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










Publication year range
1.
Nucleic Acids Res ; 49(22): 12895-12911, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34850113

ABSTRACT

Mixed lineage leukemia 1 (MLL1) is a histone methyltransferase. Kaposi's sarcoma-associated herpesvirus (KSHV) is a leading cause of malignancy in AIDS. KSHV latently infects tumor cells and its genome is decorated with epigenetic marks. Here, we show that KSHV latency-associated nuclear antigen (LANA) recruits MLL1 to viral DNA where it establishes H3K4me3 modifications at the extensive KSHV terminal repeat elements during primary infection. LANA interacts with MLL1 complex members, including WDR5, integrates into the MLL1 complex, and regulates MLL1 activity. We describe the 1.5-Å crystal structure of N-terminal LANA peptide complexed with MLL1 complex member WDR5, which reveals a potential regulatory mechanism. Disruption of MLL1 expression rendered KSHV latency establishment highly deficient. This deficiency was rescued by MLL1 but not by catalytically inactive MLL1. Therefore, MLL1 is LANA regulable and exerts a central role in virus infection. These results suggest broad potential for MLL1 regulation, including by non-host factors.


Subject(s)
Antigens, Viral/genetics , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Sarcoma, Kaposi/genetics , Virus Latency/genetics , Antigens, Viral/chemistry , Antigens, Viral/metabolism , Cell Line, Tumor , Crystallography, X-Ray , DNA, Viral/genetics , DNA, Viral/metabolism , Gene Knockdown Techniques , Herpesvirus 8, Human/metabolism , Herpesvirus 8, Human/physiology , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Host-Pathogen Interactions/genetics , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/chemistry , Myeloid-Lymphoid Leukemia Protein/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , Sarcoma, Kaposi/virology
2.
J Virol ; 92(21)2018 11 01.
Article in English | MEDLINE | ID: mdl-30111565

ABSTRACT

The latency-associated nuclear antigen from Kaposi's sarcoma-associated herpesvirus (KSHV), kLANA, and its homolog from the murid herpesvirus 4 (MuHV-4), mLANA, are essential for viral latency. kLANA is nearly four times the size of mLANA, mainly due to an extensive central repeat region that is absent in mLANA. Both proteins harbor a C-terminal DNA binding domain (DBD). The DBD binds the terminal repeat (TR) DNA sequences of the viral genome to mediate persistence. Despite structural conservation, the kLANA and mLANA DBDs differ in sequence and mode of oligomerization. kLANA DBD oligomers are flexible and bent, while mLANA DBD oligomers bind DNA in a rigid, linear conformation. We previously reported that kLANA and mLANA acted reciprocally on TR sequences. Furthermore, a MuHV-4 expressing kLANA instead of mLANA (v-kLANA) established latency in mice, albeit at a lower magnitude than the wild-type (WT) virus. Here, we asked if kLANA can accommodate the mLANA DBD and generated a fusion protein which contains kLANA but with the mLANA C-terminal region in place of that of kLANA. We report a recombinant MuHV-4 (v-KM) encoding this LANA fusion protein instead of mLANA. The fusion protein was expressed in lytic infection in vitro and assembled nuclear LANA dots in infected splenocytes. Results demonstrated that kLANA functionally accommodated mLANA's mode of DNA binding, allowing MuHV-4 chimeric virus to establish latency in vivo Notably, v-KM established latency in germinal center B cells more efficiently than did v-kLANA, although levels were reduced compared to WT MuHV-4.IMPORTANCE KSHV is a human oncogenic virus for which there is no tractable, immunocompetent animal model of infection. MuHV-4, a related rodent gammaherpesvirus, enables pathogenesis studies in mice. In latency, both viruses persist as extrachromosomal, circular genomes (episomes). LANA proteins encoded by KSHV (kLANA) and MuHV-4 (mLANA) contain a C-terminal DNA binding domain (DBD) that acts on the virus terminal repeats to enable episome persistence. mLANA is a smaller protein than kLANA. Their DBDs are structurally conserved but differ strikingly in the conformation of DNA binding. We report a recombinant, chimeric MuHV-4 which contains kLANA in place of mLANA, but in which the DBD is replaced with that of mLANA. Results showed that kLANA functionally accommodated mLANA's mode of DNA binding. In fact, the new chimeric virus established latency in vivo more efficiently than MuHV-4 expressing full-length kLANA.


Subject(s)
Antigens, Viral/metabolism , DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Herpesvirus 8, Human/genetics , Nuclear Proteins/metabolism , Rhadinovirus/genetics , Terminal Repeat Sequences/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Line , DNA, Viral/genetics , Genome, Viral/genetics , Mice , Virus Latency/genetics
3.
PLoS Pathog ; 13(9): e1006555, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28910389

ABSTRACT

Many pathogens, including Kaposi's sarcoma herpesvirus (KSHV), lack tractable small animal models. KSHV persists as a multi-copy, nuclear episome in latently infected cells. KSHV latency-associated nuclear antigen (kLANA) binds viral terminal repeat (kTR) DNA to mediate episome persistence. Model pathogen murine gammaherpesvirus 68 (MHV68) mLANA acts analogously on mTR DNA. kLANA and mLANA differ substantially in size and kTR and mTR show little sequence conservation. Here, we find kLANA and mLANA act reciprocally to mediate episome persistence of TR DNA. Further, kLANA rescued mLANA deficient MHV68, enabling a chimeric virus to establish latent infection in vivo in germinal center B cells. The level of chimeric virus in vivo latency was moderately reduced compared to WT infection, but WT or chimeric MHV68 infected cells had similar viral genome copy numbers as assessed by immunofluorescence of LANA intranuclear dots or qPCR. Thus, despite more than 60 Ma of evolutionary divergence, mLANA and kLANA act reciprocally on TR DNA, and kLANA functionally substitutes for mLANA, allowing kLANA investigation in vivo. Analogous chimeras may allow in vivo investigation of genes of other human pathogens.


Subject(s)
Antigens, Viral/metabolism , DNA, Viral/genetics , Genome, Viral/genetics , Germinal Center/metabolism , Herpesvirus 8, Human , Nuclear Proteins/metabolism , Plasmids/metabolism , Sarcoma, Kaposi/metabolism , Virus Latency/genetics , Animals , Antigens, Viral/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Mice , Nuclear Proteins/genetics , Plasmids/genetics , Sarcoma, Kaposi/virology
4.
J Virol ; 90(17): 7667-83, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27307564

ABSTRACT

UNLABELLED: Viruses have evolved mechanisms to hijack components of cellular E3 ubiquitin ligases, thus modulating the ubiquitination pathway. However, the biological relevance of such mechanisms for viral pathogenesis in vivo remains largely unknown. Here, we utilized murid herpesvirus 4 (MuHV-4) infection of mice as a model system to address the role of MuHV-4 latency-associated nuclear antigen (mLANA) E3 ligase activity in gammaherpesvirus latent infection. We show that specific mutations in the mLANA SOCS box (V199A, V199A/L202A, or P203A/P206A) disrupted mLANA's ability to recruit Elongin C and Cullin 5, thereby impairing the formation of the Elongin BC/Cullin 5/SOCS (EC5S(mLANA)) complex and mLANA's E3 ligase activity on host NF-κB and Myc. Although these mutations resulted in considerably reduced mLANA binding to viral terminal repeat DNA as assessed by electrophoretic mobility shift assay (EMSA), the mutations did not disrupt mLANA's ability to mediate episome persistence. In vivo, MuHV-4 recombinant viruses bearing these mLANA SOCS box mutations exhibited a deficit in latency amplification in germinal center (GC) B cells. These findings demonstrate that the E3 ligase activity of mLANA contributes to gammaherpesvirus-driven GC B cell proliferation. Hence, pharmacological inhibition of viral E3 ligase activity through targeting SOCS box motifs is a putative strategy to control gammaherpesvirus-driven lymphoproliferation and associated disease. IMPORTANCE: The gammaherpesviruses Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) cause lifelong persistent infection and play causative roles in several human malignancies. Colonization of B cells is crucial for virus persistence, and access to the B cell compartment is gained by virus-driven proliferation in germinal center (GC) B cells. Infection of B cells is predominantly latent, with the viral genome persisting as a multicopy episome and expressing only a small subset of viral genes. Here, we focused on latency-associated nuclear antigen (mLANA) encoded by murid herpesvirus-4 (MuHV-4), which exhibits homology in sequence, structure, and function to KSHV LANA (kLANA), thereby allowing the study of LANA-mediated pathogenesis in mice. Our experiments show that mLANA's E3 ubiquitin ligase activity is necessary for efficient expansion of latency in GC B cells, suggesting that the development of pharmacological inhibitors of LANA E3 ubiquitin ligase activity may allow strategies to interfere with gammaherpesvirus-driven lymphoproliferation and associated disease.


