Probenecid relieves LPS-induced neuroinflammation by modulating the NLRP1 inflammasome pathway in BV2 cells
Probenecid relieves LPS-induced neuroinflammation by modulating the NLRP1 inflammasome pathway in BV2 cells
35
- 10.1186/s12868-017-0399-3
- Dec 1, 2017
- BMC Neuroscience
126
- 10.1007/s12012-011-9145-8
- Sep 22, 2011
- Cardiovascular Toxicology
45
- 10.1016/j.intimp.2022.108913
- Jun 1, 2022
- International Immunopharmacology
36
- 10.4103/1673-5374.346468
- Jun 2, 2022
- Neural Regeneration Research
330
- 10.1523/jneurosci.0157-08.2008
- Mar 26, 2008
- The Journal of Neuroscience
205
- 10.1128/iai.69.4.2045-2053.2001
- Apr 1, 2001
- Infection and Immunity
23
- 10.1002/dneu.22253
- Dec 6, 2014
- Developmental Neurobiology
27
- 10.3109/10715762.2016.1167277
- Apr 25, 2016
- Free Radical Research
611
- 10.15252/emmm.201810248
- Apr 23, 2019
- EMBO Molecular Medicine
66
- 10.3389/fnmol.2016.00019
- Mar 8, 2016
- Frontiers in Molecular Neuroscience
- Research Article
- 10.3760/cma.j.issn.0412-4081.2020.01.010
- Jan 11, 2020
- Chinese journal of ophthalmology
Objective: To investigate the role and mechanism of microglial activation in the process of retinal ganglion cell (RGC) death in the oxygen-glucose deprivation/reperfusion (OGD/R) model which mimicked retinal ischemia/reperfusion injury in vitro. Methods: Experimental study. Primary RGCs from C57BL/6 mice and BV2 microglia were co-cultured or cultured alone. The OGD/R model was established in vitro (reoxygenation time was set to 6 h, 24 h, 36 h, 48 h). BV2 microglial activation was assessed by immunofluorescence staining of ionized calcium binding adapter molecule 1 (iba1), and the survival rate of RGCs was detected by the Cell Counting Kit-8. The apoptosis rate of RGC was detected by using apoptosis detection kit. The levels of Toll-like receptor-4 (TLR4) and Nod-like receptor family pyrin domain containing protein 3 (NLRP3) in BV2 cells were detected by PCR, Western-blot and immunofluorescence staining. The activity of caspase-8 in BV2 cells was detected by the CaspGLOW Kit, and the content of interleukine-1β (IL-1β) in the supernatant was detected by enzyme linked immunosorbent assay. After the corresponding pathways were blocked by TLR4 small interfering RNA (siRNA) transfection or caspase-8 inhibitor, the expression changes of TLR4 and NLRP3, the activity of caspase-8, and the difference of IL-1β content could be observed as well as the activity of RGCs co-cultured with BV2. Statistical analysis was performed using analysis of variance. Results: Under co-culture of RGC and BV2 cells, cellular immunofluorescence detection showed that the expression of iba1 in BV2 cells increased, which indicated BV2 cells were activated significantly in the OGD/R model. In the OGD/R model, the apoptosis rate of RGC co-cultured with BV2 cells (71.1%±3.2%) was significantly higher than that of RGC cultured alone (35.1%±1.8%) (t=10.10, P<0.01). Cellular immunofluorescence detection showed that the expression of TLR4 and NLRP3 in BV2 cells in the OGD/R model increased significantly when BV2 cells were cultured alone, and their mRNA levels increased significantly with prolongation of reoxygenation time (F=64.45, 72.74; P<0.01), and peaked at OGD/R 24 h (TLR4 mRNA, relative ratio to control was 2.83±0.23; NLRP3 mRNA, relative ratio to control was 3.12±0.27). Caspase-8 activity also increased with prolonged reoxygenation time, the difference was statistically significant (F=93.57, P<0.01), and peaked at OGD/R 24 h (relative ratio to control was 2.92±0.31). After transfection of BV2 cells with TLR4 siRNA, its caspase-8 activity was significantly inhibited, but using caspase-8 inhibitor did not affect the up-regulation of TLR4 expression in BV2 cells. However, the mature IL-1β secreted by BV2 cells exposed to OGD/R was significantly reduced by using caspase-8 inhibitor (from 3.52±0.55 to 1.39±0.37, t=7.19, P<0.01), meanwhile, the expression of NLRP3 was also significantly decreased after caspase-8 inhibitor pretreatment (from 2.79±0.23 to 1.37±0.19, t=9.37, P<0.01). In the OGD/R model, the activity of RGC cells co-cultured with TLR4 siRNA-transfected BV2 cells was 74.5%±1.2%, and the activity of RGC cells co-cultured with BV2 cells treated with caspase-8 inhibitor was 62.8%±1.5%, those were both higher than that of RGC cells co-cultured with untreated BV2 cells (36.7%±0.3%), and the difference was statistically significant (t=11.60, 6.83; both P<0.01). Conclusion: TLR4-caspase-8-NLRP3 inflammasome pathway is activated in microglia exposed to OGD/R, resulting in the production of IL-1β, thereby contributing to the death of RGCs. (Chin J Ophthalmol, 2020, 56: 32-40).
- Research Article
3
- 10.1016/j.tox.2024.153730
- Jan 17, 2024
- Toxicology
Autophagy alleviates hippocampal neuroinflammation by inhibiting the NLRP3 inflammasome in a juvenile rat model exposed particulate matter
- Research Article
18
- 10.1007/s12640-021-00459-2
- Jan 11, 2022
- Neurotoxicity Research
Triggering receptor expressed on myeloid cells 2 (TREM2) plays a crucial role in modulating microglial-mediated neuroinflammation. The NAD-dependent deacetylase protein Sirtuin 3 (SIRT3) regulates mitochondrial oxidative stress response and neuroinflammation. TREM2 deficiency impairs the denovo synthesis pathway of NAD+. Therefore, the aim of this study was to investigate the potential role of TREM2 and SIRT3 in LPS-induced oxidative stress and neuroinflammation in BV2 cells. Lentivirus vector-mediated TREM2 overexpression (TREM2-OE) and corresponding negative control vector (TREM2-NC) were synthesized. BV2 cells were treated with LPS and/or TREM2-OE. 3-TYP, a selective SIRT3 inhibitor, was applied to determine the role of SIRT3 in the anti-oxidant and anti-inflammatory effects of TREM2. TREM2, SIRT3, NLRP3 inflammasome, caspase-1, postsynaptic density-95 (PSD-95), and brain derived neurotrophic factor (BDNF) were measured by Western blot analysis. Superoxide dismutase (SOD) was tested by SOD Assay Kit. Reactive oxygen species (ROS) expression was examined by immunofluorescence. Interleukin 1β (IL-1β) was determined by ELISA. Contents of NAD+ and NADH were detected by WST-8 method. LPS (1ug/ml for 24h) significantly decreased TREM2 expression at both RNA and protein levels (p < 0.01 and p < 0.05, respectively). Lower levels of SIRT3 protein and NAD+ were also detected following LPS stimulation (p < 0.05 and p < 0.05, respectively). LPS significantly enhanced ROS, NLRP3, caspase-1, and IL-1β expression (p < 0.01, p < 0.05, p < 0.05, and p < 0.01, respectively). PSD-95 and BDNF expression were decreased triggered by LPS (p < 0.05 and p < 0.05, respectively). TREM2 overexpression enhanced NAD+ and SIRT3 protein expression following LPS challenge in BV2 cells (p < 0.01 and p < 0.05, respectively). TREM2 alleviated LPS-induced oxidative stress and neuroinflammation (p < 0.01 and p < 0.05, respectively). Similarly, TREM2 overexpression upregulated PSD-95 and BDNF expression (p < 0.05 and p < 0.05, respectively). The anti-oxidant and anti-inflammatory effects of TREM2 were partially abrogated by SIRT3 antagonist 3-TYP (p < 0.05 and p < 0.05, respectively). Similarly, selective SIRT3 inhibition also partially abrogated TREM2-induced BDNF protein upregulation (p < 0.05) but failed to influence PSD-95 protein expression following LPS stimulation. LPS induces oxidative stress and neuroinflammation in BV2 cells, which may be mediated in part by the downregulation of TREM2 and SIRT3. TREM2 overexpression ameliorates LPS-induced oxidative stress and neuroinflammation through enhancing SIRT3 function via NAD+.
- Research Article
10
- 10.3892/etm.2018.6590
- Aug 9, 2018
- Experimental and therapeutic medicine
Neurosurgery always results in neuroinflammation, which may activate microglial cells. Previous studies have demonstrated that fentanyl could be used for the induction or maintenance of anesthesia prior to surgery. However, it is unknown if fentanyl attenuates neuroinflammation prophylactically. Cell viability in groups that were treated with different concentrations of fentanyl (0.01, 0.1, 1 or 5 µmol/l) was analyzed by an MTT assay. BV-2 microglial cells were treated with lipopolysaccharide (LPS) at a concentration of 1 µg/ml to mimic neuroinflammation in vitro. BV-2 cells were pretreated with 5 µmol/l fentanyl prior to stimulation by LPS. The protein levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-10 in the culture medium were assessed by ELISA. The mRNA level of toll-like receptor (TLR)4 was evaluated by reverse transcription-quantitative polymerase chain reaction analysis. The protein levels of TLR4, glycogen synthase kinase (GSK)-3β and phosphorylated (p)-GSK-3β in BV-2 cells were assessed by western blot analysis. The MTT assay demonstrated that low concentrations of fentanyl (0.01, 0.1 or 1 µmol/l) did not affect the cell viability of BV-2 cells, while 5 µmol/l fentanyl significantly reduced BV-2 cell viability. The results of ELISA revealed that LPS significantly upregulated the release of TNF-α, IL-1β and IL-10, which were repressed by fentanyl pretreatment. Fentanyl pretreatment significantly reduced the LPS-induced elevation of TLR4 at mRNA and protein levels as well as p-GSK-3β protein levels in BV-2 cells. In conclusion, fentanyl pretreatment protects BV-2 cells from LPS-induced neuroinflammation by inhibiting TLR4 expression and GSK-3β activation. Neuroinflammation induced by surgery serves an important role in the development of postoperative cognitive dysfunction (POCD) and targeting the TLR4 and GSK-3β signaling pathway may provide a novel therapeutic approach for the treatment of POCD.
- Research Article
23
- 10.1016/j.abb.2021.108918
- May 13, 2021
- Archives of Biochemistry and Biophysics
Tripartite-motif protein 21 knockdown extenuates LPS-triggered neurotoxicity by inhibiting microglial M1 polarization via suppressing NF-κB-mediated NLRP3 inflammasome activation
- Research Article
1
- 10.1016/j.brainresbull.2024.111157
- Jan 1, 2025
- Brain Research Bulletin
USP22 Inhibits Microglial M1 Polarization by Regulating the PU.1/NLRP3 Inflammasome Pathway
- Research Article
7
- 10.1016/j.jep.2023.117686
- Dec 29, 2023
- Journal of Ethnopharmacology
Huang-Lian-Jie-Du decoction drug-containing serum inhibits IL-1β secretion from D-glucose and PA induced BV2 cells via autophagy/NLRP3 signaling
- Research Article
1
- 10.3881/j.issn.1000-503x.10847
- Apr 28, 2019
- Zhongguo yi xue ke xue yuan xue bao. Acta Academiae Medicinae Sinicae
Objective To explore the effects of cathepsin B(CTSB)on the activation of nucleotide-binding domain and leucine-rich-repeat-containing family and pyrin domain-containing 3(NLRP3)inflammasome via transient receptor potential mucolipin-1(TRPML1)in cell oxidative stress model and specific gene silencing cell model. Methods BV2 cells cultured in vivo were treated separately or simultaneously with hydrogen peroxide(H2O2),calcium-sensitive receptor agonist gadolinium trichloride(GdCl3),and CTSB inhibitor CA-074Me,and interleukin-1(IL-1)beta and caspase-1 protein were detected by enzyme-linked immunosorbent assay.The growth activity of BV2 cells in each group was measured by MTT.BV2 cells were treated with different concentrations of H2O2.Cystatin C mRNA and TRPML1 mRNA in BV2 cells were detected by real-time quantitative polymerase chain reaction and the proteins of TRPML1,CTSB,cathepsin D(CTSD),cathepsin L(CTSL)and cathepsin V(CTSV)were detected by Western blot.Specific small interfering RNA was designed for TRPML1 gene target sequence.TRPML1 gene silencing cell lines(named Tr-si-Bv2 cells)were established in BV2 cells and treated with or without H2O2.TRPML1,CTSB and transcription factor EB(TFEB)proteins in Tr-si-Bv2 cells or control cells were detected by Western blot. Results After treatment with H2O2,the expression of caspase-1 protein and NLRP3 mRNA in BV2 cells was increased,and IL-1beta protein in BV2 cells was significantly increased after treatment with GdCl3(P=0.0036).After treatment with CA-074Me,the doses of NLRP3 mRNA(P=0.037),caspase-1(P=0.021),and IL-1β(P= 0.036)were significantly reduced.Cells in the H2O2 group and H2O2+GdCl3 group grew more slowly.The expressions of CTSB mRNA and TRPML1 mRNA,or CTSB and TRPML1 proteins in BV2 cells in the treatment group with 200 μmol/L of H2O2 concentration were similar.H2O2-induced CTSB protein expression was inhibited after silencing TRPML1 gene.The changes of other cathepsins were not affected for the different concentration of H2O2.In the BV2 cells treated with TRPML1 gene silencing,the expression of CTSB protein was significantly reduced and the difference was statistically significant(P=0.021)between the H2O2 +siRNA treatment group and the H2O2 treatment group.<b>Conclusion</b> CTSB regulates the activation of NLRP3 inflammasome in the oxidative stress model of microglia cells,probably mediated by calcium channel protein TRPML1.
- Research Article
12
- 10.2174/1381612827666210716104606
- Sep 9, 2021
- Current pharmaceutical design
Hyperglycemia-induced microglia activation can cause a continuous release of proinflammatory cytokines, which gradually damages neurons and contributes to central diabetic neuroinflammation. This study aimed to illustrate the possible mechanism related to NLRP3 inflammasome and the aggravation of diabetes neuroinflammation. The targeted proteins from BV2 cells and brain tissues were tested by Western blot or immunohistochemistry. Cytokines from cell supernatant and serum were detected by ELISA. Meanwhile, cytoplasm and mitochondria ROS were determined by DCFHDA and Mito sox Red, respectively. In vitro, BV2 cells were stimulated by different glucose concentrations (5.5 to 65 mM/L) above physiological values and maintained for different periods (12 to 48h). The proinflammatory cytokines IL-1β, IL18, IL6, TNFα and cytoplasm ROS were significantly increased in a dose-dependent manner, while mitochondrial ROS was unaffected. NLRP3 inflammasomes, MAPKs, and NF-κB pathways were obviously activated at the concentration of 35 mM/L for 12h. Inhibition assay using specific inhibitors indicated that the treatment of glucose (35 mM/L for 12h) could stimulate NLRP3 inflammasome activation via ROS/JNK MAPKs/NF-κB pathway. In STZ induced diabetes mice models, microglia NLRP3, ASC, and caspase-1 proteins were highly expressed, and serum cytokines IL-1β, IL6, IL18, and TNFα were remarkably increased. Microglia NLRP3 inflammasomes activation involves diabetic neuroinflammation in diabetic mice and BV2 cells via ROS/JNK MAPKs/NF-κB pathways.
- Research Article
- 10.5607/en25022
- Oct 31, 2025
- Experimental Neurobiology
Parkinson’s disease (PD) is a neurodegenerative disorder associated with neuroinflammation and gut dysfunction. The G protein-coupled estrogen receptor (GPER) has showed therapeutic potential in inflammatory bowel diseases (IBD), yet its role and underlying mechanisms in PD remain unclear. Here, we aimed to investigate the role and mechanisms of GPER in protecting PD. Female mice underwent bilateral ovariectomies (OVX) and were treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to induce PD, followed by administration of GPER agonist G1. The expressions of tyrosine hydroxylase (TH) and α-synuclein (α-syn), as well as activations of inflammatory cells and NLRP3 inflammasome in the brain and ileum were evaluated. BV2 cells were pretreated with G1 and/or the antagonist G15, then treated with LPS and ATP to activate NLRP3 inflammasome. Activation of NLRP3 inflammasome in BV2 cells was assessed. Results demonstrated that G1 treatment increased TH expression, reduced α-syn expression, and suppressed inflammation and NLRP3 inflammasome in both the midbrain and ileum of MPTP-treated OVX mice. Pretreatment with G1 suppressed the activation of NLRP3 inflammasome in BV2 cells, while the effect was reversed by G15. These findings indicate that GPER activation exerts a protective effect in MPTP-induced OVX mice by modulating NLRP3 inflammasome in both brain and gut, which might provide novel insights into the pathogenesis and therapy of PD.
- Research Article
60
- 10.1016/j.biopha.2020.110575
- Aug 5, 2020
- Biomedicine & Pharmacotherapy
Lychee seed polyphenol inhibits Aβ-induced activation of NLRP3 inflammasome via the LRP1/AMPK mediated autophagy induction
- Research Article
3
- 10.1002/kjm2.12670
- Mar 28, 2023
- The Kaohsiung Journal of Medical Sciences
miR‐29a‐3p promotes the regulatory role of eicosapentaenoic acid in the NLRP3 inflammasome and autophagy in microglial cells
- Research Article
13
- 10.1007/s11064-021-03441-8
- Sep 28, 2021
- Neurochemical Research
Zinc has reported to play a neuroprotective role in the development of spinal cord injury (SCI). The protective mechanism of zinc remains to be uncovered. The aim of the current study was to investigate the neuroprotective mechanism of zinc in the progression of SCI. The C57BL/6J mouse SCI model was established to confirm the protective role of zinc in vivo, while the cellular model was induced in mouse microglial BV2 cells by using lipopolysaccharide (LPS). The expression levels of XIST, miR-374a-5p and NLRP3 inflammasome as well as the autophagy-related proteins were detected using real-time PCR and immunoblotting. Cell viability was assessed by CCK-8 assay. Apoptosis was evaluated by TUNEL staining, flow cytometry, the determination of apoptosis-related proteins. The target relationship was confirmed by luciferase reporter assays. Zinc improved locomotor function in SCI mice and alleviated LPS-induced BV2 cell injuries by inhibiting apoptosis and initiating autophagy processes. XIST and NLRP3 inflammasome was upregulated while miR-374a-5p was downregulated in spinal cords of SCI mice and LPS-treated BV2 cells. All these effects were inhibited by Zinc treatment. XIST knockdown triggered microglial autophagy-mediated NLRP3 inactivation in LPS-induced BV2 cells by regulating miR-374a-5p. Zinc treatment protected BV2 cells from LPS-induced cell injury by the downregulation of XIST. This process might be through autophagy‑mediated NLRP3 inflammasome inactivation by targeting miR-374a-5p. Zinc downregulates XIST and induces neuroprotective effects against SCI by promoting microglial autophagy-induced NLRP3 inflammasome inactivation through regulating miR-374a-5p. Our finding provides novel opportunities for the understanding of zinc-related therapy of SCI.
- Research Article
18
- 10.1111/1440-1681.13052
- Jan 8, 2019
- Clinical and Experimental Pharmacology and Physiology
Parkinson's disease (PD) is a progressive neurodegenerative disorder pathologically characterized by the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). Chronic neuroinflammation is one of the hallmarks of PD pathophysiology. Cathepsin D (CathD), a soluble aspartic protease, has been reported to play an important role in neurodegenerative diseases such as PD. This research focuses on the role of CathD and the molecular mechanisms involved in the process of neuroinflammation and neurotoxicity. We use 1-methyl-4phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-challenged mice and lipopolysaccharide (LPS)-induced murine microglia BV2 cells as the in vivo and in vitro models, respectively. The effect of CathD on the neuroinflammation, cytotoxicity and the underlying mechanisms associated with NF-κB signalling pathway are investigated. Data showed that MPTP induces motor deficit, inflammation and depletion of dopaminergic neurons in PD model mice. Notably, cathD was overexpressed in the SNpc of MPTP-induced PD mice and was highly expressing in LPS-stimulated primary microglial cells and BV-2 cells. Furthermore, knockdown of CathD with lentiviral transduction inhibited LPS-induced neuroinflammation through inhibition of NF-κB signalling pathway primarily by regulating the NF-κB p65 nuclear translocation both in BV-2 and primary microglial cells. Additionally, knockdown of CathD protected the activated-microglia induced dopaminergic neurons MN9D cells from neurotoxicity as well as apoptosis. Our findings bring a new insight into understanding the complex mechanisms underlying the pathogenesis of PD and provide a novel target to attenuate the excessive neuroinflammatory responses in the treatment of PD.
- Research Article
7
- 10.1016/j.cyto.2024.156677
- Jun 18, 2024
- Cytokine
The ROS/TXNIP/NLRP3 pathway mediates LPS-induced microglial inflammatory response
- New
- Research Article
- 10.1038/s41598-025-22646-3
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22635-6
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22867-6
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22758-w
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22854-x
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22866-7
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22895-2
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22651-6
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22678-9
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22664-1
- Nov 6, 2025
- Scientific Reports
- Ask R Discovery
- Chat PDF
AI summaries and top papers from 250M+ research sources.