Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Eur J Med Chem ; 220: 113454, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-33901900

ABSTRACT

Artemisinin-based combination therapies (ACTs) have been able to reduce the clinical and pathological malaria cases in endemic areas around the globe. However, recent reports have shown a progressive decline in malaria parasite clearance in South-east Asia after ACT treatment, thus envisaging a need for new artemisinin (ART) derivatives and combinations. To address the emergence of drug resistance to current antimalarials, here we report the synthesis of artemisinin-peptidyl vinyl phosphonate hybrid molecules that show superior efficacy than artemisinin alone against chloroquine-resistant as well as multidrug-resistant Plasmodium falciparum strains with EC50 in pico-molar ranges. Further, the compounds effectively inhibited the survival of ring-stage parasite for laboratory-adapted artemisinin-resistant parasite lines as compared to artemisinin. These hybrid molecules showed complete parasite clearance in vivo using P. berghei mouse malaria model in comparison to artemisinin alone. Studies on the mode of action of hybrid molecules suggested that these artemisinin-peptidyl vinyl phosphonate hybrid molecules possessed dual activities: inhibited falcipain-2 (FP-2) activity, a P. falciparum cysteine protease involved in hemoglobin degradation, and also blocked the hemozoin formation in the food-vacuole, a step earlier shown to be blocked by artemisinin. Since these hybrid molecules blocked multiple steps of a pathway and showed synergistic efficacies, we believe that these lead compounds can be developed as effective antimalarials to prevent the spread of resistance to current antimalarials.


Subject(s)
Antimalarials/pharmacology , Drug Resistance, Multiple/drug effects , Malaria/drug therapy , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Artemisinins/chemical synthesis , Artemisinins/chemistry , Artemisinins/pharmacology , Cysteine Endopeptidases/metabolism , Dose-Response Relationship, Drug , Heme/antagonists & inhibitors , Heme/metabolism , Malaria/metabolism , Molecular Structure , Organophosphonates/chemical synthesis , Organophosphonates/chemistry , Organophosphonates/pharmacology , Parasitic Sensitivity Tests , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Polymerization/drug effects , Structure-Activity Relationship , Vinyl Compounds/chemical synthesis , Vinyl Compounds/chemistry , Vinyl Compounds/pharmacology
2.
Bioorg Chem ; 104: 104243, 2020 11.
Article in English | MEDLINE | ID: mdl-32920360

ABSTRACT

A library of 33 polymethoxylated flavones (PMF) was evaluated for heme-binding affinity by biomimetic MS assay and in vitro antiplasmodial activity on two strains of P. falciparum. Stability of heme adducts was discussed using the dissociation voltage at 50% (DV50). No correlation was observed between the methoxylation pattern and the antiparasitic activity, either for the 3D7 chloroquine-sensitive or for the W2 chloroquine-resistant P. falciparum strains. However, in each PMF family an increased DV50 was observed for the derivatives methoxylated in position 5. Measurement of intra-erythrocytic hemozoin formation of selected derivatives was performed and hemozoin concentration was inversely correlated with heme-binding affinity. Kaempferol showed no influence on hemozoin formation, reinforcing the hypothesis that this compound may exert in vitro antiplasmodial activity mostly through other pathways. Pentamethoxyquercetin has simultaneously demonstrated a significant biological activity and a strong interaction with heme, suggesting that inhibition of hemozoin formation is totally or partially responsible for its antiparasitic effect.


Subject(s)
Antimalarials/pharmacology , Flavonoids/pharmacology , Heme/antagonists & inhibitors , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flavonoids/chemical synthesis , Flavonoids/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Molecular Structure , Structure-Activity Relationship
3.
Biomol Concepts ; 11(1): 32-56, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187011

ABSTRACT

The inefficiency of cyanide/HCN (CN) binding with heme proteins (under physiological regimes) is demonstrated with an assessment of thermodynamics, kinetics, and inhibition constants. The acute onset of toxicity and CN's mg/Kg LD50 (µM lethal concentration) suggests that the classical hemeFe binding-based inhibition rationale is untenable to account for the toxicity of CN. In vitro mechanistic probing of CN-mediated inhibition of hemeFe reductionist systems was explored as a murburn model for mitochondrial oxidative phosphorylation (mOxPhos). The effect of CN in haloperoxidase catalyzed chlorine moiety transfer to small organics was considered as an analogous probe for phosphate group transfer in mOxPhos. Similarly, inclusion of CN in peroxidase-catalase mediated one-electron oxidation of small organics was used to explore electron transfer outcomes in mOxPhos, leading to water formation. The free energy correlations from a Hammett study and IC50/Hill slopes analyses and comparison with ligands ( CO/ H 2 S/ N 3 - ) $\left( {\text{CO}}/{{{{\text{H}}_{2}}\text{S}}/{\text{N}_{3}^{\text{-}}}\;}\; \right)$ provide insights into the involvement of diffusible radicals and proton-equilibriums, explaining analogous outcomes in mOxPhos chemistry. Further, we demonstrate that superoxide (diffusible reactive oxygen species, DROS) enables in vitro ATP synthesis from ADP+phosphate, and show that this reaction is inhibited by CN. Therefore, practically instantaneous CN ion-radical interactions with DROS in matrix catalytically disrupt mOxPhos, explaining the acute lethal effect of CN.


Subject(s)
Cyanides/toxicity , Heme/chemistry , Hemeproteins/antagonists & inhibitors , Hemoglobins/antagonists & inhibitors , Mitochondria/drug effects , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Binding Sites , Catalase/metabolism , Catalysis , Cell Respiration/drug effects , Cell Respiration/physiology , Chloride Peroxidase/chemistry , Cyanides/chemistry , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/metabolism , Heme/antagonists & inhibitors , Heme/metabolism , Hemeproteins/chemistry , Hemeproteins/metabolism , Hemoglobins/chemistry , Horseradish Peroxidase/metabolism , Hydroxides/chemistry , Kinetics , Ligands , Mitochondria/chemistry , Mitochondria/enzymology , Mitochondria/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Styrenes/chemistry , Styrenes/pharmacology , Superoxides/chemistry , Thermodynamics
4.
Biophys Chem ; 258: 106327, 2020 03.
Article in English | MEDLINE | ID: mdl-31927393

ABSTRACT

Serum albumin binds avidly to heme to form heme-serum albumin complex and can protect against the potentially toxic effects of heme. Rutin is a glycoside of the bioflavonoid quercetin with various protective effects due to its antioxidant ability. Clarification of the interaction mechanisms between serum albumin and bioactive components (such as heme and flavonoid) is important to develop effective carriers for encapsulation of heme and suppression of its toxicity. In this study, bindings of bovine serum albumin (BSA) to heme and/or rutin were investigated by experimental and molecular docking techniques. The fluorescence of BSA was quenched by both heme and rutin in static mode (i.e. formation of BSA-monoligand complexes), which was confirmed by Stern-Volmer calculations. Although heme showed higher affinity to BSA than rutin, the interactions of both components with BSA did locate within subdomain IIA (site I). BSA-diligand complexes were successfully formed after the simultaneous addition of heme and rutin. Bioactive rutin in the BSA-diligand complex still kept strong free radical scavenging activity compared to free rutin or BSA-monoligand complex. Hydrogen peroxide (H2O2)-induced heme degradation and free iron release was inhibited upon BSA binding and further decreased in BSA-diligand complexes. Consistently, the cytotoxicity of heme and oxidative stress in endothelial cells was decreased in the BSA-diligand complexes relative to those of heme or BSA-heme complex, where the co-presence of rutin played an important role. These results suggest the possibility and advantage of developing BSA-based carriers for the suppression of heme toxicity in their biomedical applications.


Subject(s)
Antioxidants/pharmacology , Heme/antagonists & inhibitors , Rutin/pharmacology , Serum Albumin, Bovine/metabolism , Animals , Antioxidants/chemistry , Antioxidants/metabolism , Cattle , Heme/toxicity , Ligands , Models, Molecular , Rutin/chemistry , Rutin/metabolism , Serum Albumin, Bovine/chemistry
5.
Angew Chem Int Ed Engl ; 59(10): 4068-4074, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31854064

ABSTRACT

Atherosclerosis (AS) is a major contributor to cardiovascular diseases worldwide, and alleviating inflammation is a promising strategy for AS treatment. Here, we report molecularly engineered M2 macrophage-derived exosomes (M2 Exo) with inflammation-tropism and anti-inflammatory capabilities for AS imaging and therapy. M2 Exo are derived from M2 macrophages and further electroporated with FDA-approved hexyl 5-aminolevulinate hydrochloride (HAL). After systematic administration, the engineered M2 Exo exhibit excellent inflammation-tropism and anti-inflammation effects via the surface-bonded chemokine receptors and the anti-inflammatory cytokines released from the anti-inflammatory M2 macrophages. Moreover, the encapsulated HAL can undergo intrinsic biosynthesis and metabolism of heme to generate anti-inflammatory carbon monoxide and bilirubin, which further enhance the anti-inflammation effects and finally alleviate AS. Meanwhile, the intermediate protoporphyrin IX (PpIX) of the heme biosynthesis pathway permits the fluorescence imaging and tracking of AS.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Atherosclerosis/drug therapy , Heme/antagonists & inhibitors , Inflammation/drug therapy , Macrophages/drug effects , Tropism/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Atherosclerosis/metabolism , Exosomes/drug effects , Exosomes/metabolism , Heme/biosynthesis , Inflammation/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout
6.
Eur J Med Chem ; 187: 111973, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31881453

ABSTRACT

Heme-regulated inhibitor (HRI), a eukaryotic translation initiation factor 2 alpha (eIF2α) kinase, is critically important for coupling protein synthesis to heme availability in reticulocytes and adaptation to various environmental stressors in all cells. HRI modifies the severity of several hemoglobin misfolding disorders including ß-thalassemia. Small molecule activators of HRI are essential for studying normal- and patho-biology of this kinase as well as for the treatment of various human disorders for which activation of HRI or phosphorylation of eIF2α may be beneficial. We previously reported development of 1-((1,4-trans)-4-aryloxycyclohexyl)-3-arylureas (cHAUs) as specific HRI activators and demonstrated their potential as molecular probes for studying HRI biology and as lead compounds for treatment of various human disorders. To develop more druglike cHAUs for in vivo studies and drug development and to expand the chemical space, we undertook bioassay guided structure-activity relationship studies replacing cyclohexyl ring with various 4-6-membered rings and explored further substitutions on the N-phenyl ring. We tested all analogs in the surrogate eIF2α phosphorylation and cell proliferation assays, and a subset of analogs in secondary mechanistic assays that included endogenous eIF2α phosphorylation and expression of C/EBP homologous protein (CHOP), a downstream effector. Finally, we determined specificity of these compounds for HRI by testing their anti-proliferative activity in cells transfected with siRNA targeting HRI or mock. These compounds have significantly improved cLogPs with no loss of potencies, making them excellent candidates for lead optimization for development of investigational new drugs that potently and specifically activate HRI.


Subject(s)
Antineoplastic Agents/pharmacology , Eukaryotic Initiation Factor-2/antagonists & inhibitors , Heme/antagonists & inhibitors , Urea/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Eukaryotic Initiation Factor-2/metabolism , Heme/metabolism , Humans , Models, Molecular , Molecular Structure , Phosphorylation/drug effects , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
7.
J Pept Sci ; 25(9): e3203, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31347248

ABSTRACT

Plasmodium falciparum is the most lethal species of malaria. In infected human red blood cells, P. falciparum digests hemoglobin as a nutrient source, liberating cytotoxic free heme in the process. Sequestration and subsequent conversion of this byproduct into hemozoin, an inert biocrystalline heme aggregate, plays a key role in parasite survival. Hemozoin has been a longstanding target of antimalarials such as chloroquine (CQ), which inhibit the biocrystallization of free heme. In this study, we explore heme-binding interactions with histidine-rich-protein 2 (HRP2), a known malarial biomarker and purported player in free heme sequestration. HRP2 is notoriously challenging to target due to its highly repetitious sequence and irregular secondary structure. We started with three protein-catalyzed capture agents (PCCs) developed against epitopes of HRP2, inclusive of heme-binding motifs, and explored their ability to inhibit heme:HRP2 complex formation. Cocktails of the individual PCCs exhibit an inhibitory potency similar to CQ, while a covalently linked structure built from two separate PCCs provided considerably increased inhibition relative to CQ. Epitope-targeted disruption of heme:HRP2 binding is a novel approach towards disrupting P. falciparum-related hemozoin formation.


Subject(s)
Epitopes/drug effects , Heme/antagonists & inhibitors , Peptides/pharmacology , Protozoan Proteins/antagonists & inhibitors , Amino Acid Sequence , Antigens, Protozoan/genetics , Epitopes/genetics , Heme/genetics , Humans , Molecular Conformation , Peptides/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/genetics
8.
Sci Rep ; 9(1): 8666, 2019 06 17.
Article in English | MEDLINE | ID: mdl-31209282

ABSTRACT

5-aminolevulinic acid (5-ALA) has recently been employed for photodynamic diagnosis (ALA-PDD) and photodynamic therapy (ALA-PDT) of various types of cancer because hyperproliferating tumor cells do not utilize oxidative phosphorylation and do not efficiently produce heme; instead, they accumulate protoporphyrin IX (PpIX), which is a precursor of heme that is activated by violet light irradiation that results in the production of red fluorescence and singlet oxygen. The efficiencies of ALA-PDD and ALA-PDT depend on the efficient cellular uptake of 5-ALA and the inefficient excretion of PpIX. We employed the JFCR39 cell panel to determine whether tumor cells originating from different tissues can produce and accumulate PpIX. We also investigated cellular factors/molecules involved in PpIX excretion by tumor cells with the JFCR39 cell panel. Unexpectedly, the expression levels of ABCG2, which has been considered to play a major role in PpIX extracellular transport, did not show a strong correlation with PpIX excretion levels in the JFCR39 cell panel, although an ABCG2 inhibitor significantly increased intracellular PpIX accumulation in several tumor cell lines. In contrast, the expression levels of dynamin 2, which is a cell membrane-associated molecule involved in exocytosis, were correlated with the PpIX excretion levels. Moreover, inhibitors of dynamin significantly suppressed PpIX excretion and increased the intracellular levels of PpIX. This is the first report demonstrating the causal relationship between dynamin 2 expression and PpIX excretion in tumor cells.


Subject(s)
Aminolevulinic Acid/pharmacology , Dynamin II/metabolism , Exocytosis/drug effects , Mitochondria/drug effects , Photosensitizing Agents/metabolism , Protoporphyrins/metabolism , Cell Line, Tumor , Dynamin II/antagonists & inhibitors , Dynamin II/genetics , Exocytosis/radiation effects , Heme/antagonists & inhibitors , Heme/biosynthesis , Humans , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/radiation effects , Photochemotherapy , Trimethyl Ammonium Compounds/pharmacology , Ultraviolet Rays
9.
Toxicol Sci ; 167(1): 227-238, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30215777

ABSTRACT

Transcription factor Nrf2 protects hepatocytes against various toxicants by upregulating cytoprotective genes. The heme synthesis inhibitor 3, 5-diethoxycarbonyl-1, 4-dihydrocollidine (DDC) leads to liver injury around the portal vein, unlike other groups of toxicants that cause hemorrhage and necrosis in the centrilobular area. To examine whether and how Nrf2 protects livers from the injury, we fed DDC to Nrf2 knockout (Nrf2KO), wild-type (WT), Keap1flox/flox (Keap1-knockdown; Keap1KD), and liver-specific Keap1 knockout (Keap1-Alb) mice, as these lines of mice exhibit stepwise increases in Nrf2 protein expression levels. Liver-specific Keap1::Nrf2 double-knockout (Keap1::Nrf2-Alb) mice were also exploited to examine the contribution of Nrf2. Two weeks after DDC feeding, Keap1-Alb mice were fully recovered from body weight loss, but the WT and Nrf2KO mice were not. The liver-to-body-weight ratio of Keap1-Alb mice was significantly larger than that of WT and Nrf2KO mice. Two indicators of hepatotoxicity, alanine aminotransferase and bilirubin in plasma, were both elevated in WT mice, but downregulated in Keap1-Alb mice after the DDC-feeding. DDC-induced porphyrin accumulation was reduced in the livers of Keap1-Alb and Keap1KD mice compared with that of WT mice. When assessed by the Nqo1 level, Nrf2 expression was further enhanced by DDC in Keap1-Alb mice, suggesting that DDC may have a Keap1 independent potential to activate Nrf2. Genetic activation of Nrf2 in Keap1-Alb mice increased the extracellular excretion of porphyrins, but contrary to our expectation, hepatic damages in Nrf2KO mice appeared to be similar to that of WT mice. Based on these observations, we conclude that Nrf2 activation protects livers against DDC-elicited hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Dicarbethoxydihydrocollidine/toxicity , Heme/antagonists & inhibitors , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Female , Gene Knockdown Techniques , Gene Knockout Techniques , Heme/biosynthesis , Kelch-Like ECH-Associated Protein 1/genetics , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics
10.
Article in English | MEDLINE | ID: mdl-29109165

ABSTRACT

Malaria parasites contain a relict plastid, the apicoplast, which is considered an excellent drug target due to its bacterial-like ancestry. Numerous parasiticidals have been proposed to target the apicoplast, but few have had their actual targets substantiated. Isopentenyl pyrophosphate (IPP) production is the sole required function of the apicoplast in the blood stage of the parasite life cycle, and IPP supplementation rescues parasites from apicoplast-perturbing drugs. Hence, any drug that kills parasites when IPP is supplied in culture must have a nonapicoplast target. Here, we use IPP supplementation to discriminate whether 23 purported apicoplast-targeting drugs are on- or off-target. We demonstrate that a prokaryotic DNA replication inhibitor (ciprofloxacin), several prokaryotic translation inhibitors (chloramphenicol, doxycycline, tetracycline, clindamycin, azithromycin, erythromycin, and clarithromycin), a tRNA synthase inhibitor (mupirocin), and two IPP synthesis pathway inhibitors (fosmidomycin and FR900098) have apicoplast targets. Intriguingly, fosmidomycin and FR900098 leave the apicoplast intact, whereas the others eventually result in apicoplast loss. Actinonin, an inhibitor of bacterial posttranslational modification, does not produce a typical delayed-death response but is rescued with IPP, thereby confirming its apicoplast target. Parasites treated with putative apicoplast fatty acid pathway inhibitors could not be rescued, demonstrating that these drugs have their primary targets outside the apicoplast, which agrees with the dispensability of the apicoplast fatty acid synthesis pathways in the blood stage of malaria parasites. IPP supplementation provides a simple test of whether a compound has a target in the apicoplast and can be used to screen novel compounds for mode of action.


Subject(s)
Antimalarials/pharmacology , Apicoplasts/drug effects , Drug Evaluation, Preclinical/methods , Plasmodium falciparum/cytology , Plasmodium falciparum/drug effects , Apicoplasts/genetics , Azithromycin/pharmacology , Cells, Cultured , Fatty Acids/antagonists & inhibitors , Fatty Acids/biosynthesis , Heme/antagonists & inhibitors , Heme/biosynthesis , Hemiterpenes/pharmacology , Humans , Hydroxamic Acids/pharmacology , Malaria, Falciparum/parasitology , Organophosphorus Compounds/pharmacology , Protozoan Proteins/metabolism
11.
Annu Rev Biochem ; 86: 799-823, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28426241

ABSTRACT

Iron is essential for the survival of most bacteria but presents a significant challenge given its limited bioavailability. Furthermore, the toxicity of iron combined with the need to maintain physiological iron levels within a narrow concentration range requires sophisticated systems to sense, regulate, and transport iron. Most bacteria have evolved mechanisms to chelate and transport ferric iron (Fe3+) via siderophore receptor systems, and pathogenic bacteria have further lowered this barrier by employing mechanisms to utilize the host's hemoproteins. Once internalized, heme is cleaved by both oxidative and nonoxidative mechanisms to release iron. Heme, itself a lipophilic and toxic molecule, presents a significant challenge for transport into the cell. As such, pathogenic bacteria have evolved sophisticated cell surface signaling and transport systems to obtain heme from the host. In this review, we summarize the structure and function of the heme-sensing and transport systems of pathogenic bacteria and the potential of these systems as antimicrobial targets.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Cell Membrane/drug effects , Heme/antagonists & inhibitors , Iron/metabolism , Pseudomonas aeruginosa/drug effects , Receptors, Cell Surface/antagonists & inhibitors , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport/drug effects , Cell Membrane/metabolism , Cell Wall/drug effects , Cell Wall/metabolism , Gene Expression , Heme/metabolism , Metalloporphyrins/chemical synthesis , Metalloporphyrins/pharmacology , Models, Molecular , Protein Conformation , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Siderophores/antagonists & inhibitors , Siderophores/biosynthesis , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism
12.
Article in English | MEDLINE | ID: mdl-27956423

ABSTRACT

A novel 4-aminoquinoline derivative [(S)-7-chloro-N-(4-methyl-1-(4-methylpiperazin-1-yl)pentan-2-yl)-quinolin-4-amine triphosphate] exhibiting curative activity against chloroquine-resistant malaria parasites has been identified for preclinical development as a blood schizonticidal agent. The lead molecule selected after detailed structure-activity relationship (SAR) studies has good solid-state properties and promising activity against in vitro and in vivo experimental malaria models. The in vitro absorption, distribution, metabolism, and excretion (ADME) parameters indicate a favorable drug-like profile.


Subject(s)
Aminoquinolines/chemical synthesis , Antimalarials/chemical synthesis , Malaria/drug therapy , Plasmodium cynomolgi/drug effects , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects , Administration, Oral , Aminoquinolines/pharmacology , Animals , Antimalarials/pharmacology , Chlorocebus aethiops , Chloroquine/pharmacology , Drug Resistance/drug effects , Erythrocytes/drug effects , Erythrocytes/parasitology , Heme/antagonists & inhibitors , Heme/metabolism , Hemin/antagonists & inhibitors , Hemin/biosynthesis , Inhibitory Concentration 50 , Macaca mulatta , Malaria/parasitology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Parasitic Sensitivity Tests , Plasmodium cynomolgi/growth & development , Plasmodium cynomolgi/metabolism , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Plasmodium yoelii/growth & development , Plasmodium yoelii/metabolism , Structure-Activity Relationship , Vero Cells
13.
PLoS One ; 11(8): e0160091, 2016.
Article in English | MEDLINE | ID: mdl-27483471

ABSTRACT

Antimalarial chloroquine (CQ) prevents haematin detoxication when CQ-base concentrates in the acidic digestive vacuole through protonation of its p-aminopyridine (pAP) basic aromatic nitrogen and sidechain diethyl-N. CQ export through the variant vacuolar membrane export channel, PFCRT, causes CQ-resistance in Plasmodium falciparum but 3-methyl CQ (sontochin SC), des-ethyl amodiaquine (DAQ) and bis 4-aminoquinoline piperaquine (PQ) are still active. This is determined by changes in drug accumulation ratios in parasite lipid (LAR) and in vacuolar water (VAR). Higher LAR may facilitate drug binding to and blocking PFCRT and also aid haematin in lipid to bind drug. LAR for CQ is only 8.3; VAR is 143,482. More hydrophobic SC has LAR 143; VAR remains 68,523. Similarly DAQ with a phenol substituent has LAR of 40.8, with VAR 89,366. In PQ, basicity of each pAP is reduced by distal piperazine N, allowing very high LAR of 973,492, retaining VAR of 104,378. In another bis quinoline, dichlorquinazine (DCQ), also active but clinically unsatisfactory, each pAP retains basicity, being insulated by a 2-carbon chain from a proximal nitrogen of the single linking piperazine. While LAR of 15,488 is still high, the lowest estimate of VAR approaches 4.9 million. DCQ may be expected to be very highly lysosomotropic and therefore potentially hepatotoxic. In 11 pAP antimalarials a quadratic relationship between logLAR and logResistance Index (RI) was confirmed, while log (LAR/VAR) vs logRI for 12 was linear. Both might be used to predict the utility of structural modifications.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Hemin/antagonists & inhibitors , Plasmodium falciparum/drug effects , Vacuoles/drug effects , Amodiaquine/analogs & derivatives , Amodiaquine/chemistry , Amodiaquine/metabolism , Amodiaquine/pharmacology , Antimalarials/metabolism , Biological Transport , Chloroquine/analogs & derivatives , Chloroquine/chemistry , Chloroquine/metabolism , Chloroquine/pharmacology , Drug Design , Drug Resistance , Heme/antagonists & inhibitors , Heme/metabolism , Hemin/metabolism , Hydrophobic and Hydrophilic Interactions , Plasmodium falciparum/metabolism , Quinolines/chemistry , Quinolines/metabolism , Quinolines/pharmacology , Structure-Activity Relationship , Vacuoles/metabolism
14.
Dalton Trans ; 45(24): 10058-67, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27273757

ABSTRACT

Recent work by several groups has established that MhuD, IsdG, and IsdI are non-canonical heme oxygenases that induce significant out-of-plane ruffling distortions of their heme substrates enroute to mycobilin or staphylobilin formation. However, clear explanations for the observations of "nested" S = ½ VTVH MCD saturation magnetization curves at cryogenic temperatures, and exchange broadened (1)H NMR resonances at physiologically-relevant temperatures have remained elusive. Here, MCD and NMR data have been acquired for F23A and F23W MhuD-heme-CN, in addition to MCD data for IsdI-heme-CN, in order to complete assembly of a library of spectroscopic data for cyanide-inhibited ferric heme with a wide range of ruffling deformations. The spectroscopic data were used to evaluate a number of computational models for cyanide-inhibited ferric heme, which ultimately led to the development of an accurate NEVPT2/CASSCF model. The resulting model has a shallow, double-well potential along the porphyrin ruffling coordinate, which provides clear explanations for the unusual MCD and NMR data. The shallow, double-well potential also implies that MhuD-, IsdG-, and IsdI-bound heme is dynamic, and the functional implications of these dynamics are discussed.


Subject(s)
Bacterial Proteins/chemistry , Cyanides/chemistry , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/chemistry , Heme/antagonists & inhibitors , Heme/chemistry , Mixed Function Oxygenases/chemistry , Oxygenases/chemistry , Computational Biology , Crystallography, X-Ray , Mycobacterium tuberculosis/enzymology , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Temperature
15.
Toxicol Sci ; 151(1): 150-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26880746

ABSTRACT

Cytochrome P450 (CYP) enzymes mediate mixed-function oxidation reactions important in drug metabolism. The aromatic heterocyclic cation, diphenyleneiodonium (DPI), binds flavin in cytochrome P450 reductase and inhibits CYP-mediated activity. DPI also inhibits CYP by directly interacting with heme. Herein, we report that DPI effectively inhibits a number of CYP-related monooxygenase reactions including NADPH oxidase, a microsomal enzyme activity that generates hydrogen peroxide in the absence of metabolizing substrates. Inhibition of monooxygenase by DPI was time and concentration dependent with IC50's ranging from 0.06 to 1.9 µM. Higher (4.6-23.9 µM), but not lower (0.06-1.9 µM), concentrations of DPI inhibited electron flow via cytochrome P450 reductase, as measured by its ability to reduce cytochrome c and mediate quinone redox cycling. Similar results were observed with inducible nitric oxide synthase (iNOS), an enzyme containing a C-terminal reductase domain homologous to cytochrome P450 reductase that mediates reduction of cytochrome c, and an N-terminal heme-thiolate oxygenase domain mediating nitric oxide production. Significantly greater concentrations of DPI were required to inhibit cytochrome c reduction by iNOS (IC50 = 3.5 µM) than nitric oxide production (IC50 = 0.16 µM). Difference spectra of liver microsomes, recombinant CYPs, and iNOS demonstrated that DPI altered heme-carbon monoxide interactions. In the presence of NADPH, DPI treatment of microsomes and iNOS yielded a type II spectral shift. These data indicate that DPI interacts with both flavin and heme in CYPs and iNOS. Increased sensitivity for inhibition of CYP-mediated metabolism and nitric oxide production by iNOS indicates that DPI targets heme moieties within the enzymes.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Heme/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Onium Compounds/pharmacology , Animals , Dose-Response Relationship, Drug , Heme/metabolism , Humans , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Time Factors
16.
J Antimicrob Chemother ; 71(4): 946-52, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26747101

ABSTRACT

OBJECTIVES: During recent decades, the number of invasive fungal infections among immunosuppressed patients has increased significantly, whereas the number of effective systemic antifungal drugs remains low and unsatisfactory. The aim of this study was to characterize a novel antifungal compound, CW-8/haemofungin, which we previously identified in a screen for compounds affecting fungal cell wall integrity. METHODS: The in vitro characteristics of haemofungin were investigated by MIC evaluation against a panel of pathogenic and non-pathogenic fungi, bacteria and mammalian cells in culture. Haemofungin mode-of-action studies were performed by screening an Aspergillus nidulans overexpression genomic library for resistance-conferring plasmids and biochemical validation of the target. In vivo efficacy was tested in the Galleria mellonella and Drosophila melanogaster insect models of infection. RESULTS: We demonstrate that haemofungin causes swelling and lysis of growing fungal cells. It inhibits the growth of pathogenic Aspergillus, Candida, Fusarium and Rhizopus isolates at micromolar concentrations, while only weakly affecting the growth of mammalian cell lines. Genetic and biochemical analyses in A. nidulans and Aspergillus fumigatus indicate that haemofungin primarily inhibits ferrochelatase (HemH), the last enzyme in the haem biosynthetic pathway. Haemofungin was non-toxic and significantly reduced mortality rates of G. mellonella and D. melanogaster infected with A. fumigatus and Rhizopus oryzae, respectively. CONCLUSIONS: Further development and in vivo validation of haemofungin is warranted.


Subject(s)
Antifungal Agents/pharmacology , Heme/antagonists & inhibitors , Heme/biosynthesis , Heterocyclic Compounds, 4 or More Rings/pharmacology , Animals , Aspergillus nidulans/drug effects , Aspergillus nidulans/genetics , Bacteria/drug effects , Bacteria/growth & development , Cell Line , Drug Resistance, Fungal , Drug Synergism , Ferrochelatase/antagonists & inhibitors , Fungi/drug effects , Fungi/growth & development , Humans , Insecta , Microbial Sensitivity Tests , Mycoses/drug therapy , Mycoses/microbiology , Protoporphyrins/biosynthesis
17.
J Mol Recognit ; 29(5): 186-98, 2016 May.
Article in English | MEDLINE | ID: mdl-26686319

ABSTRACT

In invertebrates, the prophenoloxidase (proPO) pathway is involved in the phenol-like antioxidant production against invading pathogens. Overproduction of melanin and phenolic substances leads to the disruption of hemocytes (own host cells); therefore, there is a prerequisite to regulate the antioxidant production, which is performed by the proteases and proPO-associated cell adhesion protein peroxinectin (PX). PX is a macromolecular structure consisting of protein involved in the proPO pathway, which is a potential target in the regulatory mechanism in crustaceans. In the proPO cascade, pattern recognition proteins initiate the proPO cascade by the consequent reaction, and PX is involved in the key step in the regulatory mechanism of phenoloxidase enzyme synthesis. In the present study, Indian white shrimp Fenneropenaeus indicus PX (Fein-PX) gene sequence was used. Upregulation of Fein-PX was determined using immunostimulants ß-glucan (agonists) and examined its expression by quantitative RT-PCR. To find the downregulation or negative regulation of Fein-PX, inhibitors were screened, and its 3D model provides molecular insights into the rationale inhibitor design for developing an effective molecule against Fein-PX.


Subject(s)
Cell Adhesion Molecules/genetics , Gene Expression Regulation/drug effects , Heme/antagonists & inhibitors , Penaeidae/metabolism , beta-Glucans/pharmacology , Animals , Catechol Oxidase/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Cloning, Molecular , Computer Simulation , Enzyme Precursors/metabolism , In Vitro Techniques , Molecular Docking Simulation , Molecular Dynamics Simulation , Penaeidae/chemistry , Penaeidae/genetics , Phylogeny , Structure-Activity Relationship
18.
Eur J Haematol ; 96(4): 367-74, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26058344

ABSTRACT

Diamond-Blackfan anemia (DBA) is a congenital pure red cell aplasia often associated with skeletal malformations. Mutations in ribosomal protein coding genes, mainly in RPS19, account for the majority of DBA cases. The molecular mechanisms underlying DBA pathogenesis are still not completely understood. Alternative spliced isoforms of FLVCR1 (feline leukemia virus subgroup C receptor 1) transcript coding for non-functional proteins have been reported in some DBA patients. Consistently, a phenotype very close to DBA has been described in animal models of FLVCR1 deficiency. FLVCR1 gene codes for two proteins: the plasma membrane heme exporter FLVCR1a and the mitochondrial heme exporter FLVCR1b. The coordinated expression of both FLVCR1 isoforms regulates an intracellular heme pool, necessary for proper expansion and differentiation of erythroid precursors. Here, we investigate the role of FLVCR1 isoforms in a cellular model of DBA. RPS19-downregulated TF1 cells show reduced FLVCR1a and FLVCR1b mRNA levels associated with heme overload. The downregulation of FLVCR1 isoforms affects cell cycle progression and apoptosis in differentiating K562 cells, a phenotype similar to DBA. Taken together, these data suggest that alteration of heme metabolism could play a role in the pathogenesis of DBA.


Subject(s)
Gene Expression Regulation, Leukemic , Heme/biosynthesis , Membrane Transport Proteins/genetics , RNA, Messenger/genetics , Receptors, Virus/genetics , Ribosomal Proteins/genetics , Alternative Splicing , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/metabolism , Anemia, Diamond-Blackfan/pathology , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Membrane/metabolism , Heme/agonists , Heme/antagonists & inhibitors , Humans , K562 Cells , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Models, Biological , Mutation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/metabolism
19.
Free Radic Biol Med ; 85: 207-18, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25933588

ABSTRACT

Transfusion of stored red blood cells (RBCs) is associated with increased morbidity and mortality in trauma patients. Pro-oxidant, pro-inflammatory, and nitric oxide (NO) scavenging properties of stored RBCs are thought to underlie this association. In this study we determined the effects of RBC washing and nitrite and antiheme therapy on stored RBC-dependent toxicity in the setting of trauma-induced hemorrhage. A murine (C57BL/6) model of trauma-hemorrhage and resuscitation with 1 or 3 units of RBCs stored for 0-10 days was used. Tested variables included washing RBCs to remove lower MW components that scavenge NO, NO-repletion therapy using nitrite, or mitigation of free heme toxicity by heme scavenging or preventing TLR4 activation. Stored RBC toxicity was determined by assessment of acute lung injury indices (airway edema and inflammation) and survival. Transfusion with 5 day RBCs increased acute lung injury indexed by BAL protein and neutrophil accumulation. Washing 5 day RBCs prior to transfusion did not decrease this injury, whereas nitrite therapy did. Transfusion with 10 day RBCs elicited a more severe injury resulting in ~90% lethality, compared to <15% with 5 day RBCs. Both washing and nitrite therapy significantly protected against 10 day RBC-induced lethality, suggesting that washing may be protective when the injury stimulus is more severe. Finally, a spectral deconvolution assay was developed to simultaneously measure free heme and hemoglobin in stored RBC supernatants, which demonstrated significant increases of both in stored human and mouse RBCs. Transfusion with free heme partially recapitulated the toxicity mediated by stored RBCs. Furthermore, inhibition of TLR4 signaling, which is stimulated by heme, using TAK-242, or hemopexin-dependent sequestration of free heme significantly protected against both 5 day and 10 day mouse RBC-dependent toxicity. These data suggest that RBC washing, nitrite therapy, and/or antiheme and TLR4 strategies may prevent stored RBC toxicities.


Subject(s)
Erythrocytes/cytology , Heme/antagonists & inhibitors , Hemorrhage/therapy , Nitrites/administration & dosage , Wounds and Injuries/therapy , Animals , Humans , Mice , Mice, Inbred C57BL
20.
Biochem Pharmacol ; 96(3): 159-67, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25986884

ABSTRACT

Tuberculosis is still a major health problem worldwide and one of the main causes of death by a single infectious agent. Only few drugs are really effective to treat tuberculosis, hence, the emergence of multiple, extensively, and totally drug resistant bacilli compromises the already difficult antituberculosis treatments. Given the persistent global burden of tuberculosis, it is crucial to understand the underlying mechanisms required for the pathogenicity of Mycobacterium tuberculosis (Mtb), the causal agent of tuberculosis, in order to pave the way for developing better drugs and strategies to treat and prevent tuberculosis. The exclusive mycobacterial cell wall lipids such as trehalose monomycolate and dimycolate (TMM, TDM), phthiocerol dimycocerosate (PDIM), sulpholipid-1 (SL-1), diacyl trehalose (DAT), and pentacyl trehalose (PAT), among others, are known to play an important role in pathogenesis; thus, proteins responsible for their transport are potential virulence factors. MmpL and MmpS proteins mediate transport of important cell wall lipids across the mycobacterial membrane. In Mtb, MmpL3, MmpL7 and MmpL8 transport TMM, PDIM and SL-1 respectively. The translocation of DAT and biosynthesis of PAT is likely due to MmpL10. MmpL and MmpS proteins are involved in other processes such as drug efflux (MmpL5 and MmpL7), siderophore export (MmpL4/MmpS4 and MmpL5/MmpS5), and heme uptake (MmpL3 and MmpL11). Altogether, these proteins can be regarded as new potential targets for antituberculosis drug development. We will review recent advances in developing inhibitors of MmpL proteins, in the challenging context of targeting membrane proteins and the future prospects for potential antituberculosis drug candidates.


Subject(s)
Antitubercular Agents/pharmacology , Cell Wall/drug effects , Drug Design , Lipid Metabolism/drug effects , Mycobacterium tuberculosis/drug effects , Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport/drug effects , Cell Wall/chemistry , Cell Wall/metabolism , Cord Factors/metabolism , Extensively Drug-Resistant Tuberculosis/drug therapy , Extensively Drug-Resistant Tuberculosis/microbiology , Extensively Drug-Resistant Tuberculosis/pathology , Gene Expression , Glycolipids/metabolism , Heme/antagonists & inhibitors , Heme/metabolism , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Virulence , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...