Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 291
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Ethnopharmacol ; 326: 117735, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38211824

ABSTRACT

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


Subject(s)
Colorectal Neoplasms , Neurotoxicity Syndromes , Wolves , Animals , Humans , Oxaliplatin/adverse effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Colorectal Neoplasms/drug therapy , Inflammation
2.
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
3.
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
4.
Food Chem Toxicol ; 178: 113934, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37423315

ABSTRACT

Evidence suggests that furan, a widespread environmental and food contaminant, causes liver toxicity and cancer, but its implications in the brain are not well defined. We measured behavioral, glial, and biochemical responses in male juvenile rats exposed orally to 2.5, 5 and 10 mg/kg furan and vitamin E after 28 days. Furan-mediated hyperactivity peaked at 5 mg/kg and did not exacerbate at 10 mg/kg. Enhanced motor defect was also observed at 10 mg/kg. Furan-treated rats elicited inquisitive exploration but showed impaired spatial working memory. Without compromising the blood-brain barrier, furan induced glial reactivity with enhanced phagocytic activity, characterized by parenchyma-wide microglial aggregation and proliferation, which switched from hyper-ramified to rod-like morphology with increasing doses. Furan altered the glutathione-S-transferase-driven enzymatic and non-enzymatic antioxidant defence systems differentially and dose-dependently across brain regions. Redox homeostasis was most perturbed in the striatum and least disrupted in hippocampus/cerebellum. Vitamin E supplementation attenuated exploratory hyperactivity and glial reactivity but did not affect impaired working memory and oxidative imbalance. Overall, sub-chronic exposure of juvenile rats to furan triggered glial reactivity and behavioral deficits suggesting the brain's vulnerability during juvenile development to furan toxicity. It remains to be determined whether environmentally relevant furan concentrations interfere with critical brain developmental milestones.


Subject(s)
Gliosis , Neurotoxicity Syndromes , Rats , Male , Animals , Rats, Wistar , Gliosis/chemically induced , Oxidative Stress , Neurotoxicity Syndromes/etiology , Vitamin E , Furans/toxicity
5.
Environ Res ; 233: 116430, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37329943

ABSTRACT

Natural deposits and human-caused releases of uranium have led to its contamination in the nature. Toxic environmental contaminants such as uranium that harm cerebral processes specifically target the brain. Numerous experimental researches have shown that occupational and environmental uranium exposure can result in a wide range of health issues. According to the recent experimental research, uranium can enter the brain after exposure and cause neurobehavioral problems such as elevated motion related activity, disruption of the sleep-wake cycle, poor memory, and elevated anxiety. However, the exact mechanism behind the factor for neurotoxicity by uranium is still uncertain. This review primarily aims on a brief overview of uranium, its route of exposure to the central nervous system, and the likely mechanism of uranium in neurological diseases including oxidative stress, epigenetic modification, and neuronal inflammation has been described, which could present the probable state-of-the-art status of uranium in neurotoxicity. Finally, we offer some preventative strategies to workers who are exposed to uranium at work. In closing, this study highlights the knowledge of uranium's health dangers and underlying toxicological mechanisms is still in its infancy, and there is still more to learn about many contentious discoveries.


Subject(s)
Neurotoxicity Syndromes , Uranium , Humans , Uranium/toxicity , Environmental Exposure , Brain , Neurotoxicity Syndromes/etiology , Learning
6.
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
7.
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
8.
J Clin Psychopharmacol ; 43(2): 145-148, 2023.
Article in English | MEDLINE | ID: mdl-36795014

ABSTRACT

BACKGROUND: Common adverse effects of valproate include sedation, tremor, gastrointestinal effects, and weight gain. Valproate-associated hyperammonemic encephalopathy (VHE) is an uncommon adverse effect of valproate therapy, which includes symptoms such as tremors, ataxia, seizures, confusion, sedation and coma. We report clinical features and management of 10 cases of VHE in a tertiary care center. METHODS: In a retrospective chart review of case records from January 2018 to June 2021, 10 patients with VHE were identified and included in this case series. The data collected include demographic information, psychiatric diagnosis, comorbidities, liver function tests, serum ammonia and serum valproate levels, dosages and duration of valproate, management of hyperammonemia including dosage variations, discontinuation, adjuvant drugs used, and whether rechallenge was done. RESULTS: The most common indication of starting valproate was bipolar disorder (n = 5). All the patients had more than one physical comorbidity and risk factors for developing hyperammonemia. Seven patients received valproate at a dose higher than 20 mg/kg. The duration of valproate use varied from 1 week to 19 years before developing VHE. Dose reduction or discontinuation and lactulose were the most common management strategies used. All 10 patients improved. Among the 7 patients in whom valproate was discontinued, for 2 patients valproate was reinitiated in inpatient care with careful monitoring and was found to be well tolerated. CONCLUSIONS: This case series highlights the need for a high index of suspicion for VHE as it is frequently associated with a delayed diagnosis and recovery in psychiatric settings. Screening for risk factors and serial monitoring may allow earlier diagnosis and management.


Subject(s)
Brain Diseases , Hyperammonemia , Neurotoxicity Syndromes , Humans , Valproic Acid/adverse effects , Tertiary Care Centers , Retrospective Studies , Tremor/drug therapy , Neurotoxicity Syndromes/etiology , Brain Diseases/chemically induced , Anticonvulsants/adverse effects
9.
Ugeskr Laeger ; 185(5)2023 01 30.
Article in Danish | MEDLINE | ID: mdl-36760188

ABSTRACT

In this case report, a four-week-old boy, who since birth had suffered from constipation and infantile colic, presented with seizures, myoclonic jerks and irritability after being fed with a herbal mixture of star anise the day prior to admission. Chinese star anise is a globally used spice and a herbal remedy for infantile colic. The quick recovery of the boy and normal paraclinical findings supported the assumption of star anise intoxication. The frequent use of complementary medicine in children warrants awareness to inform families of the potential dangers of this home remedy.


Subject(s)
Colic , Illicium , Neurotoxicity Syndromes , Male , Humans , Child , Colic/complications , Neurotoxicity Syndromes/etiology , Seizures/complications , Eating
10.
J Integr Neurosci ; 22(1): 9, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36722237

ABSTRACT

Bilirubin neurotoxicity is a serious consequence of hyperbilirubinemia, which is the most common disease of the neonatal period. Clinically, bilirubin neurotoxicity can result in motor deficit, auditory dysfunction, cerebral palsy, seizure and neurodevelopmental diseases, amongst others. Bilirubin neurotoxicity is one of the major worldwide causes of neonatal brain injury, especially in poorer developing countries. However, the mechanisms of bilirubin neurotoxicity are still unclear. After the failure of attempts targeting neurons in many neurodegenerative disorders, neuroinflammation has become a significant target of research. Here, recent advances concerning neuroinflammation in bilirubin neurotoxicity are reported with a focus on the clinical characteristics of bilirubin neurotoxicity, including age-dependency, region-specificity and its yin-yang properties. Effects of neuroinflammation on blood brain interfaces and treatments targeting neuroinflammation in bilirubin neurotoxicity are also reviewed, which may promote the precision of future treatment of bilirubin neurotoxicity.


Subject(s)
Brain Injuries , Cerebral Palsy , Neurodevelopmental Disorders , Neurotoxicity Syndromes , Infant, Newborn , Humans , Bilirubin , Neuroinflammatory Diseases , Neurotoxicity Syndromes/etiology
11.
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
12.
Toxicol Mech Methods ; 33(3): 173-182, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35920262

ABSTRACT

The ever-expanding prevalence of adverse neurotoxic reactions of the brain in response to therapeutic and recreational drugs, dietary supplements, environmental hazards, cosmetic ingredients, a spectrum of herbals, health status, and environmental stressors continues to prompt the development of novel cell-based assays to better determine neurotoxic hazard. Neurotoxicants may cause direct and epigenetic damage to the nervous tissue and alter the chemistry, structure, or normal activity of the nervous system. In severe neurotoxicity due to exposure to physical or psychosocial toxicants, neurons are disrupted or killed, and a consistent pattern of clinical neural dysfunction appears. In utero exposure to neurotoxicants can lead to altered development of the nervous system [developmental neurotoxicity (DNT)]. Patients with certain disorders and certain genomic makeup may be particularly susceptible to neurotoxicants. Traditional cytotoxicity measurements, like cell death, are easy to measure, but insufficient at identifying current routine biomarkers of toxicity including functional impairment in cell communication, which often occurs before or even in the absence of cell death. The present paper examines some of the limitations of existing neurotoxicology in light of the increasing need to develop tools to meet the challenges of achieving greater sensitivity in detection and developing and standardizing methods for exploring the toxicologic risk of such neurotoxic entities as engineered nanomaterials and even variables associated with poverty.


Subject(s)
Neurotoxicity Syndromes , Humans , Neurotoxicity Syndromes/etiology , Neurons
13.
Cell Biol Toxicol ; 39(1): 319-343, 2023 02.
Article in English | MEDLINE | ID: mdl-35701726

ABSTRACT

Adverse outcome pathways (AOPs) are organized sequences of key events (KEs) that are triggered by a xenobiotic-induced molecular initiating event (MIE) and summit in an adverse outcome (AO) relevant to human or ecological health. The AOP framework causally connects toxicological mechanistic information with apical endpoints for application in regulatory sciences. AOPs are very useful to link endophenotypic, cellular endpoints in vitro to adverse health effects in vivo. In the field of in vitro developmental neurotoxicity (DNT), such cellular endpoints can be assessed using the human "Neurosphere Assay," which depicts different endophenotypes for a broad variety of neurodevelopmental KEs. Combining this model with large-scale transcriptomics, we evaluated DNT hazards of two selected Chinese herbal medicines (CHMs) Lei Gong Teng (LGT) and Tian Ma (TM), and provided further insight into their modes-of-action (MoA). LGT disrupted hNPC migration eliciting an exceptional migration endophenotype. Time-lapse microscopy and intervention studies indicated that LGT disturbs laminin-dependent cell adhesion. TM impaired oligodendrocyte differentiation in human but not rat NPCs and activated a gene expression network related to oxidative stress. The LGT results supported a previously published AOP on radial glia cell adhesion due to interference with integrin-laminin binding, while the results of TM exposure were incorporated into a novel putative, stressor-based AOP. This study demonstrates that the combination of phenotypic and transcriptomic analyses is a powerful tool to elucidate compounds' MoA and incorporate the results into novel or existing AOPs for a better perception of the DNT hazard in a regulatory context.


Subject(s)
Adverse Outcome Pathways , Neural Stem Cells , Neurotoxicity Syndromes , Humans , Rats , Animals , Laminin/pharmacology , Neurotoxicity Syndromes/etiology , Oxidative Stress , Risk Assessment/methods
14.
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
15.
Int J Mol Sci ; 23(23)2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36499072

ABSTRACT

The recreational use of nitrous oxide (N2O) has increased over the years. At the same time, more N2O intoxications are presented to hospitals. The incidental use of N2O is relatively harmless, but heavy, frequent and chronic use comes with considerable health risks. Most importantly, N2O can inactivate the co-factor cobalamin, which, in turn, leads to paresthesia's, partial paralysis and generalized demyelinating polyneuropathy. In some patients, these disorders are irreversible. Several metabolic cascades have been identified by which N2O can cause harmful effects. Because these effects mostly occur after prolonged use, it raises the question of whether N2O has addictive properties, explaining its prolonged and frequent use at high dose. Several lines of evidence for N2O's dependence liability can be found in the literature, but the underlying mechanism of action remains controversial. N2O interacts with the opioid system, but N2O also acts as an N-methyl-D-aspartate (NMDA) receptor antagonist, by which it can cause dopamine disinhibition. In this narrative review, we provide a detailed description of animal and human evidence for N2O-induced abuse/dependence and for N2O-induced neurotoxicity.


