Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Biol Chem ; 291(40): 20876-20890, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27481941

RESUMEN

Folding-defective mutants of the human dopamine transporter (DAT) cause a syndrome of infantile dystonia/parkinsonism. Here, we provide a proof-of-principle that the folding deficit is amenable to correction in vivo by two means, the cognate DAT ligand noribogaine and the HSP70 inhibitor, pifithrin-µ. We examined the Drosophila melanogaster (d) mutant dDAT-G108Q, which leads to a sleepless phenotype in flies harboring this mutation. Molecular dynamics simulations suggested an unstable structure of dDAT-G108Q consistent with a folding defect. This conjecture was verified; heterologously expressed dDAT-G108Q and the human (h) equivalent hDAT-G140Q were retained in the endoplasmic reticulum in a complex with endogenous folding sensors (calnexin and HSP70-1A). Incubation of the cells with noribogaine (a DAT ligand selective for the inward-facing state) and/or pifithrin-µ (an HSP70 inhibitor) restored folding of, and hence dopamine transport by, dDAT-G108Q and hDAT-G140Q. The mutated versions of DAT were confined to the cell bodies of the dopaminergic neurons in the fly brain and failed to reach the axonal compartments. Axonal delivery was restored, and sleep time was increased to normal length (from 300 to 1000 min/day) if the dDAT-G108Q-expressing flies were treated with noribogaine and/or pifithrin-µ. Rescuing misfolded versions of DAT by pharmacochaperoning is of therapeutic interest; it may provide opportunities to remedy disorders arising from folding-defective mutants of human DAT and of other related SLC6 transporters.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/química , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efectos de los fármacos , Ibogaína/análogos & derivados , Pliegue de Proteína/efectos de los fármacos , Sueño/genética , Sulfonamidas/farmacología , Animales , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Humanos , Ibogaína/administración & dosificación , Ibogaína/farmacología , Simulación de Dinámica Molecular , Mutación , Fenotipo , Sulfonamidas/administración & dosificación
2.
Psychopharmacology (Berl) ; 241(7): 1417-1426, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38467891

RESUMEN

Ibogaine is a potent atypical psychedelic that has gained considerable attention due to its antiaddictive and antidepressant properties in preclinical and clinical studies. Previous research from our group showed that ibogaine suppresses sleep and produces an altered wakefulness state, which resembles natural REM sleep. However, after systemic administration, ibogaine is rapidly metabolized to noribogaine, which also shows antiaddictive effects but with a distinct pharmacological profile, making this drug a promising therapeutic candidate. Therefore, we still ignore whether the sleep/wake alterations depend on ibogaine or its principal metabolite noribogaine. To answer this question, we conducted polysomnographic recordings in rats following the administration of pure noribogaine. Our results show that noribogaine promotes wakefulness while reducing slow-wave sleep and blocking REM sleep, similar to our previous results reported for ibogaine administration. Thus, we shed new evidence on the mechanisms by which iboga alkaloids work in the brain.


Asunto(s)
Ibogaína , Polisomnografía , Sueño REM , Vigilia , Animales , Sueño REM/efectos de los fármacos , Vigilia/efectos de los fármacos , Vigilia/fisiología , Masculino , Ratas , Ibogaína/análogos & derivados , Ibogaína/farmacología , Ibogaína/administración & dosificación , Ratas Sprague-Dawley , Sueño de Onda Lenta/efectos de los fármacos , Sueño de Onda Lenta/fisiología , Alucinógenos/farmacología , Alucinógenos/administración & dosificación , Electroencefalografía/efectos de los fármacos
4.
Neuropharmacology ; 175: 108194, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32540451

RESUMEN

The primary aim of this study was to determine the anti-neuropathic activity of (±)-18-methoxycoronaridine [(±)-18-MC] and (+)-catharanthine in mice by using the oxaliplatin-induced neuropathic pain paradigm and cold plate test. The results showed that both coronaridine congeners induce anti-neuropathic pain activity at a dose of 72 mg/kg (per os), whereas a lower dose (36 mg/kg) of (+)-catharanthine decreased the progress of oxaliplatin-induced neuropathic pain. To determine the underlying molecular mechanism, electrophysiological recordings were performed on α9α10, α3ß4, and α4ß2 nAChRs as well as voltage-gated calcium (CaV2.2) channels modulated by G protein-coupled γ-aminobutyric acid type B receptors (GABABRs). The results showed that (±)-18-MC and (+)-catharanthine competitively inhibit α9α10 nAChRs with potencies higher than that at α3ß4 and α4ß2 nAChRs and directly block CaV2.2 channels without activating GABABRs. Considering the potency of the coronaridine congeners at Cav2.2 channels and α9α10 nAChRs, and the calculated brain concentration of (+)-catharanthine, it is plausible that the observed anti-neuropathic pain effects are mediated by peripheral and central mechanisms involving the inhibition of α9α10 nAChRs and/or CaV2.2 channels.


Asunto(s)
Analgésicos/administración & dosificación , Caveolina 2/metabolismo , Ibogaína/análogos & derivados , Neuralgia/metabolismo , Receptores Nicotínicos/metabolismo , Alcaloides de la Vinca/administración & dosificación , Animales , Células HEK293 , Humanos , Ibogaína/administración & dosificación , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Xenopus laevis
5.
J Psychoactive Drugs ; 51(2): 155-165, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30967101

RESUMEN

This article examines the therapeutic potential of ibogaine, a powerful oneiric alkaloid derived from Tabernanthe iboga, through exploring the subjective experiences of 44 participants from two observational treatment studies for opioid use disorder. Following treatment with ibogaine HCl, the participants (Mexico, n = 30; New Zealand, n = 14) completed the States of Consciousness Questionnaire (SCQ) to quantify the magnitude of their psychotropic experience. Participants were asked to provide written transcripts of their experiences, with those supplied being analyzed thematically through an iterative process, to produce a set of coded themes. Mean SCQ scores in many domains exceeded 0.6, the cutoff score for a "complete mystical experience," with 43% of participants achieving this in more than five of seven domains. Qualitative data described multiple phenomenological themes, including auditory and visual phenomena. Ibogaine's strong oneiric action promoted cyclic visions leading to confronting realizations involving remorse and regret for participants' actions towards others, but also release from feelings of guilt and worthlessness. Many participants reported feeling a sense of spiritual transformation. We propose that the reported experiences support the meaningfulness of ibogaine's oneiric effects as a discrete element in its capacity for healing, which is distinct from pharmacological actions associated with reduced withdrawal and craving.


Asunto(s)
Alucinógenos/administración & dosificación , Ibogaína/administración & dosificación , Trastornos Relacionados con Opioides/tratamiento farmacológico , Espiritualidad , Adulto , Femenino , Alucinógenos/farmacología , Humanos , Ibogaína/farmacología , Masculino , México , Nueva Zelanda , Encuestas y Cuestionarios , Adulto Joven
6.
Eur J Pharmacol ; 599(1-3): 91-5, 2008 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-18930043

RESUMEN

The novel iboga alkaloid congener 18-methoxycoronaridine (18-MC) is a putative anti-addictive agent that has been shown, in rats, to decrease the self-administration of several drugs of abuse. Previous work has established that 18-MC is a potent antagonist at alpha3beta4 nicotinic receptors. Because high densities of alpha3beta4 nicotinic receptors occur in the medial habenula and the interpeduncular nucleus and moderate densities occur in the dorsolateral tegmentum, ventral tegmental area, and basolateral amygdala, the present study was conducted to determine if 18-MC could act in these brain areas to modulate methamphetamine self-administration in rats. Local administration of 18-MC into either the medial habenula, the interpeduncular area or the basolateral amygdala decreased methamphetamine self-administration. Similar results were produced by local administration into the same brain areas of two other alpha3beta4 nicotinic antagonists, mecamylamine and alpha-conotoxin AuIB. Local administration of 18-MC, or the other antagonists, into the dorsolateral tegmentum or the ventral tegmental area had no effect on methamphetamine self-administration. In contrast, local administration of 18-MC and the other antagonists decreased sucrose self-administration when administered into the dorsolateral tegmentum or basolateral amygdala but had no effect when infused into the medial habenula, interpeduncular nucleus, or ventral tegmental area. These data are consistent with the hypothesis that 18-MC decreases methamphetamine self-administration by indirectly modulating the dopaminergic mesolimbic pathway via blockade of alpha3beta4 nicotinic receptors in the habenulo-interpeduncular pathway and the basolateral amygdala. The data also suggest that the basolateral amygdala along with a different pathway involving alpha3beta4 receptors in the dorsolateral tegmentum mediate the effect of 18-MC on sucrose self-administration.


Asunto(s)
Encéfalo/efectos de los fármacos , Ibogaína/análogos & derivados , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Conducta Adictiva/tratamiento farmacológico , Encéfalo/metabolismo , Conotoxinas/farmacología , Vías de Administración de Medicamentos , Femenino , Ibogaína/administración & dosificación , Ibogaína/farmacología , Mecamilamina/farmacología , Metanfetamina/administración & dosificación , Metanfetamina/farmacología , Ratas , Ratas Long-Evans , Receptores Nicotínicos/metabolismo , Autoadministración , Sacarosa/administración & dosificación , Sacarosa/farmacología , Tegmento Mesencefálico/metabolismo
7.
Medicina (Kaunas) ; 44(12): 984-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19142057

RESUMEN

OBJECTIVE: To evaluate acute toxic effect of ibogaine and noribogaine on the survival of mice and determine median lethal doses of the substances mentioned. MATERIAL AND METHODS: White laboratory mice were used for the experiments. Ibogaine and noribogaine were administered intragastrically to mice via a stomach tube. Control animals received the same volume of saline. The median lethal dose was calculated with the help of a standard formula. RESULTS: To determine the median lethal dose of ibogaine, the doses of 100, 300, 400, and 500 mg/kg were administered intragastrically to mice. The survival time of mice after the drug administration was recorded, as well as the number of survived mice in each group. Upon administration of ibogaine at a dose of 500 mg/kg, all mice in this dose group died. Three out of four mice died in the group, which received 300 mg/kg of ibogaine. No mouse deaths were observed in the group, which received 100 mg/kg of ibogaine. The determined LD(50) value of ibogaine equals to 263 mg/kg of body mass. In order to determine the median lethal dose of noribogaine, the doses of 300, 500, 700, and 900 mg/kg were administered to mice intragastrically. Noribogaine given at a dose of 500 mg/kg had no impact on the mouse survival. The increase of noribogaine dose to 700 mg/kg of mouse body mass led to the death of three out of four mice in the group. Upon administration of noribogaine at a dose of 900 mg/kg, all mice in this group died. The LD(50) value of noribogaine in mice determined on the basis of the number of dead mice and the size of the doses used equals to 630 mg/kg of mouse body mass. The behavior of mice was observed upon administration of ibogaine or noribogaine. Low doses of ibogaine and noribogaine had no impact on the mouse behavior. External effects (convulsions, nervous behaviour, limb paralysis) were observed only when substances were administrated at higher doses. CONCLUSIONS: It has been determined that the median lethal dose of ibogaine and noribogaine equals to 263 mg and 630 mg/kg of mouse body mass, respectively. The toxicity of ibogaine is 2.4 times higher than that of noribogaine.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/toxicidad , Alucinógenos/toxicidad , Ibogaína/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Ibogaína/administración & dosificación , Ibogaína/análogos & derivados , Dosificación Letal Mediana , Ratones , Estómago
8.
J Psychoactive Drugs ; 50(3): 256-265, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29608409

RESUMEN

Opioid use disorder (OUD) is currently an epidemic in the United States (US) and ibogaine is reported to have the ability to interrupt opioid addiction by simultaneously mitigating withdrawal and craving symptoms. This study examined opioid withdrawal and drug craving scores in 50 participants with OUD undergoing a week-long detoxification treatment protocol with ibogaine. The Addiction Severity Index (ASI) was used for baseline characterization of participants' OUD. Clinical Opioid Withdrawal Scale (COWS), Subjective Opioid Withdrawal Scale (SOWS), and Brief Substance Craving Scale (BSCS) scores were collected at 48 and 24 hours prior to ibogaine administration, as well as 24 and 48 hours after ibogaine administration. At 48 hours following ibogaine administration, withdrawal and craving scores were significantly lowered in comparison to baseline: 78% of patients did not exhibit objective clinical signs of opioid withdrawal, 79% reported minimal cravings for opioids, and 68% reported subjective withdrawal symptoms in the mild range. Ibogaine appears to facilitate opioid detoxification by reducing opioid withdrawal and craving in participants with OUD. These results warrant further research using rigorous controlled trials.


Asunto(s)
Alucinógenos/administración & dosificación , Ibogaína/administración & dosificación , Trastornos Relacionados con Opioides/rehabilitación , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Adulto , Ansia/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trastornos Relacionados con Opioides/psicología , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Factores de Tiempo , Adulto Joven
9.
J Psychoactive Drugs ; 50(4): 287-297, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30020025

RESUMEN

We examined persisting effects, self-perceived challenges, and potential benefits associated with positive outcomes following ibogaine detoxification using data collected as part of a larger online retrospective study of 73 patients who received treatment for chronic opioid use in Mexico between 2012 and 2015. A mixed-methods design was used comparing treatment responders versus non-responders, as well as content coding of themes from open-ended questions. Most participants reported positive persisting effects of ibogaine detoxification (e.g., enhanced personal sense of gratitude and authenticity, and meaning and appreciation for life). Compared to non-responders, treatment responders endorsed greater persisting changes in their ability to tolerate difficult/painful feelings, capacity for coping with stress, and reduced unhealthy anger. Treatment responders reported greater change in subjective levels of inner peace, joy, feelings of love/openheartedness, and experiences of sacredness in life. Qualitative analyses revealed that treatment responders reported a heightened sense of spiritual awareness and greater connection to their intra-/interpersonal relationships after ibogaine detoxification. Notable challenges of ibogaine detoxification included psychological and health-related difficulties during treatment and challenges with post-treatment integration. Findings highlight the persisting effects associated with positive response to ibogaine detoxification and possible post-treatment needs (i.e., more integration/aftercare resources). Future research using rigorous experimental designs is needed.


Asunto(s)
Alucinógenos/administración & dosificación , Ibogaína/administración & dosificación , Trastornos Relacionados con Opioides/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Adolescente , Adulto , Femenino , Alucinógenos/efectos adversos , Humanos , Ibogaína/efectos adversos , Relaciones Interpersonales , Masculino , México , Persona de Mediana Edad , Trastornos Relacionados con Opioides/psicología , Estudios Retrospectivos , Síndrome de Abstinencia a Sustancias/psicología , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
10.
Clin Toxicol (Phila) ; 55(6): 600-602, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28489458

RESUMEN

BACKGROUND: Ibogaine is an agent that has been evaluated as an unapproved anti-addictive agent for the management of drug dependence. Sudden cardiac death has been described to occur secondary to its use. We describe the clinical effects and toxicokinetics of ibogaine and noribogaine in a single patient. For this purpose, we developed a LC-MS/MS-method to measure ibogaine and noribogaine plasma-concentrations. We used two compartments with first order absorption. CASE DETAILS: The maximum concentration of ibogaine was 1.45 mg/L. Our patient developed markedly prolonged QTc interval of 647ms maximum, several multiple cardiac arrhythmias (i.e., atrial tachycardia and ventricular tachycardia and Torsades des Pointes). QTc-prolongation remained present until 12 days after ingestion, several days after ibogaine plasma-levels were low, implicating clinically relevant noribogaine concentrations long after ibogaine had been cleared from the plasma. The ratio k12/k21 for noribogaine was 21.5 and 4.28 for ibogaine, implicating a lower distribution of noribogaine from the peripheral compartment into the central compartment compared to ibogaine. CONCLUSIONS: We demonstrated a linear relationship between the concentration of the metabolite and long duration of action, rather than with parent ibogaine. Therefore, after (prolonged) ibogaine ingestion, clinicians should beware of long-term effects due to its metabolite.


Asunto(s)
Cromatografía Liquida/métodos , Ibogaína/análogos & derivados , Ibogaína/farmacocinética , Espectrometría de Masas en Tándem/métodos , Arritmias Cardíacas/inducido químicamente , Femenino , Humanos , Ibogaína/administración & dosificación , Ibogaína/toxicidad , Internet , Síndrome de QT Prolongado/inducido químicamente , Persona de Mediana Edad , Factores de Tiempo , Distribución Tisular , Toxicocinética
11.
Biochem Pharmacol ; 138: 19-30, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28483460

RESUMEN

Indole alkaloids possess a large spectrum of biological activities including anti-protozoal action. Here we report for the first time that voacamine, isolated from the plant Tabernaemontana coronaria, is an antiprotozoal agent effective against a large array of trypanosomatid parasites including Indian strain of Leishmania donovani and Brazilian strains of Leishmania amazonensis and Trypanosoma cruzi. It inhibits the relaxation activity of topoisomerase IB of L. donovani (LdTop1B) and stabilizes the cleavable complex. Voacamine is probably the first LdTop1B-specific poison to act uncompetitively. It has no impact on human topoisomerase I and II up to 200µM concentrations. The study also provides a thorough insight into ultrastructural alterations induced in three kinetoplastid parasites by a specific inhibitor of LdTop1B. Voacamine is also effective against intracellular amastigotes of different drug unresponsive field isolates of Leishmania donovani obtained from endemic zones of India severely affected with visceral leishmaniasis. Most importantly, this is the first report demonstrating the efficacy of a compound to reduce the burden of drug resistant parasites, unresponsive to SAG, amphotericin B and miltefosine, in experimental BALB/c mice model of visceral leishmaniasis. The findings cumulatively provide a strong evidence that voacamine can be a promising drug candidate against trypanosomatid infections.


Asunto(s)
Antiprotozoarios/farmacología , ADN-Topoisomerasas de Tipo I/metabolismo , Ibogaína/análogos & derivados , Leishmania donovani/efectos de los fármacos , Leishmania mexicana/efectos de los fármacos , Inhibidores de Topoisomerasa I/farmacología , Trypanosoma cruzi/efectos de los fármacos , Animales , Antiprotozoarios/administración & dosificación , Antiprotozoarios/aislamiento & purificación , Antiprotozoarios/uso terapéutico , Forma de la Célula/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/química , ADN-Topoisomerasas de Tipo I/genética , Relación Dosis-Respuesta a Droga , Resistencia a Múltiples Medicamentos , Estabilidad de Enzimas/efectos de los fármacos , Femenino , Ibogaína/administración & dosificación , Ibogaína/aislamiento & purificación , Ibogaína/farmacología , Ibogaína/uso terapéutico , Leishmania donovani/enzimología , Leishmania donovani/crecimiento & desarrollo , Leishmania donovani/ultraestructura , Leishmania mexicana/enzimología , Leishmania mexicana/crecimiento & desarrollo , Leishmania mexicana/ultraestructura , Leishmaniasis Visceral/tratamiento farmacológico , Leishmaniasis Visceral/parasitología , Dosificación Letal Mediana , Ratones Endogámicos BALB C , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Corteza de la Planta/química , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tabernaemontana/química , Inhibidores de Topoisomerasa I/administración & dosificación , Inhibidores de Topoisomerasa I/aislamiento & purificación , Inhibidores de Topoisomerasa I/uso terapéutico , Trypanosoma cruzi/enzimología , Trypanosoma cruzi/crecimiento & desarrollo , Trypanosoma cruzi/ultraestructura
12.
J Neurosci ; 25(3): 619-28, 2005 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-15659598

RESUMEN

Alcohol addiction manifests as uncontrolled drinking despite negative consequences. Few medications are available to treat the disorder. Anecdotal reports suggest that ibogaine, a natural alkaloid, reverses behaviors associated with addiction including alcoholism; however, because of side effects, ibogaine is not used clinically. In this study, we first characterized the actions of ibogaine on ethanol self-administration in rodents. Ibogaine decreased ethanol intake by rats in two-bottle choice and operant self-administration paradigms. Ibogaine also reduced operant self-administration of ethanol in a relapse model. Next, we identified a molecular mechanism that mediates the desirable activities of ibogaine on ethanol intake. Microinjection of ibogaine into the ventral tegmental area (VTA), but not the substantia nigra, reduced self-administration of ethanol, and systemic administration of ibogaine increased the expression of glial cell line-derived neurotrophic factor (GDNF) in a midbrain region that includes the VTA. In dopaminergic neuron-like SHSY5Y cells, ibogaine treatment upregulated the GDNF pathway as indicated by increases in phosphorylation of the GDNF receptor, Ret, and the downstream kinase, ERK1 (extracellular signal-regulated kinase 1). Finally, the ibogaine-mediated decrease in ethanol self-administration was mimicked by intra-VTA microinjection of GDNF and was reduced by intra-VTA delivery of anti-GDNF neutralizing antibodies. Together, these results suggest that GDNF in the VTA mediates the action of ibogaine on ethanol consumption. These findings highlight the importance of GDNF as a new target for drug development for alcoholism that may mimic the effect of ibogaine against alcohol consumption but avoid the negative side effects.


Asunto(s)
Alcoholismo/fisiopatología , Etanol/farmacología , Ibogaína/farmacología , Mesencéfalo/efectos de los fármacos , Factores de Crecimiento Nervioso/fisiología , Alcoholismo/tratamiento farmacológico , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Dopamina/fisiología , Etanol/administración & dosificación , Factor Neurotrófico Derivado de la Línea Celular Glial , Humanos , Ibogaína/administración & dosificación , Masculino , Mesencéfalo/metabolismo , Mesencéfalo/fisiología , Ratones , Ratones Endogámicos C57BL , Microinyecciones , Factores de Crecimiento Nervioso/biosíntesis , ARN Mensajero/metabolismo , Ratas , Ratas Long-Evans , Recurrencia , Autoadministración , Sustancia Negra/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos
13.
Eur J Pharmacol ; 552(1-3): 11-4, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17054944

RESUMEN

Ibogaine is an indole alkaloid present in the root of the plant Tabernanthe iboga. It is known to attenuate abstinence syndrome in animal models of drug addiction. Since the anti-addiction effect lasts longer than the presence of ibogaine in the body, some profound metabolic changes are expected. The aim of this study was to investigate the effect of ibogaine on protein expression in rat brains. Rats were treated with ibogaine at 20 mg/kg body weight i.p. and subsequently examined at 24 and 72 h. Proteins were extracted from whole brain and separated by two-dimensional (2-D) electrophoresis. Individual proteins were identified by matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF MS). Enzymes of glycolysis and tricarboxylic acid (TCA) cycle namely glyceraldehyde-3-phosphate dehydrogenase, aldolase A, pyruvate kinase and malate dehydrogenase were induced. The results suggest that the remedial effect of ibogaine could be mediated by the change in energy availability. Since energy dissipating detoxification and reversion of tolerance to different drugs of abuse requires underlying functional and structural changes in the cell, higher metabolic turnover would be favourable. Understanding the pharmacodynamics of anti-addiction drugs highlights the subcellular aspects of addiction diseases, in addition to neurological and psychological perspectives.


Asunto(s)
Encéfalo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Ibogaína/farmacología , Animales , Encéfalo/metabolismo , Electroforesis en Gel Bidimensional , Ibogaína/administración & dosificación , Inyecciones Intraperitoneales , Masculino , Proteínas/análisis , Proteínas/aislamiento & purificación , Ratas , Ratas Wistar , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tabernaemontana/química
14.
Drug Alcohol Depend ; 166: 1-5, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27426011

RESUMEN

The indole alkaloid ibogaine, present in the root bark of the West African rain forest shrub Tabernanthe iboga, has been adopted in the West as a treatment for drug dependence. Treatment of patients requires large doses of the alkaloid to cause hallucinations, an alleged integral part of the patient's treatment regime. However, case reports and case series continue to describe evidences of ataxia, gastrointestinal distress, ventricular arrhythmias and sudden and unexplained deaths of patients undergoing treatment for drug dependence. High doses of ibogaine act on several classes of neurological receptors and transporters to achieve pharmacological responses associated with drug aversion; limited toxicology research suggests that intraperitoneal doses used to successfully treat rodents, for example, have also been shown to cause neuronal injury (purkinje cells) in the rat cerebellum. Limited research suggests lethality in rodents by the oral route can be achieved at approximately 263mg/kg body weight. To consider an appropriate and safe initial dose for humans, necessary safety factors need to be applied to the animal data; these would include factors such as intra- and inter-species variability and for susceptible people in a population (such as drug users). A calculated initial dose to treat patients could be approximated at 0.87mg/kg body weight, substantially lower than those presently being administered to treat drug users. Morbidities and mortalities will continue to occur unless practitioners reconsider doses being administered to their susceptible patients.


Asunto(s)
Alucinógenos/administración & dosificación , Alucinógenos/efectos adversos , Ibogaína/administración & dosificación , Ibogaína/efectos adversos , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Animales , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/diagnóstico , Relación Dosis-Respuesta a Droga , Humanos , Trastornos Relacionados con Sustancias/diagnóstico , Resultado del Tratamiento
15.
Clin Pharmacol Drug Dev ; 5(6): 460-468, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27870477

RESUMEN

Ibogaine is a psychoactive substance that may reduce opioid withdrawal symptoms. This was the first clinical trial of noribogaine, ibogaine's active metabolite, in patients established on methadone opioid substitution therapy (OST). In this randomized, double-blind, placebo-controlled single ascending-dose study, we evaluated the safety, tolerability, and pharmacokinetics of noribogaine in 27 patients seeking to discontinue methadone OST who had been switched to morphine during the previous week. Noribogaine doses were 60, 120, or 180 mg (n = 6/dose level) or matching placebo (n = 3/dose level). Noribogaine was well tolerated. The most frequent treatment-emergent adverse events were noneuphoric changes in light perception ∼1 hour postdose, headache, and nausea. Noribogaine had dose-linear increases for AUC and Cmax and was slowly eliminated (mean t1/2 range, 24-30 hours). There was a concentration-dependent increase in QTcI (0.17 ms/ng/mL), with the largest observed mean effect of ∼16, 28, and 42 milliseconds in the 60-, 120-, and 180-mg groups, respectively. Noribogaine showed a nonstatistically significant trend toward decreased total score in opioid withdrawal ratings, most notably at the 120-mg dose; however, the study design may have confounded evaluations of time to resumption of OST. Future exposure-controlled multiple-dose noribogaine studies are planned that will address these safety and design issues.


Asunto(s)
Ibogaína/análogos & derivados , Adulto , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Electrocardiografía/efectos de los fármacos , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Ibogaína/administración & dosificación , Ibogaína/efectos adversos , Ibogaína/farmacocinética , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/fisiopatología , Masculino , Metadona , Narcóticos , Tratamiento de Sustitución de Opiáceos , Trastornos Relacionados con Opioides/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico
16.
J Ethnopharmacol ; 189: 10-3, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27180314

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Root bark from Tabernanthe iboga has been used traditionally in West Africa as a psychoactive substance in religious rituals. In smaller doses it is reported anecdotally to have stimulant properties. AIM OF THE STUDY: To evaluate the influence of a single 20mg ibogaine dose on psychological variables reflecting subjective mood state and a range of cognitive functions. MATERIALS AND METHODS: 21 healthy male volunteers received single 20mg doses of ibogaine after 6 days pretreatment with double-blind paroxetine or placebo. We compared responses to a battery of psychometric tests and subjective mood ratings performed before and 2h after ibogaine dosing, and assessed relationships between changes in test scores and concentrations of active moiety (the sum of molar noribogaine and ibogaine concentrations). Psychological tests were chosen based on responsiveness to opioid and serotonergic ligands. RESULTS: Ibogaine had minimal influence on psychological tests and mood ratings. The ability to selectively ignore distracting spatial information showed some evidence of modulation; however because this effect was limited to the less challenging condition calls into question the reliability of this result. CONCLUSION: We were unable to identify stimulant effects after single 20mg doses of ibogaine. Future research is needed to confirm whether active moiety concentrations impact selective attention abilities while leaving other cognitive functions and mood state unaffected.


Asunto(s)
Afecto/efectos de los fármacos , Fármacos del Sistema Nervioso Central/administración & dosificación , Cognición/efectos de los fármacos , Ibogaína/administración & dosificación , Adulto , Atención/efectos de los fármacos , Fármacos del Sistema Nervioso Central/sangre , Fármacos del Sistema Nervioso Central/farmacocinética , Método Doble Ciego , Esquema de Medicación , Voluntarios Sanos , Humanos , Ibogaína/análogos & derivados , Ibogaína/sangre , Ibogaína/farmacocinética , Masculino , Pruebas Neuropsicológicas , Nueva Zelanda , Fitoterapia , Plantas Medicinales , Psicometría , Adulto Joven
17.
J Clin Pharmacol ; 56(8): 960-5, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26763764

RESUMEN

The aim of this study was to switch patients established on methadone opioid substitution therapy (OST) to morphine over 1 week. Subjects established on daily methadone OST (mean dose 60 mg/day) were switched to morphine slow-release capsules, dosed at 4× the previous total daily methadone dose, for 6 days, then given morphine syrup dosed q3h. All 27 subjects enrolled in this study completed the switch from methadone to morphine. Opioid withdrawal symptoms (OWS) peaked within 12-24 hours of starting morphine, and 24/27 subjects required higher daily morphine doses (mean 5.2× multiple). Pharmacokinetic evaluation showed that 91% of methadone was cleared during this time, with a mean elimination half-life of 59 hours. The most frequent treatment-emergent non-OWS adverse events were headache, nausea, constipation, and neck pain. The method described here appears to be a safe and acceptable approach to switch subjects from methadone to morphine.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Sustitución de Medicamentos/métodos , Metadona/administración & dosificación , Morfina/administración & dosificación , Tratamiento de Sustitución de Opiáceos/métodos , Trastornos Relacionados con Opioides/tratamiento farmacológico , Adulto , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/sangre , Método Doble Ciego , Femenino , Cefalea/inducido químicamente , Humanos , Ibogaína/administración & dosificación , Ibogaína/efectos adversos , Ibogaína/análogos & derivados , Ibogaína/sangre , Masculino , Metadona/efectos adversos , Metadona/sangre , Morfina/efectos adversos , Morfina/sangre , Náusea/inducido químicamente , Tratamiento de Sustitución de Opiáceos/efectos adversos , Trastornos Relacionados con Opioides/sangre , Resultado del Tratamiento
18.
J Med Microbiol ; 54(Pt 7): 647-653, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15947429

RESUMEN

In the present study the effect of the indole alkaloid ibogaine on the in vitro lipolytic activity and adherence to epithelial cells of Candida albicans was investigated. The substance was administered intraperitoneally at a dose of 5 mg kg(-1) day(-1) in mice with disseminated and gastrointestinal C. albicans infections. Ibogaine significantly decreased the rate of mortality and the number of C. albicans c.f.u. recovered from the kidney, liver and spleen. Ibogaine interfered with the early stages of both disseminated and gastrointestinal C. albicans infections but did not reduce the number of C. albicans c.f.u. in the organs at the late phase of infections. The development of a specific immune response was not influenced by ibogaine, since the delayed-type hypersensitivity reaction to C. albicans and the production of interferon (IFN)-gamma were similar in control and ibogaine-treated mice. The combined use of amphotericin B plus ibogaine in the treatment of mice with gastrointestinal infection reduced organ colonization more strongly than each substance alone.


Asunto(s)
Candida albicans/efectos de los fármacos , Candidiasis/tratamiento farmacológico , Enfermedades Gastrointestinales/tratamiento farmacológico , Ibogaína/farmacología , Adhesividad/efectos de los fármacos , Anfotericina B/uso terapéutico , Animales , Antifúngicos/uso terapéutico , Encéfalo/microbiología , Candida albicans/crecimiento & desarrollo , Candida albicans/fisiología , Candidiasis/microbiología , Quimioterapia Combinada , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Enfermedades Gastrointestinales/microbiología , Hipersensibilidad Tardía , Ibogaína/administración & dosificación , Ibogaína/uso terapéutico , Inyecciones Intraperitoneales , Riñón/microbiología , Lipasa/antagonistas & inhibidores , Hígado/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Bazo/microbiología
19.
Eur J Pharmacol ; 525(1-3): 98-104, 2005 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-16289028

RESUMEN

18-Methoxyroconaridine (18-MC), a synthetic derivative of ibogaine, reduces morphine self-administration and alleviates several signs of acute opioid withdrawal in rats. Although there is already well documented evidence of the mechanism mediating 18-MC's action to reduce the rewarding effects of morphine, nothing is known about the mechanism responsible for 18-MC's attenuation of opioid withdrawal. In vitro studies have demonstrated that 18-MC is a potent antagonist of alpha3beta4 nicotinic receptors (IC50=0.75 microM), which are predominantly located in the medial habenula and interpeduncular nuclei. Previous work indicating that alpha3beta4 nicotinic receptors mediate 18-MC's effects on drug self-administration prompted us to assess whether brain areas having high or moderate densities of alpha3beta4 receptors might be involved in 18-MC's modulation of opioid withdrawal. To test this possibility, 18-MC was locally administered into the medial habenula, interpeduncular nucleus and locus coeruleus of morphine-dependent rats; this treatment was followed by naltrexone to precipitate a withdrawal syndrome. Pretreatment with various doses of 18-MC into the locus coeruleus significantly reduced wet-dog shakes, teeth chattering, burying and diarrhea, while pretreatment into the medial habenula attenuated teeth chattering, burying, and weight loss. Some doses of 18-MC administered into the interpeduncular nucleus significantly ameliorated rearing, teeth chattering, and burying, while other doses exacerbated diarrhea and teeth chattering. The present findings suggest that 18-MC may act in all three nuclei to suppress various signs of opioid withdrawal.


Asunto(s)
Encéfalo/efectos de los fármacos , Ibogaína/análogos & derivados , Locus Coeruleus/efectos de los fármacos , Dependencia de Morfina/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Animales , Encéfalo/fisiopatología , Vías de Administración de Medicamentos , Femenino , Ibogaína/administración & dosificación , Ibogaína/uso terapéutico , Locus Coeruleus/fisiopatología , Morfina/farmacología , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Narcóticos/farmacología , Ratas , Ratas Sprague-Dawley
20.
J Clin Pharmacol ; 55(6): 680-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25651476

RESUMEN

Conversion of ibogaine to its active metabolite noribogaine appears to be mediated primarily by CYP2D6. We compared 168 hours pharmacokinetic profiles of both analytes after a single oral 20 mg dose of ibogaine in 21 healthy subjects who had been pretreated for 6 days with placebo or the CYP2D6 inhibitor paroxetine. In placebo-pretreated subjects, ibogaine was rapidly converted to noribogaine. Median peak noribogaine concentrations occurred at 4 hours. Compared with placebo-pretreated subjects, paroxetine-pretreated subjects had rapid (Tmax = 1.5 hours) and substantial absorption of ibogaine, with detectable levels out to 72 hours, and an elimination half-life of 10.2 hours. In this group, ibogaine was also rapidly converted to noribogaine with a median Tmax of 3 hours. Extent of noribogaine exposure was similar in both groups. CYP2D6 phenotype was robustly correlated with ibogaine AUC0-t (r = 0.82) and Cmax (r = 0.77). Active moiety (ibogaine plus noribogaine) exposure was ∼2-fold higher in paroxetine-pretreated subjects. Single 20 mg ibogaine doses were safe and well tolerated in all subjects. The doubling of exposure to active moiety in subjects with reduced CYP2D6 activity suggests it may be prudent to genotype patients awaiting ibogaine treatment, and to at least halve the intended dose of ibogaine in CYP2D6 poor metabolizers.


Asunto(s)
Citocromo P-450 CYP2D6/metabolismo , Voluntarios Sanos , Ibogaína/análogos & derivados , Ibogaína/farmacocinética , Paroxetina/farmacocinética , Adulto , Inhibidores del Citocromo P-450 CYP2D6/farmacocinética , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Antagonistas de Aminoácidos Excitadores/farmacocinética , Femenino , Semivida , Humanos , Ibogaína/administración & dosificación , Ibogaína/análisis , Ibogaína/metabolismo , Masculino , Paroxetina/administración & dosificación , Factores de Tiempo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA