Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurol Res ; 46(3): 261-271, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38122814

RESUMO

INTRODUCTION: The neuroinflammatory response was seen to impact the formation of phosphorylated tau protein in Alzheimer's disease (AD). This study aims to investigate the molecular mechanism of minocycline in reducing phosphorylated tau protein formation in the hippocampus of lipopolysaccharide (LPS)-induced rats. METHODS: Fifty adult male Sprague Dawley (SD) rats were randomly allocated to 1 of 5 groups: control, LPS (5 mg/kg), LPS + minocycline (25 mg/kg), LPS + minocycline (50 mg/kg) and LPS + memantine (10 mg/kg). Minocycline and memantine were administered intraperitoneally (i.p) for two weeks, and LPS was injected i.p. once on day 5. ELISA was used to determine the level of phosphorylated tau protein in SD rats' hippocampal tissue. The density and expression of Toll-like receptor-4 (TLR-4), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-кß), tumour necrosis factor-alpha (TNF-α), and cyclooxygenase (COX)-2 were determined using Western blot and immunohistochemistry. RESULTS: Minocycline, like memantine, prevented LPS-induced increasein phosphorylated tau protein level suggested via reduced density and expression of TLR-4, NF-кß, TNF-αand COX-2 proteins in rat hippocampal tissue. Interestingly, higher doses were shown to be more neuroprotective than lower doses. CONCLUSION: This study suggests that minocycline suppresses the neuroinflammation signalling pathway and decreased phosphorylated tau protein formation induced by LPS in a dose-dependent manner. Minocycline can be used as a preventative and therapeutic drug for neuroinflammatory diseases such as AD.


Assuntos
Doença de Alzheimer , Minociclina , Ratos , Animais , Masculino , Minociclina/farmacologia , Minociclina/uso terapêutico , Proteínas tau/metabolismo , Doenças Neuroinflamatórias , Lipopolissacarídeos , Ratos Sprague-Dawley , Memantina/farmacologia , Memantina/metabolismo , Receptor 4 Toll-Like/metabolismo , Hipocampo/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , NF-kappa B/metabolismo
2.
Brain Res ; 1821: 148577, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-37716463

RESUMO

GPR39 is a receptor involved in zincergic neurotransmission, and its role in regulating psychological functions is an active area of research. The purported roles of GPR39 at the cellular level include regulation of inflammatory and oxidative stress response, and modulation of GABAergic and endocannabinoid neurotransmission. GPR39 knock-out (KO) mice exhibit episodic-like and spatial memory (ELM and SM, respectively) deficits throughout their lifetime, and are similar in that respect to senescent wild-type (WT) conspecifics. Since a role for zinc has been postulated in neurodegenerative disorders, in this study we investigated the possibility of a pharmacological rescue of both types of declarative memory with memantine - a noncompetitive NMDAR antagonist used for slowing down dementia; or, a putative GPR39 agonist - TC-G 1008. First, we tested adult WT and GPR39KO male mice under acute 5 mg/kg memantine or vehicle treatment in an object recognition task designed to simultaneously probe the "what?", "where?" and "when?" components of ELM. Next, we investigated the impact of chronic memantine or TC-G 1008 on ELM and SM (Morris water maze, MWM) in both WT and GPR39KO mice. Following chronic experiments, we assessed with qRT-PCR hippocampal gene expression of targets previously associated with GPR39. We report: no effects of acute memantine on ELM; a tendency to improve the "where?" component of ELM in both WT and GPR39 KO mice following 12 days of memantine; and, a disruption of SM in GPR39KO mice after 24 days of memantine treatment. The latter result was associated with upregulation of Htr1a hippocampal expression.


Assuntos
Memantina , Zinco , Animais , Masculino , Camundongos , Expressão Gênica , Hipocampo/metabolismo , Memantina/farmacologia , Memantina/metabolismo , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Memória Espacial , Zinco/metabolismo
3.
Life Sci ; 323: 121695, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37062446

RESUMO

AIMS: Long-term neuroinflammation and brain dysfunction have frequently been reported in sepsis survivors. In this study, the protective effect of memantine (an NMDA receptor antagonist) on the long-term consequences of sepsis on the brain was investigated in mice. MATERIALS AND METHODS: Eighty-five male C57 mice were included. Memantine was administrated through gavage at 5, 10, and 20 mg/kg three days before sepsis and continued for three days after sepsis induction. Sepsis was induced by intraperitoneal injection of 5 mg/kg LPS. A cohort of mice was sacrificed on the 4th day post sepsis to measure NF-κB, TNF-α, and IL-1ß mRNA expression and oxidative stress markers in the brain. The second cohort was used for behavioral tests one month after sepsis induction and then sacrificed for oxidative stress markers and acetylcholinesterase (AChE) activity measurement. KEY FINDINGS: MDA levels and mRNA expression of NF-κB, TNF-α, and IL-1ß ameliorated by memantine at the early days of sepsis induction, and total thiol content and SOD activity were increased. Post-septic mice showed significant disruption of recognition memory in novel object recognition (NOR) and depressive and anxiety-like behaviors in tail suspension test, elevated plus maze (EPM), and open field tests one month after sepsis. Memantine at 10 and 20 mg/kg dose-dependently ameliorated behavioral abnormalities, reduced AChE activity and MDA levels, and enhanced SOD activity and thiol content one month after sepsis. SIGNIFICANCE: These findings suggest that early treatment of septic mice with memantine could ameliorate brain inflammation and oxidative damage and prevent long-term behavioral consequences of sepsis.


Assuntos
Memantina , Sepse , Camundongos , Masculino , Animais , Memantina/farmacologia , Memantina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Acetilcolinesterase/metabolismo , Encéfalo/metabolismo , Sepse/complicações , Sepse/tratamento farmacológico , Sepse/metabolismo , RNA Mensageiro/metabolismo , Superóxido Dismutase/metabolismo
4.
Life Sci ; 319: 121509, 2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-36822316

RESUMO

AIMS: This study investigated the possible hepatoprotective effects of memantine, compared to pioglitazone, in rat steatohepatitis, emphasizing its role in modulating hepatic autophagy. MAIN METHODS: Metabolic syndrome (MetS) was provoked in adult male Wistar rats by a high fructose/fat/salt regimen for eight weeks. Then, rats were administered either memantine or pioglitazone daily for 10 weeks (both at 20 mg/kg, orally). An oral glucose tolerance test (OGTT) was done at the end of the study, and serum liver enzymes, lipids, and fasting blood glucose were measured. Also, hepatic contents of inflammatory, oxidative, and autophagy markers were quantified. Additionally, histopathological examinations of general hepatic structure and glycogen content were performed. KEY FINDINGS: Compared to the MetS rats, memantine normalized fasting serum insulin, Homeostatic Model Assessment (HOMA-IR), serum lipids, and liver enzymes (ALT and AST). Memantine also markedly reduced hepatic inflammatory markers; NF-κB and TNF-α. In addition, hepatic NRF2 and GSH were augmented, while hepatic MDA was reduced by memantine. Interestingly, livers of the memantine group showed elevated Beclin1 and LC3 and reduced p62 contents compared to the MetS group indicating that memantine preserved hepatic autophagy. Histopathological examination revealed that memantine ameliorated hepatic steatosis and inflammation. Pioglitazone also mitigated most of the steatohepatitis-related changes, however, memantine was more effective in most of the studied parameters. SIGNIFICANCE: The hepatoprotective effect of memantine against steatohepatitis is mediated, at least partly, through conserving hepatic autophagy along with anti-inflammatory, antioxidant, and anti-fibrotic effects.


Assuntos
Fígado Gorduroso , Síndrome Metabólica , Masculino , Ratos , Animais , Síndrome Metabólica/complicações , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/metabolismo , Memantina/farmacologia , Memantina/uso terapêutico , Memantina/metabolismo , Pioglitazona/farmacologia , Pioglitazona/metabolismo , Ratos Wistar , Fígado/metabolismo , Fígado Gorduroso/metabolismo , Inflamação/patologia , Fibrose , Lipídeos , Autofagia , Estresse Oxidativo
5.
Sci Rep ; 12(1): 17908, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36284170

RESUMO

In Alzheimer's disease pathology, several neuronal processes are dysregulated by excitotoxicity including neuroinflammation and oxidative stress (OS). New therapeutic agents capable of modulating such processes are needed to foster neuroprotection. Here, the effect of an optimised NMDA receptor antagonist, UB-ALT-EV and memantine, as a gold standard, have been evaluated in 5XFAD mice. Following treatment with UB-ALT-EV, nor memantine, changes in the calcineurin (CaN)/NFAT pathway were detected. UB-ALT-EV increased neurotropic factors (Bdnf, Vgf and Ngf) gene expression. Treatments reduced astrocytic and microglial reactivity as revealed by glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (Iba-1) quantification. Interestingly, only UB-ALT-EV was able to reduce gene expression of Trem2, a marker of microglial activation and NF-κB. Pro-inflammatory cytokines Il-1ß, Ifn-γ, Ccl2 and Ccl3 were down-regulated in UB-ALT-EV-treated mice but not in memantine-treated mice. Interestingly, the anti-inflammatory markers of the M2-migroglial phenotype, chitinase-like 3 (Ym1) and Arginase-1 (Arg1), were up-regulated after treatment with UB-ALT-EV. Since iNOS gene expression decreased after UB-ALT-EV treatment, a qPCR array containing 84 OS-related genes was performed. We found changes in Il-19, Il-22, Gpx6, Ncf1, Aox1 and Vim gene expression after UB-ALT-EV. Hence, our results reveal a robust effect on neuroinflammation and OS processes after UB-ALT-EV treatment, surpassing the memantine effect in 5XFAD.


Assuntos
Doença de Alzheimer , Quitinases , Animais , Camundongos , Doença de Alzheimer/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Arginase/metabolismo , Memantina/metabolismo , NF-kappa B/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Calcineurina/metabolismo , Cálcio/metabolismo , Fator de Crescimento Neural/metabolismo , Modelos Animais de Doenças , Microglia/metabolismo , Estresse Oxidativo , Citocinas/metabolismo , Quitinases/metabolismo
6.
Toxicol In Vitro ; 84: 105453, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35944748

RESUMO

Memantine is a non-competitive antagonist with a moderate affinity to the N-methyl-d-Aspartate (NMDA) receptor. The present study assessed memantine's neuroprotective activity using electrophysiology of ex-vivo hippocampal slices. Interestingly, a nicotinic component was necessary for memantine's neuroprotection (NP). Memantine demonstrated a bell-shaped dose-response curve of NP against NMDA. Memantine was neuroprotective at concentrations below 3 µM, but the NP declined at higher concentrations (>3 µM) when memantine inhibits the NMDA receptor. Additional evidence that memantine NP is mediated by an alternate mechanism independent of the inhibition of the NMDA receptor is supported by its ability to protect neurons when applied before or after the NMDA insult and in the presence of D(-)-2-Amino-5-phosphonopentanoic acid (APV), the standard NMDA receptor inhibitor. We found several similarities between the memantine NP mechanism and the neuroprotective nicotinic drug, the 4R cembranoid. Memantine's NP requires the release of acetylcholine, the activation of α4ß2, and is independent of MEK/MAPK signaling. Both 4R and memantine require the activation of PI3K/AKT for NP against NMDA-mediated excitotoxicity, although at different concentrations. In conclusion, our studies show memantine is neuroprotective through a nicotinic pathway, similar to the nicotinic drug 4R. This information leads to a better understanding of memantine's mechanisms of action and explains its dose-dependent effectiveness in Alzheimer's and other neurological disorders.


Assuntos
Memantina , Fármacos Neuroprotetores , Hipocampo/metabolismo , Memantina/metabolismo , Memantina/farmacologia , N-Metilaspartato/toxicidade , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Nicotina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
7.
Biosens Bioelectron ; 216: 114619, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-35986984

RESUMO

Olfactory dysfunction is an early symptom of neurodegenerative disease. Amyloid-beta oligomers (AßOs), the pathologic protein of Alzheimer's disease (AD), have been confirmed to be firstly deposited in olfactory bulb (OB), causing smell to malfunction. However, the detailed mechanisms underlying pathogenic nature of AßOs-induced olfactory neuronal degeneration in AD are not completely realized. Here, an early-stage olfactory dysfunction pathological model of AD in vitro based on biomimetic OB neuronal network chip was established for dynamic multi-site detection of neuronal electrical activity and network connection. We found both spike firing and correlation of overall neuronal network change regularly displayed gradually active state and then rapidly decay state after AßOs induction. Moreover, MK-801 and memantine were administrated at early-stage to detect alteration of OB neurons simulating nasal administration for AD treatment, which showed an almost recovery through the intermittent firing pattern. Together, this neuronal network-on-chip has revealed synaptic impairment and network neurodegeneration of olfactory dysfunction in AD, providing potential mechanisms information for early-stage progressive olfactory amyloidogenic pathology.


Assuntos
Doença de Alzheimer , Técnicas Biossensoriais , Doenças Neurodegenerativas , Transtornos do Olfato , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Biomimética , Maleato de Dizocilpina/metabolismo , Humanos , Memantina/metabolismo , Neurônios/metabolismo , Transtornos do Olfato/etiologia , Transtornos do Olfato/metabolismo , Transtornos do Olfato/patologia , Bulbo Olfatório , Olfato
8.
Eur Rev Med Pharmacol Sci ; 26(12): 4409-4419, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35776042

RESUMO

OBJECTIVE: The study aimed to examine the effects of two drugs, an acetylcholinesterase inhibitor (AChEI) and an N-methyl-D-aspartate receptor (NMDAR) antagonist, on degenerated annulus fibrosus (AF) and nucleus pulposus (NP) cells and the extracellular matrix (ECM) structure in vitro. PATIENTS AND METHODS: Tissue samples were obtained from patients with intervertebral disc herniation (four males and four females; classified as Pfirmann stage IV) and used to prepare cell cultures. Untreated cell culture samples served as the control group. Study group samples were treated with donepezil, memantine or a combination of the two drugs. Cell viability, toxicity and proliferation were evaluated in all groups. Western blotting was used to examine changes in protein expression of signal transducer and activator of transcription 3 (STAT3), phospho-STAT3 (ser727), hypoxia-inducible factor (HIF)-1 alpha (HIF-1α) and nucleotide-binding oligomerisation domain (NOD) leucine-rich repeat (LRR)-containing proteins (NLR) family pyrin domain containing 3 (NLRP3) inflammasome. The alpha significance value was < 0.05. RESULTS: Analysis of the microscopy and commercial kit results revealed that cell proliferation was suppressed, and no cell death was observed. The protein expression levels of NLRP3, STAT3, ser727 and HIF-1α were lower in the samples treated with donepezil and memantine at 72 h (p < 0.05). The protein expression levels of NLRP3, STAT3, ser727 and HIF-1α were higher in the samples treated with the combination of donepezil and memantine (p < 0.05). CONCLUSIONS: The combined administration of memantine a NMDAR antagonist which can prevent neurodegeneration and donepezil an AChEI used for pain relief increased the protein expression levels in the anabolic pathway. However, it did not reduce the protein expression levels in the catabolic pathway. Therefore, further studies are needed to provide extensive insight into whether it may be among the potential targets for the therapy of intervertebral disc (IVD) diseases.


Assuntos
Disco Intervertebral , Núcleo Pulposo , Acetilcolinesterase/metabolismo , Inibidores da Colinesterase/farmacologia , Donepezila/metabolismo , Donepezila/farmacologia , Feminino , Humanos , Inflamação/metabolismo , Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral , Deslocamento do Disco Intervertebral , Masculino , Memantina/metabolismo , Memantina/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Núcleo Pulposo/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
9.
J Colloid Interface Sci ; 617: 20-31, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35255395

RESUMO

Neurofibrillary tangle, composed of aggregated tau protein, is a pathological hallmark of Alzheimer's disease (AD). The inhibition of tau aggregation is therefore an important direction for AD drug discovery. In this work, we explored the efficacy of two types of carbon dots in targeting tau aggregation, as versatile nano-carriers for the development of carbon dots (CDs)-based AD therapy. We carried out synthesis, biophysical and biochemical characterizations of two types of CDs, namely, carbon nitride dots (CNDs) and black carbon dots (B-CDs). CDs, which are biocompatible and non-toxic, were successfully conjugated with memantine hydrochloride (MH) through EDC/NHS mediated amidation reactions followed by systematic characterizations using various biophysical techniques including UV-vis spectroscopy (UV-vis), photoluminescence (PL), Fourier-transform infrared spectroscopy (FTIR), thermogravimetric analysis (TGA), mass spectrometry (MS), Transmission electron microscopy (TEM) and atomic force microscopy (AFM). The surface diversity along with small particle sizes of CDs allowed facile delivery of MH across the blood-brain barrier (BBB), as demonstrated using a zebrafish in vivo model. The tau aggregation inhibition experiments were conducted using the thioflavin-T (ThT) assay to identify the most effective inhibitor. The kinetics and magnitude of tau aggregation were measured in the presence of CDs, which demonstrates that both B-CDs-MH and B-CDs alone are the most effective inhibitors of tau aggregation with IC50 values of 1.5 ± 0.3 and 1.6 ± 1.5 µg/mL, respectively. Taken together, our findings hold therapeutic significance to enhance the efficient delivery of MH to target AD pathology in the brain for improved efficacy.


Assuntos
Doença de Alzheimer , Pontos Quânticos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Animais , Barreira Hematoencefálica , Carbono/química , Memantina/metabolismo , Memantina/farmacologia , Memantina/uso terapêutico , Pontos Quânticos/química , Peixe-Zebra , Proteínas tau/metabolismo , Proteínas tau/uso terapêutico
10.
Bioengineered ; 13(3): 6688-6697, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35235756

RESUMO

Oxaliplatin is an effective chemotherapeutic agent for the treatment of malignant tumors. However, severe oxaliplatin-induced neurotoxicity has been well documented. Memantine is a drug for the management of Alzheimer's Disease (AD) due to its promising neuroprotective properties. We hypothesize that Memantine possesses a beneficial role against chemotherapy-induced neuronal damages. In this study, we established an oxaliplatin-induced neurotoxicity assay model in human SHSY-5Y neuronal cells and investigated the protective effect of Memantine. We showed that Memantine treatment ameliorated oxaliplatin-elevated intracellular production of reactive oxygen species (ROS), lipid product malondialdehyde (MDA), and NOX-2 expression. Memantine alleviated impairment of the mitochondrial membrane potential and ATP production by oxaliplatin. As a result, Memantine showed a protective role against oxaliplatin-induced cytotoxicity. Moreover, the terminal deoxynucleotidyl Transferase-mediated dUTP nick end labeling (TUNEL) apoptosis assay revealed that Memantine protected oxaliplatin-induced apoptosis through mitigating the ratio of Bax/Bcl-2 and Caspase-3 cleavage. We concluded Memantine ameliorated the neurotoxicity of oxaliplatin in a mitochondrial-dependent pathway.


Assuntos
Memantina , Mitocôndrias , Apoptose , Humanos , Memantina/metabolismo , Memantina/farmacologia , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Oxaliplatina/farmacologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
11.
Pharmacol Res Perspect ; 9(5): e00879, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34628723

RESUMO

The unbound concentrations of 14 commercial drugs, including five non-efflux/uptake transporter substrates-Class I, five efflux transporter substrates-class II and four influx transporter substrates-Class III, were simultaneously measured in rat liver, muscle, and blood via microanalysis. Kpuu,liver and Kpuu,muscle were calculated to evaluate the membrane transport activity and cell metabolism on the unbound drug concentrations in the skeletal muscle and liver. For Class I compounds, represented by antipyrine, unbound concentrations among liver, muscle and blood are symmetrically distributed when compound hepatic clearance is low. And when compound hepatic clearance is high, unbound concentrations among liver, muscle and blood are asymmetrically distributed, such as Propranolol. For Class II and III compounds, overall, the unbound concentrations among liver, muscle, and blood are asymmetrically distributed due to a combination of hepatic metabolism and efflux and/or influx transporter activity.


Assuntos
Membrana Celular/metabolismo , Fígado/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Músculo Esquelético/metabolismo , Preparações Farmacêuticas/metabolismo , Animais , Antipirina/sangue , Antipirina/metabolismo , Atenolol/sangue , Atenolol/metabolismo , Carbamazepina/sangue , Carbamazepina/metabolismo , Digoxina/sangue , Digoxina/metabolismo , Diltiazem/sangue , Diltiazem/metabolismo , Difenidramina/sangue , Difenidramina/metabolismo , Vias de Eliminação de Fármacos , Gabapentina/sangue , Gabapentina/metabolismo , Lamotrigina/sangue , Lamotrigina/metabolismo , Memantina/sangue , Memantina/metabolismo , Microdiálise , Ofloxacino/sangue , Ofloxacino/metabolismo , Preparações Farmacêuticas/sangue , Propranolol/sangue , Propranolol/metabolismo , Pirilamina/sangue , Pirilamina/metabolismo , Quinidina/sangue , Quinidina/metabolismo , Ratos , Terfenadina/análogos & derivados , Terfenadina/sangue , Terfenadina/metabolismo
12.
Int J Biol Macromol ; 190: 660-666, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34508722

RESUMO

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.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Calorimetria , Simulação por Computador , Memantina/metabolismo , Transferrina/metabolismo , Doença de Alzheimer/patologia , Fluorescência , Humanos , Ligação de Hidrogênio , Memantina/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Análise de Componente Principal , Conformação Proteica , Termodinâmica , Transferrina/química
13.
Brain Res ; 1768: 147581, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34280372

RESUMO

Brain penetration of cationic drugs is an important determinant of their efficacy and side effects. However, the effects of alterations in the activity of uptake transporters in the brain under inflammatory conditions on the brain penetration of cationic drugs are not fully understood. The aim of this study was to examine changes in brain penetration of cationic drugs, including diphenhydramine (DPHM), memantine (MMT), and cimetidine (CMD), and changes in the expression of uptake transporters such as organic cation transporter (Oct) in brain microvascular endothelial cells (BMECs) under inflammatory conditions. To clarify the effects of inflammation on the brain penetration of DPHM, MMT, and CMD, we performed brain microdialysis studies in a rat model of adjuvant-induced arthritis (AA). Further, differences in transporter mRNA expression levels between BMECs from control and AA rats were evaluated. Brain microdialysis showed that the unbound brain-to-plasma partition coefficient (Kp,uu,brain) for DPHM and MMT was significantly lower in AA rats compared with control rats. OCT mRNA levels were increased and proton-coupled organic cation (H+/OC) antiporter mRNA levels were decreased in AA rats compared with control rats. Taken together, our findings suggest that inflammation decreases the brain penetration of H+/OC antiporter substrates such as DPHM and MMT.


Assuntos
Encéfalo/metabolismo , Difenidramina/farmacologia , Memantina/farmacologia , Animais , Antiporters/genética , Antiporters/metabolismo , Antiporters/farmacologia , Artrite/tratamento farmacológico , Transporte Biológico/genética , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Cimetidina/farmacologia , Difenidramina/metabolismo , Células Endoteliais/metabolismo , Feminino , Inflamação/tratamento farmacológico , Inflamação/fisiopatologia , Memantina/metabolismo , Microdiálise/métodos , Ratos , Ratos Sprague-Dawley
14.
Chem Biol Interact ; 342: 109463, 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-33831382

RESUMO

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.


Assuntos
Inibidores da Colinesterase/farmacologia , Memantina/farmacologia , Soman/farmacologia , Animais , Bovinos , Inibidores da Colinesterase/química , Dopaminérgicos/farmacologia , Redução da Medicação , Memantina/química , Memantina/metabolismo , Estrutura Molecular , Fatores de Tempo
15.
Life Sci ; 270: 119012, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33422543

RESUMO

AIMS: Memantine is a non-competitive antagonist of glutamatergic NMDA receptor that is mainly used in the treatment of Alzheimer's disease. The excitatory toxicity mediated by glutamate via glutamatergic receptor signals is considered to be one of the mechanisms mediating neuronal injury and cognitive impairment after exposure to a hypoxic environment at a high altitude. Therefore, in this study, we hypothesized that inhibiting glutamate signaling using memantine could alleviate neuronal injury and cognitive impairment in rats exposed to chronic hypoxia. MAIN METHODS: we made animal models in the natural environment of the Qinghai-Tibet Plateau at an altitude of 4300 m, and used animal behavior, morphology, molecular biology and other methods to evaluate the impact of chronic hypoxia exposure on cognitive function and the neuroprotective effect of Memantine. KEY FINDINGS: Our results showed that the expression of NMDA receptors increased, while the expression of AMPA receptors decreased, after 4 weeks of chronic hypoxia exposure. Concomitantly, apoptotic neuronal cell death in the hippocampus and frontal cortex was significantly increased, along with levels of oxidative stress, whereas innate ability to inhibit free radicals decreased. Moreover, after 8 weeks of hypoxia exposure, learning, memory, and space exploration abilities were significantly decreased. Notably, after treatment with memantine, apoptotic neuronal cell death, oxidative stress, and free radical levels decreased, and the cognitive function of the animals improved. SIGNIFICANCE: Present study shows that chronic hypoxia can produce the excitatory toxicity leading to neural injury and cognitive impairment that can be suppressed with memantine treatment by inhibiting excitatory toxicity.


Assuntos
Doença da Altitude/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Memantina/farmacologia , Altitude , Doença da Altitude/tratamento farmacológico , Doença de Alzheimer/tratamento farmacológico , Animais , Morte Celular/efeitos dos fármacos , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Hipóxia/metabolismo , Masculino , Memantina/metabolismo , Memória/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo
16.
CPT Pharmacometrics Syst Pharmacol ; 9(10): 571-583, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32977369

RESUMO

Chronic kidney disease (CKD) has significant effects on renal clearance (CLr ) of drugs. Physiologically-based pharmacokinetic (PBPK) models have been used to predict CKD effects on transporter-mediated renal active secretion and CLr for hydrophilic nonpermeable compounds. However, no studies have shown systematic PBPK modeling of renal passive reabsorption or CLr for hydrophobic permeable drugs in CKD. The goal of this study was to expand our previously developed and verified mechanistic kidney model to develop a universal model to predict changes in CLr in CKD for permeable and nonpermeable drugs that accounts for the dramatic nonlinear effect of CKD on renal passive reabsorption of permeable drugs. The developed model incorporates physiologically-based tubular changes of reduced water reabsorption/increased tubular flow rate per remaining functional nephron in CKD. The final adaptive kidney model successfully (absolute fold error (AFE) all < 2) predicted renal passive reabsorption and CLr for 20 permeable and nonpermeable test compounds across the stages of CKD. In contrast, use of proportional glomerular filtration rate reduction approach without addressing tubular adaptation processes in CKD to predict CLr generated unacceptable CLr predictions (AFE = 2.61-7.35) for permeable compounds in severe CKD. Finally, the adaptive kidney model accurately predicted CLr of para-amino-hippuric acid and memantine, two secreted compounds, in CKD, suggesting successful integration of active secretion into the model, along with passive reabsorption. In conclusion, the developed adaptive kidney model enables mechanistic predictions of in vivo CLr through CKD progression without any empirical scaling factors and can be used for CLr predictions prior to assessment of drug disposition in renal impairment.


Assuntos
Rim/metabolismo , Taxa de Depuração Metabólica/efeitos dos fármacos , Insuficiência Renal Crônica/sangue , Reabsorção Renal/efeitos dos fármacos , Transporte Biológico , Progressão da Doença , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Rim/fisiologia , Memantina/metabolismo , Modelos Biológicos , Farmacocinética , Valor Preditivo dos Testes , Insuficiência Renal/metabolismo , Insuficiência Renal Crônica/classificação , Insuficiência Renal Crônica/diagnóstico , Insuficiência Renal Crônica/fisiopatologia , Ácido p-Aminoipúrico/metabolismo
17.
Chem Biol Interact ; 325: 109020, 2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32092300

RESUMO

Overactivation of N-methyl-D-aspartate (NMDA) receptors has been associated with neurodegenerative disorders such as Alzheimer's disease (AD), cerebral vascular disorders and amyotrophic lateral sclerosis (ALS). We have previously designed and synthesized a series of memantine nitrate and some of them have shown vessel dilatory effects and neuroprotective effects; however, the detailed mechanisms have not been elucidated. In this study, we further demonstrated that memantine nitrate-06 (MN-06), one of the novel compounds derived from memantine, possessed significant neuroprotective effects against glutamate-induced excitotoxicity in rat primary cerebellar granule neurons (CGNs). Pretreatment of MN-06 reversed the activation of GSK3b and the suppression of phosphorylated Akt induced by glutamate. In addition, the neuroprotective effects of MN-06 could be abolished by LY294002, the specific phosphatidylinositol 3-kinase (PI3-K) inhibitor. Ca2+ imaging shown that pretreatment of MN-06 prevented Ca2+ influx induced by glutamate. Moreover, MN-06 might inhibit the NMDA-mediated current by antagonizing NDMA receptors, which was further confirmed by molecular docking simulation. Taken together, MN-06 protected against glutamate-induced excitotoxicity by blocking calcium influx and attenuating PI3-K/Akt/GSK-3b pathway, indicating that MN-06 might be a potential drug for treating neurodegenerative disorders.


Assuntos
Cálcio/metabolismo , Ácido Glutâmico/toxicidade , Glicogênio Sintase Quinase 3 beta/metabolismo , Memantina/farmacologia , Neurônios/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Contagem de Células , Cerebelo/citologia , Hipocampo/citologia , Memantina/metabolismo , Simulação de Acoplamento Molecular , Neurônios/citologia , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Conformação Proteica , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/efeitos dos fármacos
18.
Bioconjug Chem ; 30(10): 2573-2583, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31553175

RESUMO

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.


Assuntos
Cloreto de Alumínio/efeitos adversos , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Dendrímeros/química , Lactoferrina/química , Memantina/química , Memantina/metabolismo , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/efeitos dos fármacos , Cognição/efeitos dos fármacos , Dendrímeros/toxicidade , Modelos Animais de Doenças , Dopamina/metabolismo , Portadores de Fármacos/química , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos , Eritrócitos/efeitos dos fármacos , Memantina/farmacocinética , Memantina/farmacologia , Camundongos , Ratos , Distribuição Tecidual
19.
Behav Brain Res ; 372: 112005, 2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31167109

RESUMO

During hippocampal-dependent memory formation, sensory signals from the neocortex converge in the dentate gyrus. It is generally believed that the dentate gyrus decorrelates inputs in order to minimize interference between codes for similar experiences, often referred to as pattern separation. The proportion of dentate neurons that are activated by experience is therefore likely to impact how memories are stored and separated. Emerging evidence from mouse models suggests that adult-born neurons can both increase and decrease activity levels in the dentate gyrus. However, the conditions that determine the direction of this modulation, and whether it occurs in other species, remains unclear. Furthermore, since the dentate gyrus is composed of a heterogeneous population of cells that are born throughout life, newborn neurons may not modulate all cells equally. We aimed to investigate whether adult neurogenesis in rats regulates activity in dentate gyrus neurons that are born at the peak of early postnatal development. Adult neurogenesis was increased by subjecting rats to an alternating running and memantine treatment schedule, and it was decreased with a transgenic GFAP-TK rat model. Activity was measured by Fos expression in BrdU+ cells after rats explored a novel environment. Running+memantine treatment increased adult neurogenesis by only 17%, but completely blocked experience-dependent Fos expression. In contrast, GFAP-TK rats had a 68% reduction in adult neurogenesis but normal experience-dependent Fos expression. The inconsistent relationship between neurogenesis and Fos expression suggests that neurogenesis does not regulate DG activity during exploration of a novel environment. Nonetheless, running and memantine may benefit disorders where there is elevated activity in the dentate gyrus, such as anxiety and age-related memory impairments.


Assuntos
Memantina/farmacologia , Neurogênese/fisiologia , Corrida/fisiologia , Animais , Encéfalo/fisiologia , Giro Denteado/fisiologia , Hipocampo/fisiologia , Masculino , Memantina/metabolismo , Memória/fisiologia , Neurogênese/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Condicionamento Físico Animal/fisiologia , Proteínas Proto-Oncogênicas c-fos/análise , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Long-Evans
20.
Sci Rep ; 9(1): 4612, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30874573

RESUMO

Hydrogen sulphide has recently drawn much attention due to its potent anti-inflammatory and neuroprotective roles in brain functions. The purpose of the current study was to exploit these beneficial properties of H2S to design a new agent for the treatment of Alzheimer's disease (AD). To pursue our aims, we replaced the free amine group of memantine with an isothiocyanate functionality as a putative H2S-donor moiety. The new chemical entity, named memit, was then tested in vitro to determine whether it retains the pharmacological profile of the "native drug", while also providing a source of H2S in the CNS. Indeed, Memit showed the ability to release H2S through a cysteine-mediated mechanism, thus generating memantine. Moreover, the new hybrid molecule exerts protective effects against neuronal inflammation and induces a drastic fall in ROS production. In addition, memit was also able to reduce the Aß(1-42) self-induced aggregation and exerted cytoprotective effect against Aß oligomers-induced damage in both human neurons and rat microglia cells. Finally, similarly to memantine, the new compound promotes autophagy, a complex process required for cellular homeostasis in cell survival that results to be altered in neurodegenerative diseases. In conclusion, our study revealed that memit is a prodrug of memantine. Further in vivo studies will be necessary to fully investigate the synergic or cumulative effects due to the H2S-releasing moiety and the native drug.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Memantina/farmacologia , Peptídeos beta-Amiloides/farmacologia , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Humanos , Sulfeto de Hidrogênio/metabolismo , Sulfeto de Hidrogênio/farmacologia , Inflamação , Memantina/análogos & derivados , Memantina/metabolismo , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Cultura Primária de Células , Pró-Fármacos/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...