Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
PLoS One ; 13(7): e0200080, 2018.
Article in English | MEDLINE | ID: mdl-29995936

ABSTRACT

The human immunodeficiency virus (HIV) depends on cellular proteins, so-called cofactors, to complete its replication cycle. In search for new therapeutic targets we identified the DNA and RNA binding protein Y-box-binding Protein 1 (YB-1) as a cofactor supporting early and late steps of HIV replication. YB-1 depletion resulted in a 10-fold decrease in HIV-1 replication in different cell lines. Dissection of the replication defects revealed that knockdown of YB-1 is associated with a 2- to 5-fold decrease in virion production due to interference with the viral RNA metabolism. Using single-round virus infection experiments we demonstrated that early HIV-1 replication also depends on the cellular YB-1 levels. More precisely, using quantitative PCR and an in vivo nuclear import assay with fluorescently labeled viral particles, we showed that YB-1 knockdown leads to a block between reverse transcription and nuclear import of HIV-1. Interaction studies revealed that YB-1 associates with integrase, although a direct interaction with HIV integrase could not be unambiguously proven. In conclusion, our results indicate that YB-1 affects multiple stages of HIV replication. Future research on the interaction between YB-1 and the virus will reveal whether this protein qualifies as a new antiviral target.


Subject(s)
HIV-1/physiology , Virus Replication , Y-Box-Binding Protein 1/metabolism , Active Transport, Cell Nucleus , HIV Integrase/metabolism , HIV-1/enzymology , HIV-1/genetics , HeLa Cells , Humans , RNA, Viral/metabolism , Reverse Transcription , Time Factors
3.
Cell Chem Biol ; 23(10): 1193-1205, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27693059

ABSTRACT

Allostery is a phenomenon observed in many proteins where binding of a macromolecular partner or a small-molecule ligand at one location leads to specific perturbations at a site not in direct contact with the region where the binding occurs. The list of proteins under allosteric regulation includes AGC protein kinases. AGC kinases have a conserved allosteric site, the phosphoinositide-dependent protein kinase 1 (PDK1)-interacting fragment (PIF) pocket, which regulates protein ATP-binding, activity, and interaction with substrates. In this study, we identify small molecules that bind to the ATP-binding site and affect the PIF pocket of AGC kinase family members, PDK1 and Aurora kinase. We describe the mechanistic details and show that although PDK1 and Aurora kinase inhibitors bind to the conserved ATP-binding site, they differentially modulate physiological interactions at the PIF-pocket site. Our work outlines a strategy for developing bidirectional small-molecule allosteric modulators of protein kinases and other signaling proteins.


Subject(s)
Adenosine Triphosphate/metabolism , Allosteric Regulation/drug effects , Indazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Allosteric Site/drug effects , Aurora Kinases/antagonists & inhibitors , Aurora Kinases/chemistry , Aurora Kinases/metabolism , Binding Sites/drug effects , HEK293 Cells , Humans , Indazoles/chemistry , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/chemistry , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
4.
Chem Biol ; 21(6): 754-65, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24836908

ABSTRACT

Protein kinases play important regulatory roles in cells and organisms. Therefore, they are subject to specific and tight mechanisms of regulation that ultimately converge on the catalytic domain and allow the kinases to be activated or inhibited only upon the appropriate stimuli. AGC protein kinases have a pocket in the catalytic domain, the PDK1-interacting fragment (PIF)-pocket, which is a key mediator of the activation. We show here that helix αC within the PIF-pocket of atypical protein kinase C (aPKC) is the target of the interaction with its inhibitory N-terminal domains. We also provide structural evidence that the small compound PS315 is an allosteric inhibitor that binds to the PIF-pocket of aPKC. PS315 exploits the physiological dynamics of helix αC for its binding and allosteric inhibition. The results will support research on allosteric mechanisms and selective drug development efforts against PKC isoforms.


Subject(s)
Biphenyl Compounds/pharmacology , Cinnamates/pharmacology , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Allosteric Regulation/drug effects , Biphenyl Compounds/chemistry , Cinnamates/chemistry , Humans , Models, Molecular , Molecular Structure , Protein Kinase C/antagonists & inhibitors , Protein Structure, Tertiary/drug effects , Structure-Activity Relationship
5.
Cell Rep ; 5(4): 886-94, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24183673

ABSTRACT

A hallmark of retroviral replication is integration of the viral genome into host cell DNA. This characteristic makes retrovirus-based vectors attractive delivery vehicles for gene therapy. However, adverse events in gene therapeutic trials, caused by activation of proto-oncogenes due to murine leukemia virus (MLV)-derived vector integration, hamper their application. Here, we show that bromodomain and extraterminal (BET) proteins (BRD2, BRD3, and BRD4) and MLV integrase specifically interact and colocalize within the nucleus of the cell. Inhibition of the BET proteins' chromatin interaction via specific bromodomain inhibitors blocks MLV virus replication at the integration step. MLV integration site distribution parallels the chromatin binding profile of BET proteins, and expression of an artificial fusion protein of the BET integrase binding domain with the chromatin interaction domain of the lentiviral targeting factor LEDGF/p75 retargets MLV integration away from transcription start sites and into the body of actively transcribed genes, conforming to the HIV integration pattern. Together, these data validate BET proteins as MLV integration targeting factors.


Subject(s)
Leukemia Virus, Murine/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Transcription Initiation Site , Virus Integration/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Azepines/pharmacology , Cell Line , Chromosomal Proteins, Non-Histone , DNA, Viral/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Structure, Tertiary , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Virus Replication/drug effects , Virus Replication/genetics
6.
J Med Chem ; 55(22): 9817-30, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23106316

ABSTRACT

The protein kinase C-related kinase 2 (PRK2)-interacting fragment (PIF) pocket of phosphoinositide-dependent kinase-1 (PDK1) was proposed as a novel target site for allosteric modulators. In the present work, we describe the design, synthesis, and structure-activity relationship of a series of 2-(3-oxo-1,3-diphenylpropyl)malonic acids as potent allosteric activators binding to the PIF pocket. Some congeners displayed AC(50) values for PDK1 activation in the submicromolar range. The potency of the best compounds to stabilize PDK1 in a thermal stability shift assay was in the same order of magnitude as that of the PIF pocket binding peptide PIFtide, suggesting comparable binding affinities to the PIF pocket. The crystal structure of PDK1 in complex with compound 4h revealed that additional ionic interactions are mainly responsible for the increased potency compared to the monocarboxylate analogues. Notably, several compounds displayed high selectivity for PDK1. Employing a prodrug strategy, we were able to corroborate the novel mechanism of action in cells.


Subject(s)
DNA Helicases/antagonists & inhibitors , Drug Design , Malonates/chemistry , Malonates/pharmacology , Muscle Cells/drug effects , Prodrugs/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Allosteric Site , Binding Sites , DNA Helicases/metabolism , Enzyme Activation , Humans , Immunoblotting , Malonates/chemical synthesis , Models, Molecular , Molecular Structure , Muscle Cells/metabolism , Prodrugs/chemical synthesis , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Structure-Activity Relationship
7.
Chem Biol ; 19(9): 1152-63, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22999883

ABSTRACT

The PIF-pocket of AGC protein kinases participates in the physiologic mechanism of regulation by acting as a docking site for substrates and as a switch for the transduction of the conformational changes needed for activation or inhibition. We describe the effects of compounds that bind to the PIF-pocket of PDK1. In vitro, PS210 is a potent activator of PDK1, and the crystal structure of the PDK1-ATP-PS210 complex shows that PS210 stimulates the closure of the kinase domain. However, in cells, the prodrug of PS210 (PS423) acts as a substrate-selective inhibitor of PDK1, inhibiting the phosphorylation and activation of S6K, which requires docking to the PIF-pocket, but not affecting PKB/Akt. This work describes a tool to study the dynamics of PDK1 activity and a potential approach for drug discovery.


Subject(s)
Allosteric Site/drug effects , Chalcones/pharmacology , Dicarboxylic Acids/pharmacology , Prodrugs/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line , Chalcones/chemistry , Dicarboxylic Acids/chemistry , HEK293 Cells , Humans , Mice , Models, Biological , Models, Molecular , Molecular Structure , Molecular Weight , Prodrugs/chemistry , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Structure-Activity Relationship , Substrate Specificity
8.
J Mol Biol ; 410(5): 811-30, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21763490

ABSTRACT

Although LEDGF/p75 is believed to act as a cellular cofactor of lentiviral integration by tethering integrase (IN) to chromatin, there is no good in vitro model to analyze this functionality. We designed an AlphaScreen assay to study how LEDGF/p75 modulates the interaction of human immunodeficiency virus type 1 IN with DNA. IN bound with similar affinity to DNA mimicking the long terminal repeat or to random DNA. While LEDGF/p75 bound DNA strongly, a mutant of LEDGF/p75 with compromised nuclear localization signal (NLS)/AT hook interacted weakly, and the LEDGF/p75 PWWP domain did not interact, corroborating previous reports on the role of NLS and AT hooks in charge-dependent DNA binding. LEDGF/p75 stimulated IN binding to DNA 10-fold to 30-fold. Stimulation of IN-DNA binding required a direct interaction between IN and the C-terminus of LEDGF/p75. Addition of either the C-terminus of LEDGF/p75 (amino acids 325-530) or LEDGF/p75 mutated in the NLS/AT hooks interfered with IN binding to DNA. Our results are consistent with an in vitro model of LEDGF/p75-mediated tethering of IN to DNA. The inhibition of IN-DNA interaction by the LEDGF/p75 C-terminus may provide a novel strategy for the inhibition of HIV IN activity and may explain the potent inhibition of HIV replication observed after the overexpression of C-terminal fragments in cell culture.


Subject(s)
DNA/metabolism , HIV Integrase/metabolism , HIV-1/enzymology , Intercellular Signaling Peptides and Proteins/metabolism , AT Rich Sequence/genetics , Biological Assay , Enzyme-Linked Immunosorbent Assay , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Kinetics , Models, Biological , Mutant Proteins/metabolism , Nuclear Localization Signals/metabolism , Oligonucleotides/metabolism , Protein Binding , Protein Structure, Tertiary , Solubility , Structure-Activity Relationship
9.
Mol Biosyst ; 5(1): 21-31, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19081927

ABSTRACT

One of the major obstacles to pursue the discovery of small molecule inhibitors targeting protein-protein interactions is the flat nature of their interface. X-Ray structures have indeed shown that a large part of the interaction area is buried with atoms closely packed together, implying a lack of available cavities for small molecule binding. Yet, it has become clear that some protein-protein interfaces have a well-defined compact area, commonly referred to as a hot spot, that plays a major role in the affinity of the interaction. These hot spots define potential targets for the development of small molecule protein-protein interaction inhibitors (SMPPIIs). In this review we discuss the interactions between viral and host proteins that have the potential for the future development of SMPPIIs. In light of the current anti-HIV therapy a short overview of protein-protein interactions that may serve as targets for novel drugs is provided. Our hypothesis will exemplify and discuss the interaction between HIV-1 integrase and its cellular cofactor LEDGF/p75, which, as evidenced by crystallography and site directed mutagenesis, displays favourable properties needed for the development of interaction inhibitors.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/therapeutic use , Human Immunodeficiency Virus Proteins/metabolism , Animals , Anti-HIV Agents/adverse effects , HIV Infections/drug therapy , Human Immunodeficiency Virus Proteins/chemistry , Humans , Protein Binding
10.
J Mol Biol ; 372(2): 407-21, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17669426

ABSTRACT

The transcriptional co-activator lens epithelium-derived growth factor (LEDGF) has been shown to protect cells against environmental stress. The protein has been implicated in auto-immunity and cancer, and is present in cells as the p52 or p75 splice variant. Recently, LEDGF/p75, but not p52, was identified as the prominent interaction partner of human immunodeficiency virus type 1 (HIV-1) integrase. This interaction of HIV-1 integrase with the C-terminal integrase-binding domain of LEDGF/p75 is crucial for HIV-1 replication. To gain insight into the cell biology of LEDGF/p75, we were interested in identifying cellular binding partners of its C-terminal domain. By yeast-two-hybrid screening with a CEMC7 cDNA-library, we were able to identify JPO2 as a binding partner of the C-terminal part of LEDGF/p75. The specific interaction between JPO2 and LEDGF/p75 was verified by pull-down, AlphaScreen, and co-immunoprecipitation. Competition assays using recombinant proteins show a mutually exclusive binding of either JPO2 or HIV-1 integrase to LEDGF/p75. However, differing mechanisms of binding were suggested by continuing interaction of JPO2 with some LEDGF/p75 mutants (I365A, D366A, F406A) that are totally defective for interaction with HIV-1 integrase. This finding is of significance for the development of specific inhibitors targeting only the interaction between LEDGF/p75 and HIV-1 integrase, without disturbing interaction with other cellular factors. Over-expression of JPO2 resulted in a modest but reproducible inhibition of HIV-1 replication, consistent with competition between integrase and JPO2 for binding to LEDGF/p75. Furthermore, JPO2 over-expression activated transcription from the HIV-1 LTR.


Subject(s)
HIV Integrase/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , Gene Expression , HIV-1/enzymology , HIV-1/physiology , HeLa Cells , Humans , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding/drug effects , Repressor Proteins/chemistry , Repressor Proteins/genetics , Virus Replication , Zinc/pharmacology
11.
J Mol Biol ; 365(5): 1480-92, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17137594

ABSTRACT

Lens epithelium-derived growth factor (LEDGF)/p75 is an important cellular co-factor for human immunodeficiency virus (HIV) replication. We originally identified LEDGF/p75 as a binding partner of integrase (IN) in human cells. The interaction has been mapped to the integrase-binding domain (IBD) of LEDGF/p75 located in the C-terminal part. We have subsequently shown that IN carrying the Q168A mutation remains enzymatically active but is impaired for interaction with LEDGF/p75. To map the integrase/LEDGF interface in more detail, we have now identified and characterized two regions within the enzyme involved in the interaction with LEDGF/p75. The first region centers around residues W131 and W132 while the second extends from I161 up to E170. For the different IN mutants the interaction with LEDGF/p75 and the enzymatic activities were determined. IN(W131A), IN(I161A), IN(R166A), IN(Q168A) and IN(E170A) are impaired for interaction with LEDGF/p75, but retain 3' processing and strand transfer activities. Due to impaired integration, an HIV-1 strain containing the W131A mutation in IN displays reduced replication capacity, whereas virus carrying IN(Q168A) is replication defective. Comparison of the wild-type IN-LEDGF/p75 co-crystal structure with that of the modelled structure of the IN(Q168A) and IN(W131A) mutant integrases corroborated our experimental data.


Subject(s)
HIV Integrase/chemistry , HIV Integrase/metabolism , HIV-1/enzymology , Intercellular Signaling Peptides and Proteins/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , DNA, Viral/biosynthesis , HIV-1/physiology , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Protein Interaction Mapping , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Virus Replication/physiology
12.
Trends Biochem Sci ; 31(2): 98-105, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16403635

ABSTRACT

To achieve productive infection, the reverse transcribed cDNA of human immunodeficiency virus type 1 (HIV-1) is inserted in the host cell genome. The main protein responsible for this reaction is the viral integrase. However, studies indicate that the virus is assisted by cellular proteins, or co-factors, to achieve integration into the infected cell. The barrier-to-autointegration factor (BAF) might prevent autointegration. Its ability to bridge DNA and the finding that the nuclear lamina-associated polypeptide-2alpha interacts with BAF suggest a role in nuclear structure organization. Integrase interactor 1 was found to directly interact with HIV-1 integrase and to activate its DNA-joining activity, and the high mobility group chromosomal protein A1 might approximate both long terminal repeat (LTR) ends and facilitate integrase binding by unwinding the LTR termini. Furthermore, the lens-epithelium-derived growth factor (LEDGF; also known as p75) seems to tether HIV-1 integrase to the chromosomes. Although a direct role in integration has only been demonstrated for LEDGF/p75, to date, each validated cellular co-factor for HIV-1 integration could constitute a promising new target for antiviral therapy.


Subject(s)
HIV Integrase/metabolism , HIV-1/physiology , Virus Integration/physiology , Animals , Chromosomal Proteins, Non-Histone , DNA, Viral/metabolism , DNA-Binding Proteins/physiology , HMGA1a Protein/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Nuclear Proteins/physiology , SMARCB1 Protein , Transcription Factors/physiology
13.
J Biol Chem ; 280(27): 25517-23, 2005 Jul 08.
Article in English | MEDLINE | ID: mdl-15855167

ABSTRACT

The insertion of a DNA copy of its RNA genome into a chromosome of the host cell is mediated by the viral integrase with the help of mostly uncharacterized cellular cofactors. We have recently described that the transcriptional co-activator LEDGF/p75 strongly interacts with HIV-1 integrase. Here we show that interaction of HIV-1 integrase with LEDGF/p75 is important for viral replication. Using multiple approaches including two-hybrid interaction studies, random and directed mutagenesis, we could demonstrate that HIV-1 virus harboring a single mutation that disrupts integrase-LEDGF/p75 interaction, resulted in defective HIV-1 replication. Furthermore, we found that LEDGF/p75 tethers HIV-1 integrase to chromosomes and that this interaction may be important for the integration process and the replication of HIV-1.


Subject(s)
HIV Infections/virology , HIV Integrase/genetics , HIV Integrase/metabolism , HIV-1/growth & development , Intercellular Signaling Peptides and Proteins/metabolism , Chromosomes, Human/virology , Glutamine/metabolism , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/genetics , HeLa Cells , Humans , Virus Integration/physiology , Virus Replication/physiology
14.
J Biol Chem ; 280(18): 17841-7, 2005 May 06.
Article in English | MEDLINE | ID: mdl-15749713

ABSTRACT

We have previously shown that the p75 isoform of the transcriptional co-activator lens epithelium-derived growth factor (LEDGF) interacts tightly with human immunodeficiency virus (HIV)-1 integrase (IN) and is essential for nuclear targeting of this protein in human cells (Cherepanov, P., Maertens, G., Proost, P., Devreese, B., Van Beeumen, J., Engelborghs, Y., De Clercq, E., and Debyser, Z. (2003) J. Biol. Chem. 278, 372-381; Maertens, G., Cherepanov, P., Pluymers, W., Busschots, K., De Clercq, E., Debyser, Z., and Engelborghs, Y. (2003) J. Biol. Chem. 278, 33528-33539). Here the interaction between recombinant LEDGF/p75 and HIV-1 IN was examined in a pull-down binding test. LEDGF/p75 was shown to increase the solubility of HIV-1 IN. Next, fluorescent correlation spectroscopy was used to measure the interaction of LEDGF/p75 or the complex of HIV-1 IN and LEDGF/p75 with a specific double-stranded DNA oligonucleotide. Whereas LEDGF/p75 displayed only a moderate affinity for DNA, it strongly promoted the binding of HIV-1 IN to DNA. This effect was specific for the p75 isoform of LEDGF and was not seen with p52. In the pull-down assay LEDGF/p75 interacted with HIV-1, HIV-2, and feline immunodeficiency virus IN, but not with the IN of human T-cell lymphotropic virus type 2, Moloney murine leukemia virus, or Rous sarcoma virus. These results strongly suggest that the interaction of LEDGF/p75 with IN is specific to lentiviridae. LEDGF/p75 stimulated the binding of HIV-1 and HIV-2 IN, but not Moloney murine leukemia virus or Rous sarcoma virus IN, to an aspecific DNA. These results provide supporting evidence for our hypothesis that LEDGF/p75 plays a role in the tethering of lentiviral IN to the chromosomal DNA.


Subject(s)
DNA, Viral/metabolism , Integrases/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lentivirus/metabolism , Binding Sites/physiology , DNA, Viral/genetics , Humans , Integrases/genetics , Intercellular Signaling Peptides and Proteins/genetics , Lentivirus/enzymology , Lentivirus/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism
15.
J Biol Chem ; 278(35): 33528-39, 2003 Aug 29.
Article in English | MEDLINE | ID: mdl-12796494

ABSTRACT

We have reported that human immunodeficiency virus type 1 (HIV-1) integrase (IN) forms a specific nuclear complex with human lens epithelium-derived growth factor/transcription co-activator p75 (LEDGF/p75) protein. We now studied the IN-LEDGF/p75 interaction and nuclear import of IN in living cells using fusions of IN and LEDGF/p75 with enhanced green fluorescent protein and far-red fluorescent protein HcRed1. We show that both the N-terminal zinc binding domain and the central core domains of IN are involved in the interaction with LEDGF/p75. Both domains are essential for nuclear localization of IN as well as for the association of IN with condensed chromosomes during mitosis. However, upon overexpression of LEDGF/p75, the core domain fragment of IN was recruited to the nuclei and mitotic chromosomes with a distribution pattern characteristic of the full-length protein, indicating that it harbors the main determinant for interaction with LEDGF/p75. Although the C-terminal domain of IN was dispensable for nuclear/chromosomal localization, a fusion of the C-terminal IN fragment with enhanced green fluorescent protein was found exclusively in the nucleus, with a diffuse nuclear/nucleolar distribution, suggesting that the C-terminal domain may also play a role in the nuclear import of IN. In contrast to LEDGF/p75, its alternative splice variant, p52, did not interact with HIV-1 IN in vitro and in living cells. Finally, RNA interference-mediated knock-down of endogenous LEDGF/p75 expression abolished nuclear/chromosomal localization of IN. We conclude, therefore, that the interaction with LEDGF/p75 accounts for the karyophilic properties and chromosomal targeting of HIV-1 IN.


Subject(s)
Cell Nucleus/metabolism , HIV Integrase/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Active Transport, Cell Nucleus , Alternative Splicing , Base Sequence , Blotting, Western , Cells, Cultured , Cytoplasm/metabolism , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Microscopy, Confocal , Microscopy, Fluorescence , Mitosis , Models, Genetic , Molecular Sequence Data , Mutation , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...