Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 584
Filtrar
1.
Neuropharmacology ; 249: 109895, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38437913

RESUMO

Acute intoxication with organophosphate (OP) cholinesterase inhibitors poses a significant public health risk. While currently approved medical countermeasures can improve survival rates, they often fail to prevent chronic neurological damage. Therefore, there is need to develop effective therapies and quantitative metrics for assessing OP-induced brain injury and its rescue by these therapies. In this study we used a rat model of acute intoxication with the OP, diisopropylfluorophosphate (DFP), to test the hypothesis that T2 measures obtained from brain magnetic resonance imaging (MRI) scans provide quantitative metrics of brain injury and therapeutic efficacy. Adult male Sprague Dawley rats were imaged on a 7T MRI scanner at 3, 7 and 28 days post-exposure to DFP or vehicle (VEH) with or without treatment with the standard of care antiseizure drug, midazolam (MDZ); a novel antiseizure medication, allopregnanolone (ALLO); or combination therapy with MDZ and ALLO (DUO). Our results show that mean T2 values in DFP-exposed animals were: (1) higher than VEH in all volumes of interest (VOIs) at day 3; (2) decreased with time; and (3) decreased in the thalamus at day 28. Treatment with ALLO or DUO, but not MDZ alone, significantly decreased mean T2 values relative to untreated DFP animals in the piriform cortex at day 3. On day 28, the DUO group showed the most favorable T2 characteristics. This study supports the utility of T2 mapping for longitudinally monitoring brain injury and highlights the therapeutic potential of ALLO as an adjunct therapy to mitigate chronic morbidity associated with acute OP intoxication.


Assuntos
Lesões Encefálicas , Intoxicação por Organofosfatos , Ratos , Masculino , Animais , Ratos Sprague-Dawley , Isoflurofato/toxicidade , Organofosfatos , Inibidores da Colinesterase/farmacologia , Intoxicação por Organofosfatos/tratamento farmacológico , Intoxicação por Organofosfatos/patologia , Lesões Encefálicas/induzido quimicamente , Encéfalo , Midazolam/farmacologia
2.
Toxicol Appl Pharmacol ; 484: 116870, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38395364

RESUMO

The development of refractory status epilepticus (SE) following sarin intoxication presents a therapeutic challenge. Here, we evaluated the efficacy of delayed combined double or triple treatment in reducing abnormal epileptiform seizure activity (ESA) and the ensuing long-term neuronal insult. SE was induced in rats by exposure to 1.2 LD50 sarin followed by treatment with atropine and TMB4 (TA) 1 min later. Double treatment with ketamine and midazolam or triple treatment with ketamine, midazolam and levetiracetam was administered 30 min post-exposure, and the results were compared to those of single treatment with midazolam alone or triple treatment with ketamine, midazolam, and valproate, which was previously shown to ameliorate this neurological insult. Toxicity and electrocorticogram activity were monitored during the first week, and behavioral evaluations were performed 2 weeks post-exposure, followed by biochemical and immunohistopathological analyses. Both double and triple treatment reduced mortality and enhanced weight recovery compared to TA-only treatment. Triple treatment and, to a lesser extent, double treatment significantly ameliorated the ESA duration. Compared to the TA-only or the TA+ midazolam treatment, both double and triple treatment reduced the sarin-induced increase in the neuroinflammatory marker PGE2 and the brain damage marker TSPO and decreased gliosis, astrocytosis and neuronal damage. Finally, both double and triple treatment prevented a change in behavior, as measured in the open field test. No significant difference was observed between the efficacies of the two triple treatments, and both triple combinations completely prevented brain injury (no differences from the naïve rats). Delayed double and, to a greater extent, triple treatment may serve as an efficacious delayed therapy, preventing brain insult propagation following sarin-induced refractory SE.


Assuntos
Lesões Encefálicas , Ketamina , Agentes Neurotóxicos , Estado Epiléptico , Ratos , Animais , Sarina/toxicidade , Agentes Neurotóxicos/toxicidade , Midazolam/farmacologia , Midazolam/uso terapêutico , Ratos Sprague-Dawley , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/tratamento farmacológico , Colinérgicos/efeitos adversos , Lesões Encefálicas/induzido quimicamente
3.
J Pharmacol Exp Ther ; 388(2): 432-450, 2024 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-37739807

RESUMO

Acute exposure to nerve agents induces a peripheral cholinergic crisis and prolonged status epilepticus (SE), causing death or long-term brain damage. To provide preclinical data pertinent to the protection of infants and newborns, we compared the antiseizure and neuroprotective effects of treating soman-induced SE with midazolam (MDZ) versus tezampanel (LY293558) in combination with caramiphen (CRM) in 12- and 7-day-old rats. The anticonvulsants were administered 1 hour after soman exposure; neuropathology data were collected up to 6 months postexposure. In both ages, the total duration of SE within 24 hours after soman exposure was significantly shorter in the LY293558 plus CRM groups compared with the MDZ groups. Neuronal degeneration was substantial in the MDZ-treated groups but absent or minimal in the groups treated with LY293558 plus CRM. Loss of neurons and interneurons in the basolateral amygdala and CA1 hippocampal area was significant in the MDZ-treated groups but virtually absent in the LY293558 plus CRM groups. Atrophy of the amygdala and hippocampus occurred only in MDZ-treated groups. Neuronal/interneuronal loss and atrophy of the amygdala and hippocampus deteriorated over time. Reduction of inhibitory activity in the basolateral amygdala and increased anxiety were found only in MDZ groups. Spontaneous recurrent seizures developed in the MDZ groups, deteriorating over time; a small percentage of rats from the LY293558 plus CRM groups also developed seizures. These results suggest that brain damage can be long lasting or permanent if nerve agent-induced SE in infant victims is treated with midazolam at a delayed timepoint after SE onset, whereas antiglutamatergic treatment with tezampanel and caramiphen provides significant neuroprotection. SIGNIFICANCE STATEMENT: To protect the brain and the lives of infants in a mass exposure to nerve agents, an anticonvulsant treatment must be administered that will effectively stop seizures and prevent neuropathology, even if offered with a relative delay after seizure onset. The present study shows that midazolam, which was recently approved by the Food and Drug Administration for the treatment of nerve agent-induced status epilepticus, is not an effective neuroprotectant, whereas brain damage can be prevented by targeting glutamate receptors.


Assuntos
Lesões Encefálicas , Ciclopentanos , Isoquinolinas , Agentes Neurotóxicos , Fármacos Neuroprotetores , Soman , Estado Epiléptico , Tetrazóis , Humanos , Recém-Nascido , Ratos , Animais , Agentes Neurotóxicos/toxicidade , Midazolam/farmacologia , Midazolam/uso terapêutico , Soman/toxicidade , Neuroproteção , Ratos Sprague-Dawley , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/tratamento farmacológico , Convulsões/tratamento farmacológico , Anticonvulsivantes/efeitos adversos , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Encéfalo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Atrofia/tratamento farmacológico
4.
Indian J Pharmacol ; 55(4): 223-228, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37737074

RESUMO

INTRODUCTION: Traumatic brain injury (TBI) is a paramount factor in mortality and morbidity. The clinical trials conducted to investigate the efficacy of neuroprotective agents, such as citicoline, as a therapeutic alternative for TBI have presented divergent findings. Therefore, this study aimed to evaluate and compare citicoline's effect on the Barthel Index in patients with severe and moderate brain injury. MATERIALS AND METHODS: The study is a randomized clinical trial. Patients in the case group (35 patients) were treated with citicoline and the control group (34 patients) received a placebo. Data were analyzed using SPSS 16 software. RESULTS: The results showed that changes in the Glasgow Coma Scale, changes in quadriceps muscle force score, Barthel Index score changes, achieving the status without intubation, and spontaneous breathing in patients treated with citicoline were not a statistically significant difference in the two groups (P > 0.05). CONCLUSION: Findings revealed that citicoline did not impact the recovery process of severe and moderate TBI patients.


Assuntos
Lesões Encefálicas , Fármacos Neuroprotetores , Nootrópicos , Humanos , Citidina Difosfato Colina/uso terapêutico , Citidina Difosfato Colina/efeitos adversos , Nootrópicos/uso terapêutico , Nootrópicos/efeitos adversos , Método Duplo-Cego , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico
5.
J Biochem Mol Toxicol ; 37(10): e23425, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37401655

RESUMO

Mercury is a toxic, environmentally heavy metal that can cause severe damage to all organs, including the nervous system. The functions of puerarin include antioxidant, anti-inflammatory, nerve cell repair, regulation of autophagy, and so forth. But because of the limited oral absorption of puerarin, it affects the protective effect on brain tissue. The nano-encapsulation of Pue can improve its limitation. Therefore, this study investigated the protective effect of Pue drug-loaded PLGA nanoparticles (Pue-PLGA-nps) on brain injury induced by mercuric chloride (HgCl2 ) in mice. The mice were divided into normal saline (NS) group, HgCl2 (4 mg/kg) group, Pue-PLGA-nps (50 mg/kg) group, HgCl2 + Pue (4 mg/kg + 30 mg/kg) group, and HgCl2 + Pue-PLGA-nps (4 mg/kg + 50 mg/kg) group. After 28 days of treatment, the mice were observed for behavioral changes, antioxidant capacity, autophagy and inflammatory response, and mercury levels in the brain, blood, and urine were measured. The results showed that HgCl2 toxicity caused learning and memory dysfunction in mice, increased mercury content in brain and blood, and increased serum levels of interleukin (IL-6), IL-1ß, and tumor necrosis factor-α in the mice. HgCl2 exposure decreased the activity of T-AOC, superoxide dismutase, and glutathione peroxidase, and increased the expression of malondialdehyde in the brain of mice. Moreover, the expression levels of TRIM32, toll-like receptor 4 (TLR4), and LC3 proteins were upregulated. Both Pue and Pue-PLGA-nps interventions mitigated the changes caused by HgCl2 exposure, and Pue-PLGA-nps further enhanced this effect. Our results suggest that Pue-PLGA-nps can ameliorate HgCl2 -induced brain injury and reduce Hg accumulation, which is associated with inhibition of oxidative stress, inflammatory response, and TLR4/TRIM32/LC3 signaling pathway.


Assuntos
Lesões Encefálicas , Mercúrio , Nanopartículas , Camundongos , Animais , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Cloreto de Mercúrio/toxicidade , Receptor 4 Toll-Like/metabolismo , Encéfalo/metabolismo , Estresse Oxidativo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/farmacologia , Mercúrio/metabolismo , Mercúrio/farmacologia , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/prevenção & controle
6.
Am J Health Syst Pharm ; 80(18): 1213-1222, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37280158

RESUMO

PURPOSE: To summarize current literature evaluating tranexamic acid in the management of intracranial bleeding associated with traumatic and nontraumatic brain injuries and implications for clinical practice. SUMMARY: Intracranial hemorrhage, regardless of etiology, is associated with high morbidity and mortality. Tranexamic acid is an antifibrinolytic with anti-inflammatory properties shown to reduce mortality in trauma patients with extracranial injuries. In traumatic brain injury, a large randomized trial found no difference in outcomes when tranexamic acid was compared to placebo; however, subgroup analyses suggested that it may reduce head injury-related mortality in the context of mild-to-moderate injury if treatment occurs within 1 hour of symptom onset. More recent out-of-hospital data have disputed these findings and even suggested harm in severely injured patients. In spontaneous, nontraumatic intracranial hemorrhage, treatment with tranexamic acid did not result in a difference in functional status; however, rates of hematoma expansion, even though modest, were significantly reduced. In aneurysmal subarachnoid hemorrhage, tranexamic acid may prevent rebleeding, but has not led to improved outcomes or reduced mortality, and there is concern for increased incidence of delayed cerebral ischemia. Overall, tranexamic acid has not been shown to result in increased risk of thromboembolic complications across these classes of brain injury. CONCLUSION: Despite its favorable safety profile overall, tranexamic acid does not seem to improve functional outcomes and cannot be routinely recommended. More data are needed to determine which head injury subpopulations are most likely to benefit from tranexamic acid and which patients are at increased risk for harm.


Assuntos
Lesões Encefálicas Traumáticas , Lesões Encefálicas , Traumatismos Craniocerebrais , Hemorragia Subaracnóidea , Ácido Tranexâmico , Humanos , Ácido Tranexâmico/efeitos adversos , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/induzido quimicamente , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/complicações , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/tratamento farmacológico , Hemorragias Intracranianas/induzido quimicamente , Hemorragias Intracranianas/complicações , Hemorragias Intracranianas/tratamento farmacológico , Traumatismos Craniocerebrais/complicações , Traumatismos Craniocerebrais/tratamento farmacológico
7.
Acta Chim Slov ; 70(1): 131-138, 2023 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-37005613

RESUMO

Amiodarone (AMD) is a powerful antiarrhythmic drug preferred for treatments of tachycardias. Brain can be affected negatively when some drugs are used, including antiarrhythmics. S-methyl methionine sulfonium chloride (MMSC) is a well-known sulfur containing substance and a novel powerful antioxidant. It was intended to investigate the protective effects of MMSC on amiodarone induced brain damage. Rats were divided to four groups as follows, control (given corn oil), MMSC (50 mg/kg per day), AMD (100 mg/kg per day), AMD (100 mg/kg per day) + MMSC (50 mg/kg per day). The brain glutathione and total antioxidant levels, catalase, superoxide dismutase, glutathione peroxidase, paraoxonase, and Na+/K+-ATPase activities were decreased, lipid peroxidation and protein carbonyl, total oxidant status, oxidative stress index and reactive oxygen species levels, myeloperoxidase, acetylcholine esterase and lactate dehydrogenase activities were increased after AMD treatment. Administration of MMSC reversed these results. We can conclude that MMSC ameliorated AMD induced brain injury probably due to its antioxidant and cell protective effect.


Assuntos
Amiodarona , Lesões Encefálicas , Vitamina U , Ratos , Animais , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Cloretos/metabolismo , Cloretos/farmacologia , Amiodarona/farmacologia , Amiodarona/metabolismo , Vitamina U/metabolismo , Vitamina U/farmacologia , Ratos Wistar , Estresse Oxidativo , Glutationa/metabolismo , Encéfalo , Superóxido Dismutase/metabolismo , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo
8.
Chem Biol Interact ; 376: 110450, 2023 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-36925032

RESUMO

Excitotoxicity and neuroinflammation are key contributors to perinatal brain injuries. Capsaicin, an active ingredient of chili peppers, is a potent exogenous agonist for transient receptor potential vanilloid 1 receptors. Although the neuroprotective and anti-inflammatory effects of capsaicin are well-documented, its effects on excitotoxic-induced neonatal brain injury and neuroinflammation have not previously been investigated. The aim of this study was to investigate the effects of capsaicin on brain damage, brain mast cells, and inflammatory mediators in a model of ibotenate-induced excitotoxic brain injury in neonatal rats. P5 rat-pups were intraperitoneally injected with vehicle, 0.2-, 1-, and 5-mg/kg doses of capsaicin, or the NMDA (N-methyl-d-aspartate) receptor antagonist MK-801 (dizocilpine), 30 min before intracerebral injection of 10 µg ibotenate. The naive-control group received no substance administration. The rat pups were sacrificed one or five days after ibotenate injection. Levels of activin A and interleukin (IL)-1ß, IL-6, and IL-10 in brain tissue were measured using the enzyme-linked immunosorbent assay method. Cortex and white matter thicknesses, white matter lesion size, and mast cells were evaluated in brain sections stained with cresyl-violet or toluidine-blue. Capsaicin improved ibotenate-induced white matter lesions and cerebral white and gray matter thicknesses in a dose-dependent manner. In addition, it suppressed the degranulation and increased number of brain mast cells induced by ibotenate. Capsaicin also reduced the excitotoxic-induced production of neuronal survival factor activin A and of the pro-inflammatory cytokines IL-1ß, and IL-6 in brain tissue. However, IL-10 levels were not altered by the treatments. MK-801, as a positive control, reversed all these ibotenate-induced changes, further confirming the success of the model. Our findings provide, for the first time, evidence for the therapeutic effects of capsaicin against excitotoxic-induced neonatal brain injury and brain mast cell-mediated neuroinflammation. Capsaicin may therefore be a promising candidate in the prevention and/or reduction of neonatal brain damage.


Assuntos
Encefalite , Mastócitos , Animais , Ratos , Animais Recém-Nascidos , Capsaicina/farmacologia , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Encefalite/induzido quimicamente , Encefalite/tratamento farmacológico , Encefalite/patologia , Substância Branca , Substância Cinzenta , Ácido Ibotênico/toxicidade , Citocinas/metabolismo
9.
Basic Clin Pharmacol Toxicol ; 132(5): 449-453, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36808477

RESUMO

Due to a narrow therapeutic index, prolonged lithium treatment and overdose may result in neurotoxicity. Neurotoxicity is deemed reversible with lithium clearance. However, echoing the report of syndrome of irreversible lithium-effectuated neurotoxicity (SILENT) in rare severe poisonings, lithium-induced histopathological brain injuries including extensive neuronal vacuolization, spongiosis and ageing-like neurodegenerative changes were described in the rat following acute toxic and pharmacological exposure. We aimed to investigate the histopathological consequences of lithium exposure in rat models mimicking prolonged treatment and all three patterns of acute, acute-on-chronic and chronic poisonings observed in humans. We performed histopathology and immunostaining-based analyses using optic microscopy of brains obtained from male Sprague-Dawley rats randomly assigned to lithium or saline (controls) and treated according to the therapeutic or to the three poisoning models. No lesion was observed in any brain structure in any of the models. Neuron and astrocyte counts did not differ significantly between lithium-treated rats and controls. Our findings support that lithium-induced neurotoxicity is reversible and brain injury not a common feature of toxicity.


Assuntos
Lesões Encefálicas , Síndromes Neurotóxicas , Intoxicação , Humanos , Masculino , Ratos , Animais , Lítio , Ratos Sprague-Dawley , Síndromes Neurotóxicas/etiologia , Encéfalo , Lesões Encefálicas/induzido quimicamente
10.
J Biochem Mol Toxicol ; 37(5): e23319, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36811218

RESUMO

Organophosphorus pesticides (OPs) have long been used extensively on agricultural land and can lead to significant improvements in crop yields. Due to occupational exposure, humans are exposed to pesticides through dermal contact, inhalation, and ingestion. The effects of OPs on the organism are currently studied for their effects on livers, kidneys, hearts, blood indicators, neurotoxicity, and teratogenic, carcinogenic, and mutagenic effects, while studies in the direction of brain tissue damage have not been reported in detail. Previous reports have confirmed that ginsenoside Rg1 is a prominent and representative tetracyclic triterpenoid derivative rich in ginseng and has good neuroprotective activity. Considering that, the aim of this study was to establish a mouse model of brain tissue injury by using the OP-type pesticide chlorpyrifos (CPF) and to explore the therapeutic effects and possible molecular mechanisms of Rg1. Mice in the experimental group were pre-protected with Rg1 by gavage for 1 week, and brain tissue damage was induced using CPF (5 mg/kg for 1 week) to assess the effect of Rg1 (80 and 160 mg/kg for 3 weeks) in alleviating brain damage. Morris water maze and histopathological analysis were performed to assess cognitive function and pathological changes in the mouse brain, respectively. Protein expression levels of Bax, Bcl-2, Caspase-3, Cl-Cas-3, Caspase-9, Cl-Cas-9, phosphoinositide 3-kinase (PI3K), phosphorylated-PI3K, protein kinase B (AKT), and phosphorylated-AKT were quantified by protein blotting analysis. Rg1 obviously restored CPF-induced oxidative stress damage in mouse brain tissue, increased the levels of antioxidant parameters (total superoxide dismutase, total antioxidative capacity, and glutathione) in the brain, and significantly reduced the overexpression of apoptosis-related proteins induced by CPF. At the same time, Rg1 also markedly attenuated the histopathological changes in the brain induced by CPF exposure. Mechanistically, Rg1 could effectively activate the phosphorylation of PI3K/AKT. Furthermore, molecular docking studies revealed a stronger binding capacity between Rg1 and PI3K. Rg1 attenuated neurobehavioural alterations and reduced lipid peroxidation in the mouse brain to a great extent. Apart from that, Rg1 administration improved brain histopathology in CPF-induced rats. All results suggest that ginsenoside Rg1 has potential antioxidant effects on CPF-induced oxidative brain injury, and it is evident that Rg1 could be used as a promising therapeutic strategy for the study of brain injury from OP poisoning.


Assuntos
Lesões Encefálicas , Clorpirifos , Praguicidas , Animais , Camundongos , Antioxidantes , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Simulação de Acoplamento Molecular , Compostos Organofosforados , Fosfatidilinositol 3-Quinase , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt
11.
Environ Sci Pollut Res Int ; 30(17): 49014-49025, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36759409

RESUMO

This study is aimed at determining whether royal jelly (RJ) which has a powerful antioxidant property prevents fluoride-induced brain tissue damage and exploring whether Bcl-2/NF-κB/ and caspase-3/caspase-6/Bax/Erk pathways play a critical role in the neuroprotective effect of RJ. Wistar albino rats were chosen for the study, and they were randomly distributed into six groups: (i) control; (ii) royal jelly; (iii) fluoride-50; (iv) fluoride-100; (v) fluoride-50 + royal jelly; (vi) fluoride-100 + royal jelly. We established fluoride-induced brain tissue damage with 8-week-old male Wistar albino rats by administration of fluoride exposure (either 50 mg/kg or 100 mg/kg bw) through drinking water for 8 weeks. Then, the study duration is for 56 days where the rats were treated with or without RJ (100 mg/kg bw) through oral gavage. The effects of RJ on glutathione (GSH), catalase activity (CAT), and malondialdehyde (MDA) levels were determined via spectrophotometer. Western blot analysis was performed to investigate the effects of royal jelly on the protein expression levels of Bax, caspase-3, caspase-6, Bcl-2, NF-κB, COX-2, and Erk. It was also studied the effects of RJ on histopathological alterations in fluoride-induced damage to the rat brain. As a result, the Bcl-2, NF-κB, and COX-2 protein expression levels were increased in the fluoride-treated (50 and 100 mg/kg) groups but they were decreased significantly by RJ treatment in the brain tissue. Additionally, the protein expression of caspase-3, caspase-6, Bax, and Erk were decreased in fluoride-treated groups and they were significantly increased by RJ treatment compared to the un-treated rats. Our results suggested that RJ prevented fluoride-induced brain tissue damage through anti-antioxidant activities.


Assuntos
Produtos Biológicos , NF-kappa B , Animais , Masculino , Ratos , Antioxidantes/metabolismo , Proteína X Associada a bcl-2/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 6/efeitos dos fármacos , Caspase 6/metabolismo , Ciclo-Oxigenase 2/metabolismo , Ácidos Graxos/farmacologia , Fluoretos/toxicidade , Glutationa/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Estresse Oxidativo , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
12.
Toxicol Mech Methods ; 33(4): 307-315, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36177783

RESUMO

To confirm the regulation of miR-204-5p on VCAM1 and its effect on sevoflurane-induced brain injury in neonatal rats. We adopted the sevoflurane-induced brain injury model, and the double luciferase reporter gene assay was applied to explore the targeting relationship between vascular adhesion factor 1 (VCAM1) and miR-204-5p. RT-qPCR was applied to assess the levels of miR-204-5. VCAM1, LC3, P62 and cleaved-caspase 3 levels in the hippocampus were estimated by western blot. The number of autophagosomes in the cerebral cortex was assessed via Transmission electron microscopy (TEM), and histopathological changes within the hippocampus by HE staining. miR-204-5p levels were remarkably increased, but VCAM1 expression was decreased after neonatal rat brain injury. Furthermore, miR-204-5p directly targeted VCAM1. The escape latency, swimming distance, autophagosome number, neuronal apoptosis ratio, LC3 II and Cleaved-caspase 3 expression were reduced after miR-204-5p inhibition interference, whereas crossing times, and P62 expression increased in the sevoflurane-induced brain injury model. Furthermore, down-regulation of VCAM1 reversed the trend of these indices. These results suggest that down-regulation of miR-204-5p ameliorates sevoflurane-induced cognitive impairment and hippocampal pathology and inhibits neuronal autophagy and apoptosis by targeting VCAM1.


Assuntos
Lesões Encefálicas , Regulação para Baixo , MicroRNAs , Sevoflurano , Animais , Ratos , Animais Recém-Nascidos , Apoptose , Lesões Encefálicas/induzido quimicamente , Caspase 3/metabolismo , MicroRNAs/genética
13.
Jpn J Clin Oncol ; 53(4): 355-364, 2023 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-36579769

RESUMO

OBJECTIVE: Antiangiogenic vascular endothelial growth factor receptor tyrosine kinase inhibitors play an essential role in systemic therapy for renal cell carcinoma. Given the anti-edematous effect of bevacizumab, an antiangiogenic antibody targeting vascular endothelial growth factor, vascular endothelial growth factor receptor tyrosine kinase inhibitors should exert therapeutic effects on radiation-induced brain injury after stereotactic radiosurgery. This preliminary study aimed to investigate the therapeutic effect of vascular endothelial growth factor receptor tyrosine kinase inhibitor against radiation-induced brain injury. METHODS: Magnetic resonance images for six patients treated with vascular endothelial growth factor receptor tyrosine kinase inhibitors who were diagnosed with radiation-induced brain injury following gamma knife radiosurgery were retrospectively reviewed. RESULTS: The median brain edema volume and tumour mass volume in the pre-tyrosine kinase inhibitor period were 57.6 mL (range: 39.4-188.2) and 3.2 mL (range: 1.0-4.6), respectively. Axitinib, pazopanib (followed by cabozantinib) and sunitinib were administered in four, one and one cases, respectively. The median brain edema volume and tumour mass volume in the post-tyrosine kinase inhibitor period were 4.8 mL (range: 1.5-27.8) and 1.6 mL (range: 0.4-3.6), respectively. The median rates of reduction in brain edema volume and tumour mass volume were 90.8% (range: 51.9-97.6%) and 57.2% (range: 20.0-68.6%), respectively. The post-tyrosine kinase inhibitor values for brain edema volume (P = 0.027) and tumour mass volume (P = 0.008) were significantly lower than the pre-tyrosine kinase inhibitor values. Changes in volume were correlated with tyrosine kinase inhibitor use. CONCLUSION: This study is the first to demonstrate the therapeutic effects of vascular endothelial growth factor receptor tyrosine kinase inhibitors on radiation-induced brain injury in patients with brain metastases from renal cell carcinoma treated via gamma knife radiosurgery.


Assuntos
Edema Encefálico , Lesões Encefálicas , Neoplasias Encefálicas , Carcinoma de Células Renais , Neoplasias Renais , Radiocirurgia , Humanos , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/radioterapia , Radiocirurgia/efeitos adversos , Fator A de Crescimento do Endotélio Vascular , Edema Encefálico/induzido quimicamente , Edema Encefálico/tratamento farmacológico , Estudos Retrospectivos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Inibidores da Angiogênese/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/radioterapia , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico
14.
Arch Biochem Biophys ; 729: 109390, 2022 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-36067878

RESUMO

BACKGROUND: To investigate the protective effect of low-dose radiation (LDR) on brain injury in mice induced by doxorubicin (DOX). METHODS: Sixty female BALB/C mice were randomly divided into the control (CTR) group, low-dose radiation (LDR) group, doxorubicin treatment (DOX) group and low-dose radiation before doxorubicin treatment (COM) group. After 72 h of exposure to 75 mGy, the mice were intraperitoneally injected with 7.5 mg/kg of doxorubicin and sacrificed 5 days later. Neuron-specific enolase (NSE), lactate dehydrogenase (LDH), adenosine triphosphate (ATP), neurotransmitters, inflammatory mediators, apoptosis- and oxidative stress-related mediators as well as mitochondrial dysfunction were examined. RESULTS: Compared to the DOX group, the concentrations of DA, 5-HT, EPI and GABA in the COM group were significantly decreased, and the number of TUNEL-positive cells was decreased. In addition, the expression of proapoptotic proteins was downregulated in the COM group compared to the DOX group. Low-dose radiation in advance reduced reactive oxygen species and activated the SOD antioxidant defense system as indicated by significantly reduced GSH expression, increased GSSG expression, increased GPx expression and activation of the Nrf2 redox pathway. After low-dose radiation, the expression levels of ATP5f1, NDUFV1 and CYC1 were close to normal, and the mitochondrial respiratory control rate (RCR) and activity of respiratory chain complex enzymes also tended to be normal. Low-dose radiation upregulated the expression levels of IL-2 and IL-4 but downregulated the expression levels of IL-10 and TGF-ß. CONCLUSION: LDR has a protective effect on brain injury in mice treated with DOX. The mechanism is related to LDR alleviating mitochondrial dysfunction and oxidative stress, which promotes the production of antioxidant damage proteins, thus exerting an adaptive protective effect on cells.


Assuntos
Lesões Encefálicas , Fator 2 Relacionado a NF-E2 , Trifosfato de Adenosina/metabolismo , Animais , Antioxidantes/farmacologia , Apoptose , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/prevenção & controle , Doxorrubicina/farmacologia , Feminino , Dissulfeto de Glutationa/metabolismo , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Interleucina-2/farmacologia , Interleucina-4/metabolismo , Lactato Desidrogenases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Fosfopiruvato Hidratase/metabolismo , Fosfopiruvato Hidratase/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Serotonina/metabolismo , Superóxido Dismutase/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ácido gama-Aminobutírico/metabolismo
15.
Eur J Pharmacol ; 932: 175217, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36007603

RESUMO

Modulation of the inflammasome NLRP3 and SIRT1 are new combat strategy for brain injury protection. The inflammasome activates proinflammatory cytokines releasing interleukin-1ß and interleukin-18 which in turn affect the toxins release from immune cells. In addition, SIRT1 controls many biological functions, such as immune response and oxidative stress. Protocatechuic has versatile biological activities and possesses antioxidant, anti-inflammatory and neuroprotective effects. So this work aims to study immunomodulatory effect of protocatechuic acid on cyclophosphamide chemotherapy drug-induced brain injury via modulation of inflammosomes NLRP3 and SIRT1. Rats were randomly assigned to four experimental groups. Normal control group was injected with a single i.p injection of saline. Cyclophosphamide group was injected with a single i.p injection of cyclophosphamide (200 mg/kg). Protocatechuic acid groups were orally administered (50 &100 mg/kg) once daily for 10 consecutive days after cyclophosphamide injection. Protocatechuic acid administration exhibited improvements of the cognition function and memory, a reduction in brain contents of MDA, NLRP3, IL-1 ß, NF-κB, IKBKB and Galectin 3 and an elevation of GSH and SIRT1 compared to cyclophosphamide group. In addition, protocatechuic acid administration ameliorated the elevation of caspase 3 and iNOS gene expression and alleviated the neuron degeneration caused by cyclophosphamide. In conclusion, the therapeutic action of protocatechuic acid and its cellular and molecular mechanisms are new insights against various human ailments, especially, neurodegenerative disease as brain injury induced by cyclophosphamide chemotherapy drug in rats through modulation of inflammosomes NLRP3 and SIRT1.


Assuntos
Lesões Encefálicas , Doenças Neurodegenerativas , Fármacos Neuroprotetores , Animais , Anti-Inflamatórios/uso terapêutico , Antioxidantes/metabolismo , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Caspase 3 , Ciclofosfamida/efeitos adversos , Citocinas/metabolismo , Galectina 3 , Humanos , Hidroxibenzoatos , Quinase I-kappa B , Imunidade , Inflamassomos/metabolismo , Interleucina-18 , Interleucina-1beta/metabolismo , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Sirtuína 1/metabolismo
16.
Neurobiol Dis ; 171: 105814, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35817217

RESUMO

Barbiturates and benzodiazepines are GABAA-receptor agonists and potent antiseizure medications. We reported that exposure of neonatal macaques to combination of phenobarbital and midazolam (Pb/M) for 24 h, at clinically relevant doses and plasma levels, causes widespread apoptosis affecting neurons and oligodendrocytes. Notably, the extent of injury was markedly more severe compared to shorter (8 h) exposure to these drugs. We also reported that, in the infant macaque, mild hypothermia ameliorates the apoptosis response to the anesthetic sevoflurane. These findings prompted us explore whether mild hypothermia might protect infant nonhuman primates from neuro- and gliotoxicity of Pb/M. Since human infants with seizures may receive combinations of benzodiazepines and barbiturates for days, we opted for 24 h treatment with Pb/M. Neonatal rhesus monkeys received phenobarbital intravenously, followed by midazolam infusion over 24 h under normothermia (T > 36.5 °C-37.5 °C; n = 4) or mild hypothermia (T = 35 °C-36.5 °C; n = 5). Medication doses and blood levels measured were comparable to those in human infants. Animals were euthanized at 36 h and brains examined immunohistochemically and stereologically. Treatment was well tolerated. Extensive degeneration of neurons and oligodendrocytes was seen at 36 h in both groups within neocortex, basal ganglia, hippocampus and brainstem. Mild hypothermia over 36 h (maintained until terminal perfusion) conferred no protection against the neurotoxic and gliotoxic effects of Pb/M. This is in marked contrast to our previous findings that mild hypothermia is protective in the context of a 5 h-long exposure to sevoflurane in infant macaques. These findings demonstrate that brain injury caused by prolonged exposure to Pb/M in the neonatal primate cannot be ameliorated by mild hypothermia.


Assuntos
Lesões Encefálicas , Hipotermia Induzida , Hipotermia , Animais , Encéfalo , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/prevenção & controle , Humanos , Lactente , Recém-Nascido , Chumbo/farmacologia , Macaca mulatta , Midazolam/farmacologia , Fenobarbital/toxicidade , Sevoflurano/farmacologia
17.
Food Funct ; 13(15): 8304-8312, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35839080

RESUMO

Brain microvascular endothelial cells are essential components of the blood-brain barrier (BBB) that acts as a selective physical barrier and plays protective roles in maintaining brain homeostasis. Tanshinone IIA (Tan IIA), isolated from Salvia miltiorrhiza Bunge, exhibited healthy effects such as antioxidant effects, anti-inflammatory effects, and cardiovascular protective effects. Here, we tried to investigate the positive effect and the potential mechanism of Tan IIA on the lipopolysaccharide (LPS)-induced brain injury in mice and brain microvascular endothelial cells in vitro. In vivo, Tan IIA inhibited the brain injury, and the enhancement of blood-brain barrier permeability in the LPS-induced brain injury in mice. Moreover, Tan IIA suppressed inflammatory response and oxidant response in LPS-treated mice evidenced by low levels of serum TNF-α and IL-1ß, high superoxide dismutase (SOD) activity and low malondialdehyde (MDA) in the brain. In vitro, Tan IIA suppressed the generation of reactive oxygen species (ROS) and MDA, and promoted SOD activity in LPS-stimulated brain microvascular endothelial cells. Moreover, Tan IIA promoted the expression of Claudin5, ZO-1, Nrf2, HO-1 and NQO1 in LPS-stimulated brain microvascular endothelial cells. In conclusion, Tan IIA protected against the LPS-induced brain injury via the suppression of oxidant stress and inflammatory response and protective effect of the BBB through activating Nrf2 signaling pathways and rescue of the tight junction proteins in microvascular endothelial cells, supporting the application of Tan IIA and Salvia miltiorrhiza Bunge as food supplements for the treatment of brain disease.


Assuntos
Abietanos , Lesões Encefálicas , Animais , Camundongos , Abietanos/farmacologia , Barreira Hematoencefálica/metabolismo , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/metabolismo , Células Endoteliais , Lipopolissacarídeos , Fator 2 Relacionado a NF-E2/metabolismo , Superóxido Dismutase/metabolismo , Estresse Oxidativo
18.
Sci Rep ; 12(1): 9194, 2022 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-35654991

RESUMO

Toluene was widely used volatile organic compound that accumulates in tissues with high lipid content. Stem cells have been proposed as an increasingly attractive approach for repair of damaged nervous system, we aimed to evaluate the ability of breast milk mesenchymal stem cells (MSc) to ameliorate toluene-induced encephalopathy. Sixty adult male albino rats were assigned to 3 groups, control, toluene, and toluene/breast milk-MSc. Neurological assessment was evaluated as well as serum levels of glial fibrillary acidic protein (GFAP), tumor necrosis factor-alpha (TNF-α), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), tissue dopamine and oxidative markers. Gene expression of peroxisome Proliferator-Activated Receptor-Gamma (PPAR-É£), nuclear factor-kappaB (NF-kB), and interleukin-6 (IL-6) were evaluated. Moreover, histological and immunohistochemical investigation were done. Results revealed that toluene caused cerebral injury, as evidenced by a significant increase in serum GFAP, TNF-α, malondialdehyde (MDA) and nitric oxide (NO), a significant decrease in serum NGF, tissue dopamine and oxidative markers, besides, a non-significant change in VEGF. Toluene also caused changes in normal cerebral structure and cellular degeneration, including a significant decrease in the total number of neurons and thickness of frontal cortex. Meninges showing signs of inflammation with inflammatory cell infiltration and exudation, a significant decrease in MBP immunoreactivity, and increase in the percent of high motility group box protein-1 (HMGB1) positive cells. PPAR- É£, NF-kB, and IL-6 gene expression were all considerably elevated by toluene. These changes were greatly improved by breast milk MSc. Therefore, we conclude that breast milk MSc can attenuate toluene-induced encephalopathy.


Assuntos
Lesões Encefálicas , Células-Tronco Mesenquimais , Adulto , Animais , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/terapia , Dopamina , Feminino , Humanos , Fatores Imunológicos , Interleucina-6 , Masculino , Leite Humano , NF-kappa B , Fator de Crescimento Neural , PPAR gama , Ratos , Tolueno/efeitos adversos , Fator de Necrose Tumoral alfa/genética , Fator A de Crescimento do Endotélio Vascular
19.
J Neuroimmunol ; 368: 577894, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35642994

RESUMO

We investigated effects of activation of TRESK channels by selective activator cloxyquin on excitotoxic-induced brain injury and neuroinflammation involving brain mast cells and inflammatory cytokines in neonatal rats. Three different doses of cloxyquin (0.2, 1 and 5 mg/kg) were studied in ibotenate-induced perinatal brain injury (PBI) in P5 rat-pups. Cerebral lesions and mast cells in coronal brain sections were evaluated. Concentrations of activin A, IL-1ß, IL-6 and IL-10 in brain homogenates were measured using ELISA. Cloxyquin dose-dependently exerted protective effects against excitotoxic-induced neonatal brain injury and neuroinflammation. TRESK channels may be a promising new target for the treatment of PBIs.


Assuntos
Lesões Encefálicas , Canais de Potássio de Domínios Poros em Tandem , Canais de Potássio/metabolismo , Animais , Animais Recém-Nascidos , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/prevenção & controle , Cloroquinolinóis , Doenças Neuroinflamatórias , Ratos
20.
Int J Radiat Oncol Biol Phys ; 113(4): 796-804, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35378217

RESUMO

PURPOSE: The treatment of radiation-induced brain injury (RI) caused by radiation therapy for head and neck cancer is challenging. Antiangiogenic therapy is a promising treatment. Apatinib is an oral tyrosine kinase inhibitor that selectively inhibits vascular endothelial growth factor receptor 2. We aimed to assess the efficacy and safety of apatinib in patients with RI. METHODS AND MATERIALS: In this phase 2, open-label, single-arm, prospective study, we recruited patients aged 35 to 80 years with prior radiation therapy history for head and neck cancer who had newly diagnosed RI at the Sun Yat-sen Memorial Hospital, China. Apatinib was administered at a dosage of 250 mg once daily orally for 4 weeks. A Simon minimax 2-stage design was performed. The primary outcome was the proportion of patients with overall clinical efficacy, defined as a radiographic response of ≥25% reduction in baseline brain edema volume on magnetic resonance fluid attenuated inversion recovery images at week 4. Secondary end points were the overall improvement rate of brain necrosis, neurologic function, and safety. RESULTS: We screened 37 patients, 36 of whom were enrolled between October 17, 2019, and August 3, 2020. At the cutoff date, 36 patients were assessed for efficacy and safety (19 were enrolled in stage 1 and 17 in stage 2). Of the 36 patients evaluated for overall clinical efficacy, 22 patients (61.1%; 95% CI, 43.5%-76.9%) achieved the primary end point at week 4. Among the 31 patients with brain necrosis lesions, 19 patients (61.3%; 95% CI, 42.2%-78.2%) showed improvement of brain necrosis. The most common grade 1 to 2 adverse events were hand-foot syndrome, fatigue, and hypertension There were no treatment-related grade 4 to 5 toxic effects. CONCLUSIONS: Oral apatinib shows promising efficacy and is well-tolerated in patients with RI. Further randomized controlled studies are warranted.


Assuntos
Antineoplásicos , Lesões Encefálicas , Neoplasias de Cabeça e Pescoço , Lesões por Radiação , Antineoplásicos/efeitos adversos , Encéfalo , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Necrose/tratamento farmacológico , Estudos Prospectivos , Piridinas , Lesões por Radiação/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...