Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Nature ; 576(7786): 315-320, 2019 12.
Article in English | MEDLINE | ID: mdl-31776516

ABSTRACT

The emergence and spread of drug-resistant Plasmodium falciparum impedes global efforts to control and eliminate malaria. For decades, treatment of malaria has relied on chloroquine (CQ), a safe and affordable 4-aminoquinoline that was highly effective against intra-erythrocytic asexual blood-stage parasites, until resistance arose in Southeast Asia and South America and spread worldwide1. Clinical resistance to the chemically related current first-line combination drug piperaquine (PPQ) has now emerged regionally, reducing its efficacy2. Resistance to CQ and PPQ has been associated with distinct sets of point mutations in the P. falciparum CQ-resistance transporter PfCRT, a 49-kDa member of the drug/metabolite transporter superfamily that traverses the membrane of the acidic digestive vacuole of the parasite3-9. Here we present the structure, at 3.2 Å resolution, of the PfCRT isoform of CQ-resistant, PPQ-sensitive South American 7G8 parasites, using single-particle cryo-electron microscopy and antigen-binding fragment technology. Mutations that contribute to CQ and PPQ resistance localize primarily to moderately conserved sites on distinct helices that line a central negatively charged cavity, indicating that this cavity is the principal site of interaction with the positively charged CQ and PPQ. Binding and transport studies reveal that the 7G8 isoform binds both drugs with comparable affinities, and that these drugs are mutually competitive. The 7G8 isoform transports CQ in a membrane potential- and pH-dependent manner, consistent with an active efflux mechanism that drives CQ resistance5, but does not transport PPQ. Functional studies on the newly emerging PfCRT F145I and C350R mutations, associated with decreased PPQ susceptibility in Asia and South America, respectively6,9, reveal their ability to mediate PPQ transport in 7G8 variant proteins and to confer resistance in gene-edited parasites. Structural, functional and in silico analyses suggest that distinct mechanistic features mediate the resistance to CQ and PPQ in PfCRT variants. These data provide atomic-level insights into the molecular mechanism of this key mediator of antimalarial treatment failures.


Subject(s)
Cryoelectron Microscopy , Drug Resistance/drug effects , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/ultrastructure , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Protozoan Proteins/ultrastructure , Chloroquine/metabolism , Chloroquine/pharmacology , Drug Resistance/genetics , Hydrogen-Ion Concentration , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Molecular , Mutation , Plasmodium falciparum/genetics , Plasmodium falciparum/ultrastructure , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Quinolines/metabolism , Quinolines/pharmacology
2.
PLoS Pathog ; 18(2): e1010278, 2022 02.
Article in English | MEDLINE | ID: mdl-35130315

ABSTRACT

Multidrug-resistant Plasmodium falciparum parasites have emerged in Cambodia and neighboring countries in Southeast Asia, compromising the efficacy of first-line antimalarial combinations. Dihydroartemisinin + piperaquine (PPQ) treatment failure rates have risen to as high as 50% in some areas in this region. For PPQ, resistance is driven primarily by a series of mutant alleles of the P. falciparum chloroquine resistance transporter (PfCRT). PPQ resistance was reported in China three decades earlier, but the molecular driver remained unknown. Herein, we identify a PPQ-resistant pfcrt allele (China C) from Yunnan Province, China, whose genotypic lineage is distinct from the PPQ-resistant pfcrt alleles currently observed in Cambodia. Combining gene editing and competitive growth assays, we report that PfCRT China C confers moderate PPQ resistance while re-sensitizing parasites to chloroquine (CQ) and incurring a fitness cost that manifests as a reduced rate of parasite growth. PPQ transport assays using purified PfCRT isoforms, combined with molecular dynamics simulations, highlight differences in drug transport kinetics and in this transporter's central cavity conformation between China C and the current Southeast Asian PPQ-resistant isoforms. We also report a novel computational model that incorporates empirically determined fitness landscapes at varying drug concentrations, combined with antimalarial susceptibility profiles, mutation rates, and drug pharmacokinetics. Our simulations with PPQ-resistant or -sensitive parasite lines predict that a three-day regimen of PPQ combined with CQ can effectively clear infections and prevent the evolution of PfCRT variants. This work suggests that including CQ in combination therapies could be effective in suppressing the evolution of PfCRT-mediated multidrug resistance in regions where PPQ has lost efficacy.


Subject(s)
Artemisinins/therapeutic use , Chloroquine/therapeutic use , Drug Resistance, Multiple , Membrane Transport Proteins/genetics , Piperazines/therapeutic use , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Quinolines/therapeutic use , Alleles , Animals , Antimalarials/therapeutic use , Computer Simulation , Humans , Malaria, Falciparum/parasitology
3.
J Chem Inf Model ; 63(10): 3171-3185, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37167486

ABSTRACT

In the hit identification stage of drug discovery, a diverse chemical space needs to be explored to identify initial hits. Contrary to empirical scoring functions, absolute protein-ligand binding free-energy perturbation (ABFEP) provides a theoretically more rigorous and accurate description of protein-ligand binding thermodynamics and could, in principle, greatly improve the hit rates in virtual screening. In this work, we describe an implementation of an accurate and reliable ABFEP method in FEP+. We validated the ABFEP method on eight congeneric compound series binding to eight protein receptors including both neutral and charged ligands. For ligands with net charges, the alchemical ion approach is adopted to avoid artifacts in electrostatic potential energy calculations. The calculated binding free energies correlate with experimental results with a weighted average of R2 = 0.55 for the entire dataset. We also observe an overall root-mean-square error (RMSE) of 1.1 kcal/mol after shifting the zero-point of the simulation data to match the average experimental values. Through ABFEP calculations using apo versus holo protein structures, we demonstrated that the protein conformational and protonation state changes between the apo and holo proteins are the main physical factors contributing to the protein reorganization free energy manifested by the overestimation of raw ABFEP calculated binding free energies using the holo structures of the proteins. Furthermore, we performed ABFEP calculations in three virtual screening applications for hit enrichment. ABFEP greatly improves the hit rates as compared to docking scores or other methods like metadynamics. The good performance of ABFEP in rank ordering compounds demonstrated in this work confirms it as a useful tool to improve the hit rates in virtual screening, thus facilitating hit discovery.


Subject(s)
Proteins , Ligands , Protein Binding , Entropy , Proteins/chemistry , Thermodynamics
4.
Proc Natl Acad Sci U S A ; 115(29): E6863-E6870, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29967165

ABSTRACT

We describe noncovalent, reversible asparagine ethylenediamine (AsnEDA) inhibitors of the Plasmodium falciparum proteasome (Pf20S) ß5 subunit that spare all active subunits of human constitutive and immuno-proteasomes. The compounds are active against erythrocytic, sexual, and liver-stage parasites, against parasites resistant to current antimalarials, and against P. falciparum strains from patients in Africa. The ß5 inhibitors synergize with a ß2 inhibitor in vitro and in mice and with artemisinin. P. falciparum selected for resistance to an AsnEDA ß5 inhibitor surprisingly harbored a point mutation in the noncatalytic ß6 subunit. The ß6 mutant was resistant to the species-selective Pf20S ß5 inhibitor but remained sensitive to the species-nonselective ß5 inhibitors bortezomib and carfilzomib. Moreover, resistance to the Pf20S ß5 inhibitor was accompanied by increased sensitivity to a Pf20S ß2 inhibitor. Finally, the ß5 inhibitor-resistant mutant had a fitness cost that was exacerbated by irradiation. Thus, used in combination, multistage-active inhibitors of the Pf20S ß5 and ß2 subunits afford synergistic antimalarial activity with a potential to delay the emergence of resistance to artemisinins and each other.


Subject(s)
Antimalarials/chemistry , Plasmodium falciparum/enzymology , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors/chemistry , Protozoan Proteins/antagonists & inhibitors , Artemisinins/chemistry , Bortezomib/chemistry , Drug Resistance, Microbial , Humans , Lactones/chemistry , Oligopeptides/chemistry , Protozoan Proteins/chemistry
5.
Angew Chem Int Ed Engl ; 60(17): 9279-9283, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33433953

ABSTRACT

Plasmodium falciparum proteasome (Pf20S) inhibitors are active against Plasmodium at multiple stages-erythrocytic, gametocyte, liver, and gamete activation stages-indicating that selective Pf20S inhibitors possess the potential to be therapeutic, prophylactic, and transmission-blocking antimalarials. Starting from a reported compound, we developed a noncovalent, macrocyclic peptide inhibitor of the malarial proteasome with high species selectivity and improved pharmacokinetic properties. The compound demonstrates specific, time-dependent inhibition of the ß5 subunit of the Pf20S, kills artemisinin-sensitive and artemisinin-resistant P. falciparum isolates in vitro and reduces parasitemia in humanized, P. falciparum-infected mice.


Subject(s)
Antimalarials/pharmacology , Drug Development , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Malaria, Falciparum/metabolism , Mice , Models, Molecular , Molecular Conformation , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/chemistry
6.
Blood ; 129(1): 88-99, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27784673

ABSTRACT

Phosphoinositide 3-kinase (PI3K) and the proteasome pathway are both involved in activating the mechanistic target of rapamycin (mTOR). Because mTOR signaling is required for initiation of messenger RNA translation, we hypothesized that cotargeting the PI3K and proteasome pathways might synergistically inhibit translation of c-Myc. We found that a novel PI3K δ isoform inhibitor TGR-1202, but not the approved PI3Kδ inhibitor idelalisib, was highly synergistic with the proteasome inhibitor carfilzomib in lymphoma, leukemia, and myeloma cell lines and primary lymphoma and leukemia cells. TGR-1202 and carfilzomib (TC) synergistically inhibited phosphorylation of the eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1), leading to suppression of c-Myc translation and silencing of c-Myc-dependent transcription. The synergistic cytotoxicity of TC was rescued by overexpression of eIF4E or c-Myc. TGR-1202, but not other PI3Kδ inhibitors, inhibited casein kinase-1 ε (CK1ε). Targeting CK1ε using a selective chemical inhibitor or short hairpin RNA complements the effects of idelalisib, as a single agent or in combination with carfilzomib, in repressing phosphorylation of 4E-BP1 and the protein level of c-Myc. These results suggest that TGR-1202 is a dual PI3Kδ/CK1ε inhibitor, which may in part explain the clinical activity of TGR-1202 in aggressive lymphoma not found with idelalisib. Targeting CK1ε should become an integral part of therapeutic strategies targeting translation of oncogenes such as c-Myc.


Subject(s)
Casein Kinase 1 epsilon/antagonists & inhibitors , Hematologic Neoplasms , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Synergism , Humans , Mice , Oligopeptides/pharmacology , Protein Biosynthesis , Random Allocation , Xenograft Model Antitumor Assays
7.
Nature ; 475(7357): 510-3, 2011 Jul 27.
Article in English | MEDLINE | ID: mdl-21796210

ABSTRACT

Membrane-bound receptors often form large assemblies resulting from binding to soluble ligands, cell-surface molecules on other cells and extracellular matrix proteins. For example, the association of membrane proteins with proteins on different cells (trans-interactions) can drive the oligomerization of proteins on the same cell (cis-interactions). A central problem in understanding the molecular basis of such phenomena is that equilibrium constants are generally measured in three-dimensional solution and are thus difficult to relate to the two-dimensional environment of a membrane surface. Here we present a theoretical treatment that converts three-dimensional affinities to two dimensions, accounting directly for the structure and dynamics of the membrane-bound molecules. Using a multiscale simulation approach, we apply the theory to explain the formation of ordered, junction-like clusters by classical cadherin adhesion proteins. The approach features atomic-scale molecular dynamics simulations to determine interdomain flexibility, Monte Carlo simulations of multidomain motion and lattice simulations of junction formation. A finding of general relevance is that changes in interdomain motion on trans-binding have a crucial role in driving the lateral, cis-, clustering of adhesion receptors.


Subject(s)
Cadherins/chemistry , Cadherins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Molecular Dynamics Simulation , Computer Simulation , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Monte Carlo Method , Platelet Glycoprotein GPIb-IX Complex , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary
8.
Proc Natl Acad Sci U S A ; 111(40): E4175-84, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25253890

ABSTRACT

Type I cadherin cell-adhesion proteins are similar in sequence and structure and yet are different enough to mediate highly specific cell-cell recognition phenomena. It has previously been shown that small differences in the homophilic and heterophilic binding affinities of different type I family members can account for the differential cell-sorting behavior. Here we use a combination of X-ray crystallography, analytical ultracentrifugation, surface plasmon resonance and double electron-electron resonance (DEER) electron paramagnetic resonance spectroscopy to identify the molecular determinants of type I cadherin dimerization affinities. Small changes in sequence are found to produce subtle structural and dynamical changes that impact relative affinities, in part via electrostatic and hydrophobic interactions, and in part through entropic effects because of increased conformational heterogeneity in the bound states as revealed by DEER distance mapping in the dimers. These findings highlight the remarkable ability of evolution to exploit a wide range of molecular properties to produce closely related members of the same protein family that have affinity differences finely tuned to mediate their biological roles.


Subject(s)
Cadherins/chemistry , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Binding, Competitive , Cadherins/genetics , Cadherins/metabolism , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Sequence Homology, Amino Acid , Static Electricity , Xenopus , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
9.
Chembiochem ; 17(23): 2286-2292, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27653442

ABSTRACT

The bottromycins are a family of highly modified peptide natural products, which display potent antimicrobial activity against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus. Bottromycins have recently been shown to be ribosomally synthesized and post-translationally modified peptides (RiPPs). Unique amongst RiPPs, the precursor peptide BotA contains a C-terminal "follower" sequence, rather than the canonical N-terminal "leader" sequence. We report herein the structural and biochemical characterization of BotP, a leucyl-aminopeptidase-like enzyme from the bottromycin pathway. We demonstrate that BotP is responsible for the removal of the N-terminal methionine from the precursor peptide. Determining the crystal structures of both apo BotP and BotP in complex with Mn2+ allowed us to model a BotP/substrate complex and to rationalize substrate recognition. Our data represent the first step towards targeted compound modification to unlock the full antibiotic potential of bottro- mycin.


Subject(s)
Leucyl Aminopeptidase/metabolism , Crystallography, X-Ray , Models, Molecular , Molecular Conformation , Peptides, Cyclic/biosynthesis , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Substrate Specificity
10.
Proc Natl Acad Sci U S A ; 109(3): E127-34, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22171007

ABSTRACT

Vertebrate classical cadherins mediate selective calcium-dependent cell adhesion by mechanisms now understood at the atomic level. However, structures and adhesion mechanisms of cadherins from invertebrates, which are highly divergent yet function in similar roles, remain unknown. Here we present crystal structures of three- and four-tandem extracellular cadherin (EC) domain segments from Drosophila N-cadherin (DN-cadherin), each including the predicted N-terminal EC1 domain (denoted EC1') of the mature protein. While the linker regions for the EC1'-EC2' and EC3'-EC4' pairs display binding of three Ca(2+) ions similar to that of vertebrate cadherins, domains EC2' and EC3' are joined in a "kinked" orientation by a previously uncharacterized Ca(2+)-free linker. Biophysical analysis demonstrates that a construct containing the predicted N-terminal nine EC domains of DN-cadherin forms homodimers with affinity similar to vertebrate classical cadherins, whereas deleting the ninth EC domain ablates dimerization. These results suggest that, unlike their vertebrate counterparts, invertebrate cadherins may utilize multiple EC domains to form intercellular adhesive bonds. Sequence analysis reveals that similar Ca(2+)-free linkers are widely distributed in the ectodomains of both vertebrate and invertebrate cadherins.


Subject(s)
Cadherins/chemistry , Calcium/metabolism , Drosophila Proteins/chemistry , Drosophila melanogaster/metabolism , Adhesiveness , Amino Acid Sequence , Animals , Cadherins/metabolism , Crystallography, X-Ray , Drosophila Proteins/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Sequence Alignment , Sequence Analysis, Protein
11.
J Bacteriol ; 196(24): 4216-28, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25246477

ABSTRACT

RecQ family helicases function as safeguards of the genome. Unlike Escherichia coli, the Gram-positive Bacillus subtilis bacterium possesses two RecQ-like homologues, RecQ[Bs] and RecS, which are required for the repair of DNA double-strand breaks. RecQ[Bs] also binds to the forked DNA to ensure a smooth progression of the cell cycle. Here we present the first biochemical analysis of recombinant RecQ[Bs]. RecQ[Bs] binds weakly to single-stranded DNA (ssDNA) and blunt-ended double-stranded DNA (dsDNA) but strongly to forked dsDNA. The protein exhibits a DNA-stimulated ATPase activity and ATP- and Mg(2+)-dependent DNA helicase activity with a 3' → 5' polarity. Molecular modeling shows that RecQ[Bs] shares high sequence and structure similarity with E. coli RecQ. Surprisingly, RecQ[Bs] resembles the truncated Saccharomyces cerevisiae Sgs1 and human RecQ helicases more than RecQ[Ec] with regard to its enzymatic activities. Specifically, RecQ[Bs] unwinds forked dsDNA and DNA duplexes with a 3'-overhang but is inactive on blunt-ended dsDNA and 5'-overhung duplexes. Interestingly, RecQ[Bs] unwinds blunt-ended DNA with structural features, including nicks, gaps, 5'-flaps, Kappa joints, synthetic replication forks, and Holliday junctions. We discuss these findings in the context of RecQ[Bs]'s possible functions in preserving genomic stability.


Subject(s)
Bacillus subtilis/enzymology , RecQ Helicases/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , DNA/metabolism , DNA, Single-Stranded/metabolism , Magnesium/metabolism , Models, Molecular , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
12.
ACS Med Chem Lett ; 15(4): 524-532, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38628784

ABSTRACT

Eleven-nineteen leukemia (ENL) is an epigenetic reader protein that drives oncogenic transcriptional programs in acute myeloid leukemia (AML). AML is one of the deadliest hematopoietic malignancies, with an overall 5-year survival rate of 27%. The epigenetic reader activity of ENL is mediated by its YEATS domain that binds to acetyl and crotonyl marks on histone tails and colocalizes with promoters of actively transcribed genes that are essential for leukemia. Prior to the discovery of TDI-11055, existing inhibitors of ENL YEATS showed in vitro potency, but had not shown efficacy in in vivo animal models. During the course of the medicinal chemistry campaign described here, we identified ENL YEATS inhibitor TDI-11055 that has an improved pharmacokinetic profile and is appropriate for in vivo evaluation of the ENL YEATS inhibition mechanism in AML.

13.
ACS Bio Med Chem Au ; 3(6): 507-515, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38144256

ABSTRACT

Lpd (lipoamide dehydrogenase) in Mycobacterium tuberculosis (Mtb) is required for virulence and is a genetically validated tuberculosis (TB) target. Numerous screens have been performed over the last decade, yet only two inhibitor series have been identified. Recent advances in large-scale virtual screening methods combined with make-on-demand compound libraries have shown the potential for finding novel hits. In this study, the Enamine REAL library consisting of ∼1.12 billion compounds was efficiently screened using the GPU Shape screen method against Mtb Lpd to find additional chemical matter that would expand on the known sulfonamide inhibitor series. We identified six new inhibitors with IC50 in the range of 5-100 µM. While these compounds remained chemically close to the already known sulfonamide series inhibitors, some diversity was found in the cores of the hits. The two most potent hits were further validated by one-step potency optimization to submicromolar levels. The co-crystal structure of optimized analogue TDI-13537 provided new insights into the potency determinants of the series.

14.
J Med Chem ; 66(2): 1484-1508, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36630286

ABSTRACT

With increasing reports of resistance to artemisinins and artemisinin-combination therapies, targeting the Plasmodium proteasome is a promising strategy for antimalarial development. We recently reported a highly selective Plasmodium falciparum proteasome inhibitor with anti-malarial activity in the humanized mouse model. To balance the permeability of the series of macrocycles with other drug-like properties, we conducted further structure-activity relationship studies on a biphenyl ether-tethered macrocyclic scaffold. Extensive SAR studies around the P1, P3, and P5 groups and peptide backbone identified compound TDI-8414. TDI-8414 showed nanomolar antiparasitic activity, no toxicity to HepG2 cells, high selectivity against the Plasmodium proteasome over the human constitutive proteasome and immunoproteasome, improved solubility and PAMPA permeability, and enhanced metabolic stability in microsomes and plasma of both humans and mice.


Subject(s)
Antimalarials , Plasmodium , Humans , Animals , Mice , Antimalarials/pharmacology , Antimalarials/chemistry , Proteasome Endopeptidase Complex/metabolism , Structure-Activity Relationship , Plasmodium falciparum/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/chemistry
15.
J Biol Chem ; 286(7): 5242-53, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21148480

ABSTRACT

All members of the kinesin superfamily of molecular motors contain an unusual structural motif consisting of an α-helix that is interrupted by a flexible loop, referred to as L5. We have examined the function of L5 in the mitotic kinesin Eg5 by combining site-directed mutagenesis of L5 with transient state kinetics, molecular dynamics simulations, and docking using cryo electron microscopy density. We find that mutation of a proline residue located at a turn within this loop profoundly slows nucleotide-induced structural changes both at the catalytic site as well as at the microtubule binding domain and the neck linker. Molecular dynamics simulations reveal that this mutation affects the dynamics not only of L5 itself but also of the switch I structural elements that sense ATP binding to the catalytic site. Our results lead us to propose that L5 regulates the rate of conformational change in key elements of the nucleotide binding site through its interactions with α3 and in so doing controls the speed of movement and force generation in kinesin motors.


Subject(s)
Computer Simulation , Kinesins/chemistry , Models, Molecular , Animals , Catalytic Domain , Humans , Kinesins/genetics , Mutagenesis, Site-Directed , Protein Structure, Secondary
16.
Chembiochem ; 13(18): 2683-9, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23169461

ABSTRACT

Cyanobactins, including patellamides, are a group of cyanobacterial post-translationally modified ribosomal cyclic peptides. The final product should in theory be predictable from the sequence of the precursor peptide and the associated tailoring enzymes. Understanding the mechanism and recognition requirements of these enzymes will allow their rational engineering. We have identified three new cyanobactins from a Cyanothece PCC 7425 culture subjected to a heat shock. One of these compounds revealed a novel signature signal for ThcA, the subtilisin-like serine protease that is homologous to the patellamide protease PatA. The crystal structure of the latter and modelling studies allow rationalisation of the recognition determinants for both enzymes, consistent with the ribosomal biosynthetic origin of the new compounds.


Subject(s)
Cyanothece/metabolism , Peptides, Cyclic/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Peptides, Cyclic/chemistry , Protein Structure, Tertiary , Serine Proteases/chemistry , Serine Proteases/metabolism
17.
J Chem Theory Comput ; 18(9): 5710-5724, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-35972903

ABSTRACT

Homology models have been used for virtual screening and to understand the binding mode of a known active compound; however, rarely have the models been shown to be of sufficient accuracy, comparable to crystal structures, to support free-energy perturbation (FEP) calculations. We demonstrate here that the use of an advanced induced-fit docking methodology reliably enables predictive FEP calculations on congeneric series across homology models ≥30% sequence identity. Furthermore, we show that retrospective FEP calculations on a congeneric series of drug-like ligands are sufficient to discriminate between predicted binding modes. Results are presented for a total of 29 homology models for 14 protein targets, showing FEP results comparable to those obtained using experimentally determined crystal structures for 86% of homology models with template structure sequence identities ranging from 30 to 50%. Implications for the use and validation of homology models in drug discovery projects are discussed.


Subject(s)
Drug Discovery , Entropy , Ligands , Molecular Docking Simulation , Protein Binding , Retrospective Studies
18.
J Med Chem ; 65(13): 9350-9375, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35727231

ABSTRACT

With over 200 million cases and close to half a million deaths each year, malaria is a threat to global health, particularly in developing countries. Plasmodium falciparum, the parasite that causes the most severe form of the disease, has developed resistance to all antimalarial drugs. Resistance to the first-line antimalarial artemisinin and to artemisinin combination therapies is widespread in Southeast Asia and is emerging in sub-Saharan Africa. The P. falciparum proteasome is an attractive antimalarial target because its inhibition kills the parasite at multiple stages of its life cycle and restores artemisinin sensitivity in parasites that have become resistant through mutation in Kelch K13. Here, we detail our efforts to develop noncovalent, macrocyclic peptide malaria proteasome inhibitors, guided by structural analysis and pharmacokinetic properties, leading to a potent, species-selective, metabolically stable inhibitor.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemisinins/pharmacology , Drug Resistance , Humans , Malaria, Falciparum/drug therapy , Peptides/therapeutic use , Plasmodium falciparum , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Protozoan Proteins/genetics
19.
J Med Chem ; 65(3): 1996-2022, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35044775

ABSTRACT

A newly validated target for tuberculosis treatment is phosphopantetheinyl transferase, an essential enzyme that plays a critical role in the biosynthesis of cellular lipids and virulence factors in Mycobacterium tuberculosis. The structure-activity relationships of a recently disclosed inhibitor, amidinourea (AU) 8918 (1), were explored, focusing on the biochemical potency, determination of whole-cell on-target activity for active compounds, and profiling of selective active congeners. These studies show that the AU moiety in AU 8918 is largely optimized and that potency enhancements are obtained in analogues containing a para-substituted aromatic ring. Preliminary data reveal that while some analogues, including 1, have demonstrated cardiotoxicity (e.g., changes in cardiomyocyte beat rate, amplitude, and peak width) and inhibit Cav1.2 and Nav1.5 ion channels (although not hERG channels), inhibition of the ion channels is largely diminished for some of the para-substituted analogues, such as 5k (p-benzamide) and 5n (p-phenylsulfonamide).


Subject(s)
Bacterial Proteins/metabolism , Guanidine/analogs & derivatives , Mycobacterium tuberculosis/enzymology , Transferases (Other Substituted Phosphate Groups)/metabolism , Urea/analogs & derivatives , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , Guanidine/chemistry , Guanidine/metabolism , Guanidine/pharmacology , Kinetics , Microbial Sensitivity Tests , Molecular Conformation , Molecular Dynamics Simulation , Mycobacterium tuberculosis/drug effects , Structure-Activity Relationship , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Urea/chemistry , Urea/metabolism , Urea/pharmacology
20.
J Mol Biol ; 433(15): 167055, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34022208

ABSTRACT

We describe an enhancement of traditional genomics-based approaches to improve the success of structure determination of membrane proteins. Following a broad screen of sequence space to identify initial expression-positive targets, we employ a second step to select orthologs with closely related sequences to these hits. We demonstrate that a greater percentage of these latter targets express well and are stable in detergent, increasing the likelihood of identifying candidates that will ultimately yield structural information.


Subject(s)
Bacteria/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Amino Acid Sequence , Bacteria/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Genomics , Models, Molecular , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL