ligustilide and Inflammation

ligustilide has been researched along with Inflammation* in 14 studies

Other Studies

14 other study(ies) available for ligustilide and Inflammation

ArticleYear
Mechanism of ligusticum cycloprolactam against neuroinflammation based on network pharmacology and experimental verification.
    Clinical and experimental pharmacology & physiology, 2023, Volume: 50, Issue:8

    Ligustilide, a natural phthalide mainly derived from chuanxiong rhizomes and Angelica Sinensis roots, possesses anti-inflammatory activity, particularly in the context of the nervous system. However, its application is limited because of its unstable chemical properties. To overcome this limitation, ligusticum cycloprolactam (LIGc) was synthesized through structural modification of ligustilide. In this study, we combined network pharmacological methods with experimental verification to investigate the anti-neuroinflammatory effects and mechanisms of ligustilide and LIGc. Based on our network pharmacology analysis, we identified four key targets of ligustilide involved in exerting an anti-inflammatory effect, with the nuclear factor (NF)-κB signal pathway suggested as the main signalling pathway. To verify these results, we examined the expression of inflammatory cytokines and inflammation-related proteins, analysed the phosphorylation level of NF-κB, inhibitor of κBα (IκBα) and inhibitor of κB kinase α and β (IKKα+β), and evaluated the effect of BV2 cell-conditioned medium on HT22 cells in vitro. Our results, demonstrate for the first time that LIGc can downregulate the activation of the NF-κB signal pathway in BV2 cells induced by lipopolysaccharide, suppress the production of inflammatory cytokines and reduce nerve injury in HT22 cells mediated by BV2 cells. These findings suggest that LIGc inhibits the neuroinflammatory response mediated by BV2 cells, providing strong scientific support for the development of anti-inflammatory drugs based on natural ligustilide or its derivatives. However, there are some limitations to our current study. In the future, further experiments using in vivo models may provide additional evidence to support our findings.

    Topics: Anti-Inflammatory Agents; Cytokines; Inflammation; Ligusticum; Lipopolysaccharides; Microglia; Network Pharmacology; Neuroinflammatory Diseases; NF-kappa B

2023
Ligustilide, a novel SIRT1 agonist, alleviates lipopolysaccharide-induced acute lung injury through deacetylation of NICD.
    International immunopharmacology, 2023, Volume: 121

    Development and progression of sepsis-induced acute lung injury (ALI) involve apoptosis and oxidative stress in lung epithelial cells. Ligustilide (LIG) is one of the main bioactive constituents derived from the Angelica sinensis. As a novel SIRT1 agonist, LIG owns powerful anti-inflammatory and antioxidative properties, exerting remarkable therapeutic effects on cancers, neurological disorders, and diabetes mellitus. However, whether LIG could protect against lipopolysaccharide (LPS)-induced ALI by activating SIRT1 remains unclear. Mice underwent intratracheal LPS injection to mimic sepsis-induced ALI while MLE-12 cells were treated with LPS for 6 h to establish an in vitro ALI model. At the same time, mice or MLE-12 cells were treated with different doses of LIG to access its pharmacological effect. The results demonstrated that LIG pretreatment could improve LPS-induced pulmonary dysfunction and pathological injury, apart from increasing 7-day survival rate. In addition, LIG pretreatment also decreased inflammation, oxidative stress and apoptosis during LPS-induced ALI. Mechanically, LPS stimulation decreased the expression and activity of SIRT1 but increased the expression of Notch1 and NICD. And LIG could also enhance the interaction between SIRT1 and NICD, thus deacetylating NICD. In vitro experiments also unveiled that EX-527, a selective SIRT1 inhibitor, could abolish LIG-elicited protection in LPS-treated MLE-12 cells. And in SIRT1 knockout mice with ALI, LIG pretreatment also lost its effects on inflammation, apoptosis, and oxidative stress during ALI.

    Topics: Acute Lung Injury; Animals; Inflammation; Lipopolysaccharides; Lung; Mice; Receptor, Notch1; Sepsis; Sirtuin 1

2023
Alleviation of glucolipotoxicity-incurred cardiomyocyte dysfunction by Z-ligustilide involves in the suppression of oxidative insult, inflammation and fibrosis.
    Chemistry and physics of lipids, 2021, Volume: 241

    Diabetes mellitus ranks as a major risk cause for disability and death around the world due to its complications, especially diabetic cardiomyopathy (DCM). Glucolipotoxicity is one of the critical causal factors of DCM. Recent finding confirms the beneficial roles of Z-ligustilide in diabetes mellitus. Nevertheless, its efficacy in DCM remains elusive. Here, Z-ligustilide elevated high glucose/high palmitic acid (HG/P)-inhibited cell viability and attenuated HG/P-induced cell apoptosis, caspase-3 activity, pro-apoptotic Bax and anti-apoptotic Bcl-2 protein expression. Furthermore, Z-ligustilide alleviated HG/P-evoked oxidative damage by decreasing HG/P-induced elevation in ROS, lactate dehydrogenase (LDH) and malondialdehyde (MDA) leakage, but increasing antioxidant enzyme-superoxide dismutase (SOD) and glutathione (GSH) levels suppressed by HG/P. Concomitantly, Z-ligustilide attenuated HG/P-induced cardiomyocyte fibrosis by increasing MMP-14 expression and diminishing HG/P-enhanced fibrotic protein expression, including collagen I, collagen II and TGF-β. Mechanistically, Z-ligustilide offset the adverse effects of HG/P on the activation of the AMPK/GSK-3β/Nrf2 pathway. Importantly, blocking the AMPK signaling overturned the protective efficacy of Z-ligustilide against HG/P-induced cardiomyocyte oxidative damage, inflammation and fibrosis. Together, these findings highlight that Z-ligustilide may alleviate glucolipotoxicity-induced cardiomyocyte dysfunction by regulating cell oxidative injury, inflammation and fibrosis via the AMPK/GSK-3β/Nrf2 pathway. Consequently, Z-ligustilide may represent a promising therapeutic agent against DCM by restoring cardiomyocyte dysfunction.

    Topics: 4-Butyrolactone; Animals; Apoptosis; Cell Survival; Cells, Cultured; Fibrosis; Inflammation; Molecular Structure; Myocytes, Cardiac; Oxidation-Reduction; Rats

2021
Systems pharmacology approach uncovers Ligustilide attenuates experimental colitis in mice by inhibiting PPARγ-mediated inflammation pathways.
    Cell biology and toxicology, 2021, Volume: 37, Issue:1

    Inflammatory bowel disease (IBD) is a chronic idiopathic disorder causing inflammation in the gastro-intestinal tract, which is lack of effective drug targets and medications. To identify novel therapeutic agents against consistent targets, we exploited a systems pharmacology-driven framework that incorporates drug-target networks of natural product and IBD disease genes. Our in silico approach found that Ligustilide (LIG), one of the major active components of Angelica acutiloba and Cnidium Officinale, potently attenuated IBD. The following in vivo and in vitro results demonstrated that LIG prevented experimental mice colitis induced by dextran sulfate sodium (DSS) via suppressing inflammatory cell infiltration, the activity of MPO and iNOS, and the expression and production of IL-1β, IL-6, and TNF-α. Subsequently, the network analysis helped to validate that LIG alleviated colitis by inhibiting NF-κB and MAPK/AP-1 pathway through activating PPARγ, which were further confirmed in RAW 264.7 cells and bone marrow-derived macrophages in vitro. In summary, this study reveals that LIG activated PPARγ to inhibit the activation of NF-κB and AP-1 signaling thus eventually alleviated DSS-induced colitis, which has promising activities and may serve as a candidate for the treatment of IBD.Graphical abstract This study suggested novel computational and experimental pharmacology approaches to identify potential IBD therapeutic agents by exploiting polypharmacology of natural products. We demonstrated that LIG could attenuate inflammation in IBD by inhibiting NF-κB and AP-1 pathways via PPARγ activation to reduce the expression of pro-inflammatory cytokines in macrophages. These findings offer comprehensive pre-clinical evidence that LIG may serve as a promising candidate for IBD therapy in the future. Graphical headlights: 1. Systems pharmacology uncovered Ligustilide attenuates experimental colitis in mice. 2. Network-based analysis predicted the mechanism of Ligustilide against IBD, which was validated by inhibiting PPARγ-mediated inflammation pathways. 3. Ligustilide activated PPARγ to inhibit NF-κB and AP-1 activation thus eventually alleviated DSS-induced colitis.4. Ligustilide has promising activities and may serve as a candidate for the treatment of IBD.

    Topics: 4-Butyrolactone; Animals; Biological Products; Colitis; Colon; Cytokines; Dextran Sulfate; Female; Inflammation; Inflammation Mediators; Inflammatory Bowel Diseases; Mice, Inbred C57BL; Models, Biological; Network Pharmacology; NF-kappa B; PPAR gamma; Signal Transduction; Transcription Factor AP-1

2021
Ligustilide improves aging-induced memory deficit by regulating mitochondrial related inflammation in SAMP8 mice.
    Aging, 2020, 02-16, Volume: 12, Issue:4

    Alzheimer's disease (AD) is an age-related neurodegenerative disease. The main active component in

    Topics: 4-Butyrolactone; Aging; Animals; Apoptosis; Brain; Inflammation; Male; Maze Learning; Memory Disorders; Mice; Mitochondria; Neuroprotective Agents; Oxidative Stress; Spatial Memory

2020
Ligustilide Ameliorates Inflammatory Pain and Inhibits TLR4 Upregulation in Spinal Astrocytes Following Complete Freund's Adjuvant Peripheral Injection.
    Cellular and molecular neurobiology, 2016, Volume: 36, Issue:1

    Ligustilide is a major component of Radix Angelica Sinensis and reported to have anti-inflammatory and anti-nociceptive effects. Toll-like receptor 4 (TLR4) has been shown to be expressed in the spinal cord and be involved in inflammatory pain and neuropathic pain. Whether ligustilide can inhibit spinal TLR4 expression in inflammatory pain is still unknown. In the present study, we intravenously injected ligustilide daily for 4 days, with the first injection given at 1 h before complete Freund's adjuvant (CFA) injection. We tested the analgesic effect of ligustilide by behavioral test and checked the expression and distribution of TLR4 in the spinal cord by real-time quantitative PCR, Western blot, and immunofluorescence. Our data showed that repeated daily intravenous treatment with ligustilide alleviated CFA-induced heat hyperalgesia and mechanical allodynia. The same treatment also inhibited CFA-induced TLR4 mRNA and protein increase in the spinal cord. Immunofluorescence double staining showed that TLR4 was predominantly expressed in spinal astrocytes. In primary cultured astrocytes, ligustilide dose-dependently reduced lipopolysaccharide-induced upregulation of TLR4 mRNA expression. These data indicate that ligustilide treatment reduces TLR4 expression in spinal astrocytes and is an effective therapy for inflammatory pain.

    Topics: 4-Butyrolactone; Animals; Astrocytes; Cells, Cultured; Freund's Adjuvant; Hot Temperature; Hyperalgesia; Inflammation; Male; Mice, Inbred ICR; Pain; RNA, Messenger; Spinal Cord; Toll-Like Receptor 4; Up-Regulation

2016
Differences in Proinflammatory Property of Six Subtypes of Peroxiredoxins and Anti-Inflammatory Effect of Ligustilide in Macrophages.
    PloS one, 2016, Volume: 11, Issue:10

    Peroxiredoxins (Prxs) are proposed to function as damage-associated molecular patterns (DAMPs) and contribute to post-ischemic neuroinflammation and brain injury by activating Toll-like receptor (TLR) 4 at the acute and subacute phases after ischemic stroke. However, there are few studies concerning the inflammatory profiles of six distinct subtypes of Prxs (Prx1-Prx6). Our previous study demonstrated that the protective effect of ligustilide (LIG) against cerebral ischemia was associated with inhibition of neuroinflammatory response and Prx/TLR4 signaling in rats. Herein, the present study explored the inflammatory members of Prxs and the effect of LIG on their inflammatory responses in macrophages.. The murine RAW264.7 macrophages were treated with each of exogenous recombinant Prxs at a range of 1 to 50 nM for 24 h. The WST-1 test showed that Prx3 exhibited a significant cytotoxicity, whereas the rest five Prxs did not affect cellular viability. The quantitative measurements with spectrometry or ELISA indicated that three subtypes, Prx1, Prx2 and Prx4, increased production of proinflammatory mediators, including nitric oxide (NO) metabolites, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in a concentration-dependent manner. Immunostaining demonstrated that 20 nM Prx1, Prx2 or Prx4 significantly increased expression of TLR4 and iNOS and nuclear translocation of NF-κB p65. However, Prx5 and Prx6 showed no poinflammatory effect in macrophages. Remarkably, LIG treatment effectively inhibited the inflammatory response induced by Prx1, Prx2 and Prx4.. Three members of Prxs, Prx1, Prx2 and Prx4, are inflammatory DAMPs that induce TLR4 activation and inflammatory response in macrophages, which is effectively inhibited by LIG. These results suggest that inflammatory Prxs-activated macrophages may provide a novel cellular model for screening the potential inhibitors of DAMPs-associated inflammatory diseases such as stroke. Moreover, selective blocking strategies targeting the inflammatory subtypes of Prxs probably provide promising therapeutic approaches with a prolonged time window for stroke.

    Topics: 4-Butyrolactone; Animals; Anti-Inflammatory Agents; Cell Line; Inflammation; Interleukin-6; Macrophages; Mice; NF-kappa B; Nitric Oxide; Nitric Oxide Synthase Type II; Peroxiredoxins; Signal Transduction; Toll-Like Receptor 4; Tumor Necrosis Factor-alpha

2016
Ligustilide ameliorates neuroinflammation and brain injury in focal cerebral ischemia/reperfusion rats: involvement of inhibition of TLR4/peroxiredoxin 6 signaling.
    Free radical biology & medicine, 2014, Volume: 71

    Blocking TLR4/peroxiredoxin (Prx6) signaling is proposed to be a novel therapeutic strategy for ischemic stroke because extracellular Prx6 released from ischemic cells may act as an endogenous ligand for TLR4 and initiate destructive immune responses in ischemic brain. Our previous studies showed that ligustilide (LIG) exerted antineuroinflammatory and neuroprotective effects against ischemic insult, but the underlying mechanisms remain unclear. This study investigated whether the TLR4/Prx6 pathway is involved in the protective effect of LIG against postischemic neuroinflammation and brain injury induced by transient middle cerebral artery occlusion (MCAO) in rats. Intraperitoneal LIG administration (20 and 40 mg/kg/day) at reperfusion onset after MCAO resulted in a reduction of brain infarct size and improved neurological outcome over 72 h. LIG-induced neuroprotection was accompanied by improvement of neuropathological alterations, including neuron loss, astrocyte and microglia/macrophage activation, neutrophil and T-lymphocyte invasion, and regulation of inflammatory mediators expression. Moreover, LIG significantly inhibited the expression and extracellular release of Prx6 and activation of TLR4 signaling, reflected by decreased TLR4 expression, extracellular signal-regulated kinase 1/2 phosphorylation, and transcriptional activity of NF-κB and signal transducer and activator of transcription 3 in the ischemic brain. Our results demonstrate that LIG may provide an early and direct neuroprotection by inhibiting TLR4/Prx6 signaling and subsequent immunity and neuroinflammation after cerebral ischemia. These findings support the translational potential of blocking TLR4/Prx6 signaling for the treatment of ischemic stroke.

    Topics: 4-Butyrolactone; Animals; Anti-Inflammatory Agents, Non-Steroidal; Astrocytes; Brain Ischemia; Cell Movement; Gene Expression Regulation; Inflammation; Injections, Intraperitoneal; Macrophage Activation; Male; Microglia; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Neurons; NF-kappa B; Oxidative Stress; Peroxiredoxin VI; Rats; Rats, Sprague-Dawley; Reperfusion Injury; Signal Transduction; STAT3 Transcription Factor; T-Lymphocytes; Toll-Like Receptor 4

2014
Ligustilide inhibits microglia-mediated proinflammatory cytokines production and inflammatory pain.
    Brain research bulletin, 2014, Volume: 109

    Ligustilide is the main component of Danggui essential oil, and recently reported to have anti-inflammatory and neuroprotective effect. Increasing evidence suggests that glia-mediated neuroinflammation in the spinal cord plays a vital role in the pathogenesis of chronic pain. In the present study, we investigated the anti-inflammatory and anti-nociceptive effect of ligustilide both in vitro and in vivo. In microglial cell line BV2 cells, lipopolysaccharide (LPS) time-dependently increased the mRNA expression of proinflammatory cytokines (TNF-α, IL-1β, and IL-6), which was decreased by pretreatment with ligustilide in a dose-dependent manner. Ligustilide also decreased LPS-induced proinflammatory cytokines production in primary cultured microglia. In vivo, intrathecal injection of LPS induced mechanical allodynia in mice. Intravenous injection of ligustilide prevented LPS-induced mechanical allodynia, and decreased LPS-induced TNF-α, IL-1β, and IL-6 up-regulation in the spinal cord. In addition, repetitive intravenous injection of ligustilide attenuated intraplantar injection of complete Freund's adjuvant (CFA)-induced mechanical allodynia and thermal hyperalgesia. The same treatment of ligustilide also inhibited CFA-induced TNF-α, IL-1β, and IL-6 up-regulation and microglial activation in the spinal cord. Taken together, our data suggest that ligustilide can alleviate inflammatory pain partly through inhibition of microglial activation and proinflammatory cytokines production, which indicates a possible benefit from the use of ligustilide in the treatment of inflammatory pain and neuroinflammation-associated disorders.

    Topics: 4-Butyrolactone; Animals; Anti-Inflammatory Agents; Calcium-Binding Proteins; Cell Line, Transformed; Cytokines; Disease Models, Animal; Dose-Response Relationship, Drug; Freund's Adjuvant; Inflammation; Lipopolysaccharides; Male; Mice; Microfilament Proteins; Microglia; Pain; Pain Measurement; Pain Threshold; RNA, Messenger; Spinal Cord; Time Factors

2014
Anti-inflammatory ligustilides from Ligusticum chuanxiong Hort.
    Fitoterapia, 2013, Volume: 91

    Four new ligustilides chuanxiongnolide R1 (1), chuanxiongnolide R2 (2), chuanxiongdiolide R1 (3) and chuanxiongdiolide R2 (4) together with eight known derivatives (5-12) were isolated from the root of Ligusticum chuanxiong Hort. Their structures were elucidated by HR-ESI-MS, UV, IR, 1D and 2D NMR (HSQC, HMBC, (1)H-(1)H COSY, NOESY) methods. The absolute configurations were confirmed via the circular dichroism (CD) spectrum. The anti-inflammatory assay in LPS-triggered RAW 264.7 macrophages was carried out on the twelve compounds. 1, 3, 5 and 6 showed significant inhibitory effects against LPS-induced NO production.

    Topics: 4-Butyrolactone; Animals; Anti-Inflammatory Agents; Benzofurans; Drugs, Chinese Herbal; Inflammation; Ligusticum; Lipopolysaccharides; Macrophages; Mice; Molecular Structure; Nitric Oxide; Phytotherapy; Plant Roots

2013
Anti-inflammatory effects of Z-ligustilide nanoemulsion.
    Inflammation, 2013, Volume: 36, Issue:2

    The Z-ligustilide (LIG) was studied for its anti-inflammatory activities with prepared LIG nanoemulsions (LIGNE). Healthy male adult Wistar rats were used in the study. Endotoxin-induced uveitis (EIU) was induced by a footpad injection of 200 μg lipopolysaccharide. EIU rats were administered orally with saline, LIG (20 mg/kg/day), and LIGNE (20 mg LIG /kg/day), respectively. Twenty-four hours later, rats were euthanized, and blood was collected from either right marginal ear vein to estimate inflammatory cells and inflammatory mediators. The drug dissolution profiles of LIGNE in both phosphate buffer pH 6.8 and 0.1 N HCl showed complete dissolution within 20 min. Pharmacokinetic studies suggested a significant increase (P < 0.0001) in the C pmax and AUC0→24 h were observed in the LIGNE group when compared with the LIG group. LIGNE significantly reduced the levels of tumor necrosis factor alpha, interleukin 1 beta, vascular endothelial growth factor alpha, and interleukin-17. The anti-inflammatory animal testing revealed that LIGNE led to an improvement in oral bioavailability.

    Topics: 4-Butyrolactone; Angelica sinensis; Animals; Anti-Inflammatory Agents; Biological Availability; Emulsions; Inflammation; Inflammation Mediators; Interleukin-17; Interleukin-1beta; Lipopolysaccharides; Male; Nanoparticles; Plant Oils; Rats; Rats, Wistar; Tumor Necrosis Factor-alpha; Uveitis; Vascular Endothelial Growth Factor A

2013
The anti-inflammatory effect of Z-Ligustilide in experimental ovariectomized osteopenic rats.
    Inflammation, 2012, Volume: 35, Issue:6

    The purpose of the present study was to investigate the anti-inflammatory activity of Z-Ligustilide (LIG) in experimental ovariectomized (OVX) osteopenic rats. The anti-inflammatory potential of LIG in the regulation of nuclear factor kappa B (NF-κB), maleic dialdehyde (MDA), polymorphonuclear cells (PMN), interleukin-1β (IL-1β), inducible nitric oxide synthase (iNOS) and tumor necrosis factor-α (TNF-α), adhesion molecule (ICAM-1) and cyclooxygenase-2 (COX-2) was determined by ELISA. LIG significantly inhibited OVX-induced up-regulation of NF-κB activation and the production of IL-1β, TNF-α, iNOS, ICAM-1 and COX-2. Moreover, LIG suppressed MDA and infiltration of PMN. The results of the present study clearly demonstrate that there may be an inflammatory component in the etiology of osteoporosis. It revealed a significant anti-inflammatory effect of Z-Ligustilide in experimental OVX osteopenic rats.

    Topics: 4-Butyrolactone; Aldehydes; Animals; Anti-Inflammatory Agents; Cyclooxygenase 2; Female; Inflammation; Intercellular Adhesion Molecule-1; Interleukin-1beta; Neutrophils; NF-kappa B; Nitric Oxide Synthase Type II; Osteoporosis; Ovariectomy; Rats; Rats, Wistar; Tumor Necrosis Factor-alpha; Up-Regulation

2012
Inhibitory effects of Angelica sinensis ethyl acetate extract and major compounds on NF-kappaB trans-activation activity and LPS-induced inflammation.
    Journal of ethnopharmacology, 2010, May-27, Volume: 129, Issue:2

    Previous study showed that the ethyl acetate (EtOAc) fraction from Angelica sinensis (Oliv.) Diels (Apiaceae) (AS) inhibited nitric oxide (NO) and prostaglandin E(2) secretions in vitro. This study was to evaluate anti-inflammatory activity of AS EtOAc extract and its major compounds in vivo and in vitro.. NF-kappaB luciferase activity and pro-inflammatory cytokine secretions from lipopolysaccharide (LPS) plus interferon (IFN)-gamma-stimulated RAW 264.7 cells pre-treated with EtOAc extract or compounds were analyzed. For further in vivo study, BALB/c mice were tube-fed with 1.56 (AS1 group), 6.25 (AS2 group) mg/kg body weight/day in 100 microl soybean oil, while the control and PDTC (pyrrolidine dithiocarbamate, an anti-inflammatory agent) groups were tube-fed with 100 microl soybean oil/day only. After 1 week of tube-feeding, the PDTC group was injected with 50 mg/kg BW PDTC and 1 h later, all of the mice were injected with 15 mg/kg BW LPS. The pro-inflammatory cytokine levels and lifespan of LPS-challenged mice were determined.. The results showed that AS EtOAc extract significantly inhibited NF-kappaB luciferase activity and TNF-alpha, IL-6, macrophage inflammatory protein-2 (MIP-2) and NO secretions from LPS/IFN-gamma-stimulated RAW 264.7 cells. The AS1 and PDTC groups, but not AS2, had significantly higher survival rate than the control group. This was characterized by the inhibition of the serum TNF-alpha and IL-12p40 levels after LPS injection (p<0.05). The major compounds of AS, ferulic acid and Z-ligustilide, also significantly decreased NF-kappaB luciferase activity, which may contribute to the anti-inflammatory activity of AS.. Low dose of AS EtOAc extract that inhibits the production of inflammatory mediators alleviates acute inflammatory hazards and protect mice from endotoxic shock.

    Topics: 4-Butyrolactone; Angelica sinensis; Animals; Anti-Inflammatory Agents; Cell Line; Coumaric Acids; Female; Inflammation; Inflammation Mediators; Lipopolysaccharides; Luciferases; Macrophages; Mice; Mice, Inbred BALB C; NF-kappa B; Phytotherapy; Plant Extracts

2010
Z-ligustilide attenuates lipopolysaccharide-induced proinflammatory response via inhibiting NF-kappaB pathway in primary rat microglia.
    Acta pharmacologica Sinica, 2010, Volume: 31, Issue:7

    To investigate the anti-inflammatory effect of Z-ligustilide (LIG) on lipopolysaccharide (LPS)-activated primary rat microglia.. Microglia were pretreated with LIG 1 h prior to stimulation with LPS (1 microg/mL). After 24 h, cell viability was tested with MTT, nitric oxide (NO) production was assayed with Griess reagent, and the content of tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and monocyte chemoattractant protein (MCP-1) was measured with ELISA. Protein expression of the nuclear factor-kappaB (NF-kappaB) p65 subunit, cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) was detected with immunocytochemistry 1 h or 24 h after LPS treatment.. LIG showed a concentration-dependent anti-inflammatory effect in LPS-activated microglia, without causing cytotoxicity. Pretreatment with LIG at 2.5, 5, 10, and 20 micromol/L decreased LPS-induced NO production to 75.9%, 54.4%, 43.1%, and 47.6% (P<0.05 or P< 0.01), TNF-alpha content to 86.2%, 68.3%, 40.1%, and 39.9% (P<0.01, with the exception of 86.2% for 2.5 micromol/L LIG), IL-1beta content to 31.5%, 27.7%, 0.6%, and 0% (P<0.01), and MCP-1 content to 84.4%, 50.3%, 45.1%, and 42.2% (P<0.05 or P<0.01), respectively, compared with LPS treatment alone. LIG (10 micromol/L) significantly inhibited LPS-stimulated immunoreactivity of activated NF-kappaB, COX-2, and iNOS (P<0.01 vs LPS group).. LIG exerted a potent anti-inflammatory effect on microglia through inhibition of NF-kappaB pathway. The data provide direct evidence of the neuroprotective effects of LIG and the potential application of LIG for the treatment of the neuroinflammatory diseases characterized by excessive microglial activation.

    Topics: 4-Butyrolactone; Animals; Anti-Inflammatory Agents; Cyclooxygenase 2; Dose-Response Relationship, Drug; Inflammation; Lipopolysaccharides; Microglia; Neuroprotective Agents; NF-kappa B; Nitric Oxide Synthase Type II; Rats; Rats, Sprague-Dawley; Stereoisomerism

2010