Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Proc Natl Acad Sci U S A ; 117(29): 17195-17203, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32606248

ABSTRACT

The vast majority of intracellular protein targets are refractory toward small-molecule therapeutic engagement, and additional therapeutic modalities are needed to overcome this deficiency. Here, the identification and characterization of a natural product, WDB002, reveals a therapeutic modality that dramatically expands the currently accepted limits of druggability. WDB002, in complex with the FK506-binding protein (FKBP12), potently and selectively binds the human centrosomal protein 250 (CEP250), resulting in disruption of CEP250 function in cells. The recognition mode is unprecedented in that the targeted domain of CEP250 is a coiled coil and is topologically featureless, embodying both a structural motif and surface topology previously considered on the extreme limits of "undruggability" for an intracellular target. Structural studies reveal extensive protein-WDB002 and protein-protein contacts, with the latter being distinct from those seen in FKBP12 ternary complexes formed by FK506 and rapamycin. Outward-facing structural changes in a bound small molecule can thus reprogram FKBP12 to engage diverse, otherwise "undruggable" targets. The flat-targeting modality demonstrated here has the potential to expand the druggable target range of small-molecule therapeutics. As CEP250 was recently found to be an interaction partner with the Nsp13 protein of the SARS-CoV-2 virus that causes COVID-19 disease, it is possible that WDB002 or an analog may exert useful antiviral activity through its ability to form high-affinity ternary complexes containing CEP250 and FKBP12.


Subject(s)
Actinobacteria/genetics , Antiviral Agents/pharmacology , Genome, Bacterial , Macrolides/pharmacology , Protein Interaction Domains and Motifs/drug effects , Small Molecule Libraries/pharmacology , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus Binding Protein 1A/metabolism , Actinobacteria/metabolism , Amino Acid Sequence , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Calcineurin/genetics , Calcineurin/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Evolution, Molecular , HEK293 Cells , Humans , Macrolides/chemistry , Macrolides/metabolism , Models, Molecular , Protein Conformation , Sequence Homology , Sirolimus/chemistry , Sirolimus/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
2.
Appl Microbiol Biotechnol ; 102(5): 2337-2350, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29396588

ABSTRACT

We used a temperature differential assay with the opportunistic fungal pathogen Cryptococcus neoformans as a simple screening platform to detect small molecules with antifungal activity in natural product extracts. By screening of a collection extracts from two different strains of the coprophilous fungus, Amphichorda felina, we detected strong, temperature-dependent antifungal activity using a two-plate agar zone of inhibition assay at 25 and 37 °C. Bioassay-guided fractionation of the crude extract followed by liquid chromatography-mass spectrometry (LC-MS) and nuclear magnetic resonance spectroscopy (NMR) identified cyclosporin C (CsC) as the main component of the crude extract responsible for growth inhibition of C. neoformans at 37 °C. The presence of CsC was confirmed by comparison with a commercial standard. We sequenced the genome of A. felina to identify and annotate the CsC biosynthetic gene cluster. The only previously characterized gene cluster for the biosynthesis of similar compounds is that of the related immunosuppressant drug cyclosporine A (CsA). The CsA and CsC gene clusters share a high degree of synteny and sequence similarity. Amino acid changes in the adenylation domain of the CsC nonribosomal peptide synthase's sixth module may be responsible for the substitution of L-threonine compared to L-α-aminobutyric acid in the CsA peptide core. This screening strategy promises to yield additional antifungal natural products with a focused spectrum of antimicrobial activity.


Subject(s)
Antifungal Agents/pharmacology , Cryptococcus neoformans/drug effects , Cyclosporins/pharmacology , Hypocreales/chemistry , Antifungal Agents/chemistry , Antifungal Agents/metabolism , Cryptococcus neoformans/growth & development , Cyclosporins/chemistry , Cyclosporins/metabolism , Hypocreales/genetics , Hypocreales/metabolism , Temperature
3.
Proc Natl Acad Sci U S A ; 109(44): 17942-7, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23071338

