1.Research progress of signal pathways of microglia activation in sleep disorders.
Zhi-Jun SHU ; Quan-Yi ZHANG ; Yi-Peng XU ; Zheng-Yu ZHAO
Acta Physiologica Sinica 2023;75(4):569-574
Sleep is an extremely important physiological state to maintain human life. Sleep disorders can not only cause anxiety and depression, but also induce multi-system diseases that seriously affect brain function and physical health. The neuroinflammation is a key pathological process after sleep disorders, which can induce a series of nervous system diseases. In recent years, the role of microglia activation in neuroinflammation has been paid more and more attention and become a research hotspot in this field. The imbalance of the central microenvironment after sleep disorders leads to changes in the activation and polarization of microglia, which triggers neuroinflammatory response. The activation and polarization of microglia in the sleep disorders are regulated by multiple signaling pathways and complex molecular mechanisms. This paper summarizes five signaling pathways of microglia activation in central inflammation induced by sleep disorders, including P2X7 receptor (P2X7R), p38MAPK, Toll-like receptor 4 (TLR4)/NF-κB, JAK/STAT, and α7 nicotinic acetylcholine receptor (α7-nAChR) pathways, in order to provide reference for further research and clinical treatment targets selection of sleep disorders.
Humans
;
Neuroinflammatory Diseases
;
Microglia/metabolism*
;
Signal Transduction/physiology*
;
NF-kappa B/metabolism*
;
Inflammation/metabolism*
;
Sleep Wake Disorders/metabolism*
2.Leaky Gut Plays a Critical Role in the Pathophysiology of Autism in Mice by Activating the Lipopolysaccharide-Mediated Toll-Like Receptor 4-Myeloid Differentiation Factor 88-Nuclear Factor Kappa B Signaling Pathway.
Fang LI ; Haoran KE ; Siqi WANG ; Wei MAO ; Cexiong FU ; Xi CHEN ; Qingqing FU ; Xiaori QIN ; Yonghua HUANG ; Bidan LI ; Shibing LI ; Jingying XING ; Minhui WANG ; Wenlin DENG
Neuroscience Bulletin 2023;39(6):911-928
Increased intestinal barrier permeability, leaky gut, has been reported in patients with autism. However, its contribution to the development of autism has not been determined. We selected dextran sulfate sodium (DSS) to disrupt and metformin to repair the intestinal barrier in BTBR T+tf/J autistic mice to test this hypothesis. DSS treatment resulted in a decreased affinity for social proximity; however, autistic behaviors in mice were improved after the administration of metformin. We found an increased affinity for social proximity/social memory and decreased repetitive and anxiety-related behaviors. The concentration of lipopolysaccharides in blood decreased after the administration of metformin. The expression levels of the key molecules in the toll-like receptor 4 (TLR4)-myeloid differentiation factor 88 (MyD88)-nuclear factor kappa B (NF-κB) pathway and their downstream inflammatory cytokines in the cerebral cortex were both repressed. Thus, "leaky gut" could be a trigger for the development of autism via activation of the lipopolysaccharide-mediated TLR4-MyD88-NF-κB pathway.
Mice
;
Animals
;
NF-kappa B
;
Myeloid Differentiation Factor 88/metabolism*
;
Lipopolysaccharides/pharmacology*
;
Toll-Like Receptor 4/metabolism*
;
Autistic Disorder/metabolism*
;
Signal Transduction/physiology*
3.Activation of hypoxia-inducible factor 1 attenuates periapical inflammation and bone loss.
Kimito HIRAI ; Hisako FURUSHO ; Kiichi HIROTA ; Hajime SASAKI
International Journal of Oral Science 2018;10(2):12-12
Hypoxia (low oxygen level) is an important feature during infections and affects the host defence mechanisms. The host has evolved specific responses to address hypoxia, which are strongly dependent on the activation of hypoxia-inducible factor 1 (HIF-1). Hypoxia interferes degradation of HIF-1 alpha subunit (HIF-1α), leading to stabilisation of HIF-1α, heterodimerization with HIF-1 beta subunit (HIF-1β) and subsequent activation of HIF-1 pathway. Apical periodontitis (periapical lesion) is a consequence of endodontic infection and ultimately results in destruction of tooth-supporting tissue, including alveolar bone. Thus far, the role of HIF-1 in periapical lesions has not been systematically examined. In the present study, we determined the role of HIF-1 in a well-characterised mouse periapical lesion model using two HIF-1α-activating strategies, dimethyloxalylglycine (DMOG) and adenovirus-induced constitutively active HIF-1α (CA-HIF1A). Both DMOG and CA-HIF1A attenuated periapical inflammation and tissue destruction. The attenuation in vivo was associated with downregulation of nuclear factor-κappa B (NF-κB) and osteoclastic gene expressions. These two agents also suppressed NF-κB activation and subsequent production of proinflammatory cytokines by macrophages. Furthermore, activation of HIF-1α by DMOG specifically suppressed lipopolysaccharide-stimulated macrophage differentiation into M1 cells, increasing the ratio of M2 macrophages against M1 cells. Taken together, our data indicated that activation of HIF-1 plays a protective role in the development of apical periodontitis via downregulation of NF-κB, proinflammatory cytokines, M1 macrophages and osteoclastogenesis.
Alveolar Bone Loss
;
metabolism
;
prevention & control
;
Amino Acids, Dicarboxylic
;
pharmacology
;
Animals
;
Cytokines
;
metabolism
;
Down-Regulation
;
Gene Expression
;
drug effects
;
Hypoxia-Inducible Factor 1, alpha Subunit
;
physiology
;
Macrophages
;
physiology
;
Mice
;
NF-kappa B
;
metabolism
;
Osteogenesis
;
physiology
;
Periapical Periodontitis
;
metabolism
;
prevention & control
;
Real-Time Polymerase Chain Reaction
;
X-Ray Microtomography
4.Nucleocapsid protein from porcine epidemic diarrhea virus isolates can antagonize interferon-λ production by blocking the nuclear factor-κB nuclear translocation.
Ying SHAN ; Zi-Qi LIU ; Guo-Wei LI ; Cong CHEN ; Hao LUO ; Ya-Jie LIU ; Xun-Hui ZHUO ; Xing-Fen SHI ; Wei-Huan FANG ; Xiao-Liang LI
Journal of Zhejiang University. Science. B 2018;19(7):570-580
Porcine epidemic diarrhea virus (PEDV) is a highly infectious pathogen that can cause severe diseases in pigs and result in enormous economic losses in the worldwide swine industry. Previous studies revealed that PEDV exhibits an obvious capacity for modulating interferon (IFN) signaling or expression. The newly discovered type III IFN, which plays a crucial role in antiviral immunity, has strong antiviral activity against PEDV proliferation in IPEC-J2 cells. In this study, we aimed to investigate the effect of PEDV nucleocapsid (N) protein on type III IFN-λ. We found that the N proteins of ten PEDV strains isolated between 2013 and 2017 from different local farms shared high nucleotide identities, while the N protein of the CV777 vaccine strain formed a monophyletic branch in the phylogenetic tree. The N protein of the epidemic strain could antagonize type III IFN, but not type I or type II IFN expression induced by polyinosinic-polycytidylic acid (poly(I:C)) in IPEC-J2 cells. Subsequently, we demonstrated that the inhibition of poly(I:C)-induced IFN-λ3 production by PEDV N protein was dependent on the blocking of nuclear factor-κB (NF-κB) nuclear translocation. These findings might help increase understanding of the pathogenesis of PEDV and its mechanisms for evading the host immune response.
Active Transport, Cell Nucleus
;
Animals
;
Coronavirus Infections
;
immunology
;
veterinary
;
virology
;
Genes, Viral
;
Host-Pathogen Interactions
;
immunology
;
Interferons
;
antagonists & inhibitors
;
biosynthesis
;
genetics
;
Interleukins
;
antagonists & inhibitors
;
biosynthesis
;
genetics
;
NF-kappa B
;
metabolism
;
Nucleocapsid Proteins
;
genetics
;
immunology
;
physiology
;
Porcine epidemic diarrhea virus
;
genetics
;
pathogenicity
;
physiology
;
Promoter Regions, Genetic
;
Swine
;
Swine Diseases
;
immunology
;
virology
5.Effect of Bifidobacterium on the expression of β-defensin-2 in intestinal tissue of neonatal rats with necrotizing enterocolitis.
Wei-Cheng LU ; Xu ZHENG ; Jin-Fu LIU ; Wen-Chuan WU ; Xing-Yue CHEN ; Hai-Bo WEI ; Chun-Lei LI ; Ming-Jing LIN
Chinese Journal of Contemporary Pediatrics 2018;20(3):224-229
<b>OBJECTIVEb>To study the effect of Bifidobacterium on the expression of β-defensin-2 (BD-2) in intestinal tissue of neonatal rats with necrotizing enterocolitis (NEC).
<b>METHODSb>A total of 40 rats were randomly divided into four groups: normal control, Bifidobacterium control, NEC model, and Bifidobacterium treatment, with 10 rats in each group. A rat model of NEC was induced by hypoxia, cold stimulation, and artificial feeding. The rats in the Bifidobacterium control and Bifidobacterium treatment groups were given Bifidobacterium via the gastric tube after cold stimulation once a day for three consecutive days. The morphological changes of the terminal ileum were observed under a light microscope and the intestinal injury score was determined. Immunohistochemistry and qRT-PCR were used to measure the protein and mRNA expression of BD-2 in the ileal mucosal tissue.
<b>RESULTSb>The NEC model group had a significantly higher intestinal injury score than the normal control, Bifidobacterium control, and Bifidobacterium treatment groups (P<0.05). The Bifidobacterium treatment group had a significantly higher intestinal injury score than the normal control and Bifidobacterium control groups (P<0.05). The mRNA and protein expression of BD-2 in the normal control group was significantly lower than in the Bifidobacterium control, NEC model, and Bifidobacterium treatment groups (P<0.05). The Bifidobacterium control group had significantly higher mRNA and protein expression of BD-2 than the NEC model and Bifidobacterium treatment groups (P<0.05). The Bifidobacterium treatment group had significantly higher mRNA and protein expression of BD-2 than the NEC model group (P<0.05).
<b>CONCLUSIONSb>Bifidobacterium can induce the expression of BD-2 in intestinal tissue of rats and reduce inflammatory response by increasing the expression of BD-2. This provides a protective effect on neonatal rats with NEC.
Animals ; Bifidobacterium ; Disease Models, Animal ; Enterocolitis, Necrotizing ; therapy ; Humans ; Infant, Newborn ; Intestinal Mucosa ; metabolism ; NF-kappa B ; physiology ; Rats ; Rats, Sprague-Dawley ; Signal Transduction ; physiology ; beta-Defensins ; analysis ; genetics ; physiology
6.Lipopolysaccharide Stimulates Surfactant Protein-A in Human Renal Epithelial HK-2 Cells through Upregulating Toll-like Receptor 4 Dependent MEK1/2-ERK1/2-NF-κB Pathway.
Jiao LIU ; Guang LI ; Wen-Jie XIE ; Lu WANG ; Rui ZHANG ; Ke-Sheng HUANG ; Qing-Shan ZHOU ; De-Chang CHEN
Chinese Medical Journal 2017;130(10):1236-1243
<b>BACKGROUNDb>Surfactant protein-A (SP-A) contributes to the regulation of sepsis-induced acute kidney injury. In a previous study, we demonstrated that the expression of SP-A in the human renal tubular epithelial (HK-2) cells can be stimulated by lipopolysaccharide (LPS). The present study evaluated the possible signal-transducing mechanisms of LPS-induced SP-A biosynthesis in the HK-2 cells.
<b>METHODSb>Tetrazolium salt colorimetry (MTT) assay was used to detect cell viability of HK-2 cells after LPS stimulation on different time points. HK-2 cells were stimulated with 100 ng/ml of LPS for different durations to determine the effects of LPS on SP-A and toll-like receptor 4 (TLR4) messenger RNA (mRNA) expression, as well as phosphorylation of mitogen-activated/extracellular signal-regulated kinase (MEK) 1, extracellular signal-regulated kinase 1/2 (ERK1/2), p38 mitogen-activated protein kinase (p38MAPK), and nuclear factor-kappa B (NF-κB) inhibitor-alpha (IkB-α). Then, HK-2 cells were pretreated with CLI-095, a TLR4 inhibitor, to analyze mRNA and protein levels of SP-A and TLR4 and expression of NF-κB in the cytoplasm and nucleus of HK-2 before LPS exposure.
<b>RESULTSb>HK-2 cells exposed to 100 ng/ml of LPS for 1, 6, and 24 h did not affect cell viability which showed no toxic effect of 100 ng/ml LPS on cells (P = 0.16); however, the biosynthesis of SP-A mRNA and protein in HK-2 cells was significantly increased (P = 0.02). As to the mechanism, LPS enhanced transmembrane receptor TLR4 protein expression. Sequentially, LPS time dependently augmented phosphorylation of MEK1, ERK1/2, and p38MAPK. In addition, levels of phosphorylated IκB-α and nuclear NF-κB were augmented with LPS exposure for 2 h. LPS-induced SP-A and TLR4 mRNA as well as NF-κB expression were significantly inhibited by pretreatment with CLI-095.
<b>CONCLUSIONSb>The present study exhibited that LPS can increase SP-A synthesis in human renal epithelial cells through sequentially activating the TLR4-related MEK1-ERK1/2-NF-κB-dependent pathway.
Cell Line ; Cell Survival ; drug effects ; physiology ; Colorimetry ; Humans ; Kidney ; cytology ; metabolism ; Lipopolysaccharides ; toxicity ; Mitogen-Activated Protein Kinase 1 ; metabolism ; Mitogen-Activated Protein Kinase 3 ; metabolism ; NF-kappa B ; metabolism ; Pulmonary Surfactant-Associated Protein A ; metabolism ; Sulfonamides ; pharmacology ; Tetrazolium Salts ; chemistry ; Toll-Like Receptor 4 ; antagonists & inhibitors ; metabolism
7.Hypaconitine inhibits TGF-β1-induced epithelial-mesenchymal transition and suppresses adhesion, migration, and invasion of lung cancer A549 cells.
Hai-Tao FENG ; Wen-Wen ZHAO ; Jin-Jian LU ; Yi-Tao WANG ; Xiu-Ping CHEN
Chinese Journal of Natural Medicines (English Ed.) 2017;15(6):427-435
Epithelial-mesenchymal transition (EMT) has been implicated in tumor invasion and metastasis and provides novel strategies for cancer therapy. Hypaconitine (HpA), a diester-diterpenoid alkaloid isolated from the root of the Aconitum species, exhibits anti-inflammatory, analgesic, and especially, cardiotoxic activities. Here, we reported the anti-metastatic potentials of HpA in transforming growth factor-β1 (TGF-β1)-induced EMT in lung cancer A549 cells. The cytotoxic effect of HpA was determined by MTT assay. A549 cells were treated with TGF-β1 with or without HpA co-treatment, and the morphological alterations were observed with a microscopy. The expression of E-cadherin, N-cadherin, and NF-κB was determined by both Western blotting and immunofluorescence analyses. The adhesion, migration, and invasion were detected with Matrigel, wound-healing, and transwell assays, respectively. The expression of Snail was determined by Western blotting. The expression of NF-κB p65, IκBα, and p-IκBα in nuclear and cytosolic extracts was assessed by Western blotting. The results showed that low concentration of HpA (<16 μmol·L) had no obvious cytotoxicity to A549 cells. Morphologically, TGF-β1 treatment induced spindle-shaped alteration in the cells. The upregulation of N-cadherin, NF-κB, and Snail and the downregulation of E-cadherin were detected after TGF-β1 treatment. The adhesion, migration and invasion abilities were also increased by TGF-β1. Besides, TGF-β1 induced expression of Snail in a time-dependent manner. Furthermore, TGF-β1 induced nuclear translocation of NF-κB p65. All these alterations were dramatically inhibited by HpA co-treatment. In addition, the NF-κB inhibitor PDTC showed similar inhibitory effect. In conclusion, these results showed that HpA inhibited TGF-β1-induced EMT in A549 cells, which was possibly mediated by the inactivation of the NF-κB signaling pathway, providing an evidence for anti-cancer effect of HpA.
A549 Cells
;
Aconitine
;
analogs & derivatives
;
pharmacology
;
Active Transport, Cell Nucleus
;
drug effects
;
Antineoplastic Agents, Phytogenic
;
pharmacology
;
Cadherins
;
analysis
;
Cell Adhesion
;
drug effects
;
Cell Movement
;
drug effects
;
Dose-Response Relationship, Drug
;
Epithelial-Mesenchymal Transition
;
drug effects
;
Humans
;
NF-kappa B
;
antagonists & inhibitors
;
metabolism
;
Neoplasm Invasiveness
;
Transforming Growth Factor beta1
;
antagonists & inhibitors
;
physiology
8.Activation of NF-κB and AP-1 Mediates Hyperproliferation by Inducing β-Catenin and c-Myc in Helicobacter pylori-Infected Gastric Epithelial Cells.
Eunyoung BYUN ; Bohye PARK ; Joo Weon LIM ; Hyeyoung KIM
Yonsei Medical Journal 2016;57(3):647-651
PURPOSE: In the gastric mucosa of Helicobacter pylori (H. pylori)-infected patients with gastritis or adenocarcinoma, proliferation of gastric epithelial cells is increased. Hyperproliferation is related to induction of oncogenes, such as β-catenin and c-myc. Even though transcription factors NF-κB and AP-1 are activated in H. pylori-infected cells, whether NF-κB or AP-1 regulates the expression of β-catenein or c-myc in H. pylori-infected cells has not been clarified. The present study was undertaken to investigate whether H. pylori-induced activation of NF-κB and AP-1 mediates the expression of oncogenes and hyperproliferation of gastric epithelial cells. MATERIALS AND METHODS: Gastric epithelial AGS cells were transiently transfected with mutant genes for IκBα (MAD3) and c-Jun (TAM67) or treated with a specific NF-κB inhibitor caffeic acid phenethyl ester (CAPE) or a selective AP-1 inhibitor SR-11302 to suppress activation of NF-κB or AP-1, respecively. As reference cells, the control vector pcDNA was transfected to the cells. Wild-type cells or transfected cells were cultured with or without H. pylori. RESULTS: H. pylori induced activation of NF-κB and AP-1, cell proliferation, and expression of oncogenes (β-catenein, c-myc) in AGS cells, which was inhibited by transfection of MAD3 and TAM67. Wild-type cells and the cells transfected with pcDNA showed similar activities of NF-κB and AP-1, proliferation, and oncogene expression regardless of treatment with H. pylori. Both CAPE and SR-11302 inhibited cell proliferation and expression of oncogenes in H. pylori-infected cells. CONCLUSION: H. pylori-induced activation of NF-κB and AP-1 regulates transcription of oncogenes and mediates hyperproliferation in gastric epithelial cells.
Blotting, Western
;
Caffeic Acids
;
Cell Line, Tumor
;
Cell Proliferation
;
DNA, Bacterial/analysis/genetics
;
DNA-Binding Proteins/*metabolism
;
Epithelial Cells/*metabolism
;
Gastric Mucosa/*metabolism/pathology
;
Gastritis/pathology
;
Gene Expression Regulation, Bacterial
;
Helicobacter Infections/metabolism/pathology/physiopathology
;
Helicobacter pylori/pathogenicity/physiology
;
Humans
;
NF-kappa B/antagonists & inhibitors/*biosynthesis/metabolism
;
Peptide Fragments
;
Phenylethyl Alcohol/analogs & derivatives
;
Proto-Oncogene Proteins c-jun
;
Repressor Proteins
;
Transcription Factor AP-1/*biosynthesis
;
Transcription Factors/*metabolism
;
beta Catenin/*metabolism
9.Effects of Low-dose Triamcinolone Acetonide on Rat Retinal Progenitor Cells under Hypoxia Condition.
Yao XING ; Li-Jun CUI ; Qian-Yan KANG
Chinese Medical Journal 2016;129(13):1600-1606
<b>BACKGROUNDb>Retinal degenerative diseases are the leading causes of blindness in developed world. Retinal progenitor cells (RPCs) play a key role in retina restoration. Triamcinolone acetonide (TA) is widely used for the treatment of retinal degenerative diseases. In this study, we investigated the role of TA on RPCs in hypoxia condition.
<b>METHODSb>RPCs were primary cultured and identified by immunofluorescence staining. Cells were cultured under normoxia, hypoxia 6 h, and hypoxia 6 h with TA treatment conditions. For the TA treatment groups, after being cultured under hypoxia condition for 6 h, RPCs were treated with different concentrations of TA for 48-72 h. Cell viability was measured by cell counting kit-8 (CCK-8) assay. Cell cycle was detected by flow cytometry. Western blotting was employed to examine the expression of cyclin D1, Akt, p-Akt, nuclear factor (NF)-κB p65, and caspase-3.
<b>RESULTSb>CCK-8 assays indicated that the viability of RPCs treated with 0.01 mg/ml TA in hypoxia group was improved after 48 h, comparing with control group (P < 0.05). After 72 h, the cell viability was enhanced in both 0.01 mg/ml and 0.02 mg/ml TA groups compared with control group (all P < 0.05). Flow cytometry revealed that there were more cells in S-phase in hypoxia 6 h group than in normoxia control group (P < 0.05). RPCs in S and G2/M phases decreased in groups given TA, comparing with other groups (all P < 0.05). There was no significant difference in the total Akt protein expression among different groups, whereas upregulation of p-Akt and NF-κB p65 protein expression and downregulation of caspase-3 and cyclin D1 protein expression were observed in 0.01 mg/ml TA group, comparing with hypoxia 6 h group and control group (all P < 0.05).
<b>CONCLUSIONb>Low-dose TA has anti-apoptosis effect on RPCs while it has no stimulatory effect on cell proliferation.
Animals ; Apoptosis ; drug effects ; physiology ; Caspase 3 ; metabolism ; Cell Cycle ; drug effects ; physiology ; Cell Hypoxia ; drug effects ; physiology ; Cell Proliferation ; drug effects ; physiology ; Cell Survival ; drug effects ; physiology ; Cells, Cultured ; Cyclin D1 ; metabolism ; NF-kappa B ; metabolism ; Proto-Oncogene Proteins c-akt ; metabolism ; Rats ; Rats, Sprague-Dawley ; Retina ; cytology ; Stem Cells ; cytology ; drug effects ; Triamcinolone Acetonide ; pharmacology
10.Celastrol targets IRAKs to block Toll-like receptor 4-mediated nuclear factor-κB activation.
Yu-fan SHEN ; Xue ZHANG ; Ying WANG ; Fan-fan CAO ; Georges UZAN ; Bin PENG ; Deng-hai ZHANG
Journal of Integrative Medicine 2016;14(3):203-208
<b>OBJECTIVEb>Celastrol has been established as a nuclear factor-κB (NF-κB) activation inhibitor; however, the exact mechanism behind this action is still unknown. Using text-mining technology, the authors predicted that interleukin-1 receptor-associated kinases (IRAKs) are potential celastrol targets, and hypothesized that targeting IRAKs might be one way that celastrol inhibits NF-κB. This is because IRAKs are key molecules for some crucial pathways to activate NF-κB (e.g., the interleukin-1 receptor (IL-1R)/Toll-like receptor (TLR) superfamily).
<b>METHODSb>The human hepatocellular cell line (HepG2) treated with palmitic acid (PA) was used as a model for stimulating TLR4/NF-κB activation, in order to observe the potential effects of celastrol in IRAK regulation and NF-κB inhibition. The transfection of small interfering RNA was used for down-regulating TLR4, IRAK1 and IRAK4, and the Western blot method was used to detect changes in the protein expressions.
<b>RESULTSb>The results showed that celastrol could effectively inhibit PA-caused TLR4-dependent NF-κB activation in the HepG2 cells; PA also activated IRAKs, which were inhibited by celastrol. Knocking down IRAKs abolished PA-caused NF-κB activation.
<b>CONCLUSIONb>The results for the first time show that targeting IRAKs is one way in which celastrol inhibits NF-κB activation.
Hep G2 Cells ; Humans ; Interleukin-1 Receptor-Associated Kinases ; antagonists & inhibitors ; NF-kappa B ; antagonists & inhibitors ; metabolism ; Phosphorylation ; Toll-Like Receptor 4 ; antagonists & inhibitors ; physiology ; Triterpenes ; pharmacology

Result Analysis
Print
Save
E-mail