Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
bioRxiv ; 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38617369

ABSTRACT

The Rep68 protein from Adeno-Associated Virus (AAV) is a multifunctional SF3 helicase that performs most of the DNA transactions required for the viral life cycle. During AAV DNA replication, Rep68 assembles at the origin and catalyzes the DNA melting and nicking reactions during the hairpin rolling replication process to complete the second-strand synthesis of the AAV genome. Here, we report the Cryo-EM structures of Rep68 bound to double-stranded DNA (dsDNA) containing the sequence of the AAVS1 integration site in different nucleotide-bound states. In the apo state, Rep68 forms a heptameric complex around DNA, with three Origin Binding Domains (OBDs) bound to the Rep Binding Site (RBS) sequence and three other OBDs forming transient dimers with them. The AAA+ domains form an open ring with no interactions between subunits and with DNA. We hypothesize the heptameric quaternary structure is necessary to load onto dsDNA. In the ATPγS-bound state, a subset of three subunits binds the nucleotide, undergoing a large conformational change, inducing the formation of intersubunit interactions interaction and interaction with three consecutive DNA phosphate groups. Moreover, the induced conformational change positions three phenylalanine residues to come in close contact with the DNA backbone, producing a distortion in the DNA. We propose that the phenylalanine residues can potentially act as a hydrophobic wedge in the DNA melting process.

2.
STAR Protoc ; 3(3): 101615, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36035794

ABSTRACT

eIF5-mimic protein (5MP) controls translation through its interaction with eukaryotic translation initiation factor (eIF) 2 and eIF3 and alters non-AUG translation rates for oncogenes in cancer and repeat expansions in neurodegenerative disease. To precisely evaluate the effect of 5MP mutations on binding affinity against eIFs, here we describe two label-free protocols of affinity measurement for 5MP binding to eIF2 or eIF3 protein segments, termed isothermal titration calorimetry (ITC) and bio-layer interferometry (BLI), starting with how to purify proteins used. For complete details on the use and execution of this protocol, please refer to Singh et al. (2021).


Subject(s)
Eukaryotic Initiation Factor-5 , Neurodegenerative Diseases , Calorimetry , Eukaryotic Initiation Factor-2 , Eukaryotic Initiation Factor-3 , Humans , Interferometry
3.
J Virol ; 96(2): e0132621, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34669461

ABSTRACT

Parvovirus B19 (B19V) infection can cause hematological disorders and fetal hydrops during pregnancy. Currently, no antivirals or vaccines are available for the treatment or prevention of B19V infection. To identify novel small-molecule antivirals against B19V replication, we developed a high-throughput screening (HTS) assay, which is based on an in vitro nicking assay using recombinant N-terminal amino acids 1 to 176 of the viral large nonstructural protein (NS1N) and a fluorescently labeled DNA probe (OriQ) that spans the nicking site of the viral DNA replication origin. We collectively screened 17,040 compounds and identified 2,178 (12.78%) hits that possess >10% inhibition of the NS1 nicking activity, among which 84 hits were confirmed to inhibit nicking in a dose-dependent manner. Using ex vivo-expanded primary human erythroid progenitor cells (EPCs) infected by B19V, we validated 24 compounds that demonstrated >50% in vivo inhibition of B19V infection at 10 µM, which can be categorized into 7 structure scaffolds. Based on the therapeutic index (half-maximal cytotoxic concentration [CC50]/half-maximal effective concentration [EC50] ratio) in EPCs, the top 4 compounds were chosen to examine their inhibitions of B19V infection in EPCs at two times of the 90% maximal effective concentration (EC90). A purine derivative (P7) demonstrated an antiviral effect (EC50 = 1.46 µM) without prominent cytotoxicity (CC50 = 71.8 µM) in EPCs and exhibited 92% inhibition of B19V infection in EPCs at 3.32 µM, which can be used as the lead compound in future studies for the treatment of B19V infection-caused hematological disorders. IMPORTANCE B19V encodes a large nonstructural protein, NS1. Its N-terminal domain (NS1N) consisting of amino acids 1 to 176 binds to viral DNA and serves as an endonuclease to nick the viral DNA replication origins, which is a pivotal step in rolling-hairpin-dependent B19V DNA replication. For high-throughput screening (HTS) of anti-B19V antivirals, we miniaturized a fluorescence-based in vitro nicking assay, which employs a fluorophore-labeled probe spanning the terminal resolution site (trs) and the NS1N protein, into a 384-well-plate format. The HTS assay showed high reliability and capability in screening 17,040 compounds. Based on the therapeutic index (half-maximal cytotoxic concentration [CC50]/half-maximal effective concentration [EC50] ratio) in EPCs, a purine derivative demonstrated an antiviral effect of 92% inhibition of B19V infection in EPCs at 3.32 µM (two times the EC90). Our study demonstrated a robust HTS assay for screening antivirals against B19V infection.


Subject(s)
Antiviral Agents/pharmacology , Erythroid Precursor Cells/virology , High-Throughput Screening Assays/methods , Parvovirus B19, Human/drug effects , Antiviral Agents/chemistry , Cell Survival/drug effects , DNA Replication/drug effects , DNA, Viral/metabolism , Erythroid Precursor Cells/drug effects , Fluorescent Dyes , Humans , Parvovirus B19, Human/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Origin , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
4.
Cell Rep ; 36(2): 109376, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260931

ABSTRACT

eIF5-mimic protein (5MP) is a translational regulatory protein that binds the small ribosomal subunit and modulates its activity. 5MP is proposed to reprogram non-AUG translation rates for oncogenes in cancer, but its role in controlling non-AUG initiated synthesis of deleterious repeat-peptide products, such as FMRpolyG observed in fragile-X-associated tremor ataxia syndrome (FXTAS), is unknown. Here, we show that 5MP can suppress both general and repeat-associated non-AUG (RAN) translation by a common mechanism in a manner dependent on its interaction with eIF3. Essentially, 5MP displaces eIF5 through the eIF3c subunit within the preinitiation complex (PIC), thereby increasing the accuracy of initiation. In Drosophila, 5MP/Kra represses neuronal toxicity and enhances the lifespan in an FXTAS disease model. These results implicate 5MP in protecting cells from unwanted byproducts of non-AUG translation in neurodegeneration.


Subject(s)
Codon, Initiator/genetics , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-3/metabolism , Protein Biosynthesis/genetics , Trinucleotide Repeat Expansion/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, Differentiation/metabolism , DNA-Binding Proteins/chemistry , Drosophila/metabolism , Drosophila Proteins/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-3/chemistry , HEK293 Cells , Humans , Male , Models, Biological , Models, Molecular , Mutation/genetics , Peptide Chain Initiation, Translational , Protein Binding , Protein Domains , Receptors, Immunologic/metabolism
5.
Nucleic Acids Res ; 48(22): 12983-12999, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33270897

ABSTRACT

The adeno-associated virus (AAV) non-structural Rep proteins catalyze all the DNA transactions required for virus viability including, DNA replication, transcription regulation, genome packaging, and during the latent phase, site-specific integration. Rep proteins contain two multifunctional domains: an Origin Binding Domain (OBD) and a SF3 helicase domain (HD). Studies have shown that Rep proteins have a dynamic oligomeric behavior where the nature of the DNA substrate molecule modulates its oligomeric state. In the presence of ssDNA, Rep68 forms a large double-octameric ring complex. To understand the mechanisms underlying AAV Rep function, we investigated the cryo-EM and X-ray structures of Rep68-ssDNA complexes. Surprisingly, Rep68 generates hybrid ring structures where the OBD forms octameric rings while the HD forms heptamers. Moreover, the binding to ATPγS promotes a large conformational change in the entire AAA+ domain that leads the HD to form both heptamer and hexamers. The HD oligomerization is driven by an interdomain linker region that acts as a latch to 'catch' the neighboring HD subunit and is flexible enough to permit the formation of different stoichiometric ring structures. Overall, our studies show the structural basis of AAV Rep's structural flexibility required to fulfill its multifunctional role during the AAV life cycle.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , DNA, Single-Stranded/genetics , DNA-Binding Proteins/genetics , Dependovirus/genetics , Viral Proteins/genetics , Adenosine Triphosphate/genetics , Cryoelectron Microscopy , DNA Helicases/genetics , DNA Helicases/ultrastructure , DNA, Single-Stranded/ultrastructure , DNA-Binding Proteins/ultrastructure , Dependovirus/ultrastructure , Humans , Viral Proteins/ultrastructure
6.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31816635

ABSTRACT

Type I interferon (IFN-I) provides effective antiviral immunity but can exacerbate harmful inflammatory reactions and cause hematopoietic stem cell (HSC) exhaustion; therefore, IFN-I expression must be tightly controlled. While signaling mechanisms that limit IFN-I induction and function have been extensively studied, less is known about transcriptional repressors acting directly on IFN-I regulatory regions. We show that NFAT5, an activator of macrophage pro-inflammatory responses, represses Toll-like receptor 3 and virus-induced expression of IFN-I in macrophages and dendritic cells. Mice lacking NFAT5 exhibit increased IFN-I production and better control of viral burden upon LCMV infection but show exacerbated HSC activation under systemic poly(I:C)-induced inflammation. We identify IFNß as a primary target repressed by NFAT5, which opposes the master IFN-I inducer IRF3 by binding to an evolutionarily conserved sequence in the IFNB1 enhanceosome that overlaps a key IRF site. These findings illustrate how IFN-I responses are balanced by simultaneously opposing transcription factors.


Subject(s)
Interferon Type I/immunology , Transcription Factors/immunology , Animals , Dendritic Cells/immunology , Female , Inflammation/immunology , Interferon Regulatory Factor-3/immunology , Interferon-gamma/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Transcription, Genetic/immunology
7.
Acta Crystallogr D Struct Biol ; 75(Pt 4): 437-450, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30988260

ABSTRACT

Although the HcpR regulator plays a vital step in initiation of the nitrosative stress response in many Gram-negative anaerobic bacteria, the molecular mechanisms that it uses to mediate gas sensing are not well understood. Here, a 2.6 Šresolution crystal structure of the N-terminal sensing domain of the anaerobic periodontopathogen Porphyromonas gingivalis HcpR is presented. The protein has classical features of the regulators belonging to the FNR-CRP family and contains a hydrophobic pocket in its N-terminal sensing domain. It is shown that heme bound to HcpR exhibits heme iron as a hexacoordinate system in the absence of nitric oxide (NO) and that upon nitrosylation it transitions to a pentacoordinate system. Finally, small-angle X-ray scattering experiments on full-length HcpR reveal that the C-terminal DNA-binding domain of HcpR has a high degree of interdomain flexibility.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Heme/metabolism , Nitric Oxide/metabolism , Nitrosative Stress , Porphyromonas gingivalis/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray/methods , Models, Molecular , Molecular Structure , Porphyromonas gingivalis/physiology , Protein Conformation , Sequence Homology
8.
PLoS One ; 13(7): e0200841, 2018.
Article in English | MEDLINE | ID: mdl-30016371

ABSTRACT

Endonucleases of the HUH family are specialized in processing single-stranded DNA in a variety of evolutionarily highly conserved biological processes related to mobile genetic elements. They share a structurally defined catalytic domain for site-specific nicking and strand-transfer reactions, which is often linked to the activities of additional functional domains, contributing to their overall versatility. To assess if these HUH domains could be interchanged, we created a chimeric protein from two distantly related HUH endonucleases, containing the N-terminal HUH domain of the bacterial conjugative relaxase TrwC and the C-terminal DNA helicase domain of the human adeno-associated virus (AAV) replicase and site-specific integrase. The purified chimeric protein retained oligomerization properties and DNA helicase activities similar to Rep68, while its DNA binding specificity and cleaving-joining activity at oriT was similar to TrwC. Interestingly, the chimeric protein could catalyse site-specific integration in bacteria with an efficiency comparable to that of TrwC, while the HUH domain of TrwC alone was unable to catalyze this reaction, implying that the Rep68 C-terminal helicase domain is complementing the TrwC HUH domain to achieve site-specific integration into TrwC targets in bacteria. Our results illustrate how HUH domains could have acquired through evolution other domains in order to attain new roles, contributing to the functional flexibility observed in this protein superfamily.


Subject(s)
DNA Nucleotidyltransferases/chemistry , Dependovirus/enzymology , Integrases/chemistry , Computational Biology , Conjugation, Genetic , DNA/chemistry , DNA Helicases/chemistry , DNA, Bacterial/genetics , DNA, Single-Stranded , Endonucleases/chemistry , Escherichia coli/metabolism , HEK293 Cells , Humans , Plasmids , Protein Domains , Recombinant Proteins/chemistry , Ultracentrifugation
9.
Nat Neurosci ; 20(9): 1247-1259, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28783139

ABSTRACT

Antipsychotic drugs remain the standard for schizophrenia treatment. Despite their effectiveness in treating hallucinations and delusions, prolonged exposure to antipsychotic medications leads to cognitive deficits in both schizophrenia patients and animal models. The molecular mechanisms underlying these negative effects on cognition remain to be elucidated. Here we demonstrate that chronic antipsychotic drug exposure increases nuclear translocation of NF-κB in both mouse and human frontal cortex, a trafficking event triggered via 5-HT2A-receptor-dependent downregulation of the NF-κB repressor IκBα. This upregulation of NF-κB activity led to its increased binding at the Hdac2 promoter, thereby augmenting Hdac2 transcription. Deletion of HDAC2 in forebrain pyramidal neurons prevented the negative effects of antipsychotic treatment on synaptic remodeling and cognition. Conversely, virally mediated activation of NF-κB signaling decreased cortical synaptic plasticity via HDAC2. Together, these observations may aid in developing therapeutic strategies to improve the outcome of schizophrenia treatment.


Subject(s)
Antipsychotic Agents/adverse effects , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Histone Deacetylase 2/metabolism , NF-kappa B/metabolism , Synapses/metabolism , Animals , Antipsychotic Agents/toxicity , Cognition Disorders/genetics , Frontal Lobe/drug effects , Frontal Lobe/metabolism , HEK293 Cells , Histone Deacetylase 2/deficiency , Histone Deacetylase 2/genetics , Humans , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , Synapses/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/physiology
10.
Bio Protoc ; 7(6)2017 03 20.
Article in English | MEDLINE | ID: mdl-28612042

ABSTRACT

Quantitative measurement of proteins binding to DNA is a requisite to fully characterize the structural determinants of complex formation necessary to understand the DNA transactions that regulate cellular processes. Here we describe a detailed protocol to measure binding affinity of the adeno-associated virus (AAV) Rep68 protein for the integration site AAVS1 using fluorescent anisotropy. This protocol can be used to measure the binding constants of any DNA binding protein provided the substrate DNA is fluorescently labeled.

11.
Bio Protoc ; 7(9)2017 May 05.
Article in English | MEDLINE | ID: mdl-28660237

ABSTRACT

Adeno-associated virus (AAV) is a small single-stranded DNA virus that requires the presence of a helper virus, such as adenovirus or herpes virus, to efficiently replicate its genome. AAV DNA is replicated by a rolling-hairpin mechanism (Ward, 2006), and during replication several DNA intermediates can be detected. This detailed protocol describes how to analyze the AAV DNA intermediates formed during AAV replication using a modified Hirt extract (Hirt, 1967) procedure and Southern blotting (Southern, 1975).

12.
J Virol ; 90(15): 6612-6624, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27170758

ABSTRACT

UNLABELLED: The life cycle of the human parvovirus adeno-associated virus (AAV) is orchestrated by four Rep proteins. The large Rep proteins, Rep78 and Rep68, are remarkably multifunctional and display a range of biochemical activities, including DNA binding, nicking, and unwinding. Functionally, Rep78 and Rep68 are involved in transcriptional regulation, DNA replication, and genomic integration. Structurally, the Rep proteins share an AAA(+) domain characteristic of superfamily 3 helicases, with the large Rep proteins additionally containing an N-terminal origin-binding domain (OBD) that specifically binds and nicks DNA. The combination of these domains, coupled with dynamic oligomerization properties, is the basis for the remarkable multifunctionality displayed by Rep68 and Rep78 during the AAV life cycle. In this report, we describe an oligomeric interface formed by Rep68 and demonstrate how disruption of this interface has drastic effects on both the oligomerization and functionality of the Rep proteins. Our results support a role for the four-helix bundle in the helicase domain of Rep68 as a bona fide oligomerization domain (OD). We have identified key residues in the OD that are critical for the stabilization of the Rep68-Rep68 interface; mutation of these key residues disrupts the enzymatic activities of Rep68, including DNA binding and nicking, and compromises viral DNA replication and transcriptional regulation of the viral promoters. Taken together, our data contribute to our understanding of the dynamic and substrate-responsive Rep78/68 oligomerization that is instrumental in the regulation of the DNA transitions that take place during the AAV life cycle. IMPORTANCE: The limited genome size of small viruses has driven the evolution of highly multifunctional proteins that integrate different domains and enzymatic activities within a single polypeptide. The Rep68 protein from adeno-associated virus (AAV) combines a DNA binding and endonuclease domain with a helicase-ATPase domain, which together support DNA replication, transcriptional regulation, and site-specific integration. The coordination of the enzymatic activities of Rep68 remains poorly understood; however, Rep68 oligomerization and Rep68-DNA interactions have been suggested to play a crucial role. We investigated the determinants of Rep68 oligomerization and identified a hydrophobic interface necessary for Rep68 activity during the AAV life cycle. Our results provide new insights into the molecular mechanisms underlying the regulation of the versatile Rep proteins. Efficient production of AAV-based gene therapy vectors requires optimal Rep expression levels, and studies such as the one presented here could contribute to further optimization of AAV production schemes.


Subject(s)
DNA Replication , DNA, Viral/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , DNA, Viral/chemistry , DNA, Viral/metabolism , DNA-Binding Proteins/genetics , Humans , Mutagenesis, Site-Directed , Mutation/genetics , Protein Binding , Protein Conformation , Protein Multimerization , Viral Proteins/genetics
13.
Proc Natl Acad Sci U S A ; 113(16): E2267-76, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27035982

ABSTRACT

Reb1 ofSchizosaccharomyces pomberepresents a family of multifunctional proteins that bind to specific terminator sites (Ter) and cause polar termination of transcription catalyzed by RNA polymerase I (pol I) and arrest of replication forks approaching the Ter sites from the opposite direction. However, it remains to be investigated whether the same mechanism causes arrest of both DNA transactions. Here, we present the structure of Reb1 as a complex with a Ter site at a resolution of 2.7 Å. Structure-guided molecular genetic analyses revealed that it has distinct and well-defined DNA binding and transcription termination (TTD) domains. The region of the protein involved in replication termination is distinct from the TTD. Mechanistically, the data support the conclusion that transcription termination is not caused by just high affinity Reb1-Ter protein-DNA interactions. Rather, protein-protein interactions between the TTD with the Rpa12 subunit of RNA pol I seem to be an integral part of the mechanism. This conclusion is further supported by the observation that double mutations in TTD that abolished its interaction with Rpa12 also greatly reduced transcription termination thereby revealing a conduit for functional communications between RNA pol I and the terminator protein.


Subject(s)
DNA, Fungal/chemistry , DNA-Binding Proteins/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces/chemistry , Terminator Regions, Genetic , Transcription Factors/chemistry , Transcription Termination, Genetic , Crystallography, X-Ray , DNA, Fungal/metabolism , DNA-Binding Proteins/metabolism , Protein Structure, Tertiary , RNA Polymerase I/chemistry , RNA Polymerase I/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Transcription Factors/metabolism
14.
J Biol Chem ; 290(46): 27487-99, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26370092

ABSTRACT

Adeno-associated virus (AAV) is the only eukaryotic virus with the property of establishing latency by integrating site-specifically into the human genome. The integration site known as AAVS1 is located in chromosome 19 and contains multiple GCTC repeats that are recognized by the AAV non-structural Rep proteins. These proteins are multifunctional, with an N-terminal origin-binding domain (OBD) and a helicase domain joined together by a short linker. As a first step to understand the process of site-specific integration, we proceeded to characterize the recognition and assembly of Rep68 onto the AAVS1 site. We first determined the x-ray structure of AAV-2 Rep68 OBD in complex with the AAVS1 DNA site. Specificity is achieved through the interaction of a glycine-rich loop that binds the major groove and an α-helix that interacts with a downstream minor groove on the same face of the DNA. Although the structure shows a complex with three OBD molecules bound to the AAVS1 site, we show by using analytical centrifugation and electron microscopy that the full-length Rep68 forms a heptameric complex. Moreover, we determined that a minimum of two direct repeats is required to form a stable complex and to melt DNA. Finally, we show that although the individual domains bind DNA poorly, complex assembly requires oligomerization and cooperation between its OBD, helicase, and the linker domains.


Subject(s)
DNA-Binding Proteins/chemistry , Dependovirus/physiology , Viral Proteins/chemistry , Virus Integration , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , DNA, Viral/chemistry , DNA-Binding Proteins/metabolism , Dependovirus/metabolism , Humans , Molecular Sequence Data , Protein Structure, Secondary , Viral Proteins/metabolism
15.
J Biol Chem ; 290(46): 27779-90, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26405037

ABSTRACT

IRF4 is a unique member of the interferon regulatory factor (IRF) family playing critical regulatory roles in immune cell development, regulation of obesity-induced inflammation, and control of thermogenic gene expression. The ability of IRF4 to control diverse transcriptional programs arises from its proficiency to interact with numerous transcriptional partners. In this study, we present the structural characterization of full-length IRF4. Using a combination of x-ray and small angle x-ray scattering studies, we reveal unique features of the interferon activation domain, including a set of ß-sheets and loops that serve as the binding site for PU.1, and also show that unlike other IRF members, IRF4 has a flexible autoinhibitory region. In addition, we have determined the small angle x-ray scattering solution structure of full-length IRF4, which, together with circular dichroism studies, suggests that the linker region is not extended but folds into a domain structure.


Subject(s)
Interferon Regulatory Factors/antagonists & inhibitors , Interferon Regulatory Factors/chemistry , Amino Acid Sequence , Animals , Crystallography, X-Ray , Interferon Regulatory Factors/ultrastructure , Mice , Models, Molecular , Molecular Sequence Data , Protein Folding , Protein Structure, Tertiary , Scattering, Small Angle , X-Ray Diffraction
16.
Biochemistry ; 54(38): 5907-19, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26314310

ABSTRACT

Adeno-associated virus (AAV) nonstructural proteins Rep78 and Rep68 carry out all DNA transactions that regulate the AAV life cycle. They share two multifunctional domains: an N-terminal origin binding/nicking domain (OBD) from the HUH superfamily and a SF3 helicase domain. A short linker of ∼20 amino acids that is critical for oligomerization and function connects the two domains. Although X-ray structures of the AAV5 OBD and AAV2 helicase domains have been determined, information about the full-length protein and linker conformation is not known. This article presents the solution structure of AAV2 Rep68 using small-angle X-ray scattering (SAXS). We first determined the X-ray structures of the minimal AAV2 Rep68 OBD and of the OBD with the linker region. These X-ray structures reveal novel features that include a long C-terminal α-helix that protrudes from the core of the protein at a 45° angle and a partially structured linker. SAXS studies corroborate that the linker is not extended, and we show that a proline residue in the linker is critical for Rep68 oligomerization and function. SAXS-based rigid-body modeling of Rep68 confirms these observations, showing a compact arrangement of the two domains in which they acquire a conformation that positions key residues in all domains on one face of the protein, poised to interact with DNA.


Subject(s)
DNA-Binding Proteins/chemistry , Dependovirus/chemistry , Viral Proteins/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Parvoviridae Infections/virology , Protein Structure, Tertiary , Scattering, Small Angle , X-Ray Diffraction
17.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 4): 414-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25849502

ABSTRACT

The Reb1 protein from Schizosaccharomyces pombe is a member of a family of proteins that control programmed replication termination and/or transcription termination in eukaryotic cells. These events occur at naturally occurring replication fork barriers (RFBs), where Reb1 binds to termination (Ter) DNA sites and coordinates the polar arrest of replication forks and transcription approaching in opposite directions. The Reb1 DNA-binding and replication-termination domain was expressed in Escherichia coli, purified and crystallized in complex with a 26-mer DNA Ter site. Batch crystallization under oil was required to produce crystals of good quality for data collection. Crystals grew in space group P21, with unit-cell parameters a = 68.9, b = 162.9, c = 71.1 Å, ß = 94.7°. The crystals diffracted to a resolution of 3.0 Å. The crystals were mosaic and required two or three cycles of annealing. This study is the first to yield structural information about this important family of proteins and will provide insights into the mechanism of replication and transcription termination.


Subject(s)
DNA Replication/physiology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Termination, Genetic/physiology , Amino Acid Sequence , Crystallization , Crystallography, X-Ray , Molecular Sequence Data , Schizosaccharomyces/genetics
18.
J Virol ; 88(15): 8227-41, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24829354

ABSTRACT

UNLABELLED: Adeno-associated virus serotype 2 (AAV2) can efficiently replicate in cells that have been infected with helper viruses, such as adenovirus or herpesvirus. However, in the absence of helper virus infection, AAV2 establishes latency by integrating its genome site specifically into PPP1R12C, a gene located on chromosome 19. This integration target site falls into one of the most gene-dense regions of the human genome, thus inviting the question as to whether the virus has evolved mechanisms to control this complex transcriptional environment in order to facilitate integration, maintain an apparently innocuous latency, and/or establish conditions that are conducive to the rescue of the integrated viral genome. The viral replication (Rep) proteins control and direct every known aspect of the viral life cycle and have been shown to tightly control all AAV2 promoters. In addition, a number of heterologous promoters are repressed by the AAV2 Rep proteins. Here, we demonstrate that Rep proteins efficiently repress expression from the target site PPP1R12C promoter. We find evidence that this repression employs mechanisms similar to those described for Rep-mediated AAV2 p5 promoter regulation. Furthermore, we show that the repression of the cellular target site promoter is based on two distinct mechanisms, one relying on the presence of a functional Rep binding motif within the 5' untranslated region (UTR) of PPP1R12C, whereas the second pathway requires only an intact nucleoside triphosphate (NTP) binding site within the Rep proteins, indicating the possible reliance of this pathway on interactions of the Rep proteins with cellular proteins that mediate or regulate cellular transcription. IMPORTANCE: The observation that repression of transcription from the adeno-associated virus serotype 2 (AAV2) p5 and integration target site promoters is mediated by shared mechanisms highlights the possible coevolution of virus and host and could lead to the identification of host factors that the virus exploits to navigate its life cycle.


Subject(s)
DNA-Binding Proteins/metabolism , Dependovirus/physiology , Gene Expression Regulation , Host-Pathogen Interactions , Promoter Regions, Genetic , Protein Phosphatase 1/genetics , Viral Proteins/metabolism , Virus Integration , Cell Line , Humans , Virus Latency
19.
J Virol ; 87(2): 1232-41, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23152528

ABSTRACT

The adeno-associated virus (AAV) encodes four regulatory proteins called Rep. The large AAV Rep proteins Rep68 and Rep78 are essential factors required in almost every step of the viral life cycle. Structurally, they share two domains: a modified version of the AAA(+) domain that characterizes the SF3 family of helicases and an N-terminal domain that binds DNA specifically. The combination of these two domains imparts extraordinary multifunctionality to work as initiators of DNA replication and regulators of transcription, in addition to their essential role during site-specific integration. Although most members of the SF3 family form hexameric rings in vitro, the oligomeric nature of Rep68 is unclear due to its propensity to aggregate in solution. We report here a comprehensive study to determine the oligomeric character of Rep68 using a combination of methods that includes sedimentation velocity ultracentrifugation, electron microscopy, and hydrodynamic modeling. We have determined that residue Cys151 induces Rep68 to aggregate in vitro. We show that Rep68 displays a concentration-dependent dynamic oligomeric behavior characterized by the presence of two populations: one with monomers and dimers in slow equilibrium and a second one consisting of a mixture of multiple-ring structures of seven and eight members. The presence of either ATP or ADP induces formation of larger complexes formed by the stacking of multiple rings. Taken together, our results support the idea of a Rep68 molecule that exhibits the flexible oligomeric behavior needed to perform the wide range of functions occurring during the AAV life cycle.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dependovirus/chemistry , Dependovirus/physiology , Protein Multimerization , Viral Proteins/chemistry , Viral Proteins/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Hydrodynamics , Microscopy, Electron , Ultracentrifugation
20.
Science ; 338(6109): 975-80, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-22983707

ABSTRACT

Interferon regulatory factor 4 (IRF4) and IRF8 regulate B, T, macrophage, and dendritic cell differentiation. They are recruited to cis-regulatory Ets-IRF composite elements by PU.1 or Spi-B. How these IRFs target genes in most T cells is enigmatic given the absence of specific Ets partners. Chromatin immunoprecipitation sequencing in T helper 17 (T(H)17) cells reveals that IRF4 targets sequences enriched for activating protein 1 (AP-1)-IRF composite elements (AICEs) that are co-bound by BATF, an AP-1 factor required for T(H)17, B, and dendritic cell differentiation. IRF4 and BATF bind cooperatively to structurally divergent AICEs to promote gene activation and T(H)17 differentiation. The AICE motif directs assembly of IRF4 or IRF8 with BATF heterodimers and is also used in T(H)2, B, and dendritic cells. This genomic regulatory element and cognate factors appear to have evolved to integrate diverse immunomodulatory signals.


Subject(s)
Immunomodulation/genetics , Interferon Regulatory Factors/metabolism , Regulatory Elements, Transcriptional , Th17 Cells/immunology , Transcription Factor AP-1/metabolism , Transcriptional Activation , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Humans , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...