Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 129: 111627, 2024 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-38309094

RESUMO

The current research aims to study the therapeutic efficacy of alpha-lipoic acid (α-LA) and caffeine-loaded chitosan nanoparticles (Caf-CNs) against cardiovascular complications induced by obesity. Rats were divided randomly into: control, high fat diet (HFD) induced obesity rat model, obese rats treated with α-LA and/or Caf-CNs. Triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), Interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) as well as activities of lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) significantly increased in the serum of obese rats. In addition, plasma atherogenic index, atherogenic coefficient and Castelli's risk indices I and II showed a significant increase. Additionally, levels of malondialdehyde (MDA) and nitric oxide (NO) and activity of monoamine oxidase (MAO) were significantly elevated in heart tissues of obese rats. However, cardiac Na+/K+-ATPase and acetylcholinesterase (AchE) activities and reduced glutathione (GSH), serotonin (5-HT), norepinephrine (NE) and dopamine (DA) as well as serum high-density lipoprotein cholesterol (HDL-C) were significantly reduced in obese rats. Treatment with α-LA and/or Caf-CNs ameliorated almost all the biochemical and histopathological alterations caused by obesity. In conclusion, the present data revealed that α-LA and/or Caf-CNs may be an effective therapeutic approach against cardiac complications caused by obesity through their antilipemic, anti-atherogenic, antioxidant, and anti-inflammatory activities.


Assuntos
Quitosana , Nanopartículas , Ácido Tióctico , Ratos , Animais , Ácido Tióctico/farmacologia , Cafeína/farmacologia , Quitosana/uso terapêutico , Quitosana/farmacologia , Acetilcolinesterase , Estresse Oxidativo , Obesidade/tratamento farmacológico , Obesidade/complicações , LDL-Colesterol
2.
Photochem Photobiol Sci ; 22(12): 2891-2904, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37917308

RESUMO

Photobiomodulation (PBM) of deep brain structures through transcranial infrared irradiation might be an effective treatment for Parkinson's disease (PD). However, the mechanisms underlying this intervention should be elucidated to optimize the therapeutic outcome and maximize therapeutic efficacy. The present study aimed at investigating the oxidative stress-related parameters of malondialdehyde (MDA), nitric oxide (NO), and reduced glutathione (GSH) and the enzymatic activities of sodium-potassium-ATPase (Na+, K+-ATPase), Acetylcholinesterase (AChE), and monoamine oxidase (MAO) and monoamine levels (dopamine (DA), norepinephrine (NE) and serotonin (5-HT) in the midbrain and striatum of reserpine-induced PD in an animal model treated with PBM. Furthermore, the locomotor behavior of the animals has been determined by the open field test. Animals were divided into three groups; the control group, the PD-induced model group, and the PD-induced model treated with the PBM group. Non-invasive treatment of animals for 14 days with 100 mW, 830 nm laser has demonstrated successful attainment in the recovery of oxidative stress, and enzymatic activities impairments induced by reserpine (0.2 mg/kg) in both midbrain and striatum of adult male Wistar rats. PBM also improved the decrease in DA, NE, and 5-HT in the investigated brain regions. On a behavioral level, animals showed improvement in their locomotion activity. These findings have shed more light on some mechanisms underlying the treatment potential of PBM and displayed the safety, easiness, and efficacy of PBM treatment as an alternative to pharmacological treatment for PD.


Assuntos
Terapia com Luz de Baixa Intensidade , Transtornos Parkinsonianos , Ratos , Masculino , Animais , Reserpina/farmacologia , Ratos Wistar , Serotonina , Acetilcolinesterase , Mesencéfalo , Dopamina , Adenosina Trifosfatases , Modelos Animais de Doenças
3.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3017-3031, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37306714

RESUMO

The present work investigated the effect of α-lipoic acid (ALA) and caffeine-loaded chitosan nanoparticles (CAF-CS NPs) on obesity and its hepatic and renal complications in rats. Rats were divided into control, rat model of obesity induced by high fat diet (HFD), and obese rats treated with ALA and/or CAF-CS NPs. At the end of the experiment, the activities of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) and the levels of urea, creatinine, interleukin-1ß (IL-1ß), and tumor necrosis factor-α (TNF-α) were determined in the sera of animals. In addition, malondialdehyde (MDA), nitric oxide (NO), and reduced glutathione (GSH) were measured in hepatic and renal tissues. Renal Na+, K+-ATPase was assessed. The histopathological changes were examined in the hepatic and renal tissues. Obese rats showed a significant increase in AST, ALT, ALP, urea, and creatinine. This was associated with a significant increase in IL-1ß, TNF-α, MDA, and NO. A significant decrease in hepatic and renal GSH and renal Na+, K+-ATPase activity was recorded in obese rats. Obese rats also showed histopathological alterations in hepatic and renal tissues. Treatment with ALA and/or CAF-CS NPs reduced the weight of obese rats and ameliorated almost all the hepatic and renal biochemical and histopathological changes induced in obese rats. In conclusion, the present findings indicate that ALA and/or CAF-CS NPs offered an effective therapy against obesity induced by HFD and its hepatic and renal complications. The therapeutic effect of ALA and CAF-CS NPs could be mediated through their antioxidant and anti-inflammatory properties.

4.
J Diabetes Metab Disord ; 21(2): 1327-1337, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36404819

RESUMO

Objective: The current study evaluates the effect of metformin (MET) and /or alpha lipoic acid (ALA) on hypothyroidism and its adverse effects on the cardiac, renal, and, hepatic functions in rats. Materials and methods: Rats were divided into five groups: control, rat model of hypothyroidism induced by propylthiouracil (PTU), rat model of hypothyroidism treated with MET, rat model of hypothyroidism treated with ALA, and rat model of hypothyroidism treated with MET and ALA. At the end of the experiment, body weight gain was determined and the blood samples were collected from orbital plexus to measure the serum levels of thyroxine (T4), triiodothyronine (T3) and thyroid stimulating hormone (TSH) by ELISA, glucose level, the activities of lactate dehydrogenase (LDH), creatine kinase MB (CK-MB), aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP), and the levels of urea and creatinine spectrophotometrically. Results: Rat model of hypothyroidism revealed a significant decrease in T4 (p < 0.001) and T3 (p < 0.001) and a significant increase in TSH (p < 0.005). This was accompanied by a significant decrease in the body weight gain (p < 0.025) and a significant increase in LDH (p < 0.001), CK-MB (p < 0.001) AST (p < 0.01), ALT (p < 0.016), ALP (p < 0.001), glucose (p < 0.001), urea (p < 0.001) and creatinine (p < 0.001). MET restored T4, T3 and TSH to control values. Treatment with ALA restored T3 and TSH levels. Treatment with Met and /or ALA reduced the levels of glucose, urea and creatinine and the activities of LDH, CK-MB, AST, ALT, and ALP to control-like values. Only ALA improved the reduced body weight gain induced by hypothyroidism. Conclusion: The present findings indicate the ameliorative effects of MET and /or ALA on hypothyroidism and its adverse effects on cardiac, renal and hepatic functions. Supplementary information: The online version contains supplementary material available at 10.1007/s40200-022-01063-7.

5.
Neuroendocrinology ; 112(11): 1129-1142, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35354137

RESUMO

OBJECTIVE: The present study evaluates the neuroprotective effect of α-lipoic acid (ALA) and/or metformin (MET) on the behavioral and neurochemical changes induced by hypothyroidism. METHODS: Rats were divided into control, rat model of hypothyroidism induced by propylthiouracil, and rat model of hypothyroidism treated with ALA, MET, or their combination. RESULTS: Behaviorally, hypothyroid rats revealed impaired memory and reduced motor activity as indicated from the novel object recognition test and open-field test, respectively. Hypothyroidism induced a significant increase in lipid peroxidation (malondialdehyde [MDA]) and a significant decrease in reduced glutathione (GSH) and nitric oxide (NO) in the cortex and hippocampus. These were associated with a significant increase in tumor necrosis factor-α (TNF-α) and a significant decrease in brain-derived neurotrophic factor (BDNF). Hypothyroidism decreased significantly the levels of serotonin (5-HT), norepinephrine (NE), and dopamine (DA) and reduced the activities of acetylcholinesterase (AchE) and Na+, K+-ATPase in the cortex and hippocampus. Treatment of hypothyroid rats with ALA and/or MET showed an improvement in memory function and motor activity. Moreover, ALA and/or MET prevented the increase in MDA and TNF-α, and the decline in GSH, NO, BDNF, 5-HT, NE, and DA. It also restored AchE and Na+, K+-ATPase activities in the studied brain regions. CONCLUSION: ALA and/or MET has a potential neuroprotective effect against the adverse behavioral and neurochemical changes induced by hypothyroidism in rats.


Assuntos
Hipotireoidismo , Metformina , Fármacos Neuroprotetores , Ácido Tióctico , Animais , Ratos , Ácido Tióctico/farmacologia , Ácido Tióctico/uso terapêutico , Fator Neurotrófico Derivado do Encéfalo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Acetilcolinesterase , Serotonina , Fator de Necrose Tumoral alfa , Dopamina , Propiltiouracila , Metformina/farmacologia , Metformina/uso terapêutico , Óxido Nítrico , Hipotireoidismo/induzido quimicamente , Hipotireoidismo/tratamento farmacológico , Glutationa , Malondialdeído , Norepinefrina , Adenosina Trifosfatases
6.
Cardiovasc Toxicol ; 21(6): 433-443, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33548025

RESUMO

The cardiotoxicity of chemotherapeutic drugs as cisplatin has become a major issue in recent years. The present study investigates the efficacy of curcumin nanoparticles against the cardiotoxic effects of cisplatin by assessment of oxidative stress parameters, Na+,K+-ATPase, acetylcholinesterase (AchE) and tumor necrosis factor-alpha (TNF-α) in cardiac tissue in addition to serum lactate dehydrogenase (LDH). Rats were divided into three groups: control rats that received saline for 14 days; cisplatin-treated rats that received a single intraperitoneal (i.p.) injection of cisplatin (12 mg/kg) followed by a daily oral administration of saline (0.9%) for 14 days and rats treated with a single i.p. injection of cisplatin (12 mg/kg) followed by a daily oral administration of curcumin nanoparticles (50 mg/kg) for 14 days. Cisplatin resulted in a significant increase in lipid peroxidation, nitric oxide (NO), and TNF-α and a significant decrease in reduced glutathione (GSH) levels and Na+, K+- ATPase activity. Moreover, significant increases in cardiac AchE and serum lactate dehydrogenase activities were recorded. Treatment of cisplatin-injected animals with curcumin nanoparticles ameliorated all the alterations induced by cisplatin in the heart of rats. This suggests that curcumin nanoparticles can be used as an important therapeutic adjuvant in chemotherapeutic and other toxicities mediated by oxidative stress and inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Curcumina/farmacologia , Cardiopatias/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Nanopartículas , Acetilcolinesterase/metabolismo , Animais , Cisplatino , Modelos Animais de Doenças , Proteínas Ligadas por GPI/metabolismo , Cardiopatias/induzido quimicamente , Cardiopatias/metabolismo , Cardiopatias/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Estresse Oxidativo/efeitos dos fármacos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
J Diet Suppl ; 18(1): 72-91, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-31958022

RESUMO

The current aim is to evaluate the effect of ashwagandha root extract (AE) on the neurochemical changes induced in the cortex and hippocampus as a consequence of thyroid dysfunction induced by propylthiouracil (PTU). Male Wistar rats were divided into; control, AE treated rats, rat model of hypothyroidism and rat model of hypothyroidism treated with either AE or L-thyroxine (T4) for 1 month. Rat model of hypothyroidism showed a significant decrease in serum levels of tri-iodothyronine (T3) and T4 and a significant increase in cortical and hippocampal lipid peroxidation (MDA), nitric oxide (NO), superoxide dismutase (SOD) and catalase (CAT). However, reduced glutathione (GSH) decreased significantly. This was associated with a significant increase in hippocampal tumor necrosis factor-α (TNF-α) and cortical dopamine levels. Both L-thyroxine and AE restored T3 and T4 levels. In the hippocampus L-Thyroxine prevented the increase in MDA and restored GSH but failed to restore the increased NO and TNF-α. In the cortex L-thyroxine didn't change the increased MDA and NO and the decreased GSH induced by PTU. L-thyroxine increased cortical and hippocampal SOD and CAT. AE prevented the increased hippocampal MDA, NO and TNF-α and the decreased GSH level induced by PTU. In the cortex AE failed to restore MDA and NO but prevented the decrease in GSH. The increase in cortical dopamine level induced by PTU was ameliorated by L-thyroxine and improved by AE. The present data indicate that AE could prevent thyroid dysfunction and reduce its complications on the nervous system including oxidative stress and neuroinflammation.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Hipotireoidismo , Fármacos Neuroprotetores , Extratos Vegetais , Animais , Modelos Animais de Doenças , Hipotireoidismo/tratamento farmacológico , Inflamação/tratamento farmacológico , Masculino , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/administração & dosagem , Extratos Vegetais/farmacologia , Raízes de Plantas , Ratos , Ratos Wistar
8.
Drug Chem Toxicol ; 42(2): 194-202, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30257586

RESUMO

The present study is conducted to evaluate the neuroprotective effect of curcumin nanoparticles (CUR NP) against the neurotoxicity induced by cisplatin (CP) in rat. Rats were divided into control group that received saline solution, CP-treated rats that received a single i.p. injection of CP (12 mg/kg body wt), and CP-treated rats that received a single i.p injection of CP (12 mg/kg body wt) followed by a daily oral administration of CUR NP (50 mg/kg body wt) for 14 days. At the end of the experiment, the motor activity of rats was evaluated by open field test. The neurochemical and histopathological changes were investigated in the cerebral cortex. A significant decrease in motor activity was observed in CP-treated rats. This was associated with a significant increase in the cortical levels of lipid peroxidation, nitric oxide, tumor necrosis factor-α, caspase-3, and acetylcholinesterase activity. However, CP induced a significant decrease in reduced glutathione levels and Na+, K+-ATPase activity. In rats treated with CP and CUR NP, no significant changes were recorded in the parameters of the open field test as compared to control. In addition, treatment with CUR NP prevented all the neurochemical changes induced by CP except the increased value of nitric oxide. CUR NP also reduced the histopathological changes induced by CP. It is clear from the present data that CUR NP could ameliorate the neurotoxic effect induced by cisplatin.


Assuntos
Encéfalo/efeitos dos fármacos , Cisplatino/toxicidade , Curcumina/farmacologia , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/toxicidade , Acetilcolinesterase/metabolismo , Animais , Química Encefálica/efeitos dos fármacos , Curcumina/administração & dosagem , Glutationa/análise , Injeções Intraperitoneais , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Atividade Motora/efeitos dos fármacos , Nanopartículas/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Óxido Nítrico/análise , Ratos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/metabolismo , Fator de Necrose Tumoral alfa/análise
9.
Nutr Neurosci ; 22(11): 789-796, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29514562

RESUMO

Objective: The present study aims to investigate the neuroprotective effect of caffeine against aluminum chloride (AlCl3)-induced neurotoxicity in rats. Methods: Twenty-one male albino rats were divided into 3 groups: control, AlCl3-intoxicated group that received daily oral administration of AlCl3 (100 mg/kg for 30 days) and protected group injected daily with caffeine (20 mg/kg intraperitoneally) one hour before oral administration of AlCl3 for 30 days. Levels of lipid peroxidation, reduced glutathione, and nitric oxide and the activities of acetylcholinesterase (AchE) and Na+/K+-ATPase were measured spectrophotometrically. Tumor necrosis factor-α (TNF-α) was evaluated by ELISA kit. Results: The data revealed evidence of oxidative and nitrosative stress in the cerebral cortex, hippocampus, and striatum of AlCl3-intoxicated rats. This was indicated from the increased levels of lipid peroxidation and nitric oxide together with the decreased level of reduced glutathione. Moreover, the daily AlCl3 administration increased AchE and Na+/K+-ATPase activities and the level of TNF-α in the selected brain regions. Protection with caffeine ameliorated the oxidative stress induced by AlCl3 in the cerebral cortex, hippocampus, and striatum. In addition, caffeine restored the elevated level of TNF-α in the hippocampus and striatum. This was accompanied by an improvement in the activities of AchE and Na+/K+-ATPase in the studied brain regions. Discussion and conclusions: The present findings clearly indicate that caffeine provides a significant neuroprotection against AlCl3-induced neurotoxicity mediated by its antioxidant, anti-inflammatory, and anticholinesterase properties.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Antioxidantes/administração & dosagem , Encéfalo/efeitos dos fármacos , Cafeína/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Síndromes Neurotóxicas/tratamento farmacológico , Cloreto de Alumínio/toxicidade , Animais , Encéfalo/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar
11.
Behav Brain Res ; 324: 41-50, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28212939

RESUMO

Cannabis sativa is one of the most widely recreational drugs and its use is more prevalent among depressed patients. Some studies reported that Cannabis has antidepressant effects while others showed increased depressive symptoms in Cannabis users. Therefore, the present study aims to investigate the effect of Cannabis extract on the depressive-like rats. Twenty four rats were divided into: control, rat model of depression induced by reserpine and depressive-like rats treated with Cannabis sativa extract (10mg/kg expressed as Δ9-tetrahydrocannabinol). The depressive-like rats showed a severe decrease in motor activity as assessed by open field test (OFT). This was accompanied by a decrease in monoamine levels and a significant increase in acetylcholinesterase activity in the cortex and hippocampus. Na+,K+-ATPase activity increased in the cortex and decreased in the hippocampus of rat model. In addition, a state of oxidative stress was evident in the two brain regions. This was indicated from the significant increase in the levels of lipid peroxidation and nitric oxide. No signs of improvement were observed in the behavioral and neurochemical analyses in the depressive-like rats treated with Cannabis extract. Furthermore, Cannabis extract exacerbated the lipid peroxidation in the cortex and hippocampus. According to the present findings, it could be concluded that Cannabis sativa aggravates the motor deficits and neurochemical changes induced in the cortex and hippocampus of rat model of depression. Therefore, the obtained results could explain the reported increase in the depressive symptoms and memory impairment among Cannabis users.


Assuntos
Cannabis/efeitos adversos , Depressão/induzido quimicamente , Dronabinol/administração & dosagem , Acetilcolinesterase/metabolismo , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Depressão/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Atividade Motora , Norepinefrina/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos Wistar , Reserpina , Serotonina/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...