Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Trace Elem Med Biol ; 74: 127074, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36155418

RESUMO

BACKGROUND: Lead is a ubiquitous environmental heavy metal known to induce neurotoxicity. It has been postulated that substance with high antioxidant capacity could alleviate lead-induced neurotoxicity. Adansonia digitata fruit shell extract (ADFS) has been reported to have high phenolic contents and exerts antioxidant activity. This study investigated the effects of Adansonia digitata fruit shell extract on lead-induced neurotoxicity in mice. METHODS: Male balb/c mice (n = 7) were administered with Pb-acetate (50 mg/kg) 30 mins before ADFS (250 mg/kg and 500 mg/kg) or succimer (50 mg/kg) per orally for 28 days. Motor activities were evaluated on days 29 and 30 through horizontal bar and open field tests respectively. Further, spectrophotometry, atomic absorption spectrophotometry and haematoxylin and eosin staining were carried-out to determine the expression of oxidative stress biomarkers, level of lead concentration in the brain and histology of the cerebellum respectively. RESULTS: Lead acetate exposure significantly (p < 0.05) induced motor deficits in horizontal bar test and open field test, caused oxidative stress, high concentration of lead in the brain as well as histological aberration in the cerebellum. ADFS significantly (p < 0.05) reversed the motor deficits evident by increased muscle strength and number of lines crossed. Further, ADFS significantly reversed oxidative stress evident by increased levels of SOD, CAT and GSH and decreased level of MDA. There was also significant (p < 0.05) decrease in brain lead concentration as well as reduced cerebellar cells death. CONCLUSION: Findings suggest that ADFS attenuated motor deficits via inhibition of oxidative stress and chelating activity which is comparable to succimer. Hence, ADFS should be explored for possible development of chelating agent against lead and other heavy metals toxicity.


Assuntos
Adansonia , Antioxidantes , Adansonia/metabolismo , Animais , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Quelantes/farmacologia , Amarelo de Eosina-(YS)/farmacologia , Frutas/metabolismo , Chumbo/toxicidade , Masculino , Camundongos , Estresse Oxidativo , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Succímero/farmacologia , Superóxido Dismutase/metabolismo
2.
Chem Res Toxicol ; 35(10): 1701-1719, 2022 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-35972774

RESUMO

Arsenic, a metalloid, is known to cause deleterious effects in various body organs, particularly the liver, urinary bladder, and brain, and these effects are primarily mediated through oxidative stress. Chelation therapy has been considered one of the promising medical treatments for arsenic poisoning. Meso 2,3- dimercaptosuccinic acid (DMSA) has been recognized as one of the most effective chelating drugs to treat arsenic poisoning. However, the drug is compromised with a number of shortcomings, including the inability to treat chronic arsenic poisoning due to its extracellular distribution. Monoisoamyl 2,3-dimercaptosuccinic acid, one of the analogues of meso 2,3-dimeraptosuccinic acid (DMSA), is a lipophilic chelator and has shown promise to be considered as a potential future chelating agent/antidote not only for arsenic but also for a few other heavy metals like lead, mercury, cadmium, and gallium arsenide. The results from numerous studies carried out in the recent past, mainly from our group, strongly support the clinical application of MiADMSA. This review paper summarizes most of the scientific details including the chemistry, pharmacology, and safety profile of MiADMSA. The efficacy of MiADMSA mainly against arsenic toxicity but also a few other heavy metals was also discussed. We also reviewed a few other strategies in order to achieve the optimum effects of MiADMSA, like combination therapy using two chelating agents or coadministration of a natural and synthetic antioxidant (including phytomedicine) along with MiADMSA for treatment of metal/metalloid poisoning. We also briefly discussed the use of nanotechnology (nano form of MiADMSA i.e. nano-MiADMSA) and compared it with bulk MiADMSA. All these strategies have been shown to be beneficial in getting more pronounced therapeutic efficacy of MiADMSA, as an adjuvant or as a complementary agent, by significantly increasing the chelating efficacy of MiADMSA.


Assuntos
Intoxicação por Arsênico , Arsênio , Mercúrio , Animais , Antídotos , Antioxidantes/uso terapêutico , Intoxicação por Arsênico/tratamento farmacológico , Cádmio , Quelantes/farmacologia , Quelantes/uso terapêutico , Intoxicação por Metais Pesados/tratamento farmacológico , Ratos , Ratos Wistar , Succímero/análogos & derivados , Succímero/farmacologia , Succímero/uso terapêutico
3.
Sci Rep ; 11(1): 252, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33420282

RESUMO

Lead (Pb) toxicity is one of the most prevalent causes of human neurotoxicity. The available chelator drugs used now have many adverse effects. So, in this study, the protective role of Beta vulgaris juice (BVJ) on rat neurotoxicity induced by Pb was evaluated and the results were compared with the results of dimercaptosuccinic acid (DMSA, as used drug). Additionally, the synergistic effect of BVJ and DMSA against Pb-induced neurotoxicity was assessed. The study focused on the determination of the antioxidant, anti-inflammatory, and neurological potential of BVJ (alone, and with DMSA) towards lead-induced neurotoxicity. Also, the characterization of BVJ was studied. The results showed that BVJ contains considerable quantities of polyphenols, triterpenoids, and betalains which play an important role as antioxidants and anti-inflammatory. BVJ exhibited a protective effect against neurotoxicity via the reduction of Pb levels in blood and brain. Moreover, BVJ decreased the oxidative stress, inflammation, and cell death induced by Pb. Also, BVJ regulated the activities of acetylcholine esterase and monoamine oxidase-A which changed by Pb toxicity. BVJ and DMSA combination displayed a synergistic antineurotoxic effect (combination index ˂ 1). These results were in harmony with brain histopathology. Conclusion: BVJ has a powerful efficacy in the protection from brain toxicity via diminishing Pb in the brain and blood circulation, resulting in the prevention of the oxidative and inflammatory stress. Treatment with BVJ in combination with DMSA revealed a synergistic effect in the reduction of neurotoxicity induced by Pb. Also, the antioxidant and anti-inflammatory effects of the BVJ lead to the improvement of DMSA therapy.


Assuntos
Antioxidantes/uso terapêutico , Beta vulgaris/química , Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/tratamento farmacológico , Extratos Vegetais/uso terapêutico , Succímero/uso terapêutico , Animais , Antioxidantes/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Fragmentação do DNA/efeitos dos fármacos , Sinergismo Farmacológico , Inflamação/tratamento farmacológico , Chumbo/sangue , Chumbo/toxicidade , Masculino , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/etiologia , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Ratos , Succímero/farmacologia
4.
Biomed Pharmacother ; 132: 110871, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33069968

RESUMO

Chelation therapy is considered as a safe and effective strategy to combat metal poisoning. Arsenic is known to cause neurological dysfunctions such as impaired memory, encephalopathy, and peripheral neuropathy as it easily crosses the blood-brain barrier. Oxidative stress is one of the mechanisms suggested for arsenic-induced neurotoxicity. We prepared Solid Lipid nanoparticles loaded with Monoisoamyl 2, 3-dimercaptosuccinic acid (Nano-MiADMSA), and compared their efficacy with bulk MiADMSA for treating arsenic-induced neurological and other biochemical effects. Solid lipid nanoparticles entrapping MiADMSA were synthesized and particle characterization was carried out by transmission electron microscopy (TEM) and dynamic light scattering (DLS). An in vivo study was planned to investigate the therapeutic efficacy of MiADMSA-encapsulated solid lipid nanoparticles (Nano-MiADMSA; 50 mg/kg orally for 5 days) and compared it with bulk MiADMSA against sodium meta-arsenite exposed rats (25 ppm in drinking water, for 12 weeks) in male rats. The results suggested the size of Nano-MiADMSA was between 100-120 nm ranges. We noted enhanced chelating properties of Nano-MiADMSA compared with bulk MiADMSA as evident by the reversal of oxidative stress variables like blood δ-aminolevulinic acid dehydratase (δ-ALAD), Reactive Oxygen Species (ROS), Catalase activity, Superoxide Dismutase (SOD), Thiobarbituric Acid Reactive Substances (TBARS), Reduced Glutathione (GSH) and Oxidized Glutathione (GSSG), Glutathione Peroxidase (GPx), Glutathione-S-transferase (GST) and efficient removal of arsenic from the blood and tissues. Recoveries in neurobehavioral parameters further confirmed nano-MiADMSA to be more effective than bulk MiADMSA. We conclude that treatment with Nano-MiADMSA is a better therapeutic strategy than bulk MiADMSA in reducing the effects of arsenic-induced oxidative stress and associated neurobehavioral changes.


Assuntos
Antioxidantes/farmacologia , Intoxicação por Arsênico/tratamento farmacológico , Arsenitos , Encéfalo/efeitos dos fármacos , Quelantes/farmacologia , Lipídeos/química , Nanopartículas , Estresse Oxidativo/efeitos dos fármacos , Compostos de Sódio , Succímero/análogos & derivados , Animais , Antioxidantes/química , Intoxicação por Arsênico/etiologia , Intoxicação por Arsênico/metabolismo , Intoxicação por Arsênico/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Biomarcadores/sangue , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Quelantes/química , Modelos Animais de Doenças , Composição de Medicamentos , Masculino , Atividade Motora/efeitos dos fármacos , Ratos Transgênicos , Succímero/química , Succímero/farmacologia
5.
BMC Complement Med Ther ; 20(1): 268, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32873301

RESUMO

BACKGROUND: Lead (Pb) is observed in all areas of the environment, mainly derived from human operations such as mining, processing, and burning fossil fuels. Pb toxicity is one of the most prevalent causes of human hepatotoxicity. The available chelator drugs used now have many adverse effects and therefore the world is looking for natural and secure alternatives. METHODS: Here, we evaluated the hepatoprotective role of the oral administration (1 g/kg b.w.) of the lyophilized Beta vulgaris juice (BVJ) against Pb-induced rat hepatotoxicity. We also examined the possible synergistic hepatoprotective impact of the combination between BVJ and 2,3- dimercaptosuccinic acid (DMSA, the currently approved drug for Pb-toxicity). The evaluation depends on the ability of BVJ, DMSA, or their combination (BVJ-DMSA) to reduce serum and hepatic Pb level and to avoid oxidative stress and inflammation caused by Pb. The level of lipid peroxidation, reduced glutathione (GSH), total antioxidant capacity, and the activity of the antioxidant enzymes were quantified. In addition, the level of interleukin (IL)-6, nitric oxide (NO), DNA fragmentation, and liver histology were studied. RESULTS: The results showed that BVJ contained considerable amounts of betalains, vitamin C, and various types of phenolic compounds. Therefore, BVJ displayed a significant (p < 0.05) preventive influence on the elevation of Pb levels in blood and liver as well as the hepatic DNA fragmentation. In addition, it significantly (p < 0.05) improved most of the studied antioxidant and inflammatory markers in the Pb-intoxicated rats. However, the combined extract (BVJ-DMSA) revealed synergistic (combination index < 1) activities in most of the tested parameters. The histopathological results verified the biochemical findings of this research. CONCLUSION: BVJ has a potent efficiency in the protection from Pb-induced hepatotoxicity through the reduction of its accumulation in blood and liver and the prevention of the oxidative stress and inflammation induced by Pb. Additionally, the treatment of hepatotoxicity with BVJ and DMSA in combination showed a synergistic effect and reduced the adverse effects induced by DMSA. Thus, BVJ can be a promising hepatoprotective extract against lead toxicity and its combination with DMSA potentiates this effect.


Assuntos
Beta vulgaris , Sinergismo Farmacológico , Inflamação/tratamento farmacológico , Intoxicação por Chumbo/tratamento farmacológico , Fígado/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Succímero/farmacologia , Administração Oral , Animais , Antioxidantes/farmacologia , Quelantes/farmacologia , Modelos Animais de Doenças , Egito , Sucos de Frutas e Vegetais , Chumbo/sangue , Masculino , Ratos
6.
Metallomics ; 12(9): 1428-1448, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32677644

RESUMO

INTRODUCTION: copper dyshomeostasis has long been linked with several neurodegenerative disorders. The binding of Cu with amyloid beta and other neuronal proteins in the brain leads to the generation of oxidative stress, which eventually causes neurotoxicity. METHOD: the present study was aimed at elucidating the efficacy of monoisoamyl 2,3-dimercaptosuccinic acid (MiADMSA) and d-penicillamine (DPA) (0.3 mEq kg-1, oral administration for 2 weeks) against Cu(ii)-induced (20 mg kg-1, oral administration for 16 weeks) neurotoxicity in Sprague-Dawley (SD) rats. RESULTS: we observed that the MiADMSA treatment modulated the altered oxidative and nitrosative stress parameters, antioxidant enzymes, and acetylcholinesterase (AChE) activity. Significant improvements were noticed in the neurobehavioral parameters except for the memory parameter. We also observed moderate improvement of memory impairment in the rats treated with MiADMSA and DPA post Cu(ii) exposure, as assessed by a passive avoidance test. Disease progression involves multiple factors and results in the up-regulation of intra and extracellular proteins such as amyloid beta and tau proteins; the expressions of these proteins were significantly reduced by the treatment proposed in our study, and these results were confirmed by ELISA and qRT-PCR. The expression of caspase-3 was higher in Cu(ii)-exposed rats, whereas it was lower in the MiADMSA-treated group. The proposed treatment reduced the copper-induced histological changes in the cortex and hippocampus regions of the brain. CONCLUSION: it can be summarised from the present study that MiADMSA is effective in reducing Cu(ii)-induced oxido-nitrosative stress, antioxidant defense enzymes, neurobehavioral changes, neuronal markers, apoptotic markers, and their genetic expressions. We conclude that chelation therapy using MiADMSA might be a promising approach for the treatment of copper-induced neurotoxicity.


Assuntos
8-Hidroxi-2'-Desoxiguanosina/análise , Peptídeos beta-Amiloides/análise , Cobre/efeitos adversos , Fármacos Neuroprotetores/farmacologia , Succímero/análogos & derivados , Proteínas tau/análise , Animais , Encéfalo/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Masculino , Simulação de Acoplamento Molecular , Ratos Sprague-Dawley , Succímero/farmacologia
7.
J Biochem Mol Toxicol ; 34(2): e22425, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31729815

RESUMO

The effect of combined administration of calcium (Ca), iron (Fe), zinc (Zn), chrysanthemum flavonoids, and meso-2,3-dimercaptosuccinic acid (DMSA) on the treatment of lead (Pb) intoxication in mice was studied. One hundred ninety female mice (SPF level, aged 18-22 days) were randomly divided into two groups as experimental animals. Mice in group I (10 mice) served as normal control animals, and were administered deionized water containing 12.5 µL/L acetate acid for 6 weeks, whereas mice in group II (180 mice) were exposed to 0.1% (wt/vol) of lead acetate in deionized water for 6 weeks and served as experimental animals. After 6 weeks of successful modeling, 180 mice from group II (lead-exposed) were divided into 18 groups of 10 mice each, 16 of which were treated by the combined administration of Ca, Fe, Zn, chrysanthemum flavonoids, and DMSA by L16 (215 ) orthogonal design. The remaining two groups were given treatment with low and high doses of DMSA, respectively. After three weeks of intervention (ig), the optimal treatment group was identified according to its blood lead level, as well as some antioxidant indices in the blood, liver, and hippocampus. The results indicated that the combined administration of Fe, Zn, chrysanthemum flavonoids, and DMSA with low dosage had the most significant effect on increasing the activities of blood delta-aminolevulinic acid dehydratase and superoxide dismutase (SOD), hepatic SOD and hippocampus nitric oxide synthase while decreasing the blood lead level, the content of hepatic malondialdehyde and hippocampus nitric oxide; this was considered the optimal treatment group. There was no difference in the level of blood hemoglobin between the optimal treatment group and the model control group (the first group of the orthogonal experiment). The activities of blood glutathione (GSH), hepatic GSH and glutathione peroxidase of the optimal treatment group were the same as other groups', and the recovery of the related indexes in the optimal effect group closely resembled the high dosage DMSA group. It can be concluded that the coadministration of Fe, Zn, and chrysanthemum flavonoids along with a low-dose DMSA effectively reduces Pb poisoning and lead-induced oxidative damage in lead-exposed mice; the result may provide a theoretical reference for the treatment of Pb poisoning.


Assuntos
Cálcio/farmacologia , Chrysanthemum/química , Flavonoides/farmacologia , Ferro/farmacologia , Intoxicação por Chumbo/tratamento farmacológico , Extratos Vegetais/farmacologia , Succímero/farmacologia , Zinco/farmacologia , Animais , Cálcio/administração & dosagem , Modelos Animais de Doenças , Quimioterapia Combinada , Feminino , Flavonoides/administração & dosagem , Glutationa/sangue , Glutationa Peroxidase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Ferro/administração & dosagem , Chumbo/efeitos adversos , Chumbo/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Malondialdeído/metabolismo , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/administração & dosagem , Sintase do Porfobilinogênio/sangue , Succímero/administração & dosagem , Superóxido Dismutase/sangue , Resultado do Tratamento , Zinco/administração & dosagem
8.
Int J Med Mushrooms ; 21(2): 143-154, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30806221

RESUMO

Ganoderma lucidum is a hard, bitter mushroom with many ethnomedicinal uses, including conditions similar to lead (Pb) toxicity. The aim of this study was to evaluate the protective effects of a G. lucidum aqueous extract (GL) when concurrently administered with Pb. Adult Wistar rats were administered oral doses of Pb (100 mg/kg) daily for 25 consecutive days. Of the Pb-treated rats, 3 groups received 100, 200, or 400 mg/kg/day GL, respectively; one group was given only 50 mg/kg/day 2,3-dimercaptosuccinic acid (DMSA); and another group was given 400 mg/kg/day GL and 50 mg/kg/day DMSA. Body weight, Pb levels in organs, enzyme and lipid levels in serum, and antioxidant capability were evaluated. Body weights were not significantly altered by GL. All doses of GL significantly reduced the amount of Pb in the liver (P < 0.01) and kidneys (P < 0.05), but not in the spleen. Doses of GL significantly reduced (P < 0.05) amounts of low-density lipoprotein, but not high-density lipoprotein or triglycerides, in serum. Pb-induced increases in amounts of serum aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase were significantly (P < 0.01) attenuated by GL. Also, a Pb-induced reduction in the amount of superoxide dismutase was significantly (P < 0.05) reversed, but the nitric oxide level was not significantly elevated. An increased malondialdehyde level, which had been induced by Pb, was significantly (P < 0.01) reversed. In conclusion, GL protects against some of the deleterious effects of Pb ingestion, possibly through antioxidant and other mechanisms. DMSA did not enhance the beneficial effects of GL.


Assuntos
Antioxidantes/farmacologia , Compostos Organometálicos/toxicidade , Reishi , Administração Oral , Animais , Antioxidantes/administração & dosagem , Antioxidantes/química , Feminino , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar , Succímero/farmacologia
9.
J Trace Elem Med Biol ; 50: 596-600, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29706452

RESUMO

Cadmium (Cd) is an environmental pollutant shown to induce multi organ dysfunction. In this study we present novel data about the effects of meso-2,3-dimercaptosuccinic acid (DMSA), monensin and salinomycin on the concentration of Cd in skeletal muscles of mice exposed to Cd (II) acetate treatment for 14 days. The impact of Cd and the chelating agents on the endogenous concentrations of calcium (Ca), copper (Cu), iron (Fe), magnesium (Mg), phosphorous (P), selenium (Se) and zinc (Zn) was also investigated. Subacute exposure of mice to Cd (II) acetate resulted in a significant accumulation of the toxic metal ion in the skeletal muscles compared to the untreated controls. Salinomycin most effectively mobilized Cd from the muscles compared to DMSA and monensin. The Cd exposure and the tested chelating agents did not significantly alter the endogenous concentrations of the selected essential elements in mouse muscles. The presented results confirmed that among the tested chelating agents salinomycin is superior as a potential antidote to Cd poisoning.


Assuntos
Cádmio/metabolismo , Cálcio/metabolismo , Cobre/metabolismo , Monensin/farmacologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Piranos/farmacologia , Succímero/farmacologia , Animais , Quelantes/química , Homeostase/efeitos dos fármacos , Ferro/metabolismo , Magnésio/metabolismo , Camundongos , Selênio/metabolismo , Zinco/metabolismo
10.
Chem Biol Interact ; 286: 78-87, 2018 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-29548727

RESUMO

Chelation therapy is the mainstream treatment for heavy metal poisoning. Apart from this, therapy using antioxidant/herbal extracts are the other strategies now commonly being tried for the treatment. We have previously reported individual beneficial efficacy of nanoparticle mediated administration of an antioxidant like 'curcumin' and an arsenic chelator 'monoisoamyl 2,3-dimercaptosuccinic acid (MiADMSA)' for the treatment of arsenic toxicity compared to bulk drugs. The present paper investigates our hypothesis that a combination drug delivery therapy employing two nanosystems, a chelator and a strong antioxidant, may produce more pronounced therapeutic effects compared to individual effects in the treatment of arsenic toxicity. An in-vivo study was conducted wherein arsenic as sodium arsenite (100 ppm) was administered in drinking water for 5 months to Swiss albino mice. This was followed by a treatment protocol comprising of curcumin encapsulated chitosan nanoparticles (nano-curcumin, 15 mg/kg, orally for 1 month) either alone or in combination with MiADMSA encapsulated polymeric nanoparticles (nano-MiADMSA, 50 mg/kg for last 5 days) to evaluate the therapeutic potential of the combination treatment. Our results demonstrated that co-treatment with nano-curcumin and nano-MiADMSA provided beneficial effects in a synergistic way on the adverse changes in oxidative stress parameters and metal status induced by arsenic.


Assuntos
Intoxicação por Arsênico/tratamento farmacológico , Arsênio/toxicidade , Curcumina/administração & dosagem , Portadores de Fármacos/química , Nanopartículas/química , Succímero/análogos & derivados , 8-Hidroxi-2'-Desoxiguanosina , Animais , Curcumina/química , Curcumina/farmacologia , Desoxiguanosina/análogos & derivados , Desoxiguanosina/urina , Sinergismo Farmacológico , Enzimas/sangue , Enzimas/metabolismo , Enzimas/urina , Glutationa/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Succímero/administração & dosagem , Succímero/química , Succímero/farmacologia
11.
J Environ Pathol Toxicol Oncol ; 33(4): 323-37, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25404379

RESUMO

Moringa oleifera is a tree belonging to Moringaceae family and its leaves and seeds are reported to have ameliorative effects against metal toxicity. In the present investigation, M. oleifera seed powder was tested against lead-induced oxidative stress and compared against meso-2, 3-dimercaptosuccinic acid (DMSA) treatment. Male Wistar rats (100-120 g) were divided into four groups: control (2000 ppm of sodium acetate for 2 weeks), exposed (2000 ppm of lead acetate for 2 weeks), Moringa treated (500 mg/kg for 7 days after lead exposure), and DMSA treated (90 mg/kg for 7 days after lead exposure). After exposure and treatment periods, rats were sacrificed and the brain was separated into cerebellum, hippocampus, frontal cortex, and brain stem; liver, kidney, and blood were also collected. The data indicated a significant (p<0.05) increase in reactive oxygen species (ROS), lipid perioxidation products (LPP), total protein carbonyl content (TPCC), and metal content of brain regions, liver, and kidney in the exposed group compared with their respective controls. In the blood, delta-amino levulinic acid dehydratase (ALAD) activity, RBC, WBC, hemoglobin, and hematocrit showed significant (p<0.05) decrease on lead exposure. However, administration of M. oleifera restored all the parameters back to control, tissue-specifically, and also showed improvement in restoration better than DMSA treatment, indicating reduction of the negative effects of lead-induced oxidative stress.


Assuntos
Antioxidantes/farmacologia , Quelantes/farmacologia , Chumbo/toxicidade , Moringa oleifera/química , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Animais , Antioxidantes/farmacocinética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Quelantes/farmacocinética , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Extratos Vegetais/farmacocinética , Pós/farmacocinética , Ratos , Ratos Wistar , Sementes/química , Succímero/farmacocinética , Succímero/farmacologia , Distribuição Tecidual
12.
J Biomed Nanotechnol ; 10(8): 1574-87, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25016657

RESUMO

To evaluate the effects of iron oxide nanoparticles on genes from different cell lines in this study, mouse macrophage RAW264.7 and hepatocyte Hepa1-6 cell lines were treated with DMSA-coated Fe3O4 nanoparticles. Two doses were used, 50 microg/mL and 100 microg/mL, respectively, for 24 hr and gene expression profile was detected by DNA microarrays. The gene expression patterns for the two cell lines greatly differed from each other when compared, revealing distinct cell-specific effects of the nanoparticles on the genes. It was found that the nanoparticles significantly influenced expression of genes in the RAW264.7 cells, showing complete difference from those of Hepa1-6 cells. More genes were downregulated in the RAW264.7 cells by two doses of the nanoparticles but up-regulated in the Hepa1-6 cells by two doses of the same nanoparticles. Moreover, the increase of nanoparticle dose greatly decreased up-regulation of genes but increased down-regulation of genes in the RAW264.7 cells. The influence of nanoparticles did not result in similar effect in the Hepa1-6 cells. Apart from the difference in gene expression patterns in the two cell lines, there were eight and seven genes which were consistently down-regulated and up-regulated by low-dose and high-dose of nanoparticles in the two cell lines, respectively, revealing the common effects of the nanoparticles on the genes in the two cell lines. These common effects on genes were caused by homeostatic processes, influenced by down-regulation of genes and immune responses, and also cell death, influenced by up-regulation of genes. The data from this study has shed new insights into the potential in vivo nanotoxicolgy of the DMSA-coated Fe3O4 nanoparticles.


Assuntos
Expressão Gênica/efeitos dos fármacos , Nanopartículas de Magnetita/química , Succímero/química , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Perfilação da Expressão Gênica , Espaço Intracelular/química , Espaço Intracelular/metabolismo , Ferro/análise , Ferro/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Succímero/farmacologia
14.
Environ Toxicol Pharmacol ; 36(3): 1150-66, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24184500

RESUMO

Arsenic (As) widely studied for its effects as a neurotoxicant. The present study was designed to evaluate the protective effect of calcium, zinc or monoisoamyl dimercaptosuccinic acid (MiADMSA), either individually or in combination on As induced oxidative stress and apoptosis in brain regions (cerebral cortex, hippocampus and cerebellum) of postnatal day (PND) 21, 28 and 3 months old rats. Arsenic exposure significantly decreased the activities of superoxide dismutase (SOD) isoforms, catalase (CAT), glutathione peroxidase (GPx) and glutathione reductase (GR) with increase in glutathione s transferase (GST) while lipid peroxidation (LPx), arsenic levels, mRNA expression of caspase 3 and 9 were significantly increased in different brain regions. Arsenic induced alterations in these parameters were greater in PND 28 and more pronounced in cerebral cortex. From the results it is evident that combined supplementation of calcium and zinc along with MiADMSA would be most effective compared to individual administration in reducing arsenic induced neurotoxicity.


Assuntos
Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Arsênio/toxicidade , Encéfalo/enzimologia , Caspases/metabolismo , Quelantes/farmacologia , Metais/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Cálcio/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Metais/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Succímero/análogos & derivados , Succímero/farmacologia , Zinco/farmacologia
15.
Int J Dev Neurosci ; 31(7): 586-97, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23906897

RESUMO

Oxidative stress (OS) has been implicated in the pathophysiology of many neurodegenerative disorders. Several studies have shown that exposure to arsenic (As) and lead (Pb) produces oxidative stress, one of the most noted molecular mechanisms for the neurotoxicity of these metals. In the present study, we examined the effect of combined exposure to these metals (As and Pb) on the activity levels of antioxidant enzymes and apoptotic marker enzymes in brain regions (cerebral cortex, hippocampus and cerebellum) of rats at postnatal day (PND) 21, 28 and 3 months age and compared the toxicity levels with individual metals (As or Pb). Further, we also evaluated the therapeutic efficacy of a chelating agent, monoisoamyl dimercaptosuccinic acid (MiADMSA) against arsenic and lead induced developmental neurotoxicity. Pregnant rats were exposed to sodium meta-arsenite (50 ppm) and lead acetate (0.2%) individually, and in combination (As=25 ppm+Pb=0.1%) via drinking water throughout perinatal period (GD 6 to PND 21). MiADMSA (50 mg/kg, orally through gavage) was given for three consecutive days to the PND 18 pups (i.e., PND 18 to PND 20). Exposure to metal mixture resulted in a significant decrease in the activity levels of antioxidant enzymes such as manganese-superoxide dismutase (Mn-SOD), Cu/Zn superoxide dismutase (Cu/Zn-SOD), catalase (CAT) and glutathione peroxidase (GPx) while the malondialdehyde (MDA) levels and mRNA expression levels of caspase-3 and caspase-9 were significantly increased in all the three brain regions. The observed alterations were greater with exposure to metal mixture than individual metals (As or Pb) and the changes were more prominent at PND 28 and greater in cerebral cortex than hippocampus and cerebellum. Interestingly, chelation therapy with MiADMSA showed significant recovery in antioxidant enzymes, lipid peroxidation and gene expression levels of caspase-3 and caspase-9. From these findings, it can be concluded that combined exposure to As and Pb showed an additive effect on antioxidant enzymes than individual metal exposure and chelation therapy with MiADMSA significantly reversed the As and Pb induced apoptosis and oxidative stress, a major contributing factor to neurotoxicity.


Assuntos
Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Arseniatos/toxicidade , Encéfalo/efeitos dos fármacos , Chumbo/toxicidade , Succímero/análogos & derivados , Fatores Etários , Animais , Animais Recém-Nascidos , Arseniatos/metabolismo , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/ultraestrutura , Catalase/metabolismo , Embrião de Mamíferos , Feminino , Chumbo/metabolismo , Masculino , Malondialdeído/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Gravidez , Ratos , Ratos Wistar , Succímero/farmacologia , Superóxido Dismutase/metabolismo
16.
Neurotoxicology ; 35: 137-45, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23376091

RESUMO

Arsenic contaminated drinking water has affected more than 200 million people globally. Chronic arsenicism has also been associated with numerous neurological diseases. One of the prime mechanisms postulated for arsenic toxicity is reactive oxygen species (ROS) mediated oxidative stress. In this study, we explored the kinetic relationship of ROS with calcium and attempted to dissect the calcium ion channels responsible for calcium imbalance after arsenic exposure. We also explored if mono- or combinational chelation therapy prevents arsenic-induced (25ppm in drinking water for 4 months) neuronal apoptosis in a guinea pig animal model. Results indicate that chronic arsenic exposure caused a significant increase in ROS followed by NO and calcium influx. This calcium influx is mainly dependent on L-type voltage gated channels that disrupt mitochondrial membrane potential, increase bax/bcl2 levels and caspase 3 activity leading to apoptosis. Interestingly, blocking of ROS could completely reduce calcium influx whereas calcium blockage partially reduced ROS increase. While in general mono- and combinational chelation therapies were effective in reversing arsenic induced alteration, combinational therapy of DMSA and MiADMSA was most effective. Our results provide evidence for the role of L-type calcium channels in regulating arsenic-induced calcium influx and DMSA+MiADMSA combinational therapy may be a better protocol than monotherapy in mitigating chronic arsenicosis.


Assuntos
Apoptose/efeitos dos fármacos , Intoxicação por Arsênico/prevenção & controle , Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Quelantes/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Succímero/análogos & derivados , Animais , Intoxicação por Arsênico/metabolismo , Intoxicação por Arsênico/patologia , Arsenitos , Caspase 3/metabolismo , Citoproteção , Modelos Animais de Doenças , Quimioterapia Combinada , Cobaias , Cinética , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Compostos de Sódio , Succímero/farmacologia , Proteína X Associada a bcl-2/metabolismo
17.
Asian Pac J Cancer Prev ; 13(6): 2943-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22938487

RESUMO

Arsenic exposure is a serious health hazard worldwide. We have previously established that it may result in immune suppression by upregulating Th2 cytokines while downregulating Th1 cytokines and causing lymphocytic death. Treatment modalities for arsenic poisoning have mainly been restricted to the use of chelating agents in the past. Only recently have combination therapies using a chelating agent in conjunction with other compounds such as anti-oxidants, micronutrients and various plant products, been introduced. In the present study, we used T11TS, a novel immune potentiating glycopeptide alone and in combination with the sulfhydryl-containing chelator, mono-iso-amyl-dimarcaptosuccinic acid (MiADMSA) as a therapeutic regimen to combat arsenic toxicity in a mouse model. Results indicated that Th1 cytokines such as TNF-α, IFNγ, IL12 and the Th2 cytokines such as IL4, IL6, IL10 which were respectively downregulated and upregulated following arsenic induction were more efficiently restored to their near normal levels by T11TS alone in comparison with the combined regimen. Similar results were obtained with the apoptotic proteins studied, FasL, BAX, BCL2 and the caspases 3, 8 and 9, where again T11TS proved more potent than in combination with MiADMSA in preventing lymphocyte death. The results thus indicate that T11TS alone is more efficient in immune re-establishment after arsenic exposureas compared to combination therapy with T11TS+MiADMSA.


Assuntos
Intoxicação por Arsênico/tratamento farmacológico , Antígenos CD2/uso terapêutico , Quelantes/uso terapêutico , Succímero/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Arsênio/toxicidade , Antígenos CD2/farmacologia , Transformação Celular Neoplásica , Terapia por Quelação/métodos , Citocinas/metabolismo , Quimioterapia Combinada , Exposição Ambiental , Linfócitos/efeitos dos fármacos , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Succímero/farmacologia , Succímero/uso terapêutico
18.
J Ethnopharmacol ; 130(2): 414-20, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20580805

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Smilax glabra Roxb. is a traditional Chinese herb, the rhizome of Smilax glabra has been used in folk medicine for the treatment of lead poisoning. AIMS OF THE STUDY: The present study was conducted to investigate the protective role of Smilax glabra extract (SGE) individually or combined with meso-2,3-dimercaptosuccinic acid (DMSA) against the effects of lead acetate on oxidative stress and lead burden in rats. MATERIALS AND METHODS: The biochemical parameters and enzymes in different treated rats were determined by commercial kits. The metal concentrations were measured using atomic absorption spectrophotometer. RESULTS: SGE (300 mg/kg) showed very low toxicity to organs in non-lead exposed rats. Administration of SGE individually had no effect on blood zinc protoporphyrin (ZPP) level but significantly enhanced the glutathione (GSH) content and delta-aminolevulinic acid dehydratase (ALAD), alanine aminotransferase (ALT) and alkaline phosphatase (ALP) activities in lead exposed rats. The co-treatment of SGE and DMSA had a synergism in increasing brain, liver and kidney superoxide dismutase (SOD), catalase (CAT) activities and GSH level, and decreasing oxidized glutathione (GSSG) and thiobarbituric acid reactive substances (TBARS) levels. Moreover, the co-treatment could improve the hepatic and renal histopathology changes. SGE as chelating agent showed significant efficiency in reducing blood and tissue lead burden. CONCLUSIONS: The in vivo results suggested that SGE individually or combined with DMSA exhibited remarkable protective effects on lead-induced oxidative stress and lead burden in rats.


Assuntos
Antioxidantes/farmacologia , Intoxicação por Chumbo/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Smilax , Alanina Transaminase/sangue , Fosfatase Alcalina/sangue , Animais , Antídotos/farmacologia , Carga Corporal (Radioterapia) , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Catalase/metabolismo , Quelantes/farmacologia , Modelos Animais de Doenças , Flavonoides/análise , Glutationa/sangue , Rim/efeitos dos fármacos , Rim/metabolismo , Intoxicação por Chumbo/etiologia , Intoxicação por Chumbo/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Compostos Organometálicos/sangue , Fenóis/análise , Extratos Vegetais/química , Sintase do Porfobilinogênio/sangue , Protoporfirinas/sangue , Ratos , Ratos Wistar , Rizoma , Succímero/farmacologia , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
19.
Planta Med ; 76(3): 241-4, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19764011

RESUMO

It has been shown that garlic and its main bioactive component, allicin, as natural chelating agents can reduce blood and tissue lead content in animal models. In this study the effect of allicin, alone or combined with meso-2,3-dimercaptosuccinic acid (DMSA), in decreasing lead content of blood and tissues of mice was evaluated. Swiss albino mice were exposed to 1000 ppm of lead in water for 35 days and then placed in various treatment groups including groups administered oral allicin, DMSA, or their combination. The concentrations of lead in blood, kidney, liver, bone and brain were measured using atomic absorption spectrophotometry. Both, allicin and DMSA decreased the blood and tissue lead concentration. There was an additive effect of the combined administration of allicin and DMSA in reducing bone lead. No side effect was observed in all treated groups. Combined use of DMSA and allicin seems to be a better choice in the treatment of chronic lead intoxication.


Assuntos
Osso e Ossos/efeitos dos fármacos , Quelantes/uso terapêutico , Alho/química , Intoxicação por Chumbo/tratamento farmacológico , Chumbo/metabolismo , Succímero/uso terapêutico , Ácidos Sulfínicos/uso terapêutico , Animais , Osso e Ossos/metabolismo , Quelantes/farmacologia , Dissulfetos , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Chumbo/sangue , Camundongos , Fitoterapia , Succímero/farmacologia , Ácidos Sulfínicos/farmacologia
20.
Toxicol Appl Pharmacol ; 240(2): 255-64, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19595699

RESUMO

Lead, a ubiquitous and potent neurotoxicant causes oxidative stress which leads to numerous neurobehavioral and physiological alterations. The ability of lead to bind sulfhydryl groups or compete with calcium could be one of the reasons for its debilitating effects. In the present study, we addressed: i) if chelation therapy could circumvent the altered oxidative stress and prevent neuronal apoptosis in chronic lead-intoxicated rats, ii) whether chelation therapy could reverse biochemical and behavioral changes, and iii) if mono or combinational therapy with captopril (an antioxidant) and thiol chelating agents (DMSA/MiADMSA) is more effective than individual thiol chelator in lead-exposed rats. Results indicated that lead caused a significant increase in reactive oxygen species, nitric oxide, and intracellular free calcium levels along with altered behavioral abnormalities in locomotor activity, exploratory behavior, learning, and memory that were supported by changes in neurotransmitter levels. A fall in membrane potential, release of cytochrome c, and DNA damage indicated mitochondrial-dependent apoptosis. Most of these alterations showed significant recovery following combined therapy with captopril with MiADMSA and to a smaller extend with captopril+DMSA over monotherapy with these chelators. It could be concluded from our present results that co-administration of a potent antioxidant (like captopril) might be a better treatment protocol than monotherapy to counter lead-induced oxidative stress. The major highlight of the work is an interesting experimental evidence of the efficacy of combinational therapy using an antioxidant with a thiol chelator in reversing neurological dystrophy caused due to chronic lead exposure in rats.


Assuntos
Antioxidantes/farmacologia , Captopril/farmacologia , Quelantes/farmacologia , Intoxicação do Sistema Nervoso por Chumbo/tratamento farmacológico , Degeneração Neural/tratamento farmacológico , Neurônios/efeitos dos fármacos , Succímero/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Aminas Biogênicas/metabolismo , Cálcio/metabolismo , Citocromos c/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Quimioterapia Combinada , Comportamento Exploratório/efeitos dos fármacos , Intoxicação do Sistema Nervoso por Chumbo/metabolismo , Intoxicação do Sistema Nervoso por Chumbo/patologia , Intoxicação do Sistema Nervoso por Chumbo/fisiopatologia , Aprendizagem/efeitos dos fármacos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Memória/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Atividade Motora/efeitos dos fármacos , Degeneração Neural/induzido quimicamente , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico/metabolismo , Compostos Organometálicos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Succímero/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA