Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 115(40): 10040-10045, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30232260

ABSTRACT

The human T cell leukemia virus I basic leucine zipper protein (HTLV-1 HBZ) maintains chronic viral infection and promotes leukemogenesis through poorly understood mechanisms involving interactions with the KIX domain of the transcriptional coactivator CBP and its paralog p300. The KIX domain binds regulatory proteins at the distinct MLL and c-Myb/pKID sites to form binary or ternary complexes. The intrinsically disordered N-terminal activation domain of HBZ (HBZ AD) deregulates cellular signaling pathways by competing directly with cellular and viral transcription factors for binding to the MLL site and by allosterically perturbing binding of the transactivation domain of the hematopoietic transcription factor c-Myb. Crystal structures of the ternary KIX:c-Myb:HBZ complex show that the HBZ AD recruits two KIX:c-Myb entities through tandem amphipathic motifs (L/V)(V/L)DGLL and folds into a long α-helix upon binding. Isothermal titration calorimetry reveals strong cooperativity in binding of the c-Myb activation domain to the KIX:HBZ complex and in binding of HBZ to the KIX:c-Myb complex. In addition, binding of KIX to the two HBZ (V/L)DGLL motifs is cooperative; the structures suggest that this cooperativity is achieved through propagation of the HBZ α-helix beyond the first binding motif. Our study suggests that the unique structural flexibility and the multiple interaction motifs of the intrinsically disordered HBZ AD are responsible for its potency in hijacking KIX-mediated transcription pathways. The KIX:c-Myb:HBZ complex provides an example of cooperative stabilization in a transcription factor:coactivator network and gives insights into potential mechanisms through which HBZ dysregulates hematopoietic transcriptional programs and promotes T cell proliferation.


Subject(s)
Basic-Leucine Zipper Transcription Factors/chemistry , Human T-lymphotropic virus 1/chemistry , Proto-Oncogene Proteins c-myb/chemistry , Retroviridae Proteins/chemistry , Transcription, Genetic , Basic-Leucine Zipper Transcription Factors/metabolism , Human T-lymphotropic virus 1/metabolism , Humans , Protein Domains , Protein Structure, Quaternary , Protein Structure, Secondary , Proto-Oncogene Proteins c-myb/metabolism , Retroviridae Proteins/metabolism
2.
Protein Expr Purif ; 173: 105659, 2020 09.
Article in English | MEDLINE | ID: mdl-32360379

ABSTRACT

Human T-cell leukemia virus type 1 is an oncovirus that causes aggressive adult T-cell leukemia but is also responsible for severe neurodegenerative and endocrine disorders. Combatting HTLV-1 infections requires a detailed understanding of the viral mechanisms in the host. Therefore, in vitro studies of important virus-encoded proteins would be critical. Our focus herein is on the HTLV-1-encoded regulatory protein p13II, which interacts with the inner mitochondrial membrane, increasing its permeability to cations (predominantly potassium, K+). Thereby, this protein affects mitochondrial homeostasis. We report on our progress in developing specific protocols for heterologous expression of p13II in E. coli, and methods for its purification and characterization. We succeeded in producing large quantities of highly-pure full-length p13II, deemed to be its fully functional form. Importantly, our particular approach based on the fusion of ubiquitin to the p13II C-terminus was instrumental in increasing the persistently low expression of soluble p13II in its native form. We subsequently developed approaches for protein spin labeling and a conformation study using double electron-electron resonance (DEER) spectroscopy and a fluorescence-based cation uptake assay for p13II in liposomes. Our DEER results point to large protein conformation changes occurring upon transition from the soluble to the membrane-bound state. The functional assay on p13II-assisted transport of thallium (Tl+) through the membrane, wherein Tl+ substituted for K+, suggests transmembrane potential involvement in p13II function. Our study lays the foundation for expansion of in vitro functional and structural investigations on p13II and would aid in the development of structure-based protein inhibitors and markers.


Subject(s)
Escherichia coli , Human T-lymphotropic virus 1/genetics , Retroviridae Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Retroviridae Proteins/biosynthesis , Retroviridae Proteins/chemistry , Retroviridae Proteins/genetics , Retroviridae Proteins/isolation & purification
3.
Biophys J ; 113(9): 2004-2015, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29117524

ABSTRACT

Binding of the retroviral structural protein Gag to the cellular plasma membrane is mediated by the protein's matrix (MA) domain. Prominent among MA-PM interactions is electrostatic attraction between the positively charged MA domain and the negatively charged plasma membrane inner leaflet. Previously, we reported that membrane association of HIV-1 Gag, as well as purified Rous sarcoma virus (RSV) MA and Gag, depends strongly on the presence of acidic lipids and is enhanced by cholesterol (Chol). The mechanism underlying this enhancement was unclear. Here, using a broad set of in vitro and in silico techniques we addressed molecular mechanisms of association between RSV MA and model membranes, and investigated how Chol enhances this association. In neutron scattering experiments with liposomes in the presence or absence of Chol, MA preferentially interacted with preexisting POPS-rich clusters formed by nonideal lipid mixing, binding peripherally to the lipid headgroups with minimal perturbation to the bilayer structure. Molecular dynamics simulations showed a stronger MA-bilayer interaction in the presence of Chol, and a large Chol-driven increase in lipid packing and membrane surface charge density. Although in vitro MA-liposome association is influenced by disparate variables, including ionic strength and concentrations of Chol and charged lipids, continuum electrostatic theory revealed an underlying dependence on membrane surface potential. Together, these results conclusively show that Chol affects RSV MA-membrane association by making the electrostatic potential at the membrane surface more negative, while decreasing the penalty for lipid headgroup desolvation. The presented approach can be applied to other viral and nonviral proteins.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/metabolism , Cholesterol/metabolism , Retroviridae Proteins/chemistry , Retroviridae Proteins/metabolism , Solvents/chemistry , Static Electricity , Animals , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Molecular Dynamics Simulation , Phosphatidylcholines/metabolism , Phosphatidylserines/metabolism , Protein Binding , Protein Conformation , Protein Domains , Rous sarcoma virus
4.
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
5.
Biochem Biophys Res Commun ; 494(1-2): 57-62, 2017 12 09.
Article in English | MEDLINE | ID: mdl-29054407

ABSTRACT

Retroviral integrase catalyzes the integration of retroviral genome into host chromosomal DNA, which is a prerequisite of effective viral replication and infection. The human immunodeficiency virus type 1 (HIV-1) integrase has previously been reported to be regulated by the ubiquitination, but the molecular characterization of integrase ubiquitination is still unclear. In this study, we analyzed the ubiquitination of avian leukosis virus (ALV) integrase in detail. The ubiquitination assay showed that, like HIV-1, ALV integrase could also be modified by ubiquitination when expressed in 293 T and DF-1 cells. Domain mapping analysis revealed that the ubiquitination of ALV integrase might mainly occurred in the catalytic core and the N-terminal zinc-binding domains. Both lysine and non-lysine residues within integrase of ALV and HIV-1 were responsible for the ubiquitin conjugation, and the N-terminal HHCC zinc-binding motif might play an important role in mediating integrase ubiquitination. Interestingly, mass spectrometry analysis identified the Thr10 and Cys37 residues in the HHCC zinc-binding motif as the ubiquitination sites, indicating that ubiquitin may be conjugated to ALV integrase through direct interaction with the non-lysine residues. These findings revealed the detailed features of retroviral integrase ubiquitination and found a novel mechanism of ubiquitination mediated by the non-lysine residues within the N-terminal zinc-binding domain of integrase.


Subject(s)
Avian Leukosis Virus/enzymology , HIV Integrase/chemistry , HIV Integrase/metabolism , Integrases/chemistry , Integrases/metabolism , Retroviridae Proteins/chemistry , Retroviridae Proteins/metabolism , Retroviridae/enzymology , Amino Acid Motifs , Amino Acid Sequence , Animals , Avian Leukosis Virus/genetics , Avian Leukosis Virus/physiology , Cell Line , Chickens , HEK293 Cells , HIV Integrase/genetics , HIV-1/enzymology , HIV-1/genetics , HIV-1/physiology , Humans , Integrases/genetics , Lysine/chemistry , Mutagenesis, Site-Directed , Retroviridae/genetics , Retroviridae/physiology , Retroviridae Proteins/genetics , Ubiquitination , Zinc/metabolism
6.
Biochem Biophys Res Commun ; 477(4): 977-981, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27387229

ABSTRACT

SAMHD1 plays diverse roles in innate immunity, autoimmune diseases and HIV restriction, but the mechanisms involved are still unclear. SAMHD1 has been reported to have both dNTPase and RNase activities. However, whether SAMHD1 possesses RNase activity remains highly controversial. Here, we found that, unlike conventional hydrolytic exoribonucleases, SAMHD1 requires inorganic phosphate to degrade RNA substrates and produces nucleotide diphosphates rather than nucleoside monophosphates, which indicated that SAMHD1 is a phosphorolytic but not hydrolytic 3'-5' exoribonuclease. Furthermore, SAMHD1 preferentially cleaved single-stranded RNAs comprising A20 or U20, whereas neither C20 nor G20 was susceptible to SAMHD1-mediated degradation. Our findings will facilitate more advanced studies into the role of the SAMHD1 RNase function in the cellular pathogenesis implicated in nucleic acid-triggered inflammatory responses and the anti-retroviral function of SAMHD1.


Subject(s)
Autoimmune Diseases of the Nervous System/enzymology , Dinucleoside Phosphates/chemistry , Monomeric GTP-Binding Proteins/chemistry , Nervous System Malformations/enzymology , RNA/chemistry , Retroviridae Proteins/chemistry , Ribonucleases/chemistry , Binding Sites , Enzyme Activation , Humans , Hydrolysis , Phosphorylation , Protein Binding , SAM Domain and HD Domain-Containing Protein 1
7.
Postepy Biochem ; 62(3): 280-285, 2016.
Article in English | MEDLINE | ID: mdl-28132482

ABSTRACT

Collaborations between the Wlodawer and Skalka laboratories have covered a period of almost 30 years. During that time our groups have co-authored 18 publications, including several much cited journal articles, book chapters, and scholarly reviews. It has therefore been most rewarding for us to share enthusiasm, insights, and expertise with our Frederick colleagues over the years, and also to enjoy lasting friendships.


Subject(s)
Biochemistry/history , Crystallography/history , Retroviridae Proteins/chemistry , Retroviridae/enzymology , Crystallography/methods , History, 20th Century , History, 21st Century , Protein Conformation , Retroviridae Proteins/metabolism , United States
8.
Retrovirology ; 12: 59, 2015 Jul 04.
Article in English | MEDLINE | ID: mdl-26140924

ABSTRACT

BACKGROUND: Human T cell lymphotropic virus type 1 (HTLV-1) is the etiological agent of a severe form of neoplasia designated Adult T cell Leukaemia (ATL). It is widely accepted that the viral transactivator Tax-1 is the major viral product involved in the onset, but not in the maintenance, of neoplastic phenotype, as only 30-40% of ATL cells express Tax-1. It has been recently demonstrated that HBZ (HTLV-1 bZIP factor), a protein encoded by the minus strand of HTLV-1 genome, constantly expressed in infected cells and in ATL tumor cells, is also involved in the pathogenesis of leukaemia. The full role played by HBZ in oncogenesis is not clarified in detail also because of the limited availability of tools to assess quantitative expression, subcellular location and interaction of HBZ with host factors in ATL. RESULTS: By the use of the first reported monoclonal antibody against HBZ, 4D4-F3, generated in our laboratory it has been possible to carefully assess for the first time the above parameters in HTLV-1 chronically infected cells and, most importantly, in fresh leukemic cells from patients. Endogenous HBZ is expressed in speckle-like structures localized in the nucleus. The calculated number of endogenous HBZ molecules varies between 17.461 and 39.615 molecules per cell, 20- to 50-fold less than the amount expressed in HBZ transfected cells used by most investigators to assess the expression, function and subcellular localization of the viral protein. HBZ interacts in vivo with p300 and JunD and co-localizes only partially, and depending on the amount of expressed HBZ, not only with p300 and JunD but also with CBP and CREB2. CONCLUSIONS: The possibility to study endogenous HBZ in detail may significantly contribute to a better delineation of the role of HBZ during HTLV-1 infection and cellular transformation.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Human T-lymphotropic virus 1/physiology , Leukemia-Lymphoma, Adult T-Cell/virology , Retroviridae Proteins/metabolism , Adult , Animals , Antibodies, Monoclonal , Basic-Leucine Zipper Transcription Factors/chemistry , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , COS Cells , Chlorocebus aethiops , Epitope Mapping , Gene Products, tax/metabolism , HEK293 Cells , Humans , Leukemia-Lymphoma, Adult T-Cell/pathology , Mice , Recombinant Proteins , Retroviridae Proteins/chemistry , Retroviridae Proteins/genetics , Retroviridae Proteins/immunology
9.
Acc Chem Res ; 46(9): 2047-58, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23402263

ABSTRACT

In living organisms, biological molecules often organize into multicomponent complexes. Such assemblies consist of various proteins and carry out essential functions, ranging from cell division, transport, and energy transduction to catalysis, signaling, and viral infectivity. To understand the biological functions of these assemblies, in both healthy and disease states, researchers need to study their three-dimensional architecture and molecular dynamics. To date, the large size, the lack of inherent long-range order, and insolubility have made atomic resolution studies of many protein assemblies challenging or impractical using traditional structural biology methods such as X-ray diffraction and solution NMR spectroscopy. In the past 10 years, we have focused our work on the development and application of magic angle spinning solid-state NMR (MAS NMR) methods to characterize large protein assemblies at atomic-level resolution. In this Account, we discuss the rapid progress in the field of MAS NMR spectroscopy, citing work from our laboratory and others on methodological developments that have facilitated the in-depth analysis of biologically important protein assemblies. We emphasize techniques that yield enhanced sensitivity and resolution, such as fast MAS (spinning frequencies of 40 kHz and above) and nonuniform sampling protocols for data acquisition and processing. We also discuss the experiments for gaining distance restraints and for recoupling anisotropic tensorial interactions under fast MAS conditions. We give an overview of sample preparation approaches when working with protein assemblies. Following the overview of contemporary MAS NMR methods, we present case studies into the structure and dynamics of two classes of biological systems under investigation in our laboratory. We will first turn our attention to cytoskeletal microtubule motor proteins including mammalian dynactin and dynein light chain 8. We will then discuss protein assemblies from the HIV-1 retrovirus.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , Proteins/chemistry , Proteins/metabolism , Animals , Cytoskeleton/chemistry , Cytoskeleton/metabolism , HIV-1/chemistry , HIV-1/metabolism , Microscopy, Electron, Transmission , Reproducibility of Results , Retroviridae Proteins/chemistry , Retroviridae Proteins/metabolism
10.
PLoS Pathog ; 7(2): e1001268, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21304939

ABSTRACT

Refolding of viral class-1 membrane fusion proteins from a native state to a trimer-of-hairpins structure promotes entry of viruses into cells. Here we present the structure of the bovine leukaemia virus transmembrane glycoprotein (TM) and identify a group of asparagine residues at the membrane-distal end of the trimer-of-hairpins that is strikingly conserved among divergent viruses. These asparagines are not essential for surface display of pre-fusogenic envelope. Instead, substitution of these residues dramatically disrupts membrane fusion. Our data indicate that, through electrostatic interactions with a chloride ion, the asparagine residues promote assembly and profoundly stabilize the fusion-active structures that are required for viral envelope-mediated membrane fusion. Moreover, the BLV TM structure also reveals a charge-surrounded hydrophobic pocket on the central coiled coil and interactions with basic residues that cluster around this pocket are critical to membrane fusion and form a target for peptide inhibitors of envelope function. Charge-surrounded pockets and electrostatic interactions with small ions are common among class-1 fusion proteins, suggesting that small molecules that specifically target such motifs should prevent assembly of the trimer-of-hairpins and be of value as therapeutic inhibitors of viral entry.


Subject(s)
Ions/metabolism , Protein Folding , Retroviridae Proteins/chemistry , Retroviridae Proteins/physiology , Static Electricity , Amino Acid Sequence , Animals , Anti-Retroviral Agents/chemistry , Anti-Retroviral Agents/pharmacology , Catalytic Domain/drug effects , Cattle , Human T-lymphotropic virus 1/chemistry , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/metabolism , Humans , Hydrogen Bonding , Ions/chemistry , Leukemia Virus, Bovine/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology , Retroviridae/metabolism , Retroviridae/physiology , Retroviridae Proteins/metabolism , Surface Properties , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
11.
Protein Expr Purif ; 92(1): 94-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24056256

ABSTRACT

N-terminal myristoylation of retroviral matrix proteins is essential for the targeting of the Gag polyproteins to the plasma membrane. To investigate the effect of the myristoylation on the structure and membrane binding ability of the matrix proteins, it is necessary to prepare their myristoylated forms. We present purification of myristoylated matrix proteins of the mouse mammary tumor virus and murine leukemia virus, two morphogenetically distinct retroviruses. The proteins were expressed in Escherichia coli coexpressing a yeast N-myristoyltransferase. This E. coli expression system yielded a mixture of myristoylated and nonmyristoylated matrix proteins. We established efficient one-step metal affinity purification that enabled to obtain pure myristoylated matrix proteins suitable for structural and functional studies.


Subject(s)
Leukemia Virus, Murine/metabolism , Myristic Acid/metabolism , Retroviridae Proteins/isolation & purification , Retroviridae Proteins/metabolism , Animals , Chromatography, Affinity , Cloning, Molecular , Leukemia Virus, Murine/chemistry , Leukemia Virus, Murine/genetics , Mice , Myristic Acid/chemistry , Nuclear Magnetic Resonance, Biomolecular , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Retroviridae Infections/virology , Retroviridae Proteins/chemistry , Retroviridae Proteins/genetics
12.
Cell Mol Life Sci ; 69(8): 1211-59, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22033837

ABSTRACT

Many proteins and protein regions are disordered in their native, biologically active states. These proteins/regions are abundant in different organisms and carry out important biological functions that complement the functional repertoire of ordered proteins. Viruses, with their highly compact genomes, small proteomes, and high adaptability for fast change in their biological and physical environment utilize many of the advantages of intrinsic disorder. In fact, viral proteins are generally rich in intrinsic disorder, and intrinsically disordered regions are commonly used by viruses to invade the host organisms, to hijack various host systems, and to help viruses in accommodation to their hostile habitats and to manage their economic usage of genetic material. In this review, we focus on the structural peculiarities of HIV-1 proteins, on the abundance of intrinsic disorder in viral proteins, and on the role of intrinsic disorder in their functions.


Subject(s)
HIV-1/chemistry , Retroviridae Proteins/chemistry , HIV-1/enzymology , HIV-1/metabolism , Models, Molecular , Protein Conformation , Retroviridae Proteins/metabolism
13.
J Med Ethics ; 39(2): 115-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23204319

ABSTRACT

The recent success of Foldit in determining the structure of the Mason-Pfizer monkey virus (M-PMV) retroviral protease is suggestive of the power-solving potential of internet-facilitated game-like crowdsourcing. This research model is highly novel, however, and thus, deserves careful consideration of potential ethical issues. In this paper, we will demonstrate that the crowdsourcing model of research has the potential to cause harm to participants, manipulates the participant into continued participation, and uses participants as experimental subjects. We conclude that protocols relying on this model require institutional review board (IRB) scrutiny.


Subject(s)
Crowdsourcing/ethics , Ethics Committees, Research , Ethics, Research , Games, Experimental , Informed Consent , Internet , Research Design , Research Personnel , Research Subjects , Behavior, Addictive/etiology , Crowdsourcing/methods , Humans , Mason-Pfizer monkey virus/enzymology , Peptide Hydrolases/chemistry , Protein Folding , Research Personnel/psychology , Research Subjects/psychology , Retroviridae Proteins/chemistry
14.
Nucleic Acids Res ; 39(Database issue): D70-4, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21036865

ABSTRACT

This article introduces the second release of the Gypsy Database of Mobile Genetic Elements (GyDB 2.0): a research project devoted to the evolutionary dynamics of viruses and transposable elements based on their phylogenetic classification (per lineage and protein domain). The Gypsy Database (GyDB) is a long-term project that is continuously progressing, and that owing to the high molecular diversity of mobile elements requires to be completed in several stages. GyDB 2.0 has been powered with a wiki to allow other researchers participate in the project. The current database stage and scope are long terminal repeats (LTR) retroelements and relatives. GyDB 2.0 is an update based on the analysis of Ty3/Gypsy, Retroviridae, Ty1/Copia and Bel/Pao LTR retroelements and the Caulimoviridae pararetroviruses of plants. Among other features, in terms of the aforementioned topics, this update adds: (i) a variety of descriptions and reviews distributed in multiple web pages; (ii) protein-based phylogenies, where phylogenetic levels are assigned to distinct classified elements; (iii) a collection of multiple alignments, lineage-specific hidden Markov models and consensus sequences, called GyDB collection; (iv) updated RefSeq databases and BLAST and HMM servers to facilitate sequence characterization of new LTR retroelement and caulimovirus queries; and (v) a bibliographic server. GyDB 2.0 is available at http://gydb.org.


Subject(s)
Databases, Genetic , Retroelements , Retroviridae/genetics , Terminal Repeat Sequences , Caulimoviridae/classification , Caulimoviridae/genetics , Phylogeny , Retroviridae/classification , Retroviridae Proteins/chemistry , Retroviridae Proteins/classification , Retroviridae Proteins/genetics , Software
15.
Biochem J ; 439(3): 505-16, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21732913

ABSTRACT

The association of the SH3 (Src homology 3) domain of SFKs (Src family kinases) with protein partners bearing proline-rich motifs has been implicated in the regulation of SFK activity, and has been described as a possible mechanism of relocalization of SFKs to subcellular compartments. We demonstrate in the present study for the first time that p13, an accessory protein encoded by the HTLV-1 (human T-cell leukaemia virus type 1), binds the SH3 domain of SFKs via its C-terminal proline-rich motif, forming a stable heterodimer that translocates to mitochondria by virtue of its N-terminal mitochondrial localization signal. As a result, the activity of SFKs is dramatically enhanced, with a subsequent increase in mitochondrial tyrosine phosphorylation, and the recognized ability of p13 to insert itself into the inner mitochondrial membrane and to perturb the mitochondrial membrane potential is abolished. Overall, the present study, in addition to confirming that the catalytic activity of SFKs is modulated by interactors of their SH3 domain, leads us to hypothesize a general mechanism by which proteins bearing a proline-rich motif and a mitochondrial localization signal at the same time may act as carriers of SFKs into mitochondria, thus contributing to the regulation of mitochondrial functions under various pathophysiological conditions.


Subject(s)
Human T-lymphotropic virus 1/chemistry , Mitochondrial Proteins/chemistry , Proline-Rich Protein Domains , Retroviridae Proteins/chemistry , src Homology Domains , src-Family Kinases/chemistry , Amino Acid Motifs , Animals , HeLa Cells , Human T-lymphotropic virus 1/genetics , Humans , Mitochondrial Proteins/genetics , Protein Binding , Protein Multimerization/genetics , Protein Transport/genetics , Rabbits , Rats , Retroviridae Proteins/genetics , src-Family Kinases/genetics
16.
Viruses ; 14(2)2022 01 29.
Article in English | MEDLINE | ID: mdl-35215882

ABSTRACT

Stephen Oroszlan received his early education in Hungary, graduating in 1950 from the Technical University in Budapest with a degree in chemical engineering [...].


Subject(s)
Retroviridae Proteins/chemistry , Retroviridae Proteins/metabolism , History, 20th Century , History, 21st Century , Humans , Male , Retroviridae/drug effects , Retroviridae/metabolism , Viral Protease Inhibitors/pharmacology , Viral Proteases/chemistry , Viral Proteases/metabolism
17.
Biochim Biophys Acta ; 1797(6-7): 945-51, 2010.
Article in English | MEDLINE | ID: mdl-20188695

ABSTRACT

Human T cell leukemia virus type 1 (HTLV-1) encodes p13, an 87-amino-acid protein that accumulates in the inner mitochondrial membrane. Recent studies performed using synthetic p13 and isolated mitochondria demonstrated that the protein triggers an inward potassium (K+) current and inner membrane depolarization. The present study investigated the effects of p13 on mitochondrial inner membrane potential (Deltapsi) in living cells. Using the potential-dependent probe tetramethyl rhodamine methyl ester (TMRM), we observed that p13 induced dose-dependent mitochondrial depolarization in HeLa cells. This effect was abolished upon mutation of 4 arginines in p13's alpha-helical domain that were previously shown to be essential for its activity in in vitro assays. As Deltapsi is known to control mitochondrial calcium (Ca2+) uptake, we next analyzed the effect of p13 on Ca2+ homeostasis. Experiments carried out in HeLa cells expressing p13 and organelle-targeted aequorins revealed that the protein specifically reduced mitochondrial Ca2+ uptake. These observations suggest that p13 might control key processes regulated through Ca2+ signaling such as activation and death of T cells, the major targets of HTLV-1 infection.


Subject(s)
Calcium/metabolism , Human T-lymphotropic virus 1/metabolism , Membrane Potential, Mitochondrial , Retroviridae Proteins/metabolism , Calcium Signaling , HeLa Cells , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/pathogenicity , Humans , Ion Transport , Models, Biological , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retroviridae Proteins/chemistry , Retroviridae Proteins/genetics , Transfection
18.
EMBO Rep ; 10(2): 144-51, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19165139

ABSTRACT

The retroviral integrase superfamily (RISF) comprises numerous important nucleic acid-processing enzymes, including transposases, integrases and various nucleases. These enzymes are involved in a wide range of processes such as transposition, replication and repair of DNA, homologous recombination, and RNA-mediated gene silencing. Two out of the four enzymes that are encoded by the human immunodeficiency virus--RNase H1 and integrase--are members of this superfamily. RISF enzymes act on various substrates, and yet show remarkable mechanistic and structural similarities. All share a common fold of the catalytic core and the active site, which is composed primarily of carboxylate residues. Here, I present RISF proteins from a structural perspective, describing the individual members and the common and divergent elements of their structures, as well as the mechanistic insights gained from the structures of RNase H1 enzyme complexes with RNA/DNA hybrids.


Subject(s)
Integrases/chemistry , Multigene Family , Retroviridae Proteins/chemistry , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/physiology , Catalytic Domain , Dimerization , Hydrolysis , Integrases/physiology , Mammals/metabolism , Mice , Models, Molecular , Nucleic Acids/metabolism , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Retroviridae Proteins/physiology , Ribonuclease H/chemistry , Ribonuclease H/physiology , Species Specificity , Structure-Activity Relationship , Substrate Specificity , Transposases/chemistry , Transposases/physiology , Viral Proteins/chemistry , Viral Proteins/physiology
19.
Biochem J ; 432(3): 525-34, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20929444

ABSTRACT

The adaptor protein ALIX [ALG-2 (apoptosis-linked-gene-2 product)-interacting protein X] links retroviruses to ESCRT (endosomal sorting complex required for transport) machinery during retroviral budding. This function of ALIX requires its interaction with the ESCRT-III component CHMP4 (charged multivesicular body protein 4) at the N-terminal Bro1 domain and retroviral Gag proteins at the middle V domain. Since cytoplasmic or recombinant ALIX is unable to interact with CHMP4 or retroviral Gag proteins under non-denaturing conditions, we constructed ALIX truncations and mutations to define the intrinsic mechanism through which ALIX interactions with these partner proteins are prohibited. Our results demonstrate that an intramolecular interaction between Patch 2 in the Bro1 domain and the TSG101 (tumour susceptibility gene 101 protein)-docking site in the proline-rich domain locks ALIX into a closed conformation that renders ALIX unable to interact with CHMP4 and retroviral Gag proteins. Relieving the intramolecular interaction of ALIX, by ectopically expressing a binding partner for one of the intramolecular interaction sites or by deleting one of these sites, promotes ALIX interaction with these partner proteins and facilitates ALIX association with the membrane. Ectopic expression of a GFP (green fluorescent protein)-ALIX mutant with a constitutively open conformation, but not the wild-type protein, increases EIAV (equine infectious anaemia virus) budding from HEK (human embryonic kidney)-293 cells. These findings predict that relieving the autoinhibitory intramolecular interaction of ALIX is a critical step for ALIX to participate in retroviral budding.


Subject(s)
Calcium-Binding Proteins/physiology , Cell Cycle Proteins/physiology , Endosomal Sorting Complexes Required for Transport/metabolism , Retroviridae Proteins/metabolism , Virus Release/physiology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Membrane/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/physiology , Gene Products, gag/chemistry , Gene Products, gag/genetics , Gene Products, gag/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Infectious Anemia Virus, Equine/physiology , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Oncogene Protein pp60(v-src)/chemistry , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Proline-Rich Protein Domains/physiology , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Retroviridae Proteins/chemistry , Retroviridae Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Vesicular Transport Proteins/metabolism
20.
Viruses ; 13(8)2021 07 23.
Article in English | MEDLINE | ID: mdl-34452297

ABSTRACT

Antisense protein of Human T-cell Leukemia Virus Type 2 (HTLV-2), also called APH-2, negatively regulates the HTLV-2 and helps the virus to maintain latency via scheming the transcription. Despite the remarkable occurrence of HTLV-2/HIV-1 co-infection, the role of APH-2 influencing HIV-1 replication kinetics is poorly understood and needs investigation. In this study, we investigated the plausible role of APH-2 regulating HIV-1 replication. Herein, we report that the overexpression of APH-2 not only hampered the release of HIV-1 pNL4.3 from 293T cells in a dose-dependent manner but also affected the cellular gag expression. A similar and consistent effect of APH-2 overexpression was also observed in case of HIV-1 gag expression vector HXB2 pGag-EGFP. APH-2 overexpression also inhibited the ability of HIV-1 Tat to transactivate the HIV-1 LTR-driven expression of luciferase. Furthermore, the introduction of mutations in the IXXLL motif at the N-terminal domain of APH-2 reverted the inhibitory effect on HIV-1 Tat-mediated transcription, suggesting the possible role of this motif towards the downregulation of Tat-mediated transactivation. Overall, these findings indicate that the HTLV-2 APH-2 may affect the HIV-1 replication at multiple levels by (a) inhibiting the Tat-mediated transactivation and (b) hampering the virus release by affecting the cellular gag expression.


Subject(s)
HIV-1/physiology , Human T-lymphotropic virus 2/genetics , Retroviridae Proteins/metabolism , Virus Replication , Cell Line , Gene Expression Regulation, Viral , HEK293 Cells , HIV-1/genetics , Humans , Retroviridae Proteins/chemistry , Retroviridae Proteins/genetics , Transcriptional Activation , Virus Release , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL