Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters

Country/Region as subject
Publication year range
1.
NMR Biomed ; 36(5): e4888, 2023 05.
Article in English | MEDLINE | ID: mdl-36468685

ABSTRACT

Favipiravir (brand name Avigan), a widely known anti-influenza prodrug, is metabolized by endogenous enzymes of host cells to generate the active form, which exerts inhibition of viral RNA-dependent RNA polymerase activity; first, favipiravir is converted to its phosphoribosylated form, favipiravir-ribofuranosyl-5'-monophosphate (favipiravir-RMP), by hypoxanthine-guanine phosphoribosyltransferase (HGPRT). Because this phosphoribosylation reaction is the rate-determining step in the generation of the active metabolite, quantitative and real-time monitoring of the HGPRT-catalyzed reaction is essential to understanding the pharmacokinetics of favipiravir. However, assay methods enabling such monitoring have not been established. 19 F- or 31 P-based nuclear magnetic resonance (NMR) are powerful techniques for observation of intermolecular interactions, chemical reactions, and metabolism of molecules of interest, given that NMR signals of the heteronuclei sensitively reflect changes in the chemical environment of these moieties. Here, we demonstrated direct, sensitive, target-selective, nondestructive, and real-time observation of HGPRT-catalyzed conversion of favipiravir to favipiravir-RMP by performing time-lapse 19 F-NMR monitoring of the fluorine atom of favipiravir. In addition, we showed that 31 P-NMR can be used for real-time observation of the identical reaction by monitoring phosphorus atoms of the phosphoribosyl group of favipiravir-RMP and of the pyrophosphate product of that reaction. Furthermore, we demonstrated that NMR approaches permit the determination of general parameters of enzymatic activity such as Vmax and Km . This method not only can be widely employed in enzyme assays, but also may be of use in the screening and development of new favipiravir-analog antiviral prodrugs that can be phosphoribosylated more efficiently by HGPRT, which would increase the intracellular concentration of the drug's active form. The techniques demonstrated in this study would allow more detailed investigation of the pharmacokinetics of fluorinated drugs, and might significantly contribute to opening new avenues for widespread pharmaceutical studies.


Subject(s)
Prodrugs , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Time-Lapse Imaging , Amides , Magnetic Resonance Spectroscopy , Catalysis
2.
Arch Biochem Biophys ; 737: 109550, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36796662

ABSTRACT

Approaches to identify novel druggable targets for treating neglected diseases include computational studies that predict possible interactions of drugs and their molecular targets. Hypoxanthine phosphoribosyltransferase (HPRT) plays a central role in the purine salvage pathway. This enzyme is essential for the survival of the protozoan parasite T. cruzi, the causal agent of Chagas disease, and other parasites related to neglected diseases. Here we found dissimilar functional behaviours between TcHPRT and the human homologue, HsHPRT, in the presence of substrate analogues that can lie in differences in their oligomeric assemblies and structural features. To shed light on this issue, we carried out a comparative structural analysis between both enzymes. Our results show that HsHPRT is considerably more resistant to controlled proteolysis than TcHPRT. Moreover, we observed a variation in the length of two key loops depending on the structural arrangement of each protein (groups D1T1 and D1T1'). Such variations might be involved in inter-subunit communication or influencing the oligomeric state. Besides, to understand the molecular basis that govern D1T1 and D1T1' folding groups, we explored the distribution of charges on the interaction surfaces of TcHPRT and HsHPRT, respectively. To know whether the rigidity degree bears effect on the active site, we studied the flexibility of both proteins. The analysis performed here illuminates the underlying reasons and significance behind each protein's preference for one or the other quaternary arrangement that can be exploited for therapeutic approaches.


Subject(s)
Anti-Infective Agents , Trypanosoma cruzi , Humans , Trypanosoma cruzi/metabolism , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/metabolism , Hypoxanthine Phosphoribosyltransferase/pharmacology , Antiparasitic Agents/pharmacology , Neglected Diseases , Anti-Infective Agents/pharmacology
3.
Biochemistry ; 61(19): 2088-2105, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36193631

ABSTRACT

Chagas disease, caused by the parasitic protozoan Trypanosoma cruzi, affects over 8 million people worldwide. Current antiparasitic treatments for Chagas disease are ineffective in treating advanced, chronic stages of the disease, and are noted for their toxicity. Like most parasitic protozoa, T. cruzi is unable to synthesize purines de novo, and relies on the salvage of preformed purines from the host. Hypoxanthine-guanine phosphoribosyltransferases (HGPRTs) are enzymes that are critical for the salvage of preformed purines, catalyzing the formation of inosine monophosphate (IMP) and guanosine monophosphate (GMP) from the nucleobases hypoxanthine and guanine, respectively. Due to the central role of HGPRTs in purine salvage, these enzymes are promising targets for the development of new treatment methods for Chagas disease. In this study, we characterized two gene products in the T. cruzi CL Brener strain that encodes enzymes with functionally identical HGPRT activities in vitro: TcA (TcCLB.509693.70) and TcC (TcCLB.506457.30). The TcC isozyme was kinetically characterized to reveal mechanistic details on catalysis, including identification of the rate-limiting step(s) of catalysis. Furthermore, we identified and characterized inhibitors of T. cruzi HGPRTs originally developed as transition-state analogue inhibitors (TSAIs) of Plasmodium falciparum hypoxanthine-guanine-xanthine phosphoribosyltransferase (PfHGXPRT), where the most potent compound bound to T. cruzi HGPRT with low nanomolar affinity. Our results validated the repurposing of TSAIs to serve as selective inhibitors for orthologous molecular targets, where primary and secondary structures as well as putatively common chemical mechanisms are conserved.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Antiparasitic Agents , Guanine/metabolism , Guanosine Monophosphate , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Inosine Monophosphate , Isoenzymes , Purines/metabolism , Purines/pharmacology
4.
J Cell Biochem ; 122(11): 1701-1714, 2021 11.
Article in English | MEDLINE | ID: mdl-34346095

ABSTRACT

Hypoxanthine-guanine phosphoribosyltransferase (HGPRT) is the key regulatory enzyme of the purine salvage pathway present in the members of trypanosomatids. The parasite solely depends on this pathway for the synthesis of nucleotides due to the absence of the de novo pathway. This study intends to identify putative inhibitors towards Trypanosoma cruzi HGPRT (TcHGPRT). Initial virtual screening was performed with substructures of phosphoribosyl pyrophosphate (PRPP), an original substrate of HGPRT. Twenty compounds that had greater binding energy than the substrate was treated as hits and was further screened and narrowed down through induced fit docking which resulted in top five compounds which was distinguished into two groups based on the ligand occupancy within the PRPP binding site of TcHGPRT. Group-I compounds (PubChem CID 130316561 and 134978234) are analogous to PRPP structure with greater occupancy, were preferred over Group-II compounds which had lesser occupancy than the substrate. However, one compound (22404820) among Group II was chosen for further analysis considering its significant electrostatic interactions. Molecular docking studies revealed the requirement of an electronegative moiety like phosphate group to be present in the ligand due to the presence of metal ions in the substrate binding site. The three chosen compounds along with PRPP were subjected to molecular dynamics analysis, which indicated a strong presence of electrostatic interaction. Considering the dynamic stability of interactions as well as pharmacological properties of ligands based on absorption, distribution, metabolism, excretion prediction, Group-I compounds were selected as lead compounds and were subjected to molecular electrostatic potential analysis to determine the charge distribution of the compound. The overall analysis thus suggests both 130316561 and 134978234 can be used as TcHGPRT inhibitors. Furthermore, these computational results emphasize the requirement of phosphorylated ligands which are essential in mediating electrostatic interactions and to compete with the binding affinity of the original substrate.


Subject(s)
Enzyme Inhibitors/pharmacology , Hypoxanthine Phosphoribosyltransferase/antagonists & inhibitors , Hypoxanthine Phosphoribosyltransferase/chemistry , Protozoan Proteins/antagonists & inhibitors , Trypanosoma cruzi/enzymology , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Humans , Hypoxanthine Phosphoribosyltransferase/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Static Electricity
5.
Genomics ; 112(1): 304-311, 2020 01.
Article in English | MEDLINE | ID: mdl-30818063

ABSTRACT

Genetic changes in Hypoxanthine guanine phosphoribosyltransferace (HPRT1) gene can alter the expression of the dopamine neurotransmitter leads to abnormal neuron function, a disease called Lesch-Nyhan syndrome (LNS). Although different studies were conducted on LNS, information on codon usage bias (CUB) of HPRT1 gene is limited. The present study examines the genetic determinants of CUB in HPRT1 gene using twelve mammalian species. In the coding sequence of HPRT1 genes, A/T ending codons was most frequently used. A higher ENC value was observed indicating lower HPRT1 gene expression in the selected mammalian species. Correlation analysis indicates that compositional constraints under mutation pressure can involve in CUB of HPRT1 genes among the selected mammalian species. Relative synonymous codon usage (RSCU) value revealed that the codons such as ACT, AGG, ATT and AGC were over-represented in each of the mammalian species. Result from the analysis of the RSCU indicates that compositional constraint is a key driver for the variation in codon usage. Ratio of nonsynonymous (dN) and synonymous (dS) substitution further suggested that purifying selection occurs among the HPRT1 gene of studied mammals to maintain its protein function under the process of evolution. Our findings report an insight into the codon usage patterns of HPRT1 gene and will be useful for LNS management.


Subject(s)
Codon Usage , Hypoxanthine Phosphoribosyltransferase/genetics , Amino Acid Sequence , Animals , Base Composition , Cattle , Codon , DNA/chemistry , Gene Expression , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/metabolism , Mammals/genetics , Phylogeny , Rats , Selection, Genetic , Sequence Alignment
6.
J Bacteriol ; 202(5)2020 02 11.
Article in English | MEDLINE | ID: mdl-31818925

ABSTRACT

Purine metabolism plays a ubiquitous role in the physiology of Mycobacterium tuberculosis and other mycobacteria. The purine salvage enzyme hypoxanthine-guanine phosphoribosyltransferase (HGPRT) is essential for M. tuberculosis growth in vitro; however, its precise role in M. tuberculosis physiology is unclear. Membrane-permeable prodrugs of specifically designed HGPRT inhibitors arrest the growth of M. tuberculosis and represent potential new antituberculosis compounds. Here, we investigated the purine salvage pathway in the model organism Mycobacterium smegmatis Using genomic deletion analysis, we confirmed that HGPRT is the only guanine and hypoxanthine salvage enzyme in M. smegmatis but is not required for in vitro growth of this mycobacterium or survival under long-term stationary-phase conditions. We also found that prodrugs of M. tuberculosis HGPRT inhibitors displayed an unexpected antimicrobial activity against M. smegmatis that is independent of HGPRT. Our data point to a different mode of mechanism of action for these inhibitors than was originally proposed.IMPORTANCE Purine bases, released by the hydrolytic and phosphorolytic degradation of nucleic acids and nucleotides, can be salvaged and recycled. The hypoxanthine-guanine phosphoribosyltransferase (HGPRT), which catalyzes the formation of guanosine-5'-monophosphate from guanine and inosine-5'-monophosphate from hypoxanthine, represents a potential target for specific inhibitor development. Deletion of the HGPRT gene (Δhgprt) in the model organism Mycobacterium smegmatis confirmed that this enzyme is not essential for M. smegmatis growth. Prodrugs of acyclic nucleoside phosphonates (ANPs), originally designed against HGPRT from Mycobacterium tuberculosis, displayed anti-M. smegmatis activities comparable to those obtained for M. tuberculosis but also inhibited the ΔhgprtM. smegmatis strain. These results confirmed that ANPs act in M. smegmatis by a mechanism independent of HGPRT.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Mycobacterium smegmatis/genetics , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Catalysis , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hypoxanthine Phosphoribosyltransferase/antagonists & inhibitors , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/metabolism , Metabolic Networks and Pathways , Microbial Viability , Mycobacterium smegmatis/growth & development , Mycobacterium smegmatis/metabolism , Plasmids/genetics , Purines/metabolism
7.
J Biol Chem ; 294(32): 11992-11993, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31399535

ABSTRACT

Two phosphoribosyltransferases in the purine salvage pathway exhibit exquisite substrate specificity despite the chemical similarity of their distinct substrates, but the basis for this discrimination was not fully understood. Ozeir et al. now employ a complementary biochemical, structural, and computational approach to deduce the chemical constraints governing binding and propose a distinct mechanism for catalysis in one of these enzymes, adenine phosphoribosyltransferase. These insights, built on data from an unexpected finding, finally provide direct answers to key questions regarding these enzymes and substrate recognition more generally.


Subject(s)
Adenine Phosphoribosyltransferase/metabolism , Hypoxanthine Phosphoribosyltransferase/metabolism , Adenine Phosphoribosyltransferase/chemistry , Biocatalysis , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , Purines/chemistry , Purines/metabolism , Substrate Specificity
8.
Biochem Biophys Res Commun ; 532(4): 499-504, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32873391

ABSTRACT

Purine bases, synthesized de novo or recycled through the salvage pathway, are precursors of nucleotide synthesis and are essential in a variety of physiological processes including cell division, growth, signaling, energy metabolism and synthesis of vitamins/co-factor. The protozoan kinetoplastid parasites including Leishmania cannot synthesize de novo and rely solely on the purine salvage pathway, recycling the degraded products of nucleic acid metabolism. Enzymes of this pathway are thus of therapeutic importance. The enzyme Hypoxanthine-guanine phosphoribosyl transferase (HGPRT) (EC 2.4.2.8) plays a central role in this pathway, converting the purine base to its monophosphate product. Towards the elucidation of its role, we have cloned, expressed, purified and determined the crystal structure of L. donovani HGPRT at 2.76 Å. Comparative structural analysis with the human homolog indicates differences in oligomer association. Comparative analyses identify insertions in the human homolog sequence in the tetramer interface. The results suggest that this difference can be exploited for therapeutic approaches.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/chemistry , Leishmania donovani/enzymology , Protozoan Proteins/chemistry , Cloning, Molecular , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/isolation & purification , Models, Molecular , Protozoan Proteins/genetics , Protozoan Proteins/isolation & purification , Structural Homology, Protein
9.
Biochim Biophys Acta ; 1864(6): 655-666, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26969784

ABSTRACT

Hypoxanthine phosphoribosyl transferase from Trypanosoma cruzi (TcHPRT) is a critical enzyme for the survival of the parasite. This work demonstrates that the full-length form in solution adopts a stable and enzymatically active tetrameric form, exhibiting large inter-subunit surfaces. Although this protein irreversibly aggregates during unfolding, oligomerization is reversible and can be modulated by low concentrations of urea. When the C-terminal region, which is predicted as a disordered stretch, is excised by proteolysis, TcHPRT adopts a dimeric state, suggesting that the C-terminal region acts as a main guide for the quaternary arrangement. These results are in agreement with X-ray crystallographic data presented in this work. On the other hand, the C-terminal region exhibits a modulatory role on the enzyme, as attested by the enhanced activity observed for the dimeric form. Bisphosphonates act as substrate-mimetics, uncovering long-range communications among the active sites. All in all, this work contributes to establish new ways applicable to the design of novel inhibitors that could eventually result in new drugs against parasitic diseases.


Subject(s)
Biopolymers/metabolism , Hypoxanthine Phosphoribosyltransferase/metabolism , Trypanosoma cruzi/enzymology , Amino Acid Sequence , Biopolymers/chemistry , Circular Dichroism , Hypoxanthine Phosphoribosyltransferase/chemistry , Molecular Sequence Data , Proteolysis , Spectrophotometry, Ultraviolet
10.
J Struct Biol ; 194(3): 311-6, 2016 06.
Article in English | MEDLINE | ID: mdl-26968365

ABSTRACT

Hypoxanthine-guanine phosphoribosyltransferase (HGPRT) (EC 2.4.2.8) reversibly catalyzes the transfer of the 5-phophoribosyl group from 5-phosphoribosyl-alpha-1-pyrophosphate (PRPP) to hypoxanthine or guanine to form inosine monophosphate (IMP) or guanosine monophosphate (GMP) in the purine salvage pathway. To investigate the catalytic mechanism of this enzyme in the intracellular pathogen Legionella pneumophila, we determined the crystal structures of the L. pneumophila HGPRT (LpHGPRT) both in its apo-form and in complex with GMP. The structures reveal that LpHGPRT comprises a core domain and a hood domain which are packed together to create a cavity for GMP-binding and the enzymatic catalysis. The binding of GMP induces conformational changes of the stable loop II. This new binding site is closely related to the Gout arthritis-linked human HGPRT mutation site (Ser103Arg). Finally, these structures of LpHGPRT provide insights into the catalytic mechanism of HGPRT.


Subject(s)
Guanosine Monophosphate/chemistry , Hypoxanthine Phosphoribosyltransferase/chemistry , Legionella pneumophila/enzymology , Arthritis, Gouty/enzymology , Arthritis, Gouty/genetics , Binding Sites , Biocatalysis , Crystallography, X-Ray , Guanosine Monophosphate/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/metabolism , Protein Binding , Protein Conformation
11.
BMC Genomics ; 17: 548, 2016 Aug 04.
Article in English | MEDLINE | ID: mdl-27491780

ABSTRACT

BACKGROUND: Numerous human genes encode potentially active DNA transposases or recombinases, but our understanding of their functions remains limited due to shortage of methods to profile their activities on endogenous genomic substrates. RESULTS: To enable functional analysis of human transposase-derived genes, we combined forward chemical genetic hypoxanthine-guanine phosphoribosyltransferase 1 (HPRT1) screening with massively parallel paired-end DNA sequencing and structural variant genome assembly and analysis. Here, we report the HPRT1 mutational spectrum induced by the human transposase PGBD5, including PGBD5-specific signal sequences (PSS) that serve as potential genomic rearrangement substrates. CONCLUSIONS: The discovered PSS motifs and high-throughput forward chemical genomic screening approach should prove useful for the elucidation of endogenous genome remodeling activities of PGBD5 and other domesticated human DNA transposases and recombinases.


Subject(s)
Gene Rearrangement , Genetic Testing , Genome, Human , Genomics , Transposases/genetics , Base Sequence , Cell Line , Gene Expression , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/genetics , Mutation , Protein Sorting Signals/genetics , Sequence Analysis, DNA , Transposases/chemistry
12.
J Am Chem Soc ; 138(17): 5519-22, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27093234

ABSTRACT

A central intermediate in purine catabolism, the inosine nucleobase hypoxanthine is also one of the most abundant modified nucleobases in RNA and plays key roles in the regulation of gene expression and determination of cell fate. It is known that hypoxanthine acts as guanine when interacting with other nucleobases and base pairs most favorably with cytosine. However, its preferences when it comes to interactions with amino acids remain unknown. Here we present for the first time the absolute binding free energies and the associated interaction modes between hypoxanthine and all standard, non-glycyl/non-prolyl amino acid side chain analogs as derived from molecular dynamics simulations and umbrella sampling in high- and low-dielectric environments. We illustrate the biological relevance of the derived affinities by providing a quantitative explanation for the specificity of hypoxanthine-guanine phosphoribosyltransferase, a key enzyme in the purine salvage pathway. Our results demonstrate that in its affinities for protein side chains, hypoxanthine closely matches guanine, much more so than its precursor adenine.


Subject(s)
Guanine/chemistry , Inosine/chemistry , Proteins/chemistry , Hypoxanthine Phosphoribosyltransferase/chemistry
13.
J Chem Inf Model ; 56(8): 1528-38, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27404508

ABSTRACT

Atomistic molecular dynamics (MD) simulations coupled with the metadynamics technique were carried out to delineate the product (PPi.2Mg and IMP) release mechanisms from the active site of both human (Hs) and Plasmodium falciparum (Pf) hypoxanthine-guanine-(xanthine) phosphoribosyltransferase (HG(X)PRT). An early movement of PPi.2Mg from its binding site has been observed. The swinging motion of the Asp side chain (D134/D145) in the binding pocket facilitates the detachment of IMP, which triggers the opening of flexible loop II, the gateway to the bulk solvent. In PfHGXPRT, PPi.2Mg and IMP are seen to be released via the same path in all of the biased MD simulations. In HsHGPRT too, the product molecules follow similar routes from the active site; however, an alternate but minor escape route for PPi.2Mg has been observed in the human enzyme. Tyr 104 and Phe 186 in HsHGPRT and Tyr 116 and Phe 197 in PfHGXPRT are the key residues that mediate the release of IMP, whereas the motion of PPi.2Mg away from the reaction center is guided by the negatively charged Asp and Glu and a few positively charged residues (Lys and Arg) that line the product release channels. Mutations of a few key residues present in loop II of Trypanosoma cruzi (Tc) HGPRT have been shown to reduce the catalytic efficiency of the enzyme. Herein, in silico mutation of corresponding residues in loop II of HsHGPRT and PfHGXPRT resulted in partial opening of the flexible loop (loop II), thus exposing the active site to bulk water, which offers a rationale for the reduced catalytic activity of these two mutant enzymes. Investigations of the product release from these HsHGPRT and PfHGXPRT mutants delineate the role of these important residues in the enzymatic turnover.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/metabolism , Molecular Dynamics Simulation , Pentosyltransferases/metabolism , Plasmodium falciparum/enzymology , Catalytic Domain , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/genetics , Inosine Monophosphate/metabolism , Movement , Mutation , Pentosyltransferases/chemistry , Pentosyltransferases/genetics
14.
Anal Biochem ; 485: 46-8, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26050630

ABSTRACT

Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) provides a powerful tool for precise gene expression analysis. The accuracy of the results highly depends on careful selection of a reference gene for data normalization. HPRT1 (hypoxanthine phosphoribosyl transferase 1) is a frequently used housekeeping gene for normalizing relative expression values. However, the existence of processed pseudogenes for HPRT1 might interfere with reliable results obtained in qRT-PCR due to amplification of unintended products. Here, we designed a primer pair for pseudogene-free amplification of HPRT1 in qRT-PCR. We demonstrate that this primer pair specifically amplified HPRT1 messenger RNA (mRNA) sequence while avoiding coamplification of the pseudogenes.


Subject(s)
DNA Primers/chemistry , DNA Primers/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Cell Line , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , RNA, Messenger/chemistry
15.
Mol Pharmacol ; 84(4): 615-29, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23907213

ABSTRACT

6-Fluoro-3-hydroxy-2-pyrazinecarboxamide (T-705) is a novel antiviral compound with broad activity against influenza virus and diverse RNA viruses. Its active metabolite, T-705-ribose-5'-triphosphate (T-705-RTP), is recognized by influenza virus RNA polymerase as a substrate competing with GTP, giving inhibition of viral RNA synthesis and lethal virus mutagenesis. Which enzymes perform the activation of T-705 is unknown. We here demonstrate that human hypoxanthine guanine phosphoribosyltransferase (HGPRT) converts T-705 into its ribose-5'-monophosphate (RMP) prior to formation of T-705-RTP. The anti-influenza virus activity of T-705 and T-1105 (3-hydroxy-2-pyrazinecarboxamide; the analog lacking the 6-fluoro atom) was lost in HGPRT-deficient Madin-Darby canine kidney cells. This HGPRT dependency was confirmed in human embryonic kidney 293T cells undergoing HGPRT-specific gene knockdown followed by influenza virus ribonucleoprotein reconstitution. Knockdown for adenine phosphoribosyltransferase (APRT) or nicotinamide phosphoribosyltransferase did not change the antiviral activity of T-705 and T-1105. Enzymatic assays showed that T-705 and T-1105 are poor substrates for human HGPRT having Km(app) values of 6.4 and 4.1 mM, respectively. Formation of the RMP metabolites by APRT was negligible, and so was the formation of the ribosylated metabolites by human purine nucleoside phosphorylase. Phosphoribosylation and antiviral activity of the 2-pyrazinecarboxamide derivatives was shown to require the presence of the 3-hydroxyl but not the 6-fluoro substituent. The crystal structure of T-705-RMP in complex with human HGPRT showed how this compound binds in the active site. Since conversion of T-705 by HGPRT appears to be inefficient, T-705-RMP prodrugs may be designed to increase the antiviral potency of this new antiviral agent.


Subject(s)
Amides/chemistry , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/physiology , Pyrazines/chemistry , Amides/metabolism , Amides/pharmacology , Animals , Antiviral Agents/pharmacology , Crystallography, X-Ray , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Pyrazines/metabolism , Pyrazines/pharmacology
16.
J Biol Chem ; 287(5): 2997-3008, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22157001

ABSTRACT

Lesch-Nyhan disease and its attenuated variants are caused by mutations in the HPRT1 gene, which encodes the purine recycling enzyme hypoxanthine-guanine phosphoribosyltransferase. The mutations are heterogeneous, with more than 400 different mutations already documented. Prior efforts to correlate variations in the clinical phenotype with different mutations have suggested that milder phenotypes typically are associated with mutants that permit some residual enzyme function, whereas the most severe phenotype is associated with null mutants. However, multiple exceptions to this concept have been reported. In the current studies 44 HPRT1 mutations associated with a wide spectrum of clinical phenotypes were reconstructed by site-directed mutagenesis, the mutant enzymes were expressed in vitro and purified, and their kinetic properties were examined toward their substrates hypoxanthine, guanine, and phosphoribosylpyrophosphate. The results provide strong evidence for a correlation between disease severity and residual catalytic activity of the enzyme (k(cat)) toward each of its substrates as well as several mechanisms that result in exceptions to this correlation. There was no correlation between disease severity and the affinity of the enzyme for its substrates (K(m)). These studies provide a valuable model for understanding general principles of genotype-phenotype correlations in human disease, as the mechanisms involved are applicable to many other disorders.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/chemistry , Lesch-Nyhan Syndrome/enzymology , Genotype , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Kinetics , Lesch-Nyhan Syndrome/genetics , Mutagenesis, Site-Directed , Mutation , Phenotype , Substrate Specificity/genetics
17.
Biochem Biophys Res Commun ; 441(1): 114-9, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24129187

ABSTRACT

Lesch-Nyhan disease (LND) is a severe and incurable X-linked genetic syndrome caused by the deficiency of hypoxanthine-guanine phosphoribosyltransferase (HPRT), resulting in severe alterations of central nervous system, hyperuricemia and subsequent impaired renal functions. Therapeutic options consist in supportive care and treatments of complications, but the disease remains largely untreatable. Enzyme replacement of the malfunctioning cytosolic protein might represent a possible therapeutic approach for the LND treatment. Protein transduction domains, such as the TAT peptide derived from HIV TAT protein, have been used to transduce macromolecules into cells in vitro and in vivo. The present study was aimed to the generation of TAT peptide fused to human HPRT for cell transduction in enzyme deficient cells. Here we document the construction, expression and delivery of a functional HPRT enzyme into deficient cells by TAT transduction domain and by liposome mediated protein transfer. With this approach we demonstrate the correction of the enzymatic defect in HPRT deficient cells. Our data show for the first time the feasibility of the enzyme replacement therapy for the treatment of LND.


Subject(s)
HIV-1/metabolism , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/metabolism , Transduction, Genetic , tat Gene Products, Human Immunodeficiency Virus/metabolism , Genetic Complementation Test , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , Kinetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism
18.
Mol Genet Metab ; 110(3): 268-74, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24075303

ABSTRACT

We describe a family of seven boys affected by Lesch-Nyhan disease with various phenotypes. Further investigations revealed a mutation c.203T>C in the gene encoding HGprt of all members, with substitution of leucine to proline at residue 68 (p.Leu68Pro). Thus patients from this family display a wide variety of symptoms although sharing the same mutation. Mutant HGprt enzyme was prepared by site-directed mutagenesis and the kinetics of the enzyme revealed that the catalytic activity of the mutant was reduced, in association with marked reductions in the affinity towards phosphoribosylpyrophosphate (PRPP). Its Km for PRPP was increased 215-fold with hypoxanthine as substrate and 40-fold with guanine as substrate with associated reduced catalytic potential. Molecular modeling confirmed that the most prominent defect was the dramatically reduced affinity towards PRPP. Our studies suggest that the p.Leu68Pro mutation has a strong impact on PRPP binding and on stability of the active conformation. This suggests that factors other than HGprt activity per se may influence the phenotype of Lesch-Nyhan patients.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Phenotype , Adolescent , Adult , Amino Acid Substitution , Child , Codon , Enzyme Activation , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , Kinetics , Lesch-Nyhan Syndrome/diagnosis , Lesch-Nyhan Syndrome/genetics , Lesch-Nyhan Syndrome/metabolism , Male , Middle Aged , Models, Molecular , Mutation , Pedigree , Protein Binding , Protein Conformation , Protein Stability , Young Adult
19.
Biochemistry ; 50(19): 4184-93, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21486037

ABSTRACT

Enzymatic efficiency and structural discrimination of substrates from nonsubstrate analogues are attributed to the precise assembly of binding pockets. Many enzymes have the additional remarkable ability to recognize several substrates. These apparently paradoxical attributes are ascribed to the structural plasticity of proteins. A partially defined active site acquires complementarity upon encountering the substrate and completing the assembly. Human hypoxanthine guanine phosphoribosyltransferase (hHGPRT) catalyzes the phosphoribosylation of guanine and hypoxanthine, while the Plasmodium falciparum HGPRT (PfHGPRT) acts on xanthine as well. Reasons for the observed differences in substrate specificities of the two proteins are not clear. We used ultraviolet resonance Raman spectroscopy to study the complexes of HGPRT with products (IMP, GMP, and XMP), in both organisms, in resonance with the purine nucleobase electronic absorption. This led to selective enhancement of vibrations of the purine ring over those of the sugar-phosphate backbone and protein. Spectra of bound nucleotides show that HGPRT distorts the structure of the nucleotides. The distorted structure resembles that of the deprotonated nucleotide. We find that the two proteins assemble similar active sites for their common substrates. While hHGPRT does not bind XMP, PfHGPRT perturbs the pK(a) of bound XMP. The results were compared with the mutant form of hHGPRT that catalyzed xanthine but failed to perturb the pK(a) of XMP.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/metabolism , Purine Nucleotides/chemistry , Purine Nucleotides/metabolism , Ribonucleotides/chemistry , Ribonucleotides/metabolism , Animals , Catalytic Domain/genetics , Deuterium Exchange Measurement , Guanosine Monophosphate/chemistry , Humans , Hydrogen-Ion Concentration , Hypoxanthine Phosphoribosyltransferase/genetics , Inosine Monophosphate/chemistry , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Protein Binding/genetics , Spectrum Analysis, Raman , Substrate Specificity/genetics , Toxoplasma/enzymology , Xanthine
20.
J Med Chem ; 64(9): 5710-5729, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33891818

ABSTRACT

Helicobacter pylori (Hp) is a human pathogen that lives in the gastric mucosa of approximately 50% of the world's population causing gastritis, peptic ulcers, and gastric cancer. An increase in resistance to current drugs has sparked the search for new Hp drug targets and therapeutics. One target is the disruption of nucleic acid production, which can be achieved by impeding the synthesis of 6-oxopurine nucleoside monophosphates, the precursors of DNA and RNA. These metabolites are synthesized by Hp xanthine-guanine-hypoxanthine phosphoribosyltransferase (XGHPRT). Here, nucleoside phosphonates have been evaluated, which inhibit the activity of this enzyme with Ki values as low as 200 nM. The prodrugs of these compounds arrest the growth of Hp at a concentration of 50 µM in cell-based assays. The kinetic properties of HpXGHPRT have been determined together with its X-ray crystal structure in the absence and presence of 9-[(N-3-phosphonopropyl)-aminomethyl-9-deazahypoxanthine, providing a basis for new antibiotic development.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/metabolism , Pentosyltransferases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/pathology , Helicobacter Infections/drug therapy , Helicobacter Infections/pathology , Helicobacter pylori/drug effects , Helicobacter pylori/enzymology , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/metabolism , Hypoxanthines/chemistry , Hypoxanthines/metabolism , Hypoxanthines/pharmacology , Hypoxanthines/therapeutic use , Kinetics , Molecular Dynamics Simulation , Organophosphonates/chemistry , Organophosphonates/metabolism , Organophosphonates/pharmacology , Organophosphonates/therapeutic use , Pentosyltransferases/chemistry , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/pharmacology , Prodrugs/therapeutic use , Sequence Alignment , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL