Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
Biomolecules ; 13(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37627292

ABSTRACT

The treatment landscape for lysosomal storage disorders (LSDs) is rapidly evolving. An increase in the number of preclinical and clinical studies in the last decade has demonstrated that pharmacological chaperones are a feasible alternative to enzyme replacement therapy (ERT) for individuals with LSDs. A systematic search was performed to retrieve and critically assess the evidence from preclinical and clinical applications of pharmacological chaperones in the treatment of LSDs and to elucidate the mechanisms by which they could be effective in clinical practice. Publications were screened according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) reporting guidelines. Fifty-two articles evaluating 12 small molecules for the treatment of seven LSDs are included in this review. Overall, a substantial amount of preclinical and clinical data support the potential of pharmacological chaperones as treatments for Fabry disease, Gaucher disease, and Pompe disease. Most of the available clinical evidence evaluated migalastat for the treatment of Fabry disease. There was a lack of consistency in the terminology used to describe pharmacological chaperones in the literature. Therefore, the new small molecule chaperone (SMC) classification system is proposed to inform a standardized approach for new, emerging small molecule therapies in LSDs.


Subject(s)
Fabry Disease , Gaucher Disease , Lysosomal Storage Diseases , Humans , Lysosomal Storage Diseases/drug therapy , Fabry Disease/drug therapy , Gaucher Disease/drug therapy , Enzyme Replacement Therapy , Lysosomes
3.
Front Immunol ; 14: 1094279, 2023.
Article in English | MEDLINE | ID: mdl-37033976

ABSTRACT

Immune responses to human non-self transgenes can present challenges in preclinical studies of adeno-associated virus (AAV) gene therapy candidates in nonhuman primates. Although anti-transgene immune responses are usually mild and non-adverse, they can confound pharmacological readouts and complicate translation of results between species. We developed a gene therapy candidate for Pompe disease consisting of AAVhu68, a clade F AAV closely related to AAV9, that expresses an engineered human acid-alpha glucosidase (hGAA) tagged with an insulin-like growth factor 2 variant (vIGF2) peptide for enhanced cell uptake. Rhesus macaques were administered an intravenous dose of 1x1013 genome copies (GC)/kg, 5x1013 GC/kg, or 1 x 1014 GC/kg of AAVhu68.vIGF2.hGAA. Some unusually severe adaptive immune responses to hGAA presented, albeit with a high degree of variability between animals. Anti-hGAA responses ranged from absent to severe cytotoxic T-cell-mediated myocarditis with elevated troponin I levels. Cardiac toxicity was not dose dependent and affected five out of eleven animals. Upon further investigation, we identified an association between toxicity and a major histocompatibility complex class I haplotype (Mamu-A002.01) in three of these animals. An immunodominant peptide located in the C-terminal region of hGAA was subsequently identified via enzyme-linked immunospot epitope mapping. Another notable observation in this preclinical safety study cohort pertained to the achievement of robust and safe gene transfer upon intravenous administration of 5x1013 GC/kg in one animal with a low pre-existing neutralizing anti-capsid antibodies titer (1:20). Collectively, these findings may have significant implications for gene therapy inclusion criteria.


Subject(s)
Glycogen Storage Disease Type II , Myocarditis , Humans , Animals , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism , Dependovirus , Macaca mulatta/metabolism , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy
4.
Viruses ; 13(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34452360

ABSTRACT

In most cases, proteolytic processing of the retroviral Pol portion of the Gag-Pol polyprotein precursor produces protease (PR), reverse transcriptase (RT), and integrase (IN). However, foamy viruses (FVs) express Pol separately from Gag and, when Pol is processed, only the IN domain is released. Here, we report a 2.9 Å resolution crystal structure of the mature PR-RT from prototype FV (PFV) that can carry out both proteolytic processing and reverse transcription but is in a configuration not competent for proteolytic or polymerase activity. PFV PR-RT is monomeric and the architecture of PFV PR is similar to one of the subunits of HIV-1 PR, which is a dimer. There is a C-terminal extension of PFV PR (101-145) that consists of two helices which are adjacent to the base of the RT palm subdomain, and anchors PR to RT. The polymerase domain of PFV RT consists of fingers, palm, thumb, and connection subdomains whose spatial arrangements are similar to the p51 subunit of HIV-1 RT. The RNase H and polymerase domains of PFV RT are connected by flexible linkers. Significant spatial and conformational (sub)domain rearrangements are therefore required for nucleic acid binding. The structure of PFV PR-RT provides insights into the conformational maturation of retroviral Pol polyproteins.


Subject(s)
Peptide Hydrolases/chemistry , Polyproteins/chemistry , RNA-Directed DNA Polymerase/chemistry , Spumavirus/chemistry , Crystallization , Peptide Hydrolases/metabolism , Polyproteins/metabolism , RNA-Directed DNA Polymerase/metabolism , Reverse Transcription
5.
Nucleic Acids Res ; 48(5): 2709-2722, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31943114

ABSTRACT

RNA aptamers that bind HIV-1 reverse transcriptase (RT) inhibit RT in enzymatic and viral replication assays. Some aptamers inhibit RT from only a few viral clades, while others show broad-spectrum inhibition. Biophysical determinants of recognition specificity are poorly understood. We investigated the interface between HIV-1 RT and a broad-spectrum UCAA-family aptamer. SAR and hydroxyl radical probing identified aptamer structural elements critical for inhibition and established the role of signature UCAA bulge motif in RT-aptamer interaction. HDX footprinting on RT ± aptamer shows strong contacts with both subunits, especially near the C-terminus of p51. Alanine scanning revealed decreased inhibition by the aptamer for mutants P420A, L422A and K424A. 2D proton nuclear magnetic resonance and SAXS data provided constraints on the solution structure of the aptamer and enable computational modeling of the docked complex with RT. Surprisingly, the aptamer enhanced proteolytic cleavage of precursor p66/p66 by HIV-1 protease, suggesting that it stabilizes the productive conformation to allow maturation. These results illuminate features at the RT-aptamer interface that govern recognition specificity by a broad-spectrum antiviral aptamer, and they open new possibilities for accelerating RT maturation and interfering with viral replication.


Subject(s)
Aptamers, Nucleotide/metabolism , HIV Protease/metabolism , HIV Reverse Transcriptase/metabolism , Aptamers, Nucleotide/chemistry , Molecular Docking Simulation , Mutagenesis/genetics , Mutant Proteins/metabolism , Protein Binding , Protein Multimerization , Reverse Transcriptase Inhibitors/pharmacology
6.
Curr Res Struct Biol ; 2: 116-129, 2020.
Article in English | MEDLINE | ID: mdl-33870216

ABSTRACT

The high-resolution crystal structure of HIV-1 reverse transcriptase (RT) bound to a 38-mer DNA hairpin aptamer with low pM affinity was previously described. The high-affinity binding aptamer contained 2'-O-methyl modifications and a seven base-pair GC-rich tract and the structure of the RT-aptamer complex revealed specific contacts between RT and the template strand of the aptamer. Similar to all crystal structures of RT bound to nucleic acid template-primers, the aptamer bound RT with a bend in the duplex DNA. To understand the structural basis for the ultra-high-affinity aptamer binding, an integrative structural biology approach was used. Hydrogen-deuterium exchange coupled to liquid chromatography-mass spectrometry (HDX-MS) was used to examine the structural dynamics of RT alone and in the presence of the DNA aptamer. RT was selectively labeled with 15N to unambiguously identify peptides from each subunit. HDX of unliganded RT shows a mostly stable core. The p66 fingers and thumb subdomains, and the RNase H domain are relatively dynamic. HDX indicates that both the aptamer and a scrambled version significantly stabilize regions of RT that are dynamic in the absence of DNA. No substantial differences in RT dynamics are observed between aptamer and scrambled aptamer binding, despite a large difference in binding affinity. Small-angle X-ray scattering and circular dichroism spectroscopy were used to investigate the aptamer conformation in solution and revealed a pre-bent DNA that possesses both A- and B-form helical character. Both the 2'-O-methyl modifications and the GC tract appear to contribute to an energetically favorable conformation for binding to RT that contributes to the aptamer's ultra-high affinity for RT. The X-ray structure of RT with an RNA/DNA version of the aptamer at 2.8 Å resolution revealed a potential role of the hairpin positioning in affinity. Together, the data suggest that both the 2'-O-methyl modifications and the GC tract contribute to an energetically favorable conformation for high-affinity binding to RT.

7.
Protein Sci ; 25(1): 46-55, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26296781

ABSTRACT

The development of a modified DNA aptamer that binds HIV-1 reverse transcriptase (RT) with ultra-high affinity has enabled the X-ray structure determination of an HIV-1 RT-DNA complex to 2.3 Å resolution without the need for an antibody Fab fragment or RT-DNA cross-linking. The 38-mer hairpin-DNA aptamer has a 15 base-pair duplex, a three-deoxythymidine hairpin loop, and a five-nucleotide 5'-overhang. The aptamer binds RT in a template-primer configuration with the 3'-end positioned at the polymerase active site and has 2'-O-methyl modifications at the second and fourth duplex template nucleotides that interact with the p66 fingers and palm subdomains. This structure represents the highest resolution RT-nucleic acid structure to date. The RT-aptamer complex is catalytically active and can serve as a platform for studying fundamental RT mechanisms and for development of anti-HIV inhibitors through fragment screening and other approaches. Additionally, the structure allows for a detailed look at a unique aptamer design and provides the molecular basis for its remarkably high affinity for RT.


Subject(s)
Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , DNA Primers/chemistry , HIV Reverse Transcriptase/chemistry , Nucleic Acid Conformation , Anti-HIV Agents/chemistry , Aptamers, Nucleotide/metabolism , Binding Sites , DNA Primers/genetics , DNA Primers/metabolism , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Models, Molecular , Protein Conformation , Reverse Transcriptase Inhibitors/chemistry , Structure-Activity Relationship , Templates, Genetic
8.
Anal Chem ; 87(7): 4015-4022, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25763479

ABSTRACT

Hydrogen/deuterium exchange (HDX) coupled to mass spectrometry has emerged as a powerful tool for analyzing the conformational dynamics of protein-ligand and protein-protein interactions. Recent advances in instrumentation and methodology have expanded the utility of HDX for the analysis of large and complex proteins; however, asymmetric dimers with shared amino acid sequence present a unique challenge for HDX because assignment of peptides with identical sequence to their subunit of origin remains ambiguous. Here we report the use of differential isotopic labeling to facilitate HDX analysis of multimers using HIV-1 reverse transcriptase (RT) as a model. RT is an asymmetric heterodimer of 51 kDa (p51) and 66 kDa (p66) subunits. The first 440 residues of p51 and p66 are identical. In this study differentially labeled RT was reconstituted from isotopically enriched ((15)N-labeled) p51 and unlabeled p66. To enable detection of (15)N-deuterated RT peptides, the software HDX Workbench was modified to follow a 100% (15)N model. Our results demonstrated that (15)N enrichment of p51 did not affect its conformational dynamics compared to unlabeled p51, but (15)N-labeled p51 did show different conformational dynamics than p66 in the RT heterodimer. Differential HDX-MS of isotopically labeled RT in the presence of the non-nucleoside reverse transcriptase inhibitor (NNRTI) efavirenz (EFV) showed subunit-specific perturbation in the rate of HDX consistent with previously published results and the RT-EFV cocrystal structure.


Subject(s)
Deuterium Exchange Measurement , HIV Reverse Transcriptase/analysis , HIV Reverse Transcriptase/chemistry , Mass Spectrometry , Nitrogen Isotopes
9.
Elife ; 3: e02450, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24755292

ABSTRACT

Using a combination of genetic, biochemical, and structural approaches, we show that the cyclic-peptide antibiotic GE23077 (GE) binds directly to the bacterial RNA polymerase (RNAP) active-center 'i' and 'i+1' nucleotide binding sites, preventing the binding of initiating nucleotides, and thereby preventing transcription initiation. The target-based resistance spectrum for GE is unusually small, reflecting the fact that the GE binding site on RNAP includes residues of the RNAP active center that cannot be substituted without loss of RNAP activity. The GE binding site on RNAP is different from the rifamycin binding site. Accordingly, GE and rifamycins do not exhibit cross-resistance, and GE and a rifamycin can bind simultaneously to RNAP. The GE binding site on RNAP is immediately adjacent to the rifamycin binding site. Accordingly, covalent linkage of GE to a rifamycin provides a bipartite inhibitor having very high potency and very low susceptibility to target-based resistance. DOI: http://dx.doi.org/10.7554/eLife.02450.001.


Subject(s)
Nucleotides/metabolism , Peptides, Cyclic/metabolism , RNA Polymerase I/metabolism , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Binding Sites , Crystallography, X-Ray , Escherichia coli/enzymology , Models, Molecular , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Rifamycins/pharmacology , Thermus thermophilus/enzymology , Transcription, Genetic/drug effects
10.
Biochimie ; 95(10): 1880-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23806423

ABSTRACT

New antibiotics with novel mechanisms of action are urgently needed to overcome the growing bacterial resistance problem faced by clinicians today. PC190723 and related compounds represent a promising new class of antibacterial compounds that target the essential bacterial cell division protein FtsZ. While this family of compounds exhibits potent antistaphylococcal activity, they have poor activity against enterococci and streptococci. The studies described herein are aimed at investigating the molecular basis of the enterococcal and streptococcal resistance to this family of compounds. We show that the poor activity of the compounds against enterococci and streptococci correlates with a correspondingly weak impact of the compounds on the self-polymerization of the FtsZ proteins from those bacteria. In addition, computational and mutational studies identify two key FtsZ residues (E34 and R308) as being important determinants of enterococcal and streptococcal resistance to the PC190723-type class of compounds.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus subtilis/drug effects , Bacterial Proteins/genetics , Cytoskeletal Proteins/genetics , Drug Resistance, Multiple, Bacterial/drug effects , Pyridines/pharmacology , Staphylococcus aureus/drug effects , Thiazoles/pharmacology , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Cytoskeletal Proteins/metabolism , Drug Resistance, Multiple, Bacterial/genetics , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Sequence Data , Mutation , Protein Multimerization , Pyridines/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Thiazoles/chemistry
11.
J Med Chem ; 55(22): 10160-76, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23050700

ABSTRACT

The prevalence of multidrug resistance among clinically significant bacterial pathogens underscores a critical need for the development of new classes of antibiotics with novel mechanisms of action. Here we describe the synthesis and evaluation of a guanidinomethyl biaryl compound {1-((4'-(tert-butyl)-[1,1'-biphenyl]-3-yl)methyl)guanidine} that targets the bacterial cell division protein FtsZ. In vitro studies with various bacterial FtsZ proteins reveal that the compound alters the dynamics of FtsZ self-polymerization via a stimulatory mechanism, while minimally impacting the polymerization of tubulin, the closest mammalian homologue of FtsZ. The FtsZ binding site of the compound is identified through a combination of computational and mutational approaches. The compound exhibits a broad spectrum of bactericidal activity, including activity against the multidrug-resistant pathogens methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE), while also exhibiting a minimal potential to induce resistance. Taken together, our results highlight the compound as a promising new FtsZ-targeting bactericidal agent.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Biphenyl Compounds/pharmacology , Cytoskeletal Proteins/metabolism , Enterococcus/drug effects , Guanidines/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Polymerization/drug effects , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Vancomycin Resistance/drug effects , Anti-Bacterial Agents/chemical synthesis , Biphenyl Compounds/chemical synthesis , Drug Resistance, Multiple/drug effects , Guanidines/chemical synthesis , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Structure-Activity Relationship
12.
Science ; 338(6110): 1076-80, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23086998

ABSTRACT

During transcription initiation, RNA polymerase (RNAP) binds and unwinds promoter DNA to form an RNAP-promoter open complex. We have determined crystal structures at 2.9 and 3.0 Å resolution of functional transcription initiation complexes comprising Thermus thermophilus RNA polymerase, σ(A), and a promoter DNA fragment corresponding to the transcription bubble and downstream double-stranded DNA of the RNAP-promoter open complex. The structures show that σ recognizes the -10 element and discriminator element through interactions that include the unstacking and insertion into pockets of three DNA bases and that RNAP recognizes the -4/+2 region through interactions that include the unstacking and insertion into a pocket of the +2 base. The structures further show that interactions between σ and template-strand single-stranded DNA (ssDNA) preorganize template-strand ssDNA to engage the RNAP active center.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , Gene Expression Regulation, Bacterial , Sigma Factor/chemistry , Thermus thermophilus/enzymology , Thermus thermophilus/genetics , Transcription Initiation, Genetic , Crystallography, X-Ray , DNA, Single-Stranded/chemistry , Promoter Regions, Genetic , Protein Conformation
13.
Nat Struct Mol Biol ; 11(5): 469-74, 2004 May.
Article in English | MEDLINE | ID: mdl-15107837

ABSTRACT

Tenofovir, also known as PMPA, R-9-(2-(phosphonomethoxypropyl)adenine, is a nucleotide reverse transcriptase (RT) inhibitor. We have determined the crystal structures of two related complexes of HIV-1 RT with template primer and tenofovir: (i) a ternary complex at a resolution of 3.0 A of RT crosslinked to a dideoxy-terminated DNA with tenofovir-diphosphate bound as the incoming substrate; and (ii) a RT-DNA complex at a resolution of 3.1 A with tenofovir at the 3' primer terminus. The tenofovir nucleotide in the tenofovir-terminated structure seems to adopt multiple conformations. Some nucleoside reverse transcriptase inhibitors, including 3TC and AZT, have elements ('handles') that project beyond the corresponding elements on normal dNTPs (the 'substrate envelope'). HIV-1 RT resistance mechanisms to AZT and 3TC take advantage of these handles; tenofovir's structure lacks handles that could protrude through the substrate envelope to cause resistance.


Subject(s)
Adenine/analogs & derivatives , Adenine/chemistry , DNA, Viral/chemistry , HIV Reverse Transcriptase/chemistry , Organophosphonates , Organophosphorus Compounds/chemistry , Reverse Transcriptase Inhibitors/chemistry , Base Sequence , DNA Primers , Models, Molecular , Tenofovir
14.
J Biol Chem ; 278(18): 16280-8, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12554739

ABSTRACT

A disulfide cross-linking strategy was used to covalently trap as a stable complex (complex N) a short-lived, kinetic intermediate in DNA polymerization. This intermediate corresponds to the product of polymerization prior to translocation. We also prepared the trapped complex that corresponds to the product of polymerization after translocation (complex P). The cross-linking method that we used is a variation of a technique developed by the Verdine and Harrison laboratories. It involves disulfide interchange between an engineered sulfhydryl group of the protein (Q258C mutation) and a disulfide-containing tether attached at the N(2) amino group of a modified dG in either the template or the primer strand of the nucleic acid. We report here a highly efficient synthesis of the precursor, bis(3-aminopropyl)disulfide dihydrochloride, used to introduce this substituent into the oligonucleotide. Efficient cross-linking takes place when the base pair containing the substituent is positioned seven registers from the dNTP-binding site (N site) and the N site is occupied. Complex N, but not complex P, is a substrate for the ATP-based excision reaction that unblocks nucleoside reverse transcriptase inhibitor (NRTI)-terminated primers and causes resistance to several NRTIs, confirming predictions that the excision reaction takes place only when the 3'-end of the primer is bound at the N site. These techniques can be used for biochemical and structural studies of the mechanism of DNA polymerization, translocation, and excision-based resistance of RT to NRTIs. They may also be useful in studying other DNA or RNA polymerases or other enzymes.


Subject(s)
DNA/metabolism , HIV Reverse Transcriptase/metabolism , Adenosine Triphosphate/metabolism , Biological Transport , Crystallization , HIV Reverse Transcriptase/chemistry
15.
EMBO J ; 21(23): 6614-24, 2002 Dec 02.
Article in English | MEDLINE | ID: mdl-12456667

ABSTRACT

AZT (3'-azido-3'-deoxythymidine) resistance involves the enhanced excision of AZTMP from the end of the primer strand by HIV-1 reverse transcriptase. This reaction can occur when an AZTMP-terminated primer is bound at the nucleotide-binding site (pre-translocation complex N) but not at the 'priming' site (post-translocation complex P). We determined the crystal structures of N and P complexes at 3.0 and 3.1 A resolution. These structures provide insight into the structural basis of AZTMP excision and the mechanism of translocation. Docking of a dNTP in the P complex structure suggests steric crowding in forming a stable ternary complex that should increase the relative amount of the N complex, which is the substrate for excision. Structural differences between complexes N and P suggest that the conserved YMDD loop is involved in translocation, acting as a springboard that helps to propel the primer terminus from the N to the P site after dNMP incorporation.


Subject(s)
DNA/metabolism , HIV-1/metabolism , RNA-Directed DNA Polymerase/metabolism , Thymine Nucleotides/metabolism , Zidovudine/analogs & derivatives , Zidovudine/metabolism , DNA/biosynthesis , Dideoxynucleotides , Drug Resistance, Viral/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...