Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Inflammation ; 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896231

ABSTRACT

This study aimed to investigate the potential protective effects of Dexmedetomidine (DEX) against acute kidney injury (AKI) induced by acute stress (AS). Wistar rats were divided into five groups: Control, DEX, AS, AS + DEX, and AS + A438079. The results showed that AS led to AKI by increasing inflammatory biomarkers and oxidative stress-related indicators. The acute stress model in rats was successfully established. Renal function, histopathology, oxidative stress, and inflammation were assessed. Localization of P2X7 receptor (P2X7R) was determined by immunofluorescence. Additionally, the key inflammatory proteins of the P2X7R/NF-κB/NLRP3 signaling pathway were measured by Western blotting. DEX significantly improved kidney function, alleviated kidney injury, and reduced oxidative stress and inflammation. DEX inhibited the activation of the P2X7R, decreased the expression of NF-κB, NLRP3 inflammasome, and Caspase-1, and inhibited the expression of interleukin-1ß (IL-1ß) and tumor necrosis factor α (TNFα). Furthermore, DEX also alleviated AS-induced AKI by inhibiting the excessive production of reactive oxygen species (ROS) and reducing oxidative stress. In conclusion, DEX attenuates AS-induced AKI by mitigating inflammation and oxidative stress through the inhibition of the P2X7R/NF-κB/NLRP3 pathway in rats.

2.
ACS Omega ; 9(23): 25370-25380, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38882150

ABSTRACT

PFOS is a ubiquitous pollutant garnering considerable attention due to its deleterious effects on both human and animal health. Given the poultry industry's intimate link with human health, investigating PFOS's impact on quails is crucial. PFOS readily accumulates in the liver, causing hepatotoxicity, yet its molecular mechanisms remain elusive. In our study, we fed quail diets contaminated with varying PFOS concentrations (12.5, 25, and 50 mg/kg) and observed dose-dependent liver damage in quails. The results show that PFOS damages mitochondrial structure, increases ROS levels, and downregulates antioxidants to promote oxidative stress damage in hepatocytes. PFOS also upregulated pro-inflammatory molecules (TNF-α, IL-1ß, and IL-6) while downregulating the anti-inflammatory factor IL-10, activating the TLR4//MyD88/NF-κB signaling pathway, thereby potentiating liver inflammation. Then, oxidative stress and inflammation by PFOS induce apoptosis in quail hepatocytes through the mitochondrial pathway, with severity closely related to hepatotoxicity. In conclusion, PFOS induces mitochondrial apoptosis by exacerbating oxidative stress and inflammation by activating the TLR4/MyD88/NF-κB signaling pathway, ultimately leading to hepatotoxicity in quails.

3.
Environ Toxicol ; 39(1): 148-155, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37676913

ABSTRACT

Glufosinate-ammonium (GLA) is a widely used herbicide, but less research has been done on its harmful effects on non-target organisms, especially aquatic organisms. In this study, 600 adult zebrafish were exposed to different concentration of GLA (0, 1.25, 2.5, 5, 10, and 20 mg/L) for 7 days, and the livers were dissected on the eighth day to examine the changes in liver structure, function, oxidative stress, inflammation, apoptosis, and Nrf2 pathway, and finally to clarify the mechanism of GLA induced liver injury in zebrafish. The levels of alanine aminotransferase, aspartate aminotransferase, reactive oxygen species, malondialdehyde, inflammatory factors (IL-6 and TNF-α), and caspase-3 gradually increased, while the levels of superoxide dismutase, catalase, glutathione, and glutathione peroxidase gradually decreased with the increase of GLA concentration. The Nrf2 pathway was activated at low concentrations (1.25-5 mg/L) and significantly inhibited at high concentrations (10 and 20 mg/L). These results suggested that GLA could cause oxidative stress, inflammation, and apoptosis in zebrafish liver. Therefore, GLA can cause liver injury in zebrafish, and at high concentrations, the inhibition of Nrf2 pathway is one of the important causes of liver injury.


Subject(s)
NF-E2-Related Factor 2 , Zebrafish , Animals , Zebrafish/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Liver , Inflammation/chemically induced , Inflammation/metabolism
4.
J Agric Food Chem ; 71(24): 9381-9390, 2023 Jun 21.
Article in English | MEDLINE | ID: mdl-37293923

ABSTRACT

Chronic stress can cause intestinal barrier damage. MAPK and NF-κB are closely related to it. Chlorogenic acid (CGA), a dietary polyphenol, has been shown to have intestinal protective effects, but whether by regulating MAPK and NF-κB is not known. Therefore, in this experiment, 24 Wistar rats were randomly divided into 4 groups (C group, CS group, CS + SB203580, and CS + CGA group). Rats in the CS group were restrained stress for 6 h per day for 21 days. Rats in the CS + SB203580 group were given SB203582 (0.5 mg/kg, intraperitoneal injection) 1 h before restraint stress every other day. Rats in the CS + CGA group were given CGA (100 mg/kg, gavage) 1 h before restraint stress. In chronic stress, intestinal barrier damage was evident, while being restored after CGA treatment. After chronic stress, the levels of p-P38 were increased (P < 0.01), while the levels of p-JNK and p-ERK were not changed. The levels of p-p38 were elevated after CGA treatment (P < 0.01). These results suggested that p38MAPK played an important role in chronic stress-induced intestinal injury, and CGA could inhibit p38MAPK activity. Therefore, we chose SB203582 (P38MAPK inhibitor) to elucidate the role of p38. After chronic stress, intestinal tight junction key proteins Occludin, ZO-1, and Claudin3 protein and gene expression were reduced (P < 0.01), while being elevated after CGA or SB203582 intervention (P < 0.05). After CGA treatment, the levels of p-IκB, p-p65, p-p38, and TNF-α were reduced (P < 0.01). SB203582 intervention reduced p-p65 and TNF-α levels significantly (P < 0.01). These results suggested that CGA could inhibit the NF-κB pathway by suppressing p38MAPK, thereby alleviating chronic stress-induced intestinal damage.


Subject(s)
Chlorogenic Acid , NF-kappa B , Rats , Animals , NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , Tumor Necrosis Factor-alpha , Rats, Wistar
5.
Int Immunopharmacol ; 117: 109898, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36827925

ABSTRACT

Sepsis is currently the main factor of death in the ICU, and the liver, as an important organ of immunity and stable metabolism, can be acutely damaged during sepsis, and the mortality rate of patients with sepsis complicated by acute liver injury is greatly increased. Celastrol (CEL) is derived from the root bark of Tripterygium wilfordii Hook.f.. As a traditional Chinese medicine, CEL has anti-inflammatory, anti-cancer, anti-oxidant, and other biological activities. Obtain CEL and AHI intersection targets via database and construct protein-protein interaction (PPI) network by STRING. GO functional enrichment and KEGG pathway analyses were performed by R studio. Targets were finally selected to perform molecular docking simulations with CEL. In vivo experiments based on the model of AHI were established by intraperitoneal injection of Lipopolysaccharide (LPS) 4 h, and pre-treated with CEL (0.5 mg/kg, 1 mg/kg, 1.5 mg/kg). The results are as follows: 273 genes with the intersection of CEL and AHI were obtained, and GO and KEGG enrichment analysis were used to design the mechanism of inflammation, apoptosis, and oxidative stress-related injury. By constructing the PPI network selected top 10 targets are: STAT3, RELA, MAPK1, MAPK3, TP53, AKT1, HSP90AA1, JUN, TNF, MAPK14, predicted CEL protection AHI design related pathways of MAPK and PI3K/AKT-related signal pathways. In vivo experiments, CEL inhibited the activation of MAPK and PI3K/AKT related pathways, reduced inflammatory response, apoptosis, and oxidative stress, and significantly improved LPS-induced AHI. In summary, this study predicted the mechanisms involved in the protective effect of CEL on AHI through network pharmacology. In vivo, CEL inhibited MAPK and PI3K/AKT-related signaling pathways, and reduced inflammatory response, apoptosis, and oxidative stress to protect LPS-induced AHI.


Subject(s)
Drugs, Chinese Herbal , Network Pharmacology , Humans , Lipopolysaccharides , Molecular Docking Simulation , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Liver , Antioxidants
6.
J Agric Food Chem ; 70(45): 14414-14426, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36318656

ABSTRACT

The liver is the major organ of metabolism and is extremely vulnerable to chronic stress. Lycopene (LYC) is a natural carotenoid with potent antioxidant and chronic disease potential. However, whether LYC protects against chronic restraint stress (CRS)-induced liver injury and the underlying mechanisms remain unclear. In this study, rats were restrained for 21 days for 6 h per day, with or without gavage of LYC (10 mg/kg). Serum ALT (85.99 ± 4.07 U/L) and AST (181.78 ± 7.35 U/L) and scores of liver injury were significantly increased in the CRS group. LYC significantly promoted the nuclear translocation of Nrf2, elevated the expression of antioxidant genes, and attenuated reactive oxygen radicals (ROS) levels within the liver. Cellular thermal shift assay (CETSA) and molecular docking results indicated that LYC competitively binds to Keap1 with the lowest molecule affinity of -9.0 kcal/mol. Moreover, LYC significantly relieved the hepatic endoplasmic reticulum swelling and decreased the expression of endoplasmic reticulum stress (ERS) hallmarks like GRP78, CHOP, and cleaved caspase-12. Meanwhile, LYC also mitigated CRS-induced hepatocyte apoptosis. Interestingly, every other day, the intraperitoneal injection of the Nrf2 inhibitor brusatol (0.4 mg/kg) significantly counteracted the protective effect of LYC. In conclusion, LYC protects against CRS-induced liver injury by activating the Nrf2 signaling pathway, scavenging ROS, and further attenuating ERS-associated apoptosis pathways.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , NF-E2-Related Factor 2 , Rats , Animals , Lycopene/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Endoplasmic Reticulum Stress , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Molecular Docking Simulation , Chemical and Drug Induced Liver Injury, Chronic/etiology , Chemical and Drug Induced Liver Injury, Chronic/genetics , Oxidative Stress , Apoptosis
7.
Food Chem Toxicol ; 169: 113441, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36162616

ABSTRACT

Melatonin, as a dietary supplement, has a potent neuroprotective effect and exerts a certain antidepressant effect. This study explored the molecular mechanisms and targets of melatonin on chronic stress-induced hippocampal damage from the perspective of inhibiting inflammatory cytokines release. Our results indicated that melatonin alleviated chronic restraint stress (CRS)-induced inflammatory response and apoptosis, thus improving hippocampal structural damage and subsequent depression-like behaviors in rats. The radar map displayed that the change of TNF-α content was the most significant. Meanwhile, correlation analysis showed that TNF-α content was highly positively correlated with apoptosis. Molecular autodocking studies suggested that TNF-α converting enzyme ADAM17 as a potential target has a priority in docking with melatonin. Molecular mechanism studies indicated that melatonin inhibited CRS-induced activation of the ADAM17/TNF-α axis and its downstream proteins p38 and p53 phosphorylation in the hippocampus. Analogously, Both ADAM17 inhibitor TMI-1 and TNF-α inhibitor thalidomide relieved the effects of CRS on ADAM17/TNF-α axis and its downstream proteins phosphorylation, hippocampal apoptosis, hippocampal inflammatory response, and depression-like behaviors in rats. Altogether, these findings reveal that melatonin relieves CRS-induced inflammatory response and apoptosis, and subsequent depression-like behaviors by inhibiting ADAM17/TNF-α axis.


Subject(s)
ADAM17 Protein , Apoptosis , Hippocampus , Melatonin , Neuroinflammatory Diseases , Neuroprotective Agents , Stress, Psychological , Animals , Rats , ADAM17 Protein/antagonists & inhibitors , Cytokines/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Melatonin/pharmacology , Melatonin/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/ethnology , Stress, Psychological/complications , Chronic Disease
8.
J Agric Food Chem ; 70(34): 10532-10542, 2022 Aug 31.
Article in English | MEDLINE | ID: mdl-35975781

ABSTRACT

Chronic stress can cause chronic inflammatory injury to the liver. Chlorogenic acid (CGA) is known to have a wide range of biological activities and anti-inflammatory effects. Resolvin D1 (RvD1) is a polyunsaturated fatty acid derivative that has inhibitory effects on a variety of inflammatory diseases. However, whether CGA can inhibit liver inflammation in chronic stress through RvD1 remains unclear. In this work, male rats were subjected to restraint stress for 6 h every day and built a chronic stress model for 21 days. CGA (100 mg/kg) was administered intragastrically 1 h before restraint, with intraperitoneal injection of RvD1 inhibitor WRW4 (antagonist of FPR2, 0.1 mg/kg) or WRW4 solution every 2 days for 30 min before CGA administration. CGA reduced hepatic hemorrhage and inflammatory cell infiltration, alleviated hepatic injury, decreased the activation of the NF-κB pathway and the expression of interleukin 1ß, interleukin 6, and tumor necrosis factor α in the liver, and increased RvD1 in the serum and liver. The therapeutic effect of CGA was blocked after WRW4 intervention. These results suggest that the protective effects of CGA mediate the NF-κB pathway by upregulating the generation of RvD1. Above all, this research demonstrates the liver protective effect of CGA and provides a potential treatment strategy for chronic inflammatory disease.


Subject(s)
Chlorogenic Acid , NF-kappa B , Animals , Docosahexaenoic Acids/pharmacology , Inflammation/drug therapy , Inflammation/pathology , Liver/metabolism , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Rats
9.
Phytother Res ; 36(6): 2558-2571, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35570830

ABSTRACT

Acute liver injury (ALI) is a poor prognosis and high mortality complication of sepsis. Paeoniflorin (PF) has remarkable anti-inflammatory effects in different disease models. Here, we explored the protective effect and underlying molecular mechanisms of PF against lipopolysaccharide (LPS)-induced ALI. Sprague-Dawley rats received intraperitoneal (i.p.) injection of PF for 7 days, 1 h after the last administration, and rats were injected i.p. 10 mg/kg LPS. PF improved liver structure and function, reduced hepatic reactive oxygen species (ROS) and methane dicarboxylic aldehyde (MDA) levels, and increased superoxide dismutase (SOD) activity. Western blot analysis suggested that PF significantly inhibited expression of inflammatory cytokines (TNF-α, IL-1ß, and IL-18) and inhibited activation of the NLRP3 inflammasome. PF or mitochondrial ROS scavenger (mito-TEMPO) significantly improved liver mitochondrial function by scavenging mitochondrial ROS (mROS), restoring mitochondrial membrane potential loss and increasing level of ATP and enzyme activity of complex I and III. In addition, PF increased expression of sirtuin-1 (SIRT1), forkhead box O1 (FOXO1a) and manganese superoxide dismutase (SOD2), and increased FOXO1a nuclear retention. However, the inhibitor of SIRT1 (EX527) abolished the protective effect of PF. Taken together, PF promotes mROS clearance to inhibit mitochondrial damage and activation of the NLRP3 inflammasome via SIRT1/FOXO1a/SOD2 signaling.


Subject(s)
Chemical and Drug Induced Liver Injury , Glucosides , Monoterpenes , Oxidative Stress , Animals , Chemical and Drug Induced Liver Injury/drug therapy , Glucosides/pharmacology , Inflammasomes/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides , Liver/metabolism , Monoterpenes/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction , Sirtuin 1/metabolism , Superoxide Dismutase/metabolism
10.
Stoch Environ Res Risk Assess ; 36(11): 3785-3802, 2022.
Article in English | MEDLINE | ID: mdl-35599986

ABSTRACT

The increasing carbon emissions have been a major concern for most countries around the world. And as a result, every country is concerned about developing appropriate strategies to curtail it. As a major economy and the largest carbon emitter in the world, China has pledged to reduce the carbon intensity by 60-65% by 2030, compared with 2005 levels, and achieve carbon neutrality before 2060. Therefore, the analysis of the impact of China's carbon intensity is becoming an increasing important topic. Due to the spatial heterogeneity of carbon intensity, various spatial econometric models have been applied in this field. However, the existing literatures failed to consider the cross-products of relevant factors. This paper constructs our dynamic general nesting spatial panel model (GNS) with common factors to deal with the dilemma, and examines the direct and spatial-temporal spillover effects of industrial structure, GDP per capita, investment in anti-pollution projects as percentage of GDP and energy price on carbon intensity in China over the period 2003-2017. Our analysis shows that: (1) China's carbon intensity showed the spatial agglomeration and temporal "inertia" from 2003 to 2017. (2) From the time dimension, the long-term effect of industrial structure first increased and then gradually decreased. (3) From the spatial dimension, industrial structure and investment in anti-pollution projects as percentage of GDP accounted for the main spatial heterogeneity. Furthermore, this paper attempts to provide policy implications to help reduce carbon intensity and achieve carbon neutrality in China.

11.
J Agric Food Chem ; 70(14): 4353-4361, 2022 Apr 13.
Article in English | MEDLINE | ID: mdl-35380825

ABSTRACT

Chronic stress causes duodenal damage, in which iron death is likely to play an important role. Chlorogenic acid (CGA), one of the most widely consumed dietary polyphenols, has been shown to protect the intestine. However, it is unclear whether CGA exerts a duodenoprotective effect in chronic stress by inhibiting ferroptosis. In this work, rats were daily exposed to restraint stress for 6 h over 21 consecutive days, with/without CGA (100 mg/kg, gavage). CGA reduced blood hepcidin, iron, reactive oxygen species (ROS), and ferroportin 1 (FPN1) levels and upregulated the levels of ferroptosis-related biomarkers (GPX4, GSH, NADPH, etc.). These results confirmed that CGA inhibited ferroptosis in the duodenum. Furthermore, the use of S3I-201 (STAT3 inhibitor) helped to further clarify the mechanism of action of CGA. Overall, CGA could reduce hepcidin production by inhibiting the IL-6/JAK2/STAT3 pathway in the liver to increase the expression of FPN1 in the duodenum, which restored iron homeostasis and inhibited ferroptosis, alleviating chronic stress-induced duodenal injury.


Subject(s)
Ferroptosis , Animals , Chlorogenic Acid , Duodenum/metabolism , Ferroptosis/genetics , Hepcidins/genetics , Interleukin-6/genetics , Interleukin-6/pharmacology , Iron/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Rats , Signal Transduction
12.
J Agric Food Chem ; 70(9): 2889-2897, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35212537

ABSTRACT

Chronic stress induction in immunosuppression and splenocyte apoptosis is commonly associated with increased susceptibility to various diseases. Lycopene (LYC) is a member of the carotenoid family with immune restoration and anti-apoptotic function. However, little is known about the mechanisms underlying the protective roles of LYC against spleen injury induced by chronic stress. Herein, male Wistar rats were undergoing chronic restraint stress and/or administered LYC (10 mg/kg) for 21 days. The effective model establishment was validated by open-field tests and levels of corticosterone in serum. Histopathological staining observation displayed that LYC could reduce chronic stress-induced spleen structure damage. Furthermore, LYC treatment significantly reduced the number of apoptotic-positive splenocytes caused by chronic stress via the death receptor apoptotic pathway. We detected the interleukin 4 and interferon γ levels in serum and spleen to determine the ratio of Th1 and Th2 and found that LYC can alleviate the immunosuppression induced by chronic stress. Notably, western blot and real-time polymerase chain reaction indicated that LYC can reduce the expression of the Notch-pathway-related proteins and mRNA in rats exposed to chronic stress. Further study of the potential mechanisms by adding the Notch pathway inhibitor DAPT revealed that LYC alleviates the structure damage, apoptosis, and immunosuppression caused by chronic stress via the suppression of the Notch pathway. Overall, this study presents a strong rationale to target LYC as a treatment strategy to relieve chronic stress-induced spleen injury.


Subject(s)
Oxidative Stress , Spleen , Animals , Apoptosis , Immunosuppression Therapy , Lycopene/metabolism , Male , Rats , Rats, Wistar , Signal Transduction , Spleen/metabolism
13.
Oxid Med Cell Longev ; 2021: 4475968, 2021.
Article in English | MEDLINE | ID: mdl-34691354

ABSTRACT

Low-dose alcohol possesses multiple bioactivities. Accordingly, we investigated the protective effect and related molecular mechanism of low-dose alcohol against acute stress- (AS-) induced renal injury. Herein, exhaustive swimming for 15 min combined with restraint stress for 3 h was performed to establish a rat acute stress model, which was verified by an open field test. Evaluation of renal function (blood creatinine and urea nitrogen), urine test (urine leukocyte esterase and urine occult blood), renal histopathology, oxidative stress, inflammation, and apoptosis was performed. The key indicators of the cytochrome P450 (CYP) 4A1/20-hydroxystilbenetetraenoic acid (20-HETE) pathway, cyclooxygenase (COX)/prostaglandin E2 (PGE2) pathway, and leukotriene B4 (LTB4)/leukotriene B4 receptor 1 (BLT1) pathway were measured by real-time PCR and ELISA. We found that low-dose alcohol (0.05 g/kg, i.p.) ameliorated AS-induced renal dysfunction and histological damage. Low-dose alcohol also attenuated AS-induced oxidative stress and inflammation, presenting as reduced malondialdehyde and hydrogen peroxide formation, increased superoxide dismutase and glutathione activity, and decreased myeloperoxidase, interleukin-6, interleukin-1ß, and monocyte chemoattractant protein-1 levels (P < 0.05). Moreover, low-dose alcohol alleviated AS-induced apoptosis by downregulating Bax and cleaved caspase 3 protein expression and reduced numbers of terminal deoxynucleotidyl transferase-mediated dUTP nick-end label-positive cells (P < 0.01). Correlation analysis indicated that 20-HETE was strongly correlated with oxidative stress, while LTB4 was strongly correlated with inflammation. Low-dose alcohol inhibited AS-induced increases in CYP4A1, CYP4A2, CYP4A3, CYP4A8, and BLT1 mRNA levels and LTB4 and 20-HETE content (P < 0.01). Interestingly, low-dose alcohol had no effect on COX1 or COX2 mRNA expression or the concentration of PGE2. Furthermore, low-dose alcohol reduced calcium-independent phospholipase A2 mRNA expression, but did not affect secreted phospholipase A2 or cytosolic phospholipase A2 mRNA expression. Together, these results indicate that low-dose alcohol ameliorated AS-induced renal injury by inhibiting CYP4A/20-HETE and LTB4/BLT1 pathways, but not the COX/PGE2 pathway.


Subject(s)
Acute Kidney Injury/drug therapy , Alcohol Drinking/physiopathology , Cytochrome P-450 CYP4A/metabolism , Leukotriene B4/metabolism , Animals , Male , Rats , Rats, Wistar
14.
Mol Neurobiol ; 58(11): 5533-5547, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34363182

ABSTRACT

Dexmedetomidine (DEX) has multiple biological effects. Here, we investigated the neuroprotective role and molecular mechanism of DEX against lipopolysaccharide (LPS)-induced hippocampal neuronal apoptosis. Sprague Dawley rats were intraperitoneally injected with LPS (10 mg/kg) and/or DEX (30 µg/kg). We found that DEX improved LPS-induced alterations of hippocampal microstructure (necrosis and neuronal loss in the CA1 and CA3 regions) and ultrastructure (mitochondrial damage). DEX also attenuated LPS-induced inflammation and hippocampal apoptosis by inhibiting the increase of interleukin-1ß, interleukin-6, interleukin-18, and tumor necrosis factor-α levels and downregulating the expression of mitochondrial apoptosis pathway-related proteins. Moreover, DEX prevented the LPS-induced activation of the c-Myc/chloride intracellular channel 4 (CLIC4) pathway. DEX inhibited the p38 MAPK pathway, but not JNK and ERK. To further clarify whether DEX alleviated LPS-induced neuronal apoptosis through the p38 MAPK/c-Myc/CLIC4 pathway, we treated PC12 cells with p38 MAPK inhibitor SB203582 (10 µM). DEX had the same effect as SB203582 in reducing the protein and mRNA expression of c-Myc and CLIC4. Furthermore, DEX and SB203582 diminished LPS-induced apoptosis, indicated by decreased Bax and Tom20 fluorescent double-stained cells, reduced annexin V-FITC/PI apoptosis rate, and reduced protein expression levels of Bax, cytochrome C, cleaved caspase-9, and cleaved caspase-3. Taken together, the findings indicate that DEX attenuates LPS-induced hippocampal neuronal apoptosis by regulating the p38 MAPK/c-Myc/CLIC4 signaling pathway. These findings provide new insights into the mechanism of Alzheimer's disease and depression and may help aid in drug development for these diseases.


Subject(s)
Apoptosis , Hippocampus , MAP Kinase Signaling System , Neurons , Animals , Male , Rats , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Chloride Channels/physiology , Cytokines/blood , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Hippocampus/drug effects , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/pathology , PC12 Cells , Proto-Oncogene Proteins c-myc/physiology , Random Allocation , Rats, Sprague-Dawley
15.
J Hazard Mater ; 411: 125179, 2021 06 05.
Article in English | MEDLINE | ID: mdl-33858114

ABSTRACT

Alumina nanoparticles (AlNPs) exposure causes hippocampal-dependent cognitive dysfunction. However, whether chronic stress exacerbates AlNPs-induced hippocampal lesion and its mechanism remains unclear. This study was aimed to investigate the combined effects and mechanisms of AlNPs and chronic stress on the hippocampal lesion. The behavioral tests demonstrated that combined exposure to AlNPs and chronic restraint stress (CRS) worsened both cognition and depression-like behavior than exposed to AlNPs and CRS alone. Microstructural and ultrastructural observations showed that combined exposure to AlNPs and CRS exacerbated hippocampal damage. Both AlNPs and CRS induced hippocampal neuronal ferroptosis, presenting as iron and glutamate metabolism disorder, GPX4 fluorescence of neurons decrease, LPO and ROS levels increase, and FJB-positive neurons increase. Meanwhile, combined exposure to AlNPs and CRS exacerbated hippocampal neuronal ferroptosis. Mechanism investigation revealed that combined exposure to AlNPs and CRS activated IFN-γ/ASK1/JNK signaling pathway. Furthermore, IFN-γ neutralizing antibody R4-6A2 effectively inhibited the activation of IFN-γ/ASK1/JNK signaling pathway, alleviated hippocampal neuronal ferroptosis and improved cognition ability. ASK1 inhibitor GS-4997 also improved hippocampal neuronal ferroptosis and cognitive dysfunction by inhibiting ASK1/JNK signaling pathway. Together, these results demonstrate that combined exposure to AlNPs and CRS exacerbates hippocampal neuronal ferroptosis via activating IFN-γ/ASK1/JNK signaling pathway.


Subject(s)
Ferroptosis , Nanoparticles , Aluminum Oxide , Animals , Apoptosis , Hippocampus , MAP Kinase Signaling System , Neurons , Rats
16.
Chem Res Toxicol ; 34(3): 857-864, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33539076

ABSTRACT

Sodium hydrosulfide (NaHS), as an exogenous hydrogen sulfide (H2S) donor, has been used in various pathological models. NaHS is usually considered to be primarily protective, however, the toxic effect of NaHS has not been well elucidated. The aim of this study was to investigate whether NaHS (1 mg/kg) can induce acute lung injury (ALI is a disease process characterized by diffuse inflammation of the lung parenchyma) and define the mechanism by which NaHS-induced ALI involves autophagy, oxidative stress, and inflammatory response. Wistar rats were randomly divided into three groups (control group, NaHS group, and 3-MA + NaHS group), and samples from each group were collected from 2, 6, 12, and 24 h. We found that intraperitoneal injection of NaHS (1 mg/kg) increased the pulmonary levels of H2S and oxidative stress-related indicators (reactive oxygen species, myeloperoxidase, and malondialdehyde) in a time-dependent manner. Intraperitoneal injection of NaHS (1 mg/kg) induced histopathological changes of ALI and inhibition of autophagy exacerbated the lung injury. This study demonstrates that administration of NaHS (1 mg/kg) induces ALI in rats and autophagy in response to ROS is protective in NaHS-induced ALI by attenuating oxidative stress and inflammation.


Subject(s)
Acute Lung Injury/chemically induced , Autophagy/drug effects , Inflammation/chemically induced , Sulfides/pharmacology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Inflammation/metabolism , Inflammation/pathology , Injections, Intraperitoneal , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sulfides/administration & dosage
17.
Waste Manag ; 121: 255-264, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33388648

ABSTRACT

Potassium is an inorganic mineral element in biomass and has a significant catalytic effect on biomass pyrolysis. In this work, the effect of potassium on the pyrolysis of biomass components (cellulose, xylan and lignin) was investigated with the help of thermogravimetric analyzer coupled to fourier transform infrared spectrometer (TG-FTIR) and pyrolysis-gas chromatography coupled to mass spectrometry (Py-GC/MS). The results showed that potassium accelerated the start of the main pyrolysis stage of the biomass components, reduced the weight loss rate for cellulose and lignin, and increased the weight loss rate for xylan. On the other hand, potassium presented a promotion effect on the formation of char for cellulose but a suppression effect for lignin. In addition, an increasing potassium content promoted the release of volatile products for xylan. Product distribution analysis found that potassium promoted the scission of glycosidic bonds and the decomposition of glucose units, resulting in a sharp yield decrease of carbohydrates and a yield increase of furans, aldehydes and ketones. In addition, an increased production of CO2 was obtained, indicating that potassium favors the cleavage and reforming of carboxyl (COOH) and carbonyl (CO) groups. Furthermore, the effect of potassium on the pyrolysis of cellulose and xylan was stronger than that on lignin pyrolysis. The effect on the pyrolysis reaction also resulted in a higher activation energy for the decomposition of biomass components, especially at high temperature intervals. Moreover, the higher the content of potassium added, the greater the increase was in the activation energy.


Subject(s)
Potassium , Pyrolysis , Biomass , Cellulose , Hot Temperature , Lignin , Thermogravimetry
18.
Food Chem Toxicol ; 145: 111688, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32810585

ABSTRACT

The natural carotenoid lycopene (LYC) has strong antioxidant and neuroprotective capacities. This study investigated the effects and mechanisms of LYC on chronic stress-induced hippocampal lesions and learning and memory dysfunction. Rats were administered LYC and/or chronic restraint stress (CRS) for 21 days. Morris water maze results demonstrated that LYC prevented CRS-induced learning and memory dysfunction. Histopathological staining and transmission electron microscopy observation revealed that LYC ameliorated CRS-induced hippocampal microstructural and ultrastructural damage. Furthermore, LYC alleviated CRS-induced oxidative stress by reducing reactive oxygen species (ROS) production and enhancing antioxidant enzyme activities. LYC also improved CRS-induced hippocampal mitochondrial dysfunction by recovering mitochondrial membrane potential, and complex I (NADH dehydrogenase) and II (succinate dehydrogenase) activities. Moreover, LYC reduced CRS-induced apoptosis via the mitochondrial apoptotic pathway, and decreased the number of terminal deoxynucleotidyl transferase dUTP nick-end-labeled positive cells. Additionally, western blot analysis demonstrated that LYC inhibited CRS-induced activation of the c-Jun N-terminal kinase (JNK) signaling pathway. Correlation analysis indicated that ROS levels, JNK activation, and the mitochondrial apoptotic pathway were positively correlated. Further investigation of the underlying mechanisms revealed that the ROS scavenger N-acetyl-l-cysteine inhibited CRS-induced JNK activation. Furthermore, the JNK inhibitor SP600125 relieved CRS-induced hippocampal mitochondrial dysfunction, apoptosis via the mitochondrial apoptotic pathway, and learning and memory dysfunction. Together, these results suggest that LYC alleviates hippocampal oxidative stress, mitochondrial dysfunction, and apoptosis by inhibiting the ROS/JNK signaling pathway, thereby improving CRS-induced hippocampal injury and learning and memory dysfunction. This study provides a theoretical basis and new therapeutic strategies for the application of LYC to relieve chronic stress encephalopathy.


Subject(s)
Hippocampus/drug effects , Hippocampus/injuries , JNK Mitogen-Activated Protein Kinases/metabolism , Learning/drug effects , Lycopene/administration & dosage , Memory/drug effects , Reactive Oxygen Species/metabolism , Animals , Antioxidants/administration & dosage , Apoptosis/drug effects , Hippocampus/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Male , Maze Learning , Oxidative Stress/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects , Stress, Physiological/drug effects
19.
Front Physiol ; 11: 680, 2020.
Article in English | MEDLINE | ID: mdl-32714202

ABSTRACT

Chronic stress leads to immunosuppression and induces splenocyte apoptosis. STAT3 is a transcription factor that regulates immunity and apoptosis; however, it is unclear whether the increased expression of phosphorylated STAT3 (p-STAT3) observed in chronic stress is related to splenocyte apoptosis. To explore the relationship between splenocyte apoptosis and STAT3 in chronic stress, we treated rats undergoing a 21-day chronic restraint stress program with the STAT3 inhibitor S3I-201. This chronic stress model was verified by observing rats' behavior and measuring their serum corticosterone levels. Chronic stress led to increased expression of anti-inflammatory cytokines, and p-STAT3 inhibition enhanced splenocyte apoptosis in chronic stress. We detected key proteins in three apoptotic pathways to determine which pathway mediated increasing splenocyte apoptosis and found that the death receptor pathway was the main apoptotic pathway that occurred in the spleen during chronic stress. The unfolded protein response (UPR) was also activated, but the Bcl-2 family was not involved in chronic stress. P-STAT3 inhibition had no influence on the Bcl-2 family and the death receptor pathway; however, p-STAT3 inhibition disrupted the pro-survival function of the UPR by decreasing the expression of ATF6α and p-IRE1α. Furthermore, p-STAT3 inhibition activated endoplasmic reticulum stress by promoting the expression of CHOP, p-JNK, and procaspase-12. Collectively, these findings indicate that the increased p-STAT3 expression during chronic stress may promote splenocyte survival by activating the UPR. Consequently, STAT3 and the UPR may be considered as potential therapeutic targets for chronic stress in the future.

20.
Front Pharmacol ; 11: 790, 2020.
Article in English | MEDLINE | ID: mdl-32670056

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) is a severe complication of sepsis; however, no effective drugs have been found. Activation of the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome is a major pathogenic mechanism of AKI induced by lipopolysaccharide (LPS). Autophagy, a process of intracellular degradation related to renal homeostasis, effectively restricts inflammatory responses. Herein, we explored the potential protective mechanisms of dexmedetomidine (DEX), which has confirmed anti-inflammatory effects, on LPS-induced AKI. METHODS: AKI was induced in rats by injecting 10 mg/kg of LPS intraperitoneally (i.p.). Wistar rats received intraperitoneal injections of DEX (30 µg/kg) 30 min before an intraperitoneal injection of LPS. Atipamezole (ATI) (250 µg/kg) and 3-methyladenine (3-MA) (15 mg/kg) were intraperitoneally injected 30 min before the DEX injection. RESULTS: DEX significantly attenuated renal injury. Furthermore, DEX decreased activation of the NLRP3 inflammasome and expression of interleukins 1ß and 18. In addition, autophagy-related protein and gene analysis indicated that DEX could significantly enhance autophagy. Finally, we verified the pharmacological effects of DEX on the 5'-adenosine monophosphate-activated protein kinase (AMPK)/mechanistic target of rapamycin (mTOR) pathway. Atip and 3-MA significantly reversed the protective effects of DEX. CONCLUSIONS: Our results suggest that the protective effects of DEX were mediated by enhanced autophagy via the α2-adrenoreceptor/AMPK/mTOR pathway, which decreased activation of the NLRP3 inflammasome. Above all, we verified the renal protective effects of DEX and offer a new treatment strategy for AKI.

SELECTION OF CITATIONS
SEARCH DETAIL
...