Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 435
Filtrar
1.
Neurotox Res ; 42(2): 24, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38598025

RESUMO

The investigation into the hippocampal function and its response to heavy metal exposure is crucial for understanding the mechanisms underlying neurotoxicity, this can potentially inform strategies for mitigating the adverse effects associated with heavy metal exposure. Melatonin is an essential neuromodulator known for its efficacy as an antioxidant. In this study, we aimed to determine whether melatonin could protect against Nickel (Ni) neurotoxicity. To achieve this, we performed an intracerebral injection of Ni (300 µM NiCl2) into the right hippocampus of male Wistar rats, followed by melatonin treatment. Based on neurobehavioral and neurobiochemical assessments, our results demonstrate that melatonin efficiently enhances Ni-induced behavioral dysfunction and cognitive impairment. Specifically, melatonin treatment positively influences anxious behavior, significantly reduces immobility time in the forced swim test (FST), and improves learning and spatial memory abilities. Moreover, neurobiochemical assays revealed that melatonin treatment modulates the Ni-induced alterations in oxidative stress balance by increasing antioxidant enzyme activities, such as superoxide dismutase (SOD) and catalase (CAT). Additionally, we observed that melatonin significantly attenuated the increased levels of lipid peroxidation (LPO) and nitric oxide (NO). In conclusion, the data from this study suggests that melatonin attenuates oxidative stress, which is the primary mechanism responsible for Ni-induced neurotoxicity. Considering that the hippocampus is the main structure involved in the pathology associated with heavy metal intoxication, such as Ni, these findings underscore the potential therapeutic efficacy of melatonin in mitigating heavy metal-induced brain damage.


Assuntos
Melatonina , Síndromes Neurotóxicas , Masculino , Ratos , Animais , Antioxidantes/farmacologia , Melatonina/farmacologia , Melatonina/uso terapêutico , Níquel/toxicidade , Ratos Wistar , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle
2.
Hum Exp Toxicol ; 43: 9603271231218707, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38487884

RESUMO

BACKGROUND: Bupivacaine (BUP), a long-acting local anesthetic, has been widely used in analgesia and anesthesia. However, evidence strongly suggests that excessive application of BUP may lead to neurotoxicity in neurons. Sphingosine kinase 2 (SPHK2) has been reported to exert neuroprotective effects. In this study, we intended to investigate the potential role and mechanism of SPHK2 in BUP-induced neurotoxicity in dorsal root ganglion (DRG) neurons. METHODS: DRG neurons were cultured with BUP to simulate BUP-induced neurotoxicity in vitro. CCK-8, LDH, and flow cytometry assays were performed to detect the viability, LDH activity, and apoptosis of DRG neurons. RT-qPCR and western blotting was applied to measure gene and protein expression. Levels. MeRIP-qPCR was applied for quantification of m6A modification. RIP-qPCR was used to analyze the interaction between SPHK2 and YTHDF1. RESULTS: SPHK2 expression significantly declined in DRG neurons upon exposure to BUP. BUP challenge substantially reduced the cell viability and increased the apoptosis rate in DRG neurons, which was partly abolished by SPHK2 upregulation. YTHDF1, an N6-methyladenosine (m6A) reader, promoted SPHK2 expression in BUP-treated DRG neurons in an m6A-dependent manner. YTHDF1 knockdown partly eliminated the increase in SPHK2 protein level and the protection against BUP-triggered neurotoxicity in DRG neurons mediated by SPHK2 overexpression. Moreover, SPHK2 activated the PI3K/AKT signaling to protect against BUP-induced cytotoxic effects on DRG neurons. CONCLUSIONS: In sum, YTHDF1-mediated SPHK2 upregulation ameliorated BUP-induced neurotoxicity in DRG neurons via promoting activation of the PI3K/AKT signaling pathway.


Assuntos
Bupivacaína , Síndromes Neurotóxicas , Fosfotransferases (Aceptor do Grupo Álcool) , Humanos , Bupivacaína/toxicidade , Regulação para Cima , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Síndromes Neurotóxicas/prevenção & controle , Apoptose , Proteínas de Ligação a RNA
3.
J Pharm Pharmacol ; 76(4): 368-380, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38330395

RESUMO

OBJECTIVES: To evaluate whether the glycosylation of chrysin (CHR) enhances its protective effects against aluminum-induced neurotoxicity. METHODS: To compare the antioxidant, anticholinesterase, and behavioral effects of CHR with its glycosylated form (CHR bonded to ß-d-glucose tetraacetate, denoted as LQFM280), we employed an integrated approach using both in vitro (SH-SY5Y cells) and in vivo (aluminum-induced neurotoxicity in Swiss mice) models. KEY FINDINGS: LQFM280 demonstrated higher antioxidant activity than CHR in both models. Specifically, LQFM280 exhibited the ability to exert antioxidant effects in the cytoplasm of SH-SY5Y cells, indicating its competence in traversing neuronal membranes. Remarkably, LQFM280 proved more effective than CHR in recovering memory loss and counteracting neuronal death in the aluminum chloride mice model, suggesting its increased bioavailability at the brain level. CONCLUSIONS: The glycosylation of CHR with ß-d-glucose tetraacetate amplifies its neuroprotective effects, positioning LQFM280 as a promising lead compound for safeguarding against neurodegenerative processes involving oxidative stress.


Assuntos
Flavonoides , Neuroblastoma , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Camundongos , Animais , Humanos , Alumínio/toxicidade , Glucose/farmacologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo , Antioxidantes/farmacologia , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/prevenção & controle , Linhagem Celular Tumoral
4.
Mar Drugs ; 22(2)2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38393033

RESUMO

The MTS cell viability test was used to screen a mini library of natural and synthetic 1,4-naphthoquinone derivatives (1,4-NQs) from marine sources. This screening identified two highly effective compounds, U-443 and U-573, which showed potential in protecting Neuro-2a neuroblastoma cells from the toxic effects of rotenone in an in vitro model of neurotoxicity. The selected 1,4-NQs demonstrated the capability to reduce oxidative stress by decreasing the levels of reactive oxygen species (ROS) and nitric oxide (NO) in Neuro-2a neuroblastoma cells and RAW 264.7 macrophage cells and displayed significant antioxidant properties in mouse brain homogenate. Normal mitochondrial function was restored and the mitochondrial membrane potential was also regained by 1,4-NQs after exposure to neurotoxins. Furthermore, at low concentrations, these compounds were found to significantly reduce levels of proinflammatory cytokines TNF and IL-1ß and notably inhibit the activity of cyclooxygenase-2 (COX-2) in RAW 264.7 macrophages. The results of docking studies showed that the 1,4-NQs were bound to the active site of COX-2, analogically to a known inhibitor of this enzyme, SC-558. Both substances significantly improved the behavioral changes in female CD1 mice with rotenone-induced early stage of Parkinson's disease (PD) in vivo. It is proposed that the 1,4-NQs, U-443 and U-573, can protect neurons and microglia through their potent anti-ROS and anti-inflammatory activities.


Assuntos
Naftoquinonas , Neuroblastoma , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Doença de Parkinson , Feminino , Camundongos , Animais , Rotenona/toxicidade , Ciclo-Oxigenase 2 , Naftoquinonas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/prevenção & controle , Fármacos Neuroprotetores/farmacologia
5.
Toxicology ; 503: 153740, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38316350

RESUMO

Bupivacaine, a common amide local anesthetic, can provide effective analgesia or pain relief but can also cause neurotoxicity, which remains a mounting concern in clinic and animal care. However, the precise underlying mechanisms have not been fully elucidated. A natural compound, notoginsenoside R1 (NG-R1) has been reported to exhibit a neuroprotective role in stress conditions. In this study, we explored the function and mechanism of NG-R1 in alleviating bupivacaine-induced neurotoxicity in mouse hippocampal neuronal (HT-22) and mouse neuroblastoma (Neuro-2a) cell lines. Our results exhibited that NG-R1 treatment can significantly rescue the decline of cell survival induced by bupivacaine. Tunel staining and western blotting showed that NG-R1 could attenuate BPV­induced cell apoptosis. Besides, we focused on Mcl1 as a potential target as it showed opposite expression tendency in response to NG-R1 and bupivacaine exposure. Mcl1 knockdown blocked the inhibitory effect of NG-R1 on cell apoptosis against bupivacaine treatment. Intriguingly, we found that NG-R1 can upregulate Mcl1 transcription by activating Stat3 and promote its nuclear translocation. In addition, NG-R1 can also promote Jak1 phosphorylation and docking analysis provide a predicted model for interaction between NG-R1 and phosphorylated Jak1. Taken together, our results demonstrated that NG-R1 can attenuate bupivacaine induced neurotoxicity by activating Jak1/Stat3/Mcl1 pathway.


Assuntos
Ginsenosídeos , Síndromes Neurotóxicas , Camundongos , Animais , Bupivacaína/toxicidade , Ginsenosídeos/farmacologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Síndromes Neurotóxicas/metabolismo , Linhagem Celular , Apoptose
6.
J Ethnopharmacol ; 326: 117735, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38211824

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Traditional plant-based medicines (TMs) have been widely used to prevent chronic oxaliplatin-induced peripheral neurotoxicity (OIPN). However, the prevention and safety of TMs for chronic OIPN remain ambiguous. Furthermore, diverse TM prescriptions and complicated components limit in-depth research on the mechanisms of TMs. AIM OF THIS STUDY: To determine core TMs and potential pharmacological pathways on the basis of a thorough investigation into the preventive benefits and safety of oral TMs for chronic OIPN in colorectal cancer (CRC). METHODS: A search of the PubMed, Cochrane, Embase, CNKI, VIP, and Wanfang databases for RCTs reporting on TMs for chronic OIPN was conducted through December 1, 2022. Subgroup analysis, sensitivity analysis and meta-regression were applied to assess the impacts of influencing variables. The assessment of Risk of Bias was relied on Cochrane Risk of Bias tool. The funnel plot, Egger's test, and the Trim and Fill method were applied to identify potential publication bias. Trial sequential analyses (TSA) were carried out by the TSA tool to increase the robustness. The assessment of the quality of evidence was according to the GRADE system. System pharmacology analysis was employed to screen core herbal combinations to elucidate possible mechanisms for preventing chronic OIPN in CRC. RESULTS: The pooled effect estimate with robustness increased by TSA analysis demonstrated that oral TMs appeared to significantly decrease the incidence of chronic OIPN (RR = 0.66, 95% CI (0.56, 0.78); P<0.00001), leukocytopenia (RR = 0.65, 95% CI (0.54,0.79); P<0.00001), and nausea and vomiting (RR = 0.72, 95% CI (0.61,0.84); P<0.0001) as well as improve the Objective Response Rate (ORR) (RR = 1.31, 95% CI (1.09,1.56); P = 0.003). The incidence of severe chronic OIPN was revealed a significant reduction, particularly when chemotherapy was administered for periods of time shorter than six months (RR = 0.33, 95% CI (0.15,0.71); P = 0.005; actuation duration<3 months; RR = 0.33, 95% CI (0.17,0.62); P = 0.0007; actuation duration≥3 months, <6 months). The considerable heterogeneity among studies may be attributable to the severity of dysfunction categorized by grade and accumulated dosage. Using core TMs consisting of Astragalus membranaceus (Fisch.) Bunge, Atractylodes Macrocephala Koidz., Poria cocos (Schw.) Wolf, and Codonopsis pilosula (Franch.) Nannf. To regulate nuclear factor-kappa B against inflammation caused by activation of microglia might be an approach to preventing chronic OIPN. CONCLUSIONS: TMs appear to be effective and safe in the prevention of chronic OIPN, especially severe chronic OIPN. Additionally, core TMs consisting of Astragalus membranaceus (Fisch.) Bunge, Atractylodes Macrocephala Koidz., Poria cocos (Schw.) Wolf, and Codonopsis pilosula (Franch.) Nannf were presumably responsible for reducing the incidence of chronic OIPN, and the mechanism may be related to relieving inflammation. However, quality-assured trials with long-term follow-up for exploring inflammatory factors and preliminary research on core TMs and pharmacological pathways are needed.


Assuntos
Neoplasias Colorretais , Síndromes Neurotóxicas , Lobos , Animais , Humanos , Oxaliplatina/efeitos adversos , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Neoplasias Colorretais/tratamento farmacológico , Inflamação
7.
Basic Clin Pharmacol Toxicol ; 134(3): 293-309, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37697481

RESUMO

BACKGROUND: Phytocannabinoids (pCBs) have been shown to inhibit the aggregation and neurotoxicity of the neurotoxic Alzheimer's disease protein beta amyloid (Aß). We characterized the capacity of six pCBs-cannabichromene (CBC), cannabigerol (CBG), cannabinol (CBN), cannabidivarin (CBDV), cannabidiol (CBD) and Δ9 -tetrahydrocannabinol (Δ9 -THC)-to disrupt Aß aggregation and protect against Aß-evoked neurotoxicity in PC12 cells. METHODS: Neuroprotection against lipid peroxidation and Aß-induced cytotoxicity was assessed using the MTT assay. Transmission electron microscopy was used to visualize pCB effects on Aß aggregation and fluorescence microscopy, with morphometrics and principal component analysis to assess PC12 cell morphology. RESULTS: CBD inhibited lipid peroxidation with no significant effect on Aß toxicity, whilst CBN, CBDV and CBG provided neuroprotection. CBC, CBG and CBN inhibited Aß1-42 -induced neurotoxicity in PC12 cells, as did Δ9 -THC, CBD and CBDV. CBC, CBN and CBDV inhibited Aß aggregation, whilst Δ9 -THC reduced aggregate density. Aß1-42 induced morphological changes in PC12 cells, including a reduction in neuritic projections and rounded cell morphology. CBC and CBG inhibited this effect, whilst Δ9 -THC, CBD and CBDV did not alter Aß1-42 effects on cell morphology. CONCLUSIONS: These findings highlight the neuroprotective activity of CBC, CBG and CBN as novel pCBs associated with variable effects on Aß-evoked neurite damage and inhibition of amyloid ß aggregation.


Assuntos
Canabidiol , Canabinoides , Síndromes Neurotóxicas , Ratos , Animais , Canabinol , Peptídeos beta-Amiloides/toxicidade , Células PC12 , Canabidiol/farmacologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Dronabinol/farmacologia
8.
J Agric Food Chem ; 71(51): 20724-20734, 2023 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-38098161

RESUMO

Amyloid-ß (Aß) is one of the core factors in the pathogenesis of Alzheimer's disease (AD), and the accumulation of its aggregates in the brain can form age-related plaques, leading to brain cell damage and intellectual decline, which may be the common intersection of all causes of neurotoxicity. Jujuboside B (JUB) has many characteristics such as hypnosis, sedation, antianxiety, and antioxidant stress. However, it is still unclear whether JuB can alleviate the neurotoxicity caused by Aß. Our study demonstrates that JUB improves learning and memory deficits in the nematode model. At the same time, JUB increases the antioxidant activity, prevents excessive accumulation of lipid synthesis, and resists endogenous lipofuscin deposition, thereby inhibiting the toxic effect of Aß. In vitro, JUB can improve Aß1-42-induced neuronal apoptosis level through the Bax/Bcl-2/caspase-3 signaling pathway and restore mitochondrial function in SH-SY5Y cells. The network pharmacology has been used to predict the potential neuroprotective mechanism of JUB. In summary, JUB exhibits neuroprotective properties employing both a neural cell and a nematode, which provides a basis for screening candidate ingredients for preventing AD.


Assuntos
Doença de Alzheimer , Neuroblastoma , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Humanos , Farmacologia em Rede , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Linhagem Celular Tumoral , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/tratamento farmacológico , Antioxidantes/farmacologia , Apoptose , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/prevenção & controle , Fragmentos de Peptídeos/farmacologia
9.
Best Pract Res Clin Anaesthesiol ; 37(1): 52-62, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37295854

RESUMO

Over the past 20 years, hundreds of preclinical studies of the developing central nervous system have been published concluding that the common γ-aminobutryic acid and N-methyl-d-aspartate binding anesthetic agents cause neuroapoptosis and other forms of neurodegeneration. Some clinical studies, including controlled trials, both prospective and ambidirectional in design, indicate an association between any exposure (single or multiple) to anesthesia and surgery at a young age, generally less than 3-4 years, and later behavioral and neurodevelopmental problems. A consideration of neuroprotective strategies is important, as scientists and clinicians alike ponder methods to potentially improve the neurodevelopmental outcomes of the millions of infants and children who undergo surgery and anesthesia annually around the world. This review will address plausible neuroprotective strategies and include alternative anesthetics, neuroprotective nonanesthetic drugs, and physiologic neuroprotection.


Assuntos
Anestesia , Anestésicos , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Criança , Pré-Escolar , Humanos , Anestesia/efeitos adversos , Anestésicos/toxicidade , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Estudos Prospectivos , Lactente
10.
Toxicol Mech Methods ; 33(7): 607-623, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37051630

RESUMO

Tramadol (TRA) causes neurotoxicity whereas trimetazidine (TMZ) is neuroprotective. The potential involvement of the PI3K/Akt/mTOR signaling pathway in the neuroprotection of TMZ against TRA-induced neurotoxicity was evaluated. Seventy male Wistar rats were divided into groups. Groups 1 and 2 received saline or TRA (50 mg/kg). Groups 3, 4, and 5 received TRA (50 mg/kg) and TMZ (40, 80, or 160 mg/kg) for 14 days. Group 6 received TMZ (160 mg/kg). Hippocampal neurodegenerative, mitochondrial quadruple complex enzymes, phosphatidylinositol-3-kinases (PI3Ks)/protein kinase B levels, oxidative stress, inflammatory, apoptosis, autophagy, and histopathology were evaluated. TMZ decreased anxiety and depressive-like behavior induced by TRA. TMZ in tramadol-treated animals inhibited lipid peroxidation, GSSG, TNF-α, and IL-1ß while increasing GSH, SOD, GPx, GR, and mitochondrial quadruple complex enzymes in the hippocampus. TRA inhibited Glial fibrillary acidic protein expression and increased pyruvate dehydrogenase levels. TMZ reduced these changes. TRA decreased the level of JNK and increased Beclin-1 and Bax. TMZ decreased phosphorylated Bcl-2 while increasing the unphosphorylated form in tramadol-treated rats. TMZ activated phosphorylated PI3Ks, Akt, and mTOR proteins. TMZ inhibited tramadol-induced neurotoxicity by modulating the PI3K/Akt/mTOR signaling pathways and its downstream inflammatory, apoptosis, and autophagy-related cascades.


Assuntos
Fármacos Neuroprotetores , Síndromes Neurotóxicas , Tramadol , Trimetazidina , Masculino , Ratos , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/farmacologia , Trimetazidina/farmacologia , Tramadol/toxicidade , Neuroproteção , Ratos Wistar , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Serina-Treonina Quinases TOR/farmacologia , Apoptose , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Autofagia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico
11.
Support Care Cancer ; 31(5): 293, 2023 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-37086308

RESUMO

PURPOSE: Chemotherapy-induced peripheral neurotoxicity (CIPN) is a highly prevalent, dose-limiting, costly, and tough-to-treat adverse effect of several chemotherapy agents, presenting as sensory and motor dysfunction in the distal extremities. Due to limited effective treatments, CIPN can permanently reduce patient function, independence, and quality of life. One of the most promising interventions for CIPN is physical therapy which includes exercise, stretching, balance, and manual therapy interventions. Currently, there are no physical therapy guidelines for CIPN, thus limiting its uptake and potential effectiveness. METHODS: Utilizing the authors' collective expertise spanning physical therapy, symptom management research, oncology, neurology, and treating patients with CIPN, we propose a comprehensive clinical workflow for physical therapists to assess and treat CIPN. This workflow is based on (1) physical therapy guidelines for treating neurologic symptoms like those of CIPN, (2) results of clinical research on physical therapy and exercise, and (3) physical therapy clinical judgement. RESULTS: We present detailed tables of pertinent physical therapy assessment and treatment methods that can be used in clinical settings. CIPN assessment should include detailed sensory assessment, objective strength assessments of involved extremities, and validated physical performance measures incorporating static and dynamic balance, gait, and functional mobility components. CIPN treatment should involve sensorimotor, strength, balance, and endurance-focused interventions, alongside a home-based exercise prescription that includes aerobic training. We conclude with action items for oncology teams, physical therapists, patients, and researchers to best apply this framework to address CIPN. CONCLUSIONS: Physical therapists are in a unique position to help assess, prevent, and treat CIPN given their training and prevalence, yet there are no physical therapy clinical practice guidelines for CIPN. Our preliminary suggestions for CIPN assessments and treatments can catalyze the development of guidelines to assess and treat CIPN. We urge oncology teams, physical therapists, patients, and researchers to develop, adapt, and disseminate this framework to help alleviate the burden of chemotherapy on patients with cancer.


Assuntos
Antineoplásicos , Síndromes Neurotóxicas , Doenças do Sistema Nervoso Periférico , Fisioterapeutas , Humanos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/diagnóstico , Doenças do Sistema Nervoso Periférico/terapia , Qualidade de Vida , Síndromes Neurotóxicas/diagnóstico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Antineoplásicos/efeitos adversos
12.
Food Chem Toxicol ; 175: 113691, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36871878

RESUMO

Arsenic is a notorious metalloid that exists in the earth's crust and is considered toxic for humans and the environment. Both cancerous and non-cancerous complications are possible after arsenic exposure. Target organs include the liver, lungs, kidney, heart, and brain. Arsenic-induced neurotoxicity, the main focus of our study, can occur in central and peripheral nervous systems. Symptoms can develop in a few hours, weeks, or years depending on the quantity of arsenic and the duration of exposure. In this review, we aimed to gather all the compounds, natural and chemical, that have been studied as protective agents in cellular, animal, and human reports. Oxidative stress, apoptosis, and inflammation are frequently described as destructive mechanisms in heavy metal toxicity. Moreover, reduced activity of acetylcholinesterase, the altered release of monoamine neurotransmitters, down-regulation of N-methyl-D-aspartate receptors, and decreased brain-derived neurotrophic factor are important underlying mechanisms of arsenic-induced neurotoxicity. As for neuroprotection, though some compounds have yet limited data, there are others, such as curcumin, resveratrol, taurine, or melatonin which have been studied more deeply and might be closer to a reliable protective agent. We collected the available information on all protective agents and the mechanisms by which they fight against arsenic-induced neurotoxicity.


Assuntos
Arsênio , Melatonina , Síndromes Neurotóxicas , Animais , Humanos , Arsênio/toxicidade , Acetilcolinesterase , Estresse Oxidativo , Melatonina/farmacologia , Encéfalo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle
13.
J Pediatr Hematol Oncol ; 45(1): 1-11, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36598958

RESUMO

PURPOSE: To review all studies providing evidence of the correlation between folinic acid (FA) rescue inadequacy and long-term cognitive damage in neuropsychological studies of children with acute lymphoblastic leukemia or osteogenic sarcoma treated under protocols using high-dose methotrexate and FA rescue. METHODS: A comprehensive literature search was performed of all databases of the Web of Science Citation Index, during 1990-2020, for the terms: neuropsychological, neurocognitive, and cognitive, together with acute lymphoblastic (and lymphocytic) leukemia and osteogenic sarcoma. English-language peer-reviewed articles on neuropsychological assessments of children who had been treated with high-dose methotrexate without irradiation, and which included details of methotrexate and FA schedules, were selected. In addition, a personal database of over 500 reprints of articles from over 130 journals was reviewed on the subjects of methotrexate and FA and their side effects. RESULTS: Three groups of studies were found and analyzed, with (1) no evidence of cognitive deterioration, (2) evidence of cognitive deterioration, and (3) more than 1 protocol grouped together, preventing separate analysis of any protocols, Protocols without cognitive deterioration reported adequate FA rescue, and those with cognitive deterioration reported inadequate FA rescue. CONCLUSION: Neuropsychological evaluation supported inadequate FA being the cause of neurocognitive damage after high-dose methotrexate and that adequate FA rescue prevents this complication.


Assuntos
Neoplasias Ósseas , Leucovorina , Metotrexato , Síndromes Neurotóxicas , Osteossarcoma , Leucemia-Linfoma Linfoblástico de Células Precursoras , Criança , Humanos , Neoplasias Ósseas/tratamento farmacológico , Leucovorina/uso terapêutico , Metotrexato/efeitos adversos , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Síndromes Neurotóxicas/tratamento farmacológico , Osteossarcoma/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico
14.
Toxicol Mech Methods ; 33(2): 95-103, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35702031

RESUMO

Colistin is an effective antibiotic against multidrug-resistant gram-negative bacterial infections; however, neurotoxic effects are fundamental dose-limiting factors for this treatment. Stem cell therapy is a promising method for treating neuronal diseases. Multipotent mesenchymal stromal cells (MSC) represent a promising source for regenerative medicine. Identification of neuroprotective agents that can be co-administered with colistin has the potential to allow the clinical application of this essential drug. This study was conducted to assess the potential protective effects of MSC, against colistin-induced neurotoxicity, and the possible mechanisms underlying any effect. Forty adult female albino rats were randomly classified into four equal groups; the control group, the MSC-treated group (A single dose of 1 × 106/mL MSCs through the tail vein), the colistin-treated group (36 mg/kg/d colistin was given for 7 d) and the colistin and MSC treated group (36 mg/kg/d colistin was administered for 7 d, and 1 × 106/mL MSCs). Colistin administration significantly increased GFAP, NGF, Beclin-1, IL-6, and TNF-α immunreactivity intensity. MSC administration in colistin-treated rats partially restored each of these markers. Histopathological changes in brain tissues were also alleviated by MSC co-treatment. Our study reveals a critical role of inflammation, autophagy, and apoptosis in colistin-induced neurotoxicity and showed that they were markedly ameliorated by MSC co-administration. Therefore, MSC could represent a promising agent for prevention of colistin-induced neurotoxicity.


Assuntos
Células-Tronco Mesenquimais , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Animais , Feminino , Ratos , Antibacterianos/toxicidade , Apoptose , Colistina/toxicidade , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle
15.
Toxicol Mech Methods ; 33(1): 38-46, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35574720

RESUMO

This study investigated the pharmacological mechanism of kaempferol in the treatment of oxaliplatin-induced neuropathic pain by network pharmacological method and cells experiment. The kaempferol and disease target genes were obtained from several databases, including TCMSP, SwissTargetPrediction, GeneCards, and CTD. Then, the common target genes of drugs and diseases were obtained using Venny online tools. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional analyses were carried out to obtain the enriched molecular pathways associated with the kaempferol and disease. Finally, we constructed a neuropathic pain cell experiment to confirm the findings. 138 intersection targets were identified between targets of kaempferol and oxaliplatin-induced neurotoxicity. Enrichment analyses revealed that the IL-17 signaling pathway was associated with the therapeutic effects of kaempferol. Kaempferol down-regulated the mRNA expression levels of TNF-α, IL-6, and CCL2 in oxaliplatin-treated astrocytes. Our findings showed that kaempferol alleviated oxaliplatin-induced neurotoxicity via regulation of inflammation-related genes.


Assuntos
Medicamentos de Ervas Chinesas , Neuralgia , Síndromes Neurotóxicas , Humanos , Quempferóis/farmacologia , Oxaliplatina/toxicidade , Astrócitos , Bases de Dados Factuais , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Simulação de Acoplamento Molecular
16.
Pharm Biol ; 60(1): 1915-1924, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36205592

RESUMO

CONTEXT: Sevoflurane (Sev) is a commonly used surgical anaesthetic; it has neurotoxic effects on the brain. Echinatin (Ech) is reported to have anti-inflammatory and antioxidant activity. OBJECTIVE: This research confirms the effect of Ech on Sev-induced neurotoxicity and cognitive deficits. MATERIALS AND METHODS: Primary rat hippocampal neurons were treated with 4.1% Sev for 6 h in the presence of Ech (5, 10, and 20 µM) or vehicle, followed by a further 42 h of culture. Male Sprague-Dawley aged rats were divided into 6 groups (n = 6): control, Sev, Sev + Ech (20 mg/kg;), Sev + Ech (40 mg/kg), and Sev + Ech (80 mg/kg). Rats were intraperitoneally injected with Ech or vehicle 1 h before Sev exposure (2% Sev for 5 h). RESULTS: We found that Ech (5, 10, and 20 µM) elevated cell viability (1.29-, 1.51-, 1.68-fold) but mitigated apoptosis (23.87% vs. 16.48%, 12.72%, 9.02%), oxidative stress, and ferroptosis in hippocampal neurons with Sev treatment. Ech activated the Nrf2 expression in Sev-induced in vitro and in vivo models of anaesthetic neurotoxicity. Ech also weakened neurotoxicity in hippocampal neurons with Sev treatment by increasing Nrf2 expression level. Moreover, Ech alleviated hippocampus neurons apoptosis (19.38% vs. 16.05%, 11.71%, 8.88%), oxidative stress, and ferroptosis in rats with Sev treatment. Ech improved Sev-induced cognitive deficits in rats. CONCLUSIONS: Ech alleviates Sev-induced neurotoxicity and cognitive deficits by mitigation of ferroptosis and oxidative stress. Ech may be developed as a new promising therapeutic drug for treatment of cerebral nerve injury caused by surgical anaesthesia.


Assuntos
Sobrecarga de Ferro , Síndromes Neurotóxicas , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Apoptose , Chalconas , Cognição , Hipocampo , Sobrecarga de Ferro/metabolismo , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/prevenção & controle , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Sevoflurano/metabolismo , Sevoflurano/toxicidade
17.
Drug Des Devel Ther ; 16: 3447-3463, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36217449

RESUMO

Objective: The present study was designed to investigate the possible synergistic effects of melatonin with zinc in the prevention and treatment of oxaliplatin-induced neurotoxicity in rats. Methodology: Forty-eight male Wistar albino rats were used and randomly allocated into six groups: The negative control group, oxaliplatin group, zinc + oxaliplatin group, melatonin + oxaliplatin group, zinc + melatonin + oxaliplatin prevention-approach group, and zinc + melatonin + oxaliplatin treatment-approach group. The thermal nociceptive/hyperalgesia tests were performed. Brain tissue homogenate was used for measuring GFAP, NCAM, TNF α, MAPK 14, NF-kB, GPX, and SOD. Brain tissue was sent for histopathological and immunohistochemistry studies. Results: The combination therapies showed improvement in the behavioral tests. A significant increase in GPX and SOD with a significant decrease in GFAP levels resulted in the prevention approach. TNF α decreased significantly in the treatment approach. No significant changes were seen in NCAM, NFkB, and MAPK-14. The histopathological findings support the biochemical results. Additionally, immunohistochemistry revealed a significant attenuation of p53 and a non-significant decrease in Bcl2 levels in the combination groups. Conclusion: The combination of zinc with melatonin for the prevention approach was effective in attenuating neurotoxicity induced by oxaliplatin. The proposed mechanisms are boosting the antioxidant system and attenuating the expression of p53, GFAP, and TNF-α.


Assuntos
Melatonina , Síndromes Neurotóxicas , Doenças do Sistema Nervoso Periférico , Animais , Antioxidantes/metabolismo , Hiperalgesia , Masculino , Melatonina/farmacologia , NF-kappa B/metabolismo , Moléculas de Adesão de Célula Nervosa/efeitos adversos , Moléculas de Adesão de Célula Nervosa/metabolismo , Neuroproteção , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/prevenção & controle , Oxaliplatina/efeitos adversos , Estresse Oxidativo , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Zinco/farmacologia
18.
Arch Toxicol ; 96(12): 3175-3199, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36063174

RESUMO

Methylmercury (MeHg) is a global environmental pollutant with neurotoxic effects. Exposure to MeHg via consumption of seafood and fish can severely impact fetal neurobehavioral development even when MeHg levels in maternal blood are as low as about 5 µg/L, which the mother tolerates well. Persistent motor dysfunctions and cognitive deficits may result from trans-placental exposure. The present review summarizes current knowledge on the mechanisms of MeHg toxicity during the period of nervous system development. Although cerebellar Purkinje cells are MeHg targets, the actions of MeHg on thiol components in the neuronal cytoskeleton as well as on mitochondrial enzymes and induction of disturbances of glutamate signaling can impair extra-cerebellar functions, also at levels well tolerated by adult individuals. Numerous herbal substances possess neuroprotective effects, predominantly represented by natural polyphenolic molecules that might be utilized to develop natural drugs to alleviate neurotoxicity symptoms caused by MeHg or other Hg compounds.


Assuntos
Poluentes Ambientais , Mercúrio , Compostos de Metilmercúrio , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Animais , Feminino , Gravidez , Compostos de Metilmercúrio/toxicidade , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Placenta , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Poluentes Ambientais/toxicidade , Compostos Fitoquímicos , Compostos de Sulfidrila , Glutamatos
19.
Toxicology ; 477: 153260, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35850386

RESUMO

Aluminum chloride (AlCl3) and its accumulation in the brain are associated with neurodegenerative disease. Recent investigations have illustrated that silibinin is known to have neuroprotective properties. The present study investigates the neuroprotective effects of silibinin-loaded nanostructured lipid carriers (Sili-NLCs) against AlCl3-induced neurotoxicity in male mice. Sili-NLCs were prepared using the emulsification-solvent evaporation method and subjected to particle size, zeta potential, and entrapment efficiency (% EE) analysis. Mice were treated with AlCl3 (100 mg/kg/day, p.o.) and with the same concentration of silibinin and Sili-NLCs (50,100, and 200 mg/kg/day, p.o.) for 30 days in different groups. After treating animals, behavioral studies were assessed. Also, the brain tissue samples were collected from all mice to evaluate oxidative damage and histological changes. The particle size, polydispersity index, zeta potential, and entrapment efficiency (% EE) of prepared Sili-NLCs found 239.7 ± 4.04 nm, 0.082 ± 0.003, - 16.33 ± 0.15 mV, and 72.65 ± 2.03 %, respectively. Brain uptake studies showed that Sili-NLCs had a 5.7-fold greater uptake in the mice brain than the free drug. The AlCl3 caused significant cognitive impairment and increased the level of lipid peroxidation accompanied by decreasing antioxidant enzyme activity in the brain tissue. These findings correlated well with the histopathological experiments. Furthermore, treatment with Sili-NLCs significantly improved the AlCl3-induced cognitive impairment, neurochemical anomalies, and histopathological changes. Given these results, silibinin, when delivered using NLCs, is potentially more effective than free silibinin in decreasing AlCl3- induced neurotoxicity.


Assuntos
Disfunção Cognitiva , Nanoestruturas , Doenças Neurodegenerativas , Síndromes Neurotóxicas , Cloreto de Alumínio/toxicidade , Animais , Antioxidantes/farmacologia , Cognição , Portadores de Fármacos/química , Lipídeos/química , Masculino , Camundongos , Nanoestruturas/química , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Estresse Oxidativo , Tamanho da Partícula , Silibina
20.
Toxicol Appl Pharmacol ; 449: 116112, 2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35688184

RESUMO

Neurotoxicity is one of the major side effects caused by calcineurin inhibitors such as tacrolimus in clinical practice. The underlying mechanisms remain unclear, and no potential protective agents have been identified yet. Here, we aimed to investigate tacrolimus-induced neurotoxicity and assess the protective effects of ibudilast, a nonselective phosphodiesterase inhibitor with neuroprotective effects, against tacrolimus-induced neurotoxicity. An in vitro assay of human neuroblastoma SH-SY5Y cells showed that ibudilast reduced tacrolimus-induced cell death. Subsequently, using in vivo studies, we assessed the pathological mechanism of neurotoxicity and evaluated the protective effect of ibudilast. Wistar rats were subcutaneously administered tacrolimus (2.5 or 5.0 mg/kg/day) for 14 d, and ibudilast (7.5 mg/kg/day) was intraperitoneally administered once a day beginning 2 d prior to tacrolimus (5 mg/kg/day) administration. We observed that ibudilast significantly reduced the tacrolimus-induced neurotoxic events. From the assessment of excised brains, we found that tacrolimus was penetrated to brain and the brain concentration was correlated with the neurotoxicity-score, although ibudilast had no effect on this pharmacokinetics. Tacrolimus-induced neuronal damage was histopathologically evaluated using Nissl and TUNEL staining, where only the cerebral cortex and CA1 region in hippocampus exhibited neuronal death, but not the CA3 region, dendrite gyrus, and cerebellum. Co-administration of ibudilast significantly attenuated these histopathological changes. In conclusion, these results suggest that tacrolimus translocation into the brain and neuronal damage in the cerebral cortex and CA1 are the underlying mechanisms of tacrolimus-induced neurotoxicity and that ibudilast could be a protective agent against this adverse event.


Assuntos
Neuroblastoma , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Animais , Humanos , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Piridinas , Ratos , Ratos Wistar , Tacrolimo/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA