Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jarunee Vanichtanankul is active.

Publication


Featured researches published by Jarunee Vanichtanankul.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Malarial dihydrofolate reductase as a paradigm for drug development against a resistance-compromised target

Yongyuth Yuthavong; Bongkoch Tarnchompoo; Tirayut Vilaivan; Penchit Chitnumsub; Sumalee Kamchonwongpaisan; Susan A. Charman; Danielle N McLennan; Karen L. White; Livia Vivas; Emily Bongard; Chawanee Thongphanchang; Jarunee Vanichtanankul; Roonglawan Rattanajak; Uthai Arwon; Pascal Fantauzzi; Jirundon Yuvaniyama; William N. Charman; David Matthews

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.


Parasitology | 2005

Malarial (Plasmodium falciparum) dihydrofolate reductase-thymidylate synthase: structural basis for antifolate resistance and development of effective inhibitors.

Yongyuth Yuthavong; Jirundon Yuvaniyama; Penchit Chitnumsub; Jarunee Vanichtanankul; Sudsanguan Chusacultanachai; Bongkoch Tarnchompoo; Tirayut Vilaivan; Sumalee Kamchonwongpaisan

Dihydrofolate reductase-thymidylate synthase (DHFR-TS) from Plasmodium falciparum, a validated target for antifolate antimalarials, is a dimeric enzyme with interdomain interactions significantly mediated by the junction region as well as the Plasmodium-specific additional sequences (inserts) in the DHFR domain. The X-ray structures of both the wild-type and mutant enzymes associated with drug resistance, in complex with either a drug which lost, or which still retains, effectiveness for the mutants, reveal features which explain the basis of drug resistance resulting from mutations around the active site. Binding of rigid inhibitors like pyrimethamine and cycloguanil to the enzyme active site is affected by steric conflict with the side-chains of mutated residues 108 and 16, as well as by changes in the main chain configuration. The role of important residues on binding of inhibitors and substrates was further elucidated by site-directed and random mutagenesis studies. Guided by the active site structure and modes of inhibitor binding, new inhibitors with high affinity against both wild-type and mutant enzymes have been designed and synthesized, some of which have very potent anti-malarial activities against drug-resistant P. falciparum bearing the mutant enzymes.


Acta Crystallographica Section D-biological Crystallography | 2004

Characterization, crystallization and preliminary X-­ray analysis of bifunctional dihydrofolate reductase–thymidylate synthase from Plasmodium falciparum

Penchit Chitnumsub; Jirundon Yuvaniyama; Jarunee Vanichtanankul; Sumalee Kamchonwongpaisan; Walkinshaw; Yongyuth Yuthavong

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.


Antimicrobial Agents and Chemotherapy | 2012

Combined Spatial Limitation around Residues 16 and 108 of Plasmodium falciparum Dihydrofolate Reductase Explains Resistance to Cycloguanil.

Jarunee Vanichtanankul; Chayasith Uttamapinant; Penchit Chitnumsub; Tirayut Vilaivan; Yongyuth Yuthavong; Sumalee Kamchonwongpaisan

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.


ACS Chemical Biology | 2011

Trypanosomal dihydrofolate reductase reveals natural antifolate resistance

Jarunee Vanichtanankul; Jirundon Yuvaniyama; Tirayut Vilaivan; Penchit Chitnumsub; Sumalee Kamchonwongpaisan; Yongyuth Yuthavong

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.


Biotechnology and Applied Biochemistry | 2017

Characterization of the binding of a glycosylated serine protease from Euphorbia cf. lactea latex to human fibrinogen

Jaruwan Siritapetawee; Chutima Talabnin; Jarunee Vanichtanankul; Chomphunuch Songsiriritthigul; Kanjana Thumanu; Chun-Jung Chen; Nantarat Komanasin

In this study, the binding of a glycosylated serine protease (EuP‐82) with human fibrinogen was investigated by isothermal titration calorimetry (ITC). ITC analysis indicated that the binding of EuP‐82 to fibrinogen in the conditions with or without the activator (Ca2+) was an exothermic reaction (dominant negative enthalpy), which tended to be driven by hydrogen bonding and van der Waals interactions. In contrast, the binding of fibrinogen−EuP‐82 in the condition with the inhibitor (Zn2+) was an unfavorable endothermic reaction. EuP‐82 could not inhibit the platelet activity in citrated whole blood via the ADP–receptor pathways (mainly, P2Y1 and P2Y12), but it could enhance the platelet aggregation. The ITC together with whole blood platelet aggregation suggested that EuP‐82 provided multiple fibrinogen‐binding sites that were not related to the arginine‐glycine‐aspartate (RGD) and the dodecapeptide sequences of fibrinogen. In addition, EuP‐82 had neither thrombin‐like activity nor anticoagulant activity. The SR‐FTIR spectra revealed that EuP‐82 was a glycoprotein. Deglycosylation of EuP‐82 did not affect its proteolytic activity. Moreover, EuP‐82 did not exhibit any toxicity to the living cells (NIH‐3T3). This study supports that EuP‐82 may be useful for wound‐healing material through stabilizing the clot via the platelet induction for the first process.


Journal of Medicinal Chemistry | 2004

Inhibitors of Multiple Mutants of Plasmodium falciparum Dihydrofolate Reductase and Their Antimalarial Activities

Sumalee Kamchonwongpaisan; Rachel Quarrell; Netnapa Charoensetakul; Rachel Ponsinet; Tirayut Vilaivan; Jarunee Vanichtanankul; Bongkoch Tarnchompoo; Worachart Sirawaraporn; Gordon Lowe; Yongyuth Yuthavong


Journal of Medicinal Chemistry | 2002

Development of 2,4-Diaminopyrimidines as Antimalarials Based on Inhibition of the S108N and C59R+S108N Mutants of Dihydrofolate Reductase from Pyrimethamine-Resistant Plasmodium falciparum

Bongkoch Tarnchompoo; Chawanee Sirichaiwat; Worrapong Phupong; Chakapong Intaraudom; Worachart Sirawaraporn; Sumalee Kamchonwongpaisan; Jarunee Vanichtanankul; Yodhathai Thebtaranonth; Yongyuth Yuthavong


Journal of Medicinal Chemistry | 2004

Target Guided Synthesis of 5-Benzyl-2,4-diamonopyrimidines: Their Antimalarial Activities and Binding Affinities to Wild Type and Mutant Dihydrofolate Reductases from Plasmodium falciparum

Chawanee Sirichaiwat; Chakapong Intaraudom; Sumalee Kamchonwongpaisan; Jarunee Vanichtanankul; Yodhathai Thebtaranonth; Yongyuth Yuthavong


Analytical Chemistry | 2005

Stoichiometric selection of tight-binding inhibitors by wild-type and mutant forms of malarial (Plasmodium falciparum) dihydrofolate reductase.

Sumalee Kamchonwongpaisan; Jarunee Vanichtanankul; Bongkoch Tarnchompoo; Jirundon Yuvaniyama; Yongyuth Yuthavong

Collaboration


Dive into the Jarunee Vanichtanankul's collaboration.

Top Co-Authors

Avatar

Yongyuth Yuthavong

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar

Sumalee Kamchonwongpaisan

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar

Penchit Chitnumsub

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar

Tirayut Vilaivan

Medicines for Malaria Venture

View shared research outputs
Top Co-Authors

Avatar

Bongkoch Tarnchompoo

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roonglawan Rattanajak

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chawanee Thongphanchang

Thailand National Science and Technology Development Agency

View shared research outputs
Researchain Logo
Decentralizing Knowledge