Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biomacromolecules ; 19(10): 3945-3957, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30160482

ABSTRACT

The simultaneous expression in Escherichia coli cells of the Qß virus-like particle (VLP) capsid protein and protein "cargo" tagged with a positively charged Rev peptide sequence leads to the spontaneous self-assembly of VLPs with multiple copies of the cargo inside. We report the packaging of four new enzymes with potential applications in medicine and chemical manufacturing. The captured enzymes are active while inside the nanoparticle shell and are protected from environmental conditions that lead to free-enzyme destruction. We also describe genetic modifications to the packaging scheme that shed light on the self-assembly mechanism of this system and allow indirect control over the internal packaging density of cargo. The technology was extended to create, via self-assembly, VLPs that simultaneously display protein ligands on the exterior and contain enzymes within. Inverse relationships were observed between the size of both the packaged and externally displayed protein or domains and nanoparticle yield. These results provide a general method for the rapid creation of robust protein nanoparticles with desired catalytic and targeting functionalities.


Subject(s)
Capsid Proteins/metabolism , Gene Products, rev/metabolism , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/metabolism , Nanoparticles/metabolism , RNA, Viral/metabolism , Virus Assembly , Aldehyde-Lyases/chemistry , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Catalysis , Cytosine Deaminase/chemistry , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Gene Products, rev/chemistry , Gene Products, rev/genetics , HeLa Cells , Humans , Multifunctional Enzymes/genetics , Nanoparticles/chemistry , RNA, Viral/chemistry , RNA, Viral/genetics
2.
Retrovirology ; 14(1): 40, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28830558

ABSTRACT

BACKGROUND: Rev-like proteins are post-transcriptional regulatory proteins found in several retrovirus genera, including lentiviruses, betaretroviruses, and deltaretroviruses. These essential proteins mediate the nuclear export of incompletely spliced viral RNA, and act by tethering viral pre-mRNA to the host CRM1 nuclear export machinery. Although all Rev-like proteins are functionally homologous, they share less than 30% sequence identity. In the present study, we computationally assessed the extent of structural homology among retroviral Rev-like proteins within a phylogenetic framework. RESULTS: We undertook a comprehensive analysis of overall protein domain architecture and predicted secondary structural features for representative members of the Rev-like family of proteins. Similar patterns of α-helical domains were identified for Rev-like proteins within each genus, with the exception of deltaretroviruses, which were devoid of α-helices. Coiled-coil oligomerization motifs were also identified for most Rev-like proteins, with the notable exceptions of HIV-1, the deltaretroviruses, and some small ruminant lentiviruses. In Rev proteins of primate lentiviruses, the presence of predicted coiled-coil motifs segregated within specific primate lineages: HIV-1 descended from SIVs that lacked predicted coiled-coils in Rev whereas HIV-2 descended from SIVs that contained predicted coiled-coils in Rev. Phylogenetic ancestral reconstruction of coiled-coils for all Rev-like proteins predicted a single origin for the coiled-coil motif, followed by three losses of the predicted signal. The absence of a coiled-coil signal in HIV-1 was associated with replacement of canonical polar residues with non-canonical hydrophobic residues. However, hydrophobic residues were retained in the key 'a' and 'd' positions, and the α-helical region of HIV-1 Rev oligomerization domain could be modeled as a helical wheel with two predicted interaction interfaces. Moreover, the predicted interfaces mapped to the dimerization and oligomerization interfaces in HIV-1 Rev crystal structures. Helical wheel projections of other retroviral Rev-like proteins, including endogenous sequences, revealed similar interaction interfaces that could mediate oligomerization. CONCLUSIONS: Sequence-based computational analyses of Rev-like proteins, together with helical wheel projections of oligomerization domains, reveal a conserved homogeneous structural basis for oligomerization by retroviral Rev-like proteins.


Subject(s)
Gene Products, rev/chemistry , Gene Products, rev/metabolism , Models, Molecular , Retroviridae/chemistry , Retroviridae/metabolism , Amino Acid Sequence , Dimerization , Genetic Variation , Phylogeny , Protein Structure, Secondary , Retroviridae Proteins/chemistry , Retroviridae Proteins/metabolism , Sequence Homology, Amino Acid
3.
Retrovirology ; 11: 115, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25533001

ABSTRACT

BACKGROUND: The lentiviral Rev protein mediates nuclear export of intron-containing viral RNAs that encode structural proteins or serve as the viral genome. Following translation, HIV-1 Rev localizes to the nucleus and binds its cognate sequence, termed the Rev-responsive element (RRE), in incompletely spliced viral RNA. Rev subsequently multimerizes along the viral RNA and associates with the cellular Crm1 export machinery to translocate the RNA-protein complex to the cytoplasm. Equine infectious anemia virus (EIAV) Rev is functionally homologous to HIV-1 Rev, but shares very little sequence similarity and differs in domain organization. EIAV Rev also contains a bipartite RNA binding domain comprising two short arginine-rich motifs (designated ARM-1 and ARM-2) spaced 79 residues apart in the amino acid sequence. To gain insight into the topology of the bipartite RNA binding domain, a computational approach was used to model the tertiary structure of EIAV Rev. RESULTS: The tertiary structure of EIAV Rev was modeled using several protein structure prediction and model quality assessment servers. Two types of structures were predicted: an elongated structure with an extended central alpha helix, and a globular structure with a central bundle of helices. Assessment of models on the basis of biophysical properties indicated they were of average quality. In almost all models, ARM-1 and ARM-2 were spatially separated by >15 Å, suggesting that they do not form a single RNA binding interface on the monomer. A highly conserved canonical coiled-coil motif was identified in the central region of EIAV Rev, suggesting that an RNA binding interface could be formed through dimerization of Rev and juxtaposition of ARM-1 and ARM-2. In support of this, purified Rev protein migrated as a dimer in Blue native gels, and mutation of a residue predicted to form a key coiled-coil contact disrupted dimerization and abrogated RNA binding. In contrast, mutation of residues outside the predicted coiled-coil interface had no effect on dimerization or RNA binding. CONCLUSIONS: Our results suggest that EIAV Rev binding to the RRE requires dimerization via a coiled-coil motif to juxtapose two RNA binding motifs, ARM-1 and ARM-2.


Subject(s)
Gene Products, rev/chemistry , Gene Products, rev/metabolism , Infectious Anemia Virus, Equine/physiology , Protein Multimerization , RNA, Viral/metabolism , Models, Molecular , Protein Binding , Protein Conformation
4.
J Virol ; 86(9): 4892-905, 2012 May.
Article in English | MEDLINE | ID: mdl-22379104

ABSTRACT

The Rev protein is essential for the replication of lentiviruses. Rev is a shuttling protein that transports unspliced and partially spliced lentiviral RNAs from the nucleus to the cytoplasm via the nucleopore. To transport these RNAs, the human immunodeficiency virus type 1 (HIV-1) Rev uses the karyopherin ß family importin ß and CRM1 proteins that interact with the Rev nuclear localization signal (NLS) and nuclear exportation signal (NES), respectively. Recently, we reported the presence of new types of bipartite NLS and nucleolar localization signal (NoLS) in the bovine immunodeficiency virus (BIV) Rev protein. Here we report the characterization of the nuclear import and export pathways of BIV Rev. By using an in vitro nuclear import assay, we showed that BIV Rev is transported into the nucleus by a cytosolic and energy-dependent importin α/ß classical pathway. Results from glutathione S-transferase (GST) pulldown assays that showed the binding of BIV Rev with importins α3 and α5 were in agreement with those from the nuclear import assay. We also identified a leptomycin B-sensitive NES in BIV Rev, which indicates that the protein is exported via CRM1 like HIV-1 Rev. Mutagenesis experiments showed that the BIV Rev NES maps between amino acids 109 to 121 of the protein. Remarkably, the BIV Rev NES was found to be of the cyclic AMP (cAMP)-dependent protein kinase inhibitor (PKI) type instead of the HIV-1 Rev type. In summary, our data showed that the nuclear import mechanism of BIV Rev is novel among Rev proteins characterized so far in lentiviruses.


Subject(s)
Gene Products, rev/chemistry , Gene Products, rev/metabolism , Immunodeficiency Virus, Bovine/metabolism , Nuclear Export Signals , Active Transport, Cell Nucleus/physiology , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cattle , Fatty Acids, Unsaturated/pharmacology , Humans , Karyopherins/antagonists & inhibitors , Nuclear Pore/metabolism , Protein Binding , Protein Isoforms/metabolism , Protein Transport , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Signal Transduction , alpha Karyopherins/metabolism , beta Karyopherins/metabolism , ran GTP-Binding Protein/metabolism , Exportin 1 Protein
5.
Proc Natl Acad Sci U S A ; 107(28): 12481-6, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20616058

ABSTRACT

RNA is a crucial structural component of many ribonucleoprotein (RNP) complexes, including the ribosome, spliceosome, and signal recognition particle, but the role of RNA in guiding complex formation is only beginning to be explored. In the case of HIV, viral replication requires assembly of an RNP composed of the Rev protein homooligomer and the Rev response element (RRE) RNA to mediate nuclear export of unspliced viral mRNAs. Assembly of the functional Rev-RRE complex proceeds by cooperative oligomerization of Rev on the RRE scaffold and utilizes both protein-protein and protein-RNA interactions to organize complexes with high specificity. The structures of the Rev protein and a peptide-RNA complex are known, but the complete RNP is not, making it unclear to what extent RNA defines the composition and architecture of Rev-RNA complexes. Here we show that the RRE controls the oligomeric state and solubility of Rev and guides its assembly into discrete Rev-RNA complexes. SAXS and EM data were used to derive a structural model of a Rev dimer bound to an essential RRE hairpin and to visualize the complete Rev-RRE RNP, demonstrating that RRE binding drives assembly of Rev homooligomers into asymmetric particles, reminiscent of the role of RNA in organizing more complex RNP machines, such as the ribosome, composed of many different protein subunits. Thus, the RRE is not simply a passive scaffold onto which proteins bind but instead actively defines the protein composition and organization of the RNP.


Subject(s)
Gene Products, rev/chemistry , Gene Products, rev/metabolism , HIV/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , Gene Products, rev/genetics , HIV/metabolism , HIV Infections/genetics , HIV Infections/metabolism , Peptides/genetics , Peptides/metabolism , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Scattering, Small Angle , Virus Replication/genetics
6.
J Biol Chem ; 285(28): 21768-80, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20406803

ABSTRACT

The human immunodeficiency virus, type 1 (HIV-1)-encoded Rev protein is essential for the expression of late viral mRNAs. Rev forms a large organized multimeric protein-protein complex on the Rev response element of these viral mRNA species and transports them from the nucleus to the cytoplasm, exploiting the CRM1-mediated cellular machinery. Here we report the selection of a nanobody, derived from a llama heavy-chain only antibody, that efficiently blocks the assembly of Rev multimers. The nanobody inhibits HIV-1 replication in cells and specifically suppresses the Rev-dependent expression of partially spliced and unspliced HIV-1 RNA. In HIV-susceptible cells, this nanobody thus has potential as an effective anti-HIV agent using genetic immunization strategies. Its binding site was mapped to Rev residues Lys-20 and Tyr-23 located in the N-terminal alpha-helical multimerization domain. In the presence of this nanobody, we observed an accumulation of dimeric Rev species, supporting a head-to-head/tail-to-tail molecular model for Rev assembly. The results indicate that the oligomeric assembly of Rev follows an ordered stepwise process and identify a new epitope within Rev that could guide strategies for the development of novel HIV inhibitors.


Subject(s)
Gene Products, rev/chemistry , HIV Infections/drug therapy , HIV-1/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Anti-HIV Agents/pharmacology , Antibodies/chemistry , Camelids, New World , Dimerization , Fluorescence Resonance Energy Transfer , Gene Products, rev/metabolism , HeLa Cells , Humans , Lysine/chemistry , Molecular Sequence Data , Protein Interaction Mapping , Protein Structure, Secondary , Protein Structure, Tertiary , Tyrosine/chemistry
7.
J Am Chem Soc ; 133(25): 9912-22, 2011 Jun 29.
Article in English | MEDLINE | ID: mdl-21585196

ABSTRACT

A series of compounds that target reactive metal chelates to the HIV-1 Rev response element (RRE) mRNA have been synthesized. Dissociation constants and chemical reactivity toward HIV RRE RNA have been determined and evaluated in terms of reduction potential, coordination unsaturation, and overall charge associated with the metal-chelate-Rev complex. Ethylenediaminetetraacetic acid (EDTA), nitrilotriacetic acid (NTA), diethylenetriaminepentaacetic acid (DTPA), and 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) were linked to a lysine side chain of a Rev-derived peptide by either EDC/NHS or isothiocyanate coupling. The resulting chelate-Rev (EDTA-Rev, DTPA-Rev, NTA-Rev, and DOTA-Rev) conjugates were used to form coordination complexes with Fe(2+), Co(2+), Ni(2+), and Cu(2+) such that the arginine-rich Rev peptide could mediate localization of the metal chelates to the Rev peptide's high-affinity mRNA binding partner, RRE stem loop IIB. Metal complexes of the extended peptides GGH-Rev and KGHK-Rev, which also contain N-terminal peptidic chelators (ATCUN motifs), were studied for comparison. A fluorescence titration assay revealed high-affinity RRE RNA binding by all 22 metal-chelate-Rev species, with K(D) values ranging from ~0.2 to 16 nM, indicating little to no loss of RNA affinity due to the coupling of the metal chelates to the Rev peptide. Dissociation constants for binding at a previously unobserved low-affinity site are also reported. Rates of RNA modification by each metal-chelate-Rev species were determined and varied from ~0.28 to 4.9 nM/min but were optimal for Cu(2+)-NTA-Rev. Metal-chelate reduction potentials were determined and varied from -228 to +1111 mV vs NHE under similar solution conditions, allowing direct comparison of reactivity with redox thermodynamics. Optimal activity was observed when the reduction potential for the metal center was poised between those of the two principal co-reagents for metal-promoted formation of reactive oxygen species: E°(ascorbate/ascorbyl radical) = -66 mV and E°(H(2)O(2)/hydroxyl radical) = 380 mV. Given the variety of oxidative activities of these metal complexes and their high-affinity binding to the targeted RRE mRNA following coupling to the Rev peptide, this class of metal-chelate-Rev derivatives constitutes a promising step toward development of multiple-turnover reagents for selective eradication of HIV-1 RRE mRNA.


Subject(s)
Anti-HIV Agents/chemical synthesis , Chelating Agents/pharmacology , Gene Products, rev/genetics , HIV/genetics , RNA, Viral/drug effects , Response Elements/drug effects , Chelating Agents/chemistry , Gene Products, rev/chemistry , Hydrolysis , Metals, Heavy , Oxidation-Reduction , Reactive Oxygen Species
8.
J Virol ; 83(24): 12842-53, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19828621

ABSTRACT

The bovine immunodeficiency virus (BIV) Rev protein (186 amino acids [aa] in length) is involved in the nuclear exportation of partially spliced and unspliced viral RNAs. Previous studies have shown that BIV Rev localizes in the nucleus and nucleolus of infected cells. Here we report the characterization of the nuclear/nucleolar localization signals (NLS/NoLS) of this protein. Through transfection of a series of deletion mutants of BIV Rev fused to enhanced green fluorescent protein and fluorescence microscopy analyses, we were able to map the NLS region between aa 71 and 110 of the protein. Remarkably, by conducting alanine substitution of basic residues within the aa 71 to 110 sequence, we demonstrated that the BIV Rev NLS is bipartite, maps to aa 71 to 74 and 95 to 101, and is predominantly composed of arginine residues. This is the first report of a bipartite Rev (or Rev-like) NLS in a lentivirus/retrovirus. Moreover, this NLS is atypical, as the length of the sequence between the motifs composing the bipartite NLS, e.g., the spacer sequence, is 20 aa. Further mutagenesis experiments also identified the NoLS region of BIV Rev. It localizes mainly within the NLS spacer sequence. In addition, the BIV Rev NoLS sequence differs from the consensus sequence reported for other viral and cellular nucleolar proteins. In summary, we conclude that the nucleolar and nuclear localizations of BIV Rev are mediated via novel NLS and NoLS motifs.


Subject(s)
Gene Products, rev/analysis , Immunodeficiency Virus, Bovine/chemistry , Nuclear Localization Signals , Amino Acid Sequence , Animals , Cell Nucleolus/chemistry , Cell Nucleus/chemistry , Cells, Cultured , Gene Products, rev/chemistry , Humans , Molecular Sequence Data
9.
Open Biol ; 10(12): 200320, 2020 12.
Article in English | MEDLINE | ID: mdl-33352061

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) proteome is expressed from alternatively spliced and unspliced genomic RNAs. However, HIV-1 RNAs that are not fully spliced are perceived by the host machinery as defective and are retained in the nucleus. During late infection, HIV-1 bypasses this regulatory mechanism by expression of the Rev protein from a fully spliced mRNA. Once imported into the nucleus, Rev mediates the export of unprocessed HIV-1 RNAs to the cytoplasm, leading to the production of the viral progeny. While regarded as a canonical RNA export factor, Rev has also been linked to HIV-1 RNA translation, stabilization, splicing and packaging. However, Rev's functions beyond RNA export have remained poorly understood. Here, we revisit this paradigmatic protein, reviewing recent data investigating its structure and function. We conclude by asking: what remains unknown about this enigmatic viral protein?


Subject(s)
HIV Infections/virology , HIV/physiology , Animals , Drug Discovery , Gene Expression Regulation, Viral , Gene Products, rev/chemistry , Gene Products, rev/genetics , Gene Products, rev/metabolism , HIV Infections/drug therapy , HIV Infections/metabolism , Host-Pathogen Interactions , Humans , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
10.
Science ; 273(5281): 1547-51, 1996 Sep 13.
Article in English | MEDLINE | ID: mdl-8703216

ABSTRACT

The solution structure of a human immunodeficiency virus type-1 (HIV-1) Rev peptide bound to stem-loop IIB of the Rev response element (RRE) RNA was solved by nuclear magnetic resonance spectroscopy. The Rev peptide has an alpha-helical conformation and binds in the major groove of the RNA near a purine-rich internal loop. Several arginine side chains make base-specific contacts, and an asparagine residue contacts a G.A base pair. The phosphate backbone adjacent to a G.G base pair adopts an unusual structure that allows the peptide to access a widened major groove. The structure formed by the two purine-purine base pairs of the RRE creates a distinctive binding pocket that the peptide can use for specific recognition.


Subject(s)
DNA-Binding Proteins , Gene Products, rev/chemistry , Gene Products, rev/metabolism , Genes, env , HIV-1/chemistry , Nucleic Acid Conformation , Protein Structure, Secondary , RNA, Viral/chemistry , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Arginine/chemistry , Asparagine/chemistry , Base Composition , Base Sequence , Fungal Proteins/chemistry , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Kinases/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , Threonine/chemistry , rev Gene Products, Human Immunodeficiency Virus
11.
Science ; 294(5548): 1841-2, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11729289

ABSTRACT

There seem to be numerous pathways for exporting mRNAs from the nucleus to the cytoplasm. But working out which set of export adaptors and receptors transport individual mRNAs has been very difficult. In a Perspective, Moore and Rosbash discuss a new strategy using cell-penetrating peptide inhibitors for unraveling the routes of mRNA export in living cells (Gallouzi and Steitz).


Subject(s)
Antigens, Surface , Cell Nucleus/metabolism , Cytoplasm/metabolism , Karyopherins/metabolism , Nucleocytoplasmic Transport Proteins , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear , Transcription Factors , Animals , Antennapedia Homeodomain Protein , Biological Transport/drug effects , Cell Membrane Permeability , Cell Nucleus/drug effects , Cytoplasm/drug effects , ELAV Proteins , ELAV-Like Protein 1 , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Gene Products, rev/chemistry , Gene Products, rev/metabolism , HIV/genetics , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Humans , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Binding/drug effects , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , rev Gene Products, Human Immunodeficiency Virus , Exportin 1 Protein
12.
PLoS One ; 14(8): e0221505, 2019.
Article in English | MEDLINE | ID: mdl-31437223

ABSTRACT

The lentiviral Rev protein, which is a regulatory protein essential for virus replication, has been first studied in the human immunodeficiency virus type 1 (HIV-1). The main function of Rev is to mediate the nuclear exportation of viral RNAs. To fulfill its function, Rev shuttles between the cytoplasm and the nucleus. The Jembrana disease virus (JDV), a lentivirus, is the etiologic agent of the Jembrana disease which was first described in Bali cattle in Indonesia in 1964. Despite the high mortality rate associated with JDV, this virus remains poorly studied. Herein the subcellular distribution of JDV Rev, the nuclear and nucleolar localization signals (NLS and NoLS, respectively) and the nuclear export signal (NES) of the protein were examined. JDV Rev fused to the enhanced green fluorescent protein (EGFP) predominantly localized to the cytoplasm and nucleolus of transfected cells, as determined by fluorescence microscopy analyses. Through transfection of a series of deletion mutants of JDV Rev, it was possible to localize the NLS/NoLS region between amino acids (aa) 74 to 105. By substituting basic residues with alanine within this sequence, we demonstrated that the JDV Rev NLS encompasses aa 76 to 86, and is exclusively composed of arginine residues, whereas a bipartite NoLS was observed for the first time in any retroviral Rev/Rev-like proteins. Finally, a NES was identified downstream of the NLS/NoLS and encompasses aa 116 to 128 of the JDV Rev protein. The JDV Rev NES was found to be of the protein kinase A inhibitor (PKI) class instead of the HIV-1 Rev class. It also corresponds to the most optimal consensus sequence of PKI NES and, as such, is novel among lentiviral Rev NES.


Subject(s)
Cell Nucleolus/metabolism , Gene Products, rev/metabolism , Lentivirus/metabolism , Nuclear Export Signals , Amino Acid Motifs , Amino Acid Sequence , Animals , Cattle , Cell Line , Dogs , Gene Products, rev/chemistry , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mutant Proteins/metabolism , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/metabolism , Protein Transport , Recombinant Fusion Proteins/metabolism
13.
Mol Cell Biol ; 19(2): 1210-7, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9891055

ABSTRACT

Protein nuclear import is generally mediated by basic nuclear localization signals (NLSs) that serve as targets for the importin alpha (Imp alpha) NLS receptor. Imp alpha is in turn bound by importin beta (Imp beta), which targets the resultant protein complex to the nucleus. Here, we report that the arginine-rich NLS sequences present in the human immunodeficiency virus type 1 regulatory proteins Tat and Rev fail to interact with Imp alpha and instead bind directly to Imp beta. Using in vitro nuclear import assays, we demonstrate that Imp alpha is entirely dispensable for Tat and Rev nuclear import. In contrast, Imp beta proved both sufficient and necessary, in that other beta-like import factors, such as transportin, were unable to support Tat or Rev nuclear import. Using in vitro competition assays, it was demonstrated that the target sites on Imp beta for Imp alpha, Tat, and Rev binding either are identical or at least overlap. The interaction of Tat and Rev with Imp beta is also similar to Imp alpha binding in that it is inhibited by RanGTP but not RanGDP, a finding that may in part explain why the interaction of the Rev nuclear RNA export factor with target RNA species is efficient in the cell nucleus yet is released in the cytoplasm. Together, these studies define a novel class of arginine-rich NLS sequences that are direct targets for Imp beta and that therefore function independently of Imp alpha.


Subject(s)
Gene Products, rev/chemistry , Gene Products, rev/metabolism , Gene Products, tat/chemistry , Gene Products, tat/metabolism , HIV-1/metabolism , Nuclear Proteins/metabolism , Amino Acid Sequence , Arginine/chemistry , Binding Sites/genetics , Gene Products, rev/genetics , Gene Products, tat/genetics , HIV-1/genetics , HeLa Cells , Humans , In Vitro Techniques , Karyopherins , Nuclear Localization Signals , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , ran GTP-Binding Protein , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
14.
Curr Opin Struct Biol ; 9(1): 74-87, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10047585

ABSTRACT

Recently, progress has been made towards the structural characterization of the novel folds of RNA-bound arginine-rich peptides and the architecture of their peptide-binding RNA pockets in viral and phage systems. These studies are based on an approach whereby the peptide and RNA components are minimalist modular domains that undergo adaptive structural transitions upon complex formation. Such complexes are characterized by recognition alignments in which the tertiary fold of the RNA generates binding pockets with the potential to envelop minimal elements of protein secondary structure. Strikingly, the peptides fold as isolated alpha-helical or beta-hairpin folds within their RNA major-groove targets, without the necessity of additional appendages for anchorage within the binding pocket. The RNA peptide-binding pocket architectures are sculptured through precisely positioned mismatches, triples and looped-out bases, which accommodate amino acid sidechains through hydrophobic, hydrogen bonding and ionic intermolecular contacts. By contrast, protein modules associated with the HIV-1 nucleocapsid and MS2 phage coat target their RNA binding sites through the insertion of specificity-determining RNA base residues within conserved hydrophobic pockets and crevices on the protein surface, with the bases anchored through hydrogen bonding interactions. These alternative strategies of RNA recognition at the peptide and protein module level provide novel insights into the principles, patterns and diversity of the adaptive transitions associated with the recognition process.


Subject(s)
Peptides/chemistry , Peptides/metabolism , RNA, Viral/chemistry , RNA, Viral/metabolism , Amino Acid Sequence , Animals , Bacteriophages/genetics , Bacteriophages/physiology , Base Sequence , Binding Sites , Cattle , Gene Products, rev/chemistry , Gene Products, rev/genetics , Gene Products, rev/metabolism , HIV-1/genetics , HIV-1/physiology , Humans , Immunodeficiency Virus, Bovine/genetics , Immunodeficiency Virus, Bovine/physiology , Models, Molecular , Molecular Sequence Data , Peptides/genetics , Protein Conformation , RNA, Viral/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , rev Gene Products, Human Immunodeficiency Virus
15.
Retrovirology ; 3: 1, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16398928

ABSTRACT

CTL based vaccine strategies in the macaque model of AIDS have shown promise in slowing the progression to disease. However, rapid CTL escape viruses can emerge rendering such vaccination useless. We hypothesized that such escape is made more difficult if the immunizing CTL epitope falls within a region of the virus that has a high density of overlapping reading frames which encode several viral proteins. To test this hypothesis, we immunized macaques using a peptide-loaded dendritic cell approach employing epitopes in the second coding exon of SIV Tat which spans reading frames for both Env and Rev. We report here that autologous dendritic cells, loaded with SIV peptides from Tat, Rev, and Env, induced a distinct cellular immune response measurable ex vivo. However, conclusive in vivo control of a challenge inoculation of SIVmac239 was not observed suggesting that CTL epitopes within densely overlapping reading frames are also subject to escape mutations.


Subject(s)
Dendritic Cells/immunology , Gene Products, env/genetics , Gene Products, rev/genetics , Gene Products, tat/genetics , Reading Frames/genetics , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit , Viral Load , Animals , CD4 Lymphocyte Count , DNA Primers , Enzyme-Linked Immunosorbent Assay , Gene Products, env/chemistry , Gene Products, env/immunology , Gene Products, rev/chemistry , Gene Products, rev/immunology , Gene Products, tat/chemistry , Gene Products, tat/immunology , Macaca mulatta , Molecular Sequence Data , Mutation , Peptide Fragments/immunology , Polymerase Chain Reaction , Vaccination
16.
J Mol Biol ; 351(5): 982-94, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-16055152

ABSTRACT

DNA in a single-stranded form (ssDNA) exists transiently within the cell and comprises the telomeres of linear chromosomes and the genomes of some DNA viruses. As with RNA, in the single-stranded state, some DNA sequences are able to fold into complex secondary and tertiary structures that may be recognized by proteins and participate in gene regulation. To better understand how such DNA elements might fold and interact with proteins, and to compare recognition features to those of a structured RNA, we used in vitro selection to identify ssDNAs that bind an RNA-binding peptide from the HIV Rev protein with high affinity and specificity. The large majority of selected binders contain a non-Watson-Crick G.T base-pair and an adjacent C:G base-pair and both are essential for binding. This GT motif can be presented in different DNA contexts, including a nearly perfect duplex and a branched three-helix structure, and appears to be recognized in large part by arginine residues separated by one turn of an alpha-helix. Interestingly, a very similar GT motif is necessary also for protein binding and function of a well-characterized model ssDNA regulatory element from the proenkephalin promoter.


Subject(s)
DNA, Single-Stranded/chemistry , DNA/chemistry , Nucleic Acid Conformation , Amino Acid Motifs , Arginine/chemistry , Base Sequence , Circular Dichroism , Dose-Response Relationship, Drug , Enkephalins/genetics , Gene Products, rev/chemistry , Macromolecular Substances , Molecular Sequence Data , Peptides/chemistry , Promoter Regions, Genetic , Protein Binding , Protein Precursors/genetics , Protein Structure, Secondary , RNA/chemistry , Sequence Homology, Nucleic Acid
17.
Protein Pept Lett ; 13(10): 1025-7, 2006.
Article in English | MEDLINE | ID: mdl-17168825

ABSTRACT

Nuclear translocation signal has been identified as a mediator of protein shuttling between nuclear and cytoplasm. Here we report that the combination of peptides from nuclear localization signal (NLS) and nuclear export signal (NES) of HIV-1 Rev have an antiviral activity against the Herpes virus of turkey and Marek's disease virus serotype 1.


Subject(s)
Antiviral Agents/pharmacology , Gene Products, rev/physiology , HIV-1/chemistry , Nuclear Export Signals/physiology , Nuclear Localization Signals/physiology , Amino Acid Sequence , Animals , Antiviral Agents/chemical synthesis , Chick Embryo , Gene Products, rev/chemistry , HIV-1/physiology , Herpesvirus 1, Meleagrid/growth & development , Herpesvirus 1, Meleagrid/metabolism , Herpesvirus 2, Gallid/growth & development , Herpesvirus 2, Gallid/metabolism , Humans , Molecular Sequence Data , rev Gene Products, Human Immunodeficiency Virus
18.
Nucleic Acids Res ; 27(21): 4128-34, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10518602

ABSTRACT

The human immunodeficiency virus type-1 Rev protein induces the nuclear export of intron-containing viral mRNAs that harbor its binding site, the Rev response element (RRE). A leucine-rich region of Rev, the activation domain, is essential for function and has been shown to be a nuclear export signal (NES). Although Rev exports viral RNAs that resemble cellular mRNAs, competition studies performed using microinjected Xenopus laevis oocytes have previously indicated that Rev utilizes a non-mRNA export pathway. Here, we show that Rev is able to induce the export of both spliceable and non-spliceable RRE-containing pre-mRNAs and that this activity is not dependent on the location of the RRE within the RNA. Importantly, even RNA molecules of different classes, such as U3 snoRNA and U6 snRNA, which are retained in the nucleus by non-pre-mRNA mechanisms, are exported to the cytoplasm in response to Rev. Consistent with the notion that Rev-mediated export of RRE-containing RNA is mechanistically distinct from the export of processed cellular mRNA, a chimeric Rev protein in which its NES is replaced by the NES of hnRNP A1 does not induce the export of a Rev-responsive mRNA. Finally, we demonstrate that Rev/RRE-activated RNA export is, like other nuclear export pathways, linked to the Ran-GTPase cycle.


Subject(s)
Cell Nucleus/metabolism , Gene Products, rev/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , RNA/metabolism , ran GTP-Binding Protein/metabolism , Animals , Biological Transport , COS Cells , Consensus Sequence/genetics , Exons/genetics , Gene Products, rev/chemistry , Gene Products, rev/genetics , Gene Products, tat/genetics , HIV-1/genetics , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoproteins , Introns/genetics , Mutation , Oocytes/metabolism , RNA/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/genetics , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , RNA-Binding Proteins/metabolism , Response Elements/genetics , Ribonucleoproteins/genetics , Xenopus laevis , ran GTP-Binding Protein/genetics , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
19.
Structure ; 5(1): 7-11, 1997 Jan 15.
Article in English | MEDLINE | ID: mdl-9016721

ABSTRACT

The structure of a complex between the HIV Revresponsive element (RRE) RNA and a fragment of the Rev protein has recently been determined by NMR. Together with previous studies of the Tat-TAR complex, these results show how RNA elements with considerable tertiary structure are able to play a more active role in directing binding to elements of protein secondary structure.


Subject(s)
Escherichia coli Proteins , Gene Products, rev/metabolism , Genes, env/genetics , HIV/chemistry , Receptors, Cell Surface , Bacterial Proteins/chemistry , Chemoreceptor Cells , Gene Products, rev/chemistry , Gene Products, tat/chemistry , Magnetic Resonance Spectroscopy , Membrane Proteins/chemistry , Models, Molecular , Nucleic Acid Conformation , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Structure, Secondary , RNA-Binding Proteins/metabolism , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
20.
Oncogene ; 16(14): 1851-61, 1998 Apr 09.
Article in English | MEDLINE | ID: mdl-9583682

ABSTRACT

A peptide signal, which may control nucleo-cytoplasmic protein trafficking, was newly identified in human immunodeficiency virus type I (HIV-1) Rev, a lentiviral post-transcriptional transactivator. The sequence, in the amino-terminal portion of HIV-1 Rev, maintains a Rev mutant with a dysfunctional nuclear/nucleolar targeting signal outside of the nucleus, although this Rev molecule itself is small enough to pass through the nuclear pores. Transition of this sequence to the N-terminus of human T-lymphocytic leukemia/lymphoma virus type I (HTLV-I) p21x, which is usually located evenly distributed throughout the cell, resulted in capture of p21x in the cytoplasm. Mutational analysis clarified that a 14 residue peptide sequence was sufficient to display this inhibitory effect against nuclear entry. Furthermore, this HIV-1 Rev sequence was capable of inhibiting nuclear entry of a fragment of a human ribosomal protein, when it was fused to the carboxy terminus. The identified nuclear entry inhibitory signal (NIS) contains a conserved hydrophilicity motif, which forms an amphipathic helix. Significantly, this motif and its helical structure were shown to be important for NIS function and the HIV-1 Rev function itself. Possible roles for NIS as a molecular anchor are proposed herein.


Subject(s)
Cell Nucleus/metabolism , Gene Products, rev/physiology , HIV-1/physiology , Protein Sorting Signals/physiology , Amino Acid Sequence , Animals , Biological Transport/genetics , COS Cells , Conserved Sequence , Cytoplasm/metabolism , Gene Products, rev/chemistry , Humans , Molecular Sequence Data , Nuclear Localization Signals/genetics , Point Mutation , Protein Sorting Signals/chemistry , Protein Structure, Secondary , Retroviridae Proteins, Oncogenic/chemistry , Retroviridae Proteins, Oncogenic/physiology , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Sequence Deletion , rev Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL