
Cell Host Microbe ; 7(3): 197-209, 2010 Mar 18.
Article in English | MEDLINE | ID: mdl-20227663

ABSTRACT

Agrobacterium exports DNA into plant cells, eliciting neoplastic growths on many plant species. During this process, a Skp1-Cdc53-cullin-F-box (SCF) complex that contains the bacterial virulence F-box protein VirF facilitates genetic transformation by targeting for proteolysis proteins, the Agrobacterium protein VirE2 and the host protein VIP1, that coat the transferred DNA. However, some plant species do not require VirF for transformation. Here, we show that Agrobacterium induces expression of a plant F-box protein, which we designated VBF for VIP1-binding F-box protein, that can functionally replace VirF, regulating levels of the VirE2 and VIP1 proteins via a VBF-containing SCF complex. When expressed in Agrobacterium and exported into the plant cell, VBF functionally complements tumor formation by a strain lacking VirF. VBF expression is known to be induced by diverse pathogens, suggesting that Agrobacterium has co-opted a plant defense response and that bacterial VirF and plant VBF both contribute to targeted proteolysis that promotes plant genetic transformation.


Subject(s)
F-Box Proteins/biosynthesis , Host-Pathogen Interactions , Plant Proteins/biosynthesis , Plant Tumors/microbiology , Rhizobium/pathogenicity , Arabidopsis , Bacterial Proteins/metabolism , Solanum lycopersicum , Molecular Sequence Data , Protein Binding , Protein Interaction Mapping , Sequence Analysis, DNA , Nicotiana , Transformation, Genetic , Two-Hybrid System Techniques , Virulence Factors/metabolism
17.
J Gen Virol ; 91(Pt 6): 1503-13, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20147519

ABSTRACT

At the cellular level, cells infected with human immunodeficiency virus type 1 (HIV-1) exhibit immunity to a second infection by the virus that initiated the first infection or by related viruses [superinfection resistance (SIR)]. In the case of HIV infection, SIR was basically attributed to downregulation of the CD4 receptors. We have recently reported on an interaction between HIV-1 Rev and integrase (IN) proteins, which results in inhibition of IN activity in vitro and integration of cDNA in HIV-1-infected cells. A novel function for the viral Rev protein in controlling integration of HIV cDNAs was thus proposed. The results of the present work suggest involvement of the inhibitory Rev in sustaining SIR. A single exposure to wild-type HIV-1 resulted in one to two integrations per cell. The number of integrated proviral cDNA copies remained at this low level even after double infection or superinfection. SIR was dependent on Rev expression by the strain used for the first infection and was eliminated by peptides that disrupt intracellular complex formation between IN and Rev. The same lack of resistance was observed in the absence of Rev, namely following first infection with a DeltaRev HIV strain. The involvement of Rev, expressed from either unintegrated or integrated viral cDNA, in promoting SIR was clearly demonstrated. We conclude that SIR involves Rev-dependent control of HIV cDNA integration.


Subject(s)
Gene Products, rev/physiology , HIV-1/physiology , Virus Integration , Cell Line , HIV Integrase/metabolism , Humans , Protein Binding
18.
Biochem Biophys Res Commun ; 394(2): 260-5, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20171172

ABSTRACT

The HIV-1 integrase protein (IN) mediates integration of the viral cDNA into the host genome and is a target for anti-HIV drugs. We have recently described a peptide derived from residues 361-370 of the IN cellular partner protein LEDGF/p75, which inhibited IN catalytic activity in vitro and HIV-1 replication in cells. Here we performed a comprehensive study of the LEDGF 361-370 mechanism of action in vitro, in cells and in vivo. Alanine scan, fluorescence anisotropy binding studies, homology modeling and NMR studies demonstrated that all residues in LEDGF 361-370 contribute to IN binding and inhibition. Kinetic studies in cells showed that LEDGF 361-370 specifically inhibited integration of viral cDNA. Thus, the full peptide was chosen for in vivo studies, in which it inhibited the production of HIV-1 RNA in mouse model. We conclude that the full LEDGF 361-370 peptide is a potent HIV-1 inhibitor and may be used for further development as an anti-HIV lead compound.


Subject(s)
Adaptor Proteins, Signal Transducing/pharmacology , HIV Integrase Inhibitors/pharmacology , Peptide Fragments/pharmacology , Transcription Factors/pharmacology , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Sequence , Animals , HIV Integrase , HIV Integrase Inhibitors/chemistry , HIV-1/drug effects , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred Strains , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Conformation , Transcription Factors/chemistry , Virus Replication/drug effects
19.
Virus Genes ; 40(3): 341-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20151187

ABSTRACT

Expression of the human immunodeficiency virus type 1 (HIV-1) Rev protein is essential for completion of the viral life cycle. Rev mediates nuclear export of partially spliced and unspliced viral transcripts and therefore bears a nuclear localization signal (NLS) as well as a nuclear export signal (NES), which allow its nucleocytoplasmic shuttling. Attempts to express the wild-type Rev protein in eukaryotic human cultured cells have encountered difficulties and so far have failed. Here we show that accumulation of Rev, which occurs in nondividing Rev-expressing cells or when such cells reach confluency, results in death of these cells. Cell death was also promoted by addition of a cell permeable peptide bearing the Rev-NES sequence, but not by the Rev-NLS peptide. Our results probably indicate that binding of excess amounts of the Rev protein or the NES peptide to the exportin receptor CRM1 results in cells' death.


Subject(s)
Cell Death , Eukaryotic Cells/virology , Gene Expression , HIV-1/pathogenicity , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Virulence Factors/biosynthesis , rev Gene Products, Human Immunodeficiency Virus/biosynthesis , Cell Line , Humans , Protein Binding , Exportin 1 Protein
20.
Nucleus ; 1(5): 422-31, 2010.
Article in English | MEDLINE | ID: mdl-21326825

ABSTRACT

Unlike other retroviruses, human immunodeficiency virus type-1 (HIV-1) can infect terminally differentiated cells, due to the ability of its pre-integration complex (PIC) to translocate via the host nuclear pore complex (NPC). The PIC Nuclear import has been suggested to be mediated by the viral integrase protein (IN), via either the importin α or transportin 3 (TNPO3/transportin-SR2) pathways.We show that in virus-infected cells, IN interacts with both importin α and TNPO3, simultaneously or separately, suggesting a multiple use of nuclear import pathways. Disruption of either the IN-importin α or IN-TNPO3 complexes in virus-infected cells by specific cell-permeable-peptides resulted in inhibition of IN and viral cDNA nuclear import. Here we show that peptides which disrupt either one of these complexes block virus infection, indicating involvement of both pathways in efficient viral replication. Formation of IN-importin α and IN-TNPO3 complexes has also been observed in IN-transfected cultured cells. Using specific peptides, we demonstrate that in transfected cells but not in virus infected cells the importin α pathway overrides that of TNPO3. The IN-importin α and IN-TNPO3 complexes were not observed in virus-infected Rev-expressing cells, indicating the Rev protein's ability to disrupt both complexes.Our work suggests that IN nuclear import requires the involvement of both importin α and TNPO3. The ability to inhibit nuclear import of the IN-DNA complex and consequently, virus infection by peptides that interrupt IN's interaction with either importin α or TNPO3 indicates that for efficient infection, nuclear import of IN should be mediated by both nuclear-import receptors.


Subject(s)
DNA, Viral/antagonists & inhibitors , HIV Integrase/metabolism , HIV-1/physiology , Nuclear Pore/metabolism , alpha Karyopherins/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Cell Line, Tumor , DNA, Complementary/antagonists & inhibitors , DNA, Complementary/biosynthesis , DNA, Viral/biosynthesis , HIV Integrase/genetics , Host-Pathogen Interactions , Humans , Molecular Sequence Data , Nuclear Pore/genetics , Peptides/chemical synthesis , Peptides/pharmacology , Protein Binding , Transfection , Virus Replication/drug effects , alpha Karyopherins/genetics , beta Karyopherins/genetics , rev Gene Products, Human Immunodeficiency Virus/genetics , rev Gene Products, Human Immunodeficiency Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...