Subject(s)
Antigens, Viral/metabolism , B-Lymphocytes/physiology , Cell Proliferation , Germinal Center/cytology , Host-Pathogen Interactions , Nuclear Proteins/metabolism , Rhadinovirus/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Antigens, Viral/genetics , DNA Mutational Analysis , DNA, Viral/metabolism , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Nuclear Proteins/genetics , Protein Binding
5.
Anal Chem ; 88(12): 6214-22, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27238076

ABSTRACT

We present the first study of a novel, more sensitive method for the characterization of nanoparticles (NPs). This approach combines detection via a protein nanopore with modification of its interaction behavior using a molecular adaptor. We identify different populations of 3-mercapto-1-propanesulfonate (MPSA)-modified-gold NPs using the biological nanopores α-hemolysin (αHL) and its M113N mutant equipped with a noncovalently bound γ-cyclodextrin molecule as a stochastic sensor. Identification takes place on the basis of the extent of current blockades and residence times. Here, we demonstrate that noncovalently attached adaptors can be used to change the sensing properties of αHL nanopores, allowing the detection and characterization of different populations of MPSA NPs. This is an advance in sensitivity and diversity of NP sensing, as well as a promising and reliable technology to characterize NPs by using protein nanopores.

6.
J Biol Chem ; 290(47): 28084-28096, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26420481

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) has a causative role in several human malignancies. KSHV latency-associated nuclear antigen (LANA) mediates persistence of viral episomes in latently infected cells. LANA mediates KSHV DNA replication and segregates episomes to progeny nuclei. The structure of the LANA DNA binding domain was recently solved, revealing a positive electrostatic patch opposite the DNA binding surface, which is the site of BET protein binding. Here we investigate the functional role of the positive patch in LANA-mediated episome persistence. As expected, LANA mutants with alanine or glutamate substitutions in the central, peripheral, or lateral portions of the positive patch maintained the ability to bind DNA by EMSA. However, all of the substitution mutants were deficient for LANA DNA replication and episome maintenance. Mutation of the peripheral region generated the largest deficiencies. Despite these deficiencies, all positive patch mutants concentrated to dots along mitotic chromosomes in cells containing episomes, similar to LANA. The central and peripheral mutants, but not the lateral mutants, were reduced for BET protein interaction as assessed by co-immunoprecipitation. However, defects in BET protein binding were independent of episome maintenance function. Overall, the reductions in episome maintenance closely correlated with DNA replication deficiencies, suggesting that the replication defects account for the reduced episome persistence. Therefore, the electrostatic patch exerts a key role in LANA-mediated DNA replication and episome persistence and may act through a host cell partner(s) other than a BET protein or by inducing specific structures or complexes.


Subject(s)
Antigens, Viral/metabolism , DNA Replication , Herpesvirus 8, Human/genetics , Nuclear Proteins/metabolism , Plasmids/physiology , Virus Latency , Binding Sites , Cell Line, Tumor , Herpesvirus 8, Human/immunology , Humans , Static Electricity , Terminal Repeat Sequences
7.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 10): 2137-49, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26457437

ABSTRACT

Uracil-DNA N-glycosylase (UNG) is a DNA-repair enzyme in the base-excision repair (BER) pathway which removes uracil from DNA. Here, the crystal structure of UNG from the extremophilic bacterium Deinococcus radiodurans (DrUNG) in complex with DNA is reported at a resolution of 1.35 Å. Prior to the crystallization experiments, the affinity between DrUNG and different DNA oligonucleotides was tested by electrophoretic mobility shift assays (EMSAs). As a result of this analysis, two 16 nt double-stranded DNAs were chosen for the co-crystallization experiments, one of which (16 nt AU) resulted in well diffracting crystals. The DNA in the co-crystal structure contained an abasic site (substrate product) flipped into the active site of the enzyme, with no uracil in the active-site pocket. Despite the high resolution, it was not possible to fit all of the terminal nucleotides of the DNA complex into electron density owing to disorder caused by a lack of stabilizing interactions. However, the DNA which was in contact with the enzyme, close to the active site, was well ordered and allowed detailed analysis of the enzyme-DNA interaction. The complex revealed that the interaction between DrUNG and DNA is similar to that in the previously determined crystal structure of human UNG (hUNG) in complex with DNA [Slupphaug et al. (1996). Nature (London), 384, 87-92]. Substitutions in a (here defined) variable part of the leucine loop result in a shorter loop (eight residues instead of nine) in DrUNG compared with hUNG; regardless of this, it seems to fulfil its role and generate a stabilizing force with the minor groove upon flipping out of the damaged base into the active site. The structure also provides a rationale for the previously observed high catalytic efficiency of DrUNG caused by high substrate affinity by demonstrating an increased number of long-range electrostatic interactions between the enzyme and the DNA. Interestingly, specific interactions between residues in the N-terminus of a symmetry-related molecule and the complementary DNA strand facing away from the active site were also observed which seem to stabilize the enzyme-DNA complex. However, the significance of this observation remains to be investigated. The results provide new insights into the current knowledge about DNA damage recognition and repair by uracil-DNA glycosylases.


Subject(s)
DNA/metabolism , Deinococcus/enzymology , Uracil-DNA Glycosidase/chemistry , Uracil-DNA Glycosidase/metabolism , Amino Acid Sequence , Base Sequence , Crystallography, X-Ray , DNA/chemistry , Deinococcus/chemistry , Deinococcus/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment
8.
Nucleic Acids Res ; 43(20): 10039-54, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26424851

ABSTRACT

Latency-associated nuclear antigen (LANA) is central to episomal tethering, replication and transcriptional regulation of γ2-herpesviruses. LANA binds cooperatively to the terminal repeat (TR) region of the viral episome via adjacent LANA binding sites (LBS), but the molecular mechanism by which LANA assembles on the TR remains elusive. We show that KSHV LANA and MHV-68 LANA proteins bind LBS DNA using strikingly different modes. Solution structure of LANA complexes revealed that while kLANA tetramer is intrinsically bent both in the free and bound state to LBS1-2 DNA, mLANA oligomers instead adopt a rigid linear conformation. In addition, we report a novel non-ring kLANA structure that displays more flexibility at its assembly interface than previously demonstrated. We identified a hydrophobic pivot point located at the dimer-dimer assembly interface, which gives rotational freedom for kLANA to adopt variable conformations to accommodate both LBS1-2 and LBS2-1-3 DNA. Alterations in the arrangement of LBS within TR or at the tetramer assembly interface have a drastic effect on the ability of kLANA binding. We also show kLANA and mLANA DNA binding functions can be reciprocated. Although KSHV and MHV-68 are closely related, the findings provide new insights into how the structure, oligomerization, and DNA binding of LANA have evolved differently to assemble on the TR DNA.


Subject(s)
Antigens, Viral/chemistry , DNA, Viral/chemistry , Herpesvirus 8, Human , Nuclear Proteins/chemistry , Rhadinovirus , Antigens, Viral/genetics , Antigens, Viral/metabolism , Binding Sites , DNA, Viral/metabolism , Models, Molecular , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Terminal Repeat Sequences , Thermodynamics
9.
PLoS Pathog ; 9(10): e1003673, 2013.
Article in English | MEDLINE | ID: mdl-24146618

ABSTRACT

Latency-associated nuclear antigen (LANA) mediates γ2-herpesvirus genome persistence and regulates transcription. We describe the crystal structure of the murine gammaherpesvirus-68 LANA C-terminal domain at 2.2 Šresolution. The structure reveals an alpha-beta fold that assembles as a dimer, reminiscent of Epstein-Barr virus EBNA1. A predicted DNA binding surface is present and opposite this interface is a positive electrostatic patch. Targeted DNA recognition substitutions eliminated DNA binding, while certain charged patch mutations reduced bromodomain protein, BRD4, binding. Virus containing LANA abolished for DNA binding was incapable of viable latent infection in mice. Virus with mutations at the charged patch periphery exhibited substantial deficiency in expansion of latent infection, while central region substitutions had little effect. This deficiency was independent of BRD4. These results elucidate the LANA DNA binding domain structure and reveal a unique charged region that exerts a critical role in viral latent infection, likely acting through a host cell protein(s).


Subject(s)
DNA, Viral/chemistry , Protein Folding , Rhadinovirus/chemistry , Viral Proteins/chemistry , Virus Latency , DNA, Viral/genetics , DNA, Viral/metabolism , Mutation , Protein Binding , Protein Structure, Tertiary , Rhadinovirus/physiology , Viral Proteins/genetics , Viral Proteins/metabolism
10.
Anal Chem ; 85(21): 10149-58, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24053797

ABSTRACT

Gold nanoparticles are widely used in various applications in fields including chemistry, engineering, biology, medicine, and electronics. These materials can be synthesized and modified with ligands containing different functional groups. Among nanoparticles' characteristics, chemical surface composition is likely to be a crucial feature, demanding robust analytical methodologies for its assessment. Single molecule analysis using the biological nanopores α-hemolysin and its E111A mutant is presented here as a promising methodology to stochastically sense organic monolayer protected gold-nanoparticles with different ligand shell compositions. By monitoring the ionic current across a single protein nanopore, differences in the physical and chemical characteristics (e.g., size, ligand shell composition, and arrangement) of individual nanoparticles can be distinguished based on the differences in the current blockade events that they cause. Such differences are observed in the spread of both the amplitude and duration of current blockades. These values cannot be correlated with a single physical characteristic. Instead the spread represents a measure of heterogeneity within the nanoparticle population. While our results compare favorably with the more traditional analytical methodologies, further work will be required to improve the accuracy of identification of the NPs and understand the spread of values within a nanoparticle preparation as well as the overlap between similar preparations.


Subject(s)
Gold/chemistry , Hemolysin Proteins/chemistry , Metal Nanoparticles , Nanopores , Mutagenesis
11.
J Biol Chem ; 288(6): 3858-70, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23258536

ABSTRACT

γ-Herpesviruses express proteins that modulate B lymphocyte signaling to achieve persistent latent infections. One such protein is the M2 latency-associated protein encoded by the murid herpesvirus-4. M2 has two closely spaced tyrosine residues, Tyr(120) and Tyr(129), which are phosphorylated by Src family tyrosine kinases. Here we used mass spectrometry to identify the binding partners of tyrosine-phosphorylated M2. Each M2 phosphomotif is shown to bind directly and selectively to SH2-containing signaling molecules. Specifically, Src family kinases, NCK1 and Vav1, bound to the Tyr(P)(120) site, PLCγ2 and the SHP2 phosphatase bound to the Tyr(P)(129) motif, and the p85α subunit of PI3K associated with either motif. Consistent with these data, we show that M2 coordinates the formation of multiprotein complexes with these proteins. The effect of those interactions is functionally bivalent, because it can result in either the phosphorylation of a subset of binding proteins (Vav1 and PLCγ2) or in the inactivation of downstream targets (AKT). Finally, we show that translocation to the plasma membrane and subsequent M2 tyrosine phosphorylation relies on the integrity of a C-terminal proline-rich SH3 binding region of M2 and its interaction with Src family kinases. Unlike other γ-herpesviruses, that encode transmembrane proteins that mimic the activation of ITAMs, murid herpesvirus-4 perturbs B cell signaling using a cytoplasmic/membrane shuttling factor that nucleates the assembly of signaling complexes using a bilayered mechanism of phosphotyrosine and proline-rich anchoring motifs.


Subject(s)
B-Lymphocytes/metabolism , Herpesviridae Infections/metabolism , Multiprotein Complexes/metabolism , Muromegalovirus/metabolism , Signal Transduction , Viral Proteins/metabolism , Amino Acid Motifs , Animals , B-Lymphocytes/virology , Cell Membrane/genetics , Cell Membrane/metabolism , Herpesviridae Infections/genetics , Mice , Multiprotein Complexes/genetics , Muromegalovirus/genetics , Protein Binding , Protein Transport/genetics , Viral Proteins/genetics , src Homology Domains
12.
Langmuir ; 28(44): 15643-50, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23046444

ABSTRACT

Single channel recordings were used to determine the effect of direct electrostatic interactions between sulfonate-coated gold nanoparticles and the constriction of the Staphylococcus aureus α-hemolysin protein channel on the ionic current amplitude. We provide evidence that Lys147 of α-hemolysin can interact with the sulfonate groups at the nanoparticle surface, and these interactions can reversibly block 100% of the residual ionic current. Lys147 is normally involved in a salt bridge with Glu111. The capture of a nanoparticle leads to a partial current block at neutral pH values, but protonation of Glu111 at pH 2.8 results in a full current block when the nanoparticle is captured. At pH 2.8, we suggest that Lys147 is free to engage in electrostatic interactions with sulfonates at the nanoparticle surface. To verify our results, we engineered a mutation in the α-hemolysin protein, where Glu111 is substituted by Ala (E111A), thus removing Glu111-Lys147 interactions and facilitating Lys147-sulfonate electrostatic interactions. This mutation leads to a 100% current block at pH 2.8 and a 92% block at pH 8.0, showing that electrostatic interactions are formed between the nanopore and the nanoparticle surface. Besides demonstrating the effect of electrostatic interactions on cross channel ionic current, this work offers a novel approach to controlling open and closed states of the α-hemolysin nanopore as a function of external gears.


Subject(s)
Gold/chemistry , Hemolysin Proteins/chemistry , Lysine/chemistry , Metal Nanoparticles/chemistry , Nanopores , Sulfhydryl Compounds/chemistry , Hydrogen-Ion Concentration , Models, Molecular , Staphylococcus aureus/chemistry , Surface Properties
13.
J Struct Biol ; 176(3): 279-91, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21933716

ABSTRACT

RuvBL1 (RuvB-like 1) and its homolog RuvBL2 are evolutionarily highly conserved AAA(+) ATPases essential for many cellular activities. They play an important role in chromatin remodeling, transcriptional regulation and DNA damage repair. RuvBL1 and RuvBL2 are overexpressed in different types of cancer and interact with major oncogenic factors, such as ß-catenin and c-Myc regulating their function. We solved the first three-dimensional crystal structure of the human RuvBL complex with a truncated domain II and show that this complex is competent for helicase activity. The structure reveals a dodecamer consisting of two heterohexameric rings with alternating RuvBL1 and RuvBL2 monomers bound to ADP/ATP, that interact with each other via the retained part of domain II. The dodecameric quaternary structure of the R1ΔDII/R2ΔDII complex observed in the crystal structure was confirmed by small-angle X-ray scattering analysis. Interestingly, truncation of domain II led to a substantial increase in ATP consumption of RuvBL1, RuvBL2 and their complex. In addition, we present evidence that DNA unwinding of the human RuvBL proteins can be auto-inhibited by domain II, which is not present in the homologous bacterial helicase RuvB. Our data give new insights into the molecular arrangement of RuvBL1 and RuvBL2 and strongly suggest that in vivo activities of these highly interesting therapeutic drug targets are regulated by cofactors inducing conformational changes via domain II in order to modulate the enzyme complex into its active state.


Subject(s)
Carrier Proteins/chemistry , DNA Helicases/chemistry , Macromolecular Substances/chemistry , ATPases Associated with Diverse Cellular Activities , Catalytic Domain , Crystallography, X-Ray , Humans , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary
14.
J Mol Biol ; 387(3): 759-70, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19232357

ABSTRACT

SorC transcriptional regulators are common regulators in prokaryotes. Here we report the first crystal structure of a full-length SorC, the sorbitol operon regulator SorC from Klebsiella pneumoniae, the prototype of its family. SorC was found to be a homotetramer (which seems to be the biologically active form) that is able to recognize its DNA operator. The tetramer can be regarded as a dimer of dimers, with each dimer being composed of two identical subunits in different conformations. The DNA-binding domains divergently protrude from the core of the tetramer, suggesting that SorC may bind its operator in two distinct regions. The sugar-binding domain presents the same fold identified in members of the SorC family that shows some features identified as specific for sugar recognition. An in silico analysis shows that the localization of the putative sugar-binding site is close to the dimeric interfaces. This supports the proposal of a new mechanism of transcriptional regulation that is in complete agreement with functional studies recently reported on a protein belonging to the same family.


Subject(s)
Bacterial Proteins/chemistry , Gene Expression Regulation, Bacterial , Klebsiella pneumoniae/genetics , Operon , Protein Structure, Quaternary , Sorbitol/metabolism , Transcription Factors/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Klebsiella pneumoniae/metabolism , Models, Molecular , Molecular Sequence Data , Operator Regions, Genetic , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Sorbitol/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Article in English | MEDLINE | ID: mdl-18097095

ABSTRACT

The sorbitol operon regulator (SorC) regulates the metabolism of L-sorbose in Klebsiella pneumonia. SorC was overexpressed in Escherichia coli and purified, and crystals were obtained of a tetrameric form. A single crystal showed X-ray diffraction to 3.20 A. The crystal belongs to space group P2(1)2(1)2(1), with unit-cell parameters a = 91.6, b = 113.3, c = 184.1 A. Analysis of the molecular-replacement solution indicates the presence of four SorC molecules in the asymmetric unit.


Subject(s)
Bacterial Proteins/chemistry , Klebsiella pneumoniae/genetics , Operon , Recombinant Proteins/chemistry , Sorbitol/metabolism , Trans-Activators/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Cloning, Molecular , Crystallography, X-Ray , Macromolecular Substances/chemistry , Polymerase Chain Reaction , Recombinant Proteins/isolation & purification , Sorbose/metabolism , X-Ray Diffraction
16.
J Gen Virol ; 88(Pt 6): 1656-1666, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17485524

ABSTRACT

Vaccinia virus (VACV) encodes many immunomodulatory proteins, including inhibitors of apoptosis and modulators of innate immune signalling. VACV protein N1 is an intracellular homodimer that contributes to virus virulence and was reported to inhibit nuclear factor (NF)-kappaB signalling. However, analysis of NF-kappaB signalling in cells infected with recombinant viruses with or without the N1L gene showed no difference in NF-kappaB-dependent gene expression. Given that N1 promotes virus virulence, other possible functions of N1 were investigated and this revealed that N1 is an inhibitor of apoptosis in cells transfected with the N1L gene and in the context of VACV infection. In support of this finding virally expressed N1 co-precipitated with endogenous pro-apoptotic Bcl-2 proteins Bid, Bad and Bax as well as with Bad and Bax expressed by transfection. In addition, the crystal structure of N1 was solved to 2.9 A resolution (0.29 nm). Remarkably, although N1 shows no sequence similarity to cellular proteins, its three-dimensional structure closely resembles Bcl-x(L) and other members of the Bcl-2 protein family. The structure also reveals that N1 has a constitutively open surface groove similar to the grooves of other anti-apoptotic Bcl-2 proteins, which bind the BH3 motifs of pro-apoptotic Bcl-2 family members. Molecular modelling of BH3 peptides into the N1 surface groove, together with analysis of their physico-chemical properties, suggests a mechanism for the specificity of peptide recognition. This study illustrates the importance of the evolutionary conservation of structure, rather than sequence, in protein function and reveals a novel anti-apoptotic protein from orthopoxviruses.


Subject(s)
Vaccinia virus/pathogenicity , Viral Proteins/chemistry , Viral Proteins/physiology , Virulence Factors/chemistry , Virulence Factors/physiology , Amino Acid Sequence , Animals , BH3 Interacting Domain Death Agonist Protein/chemistry , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Line , Chlorocebus aethiops , Crystallography, X-Ray , Humans , Immunoprecipitation , Models, Molecular , Molecular Sequence Data , NF-kappa B/metabolism , Protein Binding , Protein Structure, Tertiary , Vaccinia virus/immunology , bcl-2-Associated X Protein/chemistry , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/chemistry , bcl-Associated Death Protein/metabolism , bcl-X Protein/chemistry
17.
Nature ; 443(7107): 110-4, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16957732

ABSTRACT

RNA degradation is a determining factor in the control of gene expression. The maturation, turnover and quality control of RNA is performed by many different classes of ribonucleases. Ribonuclease II (RNase II) is a major exoribonuclease that intervenes in all of these fundamental processes; it can act independently or as a component of the exosome, an essential RNA-degrading multiprotein complex. RNase II-like enzymes are found in all three kingdoms of life, but there are no structural data for any of the proteins of this family. Here we report the X-ray crystallographic structures of both the ligand-free (at 2.44 A resolution) and RNA-bound (at 2.74 A resolution) forms of Escherichia coli RNase II. In contrast to sequence predictions, the structures show that RNase II is organized into four domains: two cold-shock domains, one RNB catalytic domain, which has an unprecedented alphabeta-fold, and one S1 domain. The enzyme establishes contacts with RNA in two distinct regions, the 'anchor' and the 'catalytic' regions, which act synergistically to provide catalysis. The active site is buried within the RNB catalytic domain, in a pocket formed by four conserved sequence motifs. The structure shows that the catalytic pocket is only accessible to single-stranded RNA, and explains the specificity for RNA versus DNA cleavage. It also explains the dynamic mechanism of RNA degradation by providing the structural basis for RNA translocation and enzyme processivity. We propose a reaction mechanism for exonucleolytic RNA degradation involving key conserved residues. Our three-dimensional model corroborates all existing biochemical data for RNase II, and elucidates the general basis for RNA degradation. Moreover, it reveals important structural features that can be extrapolated to other members of this family.


Subject(s)
Escherichia coli/enzymology , Exoribonucleases/chemistry , Exoribonucleases/metabolism , RNA/chemistry , RNA/metabolism , Catalysis , Escherichia coli/genetics , Exoribonucleases/genetics , Hydrogen Bonding , Models, Molecular , Protein Conformation , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Static Electricity
18.
Article in English | MEDLINE | ID: mdl-16820694

ABSTRACT

RNA degradation is important in the post-transcriptional control of gene expression. The processing, degradation and quality control of RNA is performed by many different classes of ribonucleases. Ribonuclease II (RNase II) is a 643-amino-acid enzyme that degrades single-stranded RNA from its 3'-end, releasing ribonucleoside 5'-monophosphates. RNase II was expressed both as the wild type and as a D209N mutant form. The latter was also produced as an SeMet derivative. The various protein forms were crystallized using the vapour-diffusion method. Wild-type RNase II was crystallized in two crystal forms, both of which belonged to space group P2(1). X-ray diffraction data were collected to 2.44 and 2.75 angstroms resolution, with unit-cell parameters a = 56.8, b = 125.7, c = 66.2 angstroms, beta = 111.9 degrees and a = 119.6, b = 57.2, c = 121.2 angstroms, beta = 99.7 degrees, respectively. The RNase II D209N mutant gave crystals that belonged to space group P6(5), with unit-cell parameters a = b = 86.3, c = 279.2 angstroms, and diffracted to 2.74 angstroms. Diffraction data from the mutant and its SeMet derivative enabled the determination of a partial Se-atom substructure by SIRAS.


Subject(s)
Escherichia coli/enzymology , Exoribonucleases/chemistry , Exoribonucleases/isolation & purification , Crystallization , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Exoribonucleases/genetics , Mutagenesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Selenomethionine , X-Ray Diffraction
19.
J Gen Virol ; 85(Pt 6): 1401-1412, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15166422

ABSTRACT

Poxviruses express numerous proteins involved in manipulating the host immune response. Analysis of the primary sequence and predicted structure of the 134R protein of Yaba-like disease virus (Y134R) indicated that it is similar to cellular proteins of the IL-10 family, specifically IL-19, IL-20 and IL-24. A flag-tagged Y134R was expressed from mammalian cells and identified as a secreted, monomeric glycoprotein that stimulated signal transduction from class II cytokine receptors IL-20Ralpha/IL-20Rbeta (IL-20R type1) and IL-22R/IL-20Rbeta (IL-20R type 2). Y134R induced phosphorylation of signal transducers and activators of transcription, their translocation to the nucleus and the induction of reporter gene expression. In contrast, Y134R was unable to induce similar responses from either the IL-22 or IFN-lambda (IL-28A, IL-28B, IL-29) class II cytokine receptors. To examine the role Y134R plays during a poxvirus infection, a vaccinia virus recombinant expressing Y134R was constructed and tested in a murine intranasal infection model. Compared with control viruses, the virus expressing Y134R had a reduced virulence, manifested by reduced weight loss, signs of illness and virus titres in infected organs. These results demonstrate that Y134R is a new viral member of the IL-10-related cytokine family and that its activity in vivo affects virus virulence.


Subject(s)
Interleukin-10/physiology , Poxviridae/immunology , Viral Proteins/physiology , Amino Acid Sequence , Animals , DNA-Binding Proteins/physiology , Glycosylation , Humans , Interleukin-10/analysis , Interleukin-10/genetics , Molecular Sequence Data , Phosphorylation , Poxviridae/pathogenicity , Receptors, Cytokine/analysis , STAT3 Transcription Factor , Signal Transduction , Trans-Activators/physiology , Virulence
20.
Biochem J ; 371(Pt 1): 143-50, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12511194

ABSTRACT

Two mutant forms of penicillin acylase from Escherichia coli strains, selected using directed evolution for the ability to use glutaryl-L-leucine for growth [Forney, Wong and Ferber (1989) Appl. Environ. Microbiol. 55, 2550-2555], are changed within one codon, replacing the B-chain residue Phe(B71) with either Cys or Leu. Increases of up to a factor of ten in k (cat)/ K (m) values for substrates possessing a phenylacetyl leaving group are consistent with a decrease in K (s). Values of k (cat)/ K (m) for glutaryl-L-leucine are increased at least 100-fold. A decrease in k (cat)/ K (m) for the Cys(B71) mutant with increased pH is consistent with binding of the uncharged glutaryl group. The mutant proteins are more resistant to urea denaturation monitored by protein fluorescence, to inactivation in the presence of substrate either in the presence of urea or at high pH, and to heat inactivation. The crystal structure of the Leu(B71) mutant protein, solved to 2 A resolution, shows a flip of the side chain of Phe(B256) into the periphery of the catalytic centre, associated with loss of the pi-stacking interactions between Phe(B256) and Phe(B71). Molecular modelling demonstrates that glutaryl-L-leucine may bind with the uncharged glutaryl group in the S(1) subsite of either the wild-type or the Leu(B71) mutant but with greater potential freedom of rotation of the substrate leucine moiety in the complex with the mutant protein. This implies a smaller decrease in the conformational entropy of the substrate on binding to the mutant proteins and consequently greater catalytic activity.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Leucine/analogs & derivatives , Mutation , Penicillin Amidase/chemistry , Penicillin Amidase/metabolism , Binding Sites , Crystallography, X-Ray , Cysteine/genetics , Enzyme Stability , Escherichia coli Proteins/genetics , Glutarates/metabolism , Kinetics , Leucine/genetics , Leucine/metabolism , Models, Molecular , Penicillin Amidase/genetics , Phenylalanine/genetics , Protein Conformation , Protein Denaturation , Substrate Specificity , Urea/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...