Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 283
Filtrar
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.
Nanotoxicology ; 16(9-10): 829-843, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36660964

RESUMO

Iron oxide nanoparticles (IONPs) are mostly intended to be administrated intravenously, understanding the interaction of IONPs with vascular endothelial cells is extremely crucial for developing safe application regimes of IONPs. In this work, interactions of three kinds of IONPs to endothelial cells were investigated both in human umbilical vein endothelial cells (HUVECs) and in healthy mice. Both meso-2,3-dimercaptosuccinic acid (DMSA) coated Fe3O4 NPs (DMSA-Fe3O4 NPs) and DMSA-Fe2O3 NPs induced cell growth inhibition, while polyglucose sorbitol carboxymethyether coated Fe2O3 NPs(PSC-Fe2O3 NPs) did not. The PSC coating inhibited the cellular uptake of the IONPs. Both DMSA-Fe3O4 and DMSA-Fe2O3 NPs induced ferroptosis of HUVEC through upregulating phospholipid peroxides, which could be inhibited by typical ferroptosis inhibitors ferrostatin-1, Trolox and deferoxamine. Moreover, transforming growth factor beta 1 (TGFß1) was upregulated by DMSA-Fe3O4 NPs at protein and gene level. The inhibitor of TGFß1 receptor LY210 could reduce the effect. When being intravenously injected in mice, DMSA-Fe3O4 NPs were observed locating in the liver, increased the levels of lipid peroxidation (4-hydroxynonenal), acyl-CoA synthetase long-chain family member 4(ACSL4) and TGFß1, indicating ferroptosis occurrence in vivo. The ferroptosis of vascular endothelial cells in exposure with IONPs depended on the surface coating and core chemistry of the NPs. Both DMSA-Fe3O4 NPs and DMSA-Fe2O3 NPs could induce the ferroptosis of endothelial cells, while PSC-Fe2O3 NPs did not induce ferroptosis and apoptosis possibly due to the very low cellular uptake. DMSA-Fe3O4 NPs and TGFß1 formed feedforward loop to induce ferroptosis.


Assuntos
Ferroptose , Nanopartículas , Camundongos , Humanos , Animais , Compostos Férricos , Células Endoteliais da Veia Umbilical Humana , Succímero/farmacologia , Nanopartículas Magnéticas de Óxido de Ferro
4.
Drug Chem Toxicol ; 45(6): 2448-2453, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34348527

RESUMO

Tungsten (W) and its compounds have emerged as a relatively new area of environmental health concern in the last decade. Tungsten is environmentally benign due to its increasing use in armour-piercing munitions and as a replacement for lead in other ammunition. It has also been identified in various hazardous waste sites and therefore been proposed for inclusion in the Environmental Protection Agency National Priorities List. The major objective of this study was to evaluate the therapeutic efficacy of orally administered monoisoamyl 2, 3-dimercaptosuccinic acid (MiADMSA) against tungstate induced oxidative injury in blood, liver and kidneys of male Wistar rats. MiADMSA, a thiol chelator has gained wide recognition recently as a future chelating drug of choice specifically for arsenic and was chosen for this study as tungstate ions too have an affinity toward the -SH group thus, being less bioavailable in the body. We determined the effects of MiADMSA (50 mg/kg, p.o.) against sodium tungstate (500 ppm in drinking water, daily for 28 days) induced biochemical changes indicative of oxidative stress in blood, and other soft tissues of of male Wistar rats. Tungsten exposure led to an increased levels of Reactive Oxygen Species (ROS) in liver, kidney, spleen and blood accompanied also by an increase in TBARS levels. The GSH: GSSG ratio also showed a decrease on sodium tungstate intoxication. Treatment with MiADMSA restored most of the sodium tungstate-induced alterations in the biomarkers suggestive of oxidative stress. These preliminary results led us to conclude that sub-acute exposure to tungstate-induced oxidative stress could be effectively reduced by the administration of MiADMSA and thus might be a promising antidote for studying in detail its efficacy in reducing body tungstate burden and its excretion post tungstate exposure.


Assuntos
Arsênio , Succímero , Animais , Masculino , Ratos , Antídotos/farmacologia , Biomarcadores , Quelantes/farmacologia , Dissulfeto de Glutationa/farmacologia , Estresse Oxidativo , Ratos Wistar , Espécies Reativas de Oxigênio , Succímero/farmacologia , Succímero/uso terapêutico , Substâncias Reativas com Ácido Tiobarbitúrico , Tungstênio/efeitos adversos
5.
Drug Deliv ; 28(1): 1785-1794, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34470555

RESUMO

The probable nanotoxicity to human health and the environment is a significant challenge for the sustainable application of nanomaterials in medicine. The cytototoxical effect of succimer (meso-2,3-dimercaptosuccinic acid-DMSA) coated titanium dioxide (DMSA-TiO2) with cultured human aortic endothelial cells (HAoECs) was assessed in this investigation. Our findings have shown that DMSA-TiO2 can be accumulated in HAoECs and dispersed in a cytoplasm on the culture medium. DMSA-cytotoxicity TiO2 effects were dose-responsive, and the concentrations were of little toxicity, and MTT stain testing showed that they had only 0.02 mg ml-1. Meanwhile, the lactate dehydrogenase biomarker was not considerably more remarkable than the biomarker from untreated (control) cells (free DMSA-TiO2). Though, also without any apparent signs of cell damage, the endocrine functions for prostacyclin I-2 and endothelin-1 and the urea transporter functions were modified. In addition, in vitro endothelial tube development has been shown that HAoECs could induce angiogenesis even with small amounts of DMSA-TiO2 (0.01 and 0.02 mg ml-1). Further, we have examined the in vivo toxicity and biochemical parameter by animal model. Furthermore, in vivo assessments designated that the resulting DMSA-TiO2 presented synergistic activities of angiogenesis activity. Overall, these findings show the cytotoxicity of DMSA-TiO2 and could induce adverse effects on normal endothelial cells.


Assuntos
Aorta/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Nanoestruturas/química , Succímero/farmacologia , Titânio/farmacologia , Animais , Biomarcadores , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Endogâmicos ICR , Distribuição Aleatória , Succímero/química , Titânio/química
6.
Drug Deliv ; 28(1): 1455-1465, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34236248

RESUMO

Chromium poisoning has become one of the most common heavy metal poisoning occupational diseases with high morbidity and mortality. However, most antidotes detoxify the whole body and are highly toxic. To achieve hepato-targeted chromium poisoning detoxification, a novel hepato-targeted strategy was developed using aging erythrocyte membranes (AEMs) as biomimetic material coated with a dimercaptosuccinic acid (DMSA) nanostructured lipid carrier to construct a biomimetic nano-drug delivery system. The particle size, potential, drug loading, encapsulation rate, in vitro release, and stability of the nanoparticles (NPs) were characterized. Confocal microscopy and flow cytometry showed that the prepared NPs could be phagocytized by RAW264.7 macrophage cells. The efficacy of AEM-DMSA-NPs for targeted liver detoxification was evaluated by in vitro MTT analysis and an in vivo model of chromium poisoning. The results showed that the NPs could safely and efficiently achieve targeted liver chromium poisoning detoxification. All the results indicated that the biomimetic nano-drug delivery system mediated by aging erythrocyte membranes and containing DMSA nanoparticles could be used as a novel therapeutic drug delivery system potentially targeting liver detoxification.


Assuntos
Antídotos/farmacologia , Materiais Biomiméticos/metabolismo , Cromo/envenenamento , Membrana Eritrocítica/metabolismo , Nanopartículas/química , Succímero/farmacologia , Animais , Antídotos/administração & dosagem , Antídotos/farmacocinética , Química Farmacêutica , Portadores de Fármacos , Liberação Controlada de Fármacos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Tamanho da Partícula , Células RAW 264.7 , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Succímero/administração & dosagem , Succímero/farmacocinética
7.
Arch Toxicol ; 95(8): 2643-2657, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34165617

RESUMO

N,N'-bis(2-mercaptoethyl)isophthalamide (NBMI) is a novel lipophilic heavy metal chelator and thiol redox antioxidant. This study was designed to investigate the neuroprotective activity of NBMI in U-87 MG cells exposed to lead acetate (PbAc). Cells were pretreated with NBMI for 24 h prior to a 48 h exposure to PbAc. Cell death (55%, p < 0.0001) and reduction of intracellular GSH levels (0.70-fold, p < 0.005) induced by 250 µM Pb were successfully attenuated by NBMI pretreatment at concentrations as low as 10 µM. A similar pretreatment with the FDA-approved Pb chelator dimercaptosuccinic acid (DMSA) proved ineffective, indicating a superior PKPD profile for NBMI. Pretreatment with NBMI successfully counteracted Pb-induced neuroinflammation by reducing IL-1ß (0.59-fold, p < 0.05) and GFAP expression levels. NBMI alone was also found to significantly increase ferroportin expression (1.97-fold, p < 0.05) thereby enhancing cellular ability to efflux heavy metals. While no response was observed on the apoptotic pathway, this study demonstrated for the first time that necrotic cell death induced by Pb in U-87 MG cells is successfully attenuated by NBMI. Collectively these data demonstrate NBMI to be a promising neuroprotective compound in the realm of Pb poisoning.


Assuntos
Derivados de Benzeno/farmacologia , Quelantes/farmacologia , Fármacos Neuroprotetores/farmacologia , Compostos Organometálicos/toxicidade , Compostos de Sulfidrila/farmacologia , Proteínas de Transporte de Cátions/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , Doenças Neuroinflamatórias/etiologia , Doenças Neuroinflamatórias/prevenção & controle , Succímero/farmacologia
8.
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
9.
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
10.
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
11.
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
12.
Biomed Pharmacother ; 128: 110257, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32474354

RESUMO

BACKGROUND AND PURPOSE: Arsenicosis is a major threat to public health and is a major cause of the development of urinary bladder cancer. Oxidative/ nitrosative stress is one of the key factors for these effects but the involvement of other associated factors is less known. There is a lack of data for the efficacy of chelator against urinary bladder carcinogenesis. The present study demonstrates the early signs of arsenic exposed urinary bladder carcinogenesis and its attenuation by Monoisoamyl dimercaptosuccinic acid (MiADMSA). METHODS: Male rats were exposed to 50 ppm of sodium arsenite and dimethylarsinic acid (DMA) via drinking water for 18 weeks and treated with MiADMSA (50 mg/kg, orally once daily for 5 days) for 3 weeks with a gap one week between the two courses of treatments. We compared in vivo data with in vitro by co-exposing 100 nM of sodium arsenite and DMA to rat (NBT-II) as well as human transitional epithelial carcinoma (T-24) cells with 100 nM of MiADMSA. RESULTS: The data showed that sodium arsenite and DMA exposure significantly increased the tissue arsenic contents, ROS, TBARS levels, catalase, SOD activities and significantly decreased GSH level which might be responsible for an increased 8-OHdG level. These changes might have increased pro-oncogenic biomarkers like MMP-9 and survivin in serum, bladder tissues, NBT-II, and T-24 cells. High cell migration and clonogenic potential in NBT-II and T-24 cells exposed to arsenic suggest pronounced carcinogenic potential. Significant recovery in these biomarkers was noted on treatment with MiADMSA. CONCLUSION: Early signs of urinary bladder carcinogenesis were observed in arsenic and DMA exposed rats which were linked to metal accumulation, oxidative/ nitrosative stress, 8-OHdG, MMP-9 and survivin which were reduced by MiADMSA possibly via its efficient chelation abilities in vivo and in vitro.


Assuntos
Anticarcinógenos/farmacologia , Arsenitos , Ácido Cacodílico , Carcinoma de Células de Transição/prevenção & controle , Transformação Celular Neoplásica/efeitos dos fármacos , Quelantes/farmacologia , Compostos de Sódio , Succímero/análogos & derivados , Neoplasias da Bexiga Urinária/prevenção & controle , 8-Hidroxi-2'-Desoxiguanosina/metabolismo , Animais , Carcinoma de Células de Transição/induzido quimicamente , Carcinoma de Células de Transição/metabolismo , Carcinoma de Células de Transição/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Dano ao DNA , Humanos , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Estresse Nitrosativo/efeitos dos fármacos , Ratos Sprague-Dawley , Succímero/farmacologia , Survivina/metabolismo , Neoplasias da Bexiga Urinária/induzido quimicamente , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia
13.
Biomolecules ; 10(2)2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32033229

RESUMO

: High arsenic (As) levels in food and drinking water, or under some occupational conditions, can precipitate chronic toxicity and in some cases cancer. Millions of people are exposed to unacceptable amounts of As through drinking water and food. Highly exposed individuals may develop acute, subacute, or chronic signs of poisoning, characterized by skin lesions, cardiovascular symptoms, and in some cases, multi-organ failure. Inorganic arsenite(III) and organic arsenicals with the general formula R-As2+ are bound tightly to thiol groups, particularly to vicinal dithiols such as dihydrolipoic acid (DHLA), which together with some seleno-enzymes constitute vulnerable targets for the toxic action of As. In addition, R-As2+-compounds have even higher affinity to selenol groups, e.g., in thioredoxin reductase that also possesses a thiol group vicinal to the selenol. Inhibition of this and other ROS scavenging seleno-enzymes explain the oxidative stress associated with arsenic poisoning. The development of chelating agents, such as the dithiols BAL (dimercaptopropanol), DMPS (dimercapto-propanesulfonate) and DMSA (dimercaptosuccinic acid), took advantage of the fact that As had high affinity towards vicinal dithiols. Primary prevention by reducing exposure of the millions of people exposed to unacceptable As levels should be the prioritized strategy. However, in acute and subacute and even some cases with chronic As poisonings chelation treatment with therapeutic dithiols, in particular DMPS appears promising as regards alleviation of symptoms. In acute cases, initial treatment with BAL combined with DMPS should be considered.


Assuntos
Antídotos/uso terapêutico , Intoxicação por Arsênico/tratamento farmacológico , Arsênio/toxicidade , Quelantes/uso terapêutico , Animais , Antídotos/química , Antídotos/farmacologia , Arsênio/efeitos adversos , Intoxicação por Arsênico/etiologia , Intoxicação por Arsênico/metabolismo , Arsenicais/efeitos adversos , Quelantes/química , Quelantes/farmacologia , Dimercaprol/análogos & derivados , Dimercaprol/farmacologia , Dimercaprol/uso terapêutico , Água Potável/efeitos adversos , Humanos , Modelos Moleculares , Exposição Ocupacional/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Succímero/química , Succímero/farmacologia , Succímero/uso terapêutico , Unitiol/química , Unitiol/farmacologia , Unitiol/uso terapêutico , Poluentes Químicos da Água/efeitos adversos , Poluentes Químicos da Água/toxicidade
14.
J Trace Elem Med Biol ; 58: 126429, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31760328

RESUMO

BACKGROUND AND AIM: Environmental lead (Pb) exposure damages the lungs and is a risk factor for death from cardiovascular disease. Pb induces toxicity by a mechanism, which involves alteration of the essential elements homeostasis. In this study we compare the effects of salinomycin (Sal), monensin (Mon) and meso-2,3-dimercaptosuccinic acid (DMSA) on the concentrations of lead (Pb), calcium (Ca), copper (Cu), iron (Fe) and zinc (Zn) in the lungs and heart of lead-exposed mice. METHODS: Sixty days old male ICR mice were divided into five groups: control (Ctrl) - untreated mice obtained distilled water for 28 days; Pb-intoxicated group (Pb) - exposed to 80 mg/kg body weight (BW) Pb(NO3)2 during the first 14 days of the experimental protocol; DMSA-treated (Pb + DMSA) - Pb-exposed mice, subjected to treatment with an average daily dose of 20 mg/kg BW DMSA for two weeks; Monensin-treated (Pb + Mon) - Pb-exposed mice, obtained an average daily dose of 20 mg/kg BW tetraethylammonium salt of monensic acid for 14 days; Pb + Sal - Pb-exposed mice, treated with an average daily dose of 20 mg/kg BW tetraethylammonium salt of salinomycinic acid for two weeks. On the 29th day of the experiment the samples (lungs and heart) were taken for atomic absorption analysis. RESULTS: The results revealed that exposure of mice to Pb for 14 days significantly increased the concentration of the toxic metal in both organs and elevated the cardiac concentrations of Ca, Cu and Fe compared to untreated mice. Pb exposure diminished the lung concentrations of Ca and Zn compared to that of untreated controls. DMSA, monensin and salinomycin decreased the concentration of Pb in the lungs and heart. Among the tested chelating agents, only salinomycin restored the cardiac Fe concentration to normal control values. CONCLUSION: The results demonstrated the potential application of polyether ionophorous antibiotic salinomycin as antidote for treatment of Pb-induced toxicity in the lungs and heart. The possible complexation of the polyether ionophorous antibiotics with Ca(II) and Zn(II), which can diminish the endogenous concentrations of both ions in the lungs should be taken into account.


Assuntos
Intoxicação por Chumbo/metabolismo , Pulmão/metabolismo , Metais Pesados/metabolismo , Monensin/farmacologia , Miocárdio/metabolismo , Piranos/farmacologia , Succímero/farmacologia , Animais , Cálcio/metabolismo , Cobre/metabolismo , Ferro/metabolismo , Chumbo/metabolismo , Masculino , Camundongos Endogâmicos ICR , Zinco/metabolismo
15.
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
16.
Environ Sci Pollut Res Int ; 26(32): 33304-33310, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31520384

RESUMO

In this study, we present experimental data on the effects of meso-2,3-dimercaptosuccinic acid (DMSA) and tetraethylammonium salt of salinomycinic acid (Sal) on cadmium-induced spleen dysfunction and altered essential metal balance in mice. Sixty-day-old male mice (ICR line) were randomly divided into four groups: untreated control group (Ctrl)-obtained distilled water for 28 days, toxic control group (Cd)-exposed to cadmium acetate dihydrate at average daily dose of 20mg/kg body weight (BW) for 14 days, Cd + DMSA group-obtained cadmium acetate dihydrate as the toxic control group followed by treatment with 20mg/kg BW DMSA for 2 weeks, and Cd + Sal group-mice exposed to cadmium acetate dihydrate at average daily dose of 20mg/kg BW for 2 weeks followed by administration of Sal at an average daily dose of 20mg/kg BW for 2 weeks. The compounds were administered orally via the drinking water of the animals. We found that cadmium exposure caused splenomegaly and reduced the hemoglobin and hematocrit levels and total red blood cell count compared with untreated controls. Cadmium intoxication of mice induced accumulation of the toxic metal ion in the blood and spleen. Alterations in the endogenous levels of calcium (Ca) and iron (Fe) in the spleen of cadmium-exposed mice compared with those in untreated controls were observed. Treatment of cadmium-exposed mice with DMSA or Sal recovered the spleen weight and hematological parameters to normal control values, decreased cadmium concentration in the blood and spleen, and improved splenic architecture. The results prove that Sal is a potential antidote for treatment of Cd-induced spleen dysfunction.


Assuntos
Substâncias Protetoras/farmacologia , Piranos/farmacologia , Baço/efeitos dos fármacos , Succímero/farmacologia , Poluentes Químicos da Água/toxicidade , Acetatos , Animais , Cádmio/toxicidade , Cálcio , Água Potável , Ferro , Masculino , Camundongos , Camundongos Endogâmicos ICR , Baço/fisiologia
17.
Indian J Pediatr ; 86(9): 784-789, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30859438

RESUMO

OBJECTIVE: To determine the performance of late dimercaptosuccinic acid (DMSA) renal scans in identifying high-grade (III-V) vesicoureteral reflux (VUR) in children aged over 3 y with a febrile urinary tract infection (fUTI) history that has not been timely investigated. METHODS: In this retrospective study of diagnostic accuracy, the clinical records of children aged between 3 and 18 y with fUTI history evaluated consecutively at Nephrology Unit of Hospital General de Niños Pedro de Elizalde, Argentina between 2006 and 2016 were reviewed. Patients with previously diagnosed renal or urinary tract abnormalities or who underwent previous postnatal genitourinary imaging were excluded. Only those assessed by renal and bladder ultrasound (RBUS), voiding cystourethrogram (VCUG) and late 6-mo DMSA scan were analyzed. The ability of the scintigraphy in identifying high-grade VUR was determined by comparing its findings with those of VCUG. RESULTS: In 122 children (median age 5.37 y, 88.5% girls) RBUS was abnormal in 53 (43.4%) and 58 (47.5%) had VUR (30 of high-grade). Abnormal DMSA scan findings (70 patients, 57.4%) were associated with all grade (p = 0.00001) and with high-grade VUR (p = 0.00001). Sensitivity, specificity, negative (NPV) and positive (PPV) predictive values of late DMSA scans for all grades VUR were 93.1%, 75%, 92.3% and 77.1%, respectively. Only 4 patients with low-grade VUR had normal scans. For high-grade VUR, sensitivity and NPV reached 100%. CONCLUSIONS: In older children, the normal late DMSA scan predicted the absence of high-grade VUR, obviating the need for a VCUG. This approach could be a possible strategy for children not studied at acute infection time.


Assuntos
Succímero/farmacologia , Infecções Urinárias/diagnóstico , Refluxo Vesicoureteral/diagnóstico , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Rim , Masculino , Cintilografia , Estudos Retrospectivos , Sensibilidade e Especificidade , Ultrassonografia , Bexiga Urinária
18.
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
19.
Toxicol Ind Health ; 35(2): 89-108, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30526433

RESUMO

In this study, we evaluated the therapeutic efficacy of monoisoamyldimercaptosuccinic acid (MiADMSA) against individual and combined effects of arsenic (As) and lead (Pb) on the monoaminergic system and behavioral functions in rats. Pregnant rats were exposed to sodium metaarsenite (50 ppm) and lead acetate (0.2%) individually and in combination (As = 25 ppm + Pb = 0.1%) via drinking water from gestation day (GD) 6 to postnatal day (PND) 21. MiADMSA (50 mg/kg body weight) was given orally through gavage for 3 consecutive days to pups from PND 18 to PND 20. The results showed increases in synaptosomal epinephrine, dopamine, and norepinephrine levels with individual metal exposures and decreases with combined exposure to As and Pb in the cortex, cerebellum, and hippocampus in PND 21, PND 28, and 3 months age-group rats. We found decreased activity of mitochondrial monoamine oxidase in the selected brain regions following individual and combined exposures to Pb and As. In addition, rats treated with Pb and As alone or in combination showed significant deficits in open-field behavior, grip strength, locomotor activity, and exploratory behavior at PND 28 and 3 months of age. However, MiADMSA administration showed reversal effects against the As- and/or Pb-induced impairments in the monoaminergic system as well as in behavioral functions of rats. Our data demonstrated that the mixture of Pb and As induced synergistic toxicity to developing brain leading to impairments in neurobehavioral functions and also suggest therapeutic efficacy of MiADMSA against Pb- and/or As-induced developmental neurotoxicity.


Assuntos
Arsênio/toxicidade , Encéfalo/efeitos dos fármacos , Chumbo/toxicidade , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/toxicidade , Succímero/análogos & derivados , Animais , Comportamento Animal/efeitos dos fármacos , Monoaminas Biogênicas/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiologia , Comportamento Exploratório/efeitos dos fármacos , Feminino , Masculino , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Wistar , Succímero/farmacologia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...