61 results on '"Chitnumsub P"'
Search Results
2. Mechanism-guided tunnel engineering to increase the efficiency of a flavin-dependent halogenase
- Author
-
Prakinee, Kridsadakorn, Phintha, Aisaraphon, Visitsatthawong, Surawit, Lawan, Narin, Sucharitakul, Jeerus, Kantiwiriyawanitch, Chadaporn, Damborsky, Jiri, Chitnumsub, Penchit, van Pée, Karl-Heinz, and Chaiyen, Pimchai
- Published
- 2022
- Full Text
- View/download PDF
3. Thermoresponsive C22 phage stiffness modulates the phage infectivity
- Author
-
Sae-Ueng, Udom, Bhunchoth, Anjana, Phironrit, Namthip, Treetong, Alongkot, Sapcharoenkun, Chaweewan, Chatchawankanphanich, Orawan, Leartsakulpanich, Ubolsree, and Chitnumsub, Penchit
- Published
- 2022
- Full Text
- View/download PDF
4. Enhancement of catalytic activity and alkaline stability of cellobiohydrolase by structure-based protein engineering
- Author
-
Prabmark, Kanoknart, Boonyapakron, Katewadee, Bunterngsook, Benjarat, Arunrattanamook, Nattapol, Uengwetwanit, Tanaporn, Chitnumsub, Penchit, and Champreda, Verawat
- Published
- 2022
- Full Text
- View/download PDF
5. C22 podovirus infectivity is associated with intermediate stiffness
- Author
-
Sae-Ueng, Udom, Bhunchoth, Anjana, Phironrit, Namthip, Treetong, Alongkot, Sapcharoenkun, Chaweewan, Chatchawankanphanich, Orawan, Leartsakulpanich, Ubolsree, and Chitnumsub, Penchit
- Published
- 2020
- Full Text
- View/download PDF
6. Avian influenza A/H5N1 neuraminidase expressed in yeast with a functional head domain
- Author
-
Yongkiettrakul, S., Boonyapakron, K., Jongkaewwattana, A., Wanitchang, A., Leartsakulpanich, U., Chitnumsub, P., Eurwilaichitr, L., and Yuthavong, Y.
- Published
- 2009
- Full Text
- View/download PDF
7. Formation of catalytically active cross-species heterodimers of thymidylate synthase from Plasmodium falciparum and Plasmodium vivax
- Author
-
Chanama, Manee, Chanama, Suchart, Shaw, Philip J., Chitnumsub, Penchit, Leartsakulpanich, Ubolsree, and Yuthavong, Yongyuth
- Published
- 2011
- Full Text
- View/download PDF
8. Malarial (Plasmodium falciparum) dihydrofolate reductase-thymidylate synthase: structural basis for antifolate resistance and development of effective inhibitors
- Author
-
YUTHAVONG, Y., YUVANIYAMA, J., CHITNUMSUB, P., VANICHTANANKUL, J., CHUSACULTANACHAI, S., TARNCHOMPOO, B., VILAIVAN, T., and KAMCHONWONGPAISAN, S
- Published
- 2005
9. Synthesis of beta-sheet peptides and proteins incorporating templates
- Author
-
Chitnumsub, P., Deechanghit, S., Kaul, R., Schneide, J. P., Tsang, K. Y., Moretto, Alessandro, Bekele, H., Labrenz, S. R., Lashuel, H. A., and Kelly, W. J.
- Published
- 2002
10. Inhibitors of Plasmodial SerineHydroxymethyltransferase(SHMT): Cocrystal Structures of Pyrazolopyrans with Potent Blood-and Liver-Stage Activities.
- Author
-
Matthias C. Witschel, Matthias Rottmann, Anatol Schwab, Ubolsree Leartsakulpanich, Penchit Chitnumsub, Michael Seet, Sandro Tonazzi, Geoffrey Schwertz, Frank Stelzer, Thomas Mietzner, Case McNamara, Frank Thater, Céline Freymond, Aritsara Jaruwat, Chatchadaporn Pinthong, Pinpunya Riangrungroj, Mouhssin Oufir, Matthias Hamburger, Pascal Mäser, and LauraM. Sanz-Alonso
- Published
- 2015
- Full Text
- View/download PDF
11. Antimalarial Inhibitors Targeting Serine Hydroxymethyltransferase (SHMT) with in Vivo Efficacy and Analysis of their Binding Mode Based on X-ray Cocrystal Structures.
- Author
-
Schwertz, Geoffrey, Witschel, Matthias C., Rottmann, Matthias, Bonnert, Roger, Leartsakulpanich, Ubolsree, Chitnumsub, Penchit, Jaruwat, Aritsara, Ittarat, Wanwipa, Schäfer, Anja, Aponte, Raphael A., Charman, Susan A., White, Karen L., Kundu, Abhijit, Sadhukhan, Surajit, Lloyd, Mel, Freiberg, Gail M., Srikumaran, Myron, Siggel, Marc, Zwyssig, Adrian, and Chaiyen, Pimchai
- Published
- 2017
- Full Text
- View/download PDF
12. ChemInform Abstract: Improved Synthesis of the Boc and Fmoc Derivatives of 4-(2′-Aminoethyl) -6-dibenzofuranpropionic Acid: An Unnatural Amino Acid That Nucleates . beta.-Sheet Folding.
- Author
-
BEKELE, H., NESLONEY, C. L., MCWILLIAMS, K. W., ZACHARIAS, N. M., CHITNUMSUB, P., and KELLY, J. W.
- Published
- 1997
- Full Text
- View/download PDF
13. On the Mechanisms of Hypohalous Acid Formation and Electrophilic Halogenation by Non-Native Halogenases.
- Author
-
Prakinee K, Lawan N, Phintha A, Visitsatthawong S, Chitnumsub P, Jitkaroon W, and Chaiyen P
- Subjects
- Oxidoreductases metabolism, Oxidoreductases chemistry, Kinetics, Hydrogen Peroxide metabolism, Hydrogen Peroxide chemistry, Flavins metabolism, Flavins chemistry, Hydrolases metabolism, Hydrolases chemistry, Mixed Function Oxygenases metabolism, Mixed Function Oxygenases chemistry, Halogenation
- Abstract
Enzymatic electrophilic halogenation is a mild tool for functionalization of diverse organic compounds. Only a few groups of native halogenases are capable of catalyzing such a reaction. In this study, we used a mechanism-guided strategy to discover the electrophilic halogenation activity catalyzed by non-native halogenases. As the ability to form a hypohalous acid (HOX) is key for halogenation, flavin-dependent monooxygenases/oxidases capable of forming C4a-hydroperoxyflavin (Fl
C4a-OOH ), such as dehalogenase, hydroxylases, luciferase and pyranose-2-oxidase (P2O), and flavin reductase capable of forming H2 O2 were explored for their abilities to generate HOX in situ. Transient kinetic analyses using stopped-flow spectrophotometry/fluorometry and product analysis indicate that FlC4a-OOH in dehalogenases, selected hydroxylases and luciferases, but not in P2O can form HOX; however, the HOX generated from FlC4a-OOH cannot halogenate their substrates. Remarkably, in situ H2 O2 generated by P2O can form HOI and also iodinate various compounds. Because not all enzymes capable of forming FlC4a-OOH can react with halides to form HOX, QM/MM calculations, site-directed mutagenesis and structural analysis were carried out to elucidate the mechanism underlying HOX formation and characterize the active site environment. Our findings shed light on identifying new halogenase scaffolds besides the currently known enzymes and have invoked a new mode of chemoenzymatic halogenation., (© 2024 The Authors. Angewandte Chemie International Edition published by Wiley-VCH GmbH.)- Published
- 2024
- Full Text
- View/download PDF
14. Structure and biochemical characterization of an extradiol 3,4-dihydroxyphenylacetate 2,3-dioxygenase from Acinetobacter baumannii.
- Author
-
Pimviriyakul P, Buttranon S, Soithongcharoen S, Supawatkon C, Disayabootr K, Watthaisong P, Tinikul R, Jaruwat A, Chaiyen P, Chitnumsub P, and Maenpuen S
- Abstract
3,4-Dihydroxyphenylacetate (DHPA) 2,3-dioxygenase (EC 1.13.11.15) from Acinetobacter baumannii (AbDHPAO) is an enzyme that catalyzes the 2,3-extradiol ring-cleavage of DHPA in the p-hydroxyphenylacetate (HPA) degradation pathway. While the biochemical reactions of various DHPAOs have been reported, only structures of DHPAO from Brevibacterium fuscum and their homologs are available. Here, we report the X-ray structure and biochemical characterization of an Fe
2+ -specific AbDHPAO that shares 12% sequence identity to the enzyme from B. fuscum. The 1.8 Å X-ray structure of apo-AbDHPAO was determined with four subunits per asymmetric unit, consistent with a homotetrameric structure. Interestingly, the αβ-sandwiched fold of the AbDHPAO subunit is different from the dual β-barrel-like motif of the well-characterized B. fuscum DHPAO structures; instead, it is similar to the structures of non-DHPA extradiol dioxygenases from Comamonas sp. and Sphingomonas paucimobilis. Similarly, these extradiol dioxygenases share the same chemistry owing to a conserved 2-His-1-carboxylate catalytic motif. Structure analysis and molecular docking suggested that the Fe2+ cofactor and substrate binding sites consist of the conserved residues His12, His57, and Glu238 forming a 2-His-1-carboxylate motif ligating to Fe2+ and DHPA bound with Fe2+ in an octahedral coordination. In addition to DHPA, AbDHPAO can also use other 3,4-dihydroxyphenylacetate derivatives with different aliphatic carboxylic acid substituents as substrates, albeit with low reactivity. Altogether, this report provides a better understanding of the structure and biochemical properties of AbDHPAO and its homologs, which is advancing further modification of DHPAO in future applications., Competing Interests: Declaration of competing interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
15. Mangiferin is a new potential antimalarial and anticancer drug for targeting serine hydroxymethyltransferase.
- Author
-
Maenpuen S, Mee-Udorn P, Pinthong C, Athipornchai A, Phiwkaow K, Watchasit S, Pimviriyakul P, Rungrotmongkol T, Tinikul R, Leartsakulpanich U, and Chitnumsub P
- Subjects
- Humans, Glycine Hydroxymethyltransferase, Molecular Docking Simulation, Serine chemistry, Antimalarials pharmacology, Xanthones pharmacology, Folic Acid Antagonists, Antineoplastic Agents pharmacology
- Abstract
Mangiferin, a polyphenolic xanthone glycoside found in various botanical sources, including mango (Mangifera indica L.) leaves, can exhibit a variety of bioactivities. Although mangiferin has been reported to inhibit many targets, none of the studies have investigated the inhibition of serine hydroxymethyltransferase (SHMT), an attractive target for antimalarial and anticancer drugs. SHMT, one of the key enzymes in the deoxythymidylate synthesis cycle, catalyzes the reversible conversion of l-serine and (6S)-tetrahydrofolate (THF) into glycine and 5,10-methylene THF. Here, in vitro and in silico studies were used to probe how mangiferin isolated from mango leaves inhibits Plasmodium falciparum and human cytosolic SHMTs. The inhibition kinetics at pH 7.5 revealed that mangiferin is a competitive inhibitor against THF for enzymes from both organisms. Molecular docking and molecular dynamic (MD) simulations demonstrated the inhibitory effects of the deprotonated forms of mangiferin, specifically the C
6 -O- species and its resonance C9 -O- species appearing at pH 7.5, combined with two docked poses, either a xanthone or glucose moiety, placed inside the THF-binding pocket. The MD analysis revealed that both C6 -O- and its resonance-stabilized C9 -O- species can favorably bind to SHMT in a similar fashion to THF, supporting the THF competitive inhibition of mangiferin. In addition, characterization of the proton dissociation equilibria of isolated mangiferin revealed that only three hydroxy groups of the xanthone moiety, C6 -OH, C3 -OH, and C7 -OH, underwent varying degrees of deprotonation with pKa values of 6.38 ± 0.11, 8.21 ± 0.35, and 12.37 ± 0.30, respectively, while C1 -OH remained protonated. Altogether, our findings demonstrate a new bioactivity of mangiferin and provide the basis for the future development of mangiferin as a potent antimalarial and anticancer drug., Competing Interests: Declaration of competing interest The authors declare that they have no conflicts of interest with the contents of this article. The process to isolate mangiferin from mango (Mangifera indica L.) leaves was filed with the Department of Intellectual Property, Thailand, and received a Thai petty patent no. 20093 (2022–2025)., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
16. Dual role of azo compounds in inhibiting Plasmodium falciparum adenosine deaminase and hemozoin biocrystallization.
- Author
-
Kuaprasert B, Chitnumsub P, Leartsakulpanich U, Riangrungroj P, Suginta W, Leelayoova S, Mungthin M, Sitthichot N, Rattanabunyong S, Kiriwan D, and Choowongkomon K
- Subjects
- Adenosine Deaminase, Biomineralization, Chloroquine pharmacology, Drug Resistance, Ligands, Antimalarials pharmacology, Azo Compounds pharmacology, Plasmodium falciparum drug effects, Adenosine Deaminase Inhibitors pharmacology
- Abstract
Protein-ligand (GOLD) docking of the NCI compounds into the ligand-binding site of Plasmodium falciparum adenosine deaminase (PfADA) identified three most active azo compounds containing 4-[(4-hydroxy-2-oxo-1H-quinolin-3-yl) moiety. These compounds showed IC
50 of 3.7-15.4 μM against PfADA, as well as inhibited the growth of P. falciparum strains 3D7 (chloroquine (CQ)-sensitive) and K1 (CQ-resistant) with IC50 of 1.8-3.1 and 1.7-3.6 μM, respectively. The identified compounds have structures similar to the backbone structure (4-N-(7-chloroquinolin-4-yl)) in CQ, and NSC45545 could mimic CQ by inhibiting the bioformation of hemozoin in parasitic food vacuole. The amount of in situ hemozoin in the ring-stage parasite was determined using a combination of synchrotron transmission Fourier transform infrared microspectroscopy and Principal Component Analysis. Stretching of the C-O bond of hemozoin propionate group measured at 1220-1210 cm-1 in untreated intraerythrocytic P. falciparum strains 3D7 and K1 was disappeared following treatment with 1.85 and 1.74 μM NSC45545, similar to those treated with 0.02 and 0.13 μM CQ, respectively. These findings indicate a novel dual function of 4-[(4-hydroxy-2-oxo-1H-quinolin-3-yl) azo compounds in inhibiting both PfADA and in situ hemozoin biocrystallization. These lead compounds hold promise for further development of new antimalarial therapeutics that could delay the onset of parasitic drug resistance., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
17. Catalytic and structural insights into a stereospecific and thermostable Class II aldolase HpaI from Acinetobacter baumannii.
- Author
-
Watthaisong P, Binlaeh A, Jaruwat A, Lawan N, Tantipisit J, Jaroensuk J, Chuaboon L, Phonbuppha J, Tinikul R, Chaiyen P, Chitnumsub P, and Maenpuen S
- Subjects
- Bacterial Proteins, Catalysis, Catalytic Domain, Crystallography, X-Ray, Enzyme Stability, Substrate Specificity, Acinetobacter baumannii enzymology, Calcium chemistry, Fructose-Bisphosphate Aldolase chemistry, Zinc chemistry
- Abstract
Aldolases catalyze the reversible reactions of aldol condensation and cleavage and have strong potential for the synthesis of chiral compounds, widely used in pharmaceuticals. Here, we investigated a new Class II metal aldolase from the p-hydroxyphenylacetate degradation pathway in Acinetobacter baumannii, 4-hydroxy-2-keto-heptane-1,7-dioate aldolase (AbHpaI), which has various properties suitable for biocatalysis, including stereoselectivity/stereospecificity, broad aldehyde utilization, thermostability, and solvent tolerance. Notably, the use of Zn
2+ by AbHpaI as a native cofactor is distinct from other enzymes in this class. AbHpaI can also use other metal ion (M2+ ) cofactors, except Ca2+ , for catalysis. We found that Zn2+ yielded the highest enzyme complex thermostability (Tm of 87 °C) and solvent tolerance. All AbHpaI•M2+ complexes demonstrated preferential cleavage of (4R)-2-keto-3-deoxy-D-galactonate ((4R)-KDGal) over (4S)-2-keto-3-deoxy-D-gluconate ((4S)-KDGlu), with AbHpaI•Zn2+ displaying the highest R/S stereoselectivity ratio (sixfold higher than other M2+ cofactors). For the aldol condensation reaction, AbHpaI•M2+ only specifically forms (4R)-KDGal and not (4S)-KDGlu and preferentially catalyzes condensation rather than cleavage by ∼40-fold. Based on 11 X-ray structures of AbHpaI complexed with M2+ and ligands at 1.85 to 2.0 Å resolution, the data clearly indicate that the M2+ cofactors form an octahedral geometry with Glu151 and Asp177, pyruvate, and water molecules. Moreover, Arg72 in the Zn2+ -bound form governs the stereoselectivity/stereospecificity of AbHpaI. X-ray structures also show that Ca2+ binds at the trimer interface via interaction with Asp51. Hence, we conclude that AbHpaI•Zn2+ is distinctive from its homologues in substrate stereospecificity, preference for aldol formation over cleavage, and protein robustness, and is attractive for biocatalytic applications., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
18. Structural insights into a flavin-dependent dehalogenase HadA explain catalysis and substrate inhibition via quadruple π-stacking.
- Author
-
Pimviriyakul P, Jaruwat A, Chitnumsub P, and Chaiyen P
- Subjects
- Crystallography, X-Ray, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Bacterial Proteins genetics, Flavin-Adenine Dinucleotide metabolism, Flavin-Adenine Dinucleotide chemistry, Substrate Specificity, Flavins metabolism, Flavins chemistry, Mixed Function Oxygenases chemistry, Mixed Function Oxygenases metabolism, Mixed Function Oxygenases genetics, Catalysis, Mutagenesis, Site-Directed, Binding Sites, Models, Molecular, Nitrophenols metabolism, Nitrophenols chemistry
- Abstract
HadA is a flavin-dependent monooxygenase catalyzing hydroxylation plus dehalogenation/denitration, which is useful for biodetoxification and biodetection. In this study, the X-ray structure of wild-type HadA (HadA
WT ) co-complexed with reduced FAD (FADH- ) and 4-nitrophenol (4NP) (HadAWT -FADH- -4NP) was solved at 2.3-Å resolution, providing the first full package (with flavin and substrate bound) structure of a monooxygenase of this type. Residues Arg101, Gln158, Arg161, Thr193, Asp254, Arg233, and Arg439 constitute a flavin-binding pocket, whereas the 4NP-binding pocket contains the aromatic side chain of Phe206, which provides π-π stacking and also is a part of the hydrophobic pocket formed by Phe155, Phe286, Thr449, and Leu457. Based on site-directed mutagenesis and stopped-flow experiments, Thr193, Asp254, and His290 are important for C4a-hydroperoxyflavin formation with His290, also serving as a catalytic base for hydroxylation. We also identified a novel structural motif of quadruple π-stacking (π-π-π-π) provided by two 4NP and two Phe441 from two subunits. This motif promotes 4NP binding in a nonproductive dead-end complex, which prevents C4a-hydroperoxy-FAD formation when HadA is premixed with aromatic substrates. We also solved the structure of the HadAPhe441Val -FADH- -4NP complex at 2.3-Å resolution. Although 4NP can still bind to this variant, the quadruple π-stacking motif was disrupted. All HadAPhe441 variants lack substrate inhibition behavior, confirming that quadruple π-stacking is a main cause of dead-end complex formation. Moreover, the activities of these HadAPhe441 variants were improved by ⁓20%, suggesting that insights gained from the flavin-dependent monooxygenases illustrated here should be useful for future improvement of HadA's biocatalytic applications., Competing Interests: Conflict of interest The authors declare that they have no conflict of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
19. Dissecting the low catalytic capability of flavin-dependent halogenases.
- Author
-
Phintha A, Prakinee K, Jaruwat A, Lawan N, Visitsatthawong S, Kantiwiriyawanitch C, Songsungthong W, Trisrivirat D, Chenprakhon P, Mulholland A, van Pée KH, Chitnumsub P, and Chaiyen P
- Subjects
- Catalysis, Crystallography, X-Ray, Flavin-Adenine Dinucleotide metabolism, Halogenation, Hydrogen Peroxide metabolism, Kinetics, Models, Molecular, Protein Conformation, Flavins metabolism, Oxidoreductases metabolism
- Abstract
Although flavin-dependent halogenases (FDHs) are attractive biocatalysts, their practical applications are limited because of their low catalytic efficiency. Here, we investigated the reaction mechanisms and structures of tryptophan 6-halogenase (Thal) from Streptomyces albogriseolus using stopped-flow, rapid-quench flow, quantum/mechanics molecular mechanics calculations, crystallography, and detection of intermediate (hypohalous acid [HOX]) liberation. We found that the key flavin intermediate, C4a-hydroperoxyflavin (C4aOOH-FAD), formed by Thal and other FDHs (tryptophan 7-halogenase [PrnA] and tryptophan 5-halogenase [PyrH]), can react with I
- , Br- , and Cl- but not F- to form C4a-hydroxyflavin and HOX. Our experiments revealed that I- reacts with C4aOOH-FAD the fastest with the lowest energy barrier and have shown for the first time that a significant amount of the HOX formed leaks out as free HOX. This leakage is probably a major cause of low product coupling ratios in all FDHs. Site-saturation mutagenesis of Lys79 showed that changing Lys79 to any other amino acid resulted in an inactive enzyme. However, the levels of liberated HOX of these variants are all similar, implying that Lys79 probably does not form a chloramine or bromamine intermediate as previously proposed. Computational calculations revealed that Lys79 has an abnormally lower pKa compared with other Lys residues, implying that the catalytic Lys may act as a proton donor in catalysis. Analysis of new X-ray structures of Thal also explains why premixing of FDHs with reduced flavin adenine dinucleotide generally results in abolishment of C4aOOH-FAD formation. These findings reveal the hidden factors restricting FDHs capability which should be useful for future development of FDHs applications., Competing Interests: Conflict of interest The authors declare that they have no conflict of interest with the contents of this article., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
20. Enhancement of catalytic performance of a metagenome-derived thermophilic oligosaccharide-specific xylanase by binding module removal and random mutagenesis.
- Author
-
Boonyapakron K, Chitnumsub P, Kanokratana P, and Champreda V
- Subjects
- Cellulose metabolism, Gene Library, Hydrolysis, Kinetics, Saccharum metabolism, Substrate Specificity, Xylans metabolism, Biocatalysis, Endo-1,4-beta Xylanases genetics, Endo-1,4-beta Xylanases metabolism, Metagenome, Mutagenesis, Oligosaccharides metabolism, Temperature
- Abstract
Xylo-oligosaccharide (XO) is a promising pre-biotic with applications in food, feed and healthcare products. XO can be produced by enzymatic digestion of xylan with xylanase. In this study, we aimed to improve the biochemical properties relevant to catalysis and kinetics of X11, a thermophilic glycosyl hydrolase (GH) family 11 endo-β-1,4-xylanase derived from a metagenomic library isolated from sugarcane bagasse, under high-temperature conditions preferred for XO synthesis. Removal of a carbohydrate-binding module (X11C) resulted in 6.5 fold greater catalytic efficiency. X11C was further improved by a Pro71Thr mutation in the X11P variant obtained from a random mutagenesis library, which exhibited 15.9 fold greater catalytic efficiency compared with wild-type X11 under the enzyme's optimal conditions of 80°C and pH 6.0. Homology modeling suggested that the improved performance of X11P could be attributed to formation of an extra H-bond between Thr71 and Ser75, which stabilizes the key catalytic residue Glu180 at the active pocket and β-sheet layers and agrees with the respective increase in melting temperature (T
m ) where X11P >X11C >X11 as determined by differential scanning fluorimetry. The X11P variant was tested for hydrolysis of beechwood xylan, which showed X6 as the major product followed by X3 and X4 XOs. The highest yield of 5.5 g total XOs product/mg enzyme was observed for X11P, equivalent to 3.7 fold higher than that of wild-type with XO production of >800 mg/g xylan. The X11P enzyme could be developed as a thermophilic biocatalyst for XO synthesis in biorefineries., (Copyright © 2020 The Society for Biotechnology, Japan. Published by Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
21. Real-time detection of changes in yeast plasma membrane potential using genetically encoded voltage indicator proteins.
- Author
-
Limapichat W, Pornthanakasem W, Satitthammachart C, Chitnumsub P, and Leartsakulpanich U
- Subjects
- Fluorescence, Luminescent Proteins genetics, Saccharomyces cerevisiae genetics, Cell Membrane physiology, Membrane Potentials, Membrane Proteins genetics, Saccharomyces cerevisiae physiology
- Abstract
In yeast, adaptation to varying conditions often requires proper regulation of the plasma membrane potential. To determine yeast membrane potential change, optical methods involving potentiometric dyes have been supplemental to the direct electrode-based method. However, the hydrophobic nature of the dyes and their slow distribution across the membrane still limits their utilization. Genetically encoded voltage indicator (GEVI) proteins employed in neuroscience offer a tantalizing alternative for monitoring yeast membrane potential change. In this work, several widely used GEVI proteins were assessed in Saccharomyces cerevisiae for their expression and function as a voltage reporter. Among them, only ArcLight and Accelerated Sensor of Action Potential (ASAP) proteins could be expressed and transported to the plasma membrane. While the voltage-sensing capability was demonstrated for both ArcLight and ASAP, ArcLight fluorescence was sensitive to the intracellular pH change concurrently with the voltage change. Therefore, we established that ASAP is the more suitable GEVI protein for reporting yeast membrane potential change. This voltage-sensing reporter for yeast based on ASAP offers a new effective strategy for real-time optical detection of yeast membrane potential change, which potentially facilitates many areas of yeast research including optimizing growth conditions for industrial use and investigating yeast ion transport system., (© FEMS 2020.)
- Published
- 2020
- Full Text
- View/download PDF
22. The structure of Plasmodium falciparum hydroxymethyldihydropterin pyrophosphokinase-dihydropteroate synthase reveals the basis of sulfa resistance.
- Author
-
Chitnumsub P, Jaruwat A, Talawanich Y, Noytanom K, Liwnaree B, Poen S, and Yuthavong Y
- Subjects
- Amino Acid Sequence, Antimalarials pharmacology, Catalytic Domain, Crystallography, X-Ray, Dihydropteroate Synthase metabolism, Diphosphotransferases metabolism, Humans, Malaria, Falciparum parasitology, Protein Conformation, Sequence Homology, Dihydropteroate Synthase chemistry, Diphosphotransferases chemistry, Drug Resistance genetics, Malaria, Falciparum drug therapy, Mutation, Plasmodium falciparum drug effects, Plasmodium falciparum enzymology
- Abstract
The clinical efficacy of sulfa drugs as antimalarials has declined owing to the evolution of resistance in Plasmodium falciparum (Pf) malaria parasites. In order to understand the basis of this resistance and to design more effective antimalarials, we have solved 13 structures of the bifunctional enzyme 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase (HPPK)-dihydropteroate synthase (DHPS) from wild-type (WT) P. falciparum and sulfa-resistant mutants, both as apoenzyme and as complexes with pteroate (PTA) and sulfa derivatives. The structures of these complexes show that PTA, which effectively inhibits both the WT and mutants, stays in active sites without steric constraint. In contrast, parts of the sulfa compounds situated outside of the substrate envelope are in the vicinity of the resistance mutations. Steric conflict between compound and mutant residue along with increased flexibility of loop D2 in the mutants can account for the reduced compound binding affinity to the mutants. Kinetic data show that the mutants have enhanced enzyme activity compared with the WT. These PfDHPS structural insights are critical for the design of novel, substrate envelope-compliant DHPS inhibitors that are less vulnerable to resistance mutations. DATABASES: The data reported in this paper have been deposited in the Protein Data Bank, www.wwpdb.org. PDB ID codes: 6JWQ for apoWT; 6JWR, 6JWS, and 6JWT for PTA complexes of WT, A437G (3D7), and V1/S; 6JWU, 6JWV, and 6JWW for STZ-DHP complexes of WT, 3D7, and V1/S; 6JWX, 6JWY, and 6JWZ for SDX-DHP complexes of WT, 3D7, and W2; 6KCK, 6KCL, and 6KCM for Pterin/pHBA complexes of WT, TN1, and W2., (© 2020 Federation of European Biochemical Societies.)
- Published
- 2020
- Full Text
- View/download PDF
23. Identification of a Hotspot Residue for Improving the Thermostability of a Flavin-Dependent Monooxygenase.
- Author
-
Pongpamorn P, Watthaisong P, Pimviriyakul P, Jaruwat A, Lawan N, Chitnumsub P, and Chaiyen P
- Subjects
- Amino Acid Sequence, Enzyme Stability, Mixed Function Oxygenases genetics, Molecular Dynamics Simulation, Mutation, Protein Conformation, Flavins metabolism, Mixed Function Oxygenases chemistry, Mixed Function Oxygenases metabolism, Temperature
- Abstract
HadA is a flavin-dependent monooxygenase that can catalyze the denitration and dehalogenation of a wide variety of toxicants such as pesticides. Although these enzymatic reactions are useful for bioremediation or biocatalysis, the application of HadA for these purposes is not yet possible because of its low thermostability. In this work we have engineered HadA to be more thermostable through the use of structural, in silico, and rational approaches. The X-ray structure of HadA was solved to obtain a reliable three-dimensional protein model for further prediction of thermostable variants. In silico analysis by using two bioinformatic tools-FireProt and Disulfide by Design-suggested 102 variants that we then further refined by applying rational criteria including the location of a particular residue and its nearby interactions, as well as other biophysical parameters to narrow down the list to six candidates. The G513Y variant was found to be an optimal engineered candidate because it has significantly improved stability relative to the wild-type enzyme and equivalent activity. G513Y has an activity half-life 72 (50 °C) and 160 times (45 °C) longer than that of the wild-type enzyme. Coupled together with thermostable reactions of reduced flavin and NADH-regenerating systems, the G513Y variant can be used to catalyze denitration of 4nitrophenol at 45 °C. Structure/sequence alignments of HadA and its homologues indicate that several flavin-dependent monooxygenases also contain amino acid residues homologous to the G513 of HadA, hence opening up the possibility of applying this engineering approach to improving their thermostabilities as well. Molecular dynamics (MD) simulations confirmed that the improved thermostability of the G513Y variant was due to aromatic hydrocarbon interactions between Y513 and N359, L347, G348, and F349., (© 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
24. A flap motif in human serine hydroxymethyltransferase is important for structural stabilization, ligand binding, and control of product release.
- Author
-
Ubonprasert S, Jaroensuk J, Pornthanakasem W, Kamonsutthipaijit N, Wongpituk P, Mee-Udorn P, Rungrotmongkol T, Ketchart O, Chitnumsub P, Leartsakulpanich U, Chaiyen P, and Maenpuen S
- Subjects
- Amino Acid Motifs, Binding Sites, Enzyme Stability, Glycine Hydroxymethyltransferase genetics, Glycine Hydroxymethyltransferase metabolism, Humans, Kinetics, Molecular Dynamics Simulation, Mutagenesis, Protein Binding, Protein Multimerization, Protein Structure, Quaternary, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins genetics, Substrate Specificity, Tetrahydrofolates chemistry, Tetrahydrofolates metabolism, Glycine Hydroxymethyltransferase chemistry, Ligands
- Abstract
Human cytosolic serine hydroxymethyltransferase (hcSHMT) is a promising target for anticancer chemotherapy and contains a flexible "flap motif" whose function is yet unknown. Here, using size-exclusion chromatography, analytical ultracentrifugation, small-angle X-ray scattering (SAXS), molecular dynamics (MD) simulations, and ligand-binding and enzyme-kinetic analyses, we studied the functional roles of the flap motif by comparing WT hcSHMT with a flap-deleted variant (hcSHMT/Δflap). We found that deletion of the flap results in a mixture of apo-dimers and holo-tetramers, whereas the WT was mostly in the tetrameric form. MD simulations indicated that the flap stabilizes structural compactness and thereby enhances oligomerization. The hcSHMT/Δflap variant exhibited different catalytic properties in (6 S )-tetrahydrofolate (THF)-dependent reactions compared with the WT but had similar activity in THF-independent aldol cleavage of β-hydroxyamino acid. hcSHMT/Δflap was less sensitive to THF inhibition than the WT ( K
i of 0.65 and 0.27 mm THF at pH 7.5, respectively), and the THF dissociation constant of the WT was also 3-fold lower than that of hcSHMT/Δflap, indicating that the flap is important for THF binding. hcSHMT/Δflap did not display the burst kinetics observed in the WT. These results indicate that, upon removal of the flap, product release is no longer the rate-limiting step, implying that the flap is important for controlling product release. The findings reported here improve our understanding of the functional roles of the flap motif in hcSHMT and provide fundamental insight into how a flexible loop can be involved in controlling the enzymatic reactions of hcSHMT and other enzymes., (© 2019 Ubonprasert et al.)- Published
- 2019
- Full Text
- View/download PDF
25. Crystal structure of Plasmodium falciparum adenosine deaminase reveals a novel binding pocket for inosine.
- Author
-
Jaruwat A, Riangrungroj P, Ubonprasert S, Sae-Ueng U, Kuaprasert B, Yuthavong Y, Leartsakulpanich U, and Chitnumsub P
- Subjects
- Adenosine Deaminase genetics, Adenosine Deaminase metabolism, Adenosine Deaminase Inhibitors chemistry, Adenosine Deaminase Inhibitors pharmacology, Amino Acid Sequence, Amino Acid Substitution, Catalytic Domain, Crystallography, X-Ray, Drug Design, Humans, Inosine metabolism, Kinetics, Models, Molecular, Mutagenesis, Site-Directed, Plasmodium falciparum genetics, Protozoan Proteins genetics, Protozoan Proteins metabolism, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sequence Homology, Amino Acid, Substrate Specificity, Adenosine Deaminase chemistry, Plasmodium falciparum enzymology, Protozoan Proteins chemistry
- Abstract
Plasmodium falciparum (Pf), a malarial pathogen, can only synthesize purine nucleotides employing a salvage pathway because it lacks de novo biosynthesis. Adenosine deaminase (ADA), one of the three purine salvage enzymes, catalyzes the irreversible hydrolytic deamination of adenosine to inosine, which is further converted to GMP and AMP for DNA/RNA production. In addition to adenosine conversion, Plasmodium ADA also catalyzes the conversion of 5'-methylthioadenosine, derived from polyamine biosynthesis, into 5'-methylthioinosine whereas the human enzyme is not capable of this function. Here we report the crystal structure of a surface engineered PfADA at a resolution of 2.48 Å, together with results on kinetic studies of PfADA wild-type and active site variants. The structure reveals a novel inosine binding pocket linked to a distinctive PfADA substructure (residues 172-179) derived from a non-conserved gating helix loop (172-188) in Plasmodium spp. and other ADA enzymes. Variants of PfADA and human (h) ADA active site amino acids were generated in order to study their role in catalysis, including PfADA- Phe136, -Thr174, -Asp176, and -Leu179, and hADA-Met155, equivalent to PfADA-Asp176. PfADA-Leu179His showed no effect on kinetic parameters. However, kinetic results of PfADA-Asp176Met/Ala mutants and hADA-Met155Asp/Ala showed that the mutation reduced adenosine and 5'-methylthioadenosine substrate affinity in PfADA and k
cat in hADA, thereby reducing catalytic efficiency of the enzyme. Phe136Leu mutant showed increased Km (>10-fold) for both substrates whereas Thr174Ile/Ala only affected 5'-methylthioadenosine binding affinity. Together, the structure with the novel inosine binding pocket and the kinetic data provide insights for rational design of inhibitors against PfADA., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
26. Hybrid Inhibitors of Malarial Dihydrofolate Reductase with Dual Binding Modes That Can Forestall Resistance.
- Author
-
Tarnchompoo B, Chitnumsub P, Jaruwat A, Shaw PJ, Vanichtanankul J, Poen S, Rattanajak R, Wongsombat C, Tonsomboon A, Decharuangsilp S, Anukunwithaya T, Arwon U, Kamchonwongpaisan S, and Yuthavong Y
- Abstract
The S108N mutation of dihydrofolate reductase (DHFR) renders Plasmodium falciparum malaria parasites resistant to pyrimethamine through steric clash with the rigid side chain of the inhibitor. Inhibitors with flexible side chains can avoid this clash and retain effectiveness against the mutant. However, other mutations such as N108S reversion confer resistance to flexible inhibitors. We designed and synthesized hybrid inhibitors with two structural types in a single molecule, which are effective against both wild-type and multiple mutants of P. falciparum through their selective target binding, as demonstrated by X-ray crystallography. Furthermore, the hybrid inhibitors can forestall the emergence of new resistant mutants, as shown by selection of mutants resistant to hybrid compound BT1 from a diverse PfDHFR random mutant library expressed in a surrogate bacterial system. These results show that it is possible to develop effective antifolate antimalarials to which the range of parasite resistance mutations is greatly reduced., Competing Interests: The authors declare no competing financial interest.
- Published
- 2018
- Full Text
- View/download PDF
27. Potent Inhibitors of Plasmodial Serine Hydroxymethyltransferase (SHMT) Featuring a Spirocyclic Scaffold.
- Author
-
Schwertz G, Witschel MC, Rottmann M, Leartsakulpanich U, Chitnumsub P, Jaruwat A, Amornwatcharapong W, Ittarat W, Schäfer A, Aponte RA, Trapp N, Chaiyen P, and Diederich F
- Subjects
- Crystallography, X-Ray, Dose-Response Relationship, Drug, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Glycine Hydroxymethyltransferase metabolism, Humans, Indenes chemical synthesis, Indenes chemistry, Ligands, Models, Molecular, Molecular Structure, Oxindoles chemical synthesis, Oxindoles chemistry, Parasitic Sensitivity Tests, Plasmodium enzymology, Spiro Compounds chemical synthesis, Spiro Compounds chemistry, Structure-Activity Relationship, Enzyme Inhibitors pharmacology, Glycine Hydroxymethyltransferase antagonists & inhibitors, Indenes pharmacology, Oxindoles pharmacology, Plasmodium drug effects, Spiro Compounds pharmacology
- Abstract
With the discovery that serine hydroxymethyltransferase (SHMT) is a druggable target for antimalarials, the aim of this study was to design novel inhibitors of this key enzyme in the folate biosynthesis cycle. Herein, 19 novel spirocyclic ligands based on either 2-indolinone or dihydroindene scaffolds and featuring a pyrazolopyran core are reported. Strong target affinities for Plasmodium falciparum (Pf) SHMT (14-76 nm) and cellular potencies in the low nanomolar range (165-334 nm) were measured together with interesting selectivity against human cytosolic SHMT1 (hSHMT1). Four co-crystal structures with Plasmodium vivax (Pv) SHMT solved at 2.2-2.4 Å resolution revealed the key role of the vinylogous cyanamide for anchoring ligands within the active site. The spirocyclic motif in the molecules enforces the pyrazolopyran core to adopt a substantially more curved conformation than that of previous non-spirocyclic analogues. Finally, solvation of the spirocyclic lactam ring of the receptor-bound ligands is discussed., (© 2018 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2018
- Full Text
- View/download PDF
28. Conformational Aspects in the Design of Inhibitors for Serine Hydroxymethyltransferase (SHMT): Biphenyl, Aryl Sulfonamide, and Aryl Sulfone Motifs.
- Author
-
Schwertz G, Frei MS, Witschel MC, Rottmann M, Leartsakulpanich U, Chitnumsub P, Jaruwat A, Ittarat W, Schäfer A, Aponte RA, Trapp N, Mark K, Chaiyen P, and Diederich F
- Abstract
Malaria remains a major threat to mankind due to the perpetual emergence of resistance against marketed drugs. Twenty-one pyrazolopyran-based inhibitors bearing terminal biphenyl, aryl sulfonamide, or aryl sulfone motifs were synthesized and tested towards serine hydroxymethyltransferase (SHMT), a key enzyme of the folate cycle. The best ligands inhibited Plasmodium falciparum (Pf) and Arabidopsis thaliana (At) SHMT in target, as well as PfNF54 strains in cell-based assays in the low nanomolar range (18-56 nm). Seven co-crystal structures with P. vivax (Pv) SHMT were solved at 2.2-2.6 Å resolution. We observed an unprecedented influence of the torsion angle of ortho-substituted biphenyl moieties on cell-based efficacy. The peculiar lipophilic character of the sulfonyl moiety was highlighted in the complexes with aryl sulfonamide analogues, which bind in their preferred staggered orientation. The results are discussed within the context of conformational preferences in the ligands., (© 2017 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2017
- Full Text
- View/download PDF
29. Structure-based protein engineering for thermostable and alkaliphilic enhancement of endo-β-1,4-xylanase for applications in pulp bleaching.
- Author
-
Boonyapakron K, Jaruwat A, Liwnaree B, Nimchua T, Champreda V, and Chitnumsub P
- Subjects
- Biomass, Escherichia coli genetics, Hot Temperature, Hydrogen-Ion Concentration, Models, Molecular, Paper, Endo-1,4-beta Xylanases chemistry, Endo-1,4-beta Xylanases genetics, Protein Engineering methods, Recombinant Proteins chemistry, Recombinant Proteins genetics
- Abstract
In the pulp bleaching industry, enzymes with robust activity at high pH and temperatures are desirable for facilitating the pre-bleaching process with simplified processing and minimal use of chlorinated compounds. To engineer an enzyme for this purpose, we determined the crystal structure of the Xyn12.2 xylanase, a xylan-hydrolyzing enzyme derived from the termite gut symbiont metagenome, as the basis for structure-based protein engineering to improve Xyn12.2 stability in high heat and alkaline conditions. Engineered cysteine pairs that generated exterior disulfide bonds increased the k
cat of Xyn12.2 variants and melting temperature at all tested conditions. These improvements led to up to 4.2-fold increases in catalytic efficiency at pH 9.0, 50°C for 1h and up to 3-fold increases at 60°C. The most effective variants, XynTT and XynTTTE, exhibited 2-3-fold increases in bagasse hydrolysis at pH 9.0 and 60°C compared to the wild-type enzyme. Overall, engineering arginines and phenylalanines for increased pKa and hydrogen bonding improved enzyme catalytic efficiency at high stringency conditions. These modifications were the keys to enhancing thermostability and alkaliphilicity in our enzyme variants, with XynTT and XynTTTE being especially promising for their application to the pulp and paper industry., (Copyright © 2017 Elsevier B.V. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
30. Human and Plasmodium serine hydroxymethyltransferases differ in rate-limiting steps and pH-dependent substrate inhibition behavior.
- Author
-
Amornwatcharapong W, Maenpuen S, Chitnumsub P, Leartsakulpanich U, and Chaiyen P
- Subjects
- Amino Acid Motifs, Humans, Hydrogen-Ion Concentration, Species Specificity, Enzyme Inhibitors chemistry, Glycine Hydroxymethyltransferase antagonists & inhibitors, Glycine Hydroxymethyltransferase chemistry, Plasmodium falciparum enzymology, Plasmodium vivax enzymology, Protozoan Proteins antagonists & inhibitors, Protozoan Proteins chemistry
- Abstract
Serine hydroxymethyltransferase (SHMT), an essential enzyme for cell growth and development, catalyzes the transfer of -CH
2 OH from l-serine to tetrahydrofolate (THF) to form glycine and 5,10-methylenetetrahydrofolate (MTHF) which is used for nucleotide synthesis. Insights into the ligand binding and inhibition properties of human cytosolic SHMT (hcSHMT) and Plasmodium SHMT (PvSHMT) are crucial for designing specific drugs against malaria and cancer. The results presented here revealed strong and pH-dependent THF inhibition of hcSHMT. In contrast, in PvSHMT, THF inhibition and the influence of pH were not as pronounced. Ligand binding experiments performed at various pH values indicated that the hcSHMT:Gly complex binds THF more tightly at lower pH conditions, while the binding affinity of the PvSHMT:Gly complex for THF is not pH-dependent. Pre-steady state kinetic (rapid-quench) analysis of hcSHMT showed burst kinetics, indicating that glycine formation occurs fastest in the first turnover relative to the subsequent turnovers i.e. glycine release is the rate-limiting step in the hcSHMT reaction. All data suggest that excess THF likely binds E:Gly binary complex and forms the E:Gly:THF dead-end complex before glycine is released. A unique flap motif found in the structure of hcSHMT may be the key structural feature that imparts these described characteristics of hcSHMT., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
31. Role of Plasmodium vivax Dihydropteroate Synthase Polymorphisms in Sulfa Drug Resistance.
- Author
-
Pornthanakasem W, Riangrungroj P, Chitnumsub P, Ittarat W, Kongkasuriyachai D, Uthaipibull C, Yuthavong Y, and Leartsakulpanich U
- Subjects
- Animals, Diphosphotransferases genetics, Escherichia coli metabolism, Kinetics, Malaria, Vivax drug therapy, Malaria, Vivax parasitology, Mice, Mice, Inbred BALB C, Plasmids, Plasmodium berghei drug effects, Plasmodium berghei pathogenicity, Plasmodium vivax drug effects, Plasmodium vivax pathogenicity, Sulfadoxine pharmacology, Dihydropteroate Synthase genetics, Polymorphism, Genetic genetics
- Abstract
Dihydropteroate synthase (DHPS) is a known sulfa drug target in malaria treatment, existing as a bifunctional enzyme together with hydroxymethyldihydropterin pyrophosphokinase (HPPK). Polymorphisms in key residues of Plasmodium falciparum DHPS (PfDHPS) have been characterized and linked to sulfa drug resistance in malaria. Genetic sequencing of P. vivax dhps (Pvdhps) from clinical isolates has shown several polymorphisms at the positions equivalent to those in the Pfdhps genes conferring sulfa drug resistance, suggesting a mechanism for sulfa drug resistance in P. vivax similar to that seen in P. falciparum To characterize the role of polymorphisms in the PvDHPS in sulfa drug resistance, various mutants of recombinant PvHPPK-DHPS enzymes were expressed and characterized. Moreover, due to the lack of a continuous in vitro culture system for P. vivax parasites, a surrogate P. berghei model expressing Pvhppk-dhps genes was established to demonstrate the relationship between sequence polymorphisms and sulfa drug susceptibility and to test the activities of PvDHPS inhibitors on the transgenic parasites. Both enzyme activity and transgenic parasite growth were sensitive to sulfadoxine to different degrees, depending on the number of mutations that accumulated in DHPS. Ki values and 50% effective doses were higher for mutant PvDHPS enzymes than the wild-type enzymes. Altogether, the study provides the first evidence of sulfa drug resistance at the molecular level in P. vivax Furthermore, the enzyme inhibition assay and the in vivo screening system can be useful tools for screening new compounds for their activities against PvDHPS., (Copyright © 2016, American Society for Microbiology. All Rights Reserved.)
- Published
- 2016
- Full Text
- View/download PDF
32. Inhibitors of plasmodial serine hydroxymethyltransferase (SHMT): cocrystal structures of pyrazolopyrans with potent blood- and liver-stage activities.
- Author
-
Witschel MC, Rottmann M, Schwab A, Leartsakulpanich U, Chitnumsub P, Seet M, Tonazzi S, Schwertz G, Stelzer F, Mietzner T, McNamara C, Thater F, Freymond C, Jaruwat A, Pinthong C, Riangrungroj P, Oufir M, Hamburger M, Mäser P, Sanz-Alonso LM, Charman S, Wittlin S, Yuthavong Y, Chaiyen P, and Diederich F
- Subjects
- Administration, Oral, Animals, Antimalarials administration & dosage, Antimalarials pharmacokinetics, Chemistry Techniques, Synthetic, Crystallography, X-Ray, Drug Evaluation, Preclinical methods, Drug Resistance drug effects, Enzyme Inhibitors chemical synthesis, Female, Glycine Hydroxymethyltransferase chemistry, Glycine Hydroxymethyltransferase metabolism, Hep G2 Cells drug effects, Humans, Liver metabolism, Liver parasitology, Malaria, Falciparum drug therapy, Malaria, Falciparum parasitology, Mice, Inbred Strains, Mice, SCID, Microsomes, Liver drug effects, Organisms, Genetically Modified, Plasmodium berghei drug effects, Plasmodium berghei pathogenicity, Plasmodium falciparum enzymology, Plasmodium falciparum pathogenicity, Plasmodium vivax enzymology, Plasmodium vivax pathogenicity, Pyrazoles chemistry, Rats, Antimalarials chemistry, Antimalarials pharmacology, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Glycine Hydroxymethyltransferase antagonists & inhibitors, Plasmodium falciparum drug effects, Plasmodium vivax drug effects
- Abstract
Several of the enzymes related to the folate cycle are well-known for their role as clinically validated antimalarial targets. Nevertheless for serine hydroxymethyltransferase (SHMT), one of the key enzymes of this cycle, efficient inhibitors have not been described so far. On the basis of plant SHMT inhibitors from an herbicide optimization program, highly potent inhibitors of Plasmodium falciparum (Pf) and Plasmodium vivax (Pv) SHMT with a pyrazolopyran core structure were identified. Cocrystal structures of potent inhibitors with PvSHMT were solved at 2.6 Å resolution. These ligands showed activity (IC50/EC50 values) in the nanomolar range against purified PfSHMT, blood-stage Pf, and liver-stage P. berghei (Pb) cells and a high selectivity when assayed against mammalian cell lines. Pharmacokinetic limitations are the most plausible explanation for lack of significant activity of the inhibitors in the in vivo Pb mouse malaria model.
- Published
- 2015
- Full Text
- View/download PDF
33. Kinetic mechanism and the rate-limiting step of Plasmodium vivax serine hydroxymethyltransferase.
- Author
-
Maenpuen S, Amornwatcharapong W, Krasatong P, Sucharitakul J, Palfey BA, Yuthavong Y, Chitnumsub P, Leartsakulpanich U, and Chaiyen P
- Subjects
- Folic Acid chemistry, Glycine, Hydrogen-Ion Concentration, Kinetics, Protein Binding, Thermodynamics, Glycine Hydroxymethyltransferase chemistry, Plasmodium vivax enzymology, Protozoan Proteins chemistry, Serine chemistry
- Abstract
Serine hydroxymethyltransferase (SHMT) is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that catalyzes a hydroxymethyl group transfer from L-serine to tetrahydrofolate (H4folate) to yield glycine and 5,10-methylenetetrahydrofolate (CH2-H4folate). SHMT is crucial for deoxythymidylate biosynthesis and a target for antimalarial drug development. Our previous studies indicate that PvSHMT catalyzes the reaction via a ternary complex mechanism. To define the kinetic mechanism of this catalysis, we explored the PvSHMT reaction by employing various methodologies including ligand binding, transient, and steady-state kinetics as well as product analysis by rapid-quench and HPLC/MS techniques. The results indicate that PvSHMT can bind first to either L-serine or H4folate. The dissociation constants for the enzyme·L-serine and enzyme·H4folate complexes were determined as 0.18 ± 0.08 and 0.35 ± 0.06 mM, respectively. The amounts of glycine formed after single turnovers of different preformed binary complexes were similar, indicating that the reaction proceeds via a random-order binding mechanism. In addition, the rate constant of glycine formation measured by rapid-quench and HPLC/MS analysis is similar to the kcat value (1.09 ± 0.05 s(-1)) obtained from the steady-state kinetics, indicating that glycine formation is the rate-limiting step of SHMT catalysis. This information will serve as a basis for future investigation on species-specific inhibition of SHMT for antimalarial drug development., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
34. Structures of Plasmodium vivax serine hydroxymethyltransferase: implications for ligand-binding specificity and functional control.
- Author
-
Chitnumsub P, Jaruwat A, Riangrungroj P, Ittarat W, Noytanom K, Oonanant W, Vanichthanankul J, Chuankhayan P, Maenpuen S, Chen CJ, Chaiyen P, Yuthavong Y, and Leartsakulpanich U
- Subjects
- Binding Sites, Humans, Ligands, Models, Molecular, Plasmodium vivax chemistry, Plasmodium vivax metabolism, Protein Binding, Serine chemistry, Serine metabolism, Tetrahydrofolates chemistry, Tetrahydrofolates metabolism, Glycine Hydroxymethyltransferase chemistry, Glycine Hydroxymethyltransferase metabolism, Malaria, Vivax parasitology, Plasmodium vivax enzymology
- Abstract
Plasmodium parasites, the causative agent of malaria, rely heavily on de novo folate biosynthesis, and the enzymes in this pathway have therefore been explored extensively for antimalarial development. Serine hydroxymethyltransferase (SHMT) from Plasmodium spp., an enzyme involved in folate recycling and dTMP synthesis, has been shown to catalyze the conversion of L- and D-serine to glycine (Gly) in a THF-dependent reaction, the mechanism of which is not yet fully understood. Here, the crystal structures of P. vivax SHMT (PvSHMT) in a binary complex with L-serine and in a ternary complex with D-serine (D-Ser) and (6R)-5-formyltetrahydrofolate (5FTHF) provide clues to the mechanism underlying the control of enzyme activity. 5FTHF in the ternary-complex structure was found in the 6R form, thus differing from the previously reported structures of SHMT-Gly-(6S)-5FTHF from other organisms. This suggested that the presence of D-Ser in the active site can alter the folate-binding specificity. Investigation of binding in the presence of D-Ser and the (6R)- or (6S)-5FTHF enantiomers indicated that both forms of 5FTHF can bind to the enzyme but that only (6S)-5FTHF gives rise to a quinonoid intermediate. Likewise, a large surface area with a highly positively charged electrostatic potential surrounding the PvSHMT folate pocket suggested a preference for a polyglutamated folate substrate similar to the mammalian SHMTs. Furthermore, as in P. falciparum SHMT, a redox switch created from a cysteine pair (Cys125-Cys364) was observed. Overall, these results assert the importance of features such as stereoselectivity and redox status for control of the activity and specificity of PvSHMT.
- Published
- 2014
- Full Text
- View/download PDF
35. The structure of Plasmodium falciparum serine hydroxymethyltransferase reveals a novel redox switch that regulates its activities.
- Author
-
Chitnumsub P, Ittarat W, Jaruwat A, Noytanom K, Amornwatcharapong W, Pornthanakasem W, Chaiyen P, Yuthavong Y, and Leartsakulpanich U
- Subjects
- Amino Acid Sequence, Animals, Crystallization, Models, Molecular, Molecular Sequence Data, Oxidation-Reduction, Sequence Homology, Amino Acid, Spectrophotometry, Ultraviolet, Structure-Activity Relationship, X-Ray Diffraction, Glycine Hydroxymethyltransferase chemistry, Plasmodium falciparum enzymology
- Abstract
Plasmodium falciparum serine hydroxymethyltransferase (PfSHMT), an enzyme in the dTMP synthesis cycle, is an antimalarial target because inhibition of its expression or function has been shown to be lethal to the parasite. As the wild-type enzyme could not be crystallized, protein engineering of residues on the surface was carried out. The surface-engineered mutant PfSHMT-F292E was successfully crystallized and its structure was determined at 3 Å resolution. The PfSHMT-F292E structure is a good representation of PfSHMT as this variant revealed biochemical properties similar to those of the wild type. Although the overall structure of PfSHMT is similar to those of other SHMTs, unique features including the presence of two loops and a distinctive cysteine pair formed by Cys125 and Cys364 in the tetrahydrofolate (THF) substrate binding pocket were identified. These structural characteristics have never been reported in other SHMTs. Biochemical characterization and mutation analysis of these two residues confirm that they act as a disulfide/sulfhydryl switch to regulate the THF-dependent catalytic function of the enzyme. This redox switch is not present in the human enzyme, in which the cysteine pair is absent. The data reported here can be further exploited as a new strategy to specifically disrupt the activity of the parasite enzyme without interfering with the function of the human enzyme.
- Published
- 2014
- Full Text
- View/download PDF
36. Malarial dihydrofolate reductase as a paradigm for drug development against a resistance-compromised target.
- Author
-
Yuthavong Y, Tarnchompoo B, Vilaivan T, Chitnumsub P, Kamchonwongpaisan S, Charman SA, McLennan DN, White KL, Vivas L, Bongard E, Thongphanchang C, Taweechai S, Vanichtanankul J, Rattanajak R, Arwon U, Fantauzzi P, Yuvaniyama J, Charman WN, and Matthews D
- Subjects
- Animals, Antimalarials pharmacokinetics, Catalytic Domain genetics, Crystallography, X-Ray, Drug Design, Mice, Mice, SCID, Molecular Structure, Protein Conformation, Antimalarials chemistry, Antimalarials pharmacology, Folic Acid Antagonists metabolism, Models, Molecular, Plasmodium falciparum enzymology, Tetrahydrofolate Dehydrogenase chemistry, Tetrahydrofolate Dehydrogenase metabolism
- Abstract
Malarial dihydrofolate reductase (DHFR) is the target of antifolate antimalarial drugs such as pyrimethamine and cycloguanil, the clinical efficacy of which have been compromised by resistance arising through mutations at various sites on the enzyme. Here, we describe the use of cocrystal structures with inhibitors and substrates, along with efficacy and pharmacokinetic profiling for the design, characterization, and preclinical development of a selective, highly efficacious, and orally available antimalarial drug candidate that potently inhibits both wild-type and clinically relevant mutated forms of Plasmodium falciparum (Pf) DHFR. Important structural characteristics of P218 include pyrimidine side-chain flexibility and a carboxylate group that makes charge-mediated hydrogen bonds with conserved Arg122 (PfDHFR-TS amino acid numbering). An analogous interaction of P218 with human DHFR is disfavored because of three species-dependent amino acid substitutions in the vicinity of the conserved Arg. Thus, P218 binds to the active site of PfDHFR in a substantially different fashion from the human enzyme, which is the basis for its high selectivity. Unlike pyrimethamine, P218 binds both wild-type and mutant PfDHFR in a slow-on/slow-off tight-binding mode, which prolongs the target residence time. P218, when bound to PfDHFR-TS, resides almost entirely within the envelope mapped out by the dihydrofolate substrate, which may make it less susceptible to resistance mutations. The high in vivo efficacy in a SCID mouse model of P. falciparum malaria, good oral bioavailability, favorable enzyme selectivity, and good safety characteristics of P218 make it a potential candidate for further development.
- Published
- 2012
- Full Text
- View/download PDF
37. Combined spatial limitation around residues 16 and 108 of Plasmodium falciparum dihydrofolate reductase explains resistance to cycloguanil.
- Author
-
Vanichtanankul J, Taweechai S, Uttamapinant C, Chitnumsub P, Vilaivan T, Yuthavong Y, and Kamchonwongpaisan S
- Subjects
- Drug Resistance genetics, Mutation, Plasmodium falciparum genetics, Tetrahydrofolate Dehydrogenase chemistry, Tetrahydrofolate Dehydrogenase metabolism, Antimalarials pharmacology, Folic Acid Antagonists pharmacology, Plasmodium falciparum drug effects, Plasmodium falciparum enzymology, Proguanil pharmacology, Tetrahydrofolate Dehydrogenase genetics, Triazines pharmacology
- Abstract
Natural mutations of Plasmodium falciparum dihydrofolate reductase (PfDHFR) at A16V and S108T specifically confer resistance to cycloguanil (CYC) but not to pyrimethamine (PYR). In order to understand the nature of CYC resistance, the effects of various mutations at A16 on substrate and inhibitor binding were examined. Three series of mutations at A16 with or without the S108T/N mutation were generated. Only three mutants with small side chains at residue 16 (G, C, and S) were viable from bacterial complementation assay in the S108 series, whereas these three and an additional four mutants (T, V, M, and I) with slightly larger side chains were viable with simultaneous S108T mutation. Among these combinations, the A16V+S108T mutant was the most CYC resistant, and all of the S108T series ranged from being highly to moderately sensitive to PYR. In the S108N series, a strict requirement for alanine was observed at position 16. Crystal structure analyses reveal that in PfDHFR-TS variant T9/94 (A16V+S108T) complexed with CYC, the ligand has substantial steric conflicts with the side chains of both A16V and S108T, whereas in the complex with PYR, the ligand only showed mild conflict with S108T. CYC analogs designed to avoid such conflicts improved the binding affinity of the mutant enzymes. These results show that there is greater spatial limitation around the S108T/N residue when combined with the limitation imposed by A16V. The limitation of mutation of this series provides opportunities for drug design and development against antifolate-resistant malaria.
- Published
- 2012
- Full Text
- View/download PDF
38. Trypanosomal dihydrofolate reductase reveals natural antifolate resistance.
- Author
-
Vanichtanankul J, Taweechai S, Yuvaniyama J, Vilaivan T, Chitnumsub P, Kamchonwongpaisan S, and Yuthavong Y
- Subjects
- Amino Acid Sequence, Crystallography, X-Ray, Models, Molecular, Molecular Sequence Data, Protein Conformation, Sequence Alignment, Tetrahydrofolate Dehydrogenase chemistry, Tetrahydrofolate Dehydrogenase genetics, Drug Resistance, Folic Acid Antagonists pharmacology, Tetrahydrofolate Dehydrogenase metabolism, Trypanosoma brucei brucei enzymology
- Abstract
Dihydrofolate reductase (DHFR) is a potential drug target for Trypanosoma brucei, a human parasite, which is the causative agent for African sleeping sickness. No drug is available against this target, since none of the classical antifolates such as pyrimethamine (PYR), cycloguanil, or trimethoprim are effective as selective inhibitors of T. brucei DHFR (TbDHFR). In order to design effective drugs that target TbDHFR, co-crystal structures with bound antifolates were studied. On comparison with malarial Plasmodium falciparum DHFR (PfDHFR), the co-crystal structures of wild-type TbDHFR reveal greater structural similarities to a mutant PfDHFR causing antifolate resistance than the wild-type enzyme. TbDHFR imposes steric hindrance for rigid inhibitors like PYR around Thr86, which is equivalent to Ser108Asn of the malarial enzymes. In addition, a missing residue on TbDHFR active-site loop together with the presence of Ile51 widens its active site even further than the structural effect of Asn51Ile, which is observed in PfDHFR structures. The structural similarities are paralleled by the similarly poor affinities of the trypanosomal enzyme for rigid inhibitors. Mutations of TbDHFR at Thr86 resulted in 10-fold enhancement or 7-fold reduction in the rigid inhibitors affinities for Thr86Ser or Thr86Asn, respectively. The co-crystal structure of TbDHFR with a flexible antifolate WR99210 suggests that its greater affinity result from its ability to avoid such Thr86 clash and occupy the widened binding space similarly to what is observed in the PfDHFR structures. Natural resistance to antifolates of TbDHFR can therefore be explained, and potential antifolate chemotherapy of trypanosomiasis should be possible taking this into account.
- Published
- 2011
- Full Text
- View/download PDF
39. Preclinical evaluation of the antifolate QN254, 5-chloro- N'6'-(2,5-dimethoxy-benzyl)-quinazoline-2,4,6-triamine, as an antimalarial drug candidate.
- Author
-
Nzila A, Rottmann M, Chitnumsub P, Kiara SM, Kamchonwongpaisan S, Maneeruttanarungroj C, Taweechai S, Yeung BK, Goh A, Lakshminarayana SB, Zou B, Wong J, Ma NL, Weaver M, Keller TH, Dartois V, Wittlin S, Brun R, Yuthavong Y, and Diagana TT
- Subjects
- Administration, Oral, Animals, Antimalarials administration & dosage, Antimalarials pharmacokinetics, Antimalarials toxicity, Biological Availability, Drug Resistance, Female, Folic Acid Antagonists administration & dosage, Folic Acid Antagonists pharmacokinetics, Folic Acid Antagonists toxicity, Humans, In Vitro Techniques, Malaria drug therapy, Malaria, Falciparum drug therapy, Male, Mice, Models, Molecular, Mutation, Parasitic Sensitivity Tests, Plasmodium berghei drug effects, Plasmodium falciparum enzymology, Plasmodium falciparum genetics, Pyrimethamine pharmacology, Quinazolines administration & dosage, Quinazolines pharmacokinetics, Quinazolines toxicity, Rats, Rats, Wistar, Tetrahydrofolate Dehydrogenase chemistry, Tetrahydrofolate Dehydrogenase genetics, Antimalarials pharmacology, Folic Acid Antagonists pharmacology, Plasmodium falciparum drug effects, Quinazolines pharmacology
- Abstract
Drug resistance against dihydrofolate reductase (DHFR) inhibitors-such as pyrimethamine (PM)-has now spread to almost all regions where malaria is endemic, rendering antifolate-based malaria treatments highly ineffective. We have previously shown that the di-amino quinazoline QN254 [5-chloro-N'6'-(2,5-dimethoxy-benzyl)-quinazoline-2,4,6-triamine] is active against the highly PM-resistant Plasmodium falciparum V1S strain, suggesting that QN254 could be used to treat malaria in regions with a high prevalence of antifolate resistance. Here, we further demonstrate that QN254 is highly active against Plasmodium falciparum clinical isolates, displaying various levels of antifolate drug resistance, and we provide biochemical and structural evidence that QN254 binds and inhibits the function of both the wild-type and the quadruple-mutant (V1S) forms of the DHFR enzyme. In addition, we have assessed QN254 oral bioavailability, efficacy, and safety in vivo. The compound displays favorable pharmacokinetic properties after oral administration in rodents. The drug was remarkably efficacious against Plasmodium berghei and could fully cure infected mice with three daily oral doses of 30 mg/kg. In the course of these efficacy studies, we have uncovered some dose limiting toxicity at higher doses that was confirmed in rats. Thus, despite its relative in vitro selectivity toward the Plasmodium DHFR enzyme, QN254 does not show the adequate therapeutic index to justify its further development as a single agent.
- Published
- 2010
- Full Text
- View/download PDF
40. Crystallization and preliminary crystallographic studies of dihydrofolate reductase-thymidylate synthase from Trypanosoma cruzi, the Chagas disease pathogen.
- Author
-
Chitnumsub P, Yuvaniyama J, Chahomchuen T, Vilaivan T, and Yuthavong Y
- Subjects
- Crystallization, Crystallography, X-Ray, Molecular Structure, Protozoan Proteins chemistry, Trypanosoma cruzi pathogenicity, Chagas Disease parasitology, Multienzyme Complexes chemistry, Tetrahydrofolate Dehydrogenase chemistry, Thymidylate Synthase chemistry, Trypanosoma cruzi enzymology
- Abstract
Trypanosoma cruzi dihydrofolate reductase-thymidylate synthase (TcDHFR-TS) was crystallized in complexes with the dihydrotriazine-based or quinazoline-based antifolates C-448, cycloguanil (CYC) and Q-8 in order to gain insight into the interactions of this DHFR enzyme with classical and novel inhibitors. The TcDHFR-TS-C-448-NDP-dUMP crystal belonged to space group C222(1) with two molecules per asymmetric unit and diffracted to 2.37 angstrom resolution. The TcDHFR-TS-CYC, TcDHFR-TS-CYC-NDP and TcDHFR-TS-Q-8-NDP crystals belonged to space group P2(1) with four molecules per asymmetric unit and diffracted to 2.1, 2.6 and 2.8 angstrom resolution, respectively. Crystals belonging to the two different space groups were suitable for structure determination.
- Published
- 2009
- Full Text
- View/download PDF
41. Exploiting structural analysis, in silico screening, and serendipity to identify novel inhibitors of drug-resistant falciparum malaria.
- Author
-
Dasgupta T, Chitnumsub P, Kamchonwongpaisan S, Maneeruttanarungroj C, Nichols SE, Lyons TM, Tirado-Rives J, Jorgensen WL, Yuthavong Y, and Anderson KS
- Subjects
- Animals, Antimalarials chemistry, Antimalarials pharmacology, Antimalarials therapeutic use, Binding Sites, Cell Culture Techniques, Crystallography, X-Ray, Drug Discovery, Drug Resistance, Enzyme Inhibitors metabolism, Folic Acid Antagonists therapeutic use, Humans, Inhibitory Concentration 50, Molecular Structure, Multienzyme Complexes chemistry, Plasmodium falciparum enzymology, Plasmodium falciparum genetics, Protein Binding, Tetrahydrofolate Dehydrogenase chemistry, Thymidylate Synthase chemistry, Folic Acid Antagonists chemistry, Folic Acid Antagonists pharmacology, Malaria, Falciparum drug therapy, Multienzyme Complexes antagonists & inhibitors, Plasmodium falciparum drug effects, Thymidylate Synthase antagonists & inhibitors
- Abstract
Plasmodium falciparum thymidylate synthase-dihydrofolate reductase (TS-DHFR) is an essential enzyme in folate biosynthesis and a major malarial drug target. This bifunctional enzyme thus presents different design approaches for developing novel inhibitors against drug-resistant mutants. We performed a high-throughput in silico screen of a database of diverse, drug-like molecules against a non-active-site pocket of TS-DHFR. The top compounds from this virtual screen were evaluated by in vitro enzymatic and cellular culture studies. Three compounds active to 20 microM IC(50)'s in both wildtype and antifolate-resistant P. falciparum parasites were identified; moreover, no inhibition of human DHFR enzyme was observed, indicating that the inhibitory effects appeared to be parasite-specific. Notably, all three compounds had a biguanide scaffold. However, relative free energy of binding calculations suggested that the compounds might preferentially interact with the active site over the screened non-active-site region. To resolve the two possible modes of binding, co-crystallization studies of the compounds complexed with TS-DHFR enzyme were performed. Surprisingly, the structural analysis revealed that these novel, biguanide compounds do indeed bind at the active site of DHFR and additionally revealed the molecular basis by which they overcome drug resistance. To our knowledge, these are the first co-crystal structures of novel, biguanide, non-WR99210 compounds that are active against folate-resistant malaria parasites in cell culture.
- Published
- 2009
- Full Text
- View/download PDF
42. The role of tryptophan-48 in catalysis and binding of inhibitors of Plasmodium falciparum dihydrofolate reductase.
- Author
-
Kamchonwongpaisan S, Vanichtanankul J, Taweechai S, Chitnumsub P, and Yuthavong Y
- Subjects
- Animals, Catalysis, DNA, Protozoan chemistry, DNA, Protozoan genetics, Enzyme Stability, Escherichia coli growth & development, Folic Acid Antagonists metabolism, Folic Acid Antagonists pharmacology, Genetic Complementation Test, Kinetics, Mutagenesis, Insertional, Plasmodium falciparum genetics, Plasmodium falciparum metabolism, Proguanil pharmacology, Pyrimethamine pharmacology, Tetrahydrofolate Dehydrogenase genetics, Triazines pharmacology, Tryptophan genetics, Plasmodium falciparum enzymology, Tetrahydrofolate Dehydrogenase metabolism, Tryptophan metabolism
- Abstract
Dihydrofolate reductases (DHFRs) from Plasmodium falciparum (Pf) and various species of both prokaryotic and eukaryotic organisms have a conserved tryptophan (Trp) at position 48 in the active site. The role in catalysis and binding of inhibitors of the conserved Trp48 of PfDHFR has been analysed by site-specific mutagenesis, enzyme kinetics and use of a bacterial surrogate system. All 19 mutant enzymes showed undetectable or very low specific activities, with the highest value of k(cat)/K(m) from the Tyr48 (W48Y) mutant (0.12 versus 11.94M(-1)s(-1)), of about 1% of the wild-type enzyme. The inhibition constants for pyrimethamine, cycloguanil and WR99210 of the W48Y mutants are 2.5-5.3 times those of the wild-type enzyme. All mutants, except W48Y, failed to support the growth of Escherichia coli transformed with the parasite gene in the presence of trimethoprim, indicating the loss of functional activity of the parasite enzyme. Hence, Trp48 plays a crucial role in catalysis and inhibitor binding of PfDHFR. Interestingly, W48Y with an additional mutation at Asn188Tyr (N188Y) was found to promote bacterial growth and yielded a higher amount of purified enzyme. However, the kinetic parameters of the purified W48Y+N188Y enzyme were comparable with W48Y and the binding affinities for DHFR inhibitors were also similar to the wild-type enzyme. Due to its conserved nature, Trp48 of PfDHFR is a potential site for interaction with antimalarial inhibitors which would not be compromised by its mutations.
- Published
- 2007
- Full Text
- View/download PDF
43. Folate metabolism as a source of molecular targets for antimalarials.
- Author
-
Yuthavong Y, Kamchonwongpaisan S, Leartsakulpanich U, and Chitnumsub P
- Subjects
- Animals, Humans, Malaria drug therapy, Antimalarials pharmacology, Folic Acid biosynthesis, Plasmodium drug effects, Plasmodium metabolism, Protozoan Proteins antagonists & inhibitors
- Abstract
Folate metabolism of the malaria parasites provides two targets for current antimalarials: dihydrofolate reductase and dihydropteroate synthase. Dihydrofolate reductase inhibitors have been used as antimalarials over the past few decades, often in combination with dihydropteroate synthase inhibitors. Resistance to these antifolate drugs developed through mutations in both target enzymes. However, limited mutation possibilities gave opportunities for the development of new drugs. Furthermore, other enzymes in the folate and related pathways are potential new targets that remain to be exploited. These include thymidylate synthase, an enzyme fused with dihydrofolate reductase in the same protein chain, serine hydroxymethyltransferase, methylene tetrahydrofolate dehydrogenase, methionine synthase and enzymes in the glycine cleavage pathway.
- Published
- 2006
- Full Text
- View/download PDF
44. Crystal structure of dihydrofolate reductase from Plasmodium vivax: pyrimethamine displacement linked with mutation-induced resistance.
- Author
-
Kongsaeree P, Khongsuk P, Leartsakulpanich U, Chitnumsub P, Tarnchompoo B, Walkinshaw MD, and Yuthavong Y
- Subjects
- Amino Acid Substitution, Animals, Antimalarials chemistry, Antimalarials metabolism, Binding Sites, Crystallography, X-Ray, Folic Acid Antagonists chemistry, Folic Acid Antagonists metabolism, Molecular Structure, Protein Binding, Pyrimethamine metabolism, Tetrahydrofolate Dehydrogenase genetics, Tetrahydrofolate Dehydrogenase metabolism, Drug Resistance genetics, Plasmodium vivax enzymology, Pyrimethamine chemistry, Tetrahydrofolate Dehydrogenase chemistry
- Abstract
Pyrimethamine (Pyr) targets dihydrofolate reductase of Plasmodium vivax (PvDHFR) as well as other malarial parasites, but its use as antimalarial is hampered by the widespread high resistance. Comparison of the crystal structures of PvDHFR from wild-type and the Pyr-resistant (SP21, Ser-58 --> Arg + Ser-117 --> Asn) strain as complexes with NADPH and Pyr or its analog lacking p-Cl (Pyr20) clearly shows that the steric conflict arising from the side chain of Asn-117 in the mutant enzyme, accompanied by the loss of binding to Ser-120, is mainly responsible for the reduction in binding of Pyr. Pyr20 still effectively inhibits both the wild-type and SP21 proteins, and the x-ray structures of these complexes show how Pyr20 fits into both active sites without steric strain. These structural insights suggest a general approach for developing new generations of antimalarial DHFR inhibitors that, by only occupying substrate space of the active site, would retain binding affinity with the mutant enzymes.
- Published
- 2005
- Full Text
- View/download PDF
45. Subunit complementation of thymidylate synthase in Plasmodium falciparum bifunctional dihydrofolate reductase-thymidylate synthase.
- Author
-
Chanama M, Chitnumsub P, and Yuthavong Y
- Subjects
- Animals, Binding Sites, Deoxyuracil Nucleotides metabolism, Dimerization, Folic Acid metabolism, Genetic Complementation Test, Multienzyme Complexes metabolism, Mutation, Missense, Plasmodium falciparum genetics, Protein Structure, Tertiary genetics, Protein Subunits genetics, Substrate Specificity, Tetrahydrofolate Dehydrogenase metabolism, Thymidylate Synthase metabolism, Folic Acid analogs & derivatives, Multienzyme Complexes genetics, Plasmodium falciparum enzymology, Tetrahydrofolate Dehydrogenase genetics, Thymidylate Synthase genetics
- Abstract
Thymidylate synthase of Plasmodium falciparum dihydrofolate reductase-thymidylate synthase (PfDHFR-TS) functions as a dimeric enzyme with extensive contact between the two TS domains. Structural data of PfDHFR-TS shows that the formation of the two TS active sites involves contribution of the amino acid residues from both TS domains. Arg-470 donated from the adjoining domain is shown to hydrogen-bond to dUMP, while Cys-490 is a key nucleophile for TS catalysis by attacking C-6 of dUMP. However, mutants of the two series could complement one another, giving rise to active enzyme. By means of subunit complementation assay using Arg-470 and Cys-490 mutants, it is shown that co-transformants of both TS-inactive Arg-470 and Cys-490 mutants can complement the growth of thymidine auxotroph chi2913RecA(DE3) by formation of a functional TS heterodimer contributing from both Arg-470 and Cys-490 mutant subunits. 6-[3H]-FdUMP thymidylate synthase activity assay further elaborate the essence of restoration of TS activity. The TS k(cat) value of the R470D+C490A heterodimer is decreased by half from that of the wild-type PfDHFR-TS. However, the Km values for dUMP and CH2H4folate of the R470D+C490A heterodimer are similar to those of wild-type enzyme, indicating that the catalytic efficiency of the functional TS from the R470D+C490A heterodimer is similar to the wild-type TS enzyme in P. falciparum DHFR-TS.
- Published
- 2005
- Full Text
- View/download PDF
46. Characterization, crystallization and preliminary X-ray analysis of bifunctional dihydrofolate reductase-thymidylate synthase from Plasmodium falciparum.
- Author
-
Chitnumsub P, Yuvaniyama J, Vanichtanankul J, Kamchonwongpaisan S, Walkinshaw MD, and Yuthavong Y
- Subjects
- Animals, Cloning, Molecular, Crystallography, X-Ray, Deoxyuracil Nucleotides, Genetic Engineering, NADP, Polyethylene Glycols, Protozoan Proteins chemistry, Crystallization, Plasmodium falciparum enzymology, Tetrahydrofolate Dehydrogenase chemistry, Thymidylate Synthase chemistry
- Abstract
The full-length pfdhfr-ts genes of the wild-type TM4/8.2 and the double mutant K1CB1 (C59R+S108N) from the genomic DNA of the corresponding Plasmodium falciparum parasite have been cloned into a modified pET(17b) plasmid and expressed in Escherichia coli BL21 (DE3) pLysS. Conditions for the expression and purification of the P. falciparum dihydrofolate reductase-thymidylate synthase (PfDHFR-TS) have been established that yield approximately 1 mg of the soluble active enzyme per litre of culture. The purified enzymes have been crystallized using a modified microbatch method with PEG 4000 as the primary precipitating agent. X-ray diffraction data were collected to 2.50 and 2.64 A resolution under cryogenic conditions from single crystals of the two PfDHFR-TS proteins in complex with NADPH, dUMP and either Pyr30 or Pyr39. Preliminary X-ray analysis indicated that the crystals belong to the orthorhombic space group P2(1)2(1)2(1), with two molecules per asymmetric unit and approximately 52% solvent content (VM approximately 2.6 A3 Da-1). The use of a particular type of baby oil in the microbatch setup appeared to be beneficial to PfDHFR-TS crystallization and a preliminary comparison with another commonly used oil is described.
- Published
- 2004
- Full Text
- View/download PDF
47. Insights into antifolate resistance from malarial DHFR-TS structures.
- Author
-
Yuvaniyama J, Chitnumsub P, Kamchonwongpaisan S, Vanichtanankul J, Sirawaraporn W, Taylor P, Walkinshaw MD, and Yuthavong Y
- Subjects
- Amino Acid Sequence, Animals, Conserved Sequence, Drug Resistance, Models, Molecular, Molecular Sequence Data, Plasmodium enzymology, Plasmodium falciparum enzymology, Protein Conformation, Sequence Alignment, Sequence Homology, Amino Acid, Folic Acid Antagonists pharmacology, Multienzyme Complexes antagonists & inhibitors, Multienzyme Complexes chemistry, Tetrahydrofolate Dehydrogenase chemistry, Thymidylate Synthase antagonists & inhibitors, Thymidylate Synthase chemistry
- Abstract
Plasmodium falciparum dihydrofolate reductase-thymidylate synthase (PfDHFR-TS) is an important target of antimalarial drugs. The efficacy of this class of DHFR-inhibitor drugs is now compromised because of mutations that prevent drug binding yet retain enzyme activity. The crystal structures of PfDHFR-TS from the wild type (TM4/8.2) and the quadruple drug-resistant mutant (V1/S) strains, in complex with a potent inhibitor WR99210, as well as the resistant double mutant (K1 CB1) with the antimalarial pyrimethamine, reveal features for overcoming resistance. In contrast to pyrimethamine, the flexible side chain of WR99210 can adopt a conformation that fits well in the active site, thereby contributing to binding. The single-chain bifunctional PfDHFR-TS has a helical insert between the DHFR and TS domains that is involved in dimerization and domain organization. Moreover, positively charged grooves on the surface of the dimer suggest a function in channeling of substrate from TS to DHFR active sites. These features provide possible approaches for the design of new drugs to overcome antifolate resistance.
- Published
- 2003
- Full Text
- View/download PDF
48. Pyrimethamine analogs as strong inhibitors of double and quadruple mutants of dihydrofolate reductase in human malaria parasites.
- Author
-
Sardarian A, Douglas KT, Read M, Sims PF, Hyde JE, Chitnumsub P, Sirawaraporn R, and Sirawaraporn W
- Subjects
- Animals, Drug Resistance, Humans, Models, Molecular, Molecular Structure, Plasmodium falciparum drug effects, Plasmodium falciparum genetics, Point Mutation, Tetrahydrofolate Dehydrogenase genetics, Antimalarials pharmacology, Folic Acid Antagonists pharmacology, Plasmodium falciparum enzymology, Pyrimethamine analogs & derivatives, Pyrimethamine pharmacology, Tetrahydrofolate Dehydrogenase drug effects
- Abstract
Pyrimethamine acts against malarial parasites by selectively inhibiting their dihydrofolate reductase-thymidylate synthase. Resistance to pyrimethamine in Plasmodium falciparum is due to point mutations in the DHFR domain, initially at residue 108 (S108N), with additional mutations imparting much greater resistance. Our previous work, the development of a simple rational drug design strategy to overcome such resistance, used suitable meta-substituents in the pyrimethamine framework to avoid the unfavorable steric clash with mutant side chains at position 108. Interestingly, the meta-chloro analog of pyrimethamine not only overcame the resistance due to S108N, but also that contributed by the more remote mutation, C59R. The present work improves on this by means of other meta-substituents. Against wild type DHFR, double mutant types A16V + S108T and C59R + S108T, and the highly pyrimethamine/cycloguanil-resistant quadruple-mutant form N51I + C59R + S108N + I164L, pyrimethamine itself gave Ki values of 1.5, 2.4, 72.3 and 859 nM, respectively. The meta-substituted analogs, especially the meta-bromo analog, were much more powerful inhibitors of these DHFRs, including the quadruple-mutant form (meta-bromo analog, Ki 5.1 nM). For comparison, the dihydropyrazine antifolate, WR99210, gave Ki values of 0.9, 3.2, 0.8 and 0.9 nM, respectively. Ki values were also measured against recombinant human DHFR, as were their activities against the growth of Plasmodium falciparum cultures bearing the double mutations (FCB and K1 strains) and quadruple mutation (V1/S) and the wild type (3D7). The meta-analogs were highly active against all of these, with the meta-bromo again being the strongest, having an IC50 of 37 nM against V1/S, compared to > 5000 nM for pyrimethamine itself and 1.1 nM for WR99210.
- Published
- 2003
- Full Text
- View/download PDF
49. Three-dimensional structure of M. tuberculosis dihydrofolate reductase reveals opportunities for the design of novel tuberculosis drugs.
- Author
-
Li R, Sirawaraporn R, Chitnumsub P, Sirawaraporn W, Wooden J, Athappilly F, Turley S, and Hol WG
- Subjects
- Amino Acid Sequence, Antitubercular Agents pharmacology, Drug Design, Enzyme Inhibitors pharmacology, Humans, Molecular Sequence Data, NADP metabolism, Protein Binding, Protein Conformation, Sequence Homology, Amino Acid, Tetrahydrofolate Dehydrogenase metabolism, Antitubercular Agents chemical synthesis, Enzyme Inhibitors chemical synthesis, Mycobacterium tuberculosis enzymology, Tetrahydrofolate Dehydrogenase chemistry
- Abstract
Dihydrofolate reductase (DHFR) catalyzes the NADPH-dependent reduction of dihydrofolate to tetrahydrofolate and is essential for the synthesis of thymidylate, purines and several amino acids. Inhibition of the enzyme's activity leads to arrest of DNA synthesis and cell death. The enzyme has been studied extensively as a drug target for bacterial, protozoal and fungal infections, and also for neoplastic and autoimmune diseases. Here, we report the crystal structure of dihydrofolate reductase from Mycobacterium tuberculosis, a human pathogen responsible for the death of millions of human beings per year. Three crystal structures of ternary complexes of M. tuberculosis DHFR with NADP and different inhibitors have been determined, as well as the binary complex with NADP, with resolutions ranging from 1.7 to 2.0 A. The three DHFR inhibitors are the anticancer drug methotrexate, the antimicrobial trimethoprim and Br-WR99210, an analogue of the antimalarial agent WR99210. Structural comparison of these complexes with human dihydrofolate reductase indicates that the overall protein folds are similar, despite only 26 % sequence identity, but that the environments of both NADP and of the inhibitors contain interesting differences between the enzymes from host and pathogen. Specifically, residues Ala101 and Leu102 near the N6 of NADP are distinctly more hydrophobic in the M. tuberculosis than in the human enzyme. Another striking difference occurs in a region near atoms N1 and N8 of methotrexate, which is also near atom N1 of trimethoprim, and near the N1 and two methyl groups of Br-WR99210. A glycerol molecule binds here in a pocket of the M. tuberculosis DHFR:MTX complex, while this pocket is essentially filled with hydrophobic side-chains in the human enzyme. These differences between the enzymes from pathogen and host provide opportunities for designing new selective inhibitors of M. tuberculosis DHFR., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
50. The nucleation of monomeric parallel beta-sheet-like structures and their self-assembly in aqueous solution.
- Author
-
Chitnumsub P, Fiori WR, Lashuel HA, Diaz H, and Kelly JW
- Subjects
- Circular Dichroism, Hydrogen Bonding, Magnetic Resonance Spectroscopy, Models, Molecular, Molecular Conformation, Molecular Structure, Peptides chemistry, Spectroscopy, Fourier Transform Infrared, Ultracentrifugation, Benzofurans chemistry, Furans chemistry, Propionates chemistry, Protein Structure, Secondary
- Abstract
The aromatic diacid residue 4,6-dibenzofuranbispropionic acid (1) was designed to nucleate a parallel beta-sheet-like structure in small peptides in aqueous solution via a hydrogen-bonded hydrophobic cluster. Even though a 14-membered ring hydrogen bond necessary for parallel beta-sheet formation is favored in simple amides composed of 1, this hydrogen bonding interaction does not appear to be sufficient to nucleate parallel beta-sheet formation in the absence of hydrophobic clustering between the dibenzofuran portion of 1 and the hydrophobic side chains of the flanking alpha-amino acids. The subsequence --hydrophobic residue-1-hydrophobic residue-- is required for folding in the context of a nucleated two-stranded parallel beta-sheet structure. In all cases where the peptidomimetics can fold into two diastereomeric parallel beta-sheet structures having different hydrogen bonding networks, these conformations appear to exchange rapidly. The majority of the parallel beta-sheet structures evaluated herein undergo linked intramolecular folding and self-assembly, affording a fibrillar beta-sheet quaternary structure. To unlink folding and assembly, asymmetric parallel beta-sheet structures incorporating N-methylated alpha-amino acid residues have been synthesized using a new solid phase approach. Residue 1 facilitates the folding of several peptides described within affording a monomeric parallel beta-sheet-like structure in aqueous solution, as ascertained by a variety of spectroscopic and biophysical methods, increasing our understanding of parallel beta-sheet structure.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.