• Title/Summary/Keyword: Anti-inflammatory factor

Search Result 1,515, Processing Time 0.035 seconds

The Anti-inflammatory Mechanism of Protaetia brevitarsis Lewis via Suppression the Activation of NF-κB and Caspase-1 in LPS-stimulated RAW264.7 Cells

  • Myung, Noh-Yil;Ahn, Eun-Mi;Kim, Su-Jin
    • Biomedical Science Letters
    • /
    • v.26 no.4
    • /
    • pp.267-274
    • /
    • 2020
  • The larva of Protaetia brevitarsis Lewis (P. brevitarsis), edible insect, is traditionally consumed as alternative source of nutrients and has various health benefits. However, the exact pharmaceutical effects of P. brevitarsis on inflammatory response are still not well understood. Thus, we investigated the anti-inflammatory effects and mechanisms of P. brevitarsis in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. We investigated the effects of P. brevitarsis on the expression levels of inflammatory-related genes, including inflammatory cytokines, prostaglandin E2 (PGE2), cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) in LPS-stimulated RAW264.7 cells. To understand the anti-inflammatory mechanism of P. brevitarsis, we explored the regulatory effect of P. brevitarsis on nuclear factor (NF)-κB and caspase-1 activation. The findings of this study demonstrated that P. brevitarsis inhibits the LPS-induced inflammatory cytokine and PGE2 levels, as well as COX-2 and iNOS expression. Moreover, we confirmed that the anti-inflammatory effect of P. brevitarsis occurs via suppression of the activation of NF-κB and caspase-1. Conclusively, these findings provide experimental evidence that P. brevitarsis may be useful candidate for the treatment of inflammatory-related diseases.

Anti-Inflammatory Effect of Asterias amurensis Fatty Acids through NF-κB and MAPK Pathways against LPS-Stimulated RAW264.7 Cells

  • Monmai, Chaiwat;Go, Seok Hyeon;Shin, Il-sik;You, SangGuan;Kim, Dae-ok;Kang, SeokBeom;Park, Woo Jung
    • Journal of Microbiology and Biotechnology
    • /
    • v.28 no.10
    • /
    • pp.1635-1644
    • /
    • 2018
  • Asterias amurensis (starfish) is a marine organism that is harmful to the fishing industry, but is also a potential source of functional materials. The present study was conducted to analyze the profiles of fatty acids extracted from A. amurensis tissues and their anti-inflammatory effects on RAW264.7 macrophage cells. In different tissues, the component ratios of saturated fatty acids, monounsaturated fatty acids, and polyunsaturated fatty acids differed; particularly, polyunsaturated fatty acids such as dihomo-gamma-linolenic acid (20:3n-6) and eicosapentaenoic acid (20:5n-3) were considerably different. In lipopolysaccharide-stimulated RAW264.7 cells, fatty acids from A. amurensis skin, gonads, and digestive glands exhibited anti-inflammatory activities by reducing nitric oxide production and inducing nitric oxide synthase gene expression. Asterias amurensis fatty acids effectively suppressed the expression of inflammatory cytokines such as tumor necrosis $factor-{\alpha}$, interleukin-$1{\beta}$, and interleukin-6 in lipopolysaccharide-stimulated cells. Cyclooxygenase-2 and prostaglandin $E_2$, which are critical inflammation biomarkers, were also significantly suppressed. Furthermore, A. amurensis fatty acids reduced the phosphorylation of nuclear $factor-{\kappa}B$ p-65, p38, extracellular signal-related kinase 1/2, and c-Jun N-terminal kinase, indicating that these fatty acids ameliorated inflammation through the nuclear $factor-{\kappa}B$ and mitogen-activated protein kinase pathways. These results provide insight into the anti-inflammatory mechanism of A. amurensis fatty acids on immune cells and suggest that the species is a potential source of anti-inflammatory molecules.

Hyper-inflammatory responses in COVID-19 and anti-inflammatory therapeutic approaches

  • Choi, Hojun;Shin, Eui-Cheol
    • BMB Reports
    • /
    • v.55 no.1
    • /
    • pp.11-19
    • /
    • 2022
  • The coronavirus disease 2019 (COVID-19) is an ongoing global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Patients with severe COVID-19 exhibit hyper-inflammatory responses characterized by excessive activation of myeloid cells, including monocytes, macrophages, and neutrophils, and a plethora of pro-inflammatory cytokines and chemokines. Accumulating evidence also indicates that hyper-inflammation is a driving factor for severe progression of the disease, which has prompted the development of anti-inflammatory therapies for the treatment of patients with COVID-19. Corticosteroids, IL-6R inhibitors, and JAK inhibitors have demonstrated promising results in treating patients with severe disease. In addition, diverse forms of exosomes that exert anti-inflammatory functions have been tested experimentally for the treatment of COVID-19. Here, we briefly describe the immunological mechanisms of the hyper-inflammatory responses in patients with severe COVID-19. We also summarize current anti-inflammatory therapies for the treatment of severe COVID-19 and novel exosome-based therapeutics that are in experimental stages.

The Anti-depressive Effect of Rehmanniae Radix Preparata via Anti-inflammatory Activity (숙지황 추출물의 항염증 작용을 통한 항우울 효과)

  • Kim, Eung Sun;Chong, Myongsoo
    • The Journal of Korean Medicine
    • /
    • v.43 no.1
    • /
    • pp.99-111
    • /
    • 2022
  • Objectives: Rehmanniae Radix Preparata (RRP) has been used as a traditional remedy to treat gynecology and endocrine diseases. Recently, studies on antioxidant and anti-inflammatory effects of RRP have been reported, so it was judged that RRP extracts would have an anti-depressive effect. Methods: We investigated the anti-neuroinflammatory and anti-depressive effect of RRP on lipopolysaccharide (LPS)-induced depression and LPS-stimulated BV2 microglia. RRP inhibited the LPS-stimulated excessive release of nitrite in the BV2 cells. RRP also significantly inhibited the inflammatory cytokines such as tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta and IL-6 in LPS-stimulated BV2 microglial cells. Results: RRP significantly suppressed the LPS-induced mitogen-activated protein kinase (MAPKs) and nuclear factor (NF)-𝜅B activation. In addition, administration of RRP not only inhibited the immobility time in the forced swimming test (FST) but also increased the total travel distance in the open field test (OFT). Also, RRP inhibited the elevation of TNF-alpha, IL-1beta, and IL-6 in brain of LPS-injected mice. Conclusions: Considering the overall results, our study showed that RRP exhibited the anti-neuroinflammatory and anti-depressive activities via deactivation of MAPKs and NF-𝜅B.

The mechanism of human neural stem cell secretomes improves neuropathic pain and locomotor function in spinal cord injury rat models: through antioxidant, anti-inflammatory, anti-matrix degradation, and neurotrophic activities

  • I Nyoman Semita;Dwikora Novembri Utomo;Heri Suroto;I Ketut Sudiana;Parama Gandi
    • The Korean Journal of Pain
    • /
    • v.36 no.1
    • /
    • pp.72-83
    • /
    • 2023
  • Background: Globally, spinal cord injury (SCI) results in a big burden, including 90% suffering permanent disability, and 60%-69% experiencing neuropathic pain. The main causes are oxidative stress, inflammation, and degeneration. The efficacy of the stem cell secretome is promising, but the role of human neural stem cell (HNSC)-secretome in neuropathic pain is unclear. This study evaluated how the mechanism of HNSC-secretome improves neuropathic pain and locomotor function in SCI rat models through antioxidant, anti-inflammatory, anti-matrix degradation, and neurotrophic activities. Methods: A proper experimental study investigated 15 Rattus norvegicus divided into normal, control, and treatment groups (30 µL HNSC-secretome, intrathecal in the level of T10, three days post-traumatic SCI). Twenty-eight days post-injury, specimens were collected, and matrix metalloproteinase (MMP)-9, F2-Isoprostanes, tumor necrosis factor (TNF)-α, transforming growth factor (TGF)-β, and brain derived neurotrophic factor (BDNF) were analyzed. Locomotor recovery was evaluated via Basso, Beattie, and Bresnahan scores. Neuropathic pain was evaluated using the Rat Grimace Scale. Results: The HNSC-secretome could improve locomotor recovery and neuropathic pain, decrease F2-Isoprostane (antioxidant), decrease MMP-9 and TNF-α (anti-inflammatory), as well as modulate TGF-β and BDNF (neurotrophic factor). Moreover, HNSC-secretomes maintain the extracellular matrix of SCI by reducing the matrix degradation effect of MMP-9 and increasing the collagen formation effect of TGF-β as a resistor of glial scar formation. Conclusions: The present study demonstrated the mechanism of HNSC-secretome in improving neuropathic pain and locomotor function in SCI through antioxidant, anti-inflammatory, anti-matrix degradation, and neurotrophic activities.

Anti-Inflammatory Effects of Water Chestnut Extract on Cytokine Responses via Nuclear Factor-κB-signaling Pathway

  • Kim, Bora;Kim, Jin Eun;Choi, Byung-Kook;Kim, Hyun-Soo
    • Biomolecules & Therapeutics
    • /
    • v.23 no.1
    • /
    • pp.90-97
    • /
    • 2015
  • Water chestnut (Trapa japonica Flerov.) is an annual aquatic plant. In the present study, we showed that the treatment of water chestnut extracted with boiling water resulted in a significant increase 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical-scavenging activity and decrease the intracellular $H_2O_2$-induced accumulation of reactive oxygen species. In addition, water chestnut extract (WCE) inhibited lipopolysaccharide (LPS)-induced nitric oxide production and suppressed mRNA and protein expression of the inducible nitric oxide synthase gene. The cytokine array results showed that WCE inhibited inflammatory cytokine secretion. Also, WCE reduced tumor necrosis factor-${\alpha}$- and interleukin-6-induced nuclear factor-${\kappa}B$ activity. Furthermore, during sodium lauryl sulfate (SLS)-induced irritation of human skin, WCE reduced SLS-induced skin erythema and improved barrier regeneration. These results indicate that WCE may be a promising topical anti-inflammatory agent.

The Anti-inflammatory Mechanism of Xanthoangelol E is Through the Suppression of NF-${\kappa}B$/Caspase-1 Activation in LPS-stimulated Mouse Peritoneal Macrophage

  • Seoa, Jung-Ho;Kim, Su-Jin
    • Biomedical Science Letters
    • /
    • v.18 no.4
    • /
    • pp.345-354
    • /
    • 2012
  • Angelica keiskei has exhibited numerous pharmacological effects including antitumor, antimetastatic, and antidiabetic effects. However, the anti-inflammatory effects and mechanisms employed by xanthoangelol E isolated from Angelica keiskei are incompletely understood. In this study, we attempted to determine the effects of Xanthoangelol E on the lipopolysaccharide (LPS)-stimulated mouse peritoneal macrophage. The findings of this study demonstrated that xanthoangelol E inhibited the production of tumor necrosis factor (TNF)-${\alpha}$, interleukin (IL)-6, and prostaglandin $E_2$ ($PGE_2$). Xanthoangelol E inhibited the enhanced levels of cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) caused by LPS. Additionally, we showed that the anti-inflammatory effect of xanthoangelol E is through the regulation of the activation of nuclear factor (NF)-${\kappa}B$ and caspase-1. These results provide novel insights into the pharmacological actions of xanthoangelol E as a potential candidate for the development of new drugs to treat inflammatory diseases.

Analysis of Biological Experiments on the Anti-inflammatory and Antipyretic Effects of Hwangryeonhaedok-tang

  • Kim, Jung-Hoon;Shin, Hyeun-Kyoo
    • The Journal of Korean Medicine
    • /
    • v.33 no.4
    • /
    • pp.26-36
    • /
    • 2012
  • Objectives: To establish scientific and objective evidence for the use of a Korean medicine, articles regarding Hwangryeonhaedok-tang (HRHDT), a herbal medicine frequently used in Korean medical clinics and hospitals, were gathered and analyzed. Methods: The articles were classified as being from domestic or international journals, and by their year of publication. The mechanisms of the anti-inflammatory and antipyretic effects of HRHDT were investigated. Results: Of the 25 articles analyzed, 7 were published from Korea, 7 were from China, and 11 were from Japan. HRHDT showed anti-inflammatory and antipyretic effects through the regulation of the expression of Th1 cytokines including interleukin-2 (IL-2), IL-8, interferon-${\gamma}$ (IFN-${\gamma}$), and tumor necrosis factor-${\alpha}$ (TNF-${\alpha}$); and Th2 cytokines including IL-4, IL-6, and IL-12, which inhibit leukotriene B4 (LTB4), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and inflammatory cells. It also lowered preprodynorphin (PPD), and corticotropin-releasing factor (CRF) in the peripheral nerve system and hypothalamus. Conclusions: We speculate that the anti-inflammatory and antipyretic effects could be related to the therapeutic efficacy of HRHDT in removing pathogenic fire and heat.

Studies on Anti-inflammatory Effects of Yangdan-tang Extracts (양단탕 추출물의 항염증 효과에 대한 연구)

  • Choi, Su Ryeon;Hwang, Hyung Seo;Kim, Tae Yeon
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.34 no.5
    • /
    • pp.238-244
    • /
    • 2020
  • Yangdan-tang (YD) is recorded as a treatment to treat exterior-related fever illness in the Korean medicine. In this study, we examined the anti-inflammatory effects of YD, using YD water extract and lipopolysaccharide (LPS)-induced RAW 264.7 cells. First of all, we measured the amount of nitric oxide (NO) and prostaglandin E2 (PGE2), the products of inflammatory metabolism. Also, we measured enzymes such as inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), as well as cytokines such as tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), interleukin 1 alpha (IL-1α), and interleukin 1 beta (IL-1β). YD suppressed the production of NO and PGE2 in a dose dependent manner and reduced the amount of protein and the mRNA expression of iNOS and COX-2. Also, YD reduced the mRNA expression of TNF-α, IL-6, IL-1α and IL-1β. In conclusion, YD decreased production of LPS-induced inflammatory factor, which could be a clinical basic subject for inflammatory diseases.

Intestinal anti-inflammatory activity of Sasa quelpaertensis leaf extract by suppressing lipopolysaccharide-stimulated inflammatory mediators in intestinal epithelial Caco-2 cells co-cultured with RAW 264.7 macrophage cells

  • Kim, Kyung-Mi;Kim, Yoo-Sun;Lim, Ji Ye;Min, Soo Jin;Ko, Hee-Chul;Kim, Se-Jae;Kim, Yuri
    • Nutrition Research and Practice
    • /
    • v.9 no.1
    • /
    • pp.3-10
    • /
    • 2015
  • BACKGROUND/OBJECTIVES: Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, involves chronic inflammation of the gastrointestinal tract. Previously, Sasa quelpaertensis leaves have been shown to mediate anti-inflammation and anti-cancer effects, although it remains unclear whether Sasa leaves are able to attenuate inflammation-related intestinal diseases. Therefore, the aim of this study was to investigate the anti-inflammatory effects of Sasa quelpaertensis leaf extract (SQE) using an in vitro co-culture model of the intestinal epithelial environment. MATERIALS/METHODS: An in vitro co-culture system was established that consisted of intestinal epithelial Caco-2 cells and RAW 264.7 macrophages. Treatment with lipopolysaccharide (LPS) was used to induce inflammation. RESULTS: Treatment with SQE significantly suppressed the secretion of LPS-induced nitric oxide (NO), prostaglandin $E_2$ ($PGE_2$), IL-6, and IL-$1{\beta}$ in co-cultured RAW 264.7 macrophages. In addition, expressions of inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, and tumor necrosis factor (TNF)-${\alpha}$ were down-regulated in response to inhibition of $I{\kappa}B{\alpha}$ phosphorylation by SQE. Compared with two bioactive compounds that have previously been identified in SQE, tricin and P-coumaric acid, SQE exhibited the most effective anti-inflammatory properties. CONCLUSIONS: SQE exhibited intestinal anti-inflammatory activity by inhibiting various inflammatory mediators mediated through nuclear transcription factor kappa-B (NF-kB) activation. Thus, SQE has the potential to ameliorate inflammation-related diseases, including IBD, by limiting excessive production of pro-inflammatory mediators.