ABSTRACT

Aberrant activation of signaling by the Wnt pathway is strongly implicated in the onset and progression of numerous types of cancer. Owing to the persistent dependence of these tumors on Wnt signaling for growth and survival, inhibition of this pathway is considered an attractive mechanism-based therapeutic approach. Oncogenic activation of Wnt signaling can ensue from a variety of distinct aberrations in the signaling pathway, but most share the common feature of causing increased cellular levels of ß-catenin by interfering with its constitutive degradation. ß-Catenin serves as a central hub in Wnt signaling by engaging in crucial protein-protein interactions with both negative and positive effectors of the pathway. Direct interference with these protein-protein interactions is a biologically compelling approach toward suppression of ß-catenin hyperactivity, but such interactions have proven intransigent with respect to small-molecule targeting. Hence ß-catenin remains an elusive target for translational cancer therapy. Here we report the discovery of a hydrocarbon-stapled peptide that directly targets ß-catenin and interferes with its ability to serve as a transcriptional coactivator for T-cell factor (TCF) proteins, the downstream transcriptional regulators of the Wnt pathway.


Subject(s)
Oncogenes , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Amino Acid Sequence , HeLa Cells , Humans , Molecular Sequence Data , Transcription, Genetic
4.
J Biol Chem ; 287(30): 24916-28, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22511791

ABSTRACT

A poorly understood aspect of DNA repair proteins is their ability to identify exceedingly rare sites of damage embedded in a large excess of nearly identical undamaged DNA, while catalyzing repair only at the damaged sites. Progress toward understanding this problem has been made by comparing the structures and biochemical behavior of these enzymes when they are presented with either a target lesion or a corresponding undamaged nucleobase. Trapping and analyzing such DNA-protein complexes is particularly difficult in the case of base extrusion DNA repair proteins because of the complexity of the repair reaction, which involves extrusion of the target base from DNA followed by its insertion into the active site where glycosidic bond cleavage is catalyzed. Here we report the structure of a human 8-oxoguanine (oxoG) DNA glycosylase, hOGG1, in which a normal guanine from DNA has been forcibly inserted into the enzyme active site. Although the interactions of the nucleobase with the active site are only subtly different for G versus oxoG, hOGG1 fails to catalyze excision of the normal nucleobase. This study demonstrates that even if hOGG1 mistakenly inserts a normal base into its active site, the enzyme can still reject it on the basis of catalytic incompatibility.


Subject(s)
DNA Glycosylases/chemistry , DNA/chemistry , Guanine/analogs & derivatives , Catalytic Domain , DNA/genetics , DNA/metabolism , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA Repair/physiology , Guanine/chemistry , Guanine/metabolism , Humans , Substrate Specificity/physiology
5.
FASEB J ; 25(3): 907-15, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21106936

ABSTRACT

By an unknown mechanism, ß-thymosins are extracellular modulators of angiogenesis, inflammation, wound healing, and development. We were interested in identifying ß-thymosin interactors and determining their importance in ß-thymosins signaling in human vein endothelial cells (HUVECs). We performed pulldown experiments with biotinylated thymosin ß-4 (Tß4) in comparison to neutravidin beads alone and used mass spectrometric analysis to identify differentially interacting proteins. By this method, we identified F1-F0 ATP synthase, a known target of antiangiogenic angiostatin. By surface plasmon resonance, we determined for Tß4 binding to the ß subunit of ATP synthase a K(D) of 12 nM. Blocking antibodies and antagonists (oligomycin, IC(50) ∼1.8 µM; piceatannol, IC(50) ∼1.05 µM; and angiostatin, IC(50) ∼2.9 µg/ml) of ATP synthase inhibited the Tß4-induced increase in cell surface ATP levels, as measured by luciferase assay, and the Tß4-induced increase in HUVEC migration, as measured by transwell migration assay. Silencing of the ATP-responsive purinergic receptor P2X4 with siRNA also blocked Tß4-induced HUVEC migration in a transwell assay. Furthermore, in silico we identified common amphiphilic α-helical structural similarities between ß-thymosins and the inhibitory factor 1 (IF1), an inhibitor of ATP synthase hydrolysis. In summary, we have identified an extracellular signaling pathway where Tß4 increases cell surface ATP levels via ATP synthase and have shown further that ATP-responsive P2X4 receptor is required for Tß4-induced HUVEC migration.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , Receptors, Purinergic P2X4/metabolism , Signal Transduction/physiology , Thymosin/metabolism , Amino Acid Sequence , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/cytology , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Proteins/metabolism , Proton-Translocating ATPases/metabolism , Thymosin/chemistry , Thymosin/genetics , Umbilical Veins/cytology , Wound Healing/physiology , ATPase Inhibitory Protein
6.
J Biol Chem ; 285(46): 35783-91, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20843803

ABSTRACT

Because DNA damage is so rare, DNA glycosylases interact for the most part with undamaged DNA. Whereas the structural basis for recognition of DNA lesions by glycosylases has been studied extensively, less is known about the nature of the interaction between these proteins and undamaged DNA. Here we report the crystal structures of the DNA glycosylase AlkA in complex with undamaged DNA. The structures revealed a recognition mode in which the DNA is nearly straight, with no amino acid side chains inserted into the duplex, and the target base pair is fully intrahelical. A comparison of the present structures with that of AlkA recognizing an extrahelical lesion revealed conformational changes in both the DNA and protein as the glycosylase transitions from the interrogation of undamaged DNA to catalysis of nucleobase excision. Modeling studies with the cytotoxic lesion 3-methyladenine and accompanying biochemical experiments suggested that AlkA actively interrogates the minor groove of the DNA while probing for the presence of lesions.


Subject(s)
DNA Glycosylases/chemistry , DNA, Bacterial/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/metabolism , Binding Sites , Crystallography, X-Ray , DNA Damage , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Hydrogen Bonding , Models, Molecular , Molecular Structure , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , Transition Temperature
7.
Structure ; 16(8): 1166-74, 2008 Aug 06.
Article in English | MEDLINE | ID: mdl-18682218

ABSTRACT

The constant attack on DNA by endogenous and exogenous agents gives rise to nucleobase modifications that cause mutations, which can lead to cancer. Visualizing the effects of these lesions on the structure of duplex DNA is key to understanding their biologic consequences. The most definitive method of obtaining such structures, X-ray crystallography, is troublesome to employ owing to the difficulty of obtaining diffraction-quality crystals of DNA. Here, we present a crystallization system that uses a protein, the DNA glycosylase AlkA, as a scaffold to mediate the crystallization of lesion-containing duplex DNA. We demonstrate the use of this system to facilitate the rapid structure determination of DNA containing the lesion 8-oxoguanine in several different sequence contexts, and also deoxyinosine and 1,N(6)-ethenoadenine, each stabilized as the corresponding 2'-flouro analog. The structures of 8-oxoguanine provide a correct atomic-level view of this important endogenous lesion in DNA.


Subject(s)
Crystallography, X-Ray/methods , DNA Damage , DNA Glycosylases/chemistry , DNA/chemistry , Escherichia coli/enzymology , Nucleic Acid Conformation , Protein Conformation , Base Sequence , DNA/metabolism , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , Models, Molecular , Molecular Sequence Data , Nucleotides/chemistry , Nucleotides/metabolism
8.
J Am Chem Soc ; 130(35): 11570-1, 2008 Sep 03.
Article in English | MEDLINE | ID: mdl-18686953

ABSTRACT

The predominant product of aberrant DNA methylation is the genotoxic lesion N7-methyl-2'-deoxyguanosine (m7dG). M7dG is recognized and excised by lesion-specific DNA glycosylases, namely AlkA in E. coli and Aag in humans. Structural studies of m7dG recognition and catalysis by these enzymes have been hampered due to a lack of efficient means by which to incorporate the chemically labile m7dG moiety site-specifically into DNA on a preparative scale. Here we report a solution to this problem. We stabilized the lesion toward acid-catalyzed and glycosylase-catalyzed depurination by 2'-fluorination and toward base-catalyzed degradation using mild, nonaqueous conditions in the DNA deprotection reaction. Duplex DNA containing 2'-fluoro-m7dG (Fm7dG) cocrystallized with AlkA as a host-guest complex in which the lesion-containing segment of DNA was nearly devoid of protein contacts, thus enabling the first direct visualization of the N7-methylguanine lesion nucleobase in DNA. The structure reveals that the base-pairing mode of Fm7dG:C is nearly identical to that of G:C, and Fm7dG does not induce any apparent structural disturbance of the duplex structure. These observations suggest that AlkA and Aag must perform a structurally invasive interrogation of DNA in order to detect the presence of intrahelical m7dG lesions.


Subject(s)
DNA/chemistry , Guanine/analogs & derivatives , Base Pairing , DNA/chemical synthesis , DNA/genetics , Guanine/chemistry , Models, Molecular , Nucleic Acid Conformation
9.
Structure ; 14(1): 151-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16407074

ABSTRACT

The ubiquitous MRG/MORF family of proteins is involved in cell senescence, or the terminal loss of proliferative potential, a model for aging and tumor suppression at the cellular level. These proteins are defined by the approximately 20 kDa MRG domain that binds a plethora of transcriptional regulators and chromatin-remodeling factors, including the histone deacetylase transcriptional corepressor mSin3A and the novel nuclear protein PAM14, and they are also known components of the Tip60/NuA4 complex via interactions with the MRG binding protein (MRGBP). We present here the crystal structure of a prototypic MRG domain from human MRG15 whose core consists of two orthogonal helix hairpins. Despite the lack of sequence similarity, the core structure has surprisingly striking homology to a DNA-interacting domain of the tyrosine site-specific recombinases XerD, lambda integrase, and Cre. Site-directed mutagenesis studies based on the X-ray structure and bioinformatics identified key residues involved in the binding of PAM14 and MRGBP.


Subject(s)
Cell Proliferation , Cellular Senescence/physiology , DNA-Binding Proteins/chemistry , Structural Homology, Protein , Transcription Factors/chemistry , Amino Acid Sequence , Binding Sites , Carrier Proteins/metabolism , Crystallography, X-Ray , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Evolution, Molecular , Histone Acetyltransferases , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Protein Interaction Mapping , Protein Structure, Tertiary/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
10.
J Mol Biol ; 344(2): 303-10, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15522285

ABSTRACT

Nuclear transport carriers interact with proteins of the nuclear pore complex (NPC) to transport their cargo across the nuclear envelope. One such carrier is nuclear transport factor 2 (NTF2), whose import cargo is the small GTPase Ran. A domain highly homologous to the small NTF2 protein (14kDa) is also found in a number of additional proteins, which together make up the NTF2 domain containing superfamily of proteins. Using structural, computational and biochemical analysis we have identified a functional site that is present throughout this superfamily, and our results indicate that this site functions as an NPC binding site in NTF2. Previously we showed that a D23A mutant of NTF2 exhibits increased affinity for the NPC. The mechanism of this mutation, however, was unknown as this region of NTF2 had not been implicated in binding to NPC proteins. Here we show that the D23A mutation in NTF2 does not result in gross structural changes affecting other known NPC binding sites. Instead, the D23 residue is located in an evolutionarily important region in the NTF2 domain containing superfamily, that in NTF2, is involved in binding to the NPC.


Subject(s)
Cell Nucleus/metabolism , Computational Biology , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/chemistry , Nucleocytoplasmic Transport Proteins/metabolism , Active Transport, Cell Nucleus , Alanine/metabolism , Amino Acid Substitution , Binding Sites , Crystallography, X-Ray , Dimerization , Escherichia coli/genetics , Evolution, Molecular , HeLa Cells , Humans , Hydrogen Bonding , Models, Molecular , Nucleocytoplasmic Transport Proteins/genetics , Point Mutation , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , ran GTP-Binding Protein/metabolism
11.
J Mol Biol ; 331(2): 447-56, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12888351

ABSTRACT

The dispositions of 39 alpha helices of greater than 2.5 turns and four beta sheets in the major capsid protein (VP5, 149 kDa) of herpes simplex virus type 1 were identified by computational and visualization analysis from the 8.5A electron cryomicroscopy structure of the whole capsid. The assignment of helices in the VP5 upper domain was validated by comparison with the recently determined crystal structure of this region. Analysis of the spatial arrangement of helices in the middle domain of VP5 revealed that the organization of a tightly associated bundle of ten helices closely resembled that of a domain fold found in the annexin family of proteins. Structure-based sequence searches suggested that sequences in both the N and C-terminal portions of the VP5 sequence contribute to this domain. The long helices seen in the floor domain of VP5 form an interconnected network within and across capsomeres. The combined structural and sequence-based informatics has led to an architectural model of VP5. This model placed in the context of the capsid provides insights into the strategies used to achieve viral capsid stability.


Subject(s)
Capsid Proteins/chemistry , Simplexvirus/chemistry , Amino Acid Motifs , Amino Acid Sequence , Annexins/chemistry , Computational Biology , Cryoelectron Microscopy , Crystallography, X-Ray , Databases as Topic , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Software
12.
Mol Cell ; 29(1): 122-33, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-18158267

ABSTRACT

The nucleotide excision repair pathway corrects many structurally unrelated DNA lesions. Damage recognition in bacteria is performed by UvrA, a member of the ABC ATPase superfamily whose functional form is a dimer with four nucleotide-binding domains (NBDs), two per protomer. In the 3.2 A structure of UvrA from Bacillus stearothermophilus, we observe that the nucleotide-binding sites are formed in an intramolecular fashion and are not at the dimer interface as is typically found in other ABC ATPases. UvrA also harbors two unique domains; we show that one of these is required for interaction with UvrB, its partner in lesion recognition. In addition, UvrA contains three zinc modules, the number and ligand sphere of which differ from previously published models. Structural analysis, biochemical experiments, surface electrostatics, and sequence conservation form the basis for models of ATP-modulated dimerization, UvrA-UvrB interaction, and DNA binding during the search for lesions.


Subject(s)
Adenosine Triphosphatases/chemistry , Bacterial Proteins/chemistry , Crystallography, X-Ray , DNA Helicases/chemistry , DNA Repair , DNA-Binding Proteins/chemistry , Endodeoxyribonucleases/chemistry , Geobacillus stearothermophilus/enzymology , Protein Interaction Mapping , Adenosine Diphosphate/chemistry , Adenosine Triphosphatases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Bacterial Proteins/metabolism , Binding Sites , Conserved Sequence , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Endodeoxyribonucleases/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Static Electricity , Zinc Fingers
13.
Mol Cell ; 22(3): 339-49, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16678106

ABSTRACT

Polyadenylation of mRNAs in poxviruses, crucial for virion maturation, is carried out by a poly(A) polymerase heterodimer composed of a catalytic component, VP55, and a processivity factor, VP39. The ATP-gamma-S bound and unbound crystal structures of the vaccinia polymerase reveal an unusual architecture for VP55 that comprises of N-terminal, central or catalytic, and C-terminal domains with different topologies and that differs from many polymerases, including the eukaryotic poly(A) polymerases. Residues in the active site of VP55, located between the catalytic and C-terminal domains, make specific interactions with the adenine of the ATP analog, establishing the molecular basis of ATP recognition. VP55's concave surface docks the globular VP39. A model for RNA primer binding that involves all three VP55 domains and VP39 is proposed. The model supports biochemical evidence that VP39 functions as a processivity factor by partially enclosing the RNA primer at the heterodimer interface.


Subject(s)
Adenosine Triphosphate/metabolism , Polynucleotide Adenylyltransferase/chemistry , Vaccinia virus/enzymology , Binding Sites/genetics , Crystallography, X-Ray , Dimerization , Models, Molecular , Protein Binding , Protein Folding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Transport , RNA, Viral/metabolism
14.
J Virol ; 80(13): 6612-20, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16775348

ABSTRACT

Coronaviruses cause a variety of respiratory and enteric diseases in animals and humans including severe acute respiratory syndrome. In these enveloped viruses, the filamentous nucleocapsid is formed by the association of nucleocapsid (N) protein with single-stranded viral RNA. The N protein is a highly immunogenic phosphoprotein also implicated in viral genome replication and in modulating cell signaling pathways. We describe the structure of the two proteolytically resistant domains of the N protein from infectious bronchitis virus (IBV), a prototype coronavirus. These domains are located at its N- and C-terminal ends (NTD and CTD, respectively). The NTD of the IBV Gray strain at 1.3-A resolution exhibits a U-shaped structure, with two arms rich in basic residues, providing a module for specific interaction with RNA. The CTD forms a tightly intertwined dimer with an intermolecular four-stranded central beta-sheet platform flanked by alpha helices, indicating that the basic building block for coronavirus nucleocapsid formation is a dimeric assembly of N protein. The variety of quaternary arrangements of the NTD and CTD revealed by the analysis of the different crystal forms delineates possible interfaces that could be used for the formation of a flexible filamentous ribonucleocapsid. The striking similarity between the dimeric structure of CTD and the nucleocapsid-forming domain of a distantly related arterivirus indicates a conserved mechanism of nucleocapsid formation for these two viral families.


Subject(s)
Coronavirus/chemistry , Nucleocapsid Proteins/chemistry , Nucleocapsid/chemistry , Virus Assembly , Animals , Coronavirus/metabolism , Coronavirus Infections/metabolism , Coronavirus Nucleocapsid Proteins , Crystallography, X-Ray , Dimerization , Humans , Nucleocapsid/metabolism , Nucleocapsid Proteins/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Viral/chemistry , RNA, Viral/metabolism , Structure-Activity Relationship
15.
J Virol ; 80(5): 2309-17, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16474137

ABSTRACT

Herpesviruses replicate their double stranded DNA genomes as high-molecular-weight concatemers which are subsequently cleaved into unit-length genomes by a complex mechanism that is tightly coupled to DNA insertion into a preformed capsid structure, the procapsid. The herpes simplex virus type 1 UL25 protein is incorporated into the capsid during DNA packaging, and previous studies of a null mutant have demonstrated that its function is essential at the late stages of the head-filling process, either to allow packaging to proceed to completion or for retention of the viral genome within the capsid. We have expressed and purified an N-terminally truncated form of the 580-residue UL25 protein and have determined the crystallographic structure of the region corresponding to amino acids 134 to 580 at 2.1-Angstroms resolution. This structure, the first for any herpesvirus protein involved in processing and packaging of viral DNA, reveals a novel fold, a distinctive electrostatic distribution, and a unique "flexible" architecture in which numerous flexible loops emanate from a stable core. Evolutionary trace analysis of UL25 and its homologues in other herpesviruses was used to locate potentially important amino acids on the surface of the protein, leading to the identification of four putative docking regions for protein partners.


Subject(s)
Herpesvirus 1, Human/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , DNA Packaging , Herpesvirus 1, Human/genetics , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Conformation , Sequence Homology, Amino Acid , Viral Proteins/genetics
16.
EMBO J ; 22(4): 757-65, 2003 Feb 17.
Article in English | MEDLINE | ID: mdl-12574112

ABSTRACT

Herpes simplex virus-1 (HSV-1) virions are large, complex enveloped particles containing a proteinaceous tegument layer connected to an icosahedral capsid. The major capsid protein, VP5 (149 kDa), makes up both types of capsomere, pentons and hexons. Limited trypsin digestion of VP5 identified a single stable 65 kDa fragment which represents a proposed protein folding nucleus. We report the 2.9 A crystal structure of this fragment and its modeling into an 8.5 A resolution electron cryomicroscopy map of the HSV-1 capsid. The structure, the first for any capsid protein from Herpesviridae, revealed a novel fold, placing herpesviruses outside any of the structurally linked viral groupings. Alterations in the geometrical arrangements of the VP5 subunits in the capsomeres exposes different residues, resulting in the differential association of the tegument and VP26 with the pentons and hexons, respectively. The rearrangements of VP5 subunits required to form both pentavalent and hexavalent capsomeres result in structures that exhibit very different electrostatic properties. These differences may mediate the binding and release of other structural proteins during capsid maturation.


Subject(s)
Capsid Proteins/chemistry , Herpesvirus 1, Human/chemistry , Capsid Proteins/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Herpesvirus 1, Human/metabolism , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL