Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cell Mol Biol (Noisy-le-grand) ; 67(4): 106-114, 2022 Jan 02.
Article in English | MEDLINE | ID: mdl-35809297

ABSTRACT

Alzheimer's disease is a chronic neurodegenerative ailment and the most familiar type of dementia in the older population with no effective cure to date. It is characterized by a decrease in memory, associated with the mutilation of cholinergic neurotransmission. Presently, acetylcholinesterase inhibitors have emerged as the most endorsed pharmacological medications for the symptomatic treatment of mild to moderate Alzheimer's disease. This study aimed to research the molecular enzymatic inhibition of human brain acetylcholinesterase by a natural compound emetine and I3M. Molecular docking studies were used to identify superior interaction between enzyme acetylcholinesterase and ligands. Furthermore, the docked acetylcholinesterase-emetine complex was validated statistically using an analysis of variance in all tested conformers. In this interaction, H-bond, hydrophobic interaction, pi-pi, and Cation-pi interactions played a vital function in predicting the accurate conformation of the ligand that binds with the active site of acetylcholinesterase. The conformer with the lowest free energy of binding was further analyzed. The binding energy for acetylcholinesterase complex with emetine and I3M was -9.72kcal/mol and -7.09kcal/mol, respectively. In the current study, the prediction was studied to establish a relationship between binding energy and intermolecular energy (coefficient of determination [R2 linear = 0.999), and intermolecular energy and Van der wall forces (R2 linear = 0.994). These results would be useful in gaining structural insight for designing novel lead compounds against acetylcholinesterase for the effective management of Alzheimer's disease.


Subject(s)
Acetylcholinesterase , Alzheimer Disease , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Binding Sites , Brain/metabolism , Catalytic Domain , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Emetine/chemistry , Emetine/metabolism , Humans , Indoles , Ligands , Molecular Docking Simulation
2.
J Cell Mol Med ; 24(13): 7427-7438, 2020 07.
Article in English | MEDLINE | ID: mdl-32558259

ABSTRACT

Gain- or loss-of-function mutations in Janus kinase 3 (JAK3) contribute to the pathogenesis of various haematopoietic malignancies and immune disorders, suggesting that aberrant JAK3 signalling is an attractive therapeutic target to treat these disorders. In this study, we performed structure-based computational database screening using the 3D structure of the JAK3 kinase domain and the National Cancer Institute diversity set and identified tubulosine as a novel JAK3 inhibitor. Tubulosine directly blocked the catalytic activity of JAK3 by selective interacting with the JAK3 kinase domain. Consistently, tubulosine potently inhibited persistently activated and interleukin-2-dependent JAK3, and JAK3-mediated downstream targets. Importantly, it did not affect the activity of other JAK family members, particularly prolactin-induced JAK2/signal transducer and activator of transcription 5 and interferon alpha-induced JAK1-TYK2/STAT1. Tubulosine specifically decreased survival and proliferation of cancer cells, in which persistently active JAK3 is expressed, by inducing apoptotic and necrotic/autophagic cell death without affecting other oncogenic signalling. Collectively, tubulosine is a potential small-molecule compound that selectively inhibits JAK3 activity, suggesting that it may serve as a promising therapeutic candidate for treating disorders caused by aberrant activation of JAK3 signalling.


Subject(s)
Adenosine Triphosphate/metabolism , Emetine/analogs & derivatives , Janus Kinase 3/antagonists & inhibitors , Signal Transduction , Apoptosis/drug effects , Autophagy/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Emetine/chemistry , Emetine/pharmacology , Humans , Janus Kinase 3/metabolism , Models, Biological , Necrosis , Oncogenes , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects
3.
J Nat Prod ; 83(5): 1571-1576, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32323990

ABSTRACT

The remarkable biological activity of the dolastatin 10 structural modifications quinstatins and isoquinstatins prompted further investigation into drug hybrids containing biologically active isoquinoline moieties. In this study, the isoquinoline alkaloid emetine was selected as one of the structural domains of a hybrid molecule. That was accomplished by covalently bonding the Dov-Val-Dil-Dap peptide sequence of dolastatin 10 peptide at the N-2' secondary amine of emetine. Three new hybrids were synthesized, 5, 9, and 10. Comparison of the biological activity of these new peptide-emetine analogues with emetine showed complete retention of activity for 5 and a 10-fold decrease for hybrids 9 and 10. The result was surprising, as the activity of emetine is usually lost or greatly reduced when substituted at the N-2' position.


Subject(s)
Aminobenzoates/chemistry , Antineoplastic Agents/pharmacology , Depsipeptides/pharmacology , Emetine/chemistry , Oligopeptides/chemistry , Depsipeptides/chemistry , Emetine/analogs & derivatives , Molecular Structure , Structure-Activity Relationship
4.
Article in English | MEDLINE | ID: mdl-31964796

ABSTRACT

Drug repositioning offers an effective alternative to de novo drug design to tackle the urgent need for novel antimalarial treatments. The antiamoebic compound emetine dihydrochloride has been identified as a potent in vitro inhibitor of the multidrug-resistant strain K1 of Plasmodium falciparum (50% inhibitory concentration [IC50], 47 nM ± 2.1 nM [mean ± standard deviation]). Dehydroemetine, a synthetic analogue of emetine dihydrochloride, has been reported to have less-cardiotoxic effects than emetine. The structures of two diastereomers of dehydroemetine were modeled on the published emetine binding site on the cryo-electron microscopy (cryo-EM) structure with PDB code 3J7A (P. falciparum 80S ribosome in complex with emetine), and it was found that (-)-R,S-dehydroemetine mimicked the bound pose of emetine more closely than did (-)-S,S-dehydroisoemetine. (-)-R,S-dehydroemetine (IC50 71.03 ± 6.1 nM) was also found to be highly potent against the multidrug-resistant K1 strain of P. falciparum compared with (-)-S,S-dehydroisoemetine (IC50, 2.07 ± 0.26 µM), which loses its potency due to the change of configuration at C-1'. In addition to its effect on the asexual erythrocytic stages of P. falciparum, the compound exhibited gametocidal properties with no cross-resistance against any of the multidrug-resistant strains tested. Drug interaction studies showed (-)-R,S-dehydroemetine to have synergistic antimalarial activity with atovaquone and proguanil. Emetine dihydrochloride and (-)-R,S-dehydroemetine failed to show any inhibition of the hERG potassium channel and displayed activity affecting the mitochondrial membrane potential, indicating a possible multimodal mechanism of action.


Subject(s)
Antimalarials/pharmacology , Drug Repositioning , Emetine/analogs & derivatives , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Antimalarials/adverse effects , Atovaquone/pharmacology , Cell Line, Tumor , Drug Resistance, Multiple/genetics , Drug Synergism , Emetine/adverse effects , Emetine/chemistry , Emetine/pharmacology , Female , Hep G2 Cells , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Plasmodium falciparum/genetics , Proguanil/pharmacology , Stereoisomerism
5.
Antiviral Res ; 173: 104650, 2020 01.
Article in English | MEDLINE | ID: mdl-31734270

ABSTRACT

IRES-driven translation plays an essential role in picornavirus infection. However, there are seldom reports of compounds targeting this pathway with effective protection in animal models. Here, we identified emetine, an antiprotozoal drug, which inhibits EV-A71 with an EC50 value of 0.04 µM and a CC50 value of 10 µM in RD cell culture. Interestingly, emetine exhibits activities against a series of human enteroviruses, including CV-A16, CV-B1, EV-D68, Echov-6, etc., at the nanomolar level. When orally administered at 0.20 mg/kg twice a day in an EV-A71 mouse model, emetine reduced viral loads in various organs and completely prevented diseases and death. A mechanistic study demonstrated that emetine suppressed EV-A71 by inhibiting viral IRES-driven translation. Taken together, these data indicate emetine as a promising candidate to treat picornavirus infection.


Subject(s)
Antiviral Agents/pharmacology , Emetine/pharmacology , Enterovirus Infections/virology , Enterovirus/drug effects , Enterovirus/physiology , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Animals , Antiviral Agents/chemistry , Cell Line , Chlorocebus aethiops , Dose-Response Relationship, Drug , Drug Repositioning , Emetine/chemistry , Enterovirus A, Human , Enterovirus Infections/drug therapy , Humans , Mice , Protein Synthesis Inhibitors/chemistry , Vero Cells
6.
Assay Drug Dev Technol ; 17(7): 298-309, 2019 10.
Article in English | MEDLINE | ID: mdl-31634019

ABSTRACT

Multidrug resistance (MDR) is a major health issue for the treatment of infectious diseases throughout the world. Staphylococcus aureus (S. aureus) is a Gram-positive bacteria, responsible for various local and systemic infections in humans. The continuous and abrupt use of antibiotics against bacteria such as S. aureus results in the development of resistant strains. Presently, mupirocin (MUP) is the drug of choice against S. aureus and MDR (methicillin-resistant). However, S. aureus has acquired resistance against MUP as well due to isoleucyl-tRNA synthetase (IleS) mutation at sites 588 and 631. Thus, the aim of the present study was to discover novel bioactives against MUP-resistant S. aureus using in silico drug repurposing approaches. In silico drug repurposing techniques were used to obtain suitable bioactive lead molecules such as buclizine, tasosartan, emetine, medrysone, and so on. These lead molecules might be able to resolve this issue. These leads were obtained through molecular docking simulation based virtual screening, which could be promising for the treatment of MUP-resistant S. aureus. The findings of the present work need to be validated further through in vitro and in vivo studies for their clinical application.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Repositioning , Drug Resistance, Bacterial/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Molecular Docking Simulation , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Emetine/chemical synthesis , Emetine/chemistry , Emetine/pharmacology , Humans , Isoleucine-tRNA Ligase/antagonists & inhibitors , Isoleucine-tRNA Ligase/metabolism , Methicillin-Resistant Staphylococcus aureus/enzymology , Microbial Sensitivity Tests , Piperazines/chemical synthesis , Piperazines/chemistry , Piperazines/pharmacology , Pregnenediones/chemical synthesis , Pregnenediones/chemistry , Pregnenediones/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Tetrazoles/chemical synthesis , Tetrazoles/chemistry , Tetrazoles/pharmacology
7.
Bioorg Med Chem ; 25(24): 6707-6717, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29153549

ABSTRACT

Emetine is a small molecule protein synthesis inhibitor that is toxic to all cell types and therefore suitable for complete killing of all types of heterogeneous cancer cells within a tumor. It becomes significantly inactive (non-toxic) when derivatized at its N-2' secondary amine. This provides a strategy for targeting emetine to cancerous tumor without killing normal cells. In this report, PSA activatable peptide prodrugs of emetine were synthesized. To overcome steric hindrances and enhance protease specific cleavage, a 2-stage prodrug activation process was needed to release emetine in cancer cells. In this 2-stage process, emetine prodrug intermediates are coupled to PSA peptide substrate (Ac-His-Ser-Ser-Lys-Leu-Gln) to obtain the full prodrug. Both prodrug intermediates 10 (Ala-Pro-PABC-Emetine) and 14 (Ser-Leu-PABC-Emetine) were evaluated for kinetics of hydrolysis to emetine and potency [Where PABC = p-aminobenzyloxycarbonyl]. While both intermediates quantitatively liberate emetine when incubated under appropriate conditions, upon coupling of PSA substrate to give the full prodrugs, only prodrug 16, the prodrug obtained from 14 was hydrolyzable by PSA. Cytotoxicity studies in PSA producing LNCaP and CWR22Rv1 confirm the activation of the prodrug by PSA with an IC50 of 75 nM and 59 nM respectively. The cytotoxicity of 16 is significantly reduced in cell lines that do not produce PSA. Further, in vivo toxicity studies are done on these prodrugs and other derivatives of emetine. The results show the significance of conformational modulation in obtaining safe emetine prodrugs.


Subject(s)
Antineoplastic Agents/pharmacology , Emetine/pharmacology , Prodrugs/pharmacology , Prostate-Specific Antigen/antagonists & inhibitors , Proteolysis/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Emetine/chemical synthesis , Emetine/chemistry , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prostate-Specific Antigen/metabolism , Software , Structure-Activity Relationship
8.
Electrophoresis ; 37(9): 1161-5, 2016 05.
Article in English | MEDLINE | ID: mdl-27135308

ABSTRACT

The influence of sample matrix on sample sweeping in MEKC was examined in the presented manuscript. Significant focusing effect was observed for relatively hydrophobic cationic compounds (emetine, strychnine and quinine) using high ionic strength sample matrix (900 mM H3 PO4 /720 mM Tris) which conductivity was about ninefold higher than utilized BGE. Moreover, the results were obtained using BGE composed of comparatively low surfactant concentration (10 mM SDS) and 40 mM H3 PO4 /32 mM Tris buffer solution. About 200 to 300-fold preconcentration of analytes was reached with the presented method. Basing on experimental results and computer simulation using Simul5 software, hypothetical mechanism of observed phenomenon was proposed.


Subject(s)
Chromatography, Micellar Electrokinetic Capillary/methods , Surface-Active Agents/chemistry , Computer Simulation , Emetine/analysis , Emetine/chemistry , Emetine/isolation & purification , Hydrophobic and Hydrophilic Interactions , Models, Chemical , Quinine/analysis , Quinine/chemistry , Quinine/isolation & purification , Strychnine/analysis , Strychnine/chemistry , Strychnine/isolation & purification
9.
J Struct Biol ; 195(1): 72-81, 2016 07.
Article in English | MEDLINE | ID: mdl-27129418

ABSTRACT

The problem of image restoration in cryo-EM entails correcting for the effects of the Contrast Transfer Function (CTF) and noise. Popular methods for image restoration include 'phase flipping', which corrects only for the Fourier phases but not amplitudes, and Wiener filtering, which requires the spectral signal to noise ratio. We propose a new image restoration method which we call 'Covariance Wiener Filtering' (CWF). In CWF, the covariance matrix of the projection images is used within the classical Wiener filtering framework for solving the image restoration deconvolution problem. Our estimation procedure for the covariance matrix is new and successfully corrects for the CTF. We demonstrate the efficacy of CWF by applying it to restore both simulated and experimental cryo-EM images. Results with experimental datasets demonstrate that CWF provides a good way to evaluate the particle images and to see what the dataset contains even without 2D classification and averaging.


Subject(s)
Algorithms , Cryoelectron Microscopy/methods , Image Processing, Computer-Assisted/methods , Macromolecular Substances/chemistry , Animals , Bacteria/ultrastructure , Computer Simulation , Emetine/chemistry , Fourier Analysis , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Particle Size , Ribosomes/chemistry , Signal-To-Noise Ratio , TRPV Cation Channels/chemistry
10.
Comput Biol Chem ; 62: 145-54, 2016 06.
Article in English | MEDLINE | ID: mdl-26278973

ABSTRACT

Cancer is responsible for millions of deaths throughout the world every year. Increased understanding as well as advancements in the therapeutic aspect seems suboptimal to restrict the huge deaths associated with cancer. The major cause responsible for this is high resistance as well as relapse rate associated with cancers. Several evidences indicated that cancer stem cells (CSC) are mainly responsible for the resistance and relapses associated with cancer. Furthermore, agents targeting a single protein seem to have higher chances of resistance than multitargeting drugs. According to the concept of network model, partial inhibition of multiple targets is more productive than single hit agents. Thus, by fusing both the premises that CSC and single hit anticancer drugs, both are responsible for cancer related resistances and screened alkaloids for the search of leads having CSC targeting ability as well as the capability to modulating multiple target proteins. The in silico experimental data indicated that emetine and cortistatin have the ability to modulate hedgehog (Hh) pathway by binding to sonic hedgehog (Hh), smoothened (Smo) and Gli protein, involved in maintenance CSCs. Furthermore, solamargine, solasonine and tylophorine are also seems to be good lead molecules targeting towards CSCs by modulating Hh pathway. Except solamargine and solasonine, other best lead molecules also showed acceptable in silico ADME profile. The predicted lead molecules can be suitably modified to get multitargeting CSC targeting agent to get rid of associate resistances.


Subject(s)
Alkaloids/chemistry , Alkaloids/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cell- and Tissue-Based Therapy , Hedgehog Proteins , Molecular Docking Simulation , Alkaloids/therapeutic use , Drug Delivery Systems , Drug Screening Assays, Antitumor , Emetine/chemistry , Emetine/metabolism , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/chemistry , Humans , Models, Molecular , Neoplasms/therapy , Neoplastic Stem Cells , Neuropeptides/chemistry , Neuropeptides/metabolism , Signal Transduction
11.
Bioorg Med Chem ; 23(17): 5839-45, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26187015

ABSTRACT

A small library of emetine dithiocarbamate ester derivatives were synthesized in 25-86% yield via derivatization of the N2'- position of emetine. Anticancer evaluation of these compounds in androgen receptor positive LNCaP and androgen receptor negative PC3 and DU145 prostate cancer cell lines revealed time dependent and dose-dependent cytotoxicity. With the exception of compound 4c, all the dithiocarbamate ester analogs in this study showed appreciable potency in all the prostate cancer cell lines (regardless of whether it is androgen receptor positive or negative) with a cytotoxicity IC50 value ranging from 1.312 ± 0.032 µM to 5.201 ± 0.125 µM by day 7 of treatment. Compared to the sodium dithiocarbamate salt 1, all the dithiocarbamate ester analogs (2 and 4a-4 g) displayed lower cytotoxicity than compound 1 (PC3, IC50 = 0.087 ± 0.005 µM; DU145, IC50 = 0.079 ± 0.003 µM and LNCaP, IC50 = 0.079 ± 0.003 µM) on day 7 of treatment. Consequently, it appears that S-alkylation of compound 1 leads to a more stable dithiocarbamate ester derivative that resulted in lower anticancer activity in the prostate cancer cell lines.


Subject(s)
Emetine/chemistry , Emetine/chemical synthesis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Drug Design , Humans , Male , Structure-Activity Relationship
12.
Molecules ; 20(6): 11474-89, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26111177

ABSTRACT

Ipecac alkaloids are secondary metabolites produced in the medicinal plant Psychotria ipecacuanha. Emetine is the main alkaloid of ipecac and one of the active compounds in syrup of Ipecac with emetic property. Here we evaluated emetine's potential as an antiviral agent against Human Immunodeficiency Virus. We performed in vitro Reverse Transcriptase (RT) Assay and Natural Endogenous Reverse Transcriptase Activity Assay (NERT) to evaluate HIV RT inhibition. Emetine molecular docking on HIV-1 RT was also analyzed. Phenotypic assays were performed in non-lymphocytic and in Peripheral Blood Mononuclear Cells (PBMC) with HIV-1 wild-type and HIV-harboring RT-resistant mutation to Nucleoside Reverse Transcriptase Inhibitors (M184V). Our results showed that HIV-1 RT was blocked in the presence of emetine in both models: in vitro reactions with isolated HIV-1 RT and intravirion, measured by NERT. Emetine revealed a strong potential of inhibiting HIV-1 replication in both cellular models, reaching 80% of reduction in HIV-1 infection, with low cytotoxic effect. Emetine also blocked HIV-1 infection of RT M184V mutant. These results suggest that emetine is able to penetrate in intact HIV particles, and bind and block reverse transcription reaction, suggesting that it can be used as anti-HIV microbicide. Taken together, our findings provide additional pharmacological information on the potential therapeutic effects of emetine.


Subject(s)
Alkaloids/administration & dosage , Emetine/administration & dosage , HIV Infections/drug therapy , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Alkaloids/chemistry , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/chemistry , Emetine/chemistry , HIV Infections/enzymology , HIV Infections/virology , HIV Reverse Transcriptase/chemistry , HIV-1/pathogenicity , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Mutation , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/chemistry , Virus Replication/drug effects
13.
PLoS One ; 10(5): e0127011, 2015.
Article in English | MEDLINE | ID: mdl-25996685

ABSTRACT

Small molecule based therapeutic intervention of amyloids has been limited by their low solubility and poor pharmacokinetic characteristics. We report here, the use of water soluble poly lactic-co-glycolic acid (PLGA)-encapsulated curcumin and emetine nanoparticles (Cm-NPs and Em-NPs, respectively), as potential modulators of gelsolin amyloidogenesis. Using the amyloid-specific dye Thioflavin T (ThT) as an indicator along with electron microscopic imaging we show that the presence of Cm-NPs augmented amyloid formation in gelsolin by skipping the pre-fibrillar assemblies, while Em-NPs induced non-fibrillar aggregates. These two types of aggregates differed in their morphologies, surface hydrophobicity and secondary structural signatures, confirming that they followed distinct pathways. In spite of differences, both these aggregates displayed reduced toxicity against SH-SY5Y human neuroblastoma cells as compared to control gelsolin amyloids. We conclude that the cytotoxicity of gelsolin amyloids can be reduced by either stalling or accelerating its fibrillation process. In addition, Cm-NPs increased the fibrillar bulk while Em-NPs defibrillated the pre-formed gelsolin amyloids. Moreover, amyloid modulation happened at a much lower concentration and at a faster rate by the PLGA encapsulated compounds as compared to their free forms. Thus, besides improving pharmacokinetic and biocompatible properties of curcumin and emetine, PLGA conjugation elevates the therapeutic potential of both small molecules against amyloid fibrillation and toxicity.


Subject(s)
Amyloidogenic Proteins/metabolism , Curcumin/administration & dosage , Emetine/administration & dosage , Gelsolin/metabolism , Lactic Acid , Nanoparticles , Polyglycolic Acid , Amino Acid Sequence , Amyloidogenic Proteins/chemistry , Cell Line , Curcumin/chemistry , Emetine/chemistry , Gelsolin/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Lactic Acid/chemistry , Models, Molecular , Molecular Sequence Data , Nanoparticles/chemistry , Particle Size , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Protein Aggregation, Pathological , Protein Conformation
14.
ACS Chem Neurosci ; 5(10): 982-92, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25118567

ABSTRACT

The pathogenesis of protein misfolding diseases is attributed to the cytotoxicity caused by amyloidogenic prefibrillar aggregates, rather than mature fibrils. The presence of one or more amyloidogenic stretches in different proteins has been proven critical for initiating fibril formation. In the present study, we show that two natural compounds, curcumin and emetine, bind tightly (Kd < 1.6 µM) to the core amyloidogenic stretch (182-192) of gelsolin (AGel). Binding happens in different structural orientations, distinctly modulating the amyloidogenic pathway of AGel. While AGel alone undergoes sigmoidal transition to thioflavin T (ThT)-responsive fibrillar aggregates with clear lag phase, the presence of curcumin or emetine abolishes the lag phase and produces starkly different, noncytotoxic end products. Atomic force microscopy revealed that while curcumin augments fibril formation, emetine arrests it at an intermediate aggregated stage with no fibrillar morphology. FTIR spectroscopy, dynamic light scattering, and ANS fluorescence experiments also suggest that these two species are distinct. Curcumin and emetine also differentially affect the preformed amyloids with the former thickening the fibrils and the latter releasing reclusive oligomers. MD simulations further provided mechanistic insights of differential interaction by the two compounds modulating amyloid formation. The results were also confirmed on the disease-associated amyloidogenic fragment of gelsolin (fAGel). Thus, our findings suggest that targeting amyloidogenic stretches in proteins could be useful in designing novel molecules against protein misfolding diseases.


Subject(s)
Amyloid/chemistry , Amyloid/toxicity , Curcumin/chemistry , Emetine/chemistry , Gelsolin/chemistry , Benzothiazoles , Fluorescence , Gelsolin/genetics , Hydrophobic and Hydrophilic Interactions , Kinetics , Light , Microscopy, Atomic Force , Molecular Docking Simulation , Particle Size , Peptides/chemistry , Scattering, Radiation , Spectroscopy, Fourier Transform Infrared , Thiazoles/chemistry
15.
Elife ; 32014 Jun 09.
Article in English | MEDLINE | ID: mdl-24913268

ABSTRACT

Malaria inflicts an enormous burden on global human health. The emergence of parasite resistance to front-line drugs has prompted a renewed focus on the repositioning of clinically approved drugs as potential anti-malarial therapies. Antibiotics that inhibit protein translation are promising candidates for repositioning. We have solved the cryo-EM structure of the cytoplasmic ribosome from the human malaria parasite, Plasmodium falciparum, in complex with emetine at 3.2 Å resolution. Emetine is an anti-protozoan drug used in the treatment of ameobiasis that also displays potent anti-malarial activity. Emetine interacts with the E-site of the ribosomal small subunit and shares a similar binding site with the antibiotic pactamycin, thereby delivering its therapeutic effect by blocking mRNA/tRNA translocation. As the first cryo-EM structure that visualizes an antibiotic bound to any ribosome at atomic resolution, this establishes cryo-EM as a powerful tool for screening and guiding the design of drugs that target parasite translation machinery.


Subject(s)
Emetine/chemistry , Plasmodium falciparum/metabolism , Ribosomes/chemistry , Ribosomes/ultrastructure , Animals , Antimalarials/chemistry , Binding Sites , Cryoelectron Microscopy , Cytoplasm/metabolism , Drug Design , Erythrocytes/parasitology , Humans , Models, Molecular , Pactamycin/chemistry , Protein Binding , RNA, Messenger/metabolism , Ribosomal Proteins/chemistry
16.
Nat Prod Commun ; 7(1): 19-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22428233

ABSTRACT

This article reports the structural elucidation of the Alangium alkaloid, tubulosine (1) on the basis of systematic 2D-NMR analyses (DEPT, COSY, TOCSY, NOESY, ROESY, HMQC and HMBC). The data obtained allowed the unambiguous assignment of all proton and carbon signals in 1 for the first time.


Subject(s)
Emetine/analogs & derivatives , Magnetic Resonance Spectroscopy/methods , Emetine/chemistry
17.
J Biol Chem ; 285(10): 7722-38, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20061395

ABSTRACT

The medicinal plant Psychotria ipecacuanha produces ipecac alkaloids, a series of monoterpenoid-isoquinoline alkaloids such as emetine and cephaeline, whose biosynthesis derives from condensation of dopamine and secologanin. Here, we identified three cDNAs, IpeOMT1-IpeOMT3, encoding ipecac alkaloid O-methyltransferases (OMTs) from P. ipecacuanha. They were coordinately transcribed with the recently identified ipecac alkaloid beta-glucosidase Ipeglu1. Their amino acid sequences were closely related to each other and rather to the flavonoid OMTs than to the OMTs involved in benzylisoquinoline alkaloid biosynthesis. Characterization of the recombinant IpeOMT enzymes with integration of the enzymatic properties of the IpeGlu1 revealed that emetine biosynthesis branches off from N-deacetylisoipecoside through its 6-O-methylation by IpeOMT1, with a minor contribution by IpeOMT2, followed by deglucosylation by IpeGlu1. The 7-hydroxy group of the isoquinoline skeleton of the aglycon is methylated by IpeOMT3 prior to the formation of protoemetine that is condensed with a second dopamine molecule, followed by sequential O-methylations by IpeOMT2 and IpeOMT1 to form cephaeline and emetine, respectively. In addition to this central pathway of ipecac alkaloid biosynthesis, formation of all methyl derivatives of ipecac alkaloids in P. ipecacuanha could be explained by the enzymatic activities of IpeOMT1-IpeOMT3, indicating that they are sufficient for all O-methylation reactions of ipecac alkaloid biosynthesis.


Subject(s)
Cephaelis , Emetics/metabolism , Emetine/analogs & derivatives , Emetine/biosynthesis , Isoenzymes/metabolism , Methyltransferases/metabolism , Cephaelis/anatomy & histology , Cephaelis/chemistry , Cephaelis/enzymology , Chromatography, Liquid , Emetics/chemistry , Emetine/chemistry , Isoenzymes/classification , Isoenzymes/genetics , Methylation , Methyltransferases/classification , Methyltransferases/genetics , Molecular Sequence Data , Molecular Structure , Phylogeny , Plant Roots/chemistry , Plant Roots/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tandem Mass Spectrometry
18.
Biochemistry ; 45(38): 11695-702, 2006 Sep 26.
Article in English | MEDLINE | ID: mdl-16981729

ABSTRACT

A small library of bivalent agents was designed to probe the substrate binding sites of the human multidrug transporter P-glycoprotein (P-gp). The bivalent agents were composed of two copies of the P-gp substrate emetine, linked by tethers of varied composition. An optimum distance between the emetine molecules of approximately 10 A was found to be necessary for blocking transport of the known fluorescent substrate rhodamine 123. Additionally, it was determined that hydrophobic tethers were optimal for bridging the bivalent compounds; hydrophilic or cationic moieties within the tether had a detrimental effect on inhibition of transport. In addition to acting as probes of P-gp's drug binding sites, these agents were also potent inhibitors of P-gp. One agent, EmeC5, had IC50 values of 2.9 microM for inhibiting transport of rhodamine 123 and approximately 5 nM for inhibiting the binding of a known P-gp substrate, [125I]iodoarylazidoprazosin. Although EmeC5 is an inhibitor of P-gp and was shown to interact directly with P-gp in one or more of the substrate binding sites, our data suggest that it is either not a P-gp transport substrate itself or a poor one. Most significantly, EmeC5 was shown to reverse the MDR phenotype of MCF-7/DX1 cells when co-administered with a cytotoxic agent, such as doxorubicin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Emetine/metabolism , Molecular Probes/metabolism , Adenosine Triphosphate/metabolism , Azides/metabolism , Cross-Linking Reagents/metabolism , Dimerization , Doxorubicin/pharmacology , Emetine/chemical synthesis , Emetine/chemistry , Flow Cytometry , Humans , Hydrolysis/drug effects , Inhibitory Concentration 50 , Models, Biological , Molecular Probes/analysis , Molecular Probes/chemistry , Photoaffinity Labels , Prazosin/analogs & derivatives , Prazosin/metabolism , Rhodamine 123/pharmacology , Substrate Specificity/drug effects , Tumor Cells, Cultured
19.
Planta Med ; 72(12): 1121-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16783693

ABSTRACT

The cytotoxicity of the alkaloid emetine was determined in six human cell lines that differ in the expression of ABC transporters, such as multiple drug resistance protein 1 (MDR1/ABCB1) and multidrug resistance associated protein 1 (MRP1/ABCC1). Emetine reveals a substantial cytotoxicity due to apoptosis that is inversely correlated with the expression of MDR1. Confluent Caco-2 cells with high MDR1 activity and the MDR1 over-expressing leukemia cell line CEM/ADR5000 are more resistant towards emetine (EC (50) 250 microM and 2 microM, respectively) than cells with a low expression of MDR1 (Jurkat cells, CCRF-CEM cells, HL-60 cells) or cells which over-express MRP1 (HL-60/AR) (EC (50) between 0.05 microM for CCRF-CEM and 0.17 microM for Jurkat cells). Apparently emetine is a substrate for MDR1 but not for MRP1. Furthermore, emetine is able to up-regulate the expression of MDR1 as shown IN VITRO by real-time PCR and transport activity studies.


Subject(s)
Antibiotics, Antineoplastic/analysis , Emetine/chemistry , ATP-Binding Cassette Transporters/pharmacology , Cell Line, Tumor , Emetine/antagonists & inhibitors , Humans
20.
Org Lett ; 8(7): 1295-7, 2006 Mar 30.
Article in English | MEDLINE | ID: mdl-16562875

ABSTRACT

[reaction: see text] Catalytic asymmetric allylation of 3,4-dihydro-6,7-dimethoxyisoquinoline was carried out using allyltrimethoxysilane in the presence of Cu(I) and tol-BINAP. The allyl adduct thus obtained was transformed to a chiral synthetic intermediate for (-)-emetine in good yield. The procedure was applied to the total synthesis of ent-emetine.


Subject(s)
Emetine/chemical synthesis , Imines/chemistry , Catalysis , Cyclization , Emetine/chemistry , Molecular Structure , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...