Subject(s)
Neurotoxicity Syndromes , Nitrous Oxide , Substance-Related Disorders , Animals , Humans , Dopamine , Neurotoxicity Syndromes/etiology , Nitrous Oxide/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Vitamin B 12 , Substance-Related Disorders/complications
16.
Cancer Immunol Res ; 10(12): 1433-1440, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36259217

ABSTRACT

A major complication of chimeric antigen receptor (CAR) T-cell therapy is immune effector cell-associated neurotoxicity syndrome (ICANS), which presents as aphasia, confusion, weakness, somnolence, seizures, and coma. This is similar to the neurologic manifestations of hypophosphatemia, which can result from sudden increases in metabolic demand for phosphorylated intermediates (e.g., refeeding syndrome and sepsis). Given these similarities, we investigated whether CAR T-cell effector metabolic activity is associated with increased extracellular phosphate consumption and a possible association between hypophosphatemia and ICANS. In vitro 4-1BB and CD28 CD19-targeted CAR T-cell effector activity was found to be associated with increased consumption of media phosphorus, which was temporally associated with increased single-cell effector secretomic activity and increased phosphorus-dependent metabolic demand of the CAR T cells. A clinical cohort of 77 patients treated with CD19-targeted CAR T-cell therapy demonstrated a significant anticorrelation between serum phosphorus and ICANS incidence and severity, with earlier onset of hypophosphatemia after CAR T-cell infusion more likely to result in neurotoxicity. These results imply phosphorous level monitoring could alert to the development of ICANS in clinical scenarios. See related Spotlight by Tobin et al., p. 1422.


Subject(s)
Hypophosphatemia , Neurotoxicity Syndromes , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell , Antigens, CD19 , Neurotoxicity Syndromes/etiology , Hypophosphatemia/chemically induced , Phosphorus
17.
Neurotoxicology ; 91: 329-339, 2022 07.
Article in English | MEDLINE | ID: mdl-35753508

ABSTRACT

This study investigated the neuroprotective role of selenium nanoparticles (SeNPs) on deltamethrin-induced neurotoxicity in rats. A total of 32 adult male Wister rats were allocated into the following four groups: 1) control, 2) deltamethrin (0.6 mg/kg), 3) SeNPs (0.5 mg/kg), and 4) deltamethrin + SeNPs. All agents were administered orally three times per week for 2 months. Locomotor behavior, anxiety-like behavior, biochemical parameters, including brain oxidative damage biomarkers (Malondialdehyde (MDA) and reduced glutathione (GSH)), brain acetylcholinesterase (AChE), and brain genotoxicity were evaluated. The gene expression levels of IGF-1 and Bcl2 were also determined. Moreover, a brain histopathological examination associated with the immunohistochemical determination of Bax in brain tissue was performed. Deltamethrin-intoxicated rats showed a reduction in the locomotor activity associated with a highly anxious state. They also displayed a disturbance in the brain redox state with a decrease in the brain AChE levels and a high DNA fragmentation percentage. Furthermore, they showed a decrement in the immunohistochemical GFAP levels as well as IGF-1 and Bcl2 gene expression levels with an increase in the immunohistochemical Bax levels. All these changes were confirmed by brain histopathology. Interestingly, SeNPs ameliorated all these changes and restored the normal brain architecture. In conclusion. SeNPs possess a potent medicinal activity due to their antioxidant and anti-inflammatory activity. Therefore, SeNPs can be a potential agent in ameliorating deltamethrin-induced neurotoxicity.


Subject(s)
Nanoparticles , Neurotoxicity Syndromes , Selenium , Acetylcholinesterase/metabolism , Animals , Antioxidants/pharmacology , Apoptosis , Insulin-Like Growth Factor I/metabolism , Male , Nanoparticles/toxicity , Neuroprotection , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/prevention & control , Nitriles , Oxidative Stress , Pyrethrins , Rats , Rats, Wistar , Selenium/pharmacology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
18.
Toxicol Appl Pharmacol ; 449: 116137, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35750205

ABSTRACT

Workers in the oil and gas industry are at risk for exposure to a number of physical and chemical hazards at the workplace. Chemical hazard risks include inhalation of crude oil or its volatile components. While several studies have investigated the neurotoxic effects of volatile hydrocarbons, in general, there is a paucity of studies assessing the neurotoxicity of crude oil vapor (COV). Consequent to the 2010 Deepwater Horizon (DWH) oil spill, there is growing concern about the short- and long-term health effects of exposure to COV. NIOSH surveys suggested that the DWH oil spill cleanup workers experienced neurological symptoms, including depression and mood disorders, but the health effects apart from oil dispersants were difficult to discern. To investigate the potential neurological risks of COV, male Sprague-Dawley rats were exposed by whole-body inhalation to COV (300 ppm; Macondo surrogate crude oil) following an acute (6 h/d × 1 d) or sub-chronic (6 h/d × 4 d/wk. × 4 wks) exposure regimen. At 1, 28 or 90 d post-exposure, norepinephrine (NE), epinephrine (EPI), dopamine (DA) and serotonin (5-HT) were evaluated as neurotransmitter imbalances are associated with psychosocial-, motor- and cognitive- disorders. Sub-chronic COV exposure caused significant reductions in NE, EPI and DA in the dopaminergic brain regions, striatum (STR) and midbrain (MB), and a large increase in 5-HT in the STR. Further, sub-chronic exposure to COV caused upregulation of synaptic and Parkinson's disease-related proteins in the STR and MB. Whether such effects will lead to neurodegenerative outcomes remain to be investigated.


Subject(s)
Neurotoxicity Syndromes , Petroleum Pollution , Petroleum , Water Pollutants, Chemical , Animals , Gases , Male , Neurotoxicity Syndromes/etiology , Neurotransmitter Agents , Rats , Rats, Sprague-Dawley , Serotonin , Water Pollutants, Chemical/toxicity
19.
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
20.
Support Care Cancer ; 30(9): 7281-7292, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35596774

ABSTRACT

BACKGROUND AND OBJECTIVE: Paclitaxel and doxorubicin are associated with neurotoxicity and cardiotoxicity respectively. This study aimed at investigating the role of alpha-lipoic acid (ALA) in counteracting paclitaxel-induced neuropathy and doxorubicin-associated cardiotoxicity in women with breast cancer. PATIENTS AND METHODS: This randomized double-blind placebo-controlled prospective study included 64 patients with breast cancer who were randomized into control group (n = 32) which received 4 cycles of doxorubicin plus cyclophosphamide (every 21 days) followed by weekly doses of paclitaxel for 12 weeks plus placebo tablets once daily and ALA group (n = 32) which received the same chemotherapeutic regimen plus ALA 600 once daily for 6 months. Patients were assessed by National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE version 4.0) for grading of neuropathy and by 12-item neurotoxicity questionnaire (Ntx-12). The assessment included also echocardiography and evaluation of serum levels of brain natriuretic peptide (BNP), tumor necrosis factor-alpha (TNF-α), malondialdehyde (MDA), and neurotensin (NT). Data were analyzed by paired and unpaired t-test, Mann-Whitney U test, and chi-square test. RESULTS: As compared to placebo, ALA provoked significant improvement in NCI-CTCAE neuropathy grading and Ntx-12 score after the end of 9th and 12th weeks of paclitaxel intake (p = 0.039, p = 0.039, p = 0.03, p = 0.004, respectively). At the end of the chemotherapy cycles, ALA resulted in significant decline in serum levels of BNP, TNF-α, MDA, and neurotensin (p < 0.05) as compared to baseline data and placebo. CONCLUSION: Alpha-lipoic acid may represent a promising adjuvant therapy to attenuate paclitaxel-associated neuropathy and doxorubicin-induced cardiotoxicity in women with breast cancer. TRIAL REGISTRATION: ClinicalTrials.gov: NCT03908528.


Subject(s)
Breast Neoplasms , Neurotoxicity Syndromes , Peripheral Nervous System Diseases , Thioctic Acid , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/pathology , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Doxorubicin , Female , Humans , Neurotensin/blood , Neurotoxicity Syndromes/etiology , Paclitaxel , Peripheral Nervous System Diseases/chemically induced , Prospective Studies , Thioctic Acid/therapeutic use , Tumor Necrosis Factor-alpha/blood
SELECTION OF CITATIONS
SEARCH DETAIL