Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Dokl Biochem Biophys ; 512(1): 284-287, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38093132

RESUMEN

New hybrid structures based on memantine and edaravone molecules, in which the pyrazolone ring and adamantane fragments are linked by an alkyl linker, were synthesized. It was found that, in addition to the ability to block the intrachannel site of NMDA receptors, the new hybrid compounds exhibit the property of blockers of the allosteric site of NMDA receptors, which is not inherent in memantine and edaravone preparations. The most active hit compound was determined, which, along with the properties of a two-site blocker of the NMDA receptor, exhibits a pronounced activity as an inhibitor of lipid peroxidation, similarly to the drug edaravone.


Asunto(s)
Adamantano , Memantina , Memantina/farmacología , Memantina/química , Edaravona , Receptores de N-Metil-D-Aspartato , Adamantano/farmacología
2.
J Microencapsul ; 40(1): 15-28, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36622880

RESUMEN

AIM: To prepare polymer-drug conjugates containing a combination of memantine, tacrine, and E)-N-(3-aminopropyl)cinnamide, promising therapeutics for the treatment of neurodegenerative disorders. METHODS: The conjugates were characterised by 1HNMR, particle size analysis, SEM, LC-MS, TEM/EDX, and XRD, followed by in vitro anti-acetylcholinesterase and drug release studies. RESULTS: 1H NMR analysis revealed successful drug conjugation with drug mass percentages in the range of 1.3-6.0% w/w. The drug release from the conjugates was sustained for 10 h in the range of 20-36%. The conjugates' capability to inhibit acetylcholinesterase (AChE) activity was significant with IC50 values in the range of 13-44.4 µm which was more effective than tacrine (IC50 =1698.8 µm). The docking studies further confirmed that the conjugation of the drugs into the polymer improved their anti-acetylcholinesterase activity. CONCLUSION: The drug release profile, particle sizes, and in vitro studies revealed that the conjugates are promising therapeutics for treating neurodegenerative disorders.


Asunto(s)
Enfermedad de Alzheimer , Sistema de Administración de Fármacos con Nanopartículas , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Inhibidores de la Colinesterasa/química , Memantina/química , Memantina/farmacología , Memantina/uso terapéutico , Tacrina/farmacología , Tacrina/química , Tacrina/uso terapéutico , Sistema de Administración de Fármacos con Nanopartículas/química , Sistema de Administración de Fármacos con Nanopartículas/farmacología , Sistema de Administración de Fármacos con Nanopartículas/uso terapéutico , Polímeros/química , Polímeros/farmacología , Polímeros/uso terapéutico
3.
J Med Chem ; 65(13): 9193-9205, 2022 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-35729801

RESUMEN

Oxidative damage caused by upregulated nitric oxide (NO) plays an important role in the pathogenesis of Alzheimer's disease (AD). Currently, stimulus-triggered theranostic agents have received much attention due to benefits on disease imaging and targeted therapeutic effects. However, the development of a theranostic agent triggered by NO for AD remains unexplored. Herein, through the mechanism analysis of the reaction between a fluorophore of 9,14-diphenyl-9,14-dihydrodibenzo[a,c]phenazine (DPAC) and NO, which we occasionally found and thereafter structure optimization of DPAC, a theranostic agent DPAC-(peg)4-memantine was fabricated. In an AD cellular model, DPAC-(peg)4-memantine exhibits NO sensing ability for AD imaging. Meanwhile, DPAC-(peg)4-memantine shows improved therapeutic by targeted drug release triggered by NO and sustained therapeutic effects owing to the synergetic antioxidative abilities via the anti-AD drug and NO scavenging. This work provides an unprecedented avenue for the studies on not only AD but also other diseases with NO upregulation.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Humanos , Memantina/química , Memantina/uso terapéutico , Óxido Nítrico , Estrés Oxidativo , Medicina de Precisión
4.
Molecules ; 26(18)2021 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-34576998

RESUMEN

A new series of conjugates of aminoadamantane and γ-carboline, which are basic scaffolds of the known neuroactive agents, memantine and dimebon (Latrepirdine) was synthesized and characterized. Conjugates act simultaneously on several biological structures and processes involved in the pathogenesis of Alzheimer's disease and some other neurodegenerative disorders. In particular, these compounds inhibit enzymes of the cholinesterase family, exhibiting higher inhibitory activity against butyrylcholinesterase (BChE), but having almost no effect on the activity of carboxylesterase (anti-target). The compounds serve as NMDA-subtype glutamate receptor ligands, show mitoprotective properties by preventing opening of the mitochondrial permeability transition (MPT) pore, and act as microtubule stabilizers, stimulating the polymerization of tubulin and microtubule-associated proteins. Structure-activity relationships were studied, with particular attention to the effect of the spacer on biological activity. The synthesized conjugates showed new properties compared to their prototypes (memantine and dimebon), including the ability to bind to the ifenprodil-binding site of the NMDA receptor and to occupy the peripheral anionic site of acetylcholinesterase (AChE), which indicates that these compounds can act as blockers of AChE-induced ß-amyloid aggregation. These new attributes of the conjugates represent improvements to the pharmacological profiles of the separate components by conferring the potential to act as neuroprotectants and cognition enhancers with a multifunctional mode of action.


Asunto(s)
Amantadina/química , Amantadina/farmacología , Carbolinas/química , Carbolinas/farmacología , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/farmacología , Acetilcolinesterasa/química , Amantadina/análogos & derivados , Animales , Butirilcolinesterasa/química , Carboxilesterasa/química , Dominio Catalítico , Línea Celular , Inhibidores de la Colinesterasa/síntesis química , Caballos , Humanos , Cinética , Ligandos , Memantina/química , Memantina/farmacología , Necrosis por Permeabilidad de la Transmembrana Mitocondrial/efectos de los fármacos , Simulación del Acoplamiento Molecular , Propidio/química , Ratas , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/metabolismo , Relación Estructura-Actividad , Porcinos , Tubulina (Proteína)/efectos de los fármacos , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología
5.
Int J Biol Macromol ; 190: 660-666, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34508722

RESUMEN

Human transferrin (Tf) is an iron-binding blood plasma glycoprotein that controls free iron in biological fluids. Tf is a liver-produced protein that binds iron very tightly but reversibly and is the most significant iron pool. Memantine is an orally administrative N-methyl-d-aspartate glutamate receptor antagonist used to slow the progression of moderate-to-severe Alzheimer's disease (AD) and dementia. Here, we have investigated the molecular interactions of Memantine with Tf using molecular docking, dynamics simulation and in vitro binding studies. Molecular docking study revealed many close interactions of Memantine towards Tf with an appreciable binding affinity. The docking results were further validated by molecular dynamics (MD) simulation studies, followed by essential dynamics and free energy landscapes analyses. Memantine shows a good binding affinity to the Tf with a binding constant (K) of 105 M-1. Isothermal titration calorimetry (ITC) also advocated the spontaneous binding of memantine to Tf. The study proposed that the Memantine in complex with Tf is stable in the simulated trajectory with minimal structural changes. The study suggested that the Tf-Memantine interactions can be further explored in AD therapy after critical exploration.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Calorimetría , Simulación por Computador , Memantina/metabolismo , Transferrina/metabolismo , Enfermedad de Alzheimer/patología , Fluorescencia , Humanos , Enlace de Hidrógeno , Memantina/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Análisis de Componente Principal , Conformación Proteica , Termodinámica , Transferrina/química
6.
Eur J Med Chem ; 223: 113678, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34218083

RESUMEN

Sepsis, a systemic inflammatory response, caused by pathogenic factors including microorganisms, has high mortality and limited therapeutic approaches. Herein, a new soluble epoxide hydrolase (sEH) inhibitor series comprising a phenyl ring connected to a memantyl moiety via a urea or amide linkage has been designed. A preferential urea pharmacophore that improved the binding properties of the compounds was identified for those series via biochemical assay in vitro and in vivo studies. Molecular docking displayed that 3,5-dimethyl on the adamantyl group in B401 could make van der Waals interactions with residues at a hydrophobic pocket of sEH active site, which might indirectly explain the subnanomolar level activities of memantyl urea derivatives in vitro better than AR-9281. Among them, compound B401 significantly improved the inhibition potency with human and murine sEH IC50 values as 0.4 nM and 0.5 nM, respectively. Although the median survival time of C57BL/6 mice in LPS-induced sepsis model was slightly increased, the survival rate did not reach significant efficacy. Based on safety profile, metabolic stability, pharmacokinetic and in vivo efficacy, B401 demonstrated the proof of potential for this class of memantyl urea-based sEH inhibitors as therapeutic agents in sepsis.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Epóxido Hidrolasas/antagonistas & inhibidores , Memantina/química , Sepsis/tratamiento farmacológico , Urea/análogos & derivados , Animales , Sitios de Unión , Dominio Catalítico , Modelos Animales de Enfermedad , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Epóxido Hidrolasas/metabolismo , Femenino , Humanos , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Ratas , Sepsis/etiología , Sepsis/mortalidad , Relación Estructura-Actividad , Tasa de Supervivencia , Urea/metabolismo , Urea/uso terapéutico
7.
Chem Biol Interact ; 342: 109463, 2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-33831382

RESUMEN

Memantine is the non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist, used in the treatment of Alzheimer's disease. It is also known that memantine pretreatment assured protection of skeletal muscles from poisoning with nerve agents and an interaction between memantine and AChE was proposed. In the study presented we examined interactions of memantine and its main metabolite (1-amino-3-hydroxymethyl-5-methyl adamantine, Mrz 2/373) with AChE in vitro as well as their effect on kinetics of the soman-induced AChE inhibition and aging. The results have shown that memantine and Mrz 2/373 exerted concentration-dependent inhibition of AChE, with Mrz 2/373 being a more potent inhibitor than the parent compound. Addition of soman 7.5 nmol/l induced gradual AChE inhibition that became almost complete after 20 min. Memantine (0.1, 0.5 and 1 mmol/l) and Mrz 2/373 (0.1, 0.5 and 1 mmol/l) concentration-dependently slowed down the AChE inhibition. After 30 min of incubation of AChE with soman, 5 min of aging and 20 min of reactivation by asoxime (HI-6 dichloride), AChE activity was 8.1% in control medium, 30.7% and 41.9% after addition of 1 and 10 mmol/l memantine, and 16.1% after addition of 1 mmol/l Mrz 2/373. It was concluded that it is possible that memantine and Mrz 2/373 can prevent AChE from inhibition by soman, which could, along with known memantine's neuroprotective activity, explain its potent antidotal effect in soman poisoning. The potential effect on aging of the soman-AChE complex warrants further studies.


Asunto(s)
Inhibidores de la Colinesterasa/farmacología , Memantina/farmacología , Soman/farmacología , Animales , Bovinos , Inhibidores de la Colinesterasa/química , Dopaminérgicos/farmacología , Reducción Gradual de Medicamentos , Memantina/química , Memantina/metabolismo , Estructura Molecular , Factores de Tiempo
8.
Eur J Med Chem ; 217: 113338, 2021 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-33744690

RESUMEN

Hurdled and marred by the notorious nature of glioblastomas (GBM) in terms of resistance to therapy and limited drug delivery into the brain, the anti-GBM drug pipeline is required to be loaded with mechanistically diverse agents. The consideration of HDAC inhibition as a prudent approach to circumvent the resistance issue in GBM spurred us to pragmatically design and synthesizes hydroxamic acids endowed with CNS penetrating ability. By virtue of the blood brain barrier permeability (BBB), memantine was envisioned as an appropriate CAP component for the construction of the HDAC inhibitors. Diverse linkers were stapled for the tetheration of the zinc binding motif with the CAP group to pinpoint an appropriate combination (CAP and linker) that could confer inhibitory preference to HDAC6 isoform (overexpressed in GBM). Resultantly, hydroxamic acid 16 was identified as a promising compound that elicited striking antiproliferative effects against Human U87MG GBM cells as well as TMZ-resistant GBM cells and P1S cells, a concurrent chemo radiotherapy (CCRT)-resistant/patient-derived glioma cell line mediated through preferential HDAC6 inhibition (IC50 = 5.42 nM). Furthermore, 16 exerted cell cycle arrest at G2 phase, induced apoptosis in GBM cells at high concentration and exhibited high BBB permeability. To add on, in-vivo study revealed that the administration of compound 16 prolonged the survival of TMZ-resistant U87MG inoculated orthotopic mice. Overall, the cumulative results indicate that 16 is a tractable CNS penetrant preferential HDAC6 inhibitor that might emerge as a potent weapon against GBM.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Diseño de Fármacos , Glioblastoma/tratamiento farmacológico , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Memantina/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glioblastoma/metabolismo , Glioblastoma/patología , Histona Desacetilasa 6/metabolismo , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/química , Humanos , Masculino , Memantina/síntesis química , Memantina/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Simulación del Acoplamiento Molecular , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Relación Estructura-Actividad , Células Tumorales Cultivadas
9.
Anticancer Agents Med Chem ; 21(12): 1575-1580, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33081685

RESUMEN

BACKGROUND: Repurposing drug is an efficient strategy as the drug discovery process is timeconsuming, laborious and costly. Memantine is already used in Alzheimer's disease to prevent neurons from excess glutamate toxicity. As cancer cells benefit from higher amounts of cellular energetics like glucose and glutamine, we used memantine to interfere with the glutamate metabolism in order to restrict cancer cells' glutamine as a source for their growth. OBJECTIVE: To investigate the potential antitumor effect of memantine by reducing glutamate levels in 4T1 mouse breast cancer model. METHODS: 24 Balb/c female mice were subcutaneously inoculated with 4T1 cells. When tumors were palpable, memantine treatment was initiated as 5 and 10 mg/kg daily intraperitoneal injection. Tumor growth was recorded every 2-3 days. Tumor volumes, serum glutamate levels, spleen IL-6 levels, genome-wide DNA methylation levels and GSK3B. pGSK3B protein expressions were measured to enlighten the anticancer mechanism of action for memantine. RESULTS: We found that both two doses (5 and 10mg/kg) decreased tumor growth rates and serum glutamate levels significantly (p<0.05). 10mg/kg treatment increased spleen IL-6 levels (p<0.05) and decreased genomewide DNA methylation levels. Memantine treatment decreased GSK3B protein expression levels in tumor tissue samples. CONCLUSION: To the best of our knowledge, this is the first study that investigates the antitumor activity of memantine in a breast cancer tumor model. Our results suggest a potent anticancer mechanism of the action for memantine. Memantine decreased genome wide methylation and serum glutamate levels that are associated with a poor prognosis. Therefore, Memantine might be used for targeting glutamine metabolism in cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Modelos Animales de Enfermedad , Ácido Glutámico/sangre , Memantina/farmacología , Animales , Antineoplásicos/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Ácido Glutámico/metabolismo , Memantina/química , Ratones , Ratones Endogámicos BALB C , Células Tumorales Cultivadas
10.
Amino Acids ; 52(11-12): 1559-1580, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33191465

RESUMEN

Drug compounds including memantine moieties are an important group of biologically active agents for different pathologies, including the Alzheimer's disease. In the present study, a series of memantine derivatives incorporating amino acid residues have been synthesized and their neuroprotective in vitro evaluation in respect of the Alzheimer's disease, involving the effects on the resistance to Aß toxicity, excitotoxicity, oxidative stress, hypoxia, and neuroinflammation has been studied. The cytotoxicities of the compounds were detected by CPE assay. TC50 and IC50 were determined using Reed and Muench method. Solubility and distribution were measured using a shake-flask method. Permeability of the compounds was studied using Franz diffusion cell and Permeapad™ barrier. These compounds displayed apparent multi-neuroprotective effects against copper-triggered Aß toxicity, glutamate-induced excitotoxicity, and oxidative and hypoxic injuries. They also showed the ability to inhibit the inflammatory cytokine release from the activated microglia and potential anti-neuroinflammatory effects. Especially, two most promising compounds H-4-F-Phe-memantine and H-Tyr-memantine demonstrated the equivalent functional bioactivities in comparison with the positive control memantine hydrochloride. Higher solubility in muriatic buffer than in phosphate buffer was detected. The distribution coefficients showed the optimal lipophilicity for compounds. The presented results propose new class of memantine derivatives as potential drug compounds. Based on the experimental results, the correlations have been obtained between the biological, physicochemical parameters and structural descriptors. The correlation equations have been proposed to predict the properties of new memantine derivatives knowing only the structural formula.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Gripe Humana/tratamiento farmacológico , Memantina/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/toxicidad , Animales , Perros , Ácido Glutámico/metabolismo , Humanos , Gripe Humana/virología , Células de Riñón Canino Madin Darby , Memantina/análogos & derivados , Memantina/química , Fármacos Neuroprotectores/química , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/patogenicidad , Estrés Oxidativo/efectos de los fármacos
11.
Molecules ; 25(17)2020 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-32887400

RESUMEN

Memantine (3,5-dimethyladamantan-1-amine) is an orally active, noncompetitive N-methyl-D-aspartate receptor (NMDAR) antagonist approved for treatment of moderate-to-severe Alzheimer's disease (AD), a neurodegenerative condition characterized by a progressive cognitive decline. Unfortunately, memantine as well as the other class of drugs licensed for AD treatment acting as acetylcholinesterase inhibitors (AChEIs), provide only symptomatic relief. Thus, the urgent need in AD drug development is for disease-modifying therapies that may require approaching targets from more than one path at once or multiple targets simultaneously. Indeed, increasing evidence suggests that the modulation of a single neurotransmitter system represents a reductive approach to face the complexity of AD. Memantine is viewed as a privileged NMDAR-directed structure, and therefore, represents the driving motif in the design of a variety of multi-target directed ligands (MTDLs). In this review, we present selected examples of small molecules recently designed as MTDLs to contrast AD, by combining in a single entity the amantadine core of memantine with the pharmacophoric features of known neuroprotectants, such as antioxidant agents, AChEIs and Aß-aggregation inhibitors.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Memantina/uso terapéutico , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/farmacología , Diseño de Fármacos , Humanos , Memantina/química , Memantina/farmacología , Modelos Moleculares
12.
Int J Mol Sci ; 21(9)2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32380758

RESUMEN

Alzheimer's disease (AD) is the leading cause of dementia worldwide. Even though the number of AD patients is rapidly growing, there is no effective treatment for this neurodegenerative disorder. At present, implementation of effective treatment approaches for AD is vital to meet clinical needs. In AD research, priorities concern the development of disease-modifying therapeutic agents to be used in the early phases of AD and the optimization of the symptomatic treatments predominantly dedicated to the more advanced AD stages. Until now, available therapeutic agents for AD treatment only provide symptomatic treatment. Since AD pathogenesis is multifactorial, use of a multimodal therapeutic intervention addressing several molecular targets of AD-related pathological processes seems to be the most practical approach to modify the course of AD progression. It has been demonstrated through numerous studies, that the clinical efficacy of combination therapy (CT) is higher than that of monotherapy. In case of AD, CT is more effective, mostly when started early, at slowing the rate of cognitive impairment. In this review, we have covered the major studies regarding CT to combat AD pathogenesis. Moreover, we have also highlighted the safety, tolerability, and efficacy of CT in the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/etiología , Animales , Biomarcadores , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Ensayos Clínicos como Asunto , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Dopaminérgicos/química , Dopaminérgicos/farmacología , Dopaminérgicos/uso terapéutico , Quimioterapia Combinada/efectos adversos , Quimioterapia Combinada/métodos , Galantamina/farmacología , Galantamina/uso terapéutico , Humanos , Memantina/química , Memantina/farmacología , Memantina/uso terapéutico
13.
Spectrochim Acta A Mol Biomol Spectrosc ; 227: 117640, 2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-31675657

RESUMEN

Accurate, simple, sensitive, and fast spectrophotometric assay was applied for the quantification of certain anti-Alzheimer drug namely; Memantine hydrochloride, in its bulk and pharmaceutical preparation. The described assay has been established on the reaction between the primary amino moiety of the cited drug and 2,2-dihydroxyindane-1,3-dione reagent in N,N'-dimethylformamide medium in boiling water bath. Which give Ruhemann's purple color that can be determined at λmax = 595 nm. Beer's law has been obeyed within drug concentration range from 10-120 µg per milliliter. Detection limit and quantitation limit have been 1.6 & 4.9 µg per milliliter respectively. Developed procedure has been validated in agreement with International Conference of Horizon recommendations and applied successfully for detection of cited drug in bulk, pharmaceutical dosage form and content uniformity testing.


Asunto(s)
Química Farmacéutica , Indanos/química , Memantina/análisis , Límite de Detección , Memantina/química , Reproducibilidad de los Resultados , Solventes/química , Espectrofotometría Infrarroja , Temperatura
15.
Molecules ; 24(21)2019 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-31684129

RESUMEN

A zone-fluidics (ZF) based automated fluorimetric sensor for the determination of pharmaceutically active adamantine derivatives, i.e., amantadine (AMA), memantine (MEM) and rimantadine (RIM) is reported. Discrete zones of the analytes and reagents (o-phthalaldehyde and N-acetylcysteine) mix and react under stopped-flow conditions to yield fluorescent iso-indole derivatives (λex/ λem = 340/455 nm). The proposed ZF sensor was developed and validated to prove suitable for quality control tests (assay and content uniformity) of commercially available formulations purchased from the Greek market (EU licensed) and from non-EU web-pharmacies at a sampling rate of 16 h-1. Interestingly, a formulation obtained through the internet and produced in a third-non-EU-country (AMA capsules, 100 mg per cap), was found to be out of specifications (mean assay of 85.3%); a validated HPLC method was also applied for confirmatory purposes.


Asunto(s)
Amantadina/aislamiento & purificación , Fluorometría/métodos , Memantina/aislamiento & purificación , Rimantadina/aislamiento & purificación , Amantadina/química , Cromatografía Líquida de Alta Presión , Indicadores y Reactivos/química , Indoles/química , Memantina/química , Microfluídica , Rimantadina/química
16.
Bioconjug Chem ; 30(10): 2573-2583, 2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31553175

RESUMEN

Lower generation PAMAM dendrimers have an immense potential for drug delivery with lower toxicity, but these dendrimers yet need certain basic ameliorations. In this study, the brain delivery potential of the synthesized PAMAM-Lf (lower generation PAMAM and lactoferrin conjugate) loaded with memantine (MEM) was explored and evaluated in vitro and in vivo in the disease-induced mouse model. The developed nanoscaffolds were characterized for size, zeta potential and in vitro release. Increase in the average size from 11.54 ± 0.91 to 131.72 ± 4.73 nm, respectively, was observed for drug-loaded PAMAM (i.e., PAMAM-MEM) and PAMAM-Lf (i.e., MEM-PAMAM-Lf).  Release profile of MEM from MEM-PAMAM-Lf was slow and sustained up to 48 h. In vivo biodistribution in the Sprague-Dawley rat model revealed that the brain uptake of MEM-PAMAM-Lf was significantly higher than that of MEM alone. The behavioral response study in the healthy rats did not result in any significant changes. The in vivo study in an AlCl3-induced Alzheimer's (AD) mice model showed a significant improvement in behavioral responses. Optical density, which reflects the acetylcholinesterase (AChE) activity, was highest in the AL group 0.16 ± 0.01 (higher than the CON group, 0.09 ± 0.02; p < 0.05). No significant suppression of AChE activity was recorded in all the other treated groups. Similarly, the DOPAmine and 3,4 dihydroxyphenylacetic acid (DOPAC) levels were unaffected by the developed formulations. The study reported improved brain bioavailability of MEM in AlCl3-induced Alzheimer's mice leading to improved memory, with the resultant mechanism behind in a descriptive manner. This study is among the preliminary studies reporting the memory improvement aspect of PAMAM-Lf conjugates for MEM in AlCl3-AD induced mice. The formulation developed was beneficial in AD-induced mice and had a significant impact on the memory aspects.


Asunto(s)
Cloruro de Aluminio/efectos adversos , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Dendrímeros/química , Lactoferrina/química , Memantina/química , Memantina/metabolismo , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Dendrímeros/toxicidad , Modelos Animales de Enfermedad , Dopamina/metabolismo , Portadores de Fármacos/química , Portadores de Fármacos/toxicidad , Liberación de Fármacos , Eritrocitos/efectos de los fármacos , Memantina/farmacocinética , Memantina/farmacología , Ratones , Ratas , Distribución Tisular
17.
Int J Biol Macromol ; 140: 140-148, 2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-31415858

RESUMEN

A novel green magnetic molecularly imprinted solid phase extraction (MMI-SPE) for separation of memantine (MEM) from complicated matrices was proposed. The nanomaterial was synthesized via crosslinking of chitosan (CHIT) with [3-(2, 3-epoxypropoxy)-propyl] trimethoxysilane (EPPTMS) in presence of MEM as a template. The nanocomposites, in all steps, were characterized by SEM, FTIR and PXRD techniques. The adsorbed drug was removed from magnetic molecular imprinted polymer (MMIP) cavity by ethanol: acetic acid (8:2, v/v) and then, coupled with sodium 1, 2-naphthoquinone-4-sulphonate (NQS) in iodine/alkaline medium to yield highly fluorescent product, after reduction with potassium borohydride (KBH4). Variables affecting extraction of MEM from imprinted sites and its fluorometric analysis were studied. The linearity was achieved over concentration range of 1.84-95.0 ng mL-1 with LOD of 0.6 ng mL-1. The method was successfully applied for determination of MEM in its pharmaceutical tablets and human serum with recoveries of 100.8 ±â€¯3.0, 97.6 ±â€¯2.9, respectively.


Asunto(s)
Plásticos Biodegradables/química , Quitosano/química , Nanopartículas de Magnetita/química , Memantina/aislamiento & purificación , Suero/química , Agua/química , Tecnología Química Verde , Humanos , Memantina/química , Impresión Molecular
18.
Eur J Med Chem ; 180: 111-120, 2019 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-31301562

RESUMEN

N-methyl-d-aspartate receptors (NMDAR) are critically involved in the pathogenesis of Alzheimer's disease (AD). Acting as an open-channel blocker, the anti-AD drug memantine preferentially targets NMDAR overactivation, which has been proposed to trigger neurotoxic events mediated by amyloid ß peptide (Aß) and oxidative stress. In this study, we applied a multifunctional approach by conjugating memantine to ferulic acid, which is known to protect the brain from Aß neurotoxicity and neuronal death caused by ROS. The most interesting compound (7) behaved, like memantine, as a voltage-dependent antagonist of NMDAR (IC50 = 6.9 µM). In addition, at 10 µM concentration, 7 exerted antioxidant properties both directly and indirectly through the activation of the Nrf-2 pathway in SH-SY5Y cells. At the same concentration, differently from the parent compounds memantine and ferulic acid alone, it was able to modulate Aß production, as revealed by the observed increase of the non-amyloidogenic sAPPα in H4-SW cells. These findings suggest that compound 7 may represent a promising tool for investigating NMDAR-mediated neurotoxic events involving Aß burden and oxidative damage.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Ácidos Cumáricos/farmacología , Memantina/farmacología , Fármacos Neuroprotectores/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Supervivencia Celular/efectos de los fármacos , Ácidos Cumáricos/síntesis química , Ácidos Cumáricos/química , Relación Dosis-Respuesta a Droga , Humanos , Memantina/síntesis química , Memantina/química , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Estrés Oxidativo/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
19.
Br J Pharmacol ; 176(17): 3318-3335, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31180578

RESUMEN

BACKGROUND AND PURPOSE: Cerebral vasospasm and neuronal apoptosis after subarachnoid haemorrhage (SAH) is the major cause of morbidity and mortality in SAH patients. So far, single-target agents have not prevented its occurrence. Memantine, a non-competitive NMDA re3ceptor antagonist, is known to alleviate brain injury and vasospasm in experimental models of SAH. Impairment of NO availability also contributes to vasospasm. Recently, we designed and synthesized a memantine nitrate MN-08, which has potent dual functions: neuroprotection and vasodilation. Here, we have tested the therapeutic effects of MN-08 in animal models of SAH. EXPERIMENTAL APPROACH: Binding to NMDA receptors (expressed in HEK293 cells), NO release and vasodilator effects of MN-08 were assessed in vitro. Therapeutic effects of MN-08 were investigated in vivo, using rat and rabbit SAH models. KEY RESULTS: MN-08 bound to the NMDA receptor, slowly releasing NO in vitro and in vivo. Consequently, MN-08 relaxed the pre-contracted middle cerebral artery ex vivo and increased blood flow velocity in small vessels of the mouse cerebral cortex. It did not, however, lower systemic blood pressure. In an endovascular perforation rat model of SAH, MN-08 improved the neurological scores and ameliorated cerebral vasospasm. Moreover, MN-08 also alleviated cerebral vasospasm in a cisterna magna single-injection model in rabbits. MN-08 attenuated neural cell apoptosis in both rat and rabbit models of SAH. Importantly, the therapeutic benefit of MN-08 was greater than that of memantine. CONCLUSION AND IMPLICATIONS: MN-08 has neuroprotective potential and can ameliorate vasospasm in experimental SAH models.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Modelos Animales de Enfermedad , Memantina/uso terapéutico , Nitratos/uso terapéutico , Hemorragia Subaracnoidea/tratamiento farmacológico , Vasodilatadores/uso terapéutico , Vasoespasmo Intracraneal/tratamiento farmacológico , Animales , Lesiones Encefálicas/inducido químicamente , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Masculino , Memantina/administración & dosificación , Memantina/química , Ratones , Ratones Endogámicos C57BL , Nimodipina , Nitratos/administración & dosificación , Nitratos/química , Óxido Nítrico/análisis , Conejos , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Hemorragia Subaracnoidea/inducido químicamente , Vasodilatadores/administración & dosificación , Vasodilatadores/química , Vasoespasmo Intracraneal/inducido químicamente
20.
Molecules ; 24(7)2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30978991

RESUMEN

Harmine (HAR) is a beta-carboline alkaloid widely distributed in nature. It exhibits psychopharmacological effects of improving learning and memory. However, excessive dose of HAR can cause central tremor toxicity, which may be related to the glutamate system. Memantine (MEM) is a non-competitive N-methyl-d-aspartate receptor antagonist. It can be used for the treatment of Alzheimer's disease and also can block the neurotoxicity caused by glutamate. Therefore, combination of HAR and MEM would be meaningful and the pharmacokinetics investigation of HAR and MEM in combination is necessary. A ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method was established and validated for the simultaneous quantitative determination of MEM, HAR and harmol (HOL), a main metabolite of HAR, in rat plasma after oral administration of HAR and MEM in combination (5.0 mg/kg of MEM combined with 20.0, 40.0, 80.0 mg/kg of HAR). The contents of HAR and HOL were determined after oral administration of HAR (20.0, 40.0 and 80.0 mg/kg), and the content of MEM was determined after oral administration of MEM (5.0 mg/kg). Blood samples were collected from each rat at 0 (pre-dose), 0.08, 0.17, 0.25, 0.33, 0.50, 0.75, 1.0, 2.0, 4.0, 8.0, 12.0 and 24.0 h after administration. The maximum peak concentration (Cmax) of MEM was obviously decreased, and the area under the plasma concentration versus time curve from zero to time t (AUC(0-t)) and mean residence time (MRT) were significantly increased after combination with HAR. The Cmax and AUC(0-t) of HAR and its metabolite HOL were increased after combination with MEM. These findings suggested that co-administration of HAR and MEM could extend their residence time in rats, and then might increase the efficacy for treatment of Alzheimer's disease. Therefore, this study will provide a basis for the rational combined application of HAR and MEM.


Asunto(s)
Harmina/química , Memantina/química , Farmacocinética , Receptores de N-Metil-D-Aspartato/química , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/farmacocinética , Interacciones Farmacológicas , Harmina/farmacocinética , Humanos , Memantina/farmacocinética , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA