Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Ecotoxicol Environ Saf ; 270: 115841, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38113799

ABSTRACT

N-nitrosodiethylamine (ND) is an extremely toxic unavoidable environmental contaminant. CopperII-albumin (CuAB) complex, a newly developed Cu complex, showed antioxidant and anti-inflammatory potential. Hereby, we explored the plausible neuroprotective role of CuAB complex toward ND-evoked neurotoxicity in mice. Twenty-four male mice were sorted into 4 groups (6 mice each). Control group, mice were administered oral distilled water; and CuAB group, mice received CuAB complex at a dose of 817 µg/kg orally, three times weekly. In ND group, ND was given intraperitoneally (50 mg/kg body weight, once weekly for 6 w). CuAB+ND group, mice were administered a combination of CuAB and ND. The brain was quickly extracted upon completion of the experimental protocol for the evaluation of the oxidative/antioxidative markers, inflammatory cytokines, and histopathological examination. Oxidative stress was induced after ND exposure indicated by a reduction in GSH and SOD1 level, with increased MDA level. In addition, decreased expression of SOD1 proteins, Nrf2, and 5-HT mRNA expression levels were noticed. An apoptotic cascade has also been elicited, evidenced by overexpression of Cyt c, Cl. Casp 3. In addition, increased regulation of proinflammatory genes (TNF-α, IL-6, iNOS, Casp1, and NF-κB (p65/p50); besides, increment of protein expression of P-IKBα and reduced expression of IKBα. Pretreatment with CuAB complex significantly ameliorated ND neuronal damage. Our results recommend CuAB complex supplementation because it exerts neuroprotective effects against ND-induced toxicity.


Subject(s)
Copper , Neurotoxicity Syndromes , Mice , Male , Animals , Copper/toxicity , Diethylnitrosamine/pharmacology , Superoxide Dismutase-1/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress , Signal Transduction , Antioxidants/pharmacology , Antioxidants/metabolism , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , NF-E2-Related Factor 2/metabolism
2.
Neurotoxicology ; 99: 305-312, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37979660

ABSTRACT

Methamphetamine (METH) is a psychostimulant with a very high addiction rate. Prolonged use of METH has been observed as one of the root causes of neurotoxicity. Melatonin (Mel) has been found to have a significant role in METH-induced neurotoxicity. This study aimed to investigate the restorative effect of Mel on behavioral flexibility in METH-induced cognitive deficits. Male Sprague-Dawley rats were randomly assigned to be intraperitoneally injected with saline (control) or Meth at 5 mg/kg for 7 consecutive days. Then, METH injection was withdrawn and rats in each group were subcutaneously injected with saline or Mel at 10 mg/kg for 14 consecutive days. The stereotypic behavioral test and attentional set-shifting task (ASST) were used to evaluate neurological functions and cognitive flexibility, respectively. Rats developed abnormal features of stereotyped behaviors and deficits in cognitive flexibility after 7 days of METH administration. However, post-treatment with Mel for 14 days after METH withdrawal dramatically ameliorated the neurological and cognitive deficits in METH-treated rats. Blood biomarkers indicated METH-induced systemic low-grade inflammation. Moreover, METH-induced endoplasmic reticulum (ER) stress in the prefrontal cortex was diminished by melatonin supplementation. These findings might reveal the therapeutic potential of Mel in METH toxicity-induced neurological and cognitive deficits.


Subject(s)
Central Nervous System Stimulants , Melatonin , Methamphetamine , Neurotoxicity Syndromes , Rats , Male , Animals , Methamphetamine/toxicity , Melatonin/pharmacology , Melatonin/therapeutic use , Rats, Sprague-Dawley , Central Nervous System Stimulants/toxicity , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Cognition , Endoplasmic Reticulum Stress
3.
Biomed Pharmacother ; 164: 114880, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37224751

ABSTRACT

3,4-methylenedioxymethamphetamine (MDMA) is a popular recreational drug, however over 200 studies demonstrate that acute (e.g. hyperthermia, rhabdomyolysis) and chronic (e.g. neurotoxicity) toxicity effects of MDMA were observed in different animals. Methimazole (MMI), an inhibitor of thyroid hormone synthesis, was found to significantly reduce the HSP72 expression of heat stress induced in fibroblasts. Hence, we attempted to understand the effects of MMI on MDMA induced changes in vivo. Male SD rats were randomly divided into four groups as follows:(a) water-saline (b) water-MDMA (c) MMI-saline and (d) MMI-MDMA group. In the temperature analysis test, MMI was found to alleviate MDMA-induced hyperthermia and increase the heat loss index (HLI), revealing its peripheral vasodilation effect. PET experiment suggested that MDMA induced elevated glucose uptake by skeletal muscles, which was resolved by MMI pretreatment. IHC staining (serotonin transporter, SERT) showed the evidence of neurotoxicity caused by MDMA (serotonin fiber loss), which was alleviated by MMI. Furthermore, the animal behaviour test (forced swimming test, FST) showed higher swimming time but lower immobility time in MMI-MDMA and MMI-saline groups. Taken together, treatment of MMI shows benefits such as lowered body temperature, alleviation of neurotoxicity and excited behaviour. However, further investigations should be conducted in the future to provide in-depth evidence for its clinical use.


Subject(s)
Hyperthermia, Induced , N-Methyl-3,4-methylenedioxyamphetamine , Neurotoxicity Syndromes , Rats , Male , Animals , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Methimazole/toxicity , Rats, Sprague-Dawley , Body Temperature , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Hyperthermia, Induced/adverse effects
4.
Biomed Pharmacother ; 161: 114475, 2023 May.
Article in English | MEDLINE | ID: mdl-36905810

ABSTRACT

Withania somnifera (WS) is utilized in Ayurvedic medicine owing to its central and peripheral beneficial properties. Several studies have accrued indicating that the recreational amphetamine-related drug (+/-)- 3,4-methylenedioxymethamphetamine (MDMA; Ecstasy) targets the nigrostriatal dopaminergic system in mice, inducing neurodegeneration and gliosis, causing acute hyperthermia and cognitive impairment. This study aimed to investigate the effect of a standardized extract of W. somnifera (WSE) on MDMA-induced neurotoxicity, neuroinflammation, memory impairment and hyperthermia. Mice received a 3-day pretreatment with vehicle or WSE. Thereafter, vehicle- and WSE-pretreated mice were randomly divided into four groups: saline, WSE, MDMA alone, WSE plus MDMA. Body temperature was recorded throughout treatment, and memory performance was assessed by a novel object recognition (NOR) task at the end of treatment. Thereafter, immunohistochemistry was performed to evaluate in the substantia nigra pars compacta (SNc) and striatum the levels of tyrosine hydroxylase (TH), as marker of dopaminergic degeneration, and of glial fibrillary acidic protein (GFAP) and TMEM119, as markers of astrogliosis or microgliosis, respectively. MDMA-treated mice showed a decrease in TH-positive neurons and fibers in the SNc and striatum respectively, an increase in gliosis and body temperature, and a decrease in NOR performance, irrespective of vehicle or WSE pretreatment. Acute WSE plus MDMA counteracted the modifications in TH-positive cells in SNc, GFAP-positive cells in striatum, TMEM in both areas and NOR performance, as compared to MDMA alone, while no differences were observed as compared to saline. Results indicate that WSE acutely administered in combination with MDMA, but not as pretreatment, protects mice against the noxious central effects of MDMA.


Subject(s)
Hyperthermia, Induced , N-Methyl-3,4-methylenedioxyamphetamine , Neurotoxicity Syndromes , Withania , Animals , Mice , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neuroinflammatory Diseases , Gliosis , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Cognition
5.
J Pediatr Hematol Oncol ; 45(1): 1-11, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36598958

ABSTRACT

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


Subject(s)
Bone Neoplasms , Leucovorin , Methotrexate , Neurotoxicity Syndromes , Osteosarcoma , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Bone Neoplasms/drug therapy , Leucovorin/therapeutic use , Methotrexate/adverse effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Neurotoxicity Syndromes/drug therapy , Osteosarcoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
6.
Toxicol Mech Methods ; 33(1): 38-46, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35574720

ABSTRACT

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


Subject(s)
Drugs, Chinese Herbal , Neuralgia , Neurotoxicity Syndromes , Humans , Kaempferols/pharmacology , Oxaliplatin/toxicity , Astrocytes , Databases, Factual , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Molecular Docking Simulation
7.
Food Funct ; 13(11): 6180-6194, 2022 Jun 06.
Article in English | MEDLINE | ID: mdl-35583008

ABSTRACT

In the present study, we aimed to delineate the neuroprotective potential of thymol (THY) against neurotoxicity and cognitive deterioration induced by thioacetamide (TAA) in an experimental model of hepatic encephalopathy (HE). Rats received TAA (100 mg kg-1, intraperitoneally injected, three times per week) for two weeks. THY (30 and 60 mg kg-1), and Vit E (100 mg k-1) were administered daily by oral gavage for 30 days after HE induction. Supplementation with THY significantly improved liver function, reduced serum ammonia level, and ameliorated the locomotor and cognitive deficits. THY effectively modulated the alteration in oxidative stress markers, neurotransmitters, and brain ATP content. Histopathology of liver and brain tissues showed that THY had ameliorated TAA-induced damage, astrocyte swelling and brain edema. Furthermore, THY downregulated NF-kB and upregulated GFAP protein expression. In addition, THY significantly promoted CREB and BDNF expression at both mRNA and protein levels, together with enhancing brain cAMP level. In conclusion, THY exerted hepato- and neuroprotective effects against HE by mitigating hepatotoxicity, hyperammonemia and brain ATP depletion via its antioxidant, anti-inflammatory effects in addition to activation of the CREB/BDNF signaling pathway.


Subject(s)
Hepatic Encephalopathy , Neurotoxicity Syndromes , Adenosine Triphosphate/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cognition , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/metabolism , Liver/metabolism , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Oxidative Stress , Rats , Rats, Wistar , Signal Transduction , Thioacetamide/toxicity , Thymol/pharmacology
8.
Molecules ; 27(8)2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35458770

ABSTRACT

Bisphenol A (BPA), a well-known xenoestrogen, is commonly utilised in the production of polycarbonate plastics. Based on the existing evidence, BPA is known to induce neurotoxicity and behavioural issues. Flavonoids such as silibinin and naringenin have been shown to have biological activity against a variety of illnesses. The current research evaluates the neuropharmacological effects of silibinin and naringenin in a zebrafish model against neurotoxicity and oxidative stress caused by Bisphenol A. In this study, a novel tank diving test (NTDT) and light−dark preference test (LDPT) were used in neurobehavioural investigations. The experimental protocol was planned to last 21 days. The neuroprotective effects of silibinin (10 µM) and naringenin (10 µM) in zebrafish (Danio rerio) induced by BPA (17.52 µM) were investigated. In the brine shrimp lethality assay, the 50% fatal concentrations (LC50) were 34.10 µg/mL (silibinin) and 91.33 µg/mL (naringenin) compared to the standard potassium dichromate (13.15 µg/mL). The acute toxicity investigation found no mortality or visible abnormalities in the silibinin- and naringenin-treated groups (LC50 > 100 mg/L). The altered scototaxis behaviour in LDPT caused by BPA was reversed by co-supplementation with silibinin and naringenin, as shown by decreases in the number of transitions to the light zone and the duration spent in the light zone. Our findings point to BPA's neurotoxic potential in causing altered scototaxis and bottom-dwelling behaviour in zebrafish, as well as the usage of silibinin and naringenin as potential neuroprotectants.


Subject(s)
Neuroprotective Agents , Neurotoxicity Syndromes , Animals , Benzhydryl Compounds/toxicity , Drug Design , Flavanones , Flavonoids , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Phenols , Silybin/pharmacology , Zebrafish
9.
Mar Drugs ; 20(4)2022 Mar 31.
Article in English | MEDLINE | ID: mdl-35447917

ABSTRACT

As the most abundant marine carotenoid extracted from seaweeds, fucoxanthin is considered to have neuroprotective activity via its excellent antioxidant properties. Oxidative stress is regarded as an important starting factor for neuronal cell loss and necrosis, is one of the causes of Parkinson's disease (PD), and is considered to be the cause of adverse reactions caused by the current PD commonly used treatment drug levodopa (l-DA). Supplementation with antioxidants early in PD can effectively prevent neurodegeneration and inhibit apoptosis in dopaminergic neurons. At present, the effect of fucoxanthin in improving the adverse effects triggered by long-term l-DA administration in PD patients is unclear. In the present study, we found that fucoxanthin can reduce cytotoxicity and suppress the high concentration of l-DA (200 µM)-mediated cell apoptosis in the 6-OHDA-induced PC12 cells through improving the reduction in mitochondrial membrane potential, suppressing ROS over-expression, and inhibiting active of ERK/JNK-c-Jun system and expression of caspase-3 protein. These results were demonstrated by PD mice with long-term administration of l-DA showing enhanced motor ability after intervention with fucoxanthin. Our data indicate that fucoxanthin may prove useful in the treatment of PD patients with long-term l-DA administration.


Subject(s)
Neurotoxicity Syndromes , Parkinson Disease , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Humans , Levodopa/toxicity , Mice , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/prevention & control , Oxidopamine/toxicity , PC12 Cells , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Rats , Xanthophylls/pharmacology , Xanthophylls/therapeutic use
10.
Oxid Med Cell Longev ; 2022: 8400876, 2022.
Article in English | MEDLINE | ID: mdl-35387263

ABSTRACT

Chronic and long-term methamphetamine (METH) abuse is bound to cause damages to multiple organs and systems, especially the central nervous system (CNS). Icariside II (ICS), a type of flavonoid and one of the main active ingredients of the traditional Chinese medicine Epimedium, exhibits a variety of biological and pharmacological properties such as anti-inflammatory, antioxidant, and anticancer activities. However, whether ICS could protect against METH-induced neurotoxicity remains unknown. Based on a chronic METH abuse mouse model, we detected the neurotoxicity after METH exposure and determined the intervention effect of ICS and the potential mechanism of action. Here, we found that METH could trigger neurotoxicity, which was characterized by loss of dopaminergic neurons, depletion of dopamine (DA), activation of glial cells, upregulation of α-synuclein (α-syn), abnormal dendritic spine plasticity, and dysfunction of motor coordination and balance. ICS treatment, however, alleviated the above-mentioned neurotoxicity elicited by METH. Our data also indicated that when ICS combated METH-induced neurotoxicity, it was accompanied by partial correction of the abnormal Kelch 2 like ECH2 associated protein 1 (Keap1)-nuclear factor erythroid-2-related factor 2 (Nrf2) pathway and oxidative stress response. In the presence of ML385, an inhibitor of Nrf2, ICS failed to activate the Nrf2-related protein expression and reduce the oxidative stress response. More importantly, ICS could not attenuate METH-induced dopaminergic neurotoxicity and behavioral damage when the Nrf2 was inhibited, suggesting that the neuroprotective effect of ICS on METH-induced neurotoxicity was dependent on activating the Keap1-Nrf2 pathway. Although further research is needed to dig deeper into the actual molecular targets of ICS, it is undeniable that the current results imply the potential value of ICS to reduce the neurotoxicity of METH abusers.


Subject(s)
Methamphetamine , Neurotoxicity Syndromes , Animals , Mice , Dopamine/metabolism , Flavonoids/therapeutic use , Kelch-Like ECH-Associated Protein 1/metabolism , Methamphetamine/toxicity , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/metabolism , NF-E2-Related Factor 2/metabolism
11.
J Food Biochem ; 46(7): e14115, 2022 07.
Article in English | MEDLINE | ID: mdl-35246863

ABSTRACT

This study reports the protective role of the aqueous extract of Syzygium aromaticum (ESA) against lead (Pb)-induced neurotoxicity in mice. Thirty male mice weighing between 18 g and 25 g were randomly divided into five groups. (1) Group 1 (control group), (2) group 2 (Pb-test group): was administered with a solution containing 0.1% (w/v) of lead acetate (PbAc), (3) group 3 (ESA + Pb100 group): was administered with 0.1% (w/v) of PbAc followed by 100 mg/kg of S. aromaticum extract by gavage, (4) group 4 (ESA + Pb200): was administered with 0.1% (w/v) of PbAc followed by 200 mg/kg of S. aromaticum extract, and (5) group 5 (ESA-group): was administered with 100 mg/kg of S. aromaticum. Level of lead was determined by atomic absorption spectroscopy. Cerebral cortex synaptosomes prepared from mice administered orally with lead-acetate shown a significantly increased (p < .05) in tyrosine hydroxylase and protein carbonyl level and significantly decreased (p < .05) superoxide dismutase, glutathione reductase, and glutathione transferase activities. Also, there was a significant increase in brain lead concentration level, however, it was observed that S. aromaticum significantly reduced (p < .05) the level of lead at all tested doses. S. aromaticum rescued cerebral cortex synaptosomes from lead-induced neurotoxicity by relieving oxidative stress and abating elevated tyrosine hydroxylase activity. Moreover, S. aromaticum at the different dose grade (100 mg and 200 mg) abrogated the loss of motor performance in mice groups induced with lead. Altogether, our findings showed that S. aromaticum possesses antioxidant and neuro-modulatory potential against lead-induced neuronal damage. PRACTICAL APPLICATIONS: Environmental pollution with heavy metals is a known public health concern and their incremental concentrations in soil and water have risen to an unprecedented degree. Lead is one of the top 10 contaminants on the WHO's list of substances of greatest public health concern that impact the brain. However, exogenous natural bioactive supplements molecules could be one of the remedies to reduce Pb-induced toxicity. Our findings indicate therefore that, S. aromaticum could be a good fit for lowering Pb neurotoxicity and could be suggested as a neuroprotective molecule against neurodegenerative diseases involving catecholaminergic dysfunction induced by metallic elements.


Subject(s)
Neurotoxicity Syndromes , Syzygium , Animals , Lead/toxicity , Male , Mice , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Oxidative Stress , Synaptosomes , Syzygium/chemistry , Tyrosine 3-Monooxygenase , Water
12.
Biol Trace Elem Res ; 200(12): 5104-5114, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35059981

ABSTRACT

Alzheimer's disease (AD) is a brain disorder associated with a gradual weakening in neurocognitive functions, neuroinflammation, and impaired signaling pathways. Resveratrol (RSV) has neuroprotective properties, but with low bioavailability, and low solubility in vivo. Selenium (Se) is an essential micronutrient for brain function. Thus, this study aimed to evaluate the role of formulated RSV-Se nanoparticles (RSV-SeNPs) on neurochemical and histopathological approaches associated with the AD model in rats induced by Aluminum chloride (AlCl3) at a dose of 100 mg/kg/day for 60 days. RSV-SeNPs supplementation attenuates the impaired oxidative markers and mitochondrial dysfunction. The ameliorative effect of RSV-SeNPs on cholinergic deficits was associated with clearance of amyloid ß (Aß). Furthermore, activation of phosphatidylinositol 3 kinase (PI3K) deactivates glycogen synthase kinase 3 beta (GSK-3ß)-mediated tau hyperphosphorylation. Additionally, RSV-SeNPs downregulate signal transducer and activator of transcription (STAT3) expression as well as interleukin-1ß (IL-1ß) levels, therefore alleviating neuroinflammation in AD. Moreover, RSV-SeNPs upregulate the expression of Sirtuin-1 (SIRT1) and lower that of microRNA-134, consequently increasing neurite outgrowth. Eventually, the obtained results showed that nano-formulation of resveratrol with selenium maximized the therapeutic potential of RSV against Alzheimer's disease not only by their antioxidant but also by anti-inflammatory effect improving the neurocognitive function and modulating the signaling pathways.


Subject(s)
Alzheimer Disease , MicroRNAs , Nanoparticles , Neurotoxicity Syndromes , Selenium , Aluminum Chloride , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cholinergic Agents , Glycogen Synthase Kinase 3 beta , Interleukin-1beta , Micronutrients/therapeutic use , Nanoparticles/chemistry , Neuroinflammatory Diseases , Neurotoxicity Syndromes/drug therapy , Phosphatidylinositol 3-Kinases , Rats , Resveratrol/pharmacology , Selenium/pharmacology , Selenium/therapeutic use , Sirtuin 1/metabolism
13.
Brain Res Bull ; 181: 21-29, 2022 04.
Article in English | MEDLINE | ID: mdl-35065185

ABSTRACT

Chemotherapy-induced cognitive impairment such as memory impairment and concentration problems are now extensively recognized as side effects of chemotherapy. These problems reduce the quality of life in patients. Therefore, the present study aims to examine the effects of calcitriol supplementation (100 ng/kg /day for five weeks) on cognitive impairment, behavioral deficits, and hippocampal brain-derived neurotrophic factor (BDNF) changes following cisplatin treatment (5 mg/kg/ once a week for five weeks). We also determined the impact of cisplatin and calcitriol administration on reaction time against the thermal stimulus and muscle strength. Our findings showed that cisplatin administration resulted in a significant increase in anxiety-like behaviors. Treatment of rats with cisplatin also impaired performance in the passive avoidance and novel object recognition tasks which are indicating cognitive deficits. Co-administration of calcitriol prevented the cisplatin-induced behavioral and cognitive impairments. Cisplatin exposure also resulted in enhanced reaction time to the thermal stimulus and decreased muscle ability. Besides, hippocampal BDNF levels were reduced in cisplatin-treated rats; however, calcitriol alleviated these effects of cisplatin and up-regulated BDNF mRNA in the hippocampus. In addition, calcitriol alone indicated a significant change in BDNF level compared to the control group. We conclude that increased hippocampal BDNF mediates the beneficial effects of calcitriol against neurotoxicity in cisplatin-exposed rats. However, further studies are required to explore the other mechanisms that mediate the beneficial effect of calcitriol.


Subject(s)
Antineoplastic Agents/adverse effects , Behavioral Symptoms/drug therapy , Brain-Derived Neurotrophic Factor/drug effects , Calcitriol/pharmacology , Cisplatin/adverse effects , Cognitive Dysfunction/drug therapy , Neurotoxicity Syndromes/drug therapy , Animals , Behavior, Animal/drug effects , Behavioral Symptoms/chemically induced , Behavioral Symptoms/metabolism , Calcitriol/administration & dosage , Calcium-Regulating Hormones and Agents , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Dietary Supplements , Disease Models, Animal , Male , Neurotoxicity Syndromes/metabolism , Rats , Up-Regulation
14.
Environ Sci Pollut Res Int ; 29(41): 62042-62054, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34591247

ABSTRACT

Over the years, many researches have shown the potential protective effects of ginseng for preventing and treating neurological damage and their related diseases. Neuronal disturbance is one of the most common serious effects of cisplatin chemotherapy that triggers memory impairment and cognitive disability. Based on the hypothesis that mechanistic pathways of ginseng against the neurological and biochemical disturbance remain unclear, therefore, this study was designed to investigate the neuroprotective effect of ginseng extract against neurological and behavior abnormality induced by cisplatin in male rats. Animals were divided into 4 groups. Group 1 served as a control, group 2 was orally administrated with ginseng (100 mg/kg BW) daily for 90 days, group 3 was injected intraperitoneally with cisplatin (4 mg/kg BW) once a week for 90 days, and group 4 received ginseng and cisplatin. Cisplatin induced a learning and memory dysfunction in the Morris water maze task and locomotor disability in the rotarod test. In addition, cisplatin disrupted the oxidant/antioxidant systems, neuroinflammatory molecules (TNF-α, IL-6, IL-12, and IL-1ß), neurotransmitters, and apoptotic (caspase-3, P53, and Bax) and dementia markers (amyloid-ß40 and amyloid-ß 42). Co-treatment with ginseng extracts successfully ameliorated the cognitive behaviors and intramuscular strength and presented a good protective agent against neurological damage. Histopathological and histochemical studies proved the neuroprotective effect of ginseng. Our data showed that ginseng capable to counteract the memory dysfunction is induced by cisplatin via reducing oxidative stress and neuroinflammation restoring the neurological efficiency.


Subject(s)
Neuroprotective Agents , Neurotoxicity Syndromes , Panax , Animals , Cisplatin/toxicity , Male , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , Oxidative Stress , Panax/chemistry , Rats
15.
Nutr Neurosci ; 25(6): 1173-1187, 2022 Jun.
Article in English | MEDLINE | ID: mdl-33164715

ABSTRACT

OBJECTIVES: The study was aimed to evaluate the efficacy of three commonly consumed Lactuca sativa (LS) Linn. varieties viz., Grand rapid, Lollo rosso and Iceberg (Asteraceae) against 3-NP induced HD like symptoms in rats. METHODS: Ethanol extracts of leaves of three LS varieties were prepared, and standardized on the basis of quercetin content using HPLC. These extracts (100 and 200 mg kg, p.o. for 20 days) were evaluated for their neuroprotective effect against 3-NP (10 mg/kg, i.p. for 14 days) induced neurotoxicity in male Wistar rats. The extract that exhibited maximum activity was successively fractionated using hexane, ethyl acetate, n-butanol and aqueous in increasing order of polarity. These fractions were also evaluated (dose equivalent to the dose of the extract of LS variety exhibiting maximum activity) for their neuroprotective effect. The protective effect of extracts and fractions was evaluated using different behavioral (rota rod, actophotometer, beam walk and Morris water maze) and biochemical (malondialdehyde, nitrite, superoxide dismutase, catalase and reduced glutathione) parameters. RESULTS: 3-NP elicit marked deterioration in motor coordination, locomotor activity and memory in comparison to control group. Standardized ethanol extract of grand rapid (200 mg/kg) exhibited maximum activity amongst the three tested varieties. Therefore, its fractions were also evaluated, and n-butanol fraction (40 mg/kg) exhibited maximum attenuation of 3-NP induced HD like symptoms which was evident from improved behavioral and biochemical parameters. DISCUSSION: The results exhibit that LS (Grand rapid variety) prophylaxis mitigated 3-NP induced neurotoxicity and HD like symptoms in rats due to its potent antioxidant potential.


Subject(s)
Huntington Disease , Neuroprotective Agents , Neurotoxicity Syndromes , 1-Butanol/adverse effects , Animals , Ethanol/toxicity , Huntington Disease/drug therapy , Lactuca , Male , Motor Activity , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Nitro Compounds , Plant Extracts/therapeutic use , Propionates , Rats , Rats, Wistar
16.
Brain Res Bull ; 178: 144-154, 2022 01.
Article in English | MEDLINE | ID: mdl-34838852

ABSTRACT

Intracerebral hemorrhage (ICH)-induced brain injury is a continuous pathological process that involves the deterioration of neurological functions, such as sensory, cognitive or motor functions. Cytotoxic byproducts of red blood cell lysis, especially free iron, appear to be a significant pathophysiologic mechanism leading to ICH-induced injury. Free iron has a crucial role in secondary brain injury after ICH. Chelating iron may attenuate iron-induced neurotoxicity and may be developed as a therapeutic candidate for ICH treatment. In this review, we focused on the potential role of iron toxicity in ICH-induced injury and iron chelation therapy in the management of ICH. It will hopefully advance our understanding of the pathogenesis of ICH and lead to new approaches for treatment.


Subject(s)
Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Ferroptosis/drug effects , Iron Chelating Agents/pharmacology , Iron/toxicity , Neurotoxicity Syndromes/drug therapy , Animals , Humans
17.
J Cancer Res Ther ; 17(6): 1473-1478, 2021.
Article in English | MEDLINE | ID: mdl-34916380

ABSTRACT

BACKGROUND: The aim of the present study was to evaluate the efficacy of TJ-107 for oxaliplatin-induced peripheral neurotoxicity in prospective, multi-institutional, randomized, double-blind, placebo-controlled Phase II trials using the functional assessment of cancer therapy/gynecologic oncology group 12-item neurotoxicity questionnaire (FACT-GOG-NTX-12). PATIENTS AND METHODS: The patients who were registered to the Goshajinkigan oxaliplatin neurotoxicity evaluation study (UMIN000002211) were analyzed. A NTX-12 from the validated FACT/GOG-NTX-12 was assessed before treatment and at the end of every 2 cycles. RESULTS: The comparisons of the median scores for TJ-107 and the placebo at 8 and 26 weeks were as follows: numbness or tingling in the hands (P = 0.5820), numbness or tingling in the feet (P = 0.3236), feeling of discomfort in the hands (P = 0.8219), feeling of discomfort in the feet (P = 0.5361), joint pain or muscle cramps (P = 0.1974), feeling weak all over (P = 0.2771), trouble hearing (P = 0.2832), ringing or buzzing in ears (P = 0.1031), trouble buttoning buttons (P = 0.1653), trouble feeling the shape of small objects when held in hand (P = 0.2919), trouble walking (P = 0.5406), and pain in the hands or feet when exposed to cold temperatures (P = 0.1872). CONCLUSION: There might be no clinically significant difference between the use of TJ-107 and the severity and quality of life for patients treated with oxaliplatin.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Colonic Neoplasms/drug therapy , Drugs, Chinese Herbal/administration & dosage , Neurotoxicity Syndromes/drug therapy , Oxaliplatin/adverse effects , Rectal Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Double-Blind Method , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Middle Aged , Neurotoxicity Syndromes/diagnosis , Neurotoxicity Syndromes/etiology , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Oxaliplatin/administration & dosage , Prospective Studies , Treatment Outcome
18.
Inflamm Res ; 70(10-12): 1151-1164, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34661679

ABSTRACT

OBJECTIVE: Gisenoside Rg1 is a potent neuroprotectant in ginseng. The aim of this study was to investigate the elimination effect of Rg1 on cadmium (Cd)-induced neurotoxicity. MATERIALS AND METHODS: A cumulative Cd exposure mouse model was established. Also, the toxicity of Cd and the protective effect of Rg1 were examined in vitro using cultured neurons and microglia. RESULTS: We found that Cd-intoxicated mice exhibited significant injury in the liver, kidney, small intestine, and testis, along with cognitive impairment. Antioxidant enzymes such as SOD, GSH-Px and CAT were reduced in the blood and brain, and correspondingly, the lipid peroxidation product MDA was elevated. In the brain, astrocytes and microglia were activated, characterized by an increase in inflammatory factors such as TNF-α, IL-1ß and IL-6, as well as their protein markers GFAP and IBA1. However, Rg1 eliminated Cd-induced toxicity and restored oxidative stress and inflammatory responses, correspondingly restoring the behavioral performance of the animals. Meanwhile, the BDNF-TrkB/Akt and Notch/HES-1 signaling axes were involved in the Rg1-mediated elimination of Cd-induced toxicity. CONCLUSION: Rg1 is a promising agent for the elimination of Cd-induced toxicity.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cadmium , Ginsenosides/therapeutic use , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Brain/drug effects , Brain/immunology , Brain/pathology , Cell Survival/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Ginsenosides/pharmacology , Intestine, Small/drug effects , Intestine, Small/pathology , Kidney/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , Oxidative Stress/drug effects , Oxidoreductases/immunology , Testis/drug effects , Testis/pathology
19.
Sci Rep ; 11(1): 14872, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34290261

ABSTRACT

Date pits are nutritious by-products, containing high levels of indigestible carbohydrates and polyphenols. To maximize the biological effects of the active ingredients, the hard shell of the polysaccharide must be degraded. Therefore, the current study aimed to assess the protective potentials of date pits extract (DP) and fungal degraded date pits extract (FDDP) against scopolamine (SCO)-induced neurodegeneration in male rats. Date pits were subjected to fungal degradation and extraction, followed by the measurement of phytochemicals and free radical scavenging activities. Forty-two adult Sprague-Dawley male rats were divided into seven groups: three control groups administered with either saline, DP or FDDP; four groups with neurodegeneration receiving SCO (ip 2 mg/kg/day, SCO group) with no treatment, SCO with DP (oral 100 mg/kg/day, DP + SCO group), SCO with FDDP (oral, 100 mg/kg/day, FDDP + SCO group), and SCO with donepezil (DON, oral, 2.25 mg/kg/day, DON + SCO group). The treatment duration was 28 days, and in the last 14 days, SCO was administered daily. Morris water maze test, acetylcholine esterase activity, oxidative stress, markers of inflammation and amyloidogenesis, and brain histopathology were assessed.


Subject(s)
Hypocreales/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Phoeniceae/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Scopolamine/antagonists & inhibitors , Scopolamine/toxicity , Seeds/chemistry , Animals , Drug Synergism , Free Radical Scavengers , Male , Phytochemicals/analysis , Plant Extracts/administration & dosage , Plant Extracts/metabolism , Rats, Sprague-Dawley
20.
Molecules ; 26(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070220

ABSTRACT

Alzheimer's disease (AD) is one of the most common neurodegenerative diseases leading to dementia. Despite research efforts, currently there are no effective pharmacotherapeutic options for the prevention and treatment of AD. Recently, numerous studies highlighted the beneficial effects of curcumin (CUR), a natural polyphenol, in the neuroprotection. Especially, its dual antioxidant and anti-inflammatory properties attracted the interest of researchers. In fact, besides its antioxidant and anti-inflammatory properties, this biomolecule is not degraded in the intestinal tract. Additionally, CUR is able to cross the blood-brain barrier and could therefore to be used to treat neurodegenerative pathologies associated with oxidative stress, inflammation and apoptosis. The present study aimed to assess the ability of CUR to induce neuronal protective and/or recovery effects on a rat model of neurotoxicity induced by aluminum chloride (AlCl3), which mimics the sporadic form of Alzheimer's disease. Our results showed that treatment with CUR enhances pro-oxidant levels, antioxidant enzymes activities and anti-inflammatory cytokine production and decreases apoptotic cells in AlCl3-exposed hippocampus rats. Additionally, histopathological analysis of hippocampus revealed the potential of CUR in decreasing the hallmarks in the AlCl3-induced AD. We also showed that CUR post-treatment significantly improved the behavioral, oxidative stress and inflammation in AlCl3-exposed rats. Taken together, our data presented CUR as a nutraceutical potential through its protective effects that are more interesting than recovery ones in sporadic model of AD.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Curcumin/therapeutic use , Neurotoxicity Syndromes/complications , Neurotoxicity Syndromes/drug therapy , Acetylcholinesterase/metabolism , Aluminum Chloride/administration & dosage , Animals , Anxiety/complications , Anxiety/drug therapy , Apoptosis/drug effects , Body Weight/drug effects , Cell Survival/drug effects , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Curcumin/pharmacology , Cytokines/metabolism , Disease Models, Animal , Hippocampus/pathology , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Nerve Degeneration/pathology , Neuroprotective Agents/pharmacology , Organ Size/drug effects , Oxidative Stress/drug effects , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL