Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Neurochem Int ; 176: 105725, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38561151

ABSTRACT

Epilepsy constitutes a global health concern, affecting millions of individuals and approximately one-third of patients exhibit drug resistance. Recent investigations have revealed alterations in cerebral iron content in both epilepsy patients and animal models. However, the extant literature lacks a comprehensive exploration into the ramifications of modulating iron homeostasis as an intervention in epilepsy. This study investigated the impact of deferasirox, a iron ion chelator, on epilepsy. This study unequivocally substantiated the antiepileptic efficacy of deferasirox in a kainic acid-induced epilepsy model. Furthermore, deferasirox administration mitigated seizure susceptibility in a pentylenetetrazol-induced kindling model. Conversely, the augmentation of iron levels through supplementation has emerged as a potential exacerbating factor in the precipitating onset of epilepsy. Intriguingly, our investigation revealed a hitherto unreported discovery: ITPRIP was identified as a pivotal modulator of excitatory synaptic transmission, regulating seizures in response to deferasirox treatment. In summary, our findings indicate that deferasirox exerts its antiepileptic effects through the precise targeting of ITPRIP and amelioration of cerebral iron homeostasis, suggesting that deferasirox is a promising and novel therapeutic avenue for interventions in epilepsy.


Subject(s)
Anticonvulsants , Brain , Deferasirox , Epilepsy , Homeostasis , Iron Chelating Agents , Iron , Deferasirox/pharmacology , Iron/metabolism , Animals , Homeostasis/drug effects , Homeostasis/physiology , Epilepsy/drug therapy , Epilepsy/metabolism , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Male , Brain/drug effects , Brain/metabolism , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Mice , Kindling, Neurologic/drug effects , Pentylenetetrazole/toxicity , Rats, Sprague-Dawley
2.
Eur J Pharmacol ; 969: 176440, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38402930

ABSTRACT

This study investigated the effects of semaglutide (Sema) on the gut microbiota of obese mice induced with high-fat diet (HFD). Male C57BL/6 J mice aged 6 weeks were enrolled and randomly distributed to four groups, which were provided with a normal control diet (NCD,NCD + Sema) and a 60% proportion of a high-fat diet (HFD,HFD + Sema), respectively. HFD was given for 10 weeks to develop an obesity model and the intervention was lasted for 18 days. The results showed semaglutide significantly reduced body weight gain, areas under the curve (AUC) of glucose tolerance test and insulin resistance test, as well as adipose tissue weight in mice. Semaglutide effectively reduced lipid deposition and lipid droplet formation in the liver of obese mice, and regulated the expression of genes related to abnormal blood glucose regulation. Additionally, semaglutide influenced the composition of gut microbiota, mitigating the microbial dysbiosis induced by a high-fat diet by impacting the diversity of the gut microbiota. After the high-fat diet intervention, certain strains such as Akkermansia, Faecalibaculum, and Allobaculum were significantly decreased, while Lachnospiraceae and Bacteroides were significantly increased. However, the application of semaglutide restored the lost flora and suppressed excessive bacterial abundance. Moreover, semaglutide increased the content of tight junction proteins and repaired the damage to intestinal barrier function caused by the high-fat diet intervention. Furthermore, correlation analysis revealed inverse relationship among Akkermansia levels and weight gain, blood glucose levels, and various obesity indicators. Correlation analysis also showed that Akkermansia level was negatively correlated with weight gain, blood glucose levels and a range of obesity indicators. This phenomenon may explain the anti-obesity effect of semaglutide, which is linked to alterations in gut microbiota, specifically an increase in the abundance of Akkermansia. In summary, our findings indicate that semaglutide has the potential to alleviate gut microbiota dysbiosis, and the gut microbiota may contribute to the obesity-related effects of this drug.


Subject(s)
Gastrointestinal Microbiome , Glucagon-Like Peptides , Noncommunicable Diseases , Male , Mice , Animals , Diet, High-Fat/adverse effects , Blood Glucose/analysis , Dysbiosis/metabolism , Mice, Obese , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/microbiology , Weight Gain
3.
Neurotoxicology ; 94: 11-23, 2023 01.
Article in English | MEDLINE | ID: mdl-36374725

ABSTRACT

Arsenite is a well-documented neurotoxicant that widely exists in the environment. However, the detailed mechanisms of arsenite neurotoxicity are not fully clarified. Autophagy has been reported to be involved in many neurological problems induced by arsenite. Since beclin 1 is an essential mediator of autophagy, we herein used both adult wild-type (beclin 1+/+) and heterozygous disruption of beclin 1 (beclin 1+/-) mice for chronic administration of 50 mg/L arsenite via drinking water for 3 months. Our results demonstrated that exposure of arsenite caused the working memory deficit, anxiety-like behavior and motor coordination disorder in beclin 1+/+ mice, accompanied with pathological changes in morphology and electrophysiology in the cortical tissues. This treatment of arsenite significantly reduced the number of neuronal cells and induced microglia activation and synaptic transmission disorders in the wild-type mice as compared with vehicle controls. Intriguingly, by using beclin 1+/- mice, we found that heterozygous disruption of beclin 1 profoundly attenuated these neurotoxic effects induced by arsenite, mainly manifested by improvements in the neurobehavioral impairments, abnormal electrophysiologic alterations as well as dysregulation of synaptic transmission. These findings together indicate that regulation of autophagy via beclin 1 would be a potential strategy for treatment against arsenite neurotoxicity.


Subject(s)
Arsenites , Neurotoxicity Syndromes , Mice , Animals , Beclin-1/genetics , Beclin-1/pharmacology , Arsenites/toxicity , Synaptic Transmission , Neurotoxicity Syndromes/genetics , Autophagy
4.
Toxicol Mech Methods ; 33(2): 113-122, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35818324

ABSTRACT

Di (2-ethylhexyl) phthalate (DEHP) is one of the most prevalent xenoestrogen endocrine disruptor in daily life. A growing number of studies showed that DEHP could exhibit long-term adverse health effects on the human body, particularly in the liver, kidneys, heart and reproductive systems. However, the impact of oral intake of DEHP on the nervous system is extremely limited. In the present study, the adult C57BL/6J male mice were intragastrically administered with two dosages of DEHP for 35 days. The behavioral parameters were assessed using the elevated plus maze and open-field test. The mRNA expression levels of neuropeptides and the oxidative stress-associated proteins were detected by qPCR and western blot seperately. The histopathologic alterations of the brain were observed by H&E and Nissl staining. The results demonstrated that DEHP exposure could result in neurobehavioral impairments such as locomotor increase and anxiety-like behavior. Furthermore, pathological damages were clearly observed in the cerebral cortex and hippocampus, accompanied by a decrease in neuropeptides and an increase in oxidative stress, which were all positively correlated with the dose of DEHP. Together, these findings provide valuable clues into the DEHP-induced neurotoxicity.


Subject(s)
Diethylhexyl Phthalate , Mice , Animals , Humans , Male , Diethylhexyl Phthalate/toxicity , Mice, Inbred C57BL , Brain , Anxiety/chemically induced , Oxidative Stress
5.
Lipids ; 58(1): 19-32, 2023 01.
Article in English | MEDLINE | ID: mdl-36253942

ABSTRACT

Currently, there is a global trend of rapid increase in obesity, especially among adolescents. The antibiotics cocktails (ABX) therapy is commonly used as an adjunctive treatment for gut microbiota related diseases, including obesity. However, the effects of broad-spectrum antibiotics alone on young obese hosts have rarely been reported. In the present study, the 3-week-old C57BL/6J male mice fed a high-fat diet (HFD) were intragastric administration with ampicillin, vancomycin, metronidazole or neomycin for 30 days. The lipid metabolites in plasma were assessed by biochemical assay kits, and genes related to lipid metabolite in the white adipose were assessed by qPCR. To further analyze the underlying mechanisms, the expression of genes related to lipid metabolism, inflammatory reactions and oxidative stress in the liver were determined by qPCR assay. In addition, the expression of oxidative damage-associated proteins in the liver were detected by western blot. The results showed that oral antibiotics exposure could reduce body weight and fat index in HFD-fed mice, concurrent with the increase of white adipose lipolysis genes and the decrease of hepatic lipogenic genes. Furthermore, antibiotics treatment could clearly reverse the HFD-induced elevation of oxidative damage-related proteins in the liver. Together, these findings will provide valuable clues into the effects of antibiotics on obesity.


Subject(s)
Diet, High-Fat , Lipid Metabolism , Mice , Male , Animals , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Obesity/etiology , Obesity/genetics , Liver/metabolism , Anti-Bacterial Agents/pharmacology , Lipids
6.
Front Pharmacol ; 13: 841990, 2022.
Article in English | MEDLINE | ID: mdl-35401199

ABSTRACT

Traditional herbal medicine (THM) is used worldwide for its safety and effectiveness against various diseases. Huoxiang Zhengqi (HXZQ) is an extensively used Chinese THM formula targeting gastrointestinal disordered gastroenteritis via regulating the intestinal microbiome/immuno-microenvironment. However, the specific mechanisms remain largely unexplored, besides as a lifestyle drug, its safety on the gut microbiome homeostasis has never been investigated. In this study, the effects of HXZQ on the gut microbiome of healthy adults were investigated for the first time, and the antibiotic-induced gut microbiota dysbiosis mice model was applied for verification. Based on healthy adults, our results revealed that HXZQ exhibited mild and positive impacts on the bacterial diversity and the composition of the gut microbiome in a healthy state. As for an unhealthy state of the gut microbiome (with low bacterial diversity and deficient compositions), HXZQ significantly restored the bacterial diversity and recovered the abundance of Bacteroidetes. In the antibiotic-induced mice model, HXZQ distinctly revived the deficient gut microbial compositions impaired by antibiotics. At the genus level, the abundances that responded most strongly and positively to HXZQ were Bifidobacterium in healthy adults and Muribaculaceae, Lactobacillus, and Akkermansia in mice. In contrast, the abundance of Blautia in healthy adults, Enterococcus, and Klebsiella in mice showed inversely associated with HXZQ administration. At last, HXZQ might exhibit an anti-inflammatory effect by regulating the concentration of interleukin-6 in plasma while causing no significant changes in the colon tissue structure in mice. In conclusion, our results elucidate that the safety of HXZQ in daily use further reveals the modulatory effects of HXZQ on gut microbial community structure. These results will provide new insights into the interaction of THM and gut microbiome homeostasis and clues about the safe use of THM as a lifestyle drug for its further development.

7.
Phytomedicine ; 99: 153968, 2022 May.
Article in English | MEDLINE | ID: mdl-35183933

ABSTRACT

BACKGROUND: Huoxiangzhengqi oral liquid (HX), a pharmaceutical product made from traditional Chinese medicine formulas, has been commonly used in household medication for gastrointestinal disorders, but the mode of action remains largely unclear. PURPOSE: This study aims to investigate whether pretreatment with HX prevents lipopolysaccharide (LPS)-induced adverse effects and the potential mechanisms involved. METHODS: Seven-week-old male C57BL/6J mice were orally administered low (1.3 ml/kg) and high doses (2.6 ml/kg) of HX for 7 days, and subsequently subjected to a single dose of LPS at 6 mg/kg. Dexamethasone served as the positive control. Each group had ten animals. RESULTS: The data demonstrated that either a low or high dose of HX significantly reduced the levels of inflammation induced by LPS in both small intestinal and cortical tissues. LPS profoundly decreased the richness and evenness of the microbiota and disrupted the composition of the intestinal microbial community, but pretreatment with HX did not successfully prevent dysbiosis. No significant improvements in HX against LPS were observed in intestinal local immunity or the secretion of partial gut-brain peptides. In addition, pretreatment with HX prevented the alterations in the expression levels of proteins related to the NF-κB pathway, including phospho-p38, p38, phospho-p44/42, p44/42, p50 and p65 induced by LPS. CONCLUSION: Herein, we demonstrated for the first time that the preventive effects of HX against LPS mainly occur through the inhibition of inflammation. These findings provide novel evidence that HX may serve as a new agent for the prevention of gastrointestinal inflammation-related disorders.

8.
J Nanobiotechnology ; 19(1): 234, 2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34362405

ABSTRACT

BACKGROUND: Pregnancy exposure to titanium dioxide nanoparticles (TiO2NPs) is a vital consideration due to their inadvertent ingestion from environmental contamination. The potential health effects of TiO2NPs on the neurodevelopmental process should be seriously concerned in health risk assessment, especially for the pregnant women who are susceptible to the neurodevelopmental toxicity of nano-sized particles. However, the available evidence of neurodevelopmental toxicity of TiO2NPs remains very limited. METHODS: In the present study, the pregnant mice were intragastric administered with 150 mg/kg TiO2NPs from gestational day (GD) 8 to 21, the maternal behaviors and neurodevelopment-related indicators in offspring were all assessed at different time points after delivery. The gut microbial community in both dams and their offspring were detected by using 16S ribosomal RNA (rRNA) gene sequencing. The gut-brain axis related indicators were also determined in the offspring. RESULTS: The results clearly demonstrated that exposure to TiO2NPs did not affect the maternal behaviors of pregnant mice, or cause the deficits on the developmental milestones and perturbations in the early postnatal development of offspring. Intriguingly, our data revealed that pregnancy exposure of TiO2NPs did not affect locomotor function, learning and memory ability and anxiety-like behavior in offspring at postnatal day (PD) 21, but resulted in obvious impairments on these neurobehaviors at PD49. Similar phenomena were obtained in the composition of gut microbial community, intestinal and brain pathological damage in offspring in adulthood. Moreover, the intestinal dysbiosis induced by TiO2NPs might be highly associated with the delayed appearance of neurobehavioral impairments in offspring, possibly occurring through disruption of gut-brain axis. CONCLUSIONS: This is the first report elucidated that pregnancy exposure to TiO2NPs caused delayed appearance of neurobehavioral impairments in offspring when they reached adulthood, although these perturbations did not happen at early life after delivery. These findings will provide valuable insights about neurodevelopmental toxicity of TiO2NPs, and call for comprehensive health risk assessment of TiO2NPs on the susceptible population, such as pregnant women.


Subject(s)
Dysbiosis/chemically induced , Nanoparticles/adverse effects , Titanium/adverse effects , Animals , Brain-Gut Axis , Female , Gastrointestinal Microbiome/drug effects , Male , Mice , Mice, Inbred C57BL , Pregnancy
9.
J Nanobiotechnology ; 19(1): 174, 2021 Jun 10.
Article in English | MEDLINE | ID: mdl-34112173

ABSTRACT

BACKGROUND: Silicon dioxide nanoparticles (SiO2NPs) are widely used as additive in the food industry with controversial health risk. Gut microbiota is a new and hot topic in the field of nanotoxicity. It also contributes a novel and insightful view to understand the potential health risk of food-grade SiO2NPs in children, who are susceptible to the toxic effects of nanoparticles. METHODS: In current study, the young mice were orally administrated with vehicle or SiO2NPs solution for 28 days. The effects of SiO2NPs on the gut microbiota were detected by 16S ribosomal RNA (rRNA) gene sequencing, and the neurobehavioral functions were evaluated by open field test and Morris water maze. The level of inflammation, tissue integrity of gut and the classical indicators involved in gut-brain, gut-liver and gut-lung axis were all assessed. RESULTS: Our results demonstrated that SiO2NPs significantly caused the spatial learning and memory impairments and locomotor inhibition. Although SiO2NPs did not trigger evident intestinal or neuronal inflammation, they remarkably damaged the tissue integrity. The microbial diversity within the gut was unexpectedly enhanced in SiO2NPs-treated mice, mainly manifested by the increased abundances of Firmicutes and Patescibacteria. Intriguingly, we demonstrated for the first time that the neurobehavioral impairments and brain damages induced by SiO2NPs might be distinctively associated with the disruption of gut-brain axis by specific chemical substances originated from gut, such as Vipr1 and Sstr2. Unapparent changes in liver or lung tissues further suggested the absence of gut-liver axis or gut-lung axis regulation upon oral SiO2NPs exposure. CONCLUSION: This study provides a novel idea that the SiO2NPs induced neurotoxic effects may occur through distinctive gut-brain axis, showing no significant impact on either gut-lung axis or gut-liver axis. These findings raise the exciting prospect that maintenance and coordination of gastrointestinal functions may be critical for protection against the neurotoxicity of infant foodborne SiO2NPs.


Subject(s)
Brain/drug effects , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Nanoparticles/chemistry , Silicon Dioxide/pharmacology , Animals , Gastrointestinal Microbiome/genetics , Inflammation , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons , Silicon Dioxide/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...