Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Science ; 380(6652): 1349-1356, 2023 06 30.
Article in English | MEDLINE | ID: mdl-37384702

ABSTRACT

Millions who live in Latin America and sub-Saharan Africa are at risk of trypanosomatid infections, which cause Chagas disease and human African trypanosomiasis (HAT). Improved HAT treatments are available, but Chagas disease therapies rely on two nitroheterocycles, which suffer from lengthy drug regimens and safety concerns that cause frequent treatment discontinuation. We performed phenotypic screening against trypanosomes and identified a class of cyanotriazoles (CTs) with potent trypanocidal activity both in vitro and in mouse models of Chagas disease and HAT. Cryo-electron microscopy approaches confirmed that CT compounds acted through selective, irreversible inhibition of trypanosomal topoisomerase II by stabilizing double-stranded DNA:enzyme cleavage complexes. These findings suggest a potential approach toward successful therapeutics for the treatment of Chagas disease.


Subject(s)
Chagas Disease , Topoisomerase II Inhibitors , Triazoles , Trypanosoma , Trypanosomiasis, African , Animals , Humans , Mice , Chagas Disease/drug therapy , Cryoelectron Microscopy , DNA Topoisomerases, Type II/metabolism , Trypanosoma/drug effects , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/therapeutic use , Trypanosomiasis, African/drug therapy , Drug Evaluation, Preclinical
2.
Int J Parasitol Drugs Drug Resist ; 20: 121-128, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36375339

ABSTRACT

Malaria is among the tropical diseases that cause the most deaths in Africa. Around 500,000 malaria deaths are reported yearly among African children under the age of five. Chloroquine (CQ) is a low-cost antimalarial used worldwide for the treatment of Plasmodium vivax malaria. Due to resistance mechanisms, CQ is no longer effective against most malaria cases caused by P. falciparum. The World Health Organization recommends artemisinin combination therapies for P. falciparum malaria, but resistance is emerging in Southeast Asia and some parts of Africa. Therefore, new medicines for treating malaria are urgently needed. Previously, our group identified the 4-aminoquinoline DAQ, a CQ analog containing an acetylenic bond in its side chain, which overcomes CQ resistance in K1 P. falciparum strains. In this work, the antiplasmodial profile, drug-like properties, and pharmacokinetics of DAQ were further investigated. DAQ showed no cross-resistance against standard CQ-resistant strains (e.g., Dd2, IPC 4912, RF12) nor against P. falciparum and P. vivax isolates from patients in the Brazilian Amazon. Using drug pressure assays, DAQ showed a low propensity to generate resistance. DAQ showed considerable solubility but low metabolic stability. The main metabolite was identified as a mono N-deethylated derivative (DAQM), which also showed significant inhibitory activity against CQ-resistant P. falciparum strains. Our findings indicated that the presence of a triple bond in CQ-analogues may represent a low-cost opportunity to overcome known mechanisms of resistance in the malaria parasite.


Subject(s)
Antimalarials , Malaria, Falciparum , Malaria, Vivax , Malaria , Plasmodium , Child , Humans , Chloroquine/pharmacology , Chloroquine/therapeutic use , Plasmodium falciparum , Acetylene/pharmacology , Acetylene/therapeutic use , Alkynes/pharmacology , Alkynes/therapeutic use , Drug Resistance , Antimalarials/therapeutic use , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Malaria, Vivax/drug therapy , Malaria/drug therapy
3.
J Med Chem ; 64(14): 10102-10123, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34255515

ABSTRACT

CREBBP (CBP/KAT3A) and its paralogue EP300 (KAT3B) are lysine acetyltransferases (KATs) that are essential for human development. They each comprise 10 domains through which they interact with >400 proteins, making them important transcriptional co-activators and key nodes in the human protein-protein interactome. The bromodomains of CREBBP and EP300 enable the binding of acetylated lysine residues from histones and a number of other important proteins, including p53, p73, E2F, and GATA1. Here, we report a work to develop a high-affinity, small-molecule ligand for the CREBBP and EP300 bromodomains [(-)-OXFBD05] that shows >100-fold selectivity over a representative member of the BET bromodomains, BRD4(1). Cellular studies using this ligand demonstrate that the inhibition of the CREBBP/EP300 bromodomain in HCT116 colon cancer cells results in lowered levels of c-Myc and a reduction in H3K18 and H3K27 acetylation. In hypoxia (<0.1% O2), the inhibition of the CREBBP/EP300 bromodomain results in the enhanced stabilization of HIF-1α.


Subject(s)
Benzodiazepinones/pharmacology , CREB-Binding Protein/antagonists & inhibitors , Drug Design , E1A-Associated p300 Protein/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Benzodiazepinones/chemical synthesis , Benzodiazepinones/chemistry , CREB-Binding Protein/metabolism , Dose-Response Relationship, Drug , E1A-Associated p300 Protein/metabolism , HCT116 Cells , Humans , Ligands , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
4.
ACS Chem Biol ; 16(7): 1159-1163, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34165961

ABSTRACT

The nuclear receptor-related 1 protein, Nurr1, is a transcription factor critical for the development and maintenance of dopamine-producing neurons in the substantia nigra pars compacta, a cell population that progressively loses the ability to make dopamine and degenerates in Parkinson's disease. Recently, we demonstrated that Nurr1 binds directly to and is regulated by the endogenous dopamine metabolite 5,6-dihydroxyindole (DHI). Unfortunately, DHI is an unstable compound, and thus a poor tool for studying Nurr1 function. Here, we report that 5-chloroindole, an unreactive analog of DHI, binds directly to the Nurr1 ligand binding domain with micromolar affinity and stimulates the activity of Nurr1, including the transcription of genes governing the synthesis and packaging of dopamine.


Subject(s)
Enzyme Activators/pharmacology , Indoles/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/agonists , Animals , Cell Line , Enzyme Activators/metabolism , Enzyme Activators/toxicity , Indoles/metabolism , Indoles/toxicity , Mice , Mutation , Nuclear Receptor Subfamily 4, Group A, Member 2/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Protein Binding , Protein Domains/genetics
5.
Cancer Discov ; 11(6): 1424-1439, 2021 06.
Article in English | MEDLINE | ID: mdl-33563661

ABSTRACT

Despite a remarkable increase in the genomic profiling of cancer, integration of genomic discoveries into clinical care has lagged behind. We report the feasibility of rapid identification of targetable mutations in 153 pediatric patients with relapsed/refractory or high-risk leukemias enrolled on a prospective clinical trial conducted by the LEAP Consortium. Eighteen percent of patients had a high confidence Tier 1 or 2 recommendation. We describe clinical responses in the 14% of patients with relapsed/refractory leukemia who received the matched targeted therapy. Further, in order to inform future targeted therapy for patients, we validated variants of uncertain significance, performed ex vivo drug-sensitivity testing in patient leukemia samples, and identified new combinations of targeted therapies in cell lines and patient-derived xenograft models. These data and our collaborative approach should inform the design of future precision medicine trials. SIGNIFICANCE: Patients with relapsed/refractory leukemias face limited treatment options. Systematic integration of precision medicine efforts can inform therapy. We report the feasibility of identifying targetable mutations in children with leukemia and describe correlative biology studies validating therapeutic hypotheses and novel mutations.See related commentary by Bornhauser and Bourquin, p. 1322.This article is highlighted in the In This Issue feature, p. 1307.


Subject(s)
Leukemia/drug therapy , Neoplasm Recurrence, Local/drug therapy , Biomarkers, Tumor/genetics , Child , Cohort Studies , Disease Progression , Feasibility Studies , Female , Humans , Leukemia/genetics , Leukemia/mortality , Male , Molecular Targeted Therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Prospective Studies , United States
6.
Biochemistry ; 60(10): 802-812, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33635645

ABSTRACT

It was previously shown that human platelet 12S-lipoxygenase (h12-LOX) exists as a dimer; however, the specific structure is unknown. In this study, we create a model of the dimer through a combination of computational methods, experimental mutagenesis, and hydrogen-deuterium exchange (HDX) investigations. Initially, Leu183 and Leu187 were replaced by negatively charged glutamate residues and neighboring aromatic residues were replaced with alanine residues (F174A/W176A/L183E/L187E/Y191A). This quintuple mutant disrupted both the hydrophobic and π-π interactions, generating an h12-LOX monomer. To refine the determinants for dimer formation further, the L183E/L187E mutant was generated and the equilibrium shifted mostly toward the monomer. We then submitted the predicted monomeric structure to protein-protein docking to create a model of the dimeric complex. A total of nine of the top 10 most energetically favorable docking conformations predict a TOP-to-TOP dimeric arrangement of h12-LOX, with the α-helices containing a Leu-rich region (L172, L183, L187, and L194), corroborating our experimental results showing the importance of these hydrophobic interactions for dimerization. This model was supported by HDX investigations that demonstrated the stabilization of four, non-overlapping peptides within helix α2 of the TOP subdomain for wt-h12-LOX, consistent with the dimer interface. Most importantly, our data reveal that the dimer and monomer of h12-LOX have distinct biochemical properties, suggesting that the structural changes due to dimerization have allosteric effects on active site catalysis and inhibitor binding.


Subject(s)
Arachidonate 12-Lipoxygenase/chemistry , Arachidonate 12-Lipoxygenase/metabolism , Deuterium Exchange Measurement/methods , Molecular Docking Simulation/methods , Mutagenesis , Mutation , Protein Multimerization , Arachidonate 12-Lipoxygenase/genetics , Catalytic Domain , Humans , Models, Molecular , Protein Conformation
7.
Value Health Reg Issues ; 20: 191-195, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31733406

ABSTRACT

Regulation of drug prices that ensures adequate access to effective treatments and promotes innovation is a global challenge. In the United States, the government does not regulate drug prices when they come onto market. On the other hand, in countries such as France and Brazil, government agencies are responsible for setting up price limits by leveraging the interests of the companies and the countries' population. In Brazil, safety and efficacy of drugs are regulated by the Brazilian Health Regulatory Agency, and drug prices are regulated by the Pharmaceutical Market Regulation Chamber with a participation of Brazilian Health Regulatory Agency. Here, we introduce the current challenges faced by the Brazilian government in the drug price regulation and present proposed initiatives aiming to streamline access to innovative treatments for its citizens.


Subject(s)
Cost Control/legislation & jurisprudence , Drug Costs/legislation & jurisprudence , Government Regulation , Brazil , Cost Control/methods , Delivery of Health Care/legislation & jurisprudence , Delivery of Health Care/organization & administration , Humans , International Cooperation , Rare Diseases/drug therapy , Rare Diseases/economics
8.
PLoS Genet ; 15(8): e1008295, 2019 08.
Article in English | MEDLINE | ID: mdl-31398187

ABSTRACT

The progressive failure of protein homeostasis is a hallmark of aging and a common feature in neurodegenerative disease. As the enzymes executing the final stages of autophagy, lysosomal proteases are key contributors to the maintenance of protein homeostasis with age. We previously reported that expression of granulin peptides, the cleavage products of the neurodegenerative disease protein progranulin, enhance the accumulation and toxicity of TAR DNA binding protein 43 (TDP-43) in Caenorhabditis elegans (C. elegans). In this study we show that C. elegans granulins are produced in an age- and stress-dependent manner. Granulins localize to the endolysosomal compartment where they impair lysosomal protease expression and activity. Consequently, protein homeostasis is disrupted, promoting the nuclear translocation of the lysosomal transcription factor HLH-30/TFEB, and prompting cells to activate a compensatory transcriptional program. The three C. elegans granulin peptides exhibited distinct but overlapping functional effects in our assays, which may be due to amino acid composition that results in distinct electrostatic and hydrophobicity profiles. Our results support a model in which granulin production modulates a critical transition between the normal, physiological regulation of protease activity and the impairment of lysosomal function that can occur with age and disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/genetics , Granulins/metabolism , Lysosomes/metabolism , Neurodegenerative Diseases/genetics , Aging/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Disease Models, Animal , Endopeptidases/metabolism , Gene Expression Regulation , Granulins/genetics , Humans , Neurodegenerative Diseases/pathology , Stress, Physiological/genetics
9.
Cancer Res ; 79(16): 4283-4292, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31270078

ABSTRACT

KIT is a type-3 receptor tyrosine kinase that is frequently mutated at exon 11 or 17 in a variety of cancers. First-generation KIT tyrosine kinase inhibitors (TKI) are ineffective against KIT exon 17 mutations, which favor an active conformation that prevents these TKIs from binding. The ATP-competitive inhibitors, midostaurin and avapritinib, which target the active kinase conformation, were developed to inhibit exon 17-mutant KIT. Because secondary kinase domain mutations are a common mechanism of TKI resistance and guide ensuing TKI design, we sought to define problematic KIT kinase domain mutations for these emerging therapeutics. Midostaurin and avapritinib displayed different vulnerabilities to secondary kinase domain substitutions, with the T670I gatekeeper mutation being selectively problematic for avapritinib. Although gatekeeper mutations often directly disrupt inhibitor binding, we provide evidence that T670I confers avapritinib resistance indirectly by inducing distant conformational changes in the phosphate-binding loop. These findings suggest combining midostaurin and avapritinib may forestall acquired resistance mediated by secondary kinase domain mutations. SIGNIFICANCE: This study identifies potential problematic kinase domain mutations for next-generation KIT inhibitors midostaurin and avapritinib.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Pyrazoles/pharmacology , Pyrroles/pharmacology , Staurosporine/analogs & derivatives , Triazines/pharmacology , Cell Line , Drug Resistance, Neoplasm/drug effects , Exons , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/metabolism , Staurosporine/chemistry , Staurosporine/pharmacology
10.
Biochemistry ; 58(23): 2670-2674, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31099551

ABSTRACT

Progranulin (PGRN) is an evolutionarily conserved glycoprotein associated with several disease states, including neurodegeneration, cancer, and autoimmune disorders. This protein has recently been implicated in the regulation of lysosome function, whereby PGRN may bind to and promote the maturation and activity of the aspartyl protease cathepsin D (proCTSD, inactive precursor; matCTSD, mature, enzymatically active form). As the full-length PGRN protein can be cleaved into smaller peptides, called granulins, we assessed the function of these granulin peptides in binding to proCTSD and stimulating matCTSD enzyme activity in vitro. Here, we report that full-length PGRN and multi-granulin domain peptides bound to proCTSD with low to submicromolar binding affinities. This binding promoted proCTSD destabilization, the magnitude of which was greater for multi-granulin domain peptides than for full-length PGRN. Such destabilization correlated with enhanced matCTSD activity at acidic pH. The presence and function of multi-granulin domain peptides have typically been overlooked in previous studies. This work provides the first in vitro quantification of their binding and activity on proCTSD. Our study highlights the significance of multi-granulin domain peptides in the regulation of proCTSD maturation and enzymatic activity and suggests that attention to PGRN processing will be essential for the future understanding of the molecular mechanisms leading to neurodegenerative disease states with loss-of-function mutations in PGRN.


Subject(s)
Cathepsin D/metabolism , Enzyme Precursors/metabolism , Granulins/metabolism , Humans , Protein Binding , Protein Conformation , Protein Stability , Transition Temperature
11.
J Biol Chem ; 294(22): 8779-8790, 2019 05 31.
Article in English | MEDLINE | ID: mdl-30992364

ABSTRACT

Tau, a member of the MAP2/tau family of microtubule-associated proteins, stabilizes and organizes axonal microtubules in healthy neurons. In neurodegenerative tauopathies, tau dissociates from microtubules and forms neurotoxic extracellular aggregates. MAP2/tau family proteins are characterized by three to five conserved, intrinsically disordered repeat regions that mediate electrostatic interactions with the microtubule surface. Here, we used molecular dynamics, microtubule-binding experiments, and live-cell microscopy, revealing that highly-conserved histidine residues near the C terminus of each microtubule-binding repeat are pH sensors that can modulate tau-microtubule interaction strength within the physiological intracellular pH range. We observed that at low pH (<7.5), these histidines are positively charged and interact with phenylalanine residues in a hydrophobic cleft between adjacent tubulin dimers. At higher pH (>7.5), tau deprotonation decreased binding to microtubules both in vitro and in cells. Electrostatic and hydrophobic characteristics of histidine were both required for tau-microtubule binding, as substitutions with constitutively and positively charged nonaromatic lysine or uncharged alanine greatly reduced or abolished tau-microtubule binding. Consistent with these findings, tau-microtubule binding was reduced in a cancer cell model with increased intracellular pH but was rapidly restored by decreasing the pH to normal levels. These results add detailed insights into the intracellular regulation of tau activity that may be relevant in both normal and pathological conditions.


Subject(s)
Histidine/metabolism , Microtubules/metabolism , tau Proteins/metabolism , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Kinetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Static Electricity , tau Proteins/genetics
12.
Biochem Biophys Res Commun ; 512(2): 208-212, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30878184

ABSTRACT

Protein kinase CK2 has emerged as an attractive cancer therapeutic target. Previous studies have highlighted the challenge of optimizing CK2 ATP-competitive inhibitors that have low druggability due to their polycyclic ring scaffolds. Therefore the development of novel inhibitors with non-polycyclic scaffolds emerges as a promising strategy for drug discovery targeting CK2. In this current study, based on the similar predicted binding poses of the linear 2-propenone scaffold of isoliquiritigenin with that of the polycyclic inhibitor CX-4945, a series of 2-propenone derivatives containing an amine-substituted five-membered heterocycle and a benzoic acid were designed, synthesized and evaluated for their in vitro CK2 inhibition and anti-cancer activity. Compound 8b was found to be the most potent CK2 inhibitor (IC50 = 0.6 µM) with the anti-proliferative activity on HepG2 cancer cells (IC50 = 14 µM), compared to the activity of isoliquiritigenin (IC50 = 17 µM and 51 µM, respectively). Molecular docking was performed to understand the binding modes of the newly designed 2-propenone derivatives with CK2. Compound 8b formed the most favorable network of hydrogen bonds with both the hinge region and positive area. Our results indicate that CK2 derivatives with a linear 2-propenone scaffold are promising candidates for anti-cancer drug discovery.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Casein Kinase II/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Alkenes/chemistry , Alkenes/pharmacology , Casein Kinase II/metabolism , Cell Proliferation/drug effects , Drug Design , Hep G2 Cells , Humans , Molecular Docking Simulation , Neoplasms/drug therapy , Structure-Activity Relationship
13.
J Mol Biol ; 431(5): 1038-1047, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30690031

ABSTRACT

Single-copy loss-of-function mutations in the progranulin gene (PGRN) underlie the neurodegenerative disease frontotemporal lobar degeneration, while homozygous loss-of-function of PGRN results in the lysosomal storage disorder neuronal ceroid lipofuscinosis. Despite evidence that normal PGRN levels are critical for neuronal health, the function of this protein is not yet understood. Here, we show that PGRN stimulates the in vitro maturation of the lysosomal aspartyl protease cathepsin D (CTSD). CTSD is delivered to the endolysosomal system as an inactive precursor (proCTSD) and requires sequential cleavage steps via intermediate forms to achieve the mature state (matCTSD). In co-immunoprecipitation experiments, PGRN interacts predominantly with immature pro- and intermediate forms of CTSD. PGRN enhances in vitro conversion of proCTSD to matCTSD in a concentration-dependent manner. Differential scanning fluorimetry shows a destabilizing effect induced by PGRN on proCTSD folding (∆Tm = -1.7 °C at a 3:1 molar ratio). We propose a mechanism whereby PGRN binds to proCTSD, destabilizing the propeptide from the enzyme catalytic core and favoring conversion to mature forms of the enzyme. Further understanding of the role of PGRN in CTSD maturation will assist in the development of targeted therapies for neurodegenerative disease.


Subject(s)
Cathepsin D/metabolism , Enzyme Precursors/metabolism , Progranulins/metabolism , Cathepsin D/genetics , Cell Line , Enzyme Precursors/genetics , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Mutation/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Progranulins/genetics
14.
Mol Inform ; 38(3): e1800089, 2019 03.
Article in English | MEDLINE | ID: mdl-30307134

ABSTRACT

Protein kinase CK2 is considered as an emerging target in cancer therapy, and recent efforts have been made to develop its ATP-competitive inhibitors, but achieving selectivity with respect to related kinases remains challenging because of the highly conserved ATP-binding pocket of kinases. Non-ATP competitive inhibitors might solve this challenge; one such strategy is to identify compounds that target the CK2α/CK2ß interface as CK2 holoenzyme antagonists. Here we improved the binding affinity to CK2α and cell-based anti-cancer activity of a CK2ß-derived cyclic peptide (Pc) by combining structure-based computational design with experimental evaluation. By analyzing molecular dynamics simulations of Pc bound to CK2α, a series of Pc-derived peptides was rationally designed and synthesized to evaluate their binding affinity to CK2α, as well as anti-proliferative and pro-apoptotic effects against HepG2 cancer cell line. One amino acid substitutions on Pc, I192F, exhibited over 10-fold improvement in the predicted binding affinity to CK2α when compared to Pc, and a cell-permeable version, I192F-Tat, also demonstrated more potent anti-proliferative and pro-apoptotic effects against HepG2 compared to Pc. A second modification of Pc, H193W, also led to more potent cell-based activity, despite having weaker binding affinity (∼5×) to CK2α. The discovery of the I192F and H193W peptides provides new insights for further optimization of CK2 antagonist candidates as anti-cancer leads.


Subject(s)
Antineoplastic Agents/chemistry , Casein Kinase II/antagonists & inhibitors , Peptides, Cyclic/chemistry , Protein Kinase Inhibitors/chemistry , Quantitative Structure-Activity Relationship , Amino Acid Substitution , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites , Casein Kinase II/chemistry , Casein Kinase II/genetics , Casein Kinase II/metabolism , Drug Discovery , Hep G2 Cells , Humans , Molecular Docking Simulation , Peptides, Cyclic/pharmacology , Protein Binding , Protein Kinase Inhibitors/pharmacology
15.
Int J Parasitol Drugs Drug Resist ; 8(3): 459-464, 2018 12.
Article in English | MEDLINE | ID: mdl-30396013

ABSTRACT

In spite of recent efforts to eradicate malaria in the world, this parasitic disease is still considered a major public health problem, with a total of 216 million cases of malaria and 445,000 deaths in 2016. Artemisinin-based combination therapies remain effective in most parts of the world, but recent cases of resistance in Southeast Asia have urged for novel approaches to treat malaria caused by Plasmodium falciparum. In this work, we present chloroquine analogs that exhibited high activity against sensitive and chloroquine-resistant P. falciparum blood parasites and were also active against P. berghei infected mice. Among the compounds tested, DAQ, a chloroquine analog with a more linear side chain, was shown to be the most active in vitro and in vivo, with low cytotoxicity, and therefore may serve as the basis for the development of more effective chloroquine analogs to aid malaria eradication.


Subject(s)
Antimalarials/pharmacology , Antimalarials/therapeutic use , Chloroquine/analogs & derivatives , Chloroquine/chemistry , Drug Design , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Animals , Antimalarials/isolation & purification , Chloroquine/pharmacology , Chloroquine/therapeutic use , Drug Resistance , Hep G2 Cells , Humans , Malaria/drug therapy , Mice , Parasitic Sensitivity Tests
16.
Chem Sci ; 9(10): 2655-2665, 2018 Mar 14.
Article in English | MEDLINE | ID: mdl-29719674

ABSTRACT

We have studied the cation-π interactions of neutral aromatic ligands with the cationic amino acid residues arginine, histidine and lysine using ab initio calculations, symmetry adapted perturbation theory (SAPT), and a systematic meta-analysis of all available Protein Data Bank (PDB) X-ray structures. Quantum chemical potential energy surfaces (PES) for these interactions were obtained at the DLPNO-CCSD(T) level of theory and compared against the empirical distribution of 2012 unique protein-ligand cation-π interactions found in X-ray crystal structures. We created a workflow to extract these structures from the PDB, filtering by interaction type and residue pKa. The gas phase cation-π interaction of lysine is the strongest by more than 10 kcal mol-1, but the empirical distribution of 582 X-ray structures lies away from the minimum on the interaction PES. In contrast, 1381 structures involving arginine match the underlying calculated PES with good agreement. SAPT analysis revealed that underlying differences in the balance of electrostatic and dispersion contributions are responsible for this behavior in the context of the protein environment. The lysine-arene interaction, dominated by electrostatics, is greatly weakened by a surrounding dielectric medium and causes it to become essentially negligible in strength and without a well-defined equilibrium separation. The arginine-arene interaction involves a near equal mix of dispersion and electrostatic attraction, which is weakened to a much smaller degree by the surrounding medium. Our results account for the paucity of cation-π interactions involving lysine, even though this is a more common residue than arginine. Aromatic ligands are most likely to interact with cationic arginine residues as this interaction is stronger than for lysine in higher polarity surroundings.

17.
Biochemistry ; 56(32): 4219-4234, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28656748

ABSTRACT

Ligand binding is one of the most fundamental properties of proteins. Ligand functions fall into three basic types: substrates, regulatory molecules, and cofactors essential to protein stability, reactivity, or enzyme-substrate complex formation. The regulation of potassium ion movement in bacteria is predominantly under the control of regulatory ligands that gate the relevant channels and transporters, which possess subunits or domains that contain Rossmann folds (RFs). Here we demonstrate that adenosine monophosphate (AMP) is bound to both RFs of the dimeric bacterial Kef potassium efflux system (Kef), where it plays a structural role. We conclude that AMP binds with high affinity, ensuring that the site is fully occupied at all times in the cell. Loss of the ability to bind AMP, we demonstrate, causes protein, and likely dimer, instability and consequent loss of function. Kef system function is regulated via the reversible binding of comparatively low-affinity glutathione-based ligands at the interface between the dimer subunits. We propose this interfacial binding site is itself stabilized, at least in part, by AMP binding.


Subject(s)
Adenosine Monophosphate/chemistry , Potassium-Hydrogen Antiporters/chemistry , Protein Folding , Protein Multimerization , Shewanella/chemistry , Adenosine Monophosphate/genetics , Adenosine Monophosphate/metabolism , Potassium-Hydrogen Antiporters/genetics , Potassium-Hydrogen Antiporters/metabolism , Protein Binding , Protein Domains , Protein Stability , Protein Structure, Quaternary , Shewanella/genetics , Shewanella/metabolism
18.
J Biol Chem ; 292(32): 13243-13257, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28539361

ABSTRACT

Nicotinic acid adenine dinucleotide phosphate (NAADP) and cyclic ADP-ribose (cADPR) are Ca2+-mobilizing messengers important for modulating cardiac excitation-contraction coupling and pathophysiology. CD38, which belongs to the ADP-ribosyl cyclase family, catalyzes synthesis of both NAADP and cADPR in vitro However, it remains unclear whether this is the main enzyme for their production under physiological conditions. Here we show that membrane fractions from WT but not CD38-/- mouse hearts supported NAADP and cADPR synthesis. Membrane permeabilization of cardiac myocytes with saponin and/or Triton X-100 increased NAADP synthesis, indicating that intracellular CD38 contributes to NAADP production. The permeabilization also permitted immunostaining of CD38, with a striated pattern in WT myocytes, whereas CD38-/- myocytes and nonpermeabilized WT myocytes showed little or no staining, without striation. A component of ß-adrenoreceptor signaling in the heart involves NAADP and lysosomes. Accordingly, in the presence of isoproterenol, Ca2+ transients and contraction amplitudes were smaller in CD38-/- myocytes than in the WT. In addition, suppressing lysosomal function with bafilomycin A1 reduced the isoproterenol-induced increase in Ca2+ transients in cardiac myocytes from WT but not CD38-/- mice. Whole hearts isolated from CD38-/- mice and exposed to isoproterenol showed reduced arrhythmias. SAN4825, an ADP-ribosyl cyclase inhibitor that reduces cADPR and NAADP synthesis in mouse membrane fractions, was shown to bind to CD38 in docking simulations and reduced the isoproterenol-induced arrhythmias in WT hearts. These observations support generation of NAADP and cADPR by intracellular CD38, which contributes to effects of ß-adrenoreceptor stimulation to increase both Ca2+ transients and the tendency to disturb heart rhythm.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Calcium Signaling , Cyclic ADP-Ribose/metabolism , Membrane Glycoproteins/metabolism , Myocytes, Cardiac/metabolism , NADP/analogs & derivatives , Sarcoplasmic Reticulum/metabolism , ADP-ribosyl Cyclase 1/antagonists & inhibitors , Adrenergic beta-Agonists/pharmacology , Animals , Anti-Arrhythmia Agents/chemistry , Anti-Arrhythmia Agents/metabolism , Anti-Arrhythmia Agents/pharmacology , Calcium Signaling/drug effects , Cell Membrane Permeability/drug effects , Cells, Cultured , Detergents/pharmacology , Enzyme Inhibitors/pharmacology , Heart/drug effects , In Vitro Techniques , Male , Membrane Glycoproteins/antagonists & inhibitors , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , NADP/metabolism , Protein Transport/drug effects , Rabbits , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/enzymology , Single-Cell Analysis
19.
Org Biomol Chem ; 14(46): 10926-10938, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27814427

ABSTRACT

CREBBP bromodomains, epigenetic "reader" proteins that recognize acetylated histone lysine residues, are a current target for cancer therapy. We show that experimental CREBBP binding affinities of small-molecules with aromatic or heteroaromatic functional groups are strongly influenced by a cation-π interaction with a positively charged arginine residue. For a series of fifteen 5-isoxazolylbenzimidazole derivatives, the strength of this non-covalent interaction is directly related to improvements in binding to CREBBP. The aromatic substituents' inductive and resonance effects are not obviously correlated with observed structure and affinity relationships. In contrast, a coulombic electrostatic model can quantitatively predict the interaction strength. We have assessed different Molecular Mechanics (MM) and Quantum Mechanics (QM) descriptions of the protein-ligand interaction. Quantitative models for binding affinity were generated from: (1) Poisson Boltzmann Surface Area (MM-PBSA) and Generalized Born Surface Area (MM-GBSA) scoring functions that incorporated the entire ligand and (2) QM-complexation energies and (3) Electrostatic Potential Surface values (ESPs) that analyzed the varying aromatic group. A linear relationship between QM-computed ESP values is established for the cation-π interaction strength, and gives the best correlation (R2 = 0.84) with experimental binding affinities. This model also ranks ligand affinity most accurately (rs = 0.91) from the models tested. Consideration of the electrostatic potential in response to the local effects of substituents in addition to that of the aromatic ring is necessary to understand and describe the interaction with the cationic guanidinium ion. This leads to an improved understanding and the ability to quantitatively predict the magnitude of non-covalent interactions in the CREBBP active site.

SELECTION OF CITATIONS
SEARCH DETAIL
...