Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
J Bioenerg Biomembr ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38720136

ABSTRACT

Vesicle-associated membrane protein 8 (VAMP8), a soluble n-ethylmaleimide-sensitive factor receptor protein, acts as an oncogenic gene in the progression of several malignancies. Nevertheless, the roles and mechanisms of VAMP8 in colorectal cancer (CRC) progression remain unknown. The expression and prognostic significance of VAMP8 in CRC samples were analyzed through bioinformatics analyses. Cell proliferation was detected using CCK-8 and EdU incorporation assays and apoptosis was evaluated via flow cytometry. Western blot analysis was conducted to examine the protein expression. Ferroptosis was evaluated by measurement of iron metabolism, lipid peroxidation, and glutathione (GSH) content. VAMP8 was increased in CRC samples relative to normal samples on the basis of GEPIA and HPA databases. CRC patients with high level of VAMP8 had a worse overall survival. VAMP8 depletion led to a suppression of proliferation and promotion of apoptosis in CRC cells. Additionally, VAMP8 knockdown suppressed beclin1 expression and LC3-II/LC3-I ratio, elevated p62 expression, increased Fe2+, labile iron pool, lipid reactive oxygen species, and malondialdehyde levels, and repressed GSH content and glutathione peroxidase activity. Moreover, VAMP8 knockdown inhibited the activation of janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathway in CRC cells. Mechanistically, activation of the JAK/STAT3 pathway by JAK1 or JAK2 overexpression attenuated VAMP8 silencing-mediated anti-proliferative, pro-apoptotic, anti-autophagic, and pro-ferroptotic effects on CRC cells. In conclusion, VAMP8 knockdown affects the proliferation, apoptosis, autophagy, and ferroptosis by the JAK/STAT3 pathway in CRC cells.

3.
Arch Toxicol ; 98(2): 395-408, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38103071

ABSTRACT

Artificial light at night (ALAN) pollution has been regarded as a global environmental concern. More than 80% of the global population is exposed to light pollution. Exacerbating this issue, artificially lit outdoor areas are growing by 2.2% per year, while continuously lit areas have brightened by 2.2% each year due to rapid population growth and expanding urbanization. Furthermore, the increasing prevalence of night shift work and smart device usage contributes to the inescapable influence of ALAN. Studies have shown that ALAN can disrupt endogenous biological clocks, resulting in a disturbance of the circadian rhythm, which ultimately affects various physiological functions. Up until now, scholars have studied various disease mechanisms caused by ALAN that may be related to the response of the circadian system to light. This review outlines the molecular mechanisms by which ALAN causes circadian rhythm abnormalities in sleep disorders, endocrine diseases, cardiovascular disease, cancer, immune impairment, depression, anxiety and cognitive impairments.


Subject(s)
Light Pollution , Shift Work Schedule , Lighting/adverse effects , Circadian Rhythm/physiology , Environmental Pollution
4.
Food Chem Toxicol ; 182: 114190, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37967789

ABSTRACT

Methylmercury (MeHg) is a widely distributed environmental pollutant that can easily cross the blood-brain barrier and accumulate in the brain, thereby damaging the central nervous system. Studies have shown that MeHg-induced mitochondrial damage and apoptosis play a crucial role in its neurotoxic effects. Mitochondrial unfolded protein response (UPRmt) is indispensable to maintain mitochondrial protein homeostasis and ensure mitochondrial function, and the ATF4/CHOP axis is one of the signaling pathways to activate UPRmt. In this study, the role of the ATF4/CHOP axis-mediated UPRmt in the neurotoxicity of MeHg has been investigated by C57BL/6 mice and the HT22 cell line. We discovered that mice exposed to MeHg had abnormal neurobehavioral patterns. The pathological section showed a significant decrease in the number of neurons. MeHg also resulted in a reduction in mtDNA copy number and mitochondrial membrane potential (MMP). Additionally, the ATF4/CHOP axis and UPRmt were found to be significantly activated. Subsequently, we used siRNA to knock down ATF4 or CHOP and observed that the expression of UPRmt-related proteins and the apoptosis rate were significantly reduced. Our research showed that exposure to MeHg can over-activate the UPRmt through the ATF4/CHOP axis, leading to mitochondrial damage and ultimately inducing neuronal apoptosis.


Subject(s)
Activating Transcription Factor 4 , Methylmercury Compounds , Neurons , Transcription Factor CHOP , Unfolded Protein Response , Animals , Mice , Apoptosis/genetics , Methylmercury Compounds/toxicity , Mice, Inbred C57BL , Unfolded Protein Response/genetics , Activating Transcription Factor 4/metabolism , Transcription Factor CHOP/metabolism , Mitochondria/metabolism , Neurons/metabolism
5.
Ecotoxicol Environ Saf ; 267: 115660, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37948942

ABSTRACT

Exposure to nitrogen dioxide might potentially change the makeup and operation of gut microbes. Nitrogen dioxide data was procured from the IEU Open GWAS (N = 456 380). Subsequently, a two-sample Mendelian randomization study was executed, utilizing summary statistics of gut microbiota sourced from the most expansive available genome-wide association study meta-analysis, conducted by the MiBioGen consortium (N = 13 266). The causal relationship between nitrogen dioxide and gut microbiota was determined using inverse variance weighted, maximum likelihood, MR-Egger, Weighted Median, Weighted Model, Mendelian randomization pleiotropy residual sum and outlier, and constrained maximum likelihood and model averaging and Bayesian information criterion. The level of heterogeneity of instrumental variables was quantified by utilizing Cochran's Q statistic. The colocalization analysis was used to examine whether nitrogen dioxide and the identified gut microbiota shared casual variants. Inverse variance weighted estimate suggested that nitrogen dioxide was causally associated with Akkermansia (ß = -1.088, 95% CI: -1.909 to -0.267, P = 0.009). In addition, nitrogen dioxide presented a potential association with Bacteroides (ß = -0.938, 95% CI: -1.592 to -0.284, P = 0.005), Barnesiella (ß = -0.797, 95% CI: -1.538 to -0.055, P = 0.035), Coprococcus 3 (ß = 1.108, 95% CI: 0.048-2.167, P = 0.040), Eubacterium hallii group (E. hallii) (ß = 0.776, 95% CI: 0.090-1.463, P = 0.027), Holdemania (ß = -1.354, 95% CI: -2.336 to -0.372, P = 0.007), Howardella (ß = 1.698, 95% CI: 0.257-3.139, P = 0.021), Olsenella (ß = 1.599, 95% CI: 0.151-3.048, P = 0.030) and Sellimonas (ß = -1.647, 95% CI: -3.209 to -0.086, P = 0.039). No significant heterogeneity of instrumental variables or horizontal pleiotropy was found. The associations of nitrogen dioxide with Akkermansia (PH4 = 0.836) and E. hallii (PH4 = 0.816) were supported by colocalization analysis. This two-sample Mendelian randomization study found that increased exposure to nitrogen dioxide had the potential to impact the human gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/genetics , Bayes Theorem , Genome-Wide Association Study , Nitrogen Dioxide , Random Allocation
6.
Front Oncol ; 13: 1274340, 2023.
Article in English | MEDLINE | ID: mdl-37901319

ABSTRACT

Introduction: Breast cancer is the most common malignancy among women. Previous studies had shown that hepatitis C virus (HCV) infection might serve as a risk factor for breast cancer, while some studies failed to find such an association. Methods: In this study, we presented a first attempt to capture and clarify this clinical debate via a cumulative analysis (registration ID: CRD42023445888). Results: After systematically searching and excluding the irrelevant publications, five case-control or cohort studies were finally included. The synthetic effect from the eligible studies showed that patients with HCV infection had a significantly higher prevalence of breast cancer than non-HCV infected general population (combined HR= 1.382, 95%CI: 1.129 to 1.692, P=0.002). There was no evidence of statistical heterogeneity during this pooled analysis (I2 = 13.2%, P=0.33). The sensitivity analyses confirmed the above findings. No significant publication bias was observed among the included studies. The underlying pathophysiological mechanisms for this relationship might be associated with persistent infection/inflammation, host immune response, and the modulation of HCV-associated gene expression. Discussion: Though the causal association between HCV infection and breast cancer did not seem quite as strong, screening for HCV might enable the early detection of breast cancer and help to prevent the progression of the disease. Since the topic of this study remains a matter of clinical debate, further studies are still warranted to validate this potential association. Systematic review registration: https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023445888.

7.
J Mol Med (Berl) ; 101(12): 1499-1512, 2023 12.
Article in English | MEDLINE | ID: mdl-37817014

ABSTRACT

The unfolded protein response (UPR) is a cellular stress response mechanism induced by the accumulation of unfolded or misfolded proteins. Within the endoplasmic reticulum and mitochondria, a dynamic balance exists between protein folding mechanisms and unfolded protein levels under normal conditions. Disruption of this balance or an accumulation of unfolded proteins in these organelles can result in stress responses and UPR. The UPR restores organelle homeostasis and promotes cell survival by increasing the expression of chaperone proteins, regulating protein quality control systems, and enhancing the protein degradation pathway. However, prolonged or abnormal UPR can also have negative effects, including cell death. Therefore, many diseases, especially neurodegenerative diseases, are associated with UPR dysfunction. Neurodegenerative diseases are characterized by misfolded proteins accumulating and aggregating, and neuronal cells are particularly sensitive to misfolded proteins and are prone to degeneration. Many studies have shown that the UPR plays an important role in the pathogenesis of neurodegenerative diseases. Here, we will discuss the possible contributions of the endoplasmic reticulum unfolded protein response (UPRer) and the mitochondrial unfolded protein response (UPRmt) in the development of several neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Endoplasmic Reticulum Stress , Unfolded Protein Response , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism
8.
J Photochem Photobiol B ; 245: 112753, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37437439

ABSTRACT

Due to continuous exposure to ultraviolet B(UVB) radiation, eye lenses are constantly subjected to oxidative stress that induces lens epithelial cell (LEC) apoptosis, which has been associated with the inactivation of Sirtuin1 (SIRT1). It is well-established that NFE2L2 has a major protective effect on UVB-induced oxidative stress and damage. However, whether UVB radiation affects oxidative/antioxidative imbalance and damages LECs by inactivating the protective NFE2L2-mediated antioxidative stress pathway through inhibition of SIRT1 is unknown. In our research, we established in vivo and in vitro UVB exposure models in Sprague Dawley rats and SRA01/04 cells, respectively, to investigate the effect of UVB radiation on the NFE2L2/ KEAP1 pathway and the role of SIRT1 in this process. The in vivo findings revealed that UVB radiation exposure decreased Sirt1 and Nfe2l2 levels, upregulated Keap1 expression, led to an oxidative/antioxidative imbalance and increased LEC apoptosis in the eye lens. Sirt1 downregulated Keap1 expression levels, but activated Nfe2l2 and its downstream target proteins. The in vitro findings showed that UVB inhibited the deacetylation of SIRT1 target proteins and increased the acetylation levels of KEAP1 and NFE2L2. We also found that UVB radiation exposure led to a significant decrease in both co-localization levels and protein interaction between SIRT1 and KEAP1. In addition, the inhibition of SIRT1 increased KEAP1 levels, inhibited the activity of NFE2L2 and decreased co- localization levels and protein interactions between NFE2L2 and KEAP1. These results suggested that UVB radiation decreased SIRT1 levels and inhibited the KEAP1/NFE2L2 pathway, thereby reducing its antioxidant effect, which might be an important mechanism of UVB-induced cataract.

9.
Cell Mol Neurobiol ; 43(6): 2627-2642, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36988772

ABSTRACT

Programed cell death plays a key role in promoting human development and maintaining homeostasis. Ferroptosis is a recently identified pattern of programmed cell death that is closely associated with the onset and progression of neurodegenerative diseases. Ferroptosis is mainly caused by the intracellular accumulation of iron-dependent lipid peroxides. The cysteine/glutamate antibody Solute carrier family 7 member 11 (SLC7A11, also known as xCT) functions to import cysteine for glutathione biosynthesis and antioxidant defense. SLC7A11 has a significant impact on ferroptosis, and inhibition of SLC7A11 expression promotes ferroptosis. Moreover, SLC7A11 is also closely associated with neurodegenerative diseases. In this paper, we summarize the relationship between ferroptosis and neurodegenerative diseases and the role of SLC7A11 during this process. The various regulatory mechanisms of SLC7A11 are also discussed. In conclusion, we are looking forward to a theoretical basis for further understanding the occurrence and development of ferroptosis in SLC7A11 and neurodegenerative diseases, and to seek new clues for the treatment of neurodegenerative diseases.


Subject(s)
Ferroptosis , Neurodegenerative Diseases , Humans , Cysteine , Apoptosis , Iron/metabolism , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism
10.
Neurotoxicol Teratol ; 95: 107137, 2023.
Article in English | MEDLINE | ID: mdl-36403891

ABSTRACT

Methylmercury (MeHg) is a potent neurotoxicant that could induce oxidative stress and autophagy. However, the underlying mechanisms through which MeHg affects the central nervous system have not been fully elucidated, and little has been known of the interaction between oxidative stress and autophagy. Therefore, rats were administrated with different MeHg concentrations to evaluate the neurotoxic effects and autophagy in cerebral cortex. Moreover, we have investigated the neuroprotective role of N-acetyl-L-cysteine (NAC) against MeHg-induced neurotoxicity in order to estimate the regulation effects of oxidative stress on autophagy. A total of 64 rats, 40 of which were randomly divided into control and MeHg-treated (4, 8 and 12 µ mol/kg) groups. The remaining 24 rats were divided into control, NAC control (1 mmol/kg), 12 µ mol/kg MeHg, and NAC pretreatment. Administration of 12 µ mol/kg MeHg significantly increased behavioral and pathological abnormalities, and autophagy levels. In addition, the oxidative stress levels increased, together with abnormal expression of autophagy-related molecules. Pretreatment with NAC significantly prevented MeHg-induced oxidative stress and PI3K/AKT/mTOR or AMPK/TSC2/mTOR-mediated autophagy. In conclusion, the present study suggested that oxidative stress can regulate autophagy through PI3K/AKT/mTOR or AMPK/TSC2/mTOR pathways. This study provides a theoretical basis for the study and treatment of MeHg-induced neurotoxicity.


Subject(s)
Acetylcysteine , Methylmercury Compounds , Animals , Rats , Acetylcysteine/pharmacology , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Autophagy , Cerebral Cortex , Methylmercury Compounds/toxicity , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases
11.
Neuroscience ; 504: 79-92, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36202276

ABSTRACT

Sirtuin 1 (SIRT1) is an NAD+ dependent deacetylase that modify the gene expression through histone deacetylation. SIRT1 plays a crucial role in regulating a wide range of physiological processes by adjustment multiple mechanisms through the deacetylation of multiple substrates. Neurodegenerative diseases are a series of chronic diseases characterized by dysfunction and loss of neurons. Its basic pathogenesis is filamentous tangles and amyloid deposits, such as Amyloid-ß (Aß), tau protein, α-synuclein, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). This summarizes introduces the structure and function of SIRT1, and then analyzes the protective effects of SIRT1 on neurological diseases by regulating circadian rhythm, aging, oxidative stress, mitochondrial dysfunction and neuroinflammation related pathways.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Humans , Neurodegenerative Diseases/metabolism , Sirtuin 1/metabolism , Oxidative Stress/physiology , Neurons/metabolism , Parkinson Disease/metabolism
12.
Ecotoxicol Environ Saf ; 245: 114099, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36152427

ABSTRACT

Methylmercury (MeHg) is a ubiquitous environmental pollutant, which can cross the placenta and blood brain barrier, thus affecting fetal growth and development. Although previous studies have demonstrated that MeHg induces endoplasmic reticulum (ER) stress in rat cerebral cortex and primary neurons, the role of ER stress in MeHg-induced neurodevelopmental toxicity remains unclear. Here, we used ICR pregnant mice and hippocampal neurons cells (HT22 cells) to investigate the molecular mechanism by which MeHg exposure during pregnancy affects neurodevelopment. We found that prenatal MeHg exposure caused developmental delay in offspring, accompanied with ER stress, cell apoptosis, cell cycle arrest and abnormal DNA methylation. Then, we used ER stress specific inhibitor 4-PBA and CHOP siRNA to investigate the role of ER stress on HT22 cells damage caused by MeHg. The results showed that 4-PBA pretreatment restored MeHg-induced axonal shortening and alleviated apoptosis, cell cycle arrest and DNA methylation. At the same time, the activation of CHOP/c-Jun/GADD45A signaling pathway was inhibited, and the interaction between CHOP and c-Jun was weakened. In addition, CHOP siRNA reduced the expression of c-Jun and GADD45A, and relieved DNA methylation levels to some extent. In summary, our study suggested that ER stress induced by MeHg mediated cell apoptosis and cell cycle arrest, and may affect DNA methylation through activation of CHOP/c-Jun/GADD45A signaling pathway, thus leading to neuronal damage.


Subject(s)
Environmental Pollutants , Methylmercury Compounds , Animals , Apoptosis/physiology , Butylamines , Endoplasmic Reticulum Stress , Environmental Pollutants/metabolism , Environmental Pollutants/toxicity , Methylmercury Compounds/metabolism , Methylmercury Compounds/toxicity , Mice , Mice, Inbred ICR , Neurons/metabolism , RNA, Small Interfering/metabolism , Rats
13.
J Biochem Mol Toxicol ; 36(9): e23136, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35678294

ABSTRACT

Methylmercury (MeHg) is a cumulative environmental pollutant that can easily cross the blood-brain barrier and cause damage to the brain, mainly targeting the central nervous system. The purpose of this study is to investigate the role of calcium ion (Ca2+ ) homeostasis between the endoplasmic reticulum (ER) and mitochondria in MeHg-induced neurotoxicity. Rat primary cortical neurons exposed to MeHg (0.25-1 µm) underwent dose-dependent cell damage, accompanied by increased Ca2+ release from the ER and elevated levels of free Ca2+ in cytoplasm and mitochondria. MeHg also increased the protein and messenger RNA expressions of the inositol 1,4,5-triphosphate receptor, ryanodine receptor 2, and mitochondrial calcium uniporter. Ca2+ channel inhibitors 2-aminoethyl diphenylborinate and procaine reduced the release of Ca2+ from ER, while RR and 4,4'-diisothiocyanatostilbene-2,2'-disulfonate inhibited Ca2+ uptake from mitochondria. In addition, pretreatment with Ca2+ chelator BAPTA-AM effectively restored mitochondrial membrane potential levels, inhibited over opening of mitochondrial permeability transition pore, and maintained mitochondrial function stability. Meanwhile, the expression of mitochondrial apoptosis-related proteins recovered to some extent, along with the reduction of the early apoptosis ratio. These results suggest that Ca2+ homeostasis plays an essential role in mitochondrial damage and apoptosis induced by MeHg, which may be one of the important mechanisms of MeHg-induced neurotoxicity.


Subject(s)
Environmental Pollutants , Methylmercury Compounds , Animals , Apoptosis , Calcium/metabolism , Chelating Agents , Endoplasmic Reticulum , Environmental Pollutants/pharmacology , Homeostasis , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate Receptors/pharmacology , Methylmercury Compounds/metabolism , Methylmercury Compounds/toxicity , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore , Neurons/metabolism , Procaine/metabolism , Procaine/pharmacology , RNA, Messenger/metabolism , Rats , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/pharmacology
14.
Toxicol Lett ; 359: 55-64, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35122893

ABSTRACT

Methylmercury (MeHg) is a bio-accumulative global environmental contaminant present in fish and seafood. MeHg accumulates in the aquatic environment and eventually reaches the human system via the food chain by bio-magnification. The central nervous system is the primary target of toxicity and is particularly vulnerable during development. It is well documented that developmental MeHg exposure can lead to neurological alterations, including cognitive and motor dysfunction. Apoptosis is a primary characteristic of MeHg-induced neurotoxicity, and may be regulated by autophagic activity. However, mechanisms mediating the interaction between apoptosis and autophagy remains to be explored. Autophagy is an adaptive response under stressful conditions, and the basal level of autophagy ensures the physiological turnover of old and damaged organelles. Autophagy can regulate cell fate through different crosstalk signaling pathways. A complex interplay between autophagy and apoptosis determines the degree of apoptosis and the progression of MeHg-induced neurotoxicity as demonstrated by pre-clinical models and clinical trials. This review summarizes recent advances in the roles of autophagy and apoptosis in MeHg neurotoxicity and thoroughly explores the relationship between them. The autophagic pathway may be a potential therapeutic target in MeHg neurotoxicity through modulation of apoptosis.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Methylmercury Compounds/metabolism , Methylmercury Compounds/toxicity , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/physiopathology , Animals , Disease Models, Animal , Humans , Metabolic Networks and Pathways
15.
Chem Res Toxicol ; 35(1): 43-58, 2022 01 17.
Article in English | MEDLINE | ID: mdl-34989572

ABSTRACT

Mercury (Hg) is a toxic heavy-metal element, which can be enriched in fauna and flora and transformed into methylmercury (MeHg). MeHg is a widely distributed environmental pollutant that may be harmful to fish-eating populations through enrichment of aquatic food chains. The central nervous system is a primary target of MeHg. Embryos and infants are more sensitive to MeHg, and exposure to MeHg during gestational feeding can significantly impair the homeostasis of offspring, leading to long-term neurodevelopmental defects. At present, MeHg-induced neurodevelopmental toxicity has become a hotspot in the field of neurotoxicology, but its mechanisms are not fully understood. Some evidence point to oxidative damage, excitotoxicity, calcium ion imbalance, mitochondrial dysfunction, epigenetic changes, and other molecular mechanisms that play important roles in MeHg-induced neurodevelopmental toxicity. In this review, advances in the study of neurodevelopmental toxicity of MeHg exposure during pregnancy and the molecular mechanisms of related pathways are summarized, in order to provide more scientific basis for the study of neurodevelopmental toxicity of MeHg.


Subject(s)
Environmental Pollutants/adverse effects , Methylmercury Compounds/adverse effects , Neurodevelopmental Disorders/chemically induced , Animals , Humans
16.
Food Chem Toxicol ; 155: 112386, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34242720

ABSTRACT

Methylmercury (MeHg) is a environmental contaminant, which can induce neurotoxic effects. So far, the exact molecular mechanisms of autophagy and its effect on apoptosis in MeHg-induced neurotoxicity have not been elucidated. Here, rats were exposed to MeHg (4, 8, or 12 µmol/kg) for 4 weeks to evaluate the dose-effect relationship between MeHg and apoptosis, or autophagy in cerebral cortex. On this basis, rapamycin (Rapa) or 3-methyladenine (3-MA) was administrated to further explore the regulatory mechanisms of autophagy on MeHg-induced neuronal apoptosis. The pathological changes, autophagy or apoptosis levels, expression of autophagic or apoptotic-associated factors such as mTOR, S6K1, 4EBP1, Vps34, Beclin1, p62, LC3, Bcl-2/Bax, caspase, or MAPKs were investigated. Results showed that MeHg dose-dependently induced pathological changes in cerebral cortex, and the levels of autophagy and apoptosis were increased. Furthermore, Rapa pretreatment antagonized MeHg-induced apoptosis, whereas 3-MA further aggravated apoptosis, which were supported by findings that Rapa activated mTOR-mediated autophagy while 3-MA inhibited Vps34-related autophagy, further affect neuronal apoptosis through regulation of apoptotic factors mentioned above. In conclusion, the findings indicated that MeHg dose-dependently induced autophagy or apoptosis, and mTOR or Vps34 may play important roles in mediating autophagy, which further regulated apoptosis through MAPKs or mitochondrial apoptosis pathways.


Subject(s)
Apoptosis/drug effects , Autophagy/physiology , Cerebral Cortex/drug effects , Class III Phosphatidylinositol 3-Kinases/metabolism , Methylmercury Compounds/toxicity , TOR Serine-Threonine Kinases/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Cerebral Cortex/pathology , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Female , Male , Neurons/drug effects , Rats, Wistar , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
17.
Neuroscience ; 469: 175-190, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34174372

ABSTRACT

Methylmercury (MeHg) is a potential neurotoxin that is highly toxic to the human central nervous system. Although MeHg neurotoxicity has been widely studied, the mechanism of MeHg neurotoxicity has not yet been fully elucidated. Some research evidence suggests that oxidative stress and autophagy are important molecular mechanisms of MeHg-induced neurotoxicity. Researchers have widely accepted that oxidative stress regulates the autophagy pathway. The current study reviews the activation of Nuclear factor-erythroid-2-related factor (Nrf2)-related oxidative stress pathways and autophagy signaling pathways in the case of MeHg neurotoxicity. In addition, autophagy mainly plays a role in the neurotoxicity of MeHg through mTOR-dependent and mTOR-independent autophagy signaling pathways. Finally, the regulation of autophagy by reactive oxygen species (ROS) and Nrf2 in MeHg neurotoxicity was explored in this review, providing a new concept for the study of the neurotoxicity mechanism of MeHg.


Subject(s)
Methylmercury Compounds , Neurotoxicity Syndromes , Animals , Autophagy , Humans , Methylmercury Compounds/toxicity , Neurotoxicity Syndromes/etiology , Oxidative Stress , Rats , Rats, Wistar , Reactive Oxygen Species
18.
Neurotoxicology ; 85: 33-46, 2021 07.
Article in English | MEDLINE | ID: mdl-33964343

ABSTRACT

Methylmercury (MeHg) is a long-lasting organic environmental pollutant that poses a great threat to human health. Ingestion of seafood containing MeHg is the most important way by which it comes into contact with human body, where the central nervous system (CNS) is the primary target of MeHg toxicity. During periods of pre-plus postnatal, in particular, the brain of offspring is vulnerable to specific developmental insults that result in abnormal neurobehavioral development, even without symptoms in mothers. While many studies on neurotoxic effects of MeHg on the developing brain have been conducted, the mechanisms of oxidative stress in MeHg-induced neurodevelopmental toxicity is less clear. Hitherto, no single process can explain the many effects observed in MeHg-induced neurodevelopmental toxicity. This review summarizes the possible mechanisms of oxidative stress in MeHg-induced neurodevelopmental toxicity, highlighting modulation of Nrf2/Keap1/Notch1, PI3K/AKT, and PKC/MAPK molecular pathways as well as some preventive drugs, and thus contributes to the discovery of endogenous and exogenous molecules that can counteract MeHg-induced neurodevelopmental toxicity.


Subject(s)
Brain/drug effects , Methylmercury Compounds/toxicity , Neurodevelopmental Disorders/chemically induced , Oxidative Stress/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Environmental Pollutants/metabolism , Environmental Pollutants/toxicity , Female , Humans , Methylmercury Compounds/metabolism , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Oxidative Stress/physiology , Placenta/drug effects , Placenta/metabolism , Placenta/pathology , Pregnancy
19.
Environ Int ; 155: 106593, 2021 10.
Article in English | MEDLINE | ID: mdl-33962234

ABSTRACT

BACKGROUNDS: While the pernicious effects of outdoor air pollution on cognitive ability have been previously examined, evidence regarding household air pollution is scarce. METHODS: Using data from the Chinese Longitudinal Healthy Longevity Survey, we explored the relationship between cooking with biomass fuel and cognitive impairment and cognitive decline using a Cox proportional hazards model. We further assessed the correlation of biomass fuels and cognitive score using a generalized estimating equation. Cognitive ability was measured based on the Chinese version of the Mini-Mental State Examination (MMSE) and cognitive impairment was defined as MMSE < 24 points and cognitive decline was defined as a reduction of MMSE ≥ 3 points. On follow-up, we investigated the effect of switch-cooking combustibles on cognitive ability. RESULTS: The mean (SD) age of 4161 participants was 81.7 (10.0) years old. The reported cooking with biomass fuels was correlated with an elevated risk of cognitive impairment (hazard ratio (HR): 1.19, 95% confidence interval (CI): 1.04, 1.37) and cognitive decline (HR: 1.18, 95% CI: 1.04, 1.33). Besides, cooking with biomass fuels was related to a decrease in cognitive score (ß: -0.43, 95% CI: -0.73, -0.14). In comparison to persistent biomass fuel users, participants who reported changing their primary cooking fuels from biomass to clean fuels exhibited a reduced risk of cognitive impairment (HR: 0.68, 95% CI: 0.57, 0.82) and cognitive decline (HR: 0.66, 95% CI: 0.56, 0.76) and a higher cognitive score (ß: 0.72, 95% CI: 0.17, 1.26). Cooking without ventilated cookstoves was associated with a higher risk of cognitive impairment (HR: 1.31, 95% CI: 1.10, 1.58) and cognitive decline (HR: 1.18, 95% CI: 1.02, 1.38), regardless of types of cooking fuels. Interaction and stratified analyses showed relatively poor cognitive ability in participants who engaged in irregular exercise or were not living with family members. CONCLUSIONS: Cooking with biomass fuels was correlated with a higher risk of cognitive impairment and cognitive decline. Among the oldest-old population, this risk may, however, be lower for those changing their primary cooking fuels from biomass to clean fuels.


Subject(s)
Air Pollution, Indoor , Cognitive Dysfunction , Adult , Aged, 80 and over , Air Pollution, Indoor/adverse effects , Biomass , China/epidemiology , Cognitive Dysfunction/epidemiology , Cognitive Dysfunction/etiology , Cooking , Humans , Prospective Studies
20.
Sci Total Environ ; 772: 145395, 2021 Jun 10.
Article in English | MEDLINE | ID: mdl-33578144

ABSTRACT

BACKGROUND: Many households in developing countries, including China, rely on the traditional use of solid fuels for cooking and heating. Arthritis is highly prevalent in middle-aged and older adults and is a major cause of disability. However, evidence linking indoor solid fuel use with arthritis is scarce in this age group (≥45 years) in developing countries. OBJECTIVES: To investigate whether exposure to indoor solid fuel for cooking and heating is associated with arthritis in middle-aged and older adults in rural China. METHODS: Data for the present study were extracted from the China Health and Retirement Longitudinal Study (CHARLS), a longitudinal national prospective study of adults aged 45 years and older enrolled in 2010 and followed up through 2015. We included 7807 rural participants without arthritis at baseline, of whom 1548 living in a central heating area in winter were included in the heating analysis (taking the Qinling-Huaihe line as the heating boundary). Cox proportional hazards models were used to examine the association between indoor solid fuel use and arthritis, controlling for age, sex, education, marital status, smoking status, drinking status, self-reported socioeconomic status, BMI, sleep time, napping time, independent cooking, hypertension, diabetes, dyslipidemia, heart problems and stroke. We also investigated the effect of switching primary fuels and using solid fuels for both cooking and heating on arthritis risk. RESULTS: The mean (SD) age of the study participants was 59.2 (10.0) years old, and 48.0% of participants were women. A total of 64.8% and 63.0% of the participants reported primarily using solid fuel for cooking and heating, respectively. Arthritis incidence rates were lower among clean fuel users than solid fuel users. Compared to those using clean fuels, cooking and heating solid fuel users had a higher risk of arthritis, with hazard ratios (HRs) of 1.22 (95% confidence interval (CI): 1.01, 1.49) and 1.76 (95% CI: 1.07, 2.89), respectively. Switching from clean fuels to solid fuels for heating (HR: 3.28, 95% CI: 1.21, 7.91) and using solid fuels for both cooking and heating (HR, 1.71, 95% CI, 1.01-2.79) increased the risk of arthritis. CONCLUSIONS: Long-term solid fuel use for indoor cooking and heating is associated with an increased risk of arthritis events among adults aged 45 years and older in rural China. The potential benefits of reducing indoor solid fuel use in groups at high risk for arthritis merit further exploration.


Subject(s)
Air Pollution, Indoor , Arthritis , Aged , Arthritis/epidemiology , China/epidemiology , Coal , Cohort Studies , Cooking , Female , Humans , Longitudinal Studies , Middle Aged , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...