Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Nat Prod ; 86(6): 1500-1511, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37221656

ABSTRACT

The iboga alkaloids scaffold shows great potential as a pharmacophore in drug candidates for the treatment of neuropsychiatric disorders. Thus, the study of the reactivity of this type of motif is particularly useful for the generation of new analogs suitable for medicinal chemistry goals. In this article, we analyzed the oxidation pattern of ibogaine and voacangine using dioxygen, peroxo compounds, and iodine as oxidizing agents. Special focus was placed on the study of the regio- and stereochemistry of the oxidation processes according to the oxidative agent and starting material. We found that the C16-carboxymethyl ester present in voacangine stabilizes the whole molecule toward oxidation in comparison to ibogaine, especially in the indole ring, where 7-hydroxy- or 7-peroxy-indolenines can be obtained as oxidation products. Nevertheless, the ester moiety enhances the reactivity of the isoquinuclidinic nitrogen to afford C3-oxidized products through a regioselective iminium formation. This differential reactivity between ibogaine and voacangine was rationalized using computational DFT calculations. In addition, using qualitative and quantitative NMR experiments combined with theoretical calculations, the absolute stereochemistry at C7 in the 7-hydroxyindolenine of voacangine was revised to be S, which corrects previous reports proposing an R configuration.


Subject(s)
Ibogaine , Tabernaemontana , Ibogaine/pharmacology , Ibogaine/chemistry , Tabernaemontana/chemistry , Oxidation-Reduction , Skeleton
2.
Cell ; 186(10): 2160-2175.e17, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37137306

ABSTRACT

The serotonin transporter (SERT) removes synaptic serotonin and is the target of anti-depressant drugs. SERT adopts three conformations: outward-open, occluded, and inward-open. All known inhibitors target the outward-open state except ibogaine, which has unusual anti-depressant and substance-withdrawal effects, and stabilizes the inward-open conformation. Unfortunately, ibogaine's promiscuity and cardiotoxicity limit the understanding of inward-open state ligands. We docked over 200 million small molecules against the inward-open state of the SERT. Thirty-six top-ranking compounds were synthesized, and thirteen inhibited; further structure-based optimization led to the selection of two potent (low nanomolar) inhibitors. These stabilized an outward-closed state of the SERT with little activity against common off-targets. A cryo-EM structure of one of these bound to the SERT confirmed the predicted geometry. In mouse behavioral assays, both compounds had anxiolytic- and anti-depressant-like activity, with potencies up to 200-fold better than fluoxetine (Prozac), and one substantially reversed morphine withdrawal effects.


Subject(s)
Ibogaine , Selective Serotonin Reuptake Inhibitors , Serotonin Plasma Membrane Transport Proteins , Small Molecule Libraries , Animals , Mice , Fluoxetine/pharmacology , Ibogaine/chemistry , Ibogaine/pharmacology , Molecular Conformation , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/ultrastructure , Selective Serotonin Reuptake Inhibitors/pharmacology , Small Molecule Libraries/pharmacology
3.
Toxicol Appl Pharmacol ; 434: 115816, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34856211

ABSTRACT

Most women with ovarian cancer are treated with chemotherapy before or after surgery. Unfortunately, chemotherapy treatment can cause negative side effects and the onset of multidrug resistance (MDR). The aim of this study is to evaluate the chemosensitizing effect of a natural compound, voacamine (VOA), in ovarian (A2780 DX) and colon (LoVo DX) cancer drug-resistant cell lines which overexpress P-glycoprotein (P-gp), in combination with paclitaxel (PTX), or doxorubicin (DOX) or 5-fluorouracil (5-FU). VOA, a bisindole alkaloid extracted from Peschiera fuchsiaefolia, has already been shown to be effective in enhancing the effect of doxorubicin, because it interferes with the P-gp function. Ovarian cancer cytotoxicity test shows that single treatments with VOA, DOX and PTX do not modify cell viability, while pretreatment with VOA, and then PTX or DOX for 72 h, induces a decrease. In colon cancer, since 5-FU is not a-substrate for P-gp, VOA has no sensitizing effect while in VOA + DOX there is a decrease in viability. Annexin V/PI test, cell cycle analysis, activation of cleaved PARP1 confirm that VOA plus PTX induce apoptotic cell death. Confocal microscopy observations show the different localization of NF-kB after treatment with VOA + PTX, confirming the inhibition of nuclear translocation induced by VOA pretreatment. Our data show the specific effect of VOA which only works on drugs known to be substrates of P-gp.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm , Ibogaine/analogs & derivatives , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Carcinoma, Ovarian Epithelial/drug therapy , Cell Line, Tumor , Cell Survival , Colonic Neoplasms , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ibogaine/chemistry , Ibogaine/pharmacology , Molecular Structure
4.
BMC Mol Cell Biol ; 22(1): 33, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-34090331

ABSTRACT

BACKGROUND: In the present study, fatty acid synthesis is targeted to combat mammary gland carcinoma by activating prolyl hydroxylase-2 with Voacamine alone and in combination with Tamoxifen. It was hypothesized that the activation of prolyl hydroxylase-2 would inhibit the hypoxia-induced fatty acid synthesis and mammary gland carcinoma. Mammary gland carcinoma was induced with a single dose administration of N-methyl-N-nitrosourea (50 mg/kg,i.p.) and treatment with Voacamine and Tamoxifen 15 days after carcinogen administration. RESULTS: At the end of the study, hemodynamic profiling of animals was recorded to assess the cardiotoxic potential of the drug. Blood serum was separated and subjected to nuclear magnetic resonance spectroscopy. Carmine staining and histopathology of mammary gland tissue were performed to evaluate the anti-angiogenic potential of the drug. The antioxidant potential of the drug was measured with antioxidant markers. Western blotting was performed to study the effect of the drug at the molecular level. CONCLUSION: Results of the study have shown that Voacamine treatment stopped further decrease in body weight of experimental animals. The hemodynamic study evidenced that Voacamine at a low dose is safe in cardiac patients. Microscopic evaluation of mammary gland tissue documented the anti-angiogenic potential of Voacamine and Tamoxifen therapy. Perturbed serum metabolites were also restored to normal along with antioxidant markers. Immunoblotting of mammary gland tissue also depicted restoration of proteins of the hypoxic and fatty acid pathway. Conclusively, Voacamine and its combination with Tamoxifen activated prolyl hydroxylase-2 to combat mammary gland carcinoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/drug therapy , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Ibogaine/analogs & derivatives , Mammary Neoplasms, Experimental/drug therapy , Animals , Antioxidants/metabolism , Body Weight/drug effects , Carcinoma/chemically induced , Carcinoma/metabolism , Carcinoma/pathology , Computer Simulation , Electrocardiography , Fatty Acids/biosynthesis , Female , Heart Rate/drug effects , Ibogaine/chemistry , Ibogaine/pharmacokinetics , Ibogaine/therapeutic use , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Metabolome , Methylnitrosourea , Neovascularization, Pathologic/drug therapy , Rats, Wistar , Tamoxifen/therapeutic use
5.
Biomolecules ; 10(4)2020 03 27.
Article in English | MEDLINE | ID: mdl-32230857

ABSTRACT

Although natural products are an important source of drugs and drug leads, identification and validation of their target proteins have proven difficult. Here, we report the development of a systematic strategy for target identification and validation employing drug affinity responsive target stability (DARTS) and mass spectrometry imaging (MSI) without modifying or labeling natural compounds. Through a validation step using curcumin, which targets aminopeptidase N (APN), we successfully standardized the systematic strategy. Using label-free voacangine, an antiangiogenic alkaloid molecule as the model natural compound, DARTS analysis revealed vascular endothelial growth factor receptor 2 (VEGFR2) as a target protein. Voacangine inhibits VEGFR2 kinase activity and its downstream signaling by binding to the kinase domain of VEGFR2, as was revealed by docking simulation. Through cell culture assays, voacangine was found to inhibit the growth of glioblastoma cells expressing high levels of VEGFR2. Specific localization of voacangine to tumor compartments in a glioblastoma xenograft mouse was revealed by MSI analysis. The overlap of histological images with the MSI signals for voacangine was intense in the tumor regions and showed colocalization of voacangine and VEGFR2 in the tumor tissues by immunofluorescence analysis of VEGFR2. The strategy employing DARTS and MSI to identify and validate the targets of a natural compound as demonstrated for voacangine in this study is expected to streamline the general approach of drug discovery and validation using other biomolecules including natural products.


Subject(s)
Drug Evaluation, Preclinical/methods , Ibogaine/analogs & derivatives , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , CD13 Antigens/metabolism , Curcumin/pharmacology , Female , Human Umbilical Vein Endothelial Cells , Humans , Ibogaine/chemistry , Ibogaine/pharmacokinetics , Ibogaine/pharmacology , Mass Spectrometry , Mice, Inbred BALB C , Molecular Docking Simulation , Tissue Distribution , Vascular Endothelial Growth Factor Receptor-2/chemistry , Xenograft Model Antitumor Assays
6.
Chem Biodivers ; 17(5): e2000002, 2020 May.
Article in English | MEDLINE | ID: mdl-32232967

ABSTRACT

In continuation of our efforts to provide quantitative information on antiaddictive ibogan type alkaloid-producing Tabernaemontana species, we used gas chromatography-mass spectrometry (GC/MS) to compare the alkaloid profiles of the barks and/or leaves of one Mexican and one African species - T. arborea and T. crassa, respectively, with the primary sources of commercially available semisynthetic ibogaine, Voacanga africana root and stem bark. The qualitative and quantitative similarities between T. arborea and V. africana barks consolidate previous reports regarding the potential of the former as a promising alternative source of voacangine and ibogaine. The results also suggest that T. crassa could be used to produce conopharyngine and ibogaline, two compounds with the same basic skeletal structure and possibly similar antiaddictive properties as ibogaine.


Subject(s)
Ibogaine/chemistry , Tabernaemontana/chemistry , Voacanga/chemistry , Ghana , Ibogaine/analogs & derivatives , Mexico , Molecular Conformation , Species Specificity
7.
Chem Phys Lipids ; 223: 104781, 2019 09.
Article in English | MEDLINE | ID: mdl-31229409

ABSTRACT

The plant alkaloid voacamine (VOA) displays many interesting pharmacological activities thus, considering its scarce solubility in water, its encapsulation into liposome formulations for its delivery is an important goal. Different cationic liposome formulations containing a phospholipid, cholesterol and one of two diasteromeric cationic surfactants resulted able to maintain a stable transmembrane difference in ammonium sulfate concentration and/or pH gradient and to accumulate VOA in their internal aqueous bulk. The fluidity of the lipid bilayer affects both the ability to maintain a stable imbalance of protons and/or ammonium ions across the membrane and the entrapment efficiency. It was shown that VOA loaded into liposomes is more efficient than the free alkaloid to revert resistance of osteosarcoma cells resistant to doxorubicin to an extent depending on their composition.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Bone Neoplasms/drug therapy , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Ibogaine/analogs & derivatives , Lipids/chemistry , Osteosarcoma/drug therapy , Antibiotics, Antineoplastic/chemistry , Bone Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/chemistry , Drug Screening Assays, Antitumor , Humans , Ibogaine/chemistry , Liposomes/chemistry , Molecular Conformation , Osteosarcoma/pathology , Particle Size , Surface Properties , Tumor Cells, Cultured
8.
Chem Biodivers ; 16(7): e1900175, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31095891

ABSTRACT

Several species from the Apocynaceae family, such as Tabernanthe iboga, Voacanga africana, and many Tabernaemontana species, produce ibogan type alkaloids, some of which present antiaddictive properties. In this study, we used gas chromatography/mass spectrometry (GC/MS) to examine the efficiency of methanol, acetone, ethyl acetate, dichloromethane, chloroform, and hydrochloric acid in extracting the antiaddictive compounds coronaridine, ibogamine, voacangine, and ibogaine (altogether the CIVI-complex) from the root barks of Tabernaemontana alba and Tabernaemontana arborea. These Mexican species have recently shown great potential as alternative natural sources of the aforementioned substances. Methanol proved to be the most suitable solvent. Furthermore, the crude methanolic extracts could be engaged in a one-step demethoxycarbonylation process that converted coronaridine and voacangine directly into its non-carboxylic counterparts ibogamine and ibogaine, respectively, without the intermediacy of their carboxylic acids. The established protocol straightforwardly simplifies the alkaloid mixture from four to two majority compounds. In summary, our findings facilitate and improve both the qualitative and quantitative analysis of CIVI-complex-containing plant material, as well as outlining a viable method for the bulk production of these scientifically and pharmaceutically important substances from Mexican Tabernaemontana species.


Subject(s)
Bridged-Ring Compounds/isolation & purification , Ibogaine/analogs & derivatives , Ibogaine/isolation & purification , Tabernaemontana/chemistry , Bridged-Ring Compounds/chemistry , Ibogaine/chemistry , Mexico , Molecular Conformation , Plant Bark/chemistry , Plant Roots/chemistry , Species Specificity
9.
Nat Commun ; 10(1): 1687, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30976000

ABSTRACT

The serotonin transporter (SERT), a member of the neurotransmitter:sodium symporter family, is responsible for termination of serotonergic signaling by re-uptake of serotonin (5-HT) into the presynaptic neuron. Its key role in synaptic transmission makes it a major drug target, e.g. for the treatment of depression, anxiety and post-traumatic stress. Here, we apply hydrogen-deuterium exchange mass spectrometry to probe the conformational dynamics of human SERT in the absence and presence of known substrates and targeted drugs. Our results reveal significant changes in dynamics in regions TM1, EL3, EL4, and TM12 upon binding co-transported ions (Na+/K+) and ligand-mediated changes in TM1, EL3 and EL4 upon binding 5-HT, the drugs S-citalopram, cocaine and ibogaine. Our results provide a comprehensive direct view of the conformational response of SERT upon binding both biologically relevant substrate/ions and ligands of pharmaceutical interest, thus advancing our understanding of the structure-function relationship in SERT.


Subject(s)
Protein Domains , Serotonin Plasma Membrane Transport Proteins/chemistry , Cations, Monovalent/metabolism , Citalopram/chemistry , Citalopram/metabolism , Cocaine/chemistry , Cocaine/metabolism , Ibogaine/chemistry , Ibogaine/metabolism , Ligands , Mass Spectrometry , Molecular Dynamics Simulation , Potassium/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Serotonin/chemistry , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/isolation & purification , Serotonin Plasma Membrane Transport Proteins/metabolism , Sodium/metabolism , Structure-Activity Relationship
10.
Nature ; 569(7754): 141-145, 2019 05.
Article in English | MEDLINE | ID: mdl-31019304

ABSTRACT

The serotonin transporter (SERT) regulates neurotransmitter homeostasis through the sodium- and chloride-dependent recycling of serotonin into presynaptic neurons1-3. Major depression and anxiety disorders are treated using selective serotonin reuptake inhibitors-small molecules that competitively block substrate binding and thereby prolong neurotransmitter action2,4. The dopamine and noradrenaline transporters, together with SERT, are members of the neurotransmitter sodium symporter (NSS) family. The transport activities of NSSs can be inhibited or modulated by cocaine and amphetamines2,3, and genetic variants of NSSs are associated with several neuropsychiatric disorders including attention deficit hyperactivity disorder, autism and bipolar disorder2,5. Studies of bacterial NSS homologues-including LeuT-have shown how their transmembrane helices (TMs) undergo conformational changes during the transport cycle, exposing a central binding site to either side of the membrane1,6-12. However, the conformational changes associated with transport in NSSs remain unknown. To elucidate structure-based mechanisms for transport in SERT we investigated its complexes with ibogaine, a hallucinogenic natural product with psychoactive and anti-addictive properties13,14. Notably, ibogaine is a non-competitive inhibitor of transport but displays competitive binding towards selective serotonin reuptake inhibitors15,16. Here we report cryo-electron microscopy structures of SERT-ibogaine complexes captured in outward-open, occluded and inward-open conformations. Ibogaine binds to the central binding site, and closure of the extracellular gate largely involves movements of TMs 1b and 6a. Opening of the intracellular gate involves a hinge-like movement of TM1a and the partial unwinding of TM5, which together create a permeation pathway that enables substrate and ion diffusion to the cytoplasm. These structures define the structural rearrangements that occur from the outward-open to inward-open conformations, and provide insight into the mechanism of neurotransmitter transport and ibogaine inhibition.


Subject(s)
Cryoelectron Microscopy , Ibogaine/chemistry , Ibogaine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/ultrastructure , Serotonin/metabolism , Binding Sites/drug effects , Binding, Competitive , Biological Transport/drug effects , Hallucinogens/chemistry , Hallucinogens/pharmacology , Humans , Models, Molecular , Protein Conformation/drug effects , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Selective Serotonin Reuptake Inhibitors/chemistry , Structure-Activity Relationship
11.
Int J Mol Sci ; 20(3)2019 Jan 23.
Article in English | MEDLINE | ID: mdl-30678129

ABSTRACT

Ibogaine is a psychoactive indole alkaloid with high affinity for several targets including the σ2 receptor. Indeed, extensive data support the involvement of the σ2 receptor in neurological disorders, including Alzheimer's disease, schizophrenia, alcohol abuse and pain. Due to its serious side effects which prevent ibogaine from potential clinical applications, novel ibogaine derivatives endowed with improved σ2 receptor affinity may be particularly beneficial. With the purpose to facilitate the investigation of iboga alkaloid derivatives which may serve as templates for the design of selective σ2 receptor ligands, here we report a deconstruction study on the ibogaine tricyclic moiety and a successive scaffold-hopping of the indole counterpart. A 3D-QSAR model has been applied to predict the σ2 pKi values of the new compounds, whereas a molecular docking study conducted upon the σ2 receptor built by homology modeling was used to further validate the best-scored molecules. We eventually evaluated pinoline, a carboline derivative, for σ2 receptor affinity through radioligand binding assay and the results confirmed the predicted high µM range of affinity and good selectivity. The obtained results could be helpful in the drug design process of new ibogaine simplified analogs with improved σ2 receptor binding capabilities.


Subject(s)
Ibogaine/chemistry , Ibogaine/pharmacology , Hydrogen Bonding , Kinetics , Ligands , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Quantitative Structure-Activity Relationship , Receptors, sigma/chemistry , Receptors, sigma/metabolism
12.
ACS Chem Neurosci ; 9(10): 2475-2483, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30216039

ABSTRACT

The West African iboga plant has been used for centuries by the Bwiti and Mbiri tribes to induce hallucinations during religious ceremonies. Ibogaine, the principal alkaloid responsible for iboga's psychedelic properties, was isolated and sold as an antidepressant in France for decades before its adverse effects precipitated its removal from the market. An ibogaine resurgence in the 1960s was driven by U.S. heroin addicts who claimed that ibogaine cured their opiate addictions. Behavioral pharmacologic studies in animal models provided evidence that ibogaine could blunt self-administration of not only opiates but cocaine, amphetamines, and nicotine. Ibogaine displays moderate-to-weak affinities for a wide spectrum of receptor and transporter proteins; recent work suggests that its actions at nicotinic acetylcholine receptor subtypes may underlie its reputed antiopiate effects. At micromolar levels, ibogaine is neurotoxic and cardiotoxic and has been linked to several deaths by cardiac arrest. Structure-activity studies led to the isolation of the ibogaine analog 18-methoxycoronaridine (18-MC), an α3ß4 nicotinic receptor modulator that retains ibogaine's anticraving properties with few or no adverse effects. Clinical trials of 18-MC treatment of nicotine addiction are pending. Ibogaine analogs may also hold promise for treating anxiety and depression via the "psychedelic-assisted therapy" approach that employs hallucinogens including psilocybin and methylenedioxymethamphetamine ("ecstasy").


Subject(s)
Hallucinogens/chemistry , Hallucinogens/pharmacology , Ibogaine/chemistry , Ibogaine/pharmacology , Cardiotoxicity , Hallucinogens/history , Hallucinogens/therapeutic use , History, 20th Century , History, 21st Century , Humans , Ibogaine/analogs & derivatives , Ibogaine/history , Ibogaine/therapeutic use , Receptors, Nicotinic , Structure-Activity Relationship , Substance-Related Disorders/drug therapy , Tabernaemontana
13.
Neurochem Int ; 114: 26-32, 2018 03.
Article in English | MEDLINE | ID: mdl-29277577

ABSTRACT

In contrast to plasma membrane-expressed nicotinic acetylcholine receptors (nAChRs), mitochondrial nAChRs function in an ion-independent manner by triggering intra-mitochondrial kinases that regulate the release of cytochrome c (Cyt c), an important step in cellular apoptosis. The aim of this study is to determine the structural requirements for mitochondrial α3ß4* nAChR activation by measuring the modulatory effects of two noncompetitive antagonists of these receptors, (+)-catharanthine and (±)-18-methoxycoronaridine [(±)-18-MC], on Cyt c release from wild-type and α7-/- mice mitochondria. The sandwich ELISA results indicated that α3ß4* nAChRs are present in liver mitochondria in higher amounts compared to that in brain mitochondria and that these receptors are up-regulated in α7-/- mice. Correspondingly, (±)-18-MC decreased Cyt c release from liver mitochondria of wild-type mice and from brain and liver mitochondria of α7-/- mice. The effect in wild-type mice mitochondria was mediated mainly by the Src-dependent pathway, regulating the apoptogenic activity of reactive oxygen species, while in α7-/- mice mitochondria, (±)-18-MC strongly affected the calcium-calmodulin kinase II-dependent pathway. In contrast, (+)-catharanthine was much less potent than (±)-18-MC and triggered several signaling pathways, suggesting the involvement of multiple nAChR subtypes. These results show for the first time that noncompetitive antagonists can induce mitochondrial α3ß4* nAChR signaling, giving a more comprehensive understanding on the function of intracellular nAChR subtypes.


Subject(s)
Brain/metabolism , Ibogaine/analogs & derivatives , Mitochondria, Liver/metabolism , Receptors, Nicotinic/physiology , Signal Transduction/physiology , Animals , Brain/drug effects , Dose-Response Relationship, Drug , Female , Ibogaine/chemistry , Ibogaine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Liver/drug effects , Signal Transduction/drug effects
14.
Int J Biochem Cell Biol ; 92: 202-209, 2017 11.
Article in English | MEDLINE | ID: mdl-29042244

ABSTRACT

The inhibitory activity of coronaridine congeners on human (h) α4ß2 and α7 nicotinic acetylcholine receptors (AChRs) is determined by Ca2+ influx assays, whereas their effects on neurons in the ventral inferior (VI) aspect of the mouse medial habenula (MHb) are determined by patch-clamp recordings. The Ca2+ influx results clearly establish that coronaridine congeners inhibit hα3ß4 AChRs with higher selectivity compared to hα4ß2 and hα7 subtypes, and with the following potency sequence, for hα4ß2: (±)-18-methoxycoronaridine [(±)-18-MC]>(+)-catharanthine>(±)-18-methylaminocoronaridine [(±)-18-MAC] ∼ (±)-18-hydroxycoronaridine [(±)-18-HC]; and for hα7: (+)-catharanthine>(±)-18-MC>(±)-18-HC>(±)-18-MAC. Interestingly, the inhibitory potency of (+)-catharanthine (27±4µM) and (±)-18-MC (28±6µM) on MHb (VI) neurons was lower than that observed on hα3ß4 AChRs, suggesting that these compounds inhibit a variety of endogenous α3ß4* AChRs. In addition, the interaction of bupropion with (-)-ibogaine sites on hα3ß4 AChRs is tested by [3H]ibogaine competition binding experiments. The results indicate that bupropion binds to ibogaine sites at desensitized hα3ß4 AChRs with 2-fold higher affinity than at resting receptors, suggesting that these compounds share the same binding sites. In conclusion, coronaridine congeners inhibit hα3ß4 AChRs with higher selectivity compared to other AChRs, by interacting with the bupropion (luminal) site. Coronaridine congeners also inhibit α3ß4*AChRs expressed in MHb (VI) neurons, supporting the notion that these receptors are important endogenous targets for their anti-addictive activities.


Subject(s)
Habenula/drug effects , Habenula/metabolism , Ibogaine/analogs & derivatives , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/metabolism , Animals , Ibogaine/chemistry , Ibogaine/metabolism , Ibogaine/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Nicotinic Antagonists/metabolism , Protein Conformation , Receptors, Nicotinic/chemistry
15.
Planta Med ; 82(11-12): 1030-8, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27257769

ABSTRACT

Herbal preparations from Voacanga africana are used in West and Central African folk medicine and are also becoming increasingly popular as a legal high in Europe. Recently, the main alkaloid voacangine was found to be a potent human ether-à-go-go-related gene channel blocker in vitro. Blockage of this channel might imply possible cardiotoxicity. Therefore, the aim of this study was to characterise voacangine in vivo to assess its pharmacokinetics and to estimate if further studies to investigate its cardiotoxic risk are required. Male Wistar rats received different doses of voacangine as a pure compound and as a hydro-ethanolic extract of V. africana root bark with a quantified amount of 9.71 % voacangine. For the obtained data, a simultaneous population pharmacokinetics model was successfully developed, comprising a two-compartment model for i. v. dosing and a one-compartmental model with two first-order absorption rates for oral dosing. The absolute bioavailability of voacangine was determined to be 11-13 %. Model analysis showed significant differences in the first absorption rate constant for voacangine administered as a pure compound and voacangine from the extract of V. africana. Taking into account the obtained low bioavailability of voacangine, its cardiotoxic risk might be neglectable in healthy consumers, but may have a serious impact in light of drug/drug interactions and impaired health conditions.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ibogaine/analogs & derivatives , Voacanga/chemistry , Animals , Humans , Ibogaine/chemistry , Ibogaine/pharmacokinetics , Ibogaine/pharmacology , Male , Molecular Structure , Rats , Rats, Wistar , Tandem Mass Spectrometry/methods
16.
Cardiovasc Toxicol ; 16(1): 14-22, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25636206

ABSTRACT

The iboga alkaloids are a class of naturally occurring and synthetic compounds, some of which modify drug self-administration and withdrawal in humans and preclinical models. Ibogaine, the prototypic iboga alkaloid that is utilized clinically to treat addictions, has been associated with QT prolongation, torsades de pointes and fatalities. hERG blockade as IKr was measured using the whole-cell patch clamp technique in HEK 293 cells. This yielded the following IC50 values: ibogaine manufactured by semisynthesis via voacangine (4.09 ± 0.69 µM) or by extraction from T. iboga (3.53 ± 0.16 µM); ibogaine's principal metabolite noribogaine (2.86 ± 0.68 µM); and voacangine (2.25 ± 0.34 µM). In contrast, the IC50 of 18-methoxycoronaridine, a product of rational synthesis and current focus of drug development was >50 µM. hERG blockade was voltage dependent for all of the compounds, consistent with low-affinity blockade. hERG channel binding affinities (K i) for the entire set of compounds, including 18-MC, ranged from 0.71 to 3.89 µM, suggesting that 18-MC binds to the hERG channel with affinity similar to the other compounds, but the interaction produces substantially less hERG blockade. In view of the extended half-life of noribogaine, these results may relate to observations of persistent QT prolongation and cardiac arrhythmia at delayed intervals of days following ibogaine ingestion. The apparent structure-activity relationships regarding positions of substitutions on the ibogamine skeleton suggest that the iboga alkaloids might provide an informative paradigm for investigation of the structural biology of the hERG channel.


Subject(s)
Alkaloids/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Tabernaemontana/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ibogaine/analogs & derivatives , Ibogaine/chemical synthesis , Ibogaine/chemistry , Ibogaine/pharmacokinetics , Ibogaine/pharmacology , Ibogaine/toxicity , Patch-Clamp Techniques , Plant Extracts/chemistry , Structure-Activity Relationship
17.
Neuropharmacology ; 99: 675-88, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26302653

ABSTRACT

Noribogaine is the long-lived human metabolite of the anti-addictive substance ibogaine. Noribogaine efficaciously reaches the brain with concentrations up to 20 µM after acute therapeutic dose of 40 mg/kg ibogaine in animals. Noribogaine displays atypical opioid-like components in vivo, anti-addictive effects and potent modulatory properties of the tolerance to opiates for which the mode of action remained uncharacterized thus far. Our binding experiments and computational simulations indicate that noribogaine may bind to the orthosteric morphinan binding site of the opioid receptors. Functional activities of noribogaine at G-protein and non G-protein pathways of the mu and kappa opioid receptors were characterized. Noribogaine was a weak mu antagonist with a functional inhibition constants (Ke) of 20 µM at the G-protein and ß-arrestin signaling pathways. Conversely, noribogaine was a G-protein biased kappa agonist 75% as efficacious as dynorphin A at stimulating GDP-GTP exchange (EC50=9 µM) but only 12% as efficacious at recruiting ß-arrestin, which could contribute to the lack of dysphoric effects of noribogaine. In turn, noribogaine functionally inhibited dynorphin-induced kappa ß-arrestin recruitment and was more potent than its G-protein agonistic activity with an IC50 of 1 µM. This biased agonist/antagonist pharmacology is unique to noribogaine in comparison to various other ligands including ibogaine, 18-MC, nalmefene, and 6'-GNTI. We predict noribogaine to promote certain analgesic effects as well as anti-addictive effects at effective concentrations>1 µM in the brain. Because elevated levels of dynorphins are commonly observed and correlated with anxiety, dysphoric effects, and decreased dopaminergic tone, a therapeutically relevant functional inhibition bias to endogenously released dynorphins by noribogaine might be worthy of consideration for treating anxiety and substance related disorders.


Subject(s)
Analgesics, Opioid/pharmacology , Ibogaine/analogs & derivatives , Receptors, Opioid, kappa/agonists , Analgesics, Opioid/chemistry , Animals , Arrestins/metabolism , CHO Cells , Computer Simulation , Cricetulus , Drug Evaluation, Preclinical , Dynorphins/metabolism , GTP-Binding Proteins/metabolism , Humans , Ibogaine/chemistry , Ibogaine/pharmacology , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Models, Molecular , Morphinans/metabolism , Rats , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects , Substance-Related Disorders/prevention & control , beta-Arrestins
18.
Bioorg Med Chem Lett ; 25(18): 3937-40, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26231157

ABSTRACT

Four alkaloids, voacangine (1), isovoacangine (2), coronaridine (3), and coronaridine hydroxyindolenine (4), were isolated from the MeOH extract of Tabernaemontana divaricata aerial parts by activity-guided fractionation for Wnt signal inhibitory activity. Compounds 1-4 exhibited TCF/ß-catenin inhibitory activities with IC50 values of 11.5, 6.0, 5.8, and 7.3 µM, respectively. Of these, coronaridine (3) decreased ß-catenin levels in SW480 colon cancer cells, while this decrease in ß-catenin was not suppressed by a co-treatment with 3 and MG132, a proteasome inhibitor. These results suggested that the decrease observed in ß-catenin levels by coronaridine (3) did not depend on a proteasomal degradation process. On the other hand, the treatment of SW480 cells with coronaridine (3) caused a decrease in ß-catenin mRNA levels. Thus, coronaridine (3) may inhibit the Wnt signaling pathway by decreasing the mRNA expression of ß-catenin.


Subject(s)
Down-Regulation/drug effects , Ibogaine/analogs & derivatives , RNA, Messenger/biosynthesis , Tabernaemontana/chemistry , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Ibogaine/chemistry , Ibogaine/isolation & purification , Ibogaine/pharmacology , Molecular Conformation , RNA, Messenger/genetics , Structure-Activity Relationship
19.
Int J Biochem Cell Biol ; 65: 81-90, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26022277

ABSTRACT

To characterize the interaction of coronaridine congeners with human (h) α3ß4 nicotinic acetylcholine receptors (AChRs), structural and functional approaches were used. The Ca(2+) influx results established that coronaridine congeners noncompetitively inhibit hα3ß4 AChRs with the following potency (IC50's in µM) sequence: (-)-ibogamine (0.62±0.23)∼(+)-catharanthine (0.68±0.10)>(-)-ibogaine (0.95±0.10)>(±)-18-methoxycoronaridine [(±)-18-MC] (1.47±0.21)>(-)-voacangine (2.28±0.33)>(±)-18-methylaminocoronaridine (2.62±0.57 µM)∼(±)-18-hydroxycoronaridine (2.81±0.54)>(-)-noribogaine (6.82±0.78). A good linear correlation (r(2)=0.771) between the calculated IC50 values and their polar surface area was found, suggesting that this is an important structural feature for its activity. The radioligand competition results indicate that (±)-18-MC and (-)-ibogaine partially inhibit [(3)H]imipramine binding by an allosteric mechanism. Molecular docking, molecular dynamics, and in silico mutation results suggest that protonated (-)-18-MC binds to luminal [i.e., ß4-Phe255 (phenylalanine/valine ring; position 13'), and α3-Leu250 and ß4-Leu251 (leucine ring; position 9')], non-luminal, and intersubunit sites. The pharmacophore model suggests that nitrogens from the ibogamine core as well as methylamino, hydroxyl, and methoxyl moieties at position 18 form hydrogen bonds. Collectively our data indicate that coronaridine congeners inhibit hα3ß4 AChRs by blocking the ion channel's lumen and probably by additional negative allosteric mechanisms by interacting with a series of non-luminal sites.


Subject(s)
Ibogaine/analogs & derivatives , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/metabolism , Allosteric Regulation , Binding, Competitive , HEK293 Cells , Humans , Ibogaine/chemistry , Ibogaine/metabolism , Ibogaine/pharmacology , Models, Molecular , Molecular Dynamics Simulation , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/metabolism , Radioligand Assay , Receptors, Nicotinic/chemistry , Structure-Activity Relationship
20.
J Biol Chem ; 289(42): 28987-9000, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25202009

ABSTRACT

Mutations in the C terminus of the serotonin transporter (SERT) disrupt folding and export from the endoplasmic reticulum. Here we examined the hypothesis that a cytosolic heat shock protein relay was recruited to the C terminus to assist folding of SERT. This conjecture was verified by the following observations. (i) The proximal portion of the SERT C terminus conforms to a canonical binding site for DnaK/heat shock protein of 70 kDa (HSP70). A peptide covering this segment stimulated ATPase activity of purified HSP70-1A. (ii) A GST fusion protein comprising the C terminus of SERT pulled down HSP70-1A. The interaction between HSP70-1A and SERT was visualized in live cells by Förster resonance energy transfer: it was restricted to endoplasmic reticulum-resident transporters and enhanced by an inhibitor that traps HSP70-1A in its closed state. (iv) Co-immunoprecipitation confirmed complex formation of SERT with HSP70-1A and HSP90ß. Consistent with an HSP relay, co-chaperones (e.g. HSC70-HSP90-organizing protein) were co-immunoprecipitated with the stalled mutants SERT-R607A/I608A and SERT-P601A/G602A. (v) Depletion of HSP90ß by siRNA or its inhibition increased the cell surface expression of wild type SERT and SERT-F604Q. In contrast, SERT-R607A/I608A and SERT-P601A/G602A were only rendered susceptible to inhibition of HSP70 and HSP90 by concomitant pharmacochaperoning with noribogaine. (vi) In JAR cells, inhibition of HSP90 also increased the levels of SERT, indicating that endogenously expressed transporter was also susceptible to control by HSP90ß. These findings support the concept that the folding trajectory of SERT is sampled by a cytoplasmic chaperone relay.


Subject(s)
Cytosol/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Adenosine Triphosphatases/metabolism , Endoplasmic Reticulum/metabolism , Fluorescence Resonance Energy Transfer , Glutathione Transferase/metabolism , HEK293 Cells , Humans , Ibogaine/analogs & derivatives , Ibogaine/chemistry , Mutation , Protein Folding , Protein Structure, Tertiary , Protein Transport , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL