• Title/Summary/Keyword: MyD88

Search Result 70, Processing Time 0.037 seconds

Inhibitory Effects of Lactobacillus plantarum Lipoteichoic Acid (LTA) on Staphylococcus aureus LTA-Induced Tumor Necrosis Factor-Alpha Production

  • Kim, Han-Geun;Lee, Seung-Yeon;Kim, Na-Ra;Ko, Mi-Yeon;Lee, Jung-Min;Yi, Tae-Hoo;Chung, Sung-Kyun;Chung, Dae-Kyun
    • Journal of Microbiology and Biotechnology
    • /
    • v.18 no.6
    • /
    • pp.1191-1196
    • /
    • 2008
  • Staphylococcus aureus is a common etiologic agent for Gram-positive sepsis, and its lipoteichoic acid (LTA) may be important in causing Gram-positive bacterial septic shock. Here, we demonstrate that highly purified LTA (pLTA) isolated from Lactobacillus plantarum inhibited S. aureus LTA (aLTA)-induced TNF-${\alpha}$ production in THP-1 cells. Whereas pLTA scarcely induced TNF-${\alpha}$ production, aLTA induced excessive TNF-${\alpha}$ production. Interestingly, aLTA-induced TNF-${\alpha}$ production was inhibited by pLTA pretreatment. Compared with pLTA, aLTA induced a strong signal transduction through the MyD88, NF-${\kappa}B$, and MAP kinases. This signaling, however, was reduced by a pLTA pretreatment, and resulted in the inhibition of aLTA-induced TNF-${\alpha}$ production. Whereas dealanylated LTAs, as well as native LTAs, contributed to TNF-${\alpha}$ induction or TNF-${\alpha}$ reduction, deacylated LTAs did not, indicating that the acyl chain of LTA played an important role in the LTA-mediated immune regulation. These results suggest that pLTA may act as an antagonist for aLTA, and that an antagonistic pLTA may be a useful agent for suppressing the septic shock caused by Gram-positive bacteria.

Anti-inflammatory Effects of Phytochemicals Having Michael Addition Acceptors by the Modulation of Toll-like Receptor Signaling Pathways (Michael addition acceptor 그룹을 가지고 있는 phytochemicals의 toll-like receptor 신호전달체계 조절을 통한 항염증 효과)

  • Youn, Hyung-Sun
    • Korean Journal of Food Science and Technology
    • /
    • v.41 no.5
    • /
    • pp.477-482
    • /
    • 2009
  • Toll-like receptors (TLRs) play a critical role in the induction of innate immune responses that are essential for host defense against invading microbial pathogens. In general, TLRs have two major downstream signaling pathways, namely MyD88- and TRIF-dependent pathways, leading to the activation of nuclear factor-${\kappa}B$ (NF-${\kappa}B$) and interferon regulatory factor 3 (IRF3) and the expression of inflammatory mediators. TLR4 dimerization is required for the activation of downstream signaling pathways and may be one of the first lines of regulation in activating TLR-mediated signaling pathways. In this paper, the molecular targets of curcumin, 6-shogaol, and cinnamaldehyde in TLR signaling pathways will be discussed. Curcumin, 6-shogaol, and cinnamaldehyde with ${\alpha},{\beta}$-unsaturated carbonyl groups inhibit the dimerization of TLR4 induced by lipopolysaccharide, resulting in the downregulation of NF-${\kappa}B$ and IRF3. These results suggest that phytochemicals with the structural motif conferring Michael addition inhibit TLR4 dimerization, suggesting a novel mechanism for the anti-inflammatory activity of phytochemicals.

Flagellin-Stimulated Production of Interferon-β Promotes Anti-Flagellin IgG2c and IgA Responses

  • Kang, Wondae;Park, Areum;Huh, Ji-Won;You, Gihoon;Jung, Da-Jung;Song, Manki;Lee, Heung Kyu;Kim, You-Me
    • Molecules and Cells
    • /
    • v.43 no.3
    • /
    • pp.251-263
    • /
    • 2020
  • Flagellin, a major structural protein of the flagellum found in all motile bacteria, activates the TLR5- or NLRC4 inflammasome-dependent signaling pathway to induce innate immune responses. Flagellin can also serve as a specific antigen for the adaptive immune system and stimulate anti-flagellin antibody responses. Failure to recognize commensal-derived flagellin in TLR5-deficient mice leads to the reduction in anti-flagellin IgA antibodies at steady state and causes microbial dysbiosis and mucosal barrier breach by flagellated bacteria to promote chronic intestinal inflammation. Despite the important role of anti-flagellin antibodies in maintaining the intestinal homeostasis, regulatory mechanisms underlying the flagellin-specific antibody responses are not well understood. In this study, we show that flagellin induces interferon-β (IFN-β) production and subsequently activates type I IFN receptor signaling in a TLR5- and MyD88-dependent manner in vitro and in vivo. Internalization of TLR5 from the plasma membrane to the acidic environment of endolysosomes was required for the production of IFN-β, but not for other pro-inflammatory cytokines. In addition, we found that anti-flagellin IgG2c and IgA responses were severely impaired in interferon-alpha receptor 1 (IFNAR1)-deficient mice, suggesting that IFN-β produced by the flagellin stimulation regulates anti-flagellin antibody class switching. Our findings shed a new light on the regulation of flagellin-mediated immune activation and may help find new strategies to promote the intestinal health and develop mucosal vaccines.

The Early Induction of Suppressor of Cytokine Signaling 1 and the Downregulation of Toll-like Receptors 7 and 9 Induce Tolerance in Costimulated Macrophages

  • Lee, Hyo-Ji;Kim, Keun-Cheol;Han, Jeong A;Choi, Sun Shim;Jung, Yu-Jin
    • Molecules and Cells
    • /
    • v.38 no.1
    • /
    • pp.26-32
    • /
    • 2015
  • Toll-like receptors (TLR) 7 and 9 transduce a cellular signal through the MyD88-dependent pathway and induce the production of inflammatory mediators against microbial nucleotide components. The repeated stimulation of TLR4 leads to endotoxin tolerance, but the molecular mechanisms of tolerance induced through the costimulation of individual TLR has not yet been established, although endosomal TLRs share signaling pathways with TLR4. In the present study, mouse macrophages were simultaneously stimulated with the TLR7 agonist, gardiquimod (GDQ), and the TLR9 agonist, CpG ODN 1826, to examine the mechanism and effector functions of macrophage tolerance. Compared with individual stimulation, the costimulation of both TLRs reduced the secretion of TNF-${\alpha}$ and IL-6 through the delayed activation of the NF-${\kappa}B$ pathway; notably, IL-10 remained unchanged in costimulated macrophages. This tolerance reflected the early induction of suppressor of cytokine signaling-1 (SOCS-1), according to the detection of elevated TNF-${\alpha}$ secretion and restored NF-${\kappa}B$ signaling in response to the siRNA-mediated abrogation of SOCS-1 signaling. In addition, the restimulation of each TLRs using the same ligand significantly reduced the expression of both TLRs in endosomes. These findings revealed that the costimulation of TLR7 and TLR9 induced macrophage tolerance via SOCS-1, and the restimulation of each receptor or both TLR7 and TLR9 downregulated TLR expression through a negative feedback mechanisms that protects the host from excessive inflammatory responses. Moreover, the insufficient and impaired immune response in chronic viral infection might also reflect the repeated and simultaneous stimulation of those endosomal TLRs.

Modified Renshen Wumei Decoction Alleviates Intestinal Barrier Destruction in Rats with Diarrhea

  • Guan, Zhiwei;Zhao, Qiong;Huang, Qinwan;Zhao, Zhonghe;Zhou, Hongyun;He, Yuanyuan;Li, Shanshan;Wan, Shifang
    • Journal of Microbiology and Biotechnology
    • /
    • v.31 no.9
    • /
    • pp.1295-1304
    • /
    • 2021
  • Modified Renshen Wumei decoction (MRWD), a famous traditional Chinese medicine, is widely used for treating persistent diarrhea. However, as the mechanism by which MRWD regulates diarrhea remains unknown, we examined the protective effects of MRWD on intestinal barrier integrity in a diarrhea model. In total, 48 male rats were randomly distributed to four treatment groups: the blank group (CK group), model group (MC group), Medilac-Vita group (MV group) and Chinese herb group (MRWD group). After a 21-day experiment, serum and colon samples were assessed. The diarrhea index, pathological examination findings and change in ᴅ-lactate and diamine oxidase (DAO) contents illustrated that the induction of diarrhea caused intestinal injury, which was ameliorated by MV and MRWD infusion. Metabolomics analysis identified several metabolites in the serum. Some critical metabolites, such as phosphoric acid, taurine, cortisone, leukotriene B4 and calcitriol, were found to be significantly elevated by MRWD infusion. Importantly, these differences correlated with mineral absorption and metabolism and peroxisome proliferator-activated receptor (PPAR) pathways. Moreover, it significantly increased the expression levels of TLR4, MyD88 and p-NF-κB p65 proteins and the contents of IL-1 and TNF-α, while the expression levels of occludin, claudin-1 and ZO-1 proteins decreased. These deleterious effects were significantly alleviated by MV and MRWD infusion. Our findings indicate that MRWD infusion helps alleviate diarrhea, possibly by maintaining electrolyte homeostasis, improving the intestinal barrier integrity, and inhibiting the TLR4/NF-κB axis.

Nonsaponin fraction of Korean Red Ginseng attenuates cytokine production via inhibition of TLR4 expression

  • Ahn, Huijeong;Han, Byung-Cheol;Kim, Jeongeun;Kang, Seung Goo;Kim, Pyeung-Hyeun;Jang, Kyoung Hwa;So, Seung Ho;Lee, Seung-Ho;Lee, Geun-Shik
    • Journal of Ginseng Research
    • /
    • v.43 no.2
    • /
    • pp.291-299
    • /
    • 2019
  • Background: Ginsenosides of Korean Red Ginseng extracts (RGE) and its saponin components suppress secretion of inflammasome-mediating cytokines, whereas the nonsaponin fraction (NS) of RGE oppositely stimulates cytokine secretion. Although direct exposure of NS to macrophages in mice induces cytokine production, oral administration of NS has not been studied in inflammasome-related disease in animal models. Methods: Mice were fed RGE or NS for 7 days and then developed peritonitis. Peritoneal cytokines were measured, and peritoneal exudate cells (PECs) were collected to assay expression levels of a set of toll-like receptors (TLRs) and cytokines in response to NS ingestion. In addition, the role of intestinal bacteria in NS-fed mice was assessed. The effect of preexposure to NS in bone marrow-derived macrophages (BMDMs) on cytokine production was further confirmed. Results: NS ingestion attenuated secretion of peritoneal cytokines resulting from peritonitis. In addition, the isolated PECs from NS-fed mice presented lower TLR transcription levels than PECs from control diet-fed mice. BMDMs treated with NS showed downregulation of TLR4 mRNA and protein expression, which was mediated by the $TLR4-MyD88-NF{\kappa}B$ signal pathway. BMDMs pretreated with NS produced less cytokines in response to TLR4 ligands. Conclusion: NS administration directly inhibits TLR4 expression in inflammatory cells such as macrophages, thereby reducing secretion of cytokines during peritonitis.

Anti-cancer and Anti-inflammatory Effects of Curcumin by the Modulation of Toll-like Receptor 2, 3 and 4 (Toll-like receptor 2, 3, 4의 신호전달체계 조절을 통한 curcumin의 항암${\cdot}$항염증 효과)

  • Kang, Soon-Ah;Hwang, Daniel;Youn, Hyung-Sun
    • Korean Journal of Food Science and Technology
    • /
    • v.39 no.2
    • /
    • pp.175-180
    • /
    • 2007
  • Toll-like receptors induce innate immune responses recognizing conserved microbial structural molecules that are known as pathogen-associated molecular patterns (PAMPs). Ligand-induced homotypic oligomerization was found to proceed in LPS-induced activation of TLR4 signaling pathways. TLR2 is known to heterodimerize with TLR1 or TLR6 and recognize diacyl- or triacyl-lipopeptide, respectively. These results suggest that ligand-induced receptor dimerization of TLR4 and TLR2 is required for the activation of downstream signaling pathways. Therefore, receptor dimerization may be one of the first lines of regulation in the activation of TLR-mediated signaling pathways and induction of subsequent innate and adaptive immune responses. Here, we report biochemical evidence that curcumin from the plant Curcuma longa inhibits activation of $NF-{\kappa}B$, expression of COX-2, and dimerization of TLRs induced by TLR2, TLR3 and TLR4 agonists. These results imply that curcumin can modulate the activation of TLRs and subsequent immune/inflammatory responses induced by microbial pathogens.

Pyrrole-Derivative of Chalcone, (E)-3-Phenyl-1-(2-Pyrrolyl)-2-Propenone, Inhibits Inflammatory Responses via Inhibition of Src, Syk, and TAK1 Kinase Activities

  • Yang, Sungjae;Kim, Yong;Jeong, Deok;Kim, Jun Ho;Kim, Sunggyu;Son, Young-Jin;Yoo, Byong Chul;Jeong, Eun Jeong;Kim, Tae Woong;Han Lee, In-Sook;Cho, Jae Youl
    • Biomolecules & Therapeutics
    • /
    • v.24 no.6
    • /
    • pp.595-603
    • /
    • 2016
  • (E)-3-Phenyl-1-(2-pyrrolyl)-2-propenone (PPP) is a pyrrole derivative of chalcone, in which the B-ring of chalcone linked to ${\beta}$-carbon is replaced by pyrrole group. While pyrrole has been studied for possible Src inhibition activity, chalcone, especially the substituents on the B-ring, has shown pharmaceutical, anti-inflammatory, and anti-oxidant properties via inhibition of NF-${\kappa}B$ activity. Our study is aimed to investigate whether this novel synthetic compound retains or enhances the pharmaceutically beneficial activities from the both structures. For this purpose, inflammatory responses of lipopolysaccharide (LPS)-treated RAW264.7 cells were analyzed. Nitric oxide (NO) production, inducible NO synthase (iNOS) and tumor necrosis factor-${\alpha}$ (TNF-${\alpha}$) mRNA expression, and the intracellular inflammatory signaling cascade were measured. Interestingly, PPP strongly inhibited NO release in a dose-dependent manner. To further investigate this anti-inflammatory activity, we identified molecular pathways by immunoblot analyses of nuclear fractions and whole cell lysates prepared from LPS-stimulated RAW264.7 cells with or without PPP pretreatment. The nuclear levels of p50, c-Jun, and c-Fos were significantly inhibited when cells were exposed to PPP. Moreover, according to the luciferase reporter gene assay after cotransfection with either TRIF or MyD88 in HEK293 cells, NF-${\kappa}B$-mediated luciferase activity dose-dependently diminished. Additionally, it was confirmed that PPP dampens the upstream signaling cascade of NF-${\kappa}B$ and AP-1 activation. Thus, PPP inhibited Syk, Src, and TAK1 activities induced by LPS or induced by overexpression of these genes. Therefore, our results suggest that PPP displays anti-inflammatory activity via inhibition of Syk, Src, and TAK1 activity, which may be developed as a novel anti-inflammatory drug.

Induction of Dectin-1 Expression and Intracellular Signal Transduction by β-Glucan of Ganoderma lucidum (불로초의 β-Glucan에 의한 Dectin-1 발현 유도와 세포 내 신호전달)

  • Ryu, Han Wook;Kim, Ha Won
    • The Korean Journal of Mycology
    • /
    • v.46 no.2
    • /
    • pp.161-176
    • /
    • 2018
  • Fungal ${\beta}$-glucan, known to have immunostimulatory and antitumor activities, can be recognized by host immune cells as one of the pathogen-associated molecular patterns (PAMPs). Although there are several reports on the diverse immunostimulatory activities of ${\beta}$-glucan, little is known about the intracellular signal transduction of ${\beta}$-glucan. Stimulation of RAW264.7 macrophage cells with ${\beta}$-glucan from Ganoderma lucidum induced the expressions of dectin-1, toll-like receptor 2 (TLR2), TLR4, and TLR6 at the transcription stage. Treatment with ${\beta}$-glucan also induced inflammatory mediators such as macrophage inflammatory proteins (MIP)-$1{\alpha}$, MIP-$1{\beta}$, MIP-$1{\gamma}$, interleukin (IL)-$1{\beta}$, and tumor necrosis factor (TNF)-${\alpha}$. Treatment of the cells with polymyxin B, an inhibitor of lipopolysaccharides (LPS), blocked the induction of inflammatory mediators in LPS- or ${\beta}$-glucan-stimulated systems. Pretreatment of the cells in our cell culture system with LY294002, a phosphoinositide 3-kinase (PI3K) inhibitor, or U0126, a mitogen-activated protein kinase/extracellular-signal-regulated kinase (MAPK/ERK) kinase (MEK)1/MEK2 inhibitor, led to a reduction in the induction of inflammatory mediators in a concentration-dependent manner. These results show that stimulation of the macrophage cells by ${\beta}$-glucan induced the expressions of both dectin-1 and TLRs. We also found that the PI3K/Akt and MEK pathways were involved in the induction of inflammatory mediators in macrophage cells during intracellular signal transduction of ${\beta}$-glucan.

The Effects of Phenethyl Isothiocyanate on Nuclear Factor-κB Activation and Cyclooxygenase-2 and Inducible Nitric Oxide Synthase Expression Induced by Toll-like Receptor Agonists (Phenethyl Isothiocyanate가 Toll-like Receptor Agonists에 의해 유도된 Nuclear Factor-κB 활성과 Cyclooxygenase-2, Inducible Nitric Oxide Synthase 발현에 미치는 효과)

  • Kim, Soo-Jung;Park, Hye-Jeong;Shin, Hwa-Jeong;Kim, Ji-Soo;Ahn, Hee-Jin;Min, In-Soon;Youn, Hyung-Sun
    • Journal of Applied Biological Chemistry
    • /
    • v.54 no.4
    • /
    • pp.279-283
    • /
    • 2011
  • Toll-like receptors (TLRs) play an important role in induction of innate immune responses. The activation of TLRs triggers inflammatory responses that are essential for host defense against invading pathogens. Phenethyl isothiocyanate (PEITC) extracted from cruciferous vegetables has an effect on anti-inflammatory therapy. Dysregulated activation of nuclear factor-${\kappa}$B (NF-${\kappa}$B), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) has been shown to play important roles in the development of certain inflammatory disease. To evaluate the therapeutic potential of PEITC, NF-${\kappa}$B activation and COX-2 and iNOS expression induced by lipopolysaccharide (LPS, TLR4 agonist), polyinosinic-polycytidylic acid (Poly[I:C], TLR3 agonist), 2 kDa macrophageactivating lipopeptide (MALP-2, TLR2 and TLR6 agonist) or oligodeoxynucleotide 1668 (ODN1668, TLR9 agonist) were examined. PEITC inhibits the activation of NF-${\kappa}$B induced by LPS or Poly[I:C] but not by MALP-2 or ODN1668. PEITC also suppressed the iNOS expression induced by LPS or Poly[I:C]. However, PEITC did not suppress COX-2 expression induced by LPS, Poly[I:C], MALP-2, or ODN1668. These results suggest that PEITC has the specific mechanism for antiinflammatory responses.