Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
J Hazard Mater ; 472: 134559, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38735189

ABSTRACT

Parkinson's disease (PD) is a prevalent neurodegenerative disease and approximately one third of patients with PD are estimated to experience depression. Paraquat (PQ) is the most widely used herbicide worldwide and PQ exposure is reported to induce PD with depression. However, the specific brain region and neural networks underlying the etiology of depression in PD, especially in the PQ-induced model, have not yet been elucidated. Here, we report that the VGluT2-positive glutamatergic neurons in the paraventricular thalamic nucleus (PVT) promote depression in the PQ-induced PD mouse model. Our results show that PVTVGluT2 neurons are activated by PQ and their activation increases the susceptibility to depression in PD mice. Conversely, inhibition of PVTVGluT2 neurons reversed the depressive-behavioral changes induced by PQ. Similar to the effects of intervention the soma of PVTVGluT2 neurons, stimulation of their projections into the central amygdaloid nucleus (CeA) also strongly influenced depression in PD mice. PQ induced malfunctioning of the glutamate system and changes in the dendritic and synaptic morphology in the CeA through its role on PVTVGluT2 neuronal activation. In summary, our results demonstrate that PVTVGluT2 neurons are key neuronal subtypes for depression in PQ-induced PD and promote depression processes through the PVTVGluT2-CeA pathway.


Subject(s)
Midline Thalamic Nuclei , Neurons , Paraquat , Vesicular Glutamate Transport Protein 2 , Animals , Paraquat/toxicity , Male , Vesicular Glutamate Transport Protein 2/metabolism , Neurons/drug effects , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/metabolism , Depression/chemically induced , Depression/metabolism , Mice, Inbred C57BL , Herbicides/toxicity , Mice , Parkinson Disease/metabolism
2.
Free Radic Biol Med ; 220: 271-287, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38734267

ABSTRACT

Bilirubin-induced brain damage is a serious clinical consequence of hyperbilirubinemia, yet the underlying molecular mechanisms remain largely unknown. Ferroptosis, an iron-dependent cell death, is characterized by iron overload and lipid peroxidation. Here, we report a novel regulatory mechanism of demethylase AlkB homolog 5 (ALKBH5) in acyl-coenzyme A synthetase long-chain family member 4 (ACSL4)-mediated ferroptosis in hyperbilirubinemia. Hyperdifferential PC12 cells and newborn Sprague-Dawley rats were used to establish in vitro and in vivo hyperbilirubinemia models, respectively. Proteomics, coupled with bioinformatics analysis, first suggested the important role of ferroptosis in hyperbilirubinemia-induced brain damage. In vitro experiments showed that ferroptosis is activated in hyperbilirubinemia, and ferroptosis inhibitors (desferrioxamine and ferrostatin-1) treatment effectively alleviates hyperbilirubinemia-induced oxidative damage. Notably, we observed that the ferroptosis in hyperbilirubinemia was regulated by m6A modification through the downregulation of ALKBH5 expression. MeRIP-seq and RIP-seq showed that ALKBH5 may trigger hyperbilirubinemia ferroptosis by stabilizing ACSL4 mRNA via m6A modification. Further, hyperbilirubinemia-induced oxidative damage was alleviated through ACSL4 genetic knockdown or rosiglitazone-mediated chemical repression but was exacerbated by ACSL4 overexpression. Mechanistically, ALKBH5 promotes ACSL4 mRNA stability and ferroptosis by combining the 669 and 2015 m6A modified sites within 3' UTR of ACSL4 mRNA. Our findings unveil a novel molecular mechanism of ferroptosis and suggest that m6A-dependent ferroptosis could be an underlying clinical target for the therapy of hyperbilirubinemia.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Coenzyme A Ligases , Ferroptosis , RNA Stability , Rats, Sprague-Dawley , Animals , Ferroptosis/genetics , Rats , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , AlkB Homolog 5, RNA Demethylase/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , PC12 Cells , Cyclohexylamines/pharmacology , Humans , Deferoxamine/pharmacology , Oxidative Stress , Brain Injuries/metabolism , Brain Injuries/genetics , Brain Injuries/pathology , Brain Injuries/etiology , Phenylenediamines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Male , Disease Models, Animal , Lipid Peroxidation
3.
Environ Pollut ; 352: 124035, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38670424

ABSTRACT

The exact mechanisms underlying the initiation and exacerbation of Parkinson's disease (PD) by paraquat remain unclear. We have revealed that exosomes mediate neurotoxicity induced by low dose paraquat exposure by transmitting intercellular signaling. Exposure to 40 µM paraquat promoted exosome release from mouse microglia cells (BV2) in vitro. Paraquat exposure at 100 µM caused degeneration of mouse dopaminergic MN9D cells and inhibited microglia exosome uptake by fluorescently labeling exosomes. We established an incubation model for exosomes and dopaminergic neuron cells under PQ treatment. The results indicated that microglial exosomes alleviated degeneration, increasing proliferation and PD-related protein expression of dopaminergic neurons; however, paraquat reversed this effect. Then, through exosome high-throughput sequencing and qRT-PCR experiments, miR-92a-3p and miR-24-3p were observed to transfer from exosomes to dopaminergic neurons, inhibited by paraquat. The specificity of miR-92a-3p and miR-24-3p was verified in PD patients exosomes, indicating the potential diagnostic value of the exosomal miRNAs in paraquat-induced PD. These results suggest glia-neuron communication in paraquat-induced neurodegeneration and may identify stable paraquat-mediated PD biomarkers, offering clues for early recognition and prevention of pesticide-induced degenerative diseases.


Subject(s)
Biomarkers , Dopaminergic Neurons , Exosomes , MicroRNAs , Microglia , Paraquat , Parkinson Disease , Paraquat/toxicity , Exosomes/metabolism , Animals , Microglia/drug effects , Microglia/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Dopaminergic Neurons/drug effects , Biomarkers/metabolism , Neuroprotection/drug effects , Humans , Cell Line
4.
Ecotoxicol Environ Saf ; 273: 116169, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38447518

ABSTRACT

Parkinson's disease (PD) is among the most prevalent neurodegenerative diseases, and approximately one third of patients with PD are estimated to have depression. Paraquat (PQ) exposure is an important environmental risk factor for PD. In this study, we established a mouse model of PQ-induced PD with depression to comprehensively investigate cellular heterogeneity and the mechanisms underlying the progression of depression in the context of PD. We utilized single-cell RNA-seq (scRNA-seq) to acquire the transcriptomic atlas of individual cells from model mice and characterize the gene expression profiles in each differentially expressed cell type. We identified a specific glutamatergic neuron cluster responsible for the development of heterogeneous depression-associated changes and established a comprehensive gene expression atlas. Furthermore, functional enrichment and cell trajectory analyses revealed that the mechanisms underlying the progression of PD with depression were associated with specific glutamatergic neurons. Together, our findings provide a valuable resource for deciphering the cellular heterogeneity of PD with depression. The suggested connection between intrinsic transcriptional states of neurons and the progression of depression can provide insight into potential biomarkers and specific targets for anti-depression treatment in patients with PD. SYNOPSIS: Our results obtained using model mice confirm the core effects of PQ exposure on glutamatergic neurons and their potential role in the development of PD with depression.


Subject(s)
Paraquat , Parkinson Disease , Humans , Animals , Mice , Paraquat/toxicity , Parkinson Disease/genetics , Depression/chemically induced , Depression/genetics , Gene Expression Profiling , RNA
5.
Environ Pollut ; 349: 123875, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38548152

ABSTRACT

With the evidence emerging that abnormal expression of long noncoding RNAs (lncRNAs) are involved in onset of Parkinson's disease (PD), the role of NR_030777 contributing to this disease is of great interest. We recently found that a novel lncRNA "NR_030777" demonstrates protective effects on PQ-induced neurodegeneration. However, the underlying molecular mechanisms of NR_030777 in the regulation of mitochondrial fission and mitophagy involved in PQ-induced neuronal damage remain to be explored. NR_030777 brain conditional overexpressing mice as well as in vitro primary neuronal cells from cerebral cortex and Neuro2a cells were adopted. Immunofluorescence, Immunohistochemistry, qRT-PCR and Western blotting were used to evaluate the expression levels of RNA and proteins. RNA immunoprecipitation and RNA pulldown experiment were used to evaluate the interaction of NR_030777 with its target proteins. NR_030777 and mitophagy were increased, and tyrosine hydroxylase (TH) levels recovered after NR_030777 overexpression upon PQ treatment. The overexpression and knockdown of NR_030777 unveiled that NR_030777 positively regulated mitophagy such as the upregulation of LC3B-II:I, ATG12-ATG5, p62 and NBR1. Moreover, the application of mdivi-1, a DRP-1 inhibitor, in combination with NR_030777 genetic modified cells unveiled that NR_030777 promoted DRP1-mediated mitochondrial fission and mitophagy. Furthermore, NR_030777 were directly bound to CDK1 to increase p-DRP1 levels at the Ser616 site, leading to mitochondrial fission and mitophagy. On the other hand, NR_030777 acted directly on ATG12 within the ATG12-ATG5 complex in the 800-1400 nt region to modulate the membrane formation. Accordingly, NR_030777 deficiency in neuron cells compromised cell mitophagy. Finally, the above findings were confirmed using NR_030777-overexpressing mice. NR_030777 exerted a protective effect on PQ-exposed mice by enhancing mitophagy. Our data provide the first scientific evidence for the precise invention of PQ-induced PD. Our findings further propose a breakthrough for understanding the regulatory relationship between NR_030777, CDK1, ATG12 and mitophagy in PQ-induced PD.


Subject(s)
CDC2 Protein Kinase , Mitochondrial Dynamics , Mitophagy , Parkinson Disease , RNA, Long Noncoding , Animals , Mice , CDC2 Protein Kinase/metabolism , CDC2 Protein Kinase/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Mitophagy/drug effects , Neurons/metabolism , Neurons/drug effects , Paraquat/toxicity , Parkinson Disease/metabolism , Parkinson Disease/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
6.
Part Fibre Toxicol ; 21(1): 1, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38225661

ABSTRACT

BACKGROUND: As the demand and application of engineered nanomaterials have increased, their potential toxicity to the central nervous system has drawn increasing attention. Tunneling nanotubes (TNTs) are novel cell-cell communication that plays a crucial role in pathology and physiology. However, the relationship between TNTs and nanomaterials neurotoxicity remains unclear. Here, three types of commonly used engineered nanomaterials, namely cobalt nanoparticles (CoNPs), titanium dioxide nanoparticles (TiO2NPs), and multi-walled carbon nanotubes (MWCNTs), were selected to address this limitation. RESULTS: After the complete characterization of the nanomaterials, the induction of TNTs formation with all of the nanomaterials was observed using high-content screening system and confocal microscopy in both primary astrocytes and U251 cells. It was further revealed that TNT formation protected against nanomaterial-induced neurotoxicity due to cell apoptosis and disrupted ATP production. We then determined the mechanism underlying the protective role of TNTs. Since oxidative stress is a common mechanism in nanotoxicity, we first observed a significant increase in total and mitochondrial reactive oxygen species (namely ROS, mtROS), causing mitochondrial damage. Moreover, pretreatment of U251 cells with either the ROS scavenger N-acetylcysteine or the mtROS scavenger mitoquinone attenuated nanomaterial-induced neurotoxicity and TNTs generation, suggesting a central role of ROS in nanomaterials-induced TNTs formation. Furthermore, a vigorous downstream pathway of ROS, the PI3K/AKT/mTOR pathway, was found to be actively involved in nanomaterials-promoted TNTs development, which was abolished by LY294002, Perifosine and Rapamycin, inhibitors of PI3K, AKT, and mTOR, respectively. Finally, western blot analysis demonstrated that ROS and mtROS scavengers suppressed the PI3K/AKT/mTOR pathway, which abrogated TNTs formation. CONCLUSION: Despite their biophysical properties, various types of nanomaterials promote TNTs formation and mitochondrial transfer, preventing cell apoptosis and disrupting ATP production induced by nanomaterials. ROS/mtROS and the activation of the downstream PI3K/AKT/mTOR pathway are common mechanisms to regulate TNTs formation and mitochondrial transfer. Our study reveals that engineered nanomaterials share the same molecular mechanism of TNTs formation and intercellular mitochondrial transfer, and the proposed adverse outcome pathway contributes to a better understanding of the intercellular protection mechanism against nanomaterials-induced neurotoxicity.


Subject(s)
Cell Membrane Structures , Nanotubes, Carbon , Nanotubes , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Nanotubes, Carbon/toxicity , TOR Serine-Threonine Kinases/metabolism , Neuroglia/metabolism , Adenosine Triphosphate , Apoptosis
7.
Front Plant Sci ; 14: 1274943, 2023.
Article in English | MEDLINE | ID: mdl-38034557

ABSTRACT

Excessive nitrogen (N) application in wheat-maize cropping systems was adjusted towards more sustainable practices to reduce hydrological N losses while maintaining crop yield. In comprehensive quantification of N management effects on crop yield, N use efficiency (NUE), hydrological N losses, and soil nitrate residual across eight seasons, we have added to growing evidence of strategies beneficial for sustainable crop production with lower hydrological N losses. The results show that adjusted N practices enhanced crop yield and NUE, as compared to farmer's practices, but benefits varied with N rates and types. Optimized N treatment (OPT, 180 kg N ha-1 in both maize and wheat seasons) with or without straw returning produced the most crop yield. They increased maize yield by 5.5% and 7.3% and wheat yield by 6.2% and 3.2% on average, as compared to farmer's practice with huge N application (FP, 345 kg N ha-1 and 240 kg N ha-1 in maize and wheat). Regulation of N release through amendment with controlled release urea at a rate of 144 kg N ha-1 crop-1 (CRU treatment) obtained 4.4% greater maize yield than FP, and sustained a similar wheat yield with less N input, resulting in the highest crop NUE. Additionally, CRU was most effective in mitigating hydrological N loss, with 39.5% and 45.5% less leachate N and 31.9% and 35.9% less runoff N loss than FP in maize and wheat seasons. Synthetic N input correlated significantly and positively with runoff and leachate N losses, indicating it was one of the dominant factors driving hydrological N losses. Moreover, compared to OPT, additional straw returning (STR) or substituting 20% of the nutrients by duck manure (DMS) further reduced runoff N discharges due to the fact that organic matter incorporation increased resilience to rainfall. N over-application in FP caused considerable nitrate accumulation in the 0-90-cm soil profile, while the adjusted N practices, i.e., OPT, STR, CRU, and DMS treatments effectively controlled it to a range of 79.6-92.9 kg N ha-1. This study suggests that efforts using optimized N treatment integrated with CRU or straw returning should be encouraged for sustainable crop production in this region.

8.
Ecotoxicol Environ Saf ; 263: 115356, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37591128

ABSTRACT

Paraquat (PQ) is an environmental poison that causes clinical symptoms similar to those of Parkinson's disease (PD) in vitro and in rodents. It can lead to the activation of microglia and apoptosis of dopaminergic neurons. However, the exact role and mechanism of microglial activation in PQ-induced neuronal degeneration remain unknown. Here, we isolated the microglia-derived exosomes exposed with 0 and 40 µM PQ, which were subsequently co-incubated with PQ-exposed neuronal cells to simulate intercellular communication. First, we found that exosomes released from microglia caused a change in neuronal cell vitality and reversed PQ-induced neuronal apoptosis. RNA sequencing data showed that these activated microglia-derived exosomes carried large amounts of circZNRF1. Moreover, a bioinformatics method was used to study the underlying mechanism of circZNRF1 in regulating PD, and miR-17-5p was predicted to be its target. Second, an increased Bcl2/Bax ratio could play an anti-apoptotic role. Bcl2 was predicted to be a downstream target of miR-17-5p. Our results showed that circZNRF1 plays an anti-apoptotic role by absorbing miR-17-5p and regulating the binding of Bcl2 after exosomes are internalized by dopaminergic neurons. In conclusion, we demonstrated a new intercellular communication mechanism between microglia and neurons, in which circZNRF1 plays a key role in protecting against PQ-induced neuronal apoptosis through miR-17-5p to regulate the biological process of PD. These findings may offer a novel approach to preventing and treating PD.


Subject(s)
MicroRNAs , Microglia , Paraquat/toxicity , Dopaminergic Neurons , Proto-Oncogene Proteins c-bcl-2 , MicroRNAs/genetics
9.
Toxicol Sci ; 196(1): 85-98, 2023 10 30.
Article in English | MEDLINE | ID: mdl-37584706

ABSTRACT

The widespread use of nanomaterials in daily life has led to increased concern about their potential neurotoxicity. Therefore, it is particularly important to establish a simple and reproducible assessment system. Representative nanomaterials, including cobalt nanoparticles (CoNPs), titanium dioxide nanoparticles (TiO2-NPs), and multiwall carbon nanotubes (MWCNTs), were compared in terms of their neurotoxicity and underlying mechanisms. In 0, 25, 50, and 75 µg/ml of these nanomaterials, the survival, locomotion behaviors, acetylcholinesterase (AchE) activity, reactive oxygen species production, and glutathione-S transferase 4 (Gst-4) activation in wildtype and transgenic Caenorhabditis elegans (C. elegans) were evaluated. All nanomaterials induced an imbalance in oxidative stress, decreased the ratio of survival, impaired locomotion behaviors, as well as reduced the activity of AchE in C. elegans. Interestingly, CoNPs and MWCNTs activated Gst-4, but not TiO2-NPs. The reactive oxygen species scavenger, N-acetyl-l-cysteine, alleviated oxidative stress and Gst-4 upregulation upon exposure to CoNPs and MWCNTs, and rescued the locomotion behaviors. MWCNTs caused the most severe damage, followed by CoNPs and TiO2-NPs. Furthermore, oxidative stress and subsequent activation of Gst-4 were involved in nanomaterials-induced neurotoxicity. Our study provides a comprehensive comparison of the neurotoxicity and mechanisms of typical nanomaterials, which could serve as a model for hazard assessment of environmental pollutants using C. elegans as an experimental model system.


Subject(s)
Nanoparticles , Nanotubes, Carbon , Animals , Reactive Oxygen Species , Caenorhabditis elegans , Nanotubes, Carbon/toxicity , Cobalt/toxicity , Acetylcholinesterase , Oxidative Stress , Nanoparticles/toxicity
10.
Neurotoxicology ; 97: 120-132, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37302585

ABSTRACT

Copper (Cu) and Zinc (Zn) are required in small concentrations for metabolic functions, but are also toxic. There is a great concern about soil pollution by heavy metals, which may exposure the population to these toxicants, either by inhalation of dust or exposure to toxicants through ingestion of food derived from contaminated soils. In addition, the toxicity of metals in combination is questionable, as soil quality guidelines only assess them separately. It is well known that metal accumulation is often found in the pathologically affected regions of many neurodegenerative diseases, including Huntington's disease (HD). HD is caused by an autosomal dominantly inherited CAG trinucleotide repeat expansion in the huntingtin (HTT) gene. This results in the formation of a mutant huntingtin (mHTT) protein with an abnormally long polyglutamine (polyQ) repeat. The pathology of HD results in loss of neuronal cells, motor changes, and dementia. Rutin is a flavonoid found in various food sources, and previous studies indicate it has protective effects in HD models and acts as a metal chelator. However, further studies are needed to unravel its effects on metal dyshomeostasis and to discern the underlying mechanisms. In the present study, we investigated the toxic effects of long-term exposure to copper, zinc, and their mixture, and the relationship with the progression of neurotoxicity and neurodegeneration in a C. elegans-based HD model. Furthermore, we investigated the effects of rutin post metal exposure. Overall, we demonstrate that chronic exposure to the metals and their mixture altered body parameters, locomotion, and developmental delay, in addition to increasing polyQ protein aggregates in muscles and neurons causing neurodegeneration. We also propose that rutin has protective effects acting through mechanisms involving antioxidant and chelating properties. Altogether, our data provides new indications about the higher toxicity of metals in combination, the chelating potential of rutin in the C. elegans model of HD and possible strategies for future treatments of neurodegenerative diseases caused by the aggregation of proteins related to metals.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Animals , Humans , Huntington Disease/chemically induced , Huntington Disease/prevention & control , Huntington Disease/genetics , Caenorhabditis elegans , Copper/toxicity , Zinc , Rutin/pharmacology , Disease Models, Animal
11.
Sci Total Environ ; 881: 163429, 2023 Jul 10.
Article in English | MEDLINE | ID: mdl-37072102

ABSTRACT

Cobalt exposure, even at low concentrations, induces neurodegenerative damage, such as Alzheimer's disease (AD). The specific underlying mechanisms remain unclear. Our previous study demonstrated that m6A methylation alteration is involved in cobalt-induced neurodegenerative damage, such as in AD. However, the role of m6A RNA methylation and its underlying mechanisms are poorly understood. In this study, both epidemiological and laboratory studies showed that cobalt exposure could downregulate the expression of the m6A demethylase ALKBH5, suggesting a key role for ALKBH5. Moreover, Methylated RNA immunoprecipitation and sequencing (MeRIP-seq) analysis revealed that ALKBH5 deficiency is associated with neurodegenerative diseases. KEGG pathway and Gene ontology analyses further revealed that the differentially m6A-modified genes resulting from ALKBH5 downregulation and cobalt exposure were aggregated in the pathways of proliferation, apoptosis, and autophagy. Subsequently, ALKBH5 deficiency was shown to exacerbate cell viability decline, motivate cell apoptosis and attenuate cell autophagy induced by cobalt with experimental techniques of gene overexpression/inhibition. In addition, morphological changes in neurons and the expression of AD-related proteins, such as APP, P-Tau, and Tau, in the cerebral hippocampus of wild-type and ALKBH5 knockout mice after chronic cobalt exposure were also investigated. Both in vitro and in vivo results showed that lower expression of ALKBH5 aggravated cobalt-induced neurodegenerative damage. These results suggest that ALKBH5, as an epigenetic regulator, could be a potential target for alleviating cobalt-induced neurodegenerative damage. In addition, we propose a novel strategy for the prevention and treatment of environmental toxicant-related neurodegeneration from an epigenetic perspective.


Subject(s)
Cobalt , RNA , Mice , Animals , Cobalt/toxicity , Methylation
12.
J Hazard Mater ; 453: 131354, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37054644

ABSTRACT

Cobalt is the most widely used heavy metal pollutant in medicine and industry. Excessive cobalt exposure can adversely affect human health. Neurodegenerative symptoms have been observed in cobalt-exposed populations; however, the underlying mechanisms remain largely unknown. In this study, we demonstrate that the N6-methyladenosine (m6A) demethylase fat mass and obesity-associated gene (FTO) mediates cobalt-induced neurodegeneration by impairing autophagic flux. Cobalt-induced neurodegeneration was exacerbated through FTO genetic knockdown or repression of demethylase activity, but was alleviated by FTO overexpression. Mechanistically, we showed that FTO regulates TSC1/2-mTOR signaling pathway by targeting TSC1 mRNA stability in an m6A-YTHDF2 manner, which resulted in autophagosome accumulation. Furthermore, FTO decreases lysosome-associated membrane protein-2 (LAMP2) to inhibit the integration of autophagosomes and lysosomes, leading to autophagic flux damage. In vivo experiments further identified that central nervous system (CNS)-Fto-specific knockout resulted in serious neurobehavioral and pathological damage as well as TSC1-related autophagy impairment in cobalt-exposed mice. Interestingly, FTO-regulated autophagy impairment has been confirmed in patients with hip replacement. Collectively, our results provide novel insights into m6A-modulated autophagy through FTO-YTHDF2 targeted TSC1 mRNA stability, revealing cobalt is a novel epigenetic hazard that induces neurodegeneration. These findings suggest the potential therapeutic targets for hip replacement in patients with neurodegenerative damage.


Subject(s)
Autophagy , Cobalt , Animals , Humans , Mice , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Cobalt/toxicity , Obesity , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism
13.
Ecotoxicol Environ Saf ; 255: 114804, 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-36948007

ABSTRACT

Paraquat (PQ) has been widely acknowledged as an environmental risk factor for Parkinson's disease (PD). However, the interaction between splicing factor and long non-coding RNA (lncRNA) in the process of PQ-induced PD has rarely been studied. Based on previous research, this study focused on splicing factor 3 subunit 3 (SF3B3) and lncRNA NR_030777. After changing the target gene expression level by lentiviral transfection technology, the related gene expression was detected by western blot and qRT-PCR. The expression of SF3B3 protein was reduced in Neuro-2a cells after PQ exposure, and the reactive oxygen species (ROS) scavenger N-acetylcysteine prevented this decline. Knockdown of SF3B3 reduced the PQ-triggered NR_030777 expression increase, and overexpression of NR_030777 reduced the transcriptional and translational level of Sf3b3. Then, knockdown of SF3B3 exacerbated the PQ-induced decrease in cell viability and aggravated the reduction of tyrosine hydroxylase (TH) protein expression. Overexpressing SF3B3 reversed the reduction of TH expression caused by PQ. Moreover, after intervention with the autophagy inhibitor Bafilomycin A1, LC3B-II protein expression was further increased in Neuro-2a cells with the knockdown of SF3B3, indicating that autophagy was enhanced. In conclusion, PQ modulated the interplay between NR_030777 and SF3B3 through ROS production, thereby impairing autophagic flux and causing neuronal damage.


Subject(s)
Paraquat , RNA, Long Noncoding , Acetylcysteine/pharmacology , Neurons/metabolism , Paraquat/toxicity , Reactive Oxygen Species/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA Splicing Factors/metabolism
14.
Neurotoxicology ; 95: 155-163, 2023 03.
Article in English | MEDLINE | ID: mdl-36716931

ABSTRACT

Exposure to cobalt nanoparticles (CoNPs) has been associated with neurodegenerative disorders, while the mitochondrial-associated mechanisms that mediate their neurotoxicity have yet to be fully characterized. In this study, we reported that CoNPs exposure reduced the survival and lifespan in the nematodes, Caenorhabditis elegans (C. elegans). Moreover, exposure to CoNPs aggravated the induction of paralysis and the aggregation of ß-amyloid (Aß). These effects were accompanied by reactive oxygen species (ROS) overproduction, ATP reduction as well as mitochondrial fragmentation. Dynamin-related protein 1 (drp-1) activation and ensuing mitochondrial fragmentation have been shown to be associated with CoNPs-reduced survival. In order to address the role of mitochondrial damage and ROS production in CoNPs-induced Aß toxicity, the mitochondrial reactive oxygen species scavenger mitoquinone (Mito Q) was used. Our results showed that Mito Q pretreatment alleviated CoNPs-induced ROS generation, rescuing mitochondrial dysfunction, thereby lessening the CoNPs-induced Aß toxicity. Taken together, we show for the first time, that increasing of ROS and the upregulation of drp-1 lead to CoNPs-induced Aß toxicity. Our novel findings provide in vivo evidence for the mechanisms of environmental toxicant-induced Aß toxicity, and can afford new modalities for the prevention and treatment of CoNPs-induced neurodegeneration.


Subject(s)
Amyloid beta-Peptides , Nanoparticles , Animals , Reactive Oxygen Species/metabolism , Amyloid beta-Peptides/toxicity , Cobalt/toxicity , Caenorhabditis elegans/metabolism , Nanoparticles/toxicity
15.
Sci Total Environ ; 857(Pt 2): 159432, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36243078

ABSTRACT

Cobalt is an environmental toxicant, and excessive bodily exposure can damage the nervous system. Particularly, our previous study reported that low-dose cobalt (significantly less than the safety threshold) is still able to induce neurodegenerative changes. However, the underlying molecular mechanism is still insufficient revealed. Herein, we further investigate the molecular mechanism between cobalt-induced neurodegeneration and autophagy, as well as explore the interplay between hypoxia-inducible factor-1α (HIF-1α), reactive oxygen species (ROS), and autophagy in cobalt-exposed mice and human neuroglioma cells. We first reveal cobalt as an environmental toxicant to severely induce ß amyloid (Aß) deposition, tau hyperphosphorylation, and dysregulated autophagy in the hippocampus and cortex of mice. In particular, we further identify that cobalt-induced neurotoxicity is triggered by the impairment of autophagic flux in vitro experiments. Moreover, the mechanistic study reveals that cobalt exposure extremely activates HIF-1α expression to facilitate the overproduction of ROS. Then, elevated ROS can target the amino-threonine kinase (AKT)-mammalian target of rapamycin (mTOR)-Unc-51 like autophagy activating kinase 1 (ULK1) signaling pathway to participate in cobalt-induced impairment of autophagic flux. Subsequently, defected autophagy further exacerbates cobalt-induced neurotoxicity for its unable to eliminate the deposition of pathological protein. Therefore, our data provide scientific evidence for cobalt safety evaluation and risk assessment and propose a breakthrough for understanding the regulatory relationship between HIF-1α, ROS, and autophagy in cobalt-induced neurodegeneration.


Subject(s)
Cobalt , Hypoxia-Inducible Factor 1, alpha Subunit , Reactive Oxygen Species , Animals , Humans , Mice , Autophagy/physiology , Cobalt/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
16.
Huan Jing Ke Xue ; 43(10): 4755-4764, 2022 Oct 08.
Article in Chinese | MEDLINE | ID: mdl-36224161

ABSTRACT

To illustrate the effects of long-term straw returning on the fungal community, soil enzyme activity, and crop yield in a fluvo-aquic soil area typical of the Huang-Huai-Hai Plain, a 10-year field experiment (established in 2010) located in Dezhou City, Shandong province, was performed, including three fertilization regimes (NF, no fertilization control; NPK, fertilization with chemical N, P, and K fertilizers; NPKS, straw returning combined with chemical N, P, and K fertilizers). This study aimed to explore the regulation mechanisms of fungal communities on soil fertility, enzyme activities, and crop yield by employing co-occurrence network and structural equation model analyses. Our results showed that long-term straw returning significantly improved soil nutrients, enzyme activity, and wheat yield. Compared with the NPK and NF treatments, soil organic matter (SOM) increased by 9.20% and 34.75%, alkali-hydrolyzed nitrogen (AN) increased by 12.03% and 39.17%, dehydrogenase (DHA) increased by 37.21% and 50.91%, ß-glucosidase (ß-GC) increased by 17.29% and 73.48%, and wheat production increased by 16.22% and 125.53%, respectively. Different long-term fertilization regimes did not significantly change soil fungal α-diversity but resulted in significant differences in ß-diversity. Available phosphorus (AP), SOM, and AN were the main driving factors of fungal community differentiation based on redundancy analysis and hierarchical partitioning analysis. Different abundance analyses revealed significantly different fungal community compositions among fertilization regimes. The long-term NF treatment resulted in a significant enrichment of phosphate/potassium-solubilizing species (i.e., Mortierella, Aspergillus, Ceriporia, and Acremonium) and symbiotic species (i.e., Leohumicola and Hyalodendriella). The relative abundance of pathogenic fungi, namely Sarocladium, Fusarium, and Fusicolla, increased significantly in the NPK treatment. Long-term straw returning in the NPKS treatment significantly stimulated the growth of plant growth-promoting species (i.e., Pseudogymnoascus and Schizothecium) and straw-degrading species (i.e., Trichocladium and Lobulomyces). Co-occurrence network analysis showed that the fungal network was composed of four main modules; the cumulative relative abundance of module 2 was significantly increased under the NPKS treatment and showed a positive linear correlation with DHA and ß-GC. The structural equation model further indicated that the wheat yield was mainly regulated by SOM, whereas species of module 2 could indirectly affect SOM and wheat yield by positively regulating DHA and ß-GC. Taken together, long-term straw returning to the fluvo-aquic soil area of the Huang-Huai-Hai Plain could regulate fungal interspecific interactions, stimulate the growth of specific species groups, inhibit the activity of pathogens, increase the activity of soil enzymes, promote the accumulation of SOM, and achieve high crop yield.


Subject(s)
Mycobiome , Soil , Agriculture/methods , Alkalies , Fertilizers/analysis , Nitrogen/analysis , Oxidoreductases , Phosphates/analysis , Phosphorus/analysis , Potassium/chemistry , Soil/chemistry , Soil Microbiology , Triticum , beta-Glucosidase
17.
Curr Protoc ; 2(7): e496, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35849041

ABSTRACT

Nanomaterials are broadly used in a variety of industries and consumer products. However, studies have demonstrated that many nanomaterials, including metal-containing nanoparticles and nanoplastics, have neurotoxic effects. Caenorhabditis elegans (C. elegans) is a widely used model organism with numerous advantages for research, including transparency, short life span, well-characterized nervous system, complete connectome, available genome, and numerous genetic tools. C. elegans has been extensively used to assess the neurotoxicity of multiple chemicals via survival assays, behavioral tests, neuronal morphology studies, and various molecular and mechanistic analyses. However, detailed protocols describing general assays in C. elegans to examine the neurotoxic effects of nanomaterials are limited. Here, we describe protocols for assessing nanomaterial neurotoxicity in C. elegans. We describe the steps for exposure and subsequent evaluation of survival, locomotion behavior, and oxidative stress. Survival and locomotion behavior are measured in wild-type N2 strains to assess acute neurotoxicity. Oxidative stress is used as an endpoint here since it is one of the most predominant and common changes induced by nanomaterials. VP596 nematodes, which express GFP upon activation of skn-1 (the worm homolog of Nrf2), are evaluated for assays of oxidative stress in response to test nanomaterials. These assays can be readily used to quickly examine the neurotoxicity of nanomaterials in vivo, laying the foundation for mechanistic studies of nanomaterials and their impacts on health and physiology. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Exposure of C. elegans to nanomaterials Basic Protocol 2: Survival assessment Basic Protocol 3: Assessment of locomotion behavior Basic Protocol 4: Analysis of oxidative stress.


Subject(s)
Caenorhabditis elegans Proteins , Nanostructures , Neurotoxicity Syndromes , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Locomotion , Nanostructures/toxicity , Oxidative Stress
18.
Talanta ; 247: 123596, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35640476

ABSTRACT

L-tryptophan (Trp) is an essential amino acid for humans and plays crucial roles in many metabolic functions. Trp levels can be used for diagnosing different kinds of metabolic disorders and the symptoms associated with those diseases. Herein, a novel, simple and sensitive sensor based on 3D peony-like bimetallic conductive MOFs (Co-Ni-MOFs) was fabricated for the electrochemical determination of Trp. The bimetallic conductive MOFs were synthesized by a facile one-pot hydrothermal process. On account of the synergy between the Ni2+ and Co2+ ions, the bimetallic Co-Ni-MOFs showed excellent electrochemical performance, including good conductivity, large effective surface areas, and high electrocatalytic reactivity toward the oxidation of Trp. Consequently, the Co-Ni-MOFs-modified electrodes obtained a wide linear range from 10 nmol L-1 to 300 µmol L-1 and a low detection limit of 8.7 nmol L-1 (S/N = 3) for Trp. Additionally, the prepared sensor also displayed high selectivity, long-term stability and reproducibility. Moreover, the proposed sensor was successfully applied to determine the levels of Trp in the plasma of mice after cadmium intoxication.


Subject(s)
Electrochemical Techniques , Tryptophan , Animals , Electrodes , Limit of Detection , Mice , Reproducibility of Results , Tryptophan/chemistry
19.
Ecotoxicol Environ Saf ; 237: 113503, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35453019

ABSTRACT

Paraquat (PQ) is a ubiquitously applied herbicide. Long-term PQ exposure with low dose has been reported to induce abnormal expression of long non-coding RNAs (lncRNAs) in brain nerve cells, which could further lead to Parkinson's disease (PD). N6-methyladenosine (m6A) modification has recently been identified as having an important role in regulating the function of lncRNAs. However, how m6A modification regulates lncRNAs following PQ exposure remains largely unknown. Herein, this study reported m6A modification of lncRNAs in mouse neuroblastoma cells (Neuro-2a) following PQ induced reactive oxide species (ROS). M6A sequencing was performed to explore the m6A modificated pattern of lncRNAs in Neuro-2a cells which were treated with 200 µM PQ for 3 h. It was found that PQ hypermethylated total RNA and changed the expression of m6A methyltransferase and demethylase proteins, which leading to the alteration of m6A modification of lncRNAs. Furthermore, the functional analysis further revealed that N-acetyl-L-cysteine (NAC),a ROS scavengers, partly reversed PQ-induced distinct m6A modificated pattern of lncRNAs. In addition, tow specific m6A modified lncRNAs were identified: cell division cycle 5-like (lncRNA CDC5L) and signal transducer and activator of transcription 3 (lncRNA STAT3), which could influence downstream autophagy related biological function. In summary, this work could potentially contribute to the new insight of lncRNAs m6A modification mechanism in the field of environmental toxicology.


Subject(s)
Paraquat , RNA, Long Noncoding , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Mice , Oxidative Stress/genetics , Paraquat/toxicity , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Reactive Oxygen Species/metabolism
20.
Antioxidants (Basel) ; 11(4)2022 Apr 02.
Article in English | MEDLINE | ID: mdl-35453390

ABSTRACT

Natural products are small molecules naturally produced by multiple sources such as plants, animals, fungi, bacteria and archaea. They exert both beneficial and detrimental effects by modulating biological targets and pathways involved in oxidative stress and antioxidant response. Natural products' oxidative or antioxidative properties are usually investigated in preclinical experimental models, including virtual computing simulations, cell and tissue cultures, rodent and nonhuman primate animal models, and human studies. Due to the renewal of the concept of experimental animals, especially the popularization of alternative 3R methods for reduction, replacement and refinement, many assessment experiments have been carried out in new alternative models. The model organism Caenorhabditis elegans has been used for medical research since Sydney Brenner revealed its genetics in 1974 and has been introduced into pharmacology and toxicology in the past two decades. The data from C. elegans have been satisfactorily correlated with traditional experimental models. In this review, we summarize the advantages of C. elegans in assessing oxidative and antioxidative properties of natural products and introduce methods to construct an oxidative damage model in C. elegans. The biomarkers and signaling pathways involved in the oxidative stress of C. elegans are summarized, as well as the oxidation and antioxidation in target organs of the muscle, nervous, digestive and reproductive systems. This review provides an overview of the oxidative and antioxidative properties of natural products based on the model organism C. elegans.

SELECTION OF CITATIONS
SEARCH DETAIL
...