Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Proc Natl Acad Sci U S A ; 114(3): E386-E395, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28049853

ABSTRACT

Recoding viral genomes by numerous synonymous but suboptimal substitutions provides live attenuated vaccine candidates. These vaccine candidates should have a low risk of deattenuation because of the many changes involved. However, their genetic stability under selective pressure is largely unknown. We evaluated phenotypic reversion of deoptimized human respiratory syncytial virus (RSV) vaccine candidates in the context of strong selective pressure. Codon pair deoptimized (CPD) versions of RSV were attenuated and temperature-sensitive. During serial passage at progressively increasing temperature, a CPD RSV containing 2,692 synonymous mutations in 9 of 11 ORFs did not lose temperature sensitivity, remained genetically stable, and was restricted at temperatures of 34 °C/35 °C and above. However, a CPD RSV containing 1,378 synonymous mutations solely in the polymerase L ORF quickly lost substantial attenuation. Comprehensive sequence analysis of virus populations identified many different potentially deattenuating mutations in the L ORF as well as, surprisingly, many appearing in other ORFs. Phenotypic analysis revealed that either of two competing mutations in the virus transcription antitermination factor M2-1, outside of the CPD area, substantially reversed defective transcription of the CPD L gene and substantially restored virus fitness in vitro and in case of one of these two mutations, also in vivo. Paradoxically, the introduction into Min L of one mutation each in the M2-1, N, P, and L proteins resulted in a virus with increased attenuation in vivo but increased immunogenicity. Thus, in addition to providing insights on the adaptability of genome-scale deoptimized RNA viruses, stability studies can yield improved synthetic RNA virus vaccine candidates.


Subject(s)
Genome, Viral/genetics , RNA Viruses/genetics , Viral Vaccines/genetics , Animals , Cell Line , Chlorocebus aethiops/genetics , Codon/genetics , Humans , Mice , Mice, Inbred BALB C , Mutation/genetics , Open Reading Frames/genetics , Respiratory Syncytial Virus, Human , Vaccines, Attenuated/genetics , Vaccines, Synthetic/genetics , Vero Cells , Viral Proteins/genetics , Virus Replication/genetics
2.
J Virol ; 92(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29976658

ABSTRACT

Host-influenza virus interplay at the transcript level has been extensively characterized in epithelial cells. Yet, there are no studies that simultaneously characterize human host and influenza A virus (IAV) genomes. We infected human bronchial epithelial BEAS-2B cells with two seasonal IAV/H3N2 strains, Brisbane/10/07 and Perth/16/09 (reference strains for past vaccine seasons) and the well-characterized laboratory strain Udorn/307/72. Strand-specific RNA sequencing (RNA-seq) of the infected BEAS-2B cells allowed for simultaneous analysis of host and viral transcriptomes, in addition to pathogen genomes, to reveal changes in mRNA expression and alternative splicing (AS). In general, patterns of global and immune gene expression induced by the three IAVs were mostly shared. However, AS of host transcripts and small nuclear RNAs differed between the seasonal and laboratory strains. Analysis of viral transcriptomes showed deletions of the polymerase components (defective interfering-like RNAs) within the genome. Surprisingly, we found that the neuraminidase gene undergoes AS and that the splicing event differs between seasonal and laboratory strains. Our findings reveal novel elements of the host-virus interaction and highlight the importance of RNA-seq in identifying molecular changes at the genome level that may contribute to shaping RNA-based innate immunity.IMPORTANCE The use of massively parallel RNA sequencing (RNA-seq) has revealed insights into human and pathogen genomes and their evolution. Dual RNA-seq allows simultaneous dissection of host and pathogen genomes and strand-specific RNA-seq provides information about the polarity of the RNA. This is important in the case of negative-strand RNA viruses like influenza virus, which generate positive (complementary and mRNA) and negative-strand RNAs (genome) that differ in their potential to trigger innate immunity. Here, we characterize interactions between human bronchial epithelial cells and three influenza A/H3N2 strains using strand-specific dual RNA-seq. We focused on this subtype because of its epidemiological importance in causing significant morbidity and mortality during influenza epidemics. We report novel elements that differ between seasonal and laboratory strains highlighting the complexity of the host-virus interplay at the RNA level.


Subject(s)
Genome, Human/genetics , Genome, Viral/genetics , Host-Pathogen Interactions/genetics , Immunity, Innate/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/immunology , Bronchi/cytology , Bronchi/virology , Epithelial Cells/virology , Gene Expression Profiling , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Host-Pathogen Interactions/immunology , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/virology , Neuraminidase/genetics , RNA Splicing/genetics , Seasons , Sequence Analysis, RNA/methods
3.
Proc Natl Acad Sci U S A ; 113(8): E1006-15, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26869717

ABSTRACT

Peptide loading of major histocompatibility complex class I (MHC-I) molecules is central to antigen presentation, self-tolerance, and CD8(+) T-cell activation. TAP binding protein, related (TAPBPR), a widely expressed tapasin homolog, is not part of the classical MHC-I peptide-loading complex (PLC). Using recombinant MHC-I molecules, we show that TAPBPR binds HLA-A*02:01 and several other MHC-I molecules that are either peptide-free or loaded with low-affinity peptides. Fluorescence polarization experiments establish that TAPBPR augments peptide binding by MHC-I. The TAPBPR/MHC-I interaction is reversed by specific peptides, related to their affinity. Mutational and small-angle X-ray scattering (SAXS) studies confirm the structural similarities of TAPBPR with tapasin. These results support a role of TAPBPR in stabilizing peptide-receptive conformation(s) of MHC-I, permitting peptide editing.


Subject(s)
Antigen Presentation , HLA-A2 Antigen/immunology , Immunoglobulins/immunology , Membrane Proteins/immunology , Peptides/immunology , Animals , Cell Line , Drosophila melanogaster , HLA-A2 Antigen/genetics , Humans , Immunoglobulins/genetics , Membrane Proteins/genetics , Peptides/genetics
4.
J Virol ; 91(21)2017 11 01.
Article in English | MEDLINE | ID: mdl-28794032

ABSTRACT

Ecotropic, xenotropic, and polytropic mouse leukemia viruses (E-, X-, and P-MLVs) exist in mice as infectious viruses and endogenous retroviruses (ERVs) inserted into mouse chromosomes. All three MLV subgroups are linked to leukemogenesis, which involves generation of recombinants with polytropic host range. Although P-MLVs are deemed to be the proximal agents of disease induction, few biologically characterized infectious P-MLVs have been sequenced for comparative analysis. We analyzed the complete genomes of 16 naturally occurring infectious P-MLVs, 12 of which were typed for pathogenic potential. We sought to identify ERV progenitors, recombinational hot spots, and segments that are always replaced, never replaced, or linked to pathogenesis or host range. Each P-MLV has an E-MLV backbone with P- or X-ERV replacements that together cover 100% of the recombinant genomes, with different substitution patterns for X- and P-ERVs. Two segments are always replaced, both coding for envelope (Env) protein segments: the N terminus of the surface subunit and the cytoplasmic tail R peptide. Viral gag gene replacements are influenced by host restriction genes Fv1 and Apobec3 Pathogenic potential maps to the env transmembrane subunit segment encoding the N-heptad repeat (HR1). Molecular dynamics simulations identified three novel interdomain salt bridges in the lymphomagenic virus HR1 that could affect structural stability, entry or sensitivity to host immune responses. The long terminal repeats of lymphomagenic P-MLVs are differentially altered by recombinations, duplications, or mutations. This analysis of the naturally occurring, sometimes pathogenic P-MLV recombinants defines the limits and extent of intersubgroup recombination and identifies specific sequence changes linked to pathogenesis and host interactions.IMPORTANCE During virus-induced leukemogenesis, ecotropic mouse leukemia viruses (MLVs) recombine with nonecotropic endogenous retroviruses (ERVs) to produce polytropic MLVs (P-MLVs). Analysis of 16 P-MLV genomes identified two segments consistently replaced: one at the envelope N terminus that alters receptor choice and one in the R peptide at the envelope C terminus, which is removed during virus assembly. Genome-wide analysis shows that nonecotropic replacements in the progenitor ecotropic MLV genome are more extensive than previously appreciated, covering 100% of the genome; contributions from xenotropic and polytropic ERVs differentially alter the regions responsible for receptor determination or subject to APOBEC3 and Fv1 restriction. All pathogenic viruses had modifications in the regulatory elements in their long terminal repeats and differed in a helical segment of envelope involved in entry and targeted by the host immune system. Virus-induced leukemogenesis thus involves generation of complex recombinants, and specific replacements are linked to pathogenesis and host restrictions.


Subject(s)
Host Specificity/genetics , Leukemia Virus, Murine/classification , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/virology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Viral Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Evolution, Molecular , Genome, Viral , Leukemia Virus, Murine/genetics , Mice , Molecular Dynamics Simulation , Protein Conformation , Receptors, Virus/genetics , Receptors, Virus/metabolism , Sequence Homology , Terminal Repeat Sequences , Viral Proteins/chemistry , Viral Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 111(8): 3152-7, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24569807

ABSTRACT

Elicitation of broadly neutralizing antibodies is essential for the development of a protective vaccine against HIV-1. However, the native HIV-1 envelope adopts a protected conformation that conceals highly conserved sites of vulnerability from antibody recognition. Although high-definition structures of the monomeric core of the envelope glycoprotein subunit gp120 and, more recently, of a stabilized soluble gp140 trimer have been solved, fundamental aspects related to the conformation and function of the native envelope remain unresolved. Here, we show that the conserved central region of the second variable loop (V2) of gp120 contains sulfated tyrosines (Tys173 and Tys177) that in the CD4-unbound prefusion state mediate intramolecular interaction between V2 and the conserved base of the third variable loop (V3), functionally mimicking sulfated tyrosines in CCR5 and anti-coreceptor-binding-site antibodies such as 412d. Recombinant gp120 expressed in continuous cell lines displays low constitutive levels of V2 tyrosine sulfation, which can be enhanced markedly by overexpression of the tyrosyl sulfotransferase TPST2. In contrast, virion-associated gp120 produced by primary CD4(+) T cells is inherently highly sulfated. Consistent with a functional role of the V2 sulfotyrosines, enhancement of tyrosine sulfation decreased binding and neutralization of HIV-1 BaL by monomeric soluble CD4, 412d, and anti-V3 antibodies and increased recognition by the trimer-preferring antibodies PG9, PG16, CH01, and PGT145. Conversely, inhibition of tyrosine sulfation increased sensitivity to soluble CD4, 412d, and anti-V3 antibodies and diminished recognition by trimer-preferring antibodies. These results identify the sulfotyrosine-mediated V2-V3 interaction as a critical constraint that stabilizes the native HIV-1 envelope trimer and modulates its sensitivity to neutralization.


Subject(s)
HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/metabolism , HIV-1/genetics , HIV-1/immunology , Protein Conformation , Tyrosine/analogs & derivatives , Blotting, Western , Flow Cytometry , HEK293 Cells , HIV Envelope Protein gp120/genetics , Humans , Neutralization Tests , Surface Plasmon Resonance , Tyrosine/metabolism
6.
J Biol Chem ; 290(2): 1119-28, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25416779

ABSTRACT

The structure of the infectious form of prion protein, PrP(Sc), remains unclear. Most pure recombinant prion protein (PrP) amyloids generated in vitro are not infectious and lack the extent of the protease-resistant core and solvent exclusion of infectious PrP(Sc), especially within residues ∼90-160. Polyanionic cofactors can enhance infectivity and PrP(Sc)-like characteristics of such fibrils, but the mechanism of this enhancement is unknown. In considering structural models of PrP(Sc) multimers, we identified an obstacle to tight packing that might be overcome with polyanionic cofactors, namely, electrostatic repulsion between four closely spaced cationic lysines within a central lysine cluster of residues 101-110. For example, in our parallel in-register intermolecular ß-sheet model of PrP(Sc), not only would these lysines be clustered within the 101-110 region of the primary sequence, but they would have intermolecular spacings of only ∼4.8 Å between stacked ß-strands. We have now performed molecular dynamics simulations predicting that neutralization of the charges on these lysine residues would allow more stable parallel in-register packing in this region. We also show empirically that substitution of these clustered lysine residues with alanines or asparagines results in recombinant PrP amyloid fibrils with extended proteinase-K resistant ß-sheet cores and infrared spectra that are more reminiscent of bona fide PrP(Sc). These findings indicate that charge neutralization at the central lysine cluster is critical for the folding and tight packing of N-proximal residues within PrP amyloid fibrils. This charge neutralization may be a key aspect of the mechanism by which anionic cofactors promote PrP(Sc) formation.


Subject(s)
Amyloid/chemistry , Lysine/chemistry , PrPSc Proteins/chemistry , Prion Diseases/metabolism , Amyloid/ultrastructure , Animals , Humans , Mesocricetus , Molecular Dynamics Simulation , Mutagenesis , Polyelectrolytes , Polymers/chemistry , PrPSc Proteins/genetics , PrPSc Proteins/ultrastructure , Prion Diseases/etiology , Prion Diseases/pathology , Protein Conformation , Protein Folding , Protein Multimerization , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/ultrastructure , Static Electricity
7.
J Infect Dis ; 212(5): 684-93, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-25657255

ABSTRACT

BACKGROUND: Elimination of onchocerciasis and lymphatic filariasis is targeted for 2020. Given the coincident Loa loa infections in Central Africa and the potential for drug resistance development, the need for new microfilaricides and macrofilaricides has never been greater. With the genomes of L. loa, Onchocerca volvulus, Wuchereria bancrofti, and Brugia malayi available, new drug targets have been identified. METHODS: The effects of the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib on B. malayi adult males, adult females, L3 larvae, and microfilariae were assessed using a wide dose range (0-100 µM) in vitro. RESULTS: For microfilariae, median inhibitory concentrations (IC50 values) on day 6 were 6.06 µM for imatinib, 3.72 µM for dasatinib, and 81.35 µM for nilotinib; for L3 larvae, 11.27 µM, 13.64 µM, and 70.98 µM, respectively; for adult males, 41.6 µM, 3.87 µM, and 68.22 µM, respectively; and for adult females, 42.89 µM, 9.8 µM, and >100 µM, respectively. Three-dimensional modeling suggests how these tyrosine kinase inhibitors bind and inhibit filarial protein activity. CONCLUSIONS: Given the safety of imatinib in humans, plans are underway for pilot clinical trials to assess its efficacy in patients with filarial infections.


Subject(s)
Brugia malayi/drug effects , Filaricides/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Benzamides/pharmacology , Dasatinib , Female , Imatinib Mesylate , Inhibitory Concentration 50 , Larva/drug effects , Male , Piperazines/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology
8.
J Biol Chem ; 289(35): 24129-42, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25028516

ABSTRACT

Structures of the infectious form of prion protein (e.g. PrP(Sc) or PrP-Scrapie) remain poorly defined. The prevalent structural models of PrP(Sc) retain most of the native α-helices of the normal, noninfectious prion protein, cellular prion protein (PrP(C)), but evidence is accumulating that these helices are absent in PrP(Sc) amyloid. Moreover, recombinant PrP(C) can form amyloid fibrils in vitro that have parallel in-register intermolecular ß-sheet architectures in the domains originally occupied by helices 2 and 3. Here, we provide solid-state NMR evidence that the latter is also true of initially prion-seeded recombinant PrP amyloids formed in the absence of denaturants. These results, in the context of a primarily ß-sheet structure, led us to build detailed models of PrP amyloid based on parallel in-register architectures, fibrillar shapes and dimensions, and other available experimentally derived conformational constraints. Molecular dynamics simulations of PrP(90-231) octameric segments suggested that such linear fibrils, which are consistent with many features of PrP(Sc) fibrils, can have stable parallel in-register ß-sheet cores. These simulations revealed that the C-terminal residues ∼124-227 more readily adopt stable tightly packed structures than the N-terminal residues ∼90-123 in the absence of cofactors. Variations in the placement of turns and loops that link the ß-sheets could give rise to distinct prion strains capable of faithful template-driven propagation. Moreover, our modeling suggests that single PrP monomers can comprise the entire cross-section of fibrils that have previously been assumed to be pairs of laterally associated protofilaments. Together, these insights provide a new basis for deciphering mammalian prion structures.


Subject(s)
Amyloid/metabolism , Prions/metabolism , Carbon-13 Magnetic Resonance Spectroscopy , Disulfides/chemistry , Microscopy, Electron, Scanning Transmission , Models, Molecular , Polysaccharides/chemistry , Prions/chemistry , Proteolysis
9.
J Biol Chem ; 289(36): 25041-53, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25028517

ABSTRACT

The three isoforms of antigen 85 (A, B, and C) are the most abundant secreted mycobacterial proteins and catalyze transesterification reactions that synthesize mycolated arabinogalactan, trehalose monomycolate (TMM), and trehalose dimycolate (TDM), important constituents of the outermost layer of the cellular envelope of Mycobacterium tuberculosis. These three enzymes are nearly identical at the active site and have therefore been postulated to exist to evade host immunity. Distal to the active site is a second putative carbohydrate-binding site of lower homology. Mutagenesis of the three isoforms at this second site affected both substrate selectivity and overall catalytic activity in vitro. Using synthetic and natural substrates, we show that these three enzymes exhibit unique selectivity; antigen 85A more efficiently mycolates TMM to form TDM, whereas C (and to a lesser extent B) has a higher rate of activity using free trehalose to form TMM. This difference in substrate selectivity extends to the hexasaccharide fragment of cell wall arabinan. Mutation of secondary site residues from the most active isoform (C) into those present in A or B partially interconverts this substrate selectivity. These experiments in combination with molecular dynamics simulations reveal that differences in the N-terminal helix α9, the adjacent Pro(216)-Phe(228) loop, and helix α5 are the likely cause of changes in activity and substrate selectivity. These differences explain the existence of three isoforms and will allow for future work in developing inhibitors.


Subject(s)
Acyltransferases/metabolism , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Acyltransferases/chemistry , Acyltransferases/genetics , Amino Acid Sequence , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites/genetics , Biocatalysis , Carbohydrate Sequence , Catalytic Domain , Cell Wall/enzymology , Cell Wall/metabolism , Cord Factors/metabolism , Galactans/metabolism , Molecular Dynamics Simulation , Molecular Sequence Data , Mutation , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Polysaccharides/metabolism , Protein Binding , Protein Structure, Secondary , Sequence Homology, Amino Acid , Substrate Specificity
10.
PLoS Pathog ; 9(10): e1003677, 2013.
Article in English | MEDLINE | ID: mdl-24146620

ABSTRACT

Ebolavirus (EBOV), the causative agent of a severe hemorrhagic fever and a biosafety level 4 pathogen, increases its genome coding capacity by producing multiple transcripts encoding for structural and nonstructural glycoproteins from a single gene. This is achieved through RNA editing, during which non-template adenosine residues are incorporated into the EBOV mRNAs at an editing site encoding for 7 adenosine residues. However, the mechanism of EBOV RNA editing is currently not understood. In this study, we report for the first time that minigenomes containing the glycoprotein gene editing site can undergo RNA editing, thereby eliminating the requirement for a biosafety level 4 laboratory to study EBOV RNA editing. Using a newly developed dual-reporter minigenome, we have characterized the mechanism of EBOV RNA editing, and have identified cis-acting sequences that are required for editing, located between 9 nt upstream and 9 nt downstream of the editing site. Moreover, we show that a secondary structure in the upstream cis-acting sequence plays an important role in RNA editing. EBOV RNA editing is glycoprotein gene-specific, as a stretch encoding for 7 adenosine residues located in the viral polymerase gene did not serve as an editing site, most likely due to an absence of the necessary cis-acting sequences. Finally, the EBOV protein VP30 was identified as a trans-acting factor for RNA editing, constituting a novel function for this protein. Overall, our results provide novel insights into the RNA editing mechanism of EBOV, further understanding of which might result in novel intervention strategies against this viral pathogen.


Subject(s)
Ebolavirus/metabolism , RNA Editing/physiology , RNA, Messenger/metabolism , RNA, Viral/metabolism , Cell Line , Ebolavirus/genetics , Humans , Nucleic Acid Conformation , RNA, Messenger/genetics , RNA, Viral/genetics
11.
J Immunol ; 189(3): 1391-9, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22753930

ABSTRACT

MHC class I (MHC-I) proteins of the adaptive immune system require antigenic peptides for maintenance of mature conformation and immune function via specific recognition by MHC-I-restricted CD8(+) T lymphocytes. New MHC-I molecules in the endoplasmic reticulum are held by chaperones in a peptide-receptive (PR) transition state pending release by tightly binding peptides. In this study, we show, by crystallographic, docking, and molecular dynamics methods, dramatic movement of a hinged unit containing a conserved 3(10) helix that flips from an exposed "open" position in the PR transition state to a "closed" position with buried hydrophobic side chains in the peptide-loaded mature molecule. Crystallography of hinged unit residues 46-53 of murine H-2L(d) MHC-I H chain, complexed with mAb 64-3-7, demonstrates solvent exposure of these residues in the PR conformation. Docking and molecular dynamics predict how this segment moves to help form the A and B pockets crucial for the tight peptide binding needed for stability of the mature peptide-loaded conformation, chaperone dissociation, and Ag presentation.


Subject(s)
H-2 Antigens/metabolism , Molecular Dynamics Simulation , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , H-2 Antigens/chemistry , Histocompatibility Antigen H-2D , Humans , Ligands , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Structure-Activity Relationship , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/metabolism
12.
Blood ; 118(9): 2567-77, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21757613

ABSTRACT

IFNα, a cytokine with multiple functions in innate and adaptive immunity and a potent inhibitor of HIV, exerts antiviral activity, in part, by enhancing apolipoprotein B mRNA-editing enzyme-catalytic polypeptide-like 3 (APOBEC3) family members. Although IFNα therapy is associated with reduced viral burden, this cytokine also mediates immune dysfunction and toxicities. Through detailed mapping of IFNα receptor binding sites, we generated IFNα hybrids and mutants and determined that structural changes in the C-helix alter the ability of IFN to limit retroviral activity. Selective IFNα constructs differentially block HIV replication and their directional magnitude of inhibition correlates with APOBEC3 levels. Importantly, certain mutants exhibited reduced toxicity as reflected by induced indoleamine 2,3-dioxygenase (IDO), suggesting discreet and shared intracellular signaling pathways. Defining IFN structure and function relative to APOBEC and other antiviral genes may enable design of novel IFN-related molecules preserving beneficial antiviral roles while minimizing negative effects.


Subject(s)
Cytosine Deaminase/biosynthesis , HIV-1/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Interferon-alpha/chemistry , Macrophages/immunology , Virus Replication/physiology , APOBEC Deaminases , Calmodulin/physiology , Cytidine Deaminase , Cytosine Deaminase/genetics , Gene Expression Regulation , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-alpha/genetics , Interferon-alpha/physiology , Macrophages/virology , Models, Molecular , NF-kappa B/physiology , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Receptor, Interferon alpha-beta/chemistry , Receptor, Interferon alpha-beta/physiology , Recombinant Fusion Proteins/physiology , Sequence Homology, Amino Acid , Signal Transduction , Structure-Activity Relationship
13.
Antimicrob Agents Chemother ; 56(3): 1162-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22155829

ABSTRACT

Cryptococcus neoformans strains resistant to azoles due to mutations causing alterations in the ERG11 gene, encoding lanosterol 14α-demethylase, have rarely been reported. In this study, we have characterized a C. neoformans serotype A strain that is resistant to high concentrations of fluconazole (FLC). This strain, which was isolated from an FLC-treated patient, contained five missense mutations in the ERG11 gene compared to the sequence of reference strain H99. Molecular manipulations of the ERG11 gene coupled with susceptibility to triazole revealed that a single missense mutation resulting in the replacement of tyrosine by phenylalanine at amino acid 145 was sufficient to cause the high FLC resistance of the strain. Importantly, this newly identified point mutation in the ERG11 gene of C. neoformans afforded resistance to voriconazole (VRC) but increased susceptibility to itraconazole (ITC) and posaconazole (PSC), which are structurally similar to each other but distinct from FLC/VRC. The in vitro susceptibility/resistance of the strains with or without the missense mutation was reflected in the therapeutic efficacy of FLC versus ITC in the animals infected with the strains. This study shows the importance of the Y145F alteration of Erg11 in C. neoformans for manifestation of differential susceptibility toward different triazoles. It underscores the necessity of in vitro susceptibility testing for each FLC-resistant C. neoformans clinical isolate against different groups of azoles in order to assist patient management.


Subject(s)
Antifungal Agents/administration & dosage , Cryptococcosis/drug therapy , Cryptococcus neoformans/genetics , Drug Resistance, Fungal/genetics , Fungal Proteins/genetics , Sterol 14-Demethylase/genetics , 14-alpha Demethylase Inhibitors/administration & dosage , Animals , Cryptococcosis/microbiology , Cryptococcosis/mortality , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/enzymology , Drug Resistance, Fungal/drug effects , Female , Fluconazole/administration & dosage , Fungal Proteins/metabolism , Genotype , Humans , Itraconazole/administration & dosage , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mutation, Missense , Phenylalanine/genetics , Phenylalanine/metabolism , Pyrimidines/administration & dosage , Sterol 14-Demethylase/metabolism , Survival Rate , Triazoles/administration & dosage , Tyrosine/genetics , Tyrosine/metabolism , Voriconazole
14.
J Virol ; 85(6): 2512-23, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21228243

ABSTRACT

Cellular RNA interference (RNAi) provides a natural response against viral infection, but some viruses have evolved mechanisms to antagonize this form of antiviral immunity. To determine whether Ebolavirus (EBOV) counters RNAi by encoding suppressors of RNA silencing (SRSs), we screened all EBOV proteins using an RNAi assay initiated by exogenously delivered small interfering RNAs (siRNAs) against either an EBOV or a reporter gene. In addition to viral protein 35 (VP35), we found that VP30 and VP40 independently act as SRSs. Here, we present the molecular mechanisms of VP30 and VP35. VP30 interacts with Dicer independently of siRNA and with one Dicer partner, TRBP, only in the presence of siRNA. VP35 directly interacts with Dicer partners TRBP and PACT in an siRNA-independent fashion and in the absence of effects on interferon (IFN). Taken together, our findings elucidate a new mechanism of RNAi suppression that extends beyond the role of SRSs in double-stranded RNA (dsRNA) binding and IFN antagonism. The presence of three suppressors highlights the relevance of host RNAi-dependent antiviral immunity in EBOV infection and illustrates the importance of RNAi in shaping the evolution of RNA viruses.


Subject(s)
Ebolavirus/immunology , Immune Evasion , RNA Interference , RNA, Small Interfering/antagonists & inhibitors , Transcription Factors/metabolism , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Animals , Chlorocebus aethiops , Ebolavirus/pathogenicity , RNA, Small Interfering/metabolism , Vero Cells , Virulence Factors/metabolism
15.
PLoS Pathog ; 6: e1000974, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20617165

ABSTRACT

Mouse APOBEC3 (mA3) is a cytidine deaminase with antiviral activity. mA3 is linked to the Rfv3 virus resistance factor, a gene responsible for recovery from infection by Friend murine leukemia virus, and mA3 allelic variants differ in their ability to restrict mouse mammary tumor virus. We sequenced mA3 genes from 38 inbred strains and wild mouse species, and compared the mouse sequence and predicted structure with human APOBEC3G (hA3G). An inserted sequence was identified in the virus restrictive C57BL strain allele that disrupts a splice donor site. This insertion represents the long terminal repeat of the xenotropic mouse gammaretrovirus, and was acquired in Eurasian mice that harbor xenotropic retrovirus. This viral regulatory sequence does not alter splicing but is associated with elevated mA3 expression levels in spleens of laboratory and wild-derived mice. Analysis of Mus mA3 coding sequences produced evidence of positive selection and identified 10 codons with very high posterior probabilities of having evolved under positive selection. Six of these codons lie in two clusters in the N-terminal catalytically active cytidine deaminase domain (CDA), and 5 of those 6 codons are polymorphic in Rfv3 virus restrictive and nonrestrictive mice and align with hA3G CDA codons that are critical for deaminase activity. Homology models of mA3 indicate that the two selected codon clusters specify residues that are opposite each other along the predicted CDA active site groove, and that one cluster corresponds to an hAPOBEC substrate recognition loop. Substitutions at these clustered mA3 codons alter antiviral activity. This analysis suggests that mA3 has been under positive selection throughout Mus evolution, and identified an inserted retroviral regulatory sequence associated with enhanced expression in virus resistant mice and specific residues that modulate antiviral activity.


Subject(s)
Cytidine Deaminase/genetics , APOBEC-3G Deaminase , Amino Acid Sequence , Animals , Animals, Wild/genetics , Anti-Retroviral Agents/chemistry , Base Sequence , Evolution, Molecular , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Mutagenesis, Insertional , Selection, Genetic , Sequence Homology, Nucleic Acid
16.
Proteins ; 79(1): 282-93, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21064130

ABSTRACT

Interleukin-13 is a Th2-associated cytokine responsible for many pathological responses in allergic asthma including mucus production, inflammation, and extracellular matrix remodeling. In addition, IL-13 is required for immunity to many helminth infections. IL-13 signals via the type-II IL-4 receptor, a heterodimeric receptor of IL-13Rα1 and IL-4Rα, which is also used by IL-4. IL-13 also binds to IL-13Rα2, but with much higher affinity than the type-II IL-4 receptor. Binding of IL-13 to IL-13Rα2 has been shown to attenuate IL-13 signaling through the type-II IL-4 receptor. However, molecular determinants that dictate the specificity and affinity of mouse IL-13 for the different receptors are largely unknown. Here, we used high-density overlapping peptide arrays, structural modeling, and molecular docking methods to map IL-13 binding sequences on its receptors. Predicted binding sequences on mouse IL-13Rα1 and IL-13Rα2 were in agreement with the reported human IL-13 receptor complex structures and site-directed mutational analysis. Novel structural differences were identified between IL-13 receptors, particularly at the IL-13 binding interface. Notably, additional binding sites were observed for IL-13 on IL-13Rα2. In addition, the identification of peptide sequences that are unique to IL-13Rα1 allowed us to generate a monoclonal antibody that selectively binds IL-13Rα1. Thus, high-density peptide arrays combined with molecular docking studies provide a novel, rapid, and reliable method to map cytokine-receptor interactions that may be used to generate signaling and decoy receptor-specific antagonists.


Subject(s)
Interleukin-13 Receptor alpha1 Subunit/metabolism , Interleukin-13 Receptor alpha2 Subunit/metabolism , Interleukin-13/metabolism , Amino Acid Sequence , Animals , Binding Sites , Computer Simulation , Humans , Interleukin-13 Receptor alpha1 Subunit/chemistry , Interleukin-13 Receptor alpha2 Subunit/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Protein Array Analysis , Protein Binding , Protein Interaction Domains and Motifs , Sequence Alignment , Structural Homology, Protein
17.
mBio ; 12(2)2021 03 30.
Article in English | MEDLINE | ID: mdl-33785631

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) envelope trimer maintains a closed, metastable configuration to protect vulnerable epitopes from neutralizing antibodies. Here, we identify key hydrophobic constraints at the trimer apex that function as global stabilizers of the HIV-1 envelope spike configuration. Mutation of individual residues within four hydrophobic clusters that fasten together the V1V2, V3, and C4 domains at the apex of gp120 dramatically increases HIV-1 sensitivity to weak and restricted neutralizing antibodies targeting epitopes that are largely concealed in the prefusion Env spike, consistent with the adoption of a partially open trimer configuration. Conversely, the same mutations decrease the sensitivity to broad and potent neutralizing antibodies that preferentially recognize the closed trimer. Sera from chronically HIV-infected patients neutralize open mutants with enhanced potency, compared to the wild-type virus, suggesting that a large fraction of host-generated antibodies target concealed epitopes. The identification of structural constraints that maintain the HIV-1 envelope in an antibody-protected state may inform the design of a protective vaccine.IMPORTANCE Elucidating the structure and function of the HIV-1 envelope proteins is critical for the design of an effective vaccine. Despite the availability of many high-resolution structures, key functional correlates in the envelope trimer remain undefined. We utilized a combination of structural analysis, in silico energy calculation, mutagenesis, and neutralization profiling to dissect the functional anatomy of the trimer apex, which acts as a global regulator of the HIV-1 spike conformation. We identify four hydrophobic clusters that stabilize the spike in a tightly closed configuration and, thereby, play a critical role in protecting it from the reach of neutralizing antibodies.


Subject(s)
HIV-1/genetics , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/immunology , Amino Acid Motifs , Amino Acid Sequence , Antibodies, Neutralizing/immunology , HIV Antibodies , HIV Infections/virology , HIV-1/chemistry , HIV-1/immunology , Humans , Hydrophobic and Hydrophilic Interactions , Mutation , env Gene Products, Human Immunodeficiency Virus/genetics
18.
J Carcinog ; 8: 12, 2009.
Article in English | MEDLINE | ID: mdl-19661678

ABSTRACT

AIM: The xeroderma pigmentosum D (XPD) protein is a DNA helicase involved in the repair of DNA damage, including nucleotide excision repair (NER) and transcription-coupled repair (TCR). The C-terminal domain of XPD has been implicated in interactions with other components of the TFIIH complex, and it is also the site of a common genetic polymorphism in XPD at amino acid residue 751 (Lys->Gln). Some evidence suggests that this polymorphism may alter DNA repair capacity and increase cancer risk. The aim of this study was to investigate whether these effects could be attributable to conformational changes in XPD induced by the polymorphism. MATERIALS AND METHODS: Molecular dynamics techniques were used to predict the structure of the wild-type and polymorphic forms of the C-terminal domain of XPD and differences in structure produced by the polymorphic substitution were determined. RESULTS: The results indicate that, although the general configuration of both proteins is similar, the substitution produces a significant conformational change immediately N-terminal to the site of the polymorphism. CONCLUSION: These results provide support for the hypothesis that this polymorphism in XPD could affect DNA repair capability, and hence cancer risk, by altering the structure of the C-terminal domain.

19.
Biophys J ; 94(8): 3208-16, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18178652

ABSTRACT

Although internal water molecules are essential for the structure and function of many proteins, the structural and physical factors that govern internal hydration are poorly understood. We have examined the molecular determinants of internal hydration systematically, by solving the crystal structures of variants of staphylococcal nuclease with Gln-66, Asn-66, and Tyr-66 at cryo (100 K) and room (298 K) temperatures, and comparing them with existing cryo and room temperature structures of variants with Glu-66, Asp-66, Lys-66, Glu-92 or Lys-92 obtained under conditions of pH where the internal ionizable groups are in the neutral state. At cryogenic temperatures the polar moieties of all these internal side chains are hydrated except in the cases of Lys-66 and Lys-92. At room temperature the internal water molecules were observed only in variants with Glu-66 and Tyr-66; water molecules in the other variants are probably present but they are disordered and therefore undetectable crystallographically. Each internal water molecule establishes between 3 and 5 hydrogen bonds with the protein or with other internal water molecules. The strength of interactions between internal polar side chains and water molecules seems to decrease from carboxylic acids to amides to amines. Low temperature, low cavity volume, and the presence of oxygen atoms in the cavity increase the positional stability of internal water molecules. This set of structures and the physical insight they contribute into internal hydration will be useful for the development and benchmarking of computational methods for artificial hydration of pockets, cavities, and active sites in proteins.


Subject(s)
Crystallography/methods , Models, Chemical , Models, Molecular , Recombinant Proteins/chemistry , Recombinant Proteins/ultrastructure , Water/chemistry , Computer Simulation , Molecular Conformation , Porosity
20.
Proteins ; 72(4): 1243-58, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18361454

ABSTRACT

Although the number of known protein structures is increasing, the number of protein sequences without determined structures is still much larger. Three-dimensional (3D) protein structure information helps in the understanding of functional mechanisms, but solving structures by X-ray crystallography or NMR is often a lengthy and difficult process. A relatively fast way of determining a protein's 3D structure is to construct a computer model using homologous sequence and structure information. Much work has gone into algorithms that comprise the ORCHESTRAR homology modeling program in the SYBYL software package. This novel homology modeling tool combines algorithms for modeling conserved cores, variable regions, and side chains. The paradigm of using existing knowledge from multiple templates and the underlying protein environment knowledgebase is used in all of these algorithms, and will become even more powerful as the number of experimentally derived protein structures increases. To determine how ORCHESTRAR compares to Composer (a broadly used, but an older tool), homology models of 18 proteins were constructed using each program so that a detailed comparison of each step in the modeling process could be carried out. Proteins modeled include kinases, dihydrofolate reductase, HIV protease, and factor Xa. In almost all cases ORCHESTRAR produces models with lower root-mean-squared deviation (RMSD) values when compared with structures determined by X-ray crystallography or NMR. Moreover, ORCHESTRAR produced a homology model for three target sequences where Composer failed to produce any. Data for RMSD comparisons between structurally conserved cores, structurally variable regions, side-chain conformations are presented, as well as analyses of active site and protein-protein interface configurations.


Subject(s)
Algorithms , Computer Simulation , Protein Conformation , Proteins/chemistry , Sequence Alignment/methods , Software , Models, Molecular , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL