Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Chem Res Toxicol ; 34(5): 1348-1354, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33913699

RESUMEN

Linezolid, the principal oxazolidinone antibiotic for therapy of Gram-positive infections, is limited by its myelosuppression and monoamine oxidase (MAO) inhibition, with the latter manifested as serotonergic neurotoxicity. The oral oxazolidinone contezolid and its injectable prodrug contezolid acefosamil are developed to overcome the above limitations. Serotonergic profiles for contezolid in vitro and for orally administered contezolid acefosamil in rodents are reported. Contezolid exhibited 2- and 148-fold reduction over linezolid reversible inhibition of MAO-A and MAO-B human enzyme isoforms. In the mouse head-twitch model, contezolid acefosamil was devoid of neurotoxicity at supratherapeutic oral doses of 40, 80, and 120 mg/kg. In the rat tyramine challenge model, no significant increase in arterial blood pressure was observed for contezolid acefosamil up to 120 mg/kg oral dosing. In these tests, the comparator linezolid has elicited serotonergic responses. Thus, contezolid and contezolid acefosamil exhibited an attenuated propensity to induce MAO-related serotonergic neurotoxicity. The data support a continued clinical evaluation of these agents, with potential to expand oxazolidinone therapies to patient populations on concurrent selective serotonin reuptake inhibitor medications or where MAO inhibitors are contraindicated.


Asunto(s)
Antibacterianos/farmacología , Presión Sanguínea/efectos de los fármacos , Inhibidores de la Monoaminooxidasa/farmacología , Síndromes de Neurotoxicidad/tratamiento farmacológico , Oxazolidinonas/farmacología , Piridonas/farmacología , Administración Oral , Animales , Antibacterianos/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratones , Estructura Molecular , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/administración & dosificación , Oxazolidinonas/administración & dosificación , Piridonas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Tiramina/metabolismo
2.
Pharm Res ; 38(3): 461-471, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33709330

RESUMEN

PURPOSE: The biodistribution imaging kinetics of near-infrared monoamine oxidase inhibitor (NMI) are reported here. METHODS: NMI was administered intravenously or orally to mice and detected by NIR fluorescence optical imaging within minutes and the longitudinal signal distribution was measured for up to 1 week after a single dose. RESULTS: NMI rapidly reached 3.7-fold higher ventral and 3.2-fold higher brain region fluorescent signal intensity compared to oral route at 24 h. Similar patterns of NMI biodistribution were found in mice with or without intracranial implanted GL26 brain tumors. NMI was highly associated with tumors in contrast to adjacent non-tumor brain, confirming diagnostic utility. NMI 5 mg/kg imaging signal in brain at 48 h was optimal (tumor/non-tumor ratio > 3.5) with minimum off-target distribution. Intravenous NMI imaging signal peaked between 24 h and 48 h for lung, liver, kidney, blood, brain, and most other tissues. Clearance (signal weaker, but still present) from most tissues occurred by day 7. Intravenous low dose (0.5 mg/kg) minimally labeled tumor and other tissues, 5 mg/kg showed optimal imaging signal in glioma at a dose we previously reported as efficacious, and 50 mg/kg was tolerable but saturated the tissue signals beyond tumor specificity. Gel electrophoresis showed two major bands present in brain tumor and tissue protein lysates. CONCLUSIONS: Intravenous 5 mg/kg was optimal dose to target brain tumor and identified off-target organs of concern: lungs, liver, and kidneys. These results demonstrate the biodistribution and optimal dose range of NMI for treatment and diagnostic monitoring of glioma.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Colorantes Fluorescentes/química , Glioma/diagnóstico por imagen , Inhibidores de la Monoaminooxidasa/farmacocinética , Imagen Óptica/métodos , Animales , Encéfalo , Relación Dosis-Respuesta a Droga , Rayos Infrarrojos , Riñón , Hígado , Pulmón , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de la Monoaminooxidasa/administración & dosificación , Neoplasias Experimentales , Distribución Tisular
3.
Prostate Cancer Prostatic Dis ; 24(1): 61-68, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32123315

RESUMEN

PURPOSE: Monoamine oxidase A (MAOA) influences prostate cancer growth and metastasis in pre-clinical models. We examined effects of phenelzine (a monoamine oxidase inhibitor) in patients with biochemical recurrent castrate-sensitive prostate cancer. MATERIALS AND METHODS: An open-label single arm clinical trial enrolled subjects with biochemical recurrent prostate cancer defined by PSA ≥ 0.4 ng/ml (post prostatectomy) or PSA ≥ 2 ng/ml above nadir (post-radiation therapy); no evidence of metastasis on imaging; and normal androgen levels. Subjects received phenelzine 30 mg orally twice daily. Mood symptoms were assessed with the hospital anxiety depression score (HADS) questionnaire. The primary endpoint was the proportion of patients who achieved a PSA decline of ≥50% from baseline. RESULTS: Characteristics of the 20 eligible patients enrolled included: mean ± SD age 66.9 ± 4.8 years and PSA 4.7 ± 5.8 ng/dl. Maximal PSA declines ≥30% and ≥50% were observed in 25% (n = 5/20) and 10% (n = 2/20) of subjects, respectively. At 12 weeks, 17 subjects remained on treatment with PSA declines ≥30% and ≥50% of 24% (n = 4/17) and 6% (n = 1/17), respectively. Common toxicities observed included dizziness (grade 1 = 45%, grade 2 = 35%), hypertension (grade ≥ 2 = 30%), and edema (grade 1 = 25%, grade 2 = 10%). There was one episode of grade 4 hypertension (cycle 4) and two episodes of grade 3 syncope (cycle 12 and cycle 14) requiring treatment discontinuation. HADS questionnaires demonstrated a significant decrease in anxiety with no change in depressive symptoms on treatment. CONCLUSIONS: Phenelzine demonstrated efficacy in patients with biochemical recurrent castrate-sensitive prostate cancer. Most treatment-related toxicities were mild, but rare significant and reversible cardiovascular toxicities were observed. Therapies directed at MAOA may represent a new avenue for treatment in patients with recurrent prostate cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Fenelzina/administración & dosificación , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/tratamiento farmacológico , Adenocarcinoma/sangre , Adenocarcinoma/patología , Anciano , Biomarcadores de Tumor/sangre , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de la Monoaminooxidasa/administración & dosificación , Recurrencia Local de Neoplasia/sangre , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología , Resultado del Tratamiento
4.
Bioorg Chem ; 104: 104203, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32932120

RESUMEN

The 3-pyridazinylcoumarin scaffold was previously reported as an efficient core for the discovery of reversible and selective inhibitors of MAO-B, a validated drug target for PD therapy which also plays an important role in the AD progress. Looking for its structural optimization, novel compounds of hybrid structure coumarin-pyridazine, differing in polarizability and lipophilicity properties, were synthesized and tested against the two MAO isoforms, MAO-A and MAO-B (compounds 17a-f and 18a-f). All the designed compounds selectively inhibited the MAO-B isoenzyme, exhibiting many of them IC50 values ranging from sub-micromolar to nanomolar grade and lacking neuronal toxicity. The 7-bromo-3-(6-bromopyridazin-3-yl)coumarin (18c), the most potent compound of these series (IC50 = 60 nM), was subjected to further in vivo studies in a reserpine-induced mouse PD model. The obtained results suggest a promising potential for 18c as antiparkinsonian agent. Molecular modeling studies also provided valuable information about the enzyme-drug interactions and the potential pharmacokinetic profile of the novel compounds.


Asunto(s)
Cumarinas/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Monoaminooxidasa/metabolismo , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Piridazinas/farmacología , Animales , Cumarinas/administración & dosificación , Cumarinas/química , Relación Dosis-Respuesta a Droga , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Modelos Moleculares , Estructura Molecular , Inhibidores de la Monoaminooxidasa/administración & dosificación , Inhibidores de la Monoaminooxidasa/química , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química , Enfermedad de Parkinson/metabolismo , Piridazinas/administración & dosificación , Piridazinas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
5.
Pharmazie ; 75(9): 412-416, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32797765

RESUMEN

Recent studies have shown that monoamine oxidase A (MAOA) is significantly expressed in malignant prostate cancer (PCa) and plays an important role in tumorigenesis indicating its potential to serve as a target for PCa treatment. Here, we choose the small molecule isoniazid as the MAOA inhibition functionality and incorporated it in the tumor-targeting moiety of heptamethine carbocyanine dyes via a pH sensitive hydrazone bond to design and synthesize novel MAOA inhibitor isoniazid-heptamethine carbocyanine dye conjugates. Cytotoxicity assay in PC-3 cells shows that all conjugates possessed improved antitumor efficacy compared with isoniazid. The tested compounds also demonstrated a moderate MAOA inhibitory effect. In conclusion, these results indicate that these conjugates exert antitumor effects by delivering the MAOA-inhibiting moiety to PCa cells.


Asunto(s)
Carbocianinas/química , Isoniazida/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Isoniazida/administración & dosificación , Isoniazida/química , Masculino , Inhibidores de la Monoaminooxidasa/administración & dosificación , Inhibidores de la Monoaminooxidasa/química , Neoplasias de la Próstata/patología
6.
J Integr Neurosci ; 19(2): 229-237, 2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32706187

RESUMEN

Multiple sclerosis is a progressive autoimmune disorder of the myelin sheath and is the most common inflammatory disease of young adults. Up to 65% of multiple sclerosis patients have cognitive impairments such as memory loss and difficulty in understanding and maintaining attention and concentration. Many pharmacological interventions have been used to reverse motor impairments in multiple sclerosis patients; however, none of these drugs improve cognitive function. Melatonin can diffuse through the blood-brain barrier and has well-known antioxidant and anti-inflammatory properties with almost no side effects; it is, therefore, a promising neuroprotective supplement for many neurological diseases, such as multiple sclerosis, Alzheimer's disease, Parkinson's disease, ischemic stroke, and fibromyalgia. However, only some researches have assessed the effect of melatonin on cognitive dysfunction in multiple sclerosis. Here, we evaluated the effects of melatonin supplementation on memory defects induced by cuprizone in a mouse model of multiple sclerosis. Cuprizone (400 mg/kg) and melatonin (80 mg/kg) were administered to SWR/J mice daily for 5 weeks. Open field, tail-flick, and novel object recognition behavioral tests were performed. Also, expression of cAMP-response element-binding protein, synaptophysin, and postsynaptic density protein 95 were measured in the prefrontal cortex. Melatonin significantly improved the memory defects induced by cuprizone toxicity by up-regulating cAMP-response element-binding protein and by increasing expression of the synapse-associated synaptophysin and postsynaptic density protein 95 genes in the prefrontal cortex. These results indicate that melatonin may provide protective effects against memory impairments associated with multiple sclerosis.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Homólogo 4 de la Proteína Discs Large/efectos de los fármacos , Melatonina/farmacología , Trastornos de la Memoria/tratamiento farmacológico , Esclerosis Múltiple/complicaciones , Fármacos Neuroprotectores/farmacología , Corteza Prefrontal/efectos de los fármacos , Sinaptofisina/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Cuprizona/administración & dosificación , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Expresión Génica/efectos de los fármacos , Melatonina/administración & dosificación , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Ratones , Inhibidores de la Monoaminooxidasa/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Corteza Prefrontal/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Aprendizaje Espacial/efectos de los fármacos , Sinaptofisina/metabolismo
7.
J Parkinsons Dis ; 10(1): 173-178, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31868682

RESUMEN

BACKGROUND: A wide variety of conversion factors for a levodopa-equivalent-dose (LED) have been proposed for each Parkinson's disease (PD) medication. The currently-used set of conversion factors is based on studies that relied on subjective experience or theoretical assumptions. This set was never validated in patients receiving polytherapy. OBJECTIVES: To use real-life data to identify an optimal set of conversion factors independent of prior assumptions regarding clinical efficacy of different medications. METHODS: Retrospective analysis of data from 206 cognitively-preserved patients with advanced PD receiving polytherapy before deep brain stimulation (DBS) surgery. A nonlinear automated problem solver was used to find a set of conversion factors that, when applied, minimized the coefficient of variation of LEDs in a relatively homogenous cohort of patients. RESULTS: Independent and model-free evaluation of a wide range of possible sets of conversion factors to LED suggested a set of normalized conversion factors for immediate release levodopa (1.00), controlled release levodopa (0.88), and amantadine (1.23). A minimal clinical benefit of entacapone was observed for patients with motor fluctuations. Our analysis could not detect conversion factors for dopamine agonists and MAO-B inhibitors, possibly because their clinical contribution when added to levodopa is limited. CONCLUSIONS: Independent from previous studies and prior assumptions we show that the currently-used LED conversion factors for immediate release levodopa, controlled release levodopa and amantadine are largely correct and that dopamine agonists, MAO-B inhibitors and entacapone, given in addition to levodopa, have little additional clinical value for PD patients with motor fluctuations.


Asunto(s)
Amantadina/farmacología , Antiparkinsonianos/farmacología , Agonistas de Dopamina/farmacología , Levodopa/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Evaluación de Resultado en la Atención de Salud/normas , Enfermedad de Parkinson/tratamiento farmacológico , Anciano , Amantadina/administración & dosificación , Antiparkinsonianos/administración & dosificación , Catecoles/farmacología , Agonistas de Dopamina/administración & dosificación , Quimioterapia Combinada , Femenino , Humanos , Levodopa/administración & dosificación , Masculino , Persona de Mediana Edad , Inhibidores de la Monoaminooxidasa/administración & dosificación , Nitrilos/farmacología , Estudios Retrospectivos
8.
Nutr Neurosci ; 23(7): 516-525, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30289026

RESUMEN

Emerging evidence from human intervention trials indicates health benefits of consuming blackcurrant fruit, including improvements to cognitive performance, modulation of blood flow, regulation of blood glucose and inhibition of enzymes underpinning normal cognitive function. Of particular relevance is our previous demonstration of monoamine oxidase (MAO)-A and B inhibition after the consumption of a New Zealand "Blackadder" blackcurrant juice in humans. The current study uses a double-blind, placebo-controlled, randomised cross- over design to assess the pharmacodynamics of the effects on platelet MAO-B inhibition and associated substrates, plasma prolactin levels and blood glucose levels after consumption of a single serve of "Blackadder" blackcurrant juice standardised to 500 mg polyphenols. Eight healthy male (20--35 years) participants completed the trial. Measurements were obtained at baseline 15, 30, 45, 60, 100, 120, 150, 180, 240 mins and 24 h post dose. A fast, absolute and reversible inhibition of blood platelet MAO-B (P < 0.001) and a significant but delayed reduction in plasma prolactin (P < 0.001) were observed following the consumption of "Blackadder" blackcurrant juice when compared to a placebo control. No interpretable changes in substrates of MAO or associated metabolites were seen. These data provide a clear time course of the reversible inhibition of MAO-B after the single consumption of a of New Zealand "Blackadder" blackcurrant juice standardised at 500 mg of polyphenols and, therefore, provide a therapeutic window on which to base future nutritional interventions.


Asunto(s)
Inhibidores de la Monoaminooxidasa/administración & dosificación , Ácido gammalinolénico/farmacocinética , Adulto , Glucemia/efectos de los fármacos , Plaquetas , Estudios Cruzados , Método Doble Ciego , Humanos , Masculino , Polifenoles/farmacocinética , Prolactina/sangre , Adulto Joven
9.
Biomédica (Bogotá) ; 39(3): 491-501, jul.-set. 2019. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1038809

RESUMEN

Abstract Introduction: Parkinson's disease is the second most common neurodegenerative disease. Monoamine oxidase B inhibitors are used in the treatment of this disease concomitantly with levodopa or as monotherapy. Several substituted coumarins have shown activity as inhibitors of monoamine oxidase B. Objective: To evaluate the possible antiparkinsonian effects of the coumarin analogue FCS005 (3-methyl-7H-furo[3,2-g]chromen-7-one) in mouse models, as well as its inhibitory activity towards monoamine oxidases (MAO) and its antioxidant activity. Materials and methods: FCS005 was synthesized and the reversal of hypokinesia was evaluated in the reserpine and levodopa models. Moreover, in the haloperidol model, its anticataleptic effects were evaluated. Additionally, the monoamine oxidase inhibitory activity and antioxidant activity of FCS005 were evaluated using in vitro and ex vivo studies, respectively. Results: FCS005 (100 mg/kg) caused the reversal of hypokinesia in the reserpine and levodopa models. This furocoumarin also presented anti-cataleptic effects at the same dose. Besides, it showed selective inhibitory activity towards the MAO-B isoform and antioxidant activity. Conclusion: These results attribute interesting properties to the compound FCS005. It is important to continue research on this molecule considering that it could be a potential antiparkinsonian agent.


Resumen Introducción. El segundo trastorno neurodegenerativo más común es la enfermedad de Parkinson. Los inhibidores de la monoamino oxidasa B se emplean en el tratamiento de esta enfermedad en monoterapia o concomitantemente con levodopa. Varios compuestos cumarínicos han mostrado actividad como inhibidores de la monoamino oxidasa B. Objetivo. Evaluar los posibles efectos antiparkinsonianos del análogo de la cumarina FCS005 (3-methyl-7H-furo [3,2-g ] chromen-7-one) en modelos de ratones, la actividad inhibitoria frente a las monoamino oxidasas (MAO) y la actividad antioxidante. Materiales y métodos. Se sintetizó la furanocumarina FCS005 y, en los modelos de reserpina y levodopa, se evaluó si producía reversión de la hipocinesia; en el modelo de haloperidol se evaluaron sus efectos anticatalépticos. Además, se evaluó in vitro la actividad inhibidora de MAO y, ex vivo, la actividad antioxidante del compuesto FCS005. Resultados. El compuesto FCS005 en dosis de 100 mg/kg produjo la remisión de la hipocinesia en los modelos de reserpina y de levodopa. Esta furanocumarina presentó efectos anticatalépticos con la misma dosis. Además, mostró tener actividad inhibitoria selectiva sobre la MAO B, así como efectos antioxidantes. Conclusión. Los resultados evidenciaron propiedades interesantes del compuesto FCS005. Es importante continuar investigando esta molécula porque puede ser un potencial agente antiparkinsoniano.


Asunto(s)
Animales , Masculino , Ratones , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Inhibidores de la Monoaminooxidasa/uso terapéutico , Antiparkinsonianos/uso terapéutico , Enfermedad de Parkinson Secundaria/inducido químicamente , Reserpina/administración & dosificación , Carbidopa/administración & dosificación , Catalepsia/inducido químicamente , Levodopa/administración & dosificación , Cumarinas , Modelos Animales de Enfermedad , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Haloperidol , Locomoción/efectos de los fármacos , Ratones Endogámicos ICR , Inhibidores de la Monoaminooxidasa/administración & dosificación , Antiparkinsonianos/administración & dosificación
10.
Drug Deliv Transl Res ; 9(5): 891-905, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30877626

RESUMEN

Rasagiline mesylate is an irreversible MAO-B inhibitor which requires daily oral administration for treatment of Parkinson's disease due to its short half-life. Patients with Parkinson's disease also develop dysphagia, i.e., difficulty in swallowing. Encapsulating rasagiline in polycaprolactone microspheres can alleviate the problem of daily oral administration by prolonging drug release from polymeric microspheres for 1 month by single subcutaneous administration. Polycaprolactone shows absence of any acidic environment generation during its degradation in body which is its advantage over poly (lactic-co-glycolic) acid. Exploiting pH-based solubility of rasagiline mesylate pH changes during microencapsulation process was performed to fabricate rasagiline mesylate-loaded polycaprolactone microspheres. Particle size analysis of microspheres showed mean particle size range of 24.18-47.87 µm. Scanning electron micrographs revealed spherical non-porous particles with small pits and depressions on the surface. In vitro release studies of formulations were performed to get an idea about in vivo behavior of prepared formulations. Stereotaxic rotenone model was used to study in vivo efficacy of formulation in rats. Selected formulation significantly (p < 0.05) improved various behavioral (locomotor activity, grip strength, etc.) and biochemical (lipid peroxidation, reduced glutathione, etc.) changes. Polymeric microspheres showed robust effect on all outcomes assessed with non-significant difference between daily administration of rasagiline mesylate solution and drug-loaded polymeric microspheres administered once in a month. With prepared controlled release injectable once a month, administration is required making it an interesting and convenient approach in treatment of Parkinson's disease with dysphagia. Patient compliant system can be achieved by exploiting this approach for future use.


Asunto(s)
Indanos/administración & dosificación , Microesferas , Inhibidores de la Monoaminooxidasa/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Trastornos Parkinsonianos/tratamiento farmacológico , Poliésteres/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Preparaciones de Acción Retardada/administración & dosificación , Liberación de Fármacos , Glutatión/metabolismo , Concentración de Iones de Hidrógeno , Indanos/química , Inyecciones Subcutáneas , Peroxidación de Lípido/efectos de los fármacos , Locomoción , Masculino , Inhibidores de la Monoaminooxidasa/química , Fármacos Neuroprotectores/química , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/fisiopatología , Poliésteres/química , Ratas Sprague-Dawley , Rotenona , Superóxido Dismutasa/metabolismo
11.
Drug Test Anal ; 11(6): 898-905, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30614204

RESUMEN

BACKGROUND: Chiral analysis is a crucial way to differentiate selegiline (SG) intake from drug abuse. Oral fluid (OF) has been successfully used as an alternative matrix for blood testing in several pharmacokinetic studies. OBJECTIVE: The aim of this study is to describe the pharmacokinetics of SG and its main metabolites in OF after a single oral administration of SG which is meaningful for results interpretation in forensic analysis. METHODS: Ten milligrams of SG were orally administered to 8 volunteers, and OF samples were collected for up to 96 hours by having participants spit into polypropylene tubes without stimulation. These samples were submitted to liquid-liquid extraction before analysis by liquid chromatography-tandem mass spectrometry operating in positive ion multiple-reaction monitoring mode. RESULTS AND CONCLUSIONS: After oral administration, each analyte could be detected in OF specimens from all volunteers with an initial detection time of 0.50 hours. The Cmax values of SG, R-MA, R-AM and DM-SG were 50.93-992.67 ng/mL, 29.78-653.64 ng/mL, 8.22-150.15 ng/mL, and 4.34-16.25 ng/mL, respectively, at 0.5 hours, 1-11 hours, 1.5-11 hours, and 0.5-6 hours post dose. The times when the compounds were last determined in OF were 5-24 hours for SG, 52-96 hours for R-MA, 31-96 hours for R-AM, and 13-31 hours for DM-SG after oral administration. There is a period of time in OF in which only MA and AM are present without SG and DM-SG after a single dose of SG. The pharmacokinetic data could provide supplementary interpretation for OF tests in forensic science and drug treatment programs.


Asunto(s)
Anfetamina/farmacocinética , Anfetaminas/farmacocinética , Metanfetamina/farmacocinética , Inhibidores de la Monoaminooxidasa/farmacocinética , Saliva/metabolismo , Selegilina/farmacocinética , Administración Oral , Anfetamina/metabolismo , Anfetaminas/metabolismo , Humanos , Extracción Líquido-Líquido , Metanfetamina/metabolismo , Inhibidores de la Monoaminooxidasa/administración & dosificación , Inhibidores de la Monoaminooxidasa/metabolismo , Selegilina/administración & dosificación , Selegilina/metabolismo , Detección de Abuso de Sustancias
12.
J Control Release ; 279: 234-242, 2018 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-29679664

RESUMEN

Prostate cancer (PCa) is the most frequent malignant cancer among men in the USA, leading to substantial morbidity and mortality, while the existing treatments have restricted therapeutic benefits for patients with hormone-refractory PCa (HRPC) and metastatic PCa. Recent studies show that advanced PCa exhibits an increase in the expression of monoamine oxidase A (MAOA) which is a mitochondria enzyme, and MAOA activity inhibition could restrict metastasis and extend mice survival in PCa xenografts. These findings suggest MAOA can be a potential target to treat PCa. For this reason, we identify and synthesize a near-infrared fluorescence (NIRF) heptamethine dye­MAOA inhibitor conjugate (NIR-INH) for simultaneous PCa imaging, targeting and therapy. The conjugate combines a NIRF dye for mitochondria targeting with the MAOA inhibitor isoniazid (INH). NIR-INH exhibits specific targeting in PCa xenografts and markedly inhibited tumor growth. Furthermore, there is no obvious toxicity with NIR-INH treatment, which is a remarkable superiority towards traditional chemotherapy. These results indicate that NIR-INH has PCa targeting, imaging and high anticancer effectiveness, suggesting it is a potentially valuable image-guided anti-tumor strategy.


Asunto(s)
Colorantes Fluorescentes/química , Isoniazida/administración & dosificación , Inhibidores de la Monoaminooxidasa/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Humanos , Isoniazida/química , Isoniazida/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/metabolismo , Inhibidores de la Monoaminooxidasa/química , Inhibidores de la Monoaminooxidasa/farmacología , Células PC-3 , Neoplasias de la Próstata/patología , Espectroscopía Infrarroja Corta , Ensayos Antitumor por Modelo de Xenoinjerto
13.
ACS Chem Neurosci ; 8(12): 2746-2758, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28857544

RESUMEN

Initial work in Drosophila and mice demonstrated that the transcription factor cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) is a master control gene for memory formation. The relationship between CREB and memory has also been found to be true in other species, including aplysia and rats. It is thus well-established that CREB activation plays a central role in memory enhancement and that CREB is activated during memory formation. On the basis of these findings, a phenotypic high-throughput screening campaign utilizing a CRE-luciferase (CRE-Luci) SK-N-MC cell line was performed to identify compounds that enhance transcriptional activation of the CRE promoter with a suboptimal dose of forskolin. A number of small-molecule hits of unknown mechanisms of action were identified in the screening campaign, including HT-0411. Follow-up studies suggested that the CREB activation by HT-0411 is attributed to its specific and selective inhibition of monoamine oxidase B (MAO-B). Further, HT-0411 was shown to improve 24 h memory in rodents in a contextual fear conditioning model. This report describes the lead optimization of a series of 5-(1-methyl-5-(trifluoromethyl)-1H-pyrazol-3-yl) thiophene-2-carboxamides that were identified as novel, potent, and selective inhibitors of MAO-B. Extensive SAR studies and in vivo behavioral evaluations of this and other related analogue series identified a number of potential clinical development candidates; ultimately, compound 8f was identified as a candidate molecule with high selectivity toward MAO-B (29-56 nM) over MAO-A (19% inhibition at a screening concentration of 50 µM), an excellent profile against a panel of other enzymes and receptors, good pharmacokinetic properties in rodents and dogs, and efficacy in multiple rodent memory models.


Asunto(s)
Ácidos Carboxílicos/administración & dosificación , Ácidos Carboxílicos/química , Cognición/efectos de los fármacos , Memoria/efectos de los fármacos , Monoaminooxidasa/metabolismo , Nootrópicos/administración & dosificación , Nootrópicos/química , Amidas/administración & dosificación , Amidas/química , Animales , Cognición/fisiología , Relación Dosis-Respuesta a Droga , Masculino , Memoria/fisiología , Ratones Endogámicos C57BL , Inhibidores de la Monoaminooxidasa/administración & dosificación , Inhibidores de la Monoaminooxidasa/química , Ratas , Resultado del Tratamiento
14.
Neuropharmacology ; 126: 12-24, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28807675

RESUMEN

We have previously demonstrated the unexpected neuroprotection of the anti-cancer agent SU4312 in cellular models associated with Parkinson's disease (PD). However, the precise mechanisms underlying its neuroprotection are still unknown, and the effects of SU4312 on rodent models of PD have not been characterized. In the current study, we found that the protection of SU4312 against 1-methyl-4-phenylpyridinium ion (MPP+)-induced neurotoxicity in PC12 cells was achieved through the activation of transcription factor myocyte enhancer factor 2D (MEF2D), as evidenced by the fact that SU4312 stimulated myocyte enhancer factor 2 (MEF2) transcriptional activity and prevented the inhibition of MEF2D protein expression caused by MPP+, and that short hairpin RNA (ShRNA)-mediated knockdown of MEF2D significantly abolished the neuroprotection of SU4312. Additionally, Western blotting analysis revealed that SU4312 potentiated pro-survival PI3-K/Akt pathway to down-regulate MEF2D inhibitor glycogen synthase kinase-3beta (GSK3ß). Furthermore, using the in vivo PD model of C57BL/6 mice insulted with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), we found that intragastrical administration of SU4312 (0.2 and 1 mg/kg) greatly ameliorated Parkinsonian motor defects, and restored protein levels of MEF2D, phosphorylated-Ser473-Akt and phosphorylated-Ser9-GSK3ß. Meanwhile, SU4312 effectively reversed the decrease in protein expression of tyrosine hydroxylase in substantia nigra pars compacta dopaminergic neurons, inhibited oxidative stress, maintained mitochondrial biogenesis and partially prevented the depletion of dopamine and its metabolites. Very encouragingly, SU4312 was able to selectively inhibit monoamine oxidase-B (MAO-B) activity both in vitro and in vivo, with an IC50 value of 0.2 µM. These findings suggest that SU4312 provides therapeutic benefits in cellular and animal models of PD, possibly through multiple mechanisms including enhancement of MEF2D through the activation of PI3-K/Akt pathway, maintenance of mitochondrial biogenesis and inhibition of MAO-B activity. SU4312 thus may be an effective drug candidate for the prevention or even modification of the pathological processes of PD.


Asunto(s)
Antiparkinsonianos/administración & dosificación , Indoles/administración & dosificación , Intoxicación por MPTP/metabolismo , Monoaminooxidasa/metabolismo , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Hueso Cortical/efectos de los fármacos , Hueso Cortical/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Técnicas In Vitro , Factores de Transcripción MEF2/metabolismo , Intoxicación por MPTP/tratamiento farmacológico , Masculino , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Inhibidores de la Monoaminooxidasa/administración & dosificación , Células PC12 , Ratas , Transducción de Señal/efectos de los fármacos
15.
Life Sci ; 182: 57-64, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28623006

RESUMEN

AIMS: First proof to show that (-)-deprenyl/selegiline (DEP), the first selective inhibitor of MAO-B, later identified as the first ß-phenylethylamine (PEA)-derived synthetic catecholaminergic activity enhancer (CAE) substance and (2R)-1-(1-benzofuran-2-yl)-N-propylpentane-2-amine (BPAP), the tryptamine-derived presently known most potent, selective, synthetic enhancer substance, are specific markers of unknown enhancer-sensitive brain regulations. MAIN METHODS: Longevity study disclosing the operation of tumor-manifestation-suppressing (TMS) regulation in rat brain. Immonohistochemical identification of a fibromyxosarcoma in rats. Experiments with human medulloblastoma cell lines. Analysis of the mechanism of action of enhancer substances. KEY FINDINGS: Whereas 20/40 saline-treated rats manifested a fibromyxosarcoma, in groups of rats treated with 0.001mg/kg DEP: 15/40 rats; with 0.1mg/kg DEP: 11/40 rats (P<0.01); with 0.0001mg/kg BPAP: 8/40 rats (P<0.001); with 0.05mg/kg BPAP: 7/40 rats (P<0.01) manifested the tumor. Experiments with human medulloblastoma cell lines, HTB-186 (Daoy); UW-228-2, showed that BPAP was devoid of direct cytotoxic effect on tumor cells, and did not alter the direct cytotoxic effectiveness of temozolomide, cisplatin, etoposide, or vincristine. Interaction with distinct sites on vesicular monoamine-transporter-2 (VMAT2) is the main mechanism of action of the enhancer substances which clarifies the highly characteristic bi-modal, bell-shaped concentration-effect curves of DEP and BPAP. SIGNIFICANCE: Considering of the safeness of the enhancer substances and the finding that DEP and BPAP, specific markers of unknown enhancer sensitive brain regulations, detected the operation of an enhancer-sensitive TMS-regulation in rat brain, it seems reasonable to test in humans low dose DEP or BPAP treatment against the spreading of a malignant tumor.


Asunto(s)
Benzofuranos/farmacología , Encéfalo/efectos de los fármacos , Longevidad/efectos de los fármacos , Inhibidores de la Monoaminooxidasa/farmacología , Selegilina/farmacología , Animales , Antineoplásicos/farmacología , Benzofuranos/administración & dosificación , Encéfalo/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Fibrosarcoma/prevención & control , Humanos , Masculino , Meduloblastoma/tratamiento farmacológico , Inhibidores de la Monoaminooxidasa/administración & dosificación , Ratas , Ratas Wistar , Selegilina/administración & dosificación
16.
Neuropharmacology ; 113(Pt A): 445-456, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27984078

RESUMEN

Previous study demonstrated that the novel multitarget compound, MT-031 preserved in one molecule entity the beneficial properties of its parent drugs, rasagiline and rivastigmine, and exerted high dual potencies of monoamine oxidase-A (MAO-A) and cholinesterase (ChE) inhibition in acute-treated mice and neuroprotective effects against H2O2-induced neurotoxicity in human neuroblastoma SH-SY5Y cells. The present study aimed to further investigate the anti-inflammatory and protective effects of MT-031 in scopolamine mouse model and inflammatory cell cultures. Our findings demonstrated that once daily chronic administration of MT-031 (5-10 mg/kg) to mice antagonized scopolamine-induced memory and cognitive impairments, displayed brain selective MAO-A and AChE/BuChE inhibition, increased the levels of striatal dopamine (DA), serotonin (5-HT) and norepinephrine and prevented the metabolism of DA and 5-HT. In addition, MT-031 upregulated mRNA expression levels of Bcl-2, the neurotrophic factors, (e.g., brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF) and nerve growth factor (NGF)), the antioxidant enzyme catalase and the anti-inflammatory cytokine, neurotrophic tyrosine kinase receptor (Ntrk), and down-regulated the mRNA expression levels of the pro-inflammatory interleukin (IL)-6 in scopolamine-induced mice. In accordance, MT-031 was shown to reduce reactive oxygen species accumulation, increase the levels of anti-inflammatory cytokines, IL-10 and decrease the levels of the pro-inflammatory cytokines, IL-1ß, IL-6, IL-17 and interferon-gamma (IFN-γ) in activated mouse splenocytes and microglial cells. Taken together, these pharmacological properties of MT-031 can be of clinical importance for developing this novel multitarget compound as a novel drug candidate for the treatment of Alzheimer's disease.


Asunto(s)
Antiinflamatorios/administración & dosificación , Butirilcolinesterasa , Inhibidores de la Colinesterasa/administración & dosificación , Inhibidores de la Monoaminooxidasa/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Escopolamina/toxicidad , Acetilcolinesterasa/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Butirilcolinesterasa/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos/métodos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Monoaminooxidasa/metabolismo
17.
Physiol Behav ; 170: 100-105, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28012831

RESUMEN

Depression is a leading cause of disability worldwide. For this reason, the aim of this study was to investigate the possible antidepressant-like activity of 2-benzoyl-4-iodoselenophene (C17H11IOSe), a selenophene compound, in two well-consolidated behavioral assays for screening antidepressant activity (forced swimming test and tail suspension test) in mice. In order to investigate the mechanism of action of C17H11IOSe, it was investigated the activities of cerebral enzymes: monoamine oxidase MAO A and B and Na+, K+ ATPase, and if an inhibitor of serotonin synthesis, p-chlorophenylalanine (pCPA) (100mg/kg) blocks the antidepressant-like effect of C17H11IOSe. Swiss mice received (C17H11IOSe) (5-50mg/kg) or canola oil by the intragastric (i.g.) route before behavioral tests. The results showed that C17H11IOSe at dose range of 5-50mg/kg decreased immobility time in the tail suspension test. In the forced swimming test, C17H11IOSe reduced the immobility time at the doses of 10 and 50mg/kg. C17H11IOSe differently affected the cerebral cortical Na+, K+ ATPase; the effects on this enzyme were dependent of the dose tested. At a dose of 10mg/kg, the compound increased Na+, K+ ATPase activity, while the activity was inhibited at a dose of 50mg/kg. pCPA blocked the antidepressant-like action of C17H11IOSe in mice. Therefore, C17H11IOSe (5-50mg/kg) selectively inhibited MAO-A activity in cerebral cortices of mice. The modulation of serotonergic system contributed to the antidepressant-like action of C17H11IOSe in mice.


Asunto(s)
Antidepresivos/administración & dosificación , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/enzimología , Inhibidores de la Monoaminooxidasa/administración & dosificación , Compuestos de Organoselenio/administración & dosificación , Administración Oral , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Masculino , Ratones , Estructura Molecular , Monoaminooxidasa/metabolismo , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/enzimología
18.
Neuropharmacology ; 109: 376-385, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27318273

RESUMEN

Current novel therapeutic approach suggests that multifunctional compounds with diverse biological properties and a single bioavailability and pharmacokinetic metabolism, will produce higher significant advantages in treatment of neurodegenerative diseases, such as Alzheimer's disease (AD). Based on this rational, a new class of cholinesterase (ChE)-monoamine oxidase (MAO) inhibitors were designed and synthesized by amalgamating the propargyl moiety of the irreversible selective MAO-B inhibitor, neuroprotective/neurorestorative anti-Parkinsonian drug, rasagiline, into the "N-methyl" position of the ChE inhibitor, anti-AD drug rivastigmine. Initially, we examined the MAO and ChE inhibitory effect of these novel compounds, MT series in vitro and in vivo. Among MT series, MT-031 exhibited higher potency as a dual MAO-A and ChE inhibitor compared to other compounds in acute-treated mice. Additionally, MT-031 was found to increase the striatal levels of dopamine (DA), serotonin (5-HT) and norepinephrine (NE), and prevent the metabolism of DA and 5-HT. Finally, we have demonstrated that MT-031 exerted neuroprotective effect against H2O2-induced neurotoxicity and reactive oxygen species generation in human neuroblastoma SH-SY5Y cells. These findings provide evidence that MT-031 is a potent brain permeable novel multifunctional, neuroprotective and MAO-A/ChE inhibitor, preserves in one molecule entity some of the beneficial properties of its parent drugs, rasagiline and rivastigmine, and thus may be indicated as novel therapeutic approach for AD.


Asunto(s)
Enfermedad de Alzheimer , Inhibidores de la Colinesterasa/administración & dosificación , Inhibidores de la Colinesterasa/síntesis química , Diseño de Fármacos , Inhibidores de la Monoaminooxidasa/administración & dosificación , Inhibidores de la Monoaminooxidasa/síntesis química , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
19.
Neuropsychopharmacology ; 41(9): 2335-43, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26955970

RESUMEN

The Food and Drug Administration (FDA) has the authority to regulate cigarette smoke constituents, and a reduction in nicotine content might benefit public health by reducing the prevalence of smoking. Research suggests that cigarette smoke constituents that inhibit monoamine oxidase (MAO) may increase the reinforcing value of low doses of nicotine. The aim of the present experiments was to further characterize the impact of MAO inhibition on the primary reinforcing and reinforcement enhancing effects of nicotine in rats. In a series of experiments, rats responded for intravenous nicotine infusions or a moderately-reinforcing visual stimulus in daily 1-h sessions. Rats received pre-session injections of known MAO inhibitors. The results show that (1) tranylcypromine (TCP), a known MAO inhibitor, increases sensitivity to the primary reinforcing effects of nicotine, shifting the dose-response curve for nicotine to the left, (2) inhibition of MAO-A, but not MAO-B, increases low-dose nicotine self-administration, (3) partial MAO-A inhibition, to the degree observed in chronic cigarette smokers, also increases low-dose nicotine self-administration, and (4) TCP decreases the threshold nicotine dose required for reinforcement enhancement. The results of the present experiments suggest cigarette smoke constituents that inhibit MAO-A, in the range seen in chronic smokers, are likely to increase the primary reinforcing and reinforcement enhancing effects of low doses of nicotine. If the FDA reduces the nicotine content of cigarettes, then variability in constituents that inhibit MAO-A could impact smoking.


Asunto(s)
Condicionamiento Operante/efectos de los fármacos , Inhibidores de la Monoaminooxidasa/administración & dosificación , Nicotina/administración & dosificación , Refuerzo en Psicología , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Relación Dosis-Respuesta a Droga , Masculino , Monoaminooxidasa/metabolismo , Ratas Sprague-Dawley , Autoadministración , Tranilcipromina/administración & dosificación
20.
J Psychopharmacol ; 30(1): 56-62, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26537155

RESUMEN

While nicotine is often associated with the neuropsychological effects of tobacco smoke, the robust monoamine oxidase (MAO) inhibition observed in chronic smokers is also likely to play a role. Electroencephalographically-indexed alterations in baseline neural oscillations by nicotine have previously been reported in both smokers and non-smokers, however, little is known about the effects of MAO inhibition in combination with nicotine on resting state EEG. In a sample of 24 healthy non-smoking males, the effects of 6 mg nicotine gum, as well as MAO-A inhibition via 75 mg moclobemide, were investigated in separate and combined conditions over four separate test sessions. Drug effects were observed in the alpha2, beta2, and theta band frequencies. Nicotine increased alpha2 power, and moclobemide decreased beta2 power. Theta power was decreased most robustly by the combination of both drugs. Therefore, this study demonstrated that the nicotinic and MAO inhibiting properties of tobacco may differentially influence fast-wave oscillations (alpha2 and beta2), while acting in synergy to influence theta oscillations.


Asunto(s)
Moclobemida/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Estudios Cruzados , Método Doble Ciego , Interacciones Farmacológicas , Sinergismo Farmacológico , Electroencefalografía , Humanos , Masculino , Moclobemida/administración & dosificación , Inhibidores de la Monoaminooxidasa/administración & dosificación , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Dispositivos para Dejar de Fumar Tabaco
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA