Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 589
Filter
1.
J Biochem Mol Toxicol ; 38(7): e23760, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38953502

ABSTRACT

Cyclophosphamide (CP) is an antineoplastic drug widely used in chemotherapy. Curcumin (CUR) and piperine (PP) show a protective effect on neurodegenerative and neurological diseases. This research was designed to measure several biochemical parameters in the brain tissue of CP-applied rats to investigate the impact of combined CUR-PP administration. The study evaluated six groups of eight rats: Group 1 was the control; Groups 2 and 3 were administered 200 or 300 mg/kg CUR-PP via oral gavage; Group 4 received only 200 mg/kg CP on day 1; Groups 5 and 6 received CP + CUR-PP for 7 days. Data from all parameters indicated that CP caused brain damage. Phosphorylated TAU (pTAU), amyloid-beta peptide 1-42 (Aß1-42), glutamate (GLU), and gamma amino butyric acid (GABA) parameters were the same in Groups 4, 5, and 6. On the other hand, 8-hydroxy-2-deoxyguanosine (8-OHdG), nitric oxide (NO), interleukin-6 (IL-6), nuclear factor kappa beta (NF-kß), malondialdehyde (MDA), and tumor necrosis factor-alpha (TNF-α) levels in the CP + CUR-PP groups were lower than those in the CP group (p < 0.05). However, superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and reduced glutathione (GSH) parameters were higher in the CP + CUR-PP groups compared to the CP group (p < 0.05). It is thought that the similarity of Groups 5 and 6 with Group 4 in Aß1-42, pTAU, GLU, and GABA parameters hinder the determination of treatment protection however, they might have a therapeutic effect if the applied dose or study duration were changed. This study attempted to evaluate the effects of a CUR-PP combination on CP-induced brain damage in rats by measuring biochemical parameters and performing histopathological examinations. Based on the findings, this CUR-PP combination could be considered an alternative medicine option in cases with conditions similar to those evaluated in this study.


Subject(s)
Alkaloids , Benzodioxoles , Brain Injuries , Curcumin , Cyclophosphamide , Piperidines , Polyunsaturated Alkamides , Animals , Polyunsaturated Alkamides/pharmacology , Benzodioxoles/pharmacology , Curcumin/pharmacology , Piperidines/pharmacology , Alkaloids/pharmacology , Rats , Cyclophosphamide/toxicity , Cyclophosphamide/adverse effects , Male , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/prevention & control , Rats, Wistar , Brain/metabolism , Brain/drug effects , Brain/pathology , Oxidative Stress/drug effects , Neuroprotective Agents/pharmacology
2.
Int J Biol Macromol ; 273(Pt 1): 132887, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38851621

ABSTRACT

Acute alcohol intoxication is a harmful clinical condition characterized by behavioral and neurological symptoms, for which few effective therapies are available at present. Dysfunction of microglial BV-2 cells has been reported to be associated with acute alcohol-induced brain injuries. In the present study, the protective effects of Eucommia ulmoides Oliv. leaves polysaccharides (EULP) on acute alcoholic brain injury and microglial dysfunction were investigated. 14-day pretreatment of EULP significantly attenuated neurobehavioral deficit and neurotransmitter damage in the brain tissue of mice caused by acute alcohol exposure. Additionally, EULP regulated the metabolic disorder of brain tissue. Consistently, it was shown that EULP pretreatment significantly improved alcohol-induced phagocytosis decrease, oxidative stress and inflammation in BV-2 cells. Therefore, EULP may be proposed and employed as a potential therapeutic agent for alcohol-induced brain damage.


Subject(s)
Eucommiaceae , Microglia , Oxidative Stress , Plant Leaves , Polysaccharides , Animals , Polysaccharides/pharmacology , Polysaccharides/chemistry , Microglia/drug effects , Microglia/metabolism , Mice , Eucommiaceae/chemistry , Plant Leaves/chemistry , Oxidative Stress/drug effects , Male , Ethanol , Brain Injuries/drug therapy , Brain Injuries/chemically induced , Brain Injuries/metabolism , Brain Injuries/etiology , Brain Injuries/pathology , Cell Line , Phagocytosis/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Plant Extracts/pharmacology , Plant Extracts/chemistry
3.
Brain Inj ; 38(10): 848-857, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-38711413

ABSTRACT

BACKGROUND: Organophosphorus pesticide poisoning can lead to severe brain damage, but the specific mechanisms involved are not fully understood. Our research aims to elucidate the function of the TRPV4 ion channel in the development of brain injury induced by paraoxon (POX). METHODS: In vivo, we examined the survival rate, behavioral seizures, histopathological alterations, NMDA receptor phosphorylation, as well as the expression of the NLRP3-ASC-caspase-1 complex and downstream inflammatory factors in the POX poisoning model following intervention with the TRPV4 antagonist GSK2193874. In vitro, we investigated the effects of GSK2193874 on NMDA-induced inward current, cell viability, cell death rate, and Ca2+ accumulation in primary hippocampal neurons. RESULTS: The treatment with the TRPV4 antagonist increased the survival rate, suppressed the status epilepticus, improved pathological damage, and reduced the phosphorylation level of NMDA receptors after POX exposure. Additionally, it inhibited the upregulation of NLRP3 inflammasome and inflammatory cytokines expression after POX exposure. Moreover, the TRPV4 antagonist corrected the NMDA-induced increase in inward current and cell death rate, decrease in cell viability, and Ca2+ accumulation. CONCLUSION: TRPV4 participates in the mechanisms of brain injury induced by POX exposure through NMDA-mediated excitotoxicity and NLRP3-mediated inflammatory response.


Subject(s)
Brain Injuries , NLR Family, Pyrin Domain-Containing 3 Protein , Paraoxon , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Paraoxon/toxicity , Male , Mice , Brain Injuries/metabolism , Brain Injuries/chemically induced , Hippocampus/metabolism , Hippocampus/drug effects , N-Methylaspartate , Neurons/metabolism , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Inflammasomes/metabolism
4.
Neuropharmacology ; 249: 109895, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38437913

ABSTRACT

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.


Subject(s)
Brain Injuries , Organophosphate Poisoning , Rats , Male , Animals , Rats, Sprague-Dawley , Isoflurophate/toxicity , Organophosphates , Cholinesterase Inhibitors/pharmacology , Organophosphate Poisoning/drug therapy , Organophosphate Poisoning/pathology , Brain Injuries/chemically induced , Brain , Midazolam/pharmacology
5.
Toxicol Appl Pharmacol ; 484: 116870, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38395364

ABSTRACT

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.


Subject(s)
Brain Injuries , Ketamine , Nerve Agents , Status Epilepticus , Rats , Animals , Sarin/toxicity , Nerve Agents/toxicity , Midazolam/pharmacology , Midazolam/therapeutic use , Rats, Sprague-Dawley , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Cholinergic Agents/adverse effects , Brain Injuries/chemically induced
6.
Mol Neurobiol ; 61(8): 5787-5801, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38233687

ABSTRACT

Perioperative neurocognitive impairment (PND) is a common medical complication in the postoperative period. General anesthesia through volatile anesthetics poses a high risk of POCD. Moreover, the developing brain is especially vulnerable to anesthesia-induced neurotoxicity. Therefore, finding a practical approach to prevent or alleviate neonatal isoflurane (ISO) exposure-induced brain injury and cognitive decline is essential for reducing medical complications following major surgery during the early postnatal period. Using a repeated neonatal ISO exposure-induced PND rat model, we investigated the effects of methylene blue (MB) pretreatment on repeated neonatal isoflurane exposure-induced brain injury and memory loss. Intraperitoneal injection of low-dose MB (1 mg/kg) was conducted three times 24 h before each ISO exposure. The Barnes maze and novel objection test were conducted to assess learning and memory. Immunofluorescence staining, F-Jade C staining, TUNEL staining, and Western blot analysis were performed to determine mitochondrial fragmentation, neuronal injury, degeneration, and apoptosis. Evans blue extravasation assay, total antioxidant capacity assay, MDA assay kit, and related inflammatory assay kits were used to test blood-brain barrier (BBB) disruption, antioxidant capacity, and neuroinflammation. Behavioral tests revealed that MB pretreatment significantly ameliorated ISO exposure-induced cognitive deficits. In addition, MB pretreatment alleviates neuronal injury, apoptosis, and degeneration. Furthermore, the BBB integrity was preserved by MB pretreatment. Additional studies revealed that ISO-induced excessive mitochondrial fragmentation, oxidative stress, and neuroinflammation were significantly attenuated by MB pretreatment in the PND rat model. Our findings suggest that MB pretreatment alleviates ISO exposure-induced brain injury and memory loss for the first time, supporting MB pretreatment as a promising approach to protect the brain against neonatal ISO exposure-induced postoperative cognitive dysfunction.


Subject(s)
Animals, Newborn , Brain Injuries , Isoflurane , Memory Disorders , Methylene Blue , Rats, Sprague-Dawley , Animals , Isoflurane/pharmacology , Isoflurane/toxicity , Memory Disorders/prevention & control , Memory Disorders/chemically induced , Methylene Blue/pharmacology , Brain Injuries/prevention & control , Brain Injuries/pathology , Brain Injuries/chemically induced , Male , Apoptosis/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Rats , Oxidative Stress/drug effects , Maze Learning/drug effects , Mitochondria/drug effects , Mitochondria/metabolism
7.
J Pharmacol Exp Ther ; 388(2): 432-450, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37739807

ABSTRACT

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.


Subject(s)
Brain Injuries , Cyclopentanes , Isoquinolines , Nerve Agents , Neuroprotective Agents , Soman , Status Epilepticus , Tetrazoles , Humans , Infant, Newborn , Rats , Animals , Nerve Agents/toxicity , Midazolam/pharmacology , Midazolam/therapeutic use , Soman/toxicity , Neuroprotection , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Seizures/drug therapy , Anticonvulsants/adverse effects , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Atrophy/drug therapy
8.
Indian J Pharmacol ; 55(4): 223-228, 2023.
Article in English | MEDLINE | ID: mdl-37737074

ABSTRACT

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.


Subject(s)
Brain Injuries , Neuroprotective Agents , Nootropic Agents , Humans , Cytidine Diphosphate Choline/therapeutic use , Cytidine Diphosphate Choline/adverse effects , Nootropic Agents/therapeutic use , Nootropic Agents/adverse effects , Double-Blind Method , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Neuroprotective Agents/therapeutic use
9.
J Biochem Mol Toxicol ; 37(10): e23425, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37401655

ABSTRACT

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.


Subject(s)
Brain Injuries , Mercury , Nanoparticles , Mice , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Mercuric Chloride/toxicity , Toll-Like Receptor 4/metabolism , Brain/metabolism , Oxidative Stress , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Mercury/metabolism , Mercury/pharmacology , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain Injuries/prevention & control
10.
Am J Health Syst Pharm ; 80(18): 1213-1222, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37280158

ABSTRACT

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.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Craniocerebral Trauma , Subarachnoid Hemorrhage , Tranexamic Acid , Humans , Tranexamic Acid/adverse effects , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/chemically induced , Brain Injuries/chemically induced , Brain Injuries/complications , Brain Injuries/drug therapy , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/complications , Intracranial Hemorrhages/drug therapy , Craniocerebral Trauma/complications , Craniocerebral Trauma/drug therapy
11.
Acta Chim Slov ; 70(1): 131-138, 2023 Mar 20.
Article in English | MEDLINE | ID: mdl-37005613

ABSTRACT

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.


Subject(s)
Amiodarone , Brain Injuries , Vitamin U , Rats , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Chlorides/metabolism , Chlorides/pharmacology , Amiodarone/pharmacology , Amiodarone/metabolism , Vitamin U/metabolism , Vitamin U/pharmacology , Rats, Wistar , Oxidative Stress , Glutathione/metabolism , Brain , Superoxide Dismutase/metabolism , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain Injuries/metabolism
12.
Chem Biol Interact ; 376: 110450, 2023 May 01.
Article in English | MEDLINE | ID: mdl-36925032

ABSTRACT

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.


Subject(s)
Encephalitis , Mast Cells , Animals , Rats , Animals, Newborn , Capsaicin/pharmacology , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Encephalitis/chemically induced , Encephalitis/drug therapy , Encephalitis/pathology , White Matter , Gray Matter , Ibotenic Acid/toxicity , Cytokines/metabolism
13.
J Biochem Mol Toxicol ; 37(5): e23319, 2023 May.
Article in English | MEDLINE | ID: mdl-36811218

ABSTRACT

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.


Subject(s)
Brain Injuries , Chlorpyrifos , Pesticides , Animals , Mice , Antioxidants , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Molecular Docking Simulation , Organophosphorus Compounds , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt
14.
Basic Clin Pharmacol Toxicol ; 132(5): 449-453, 2023 May.
Article in English | MEDLINE | ID: mdl-36808477

ABSTRACT

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.


Subject(s)
Brain Injuries , Neurotoxicity Syndromes , Poisoning , Humans , Male , Rats , Animals , Lithium , Rats, Sprague-Dawley , Neurotoxicity Syndromes/etiology , Brain , Brain Injuries/chemically induced
15.
Environ Sci Pollut Res Int ; 30(17): 49014-49025, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36759409

ABSTRACT

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.


Subject(s)
Biological Products , NF-kappa B , Animals , Male , Rats , Antioxidants/metabolism , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Caspase 3/drug effects , Caspase 3/metabolism , Caspase 6/drug effects , Caspase 6/metabolism , Cyclooxygenase 2/metabolism , Fatty Acids/pharmacology , Fluorides/toxicity , Glutathione/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B/drug effects , NF-kappa B/metabolism , Oxidative Stress , Rats, Wistar , Signal Transduction/drug effects , Biological Products/pharmacology , Biological Products/therapeutic use , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism
16.
Jpn J Clin Oncol ; 53(4): 355-364, 2023 Mar 30.
Article in English | MEDLINE | ID: mdl-36579769

ABSTRACT

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.


Subject(s)
Brain Edema , Brain Injuries , Brain Neoplasms , Carcinoma, Renal Cell , Kidney Neoplasms , Radiosurgery , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/radiotherapy , Radiosurgery/adverse effects , Vascular Endothelial Growth Factor A , Brain Edema/chemically induced , Brain Edema/drug therapy , Retrospective Studies , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Vascular Endothelial Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Angiogenesis Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/radiotherapy , Brain Injuries/chemically induced , Brain Injuries/drug therapy
17.
Toxicol Mech Methods ; 33(4): 307-315, 2023 May.
Article in English | MEDLINE | ID: mdl-36177783

ABSTRACT

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.


Subject(s)
Brain Injuries , Down-Regulation , MicroRNAs , Sevoflurane , Animals , Rats , Animals, Newborn , Apoptosis , Brain Injuries/chemically induced , Caspase 3/metabolism , MicroRNAs/genetics
18.
Turk J Med Sci ; 53(5): 1058-1066, 2023.
Article in English | MEDLINE | ID: mdl-38813010

ABSTRACT

Background/aim: Propofol is a positive allosteric modulator of GABAA receptor (GABAAR) and has potent antioxidant activity. The aim of this study was to investigate the effect of propofol on damage to the cerebral cortex and hippocampus in a lithium chloride (LiCl)-pilocarpine animal model of status epilepticus (SE). Materials and methods: Adult male Sprague Dawley rats were injected with LiCl-pilocarpine to induce SE. They were then randomized and injected 30 min later with vehicle saline (SE+saline), propofol (SE+PPF, 50 mg/kg), Diazepam (SE+DZP, 10 mg/kg), Scopolamine (SE+SCOP, 10 mg/kg), or MK-801 (SE+MK-801, 2 mg/kg). Another group of rats received saline only and served as the naïve control (BLK). The levels of superoxide dismutase (SOD), glutathione (GSH) and malondialdehyde (MDA) in the serum, cortex and hippocampus were analyzed 2 and 24 h posttreatment. The degree of tissue damage in the cortex and hippocampus of individual rats was assessed 24 h posttreatment, together with expression of the GABAAR α1 subunit. Results: The propofol group showed reduced levels of tissue damage in the cerebral cortex and hippocampus, decreased levels of MDA, and increased levels of GSH compared to the SE+saline group. No changes in SOD level were observed in serum and tissue samples from the cortex and hippocampus of SE+saline rats. Immunohistochemistry and Western blot assays showed that propofol treatment significantly increased the expression of GABAAR α1 subunit in the cortical and hippocampal tissues of SE rats. Conclusion: Propofol treatment protected against SE-induced tissue injury in the cortex and hippocampus of rats. This was due at least in part to its antioxidant activity and to its induction of GABAAR α1 subunit expression in the brain.


Subject(s)
Disease Models, Animal , Lithium Chloride , Oxidative Stress , Pilocarpine , Propofol , Rats, Sprague-Dawley , Receptors, GABA-A , Status Epilepticus , Animals , Propofol/pharmacology , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Status Epilepticus/drug therapy , Pilocarpine/toxicity , Male , Lithium Chloride/pharmacology , Oxidative Stress/drug effects , Rats , Hippocampus/metabolism , Hippocampus/drug effects , Brain Injuries/metabolism , Brain Injuries/drug therapy , Brain Injuries/chemically induced , Malondialdehyde/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects
19.
Arch Biochem Biophys ; 729: 109390, 2022 10 30.
Article in English | MEDLINE | ID: mdl-36067878

ABSTRACT

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.


Subject(s)
Brain Injuries , NF-E2-Related Factor 2 , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Apoptosis , Brain Injuries/chemically induced , Brain Injuries/prevention & control , Doxorubicin/pharmacology , Female , Glutathione Disulfide/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Interleukin-2/pharmacology , Interleukin-4/metabolism , Lactate Dehydrogenases/metabolism , Mice , Mice, Inbred BALB C , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Phosphopyruvate Hydratase/metabolism , Phosphopyruvate Hydratase/pharmacology , Reactive Oxygen Species/metabolism , Serotonin/metabolism , Superoxide Dismutase/metabolism , Transforming Growth Factor beta/metabolism , gamma-Aminobutyric Acid/metabolism
20.
Eur J Pharmacol ; 932: 175217, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36007603

ABSTRACT

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.


Subject(s)
Brain Injuries , Neurodegenerative Diseases , Neuroprotective Agents , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/metabolism , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Caspase 3 , Cyclophosphamide/adverse effects , Cytokines/metabolism , Galectin 3 , Humans , Hydroxybenzoates , I-kappa B Kinase , Immunity , Inflammasomes/metabolism , Interleukin-18 , Interleukin-1beta/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Sirtuin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...