• Title/Summary/Keyword: inflammasome

검색결과 89건 처리시간 0.021초

NLRP3 Inflammasome as Therapeutic Targets in Inflammatory Diseases

  • Annamneedi Venkata Prakash;Il-Ho Park;Jun Woo Park;Jae Pil Bae;Geum Seon Lee;Tae Jin Kang
    • Biomolecules & Therapeutics
    • /
    • 제31권4호
    • /
    • pp.395-401
    • /
    • 2023
  • Innate immunity is a first line defence system in the body which is for sensing signals of danger such as pathogenic microbes or host-derived signals of cellular stress. Pattern recognition receptors (PRR's), which present in the cell memebrane, are suspect the infection through pathogen-associated molecular patterns (PAMP), and activate innate immunity with response to promote inflammation via inflammatory cells such as macrophages and neutrophils, and cytokines. Inflammasome are protein complexes which are part of innate immunity in inflammation to remove pathogens and repair damaged tissues. What is the important role of inflammation in disease? In this review, we are focused on the action mechanism of NLRP3 inflammasome in inflammatory diseases such as asthma, atopic dermatitis, and sepsis.

Cobalt Chloride-induced Hypoxia Ameliorates NLRP3-Mediated Caspase-1 Activation in Mixed Glial Cultures

  • Kim, Eun-Hee;Won, Ji-Hee;Hwang, Inhwa;Yu, Je-Wook
    • IMMUNE NETWORK
    • /
    • 제13권4호
    • /
    • pp.141-147
    • /
    • 2013
  • Hypoxia has been shown to promote inflammation, including the release of proinflammatory cytokines, but it is poorly investigated how hypoxia directly affects inflammasome signaling pathways. To explore whether hypoxic stress modulates inflammasome activity, we examined the effect of cobalt chloride ($CoCl_2$)-induced hypoxia on caspase-1 activation in primary mixed glial cultures of the neonatal mouse brain. Unexpectedly, hypoxia induced by oxygen-glucose deprivation or $CoCl_2$ treatment failed to activate caspase-1 in microglial BV-2 cells and primary mixed glial cultures. Of particular interest, $CoCl_2$-induced hypoxic condition considerably inhibited NLRP3-dependent caspase-1 activation in mixed glial cells, but not in bone marrow-derived macrophages. $CoCl_2$-mediated inhibition of NLRP3 inflammasome activity was also observed in the isolated brain microglial cells, but $CoCl_2$ did not affect poly dA:dT-triggered AIM2 inflammasome activity in mixed glial cells. Our results collectively demonstrate that $CoCl_2$-induced hypoxia may negatively regulate NLRP3 inflammasome signaling in brain glial cells, but its physiological significance remains to be determined.

Triptolide improves myocardial fibrosis in rats through inhibition of nuclear factor kappa B and NLR family pyrin domain containing 3 inflammasome pathway

  • Shen, Jianyao;Ma, Hailiang;Wang, Chaoquan
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제25권6호
    • /
    • pp.533-543
    • /
    • 2021
  • Myocardial fibrosis (MF) is the result of persistent and repeated aggravation of myocardial ischemia and hypoxia, leading to the gradual development of heart failure of chronic ischemic heart disease. Triptolide (TPL) is identified to be involved in the treatment for MF. This study aims to explore the mechanism of TPL in the treatment of MF. The MF rat model was established, subcutaneously injected with isoproterenol and treated by subcutaneous injection of TPL. The cardiac function of each group was evaluated, including LVEF, LVFS, LVES, and LVED. The expressions of ANP, BNP, inflammatory related factors (IL-1β, IL-18, TNF-α, MCP-1, VCAM1), NLRP3 inflammasome factors (NLRP3, ASC) and fibrosis related factors (TGF-β1, COL1, and COL3) in rats were dete cted. H&E staining and Masson staining were used to observe myocardial cell inflammation and fibrosis of rats. Western blot was used to detect the p-P65 and t-P65 levels in nucleoprotein of rat myocardial tissues. LVED and LVES of MF group were significantly upregulated, LVEF and LVFS were significantly downregulated, while TPL treatment reversed these trends; TPL treatment downregulated the tissue injury and improved the pathological damage of MF rats. TPL treatment downregulated the levels of inflammatory factors and fibrosis factors, and inhibited the activation of NLRP3 inflammasome. Activation of NLRP3 inflammasome or NF-κB pathway reversed the effect of TPL on MF. Collectively, TPL inhibited the activation of NLRP3 inflammasome by inhibiting NF-κB pathway, and improved MF in MF rats.

Cardamonin Inhibited IL-1β Induced Injury by Inhibition of NLRP3 Inflammasome via Activating Nrf2/NQO-1 Signaling Pathway in Chondrocyte

  • Jiang, Jianqing;Cai, Mingsong
    • Journal of Microbiology and Biotechnology
    • /
    • 제31권6호
    • /
    • pp.794-802
    • /
    • 2021
  • In this study we investigated the role and mechanism of cardamonin on IL-1β induced injury in OA. CHON-001 cells were treated with cardamonin and IL-1β and transfected with silencing nuclear factor erythroid 2-related factor 2 (siNrf2). Cell viability was detected by Cell Counting Kit-8 assay and flow cytometer assay was utilized for cell apoptosis assessment. IL-6, IL-8, TNF-α and Nrf2 mRNA expression was tested by qRT-PCR. Western blot was employed to evaluate MMP-3, MMP-13, Collagen II, Nrf2, NQO-1, NLRP3, Caspase 1 and apoptosis-associated speck-like protein containing a caspase-1 recruitment domain (ASC) protein levels. In CHON-001 cells, IL-1β suppressed cell viability and Collagen II level while promoting cell apoptosis and expression of pro-inflammatory cytokines (IL-6, IL-8, TNF-α), MMPs (MMP-3, MMP-13), NQO-1, and NLRP3 inflammasome (NLRP3, Caspase 1 and ASC), with no significant influence on Nrf2. Cardamonin reversed the effect of IL-1β on cell viability, cell apoptosis, pro-inflammatory cytokines, MMPs, Collagen II, and NLRP3 inflammasome levels. In addition, cardamonin advanced Nrf2 and NQO-1 expression of CHON-001 cells. SiNrf2 reversed the function of cardamonin on IL-1β-induced cell apoptosis and expression of pro-inflammatory cytokines, Nrf2, NQO-1, and NLRP3 inflammasome in chondrocytes. Taken together Cardamonin inhibited IL-1β induced injury by inhibition of NLRP3 inflammasome via activating Nrf2/NQO1 signaling pathway in chondrocyte.

Korean Red Ginseng attenuates ultraviolet-mediated inflammasome activation in keratinocytes

  • Ahn, Huijeong;Han, Byung-Cheol;Hong, Eui-Ju;An, Beum-Soo;Lee, Eunsong;Lee, Seung-Ho;Lee, Geun-Shik
    • Journal of Ginseng Research
    • /
    • 제45권3호
    • /
    • pp.456-463
    • /
    • 2021
  • Background: Keratinocytes form a physical barrier and act as an innate immune cell in skin. Keratinocytes secrete pro-inflammatory cytokines, such as interleukin (IL)-1β, resulting from inflammasome activation when exposed to ultraviolet (UV) irradiation. Korean Red Ginseng extracts (RGE) have been well-studied as modulators of inflammasome activation in immune cells, such as macrophages. In the study, we elucidated the role of RGE on the UV-mediated inflammasome activation in keratinocytes compared with that in macrophages. Methods: Human skin keratinocyte cells (HaCaT), human epidermal keratinocytes (HEK), human monocyte-like cells (THP-1), and mouse macrophages were treated with RGE or a saponin fraction (SF) or non-saponin fraction (NS) of RGE before and after UV irradiation. The secretion levels of IL-1β, as an indicator of inflammasome activation, were analyzed. Results: The treatment of RGE or SF in macrophages after UV irradiation inhibited IL-1β secretion, but similar treatment in HaCaT cells did not. However, the treatment of RGE or SF in HaCaT cells in the presence of poly I:C, a toll-like receptor (TLR) 3 ligand, before UV exposure elicited the inhibition of the IL-1β secretion. The inhibition was caused by the disruption by RGE or SF of the TLR mediating up-regulation of the pro-IL-1β and NLRP3 genes during the priming step. Conclusion: RGE and its saponins inhibit IL-1β secretion in response to UV exposure in both keratinocytes and macrophages. In particular, RGE treatment interrupted only the priming step in keratinocytes, although it did attenuate both the priming and activation steps in macrophages.

NLRP3 인플라마좀 작용 기전 및 신경 질환에서의 역할 (NLRP3 Inflammasome in Neuroinflammatory Disorders)

  • 김지희;김영희
    • 생명과학회지
    • /
    • 제31권2호
    • /
    • pp.237-247
    • /
    • 2021
  • 신경염증(neuroinflammation)은 여러 신경 질환의 원인 인자로 확인되고있다. 중추 신경계에 발현되는 단백질 복합체인 NLRP3 인플라마좀은 미생물, 응집되고 잘못 접힌 단백질, ATP와 같은 광범위한 외인성 및 내인성 자극에 의해 감지되고 캐스페이즈-1(capase-1)을 활성화할 수 있다. 활성을 띠는 캐스페이즈-1은 IL-1b와 IL-18과 같은 염증성 사이토카인(pro-inflammatory cytokine)을 활성화시키고 급속한 세포사멸(파이롭토시스, pyroptosis)를 야기한다. IL-1b와 IL-18, 그리고 파이롭토시스를 통해 분비된 DAMPs은 다양한 신호 전달 경로를 통해 신경염증 반응을 유도하여 신경 손상을 유발한다. 따라서 NLRP3 인플라마좀은 신경염증으로 인한 여러 가지 신경질환 발병에 중요한 역할을 할 것으로 여겨진다. 본 리뷰 에서는 NLRP3 인플라마좀의 구조와 활성화에 대해 간략히 알아 보고 다양한 형태의 신경 질환에서 NLRP3 인플라마좀의 역할에 대해 논의하고자 한다.

The activation of NLRP3 inflammasome potentiates the immunomodulatory abilities of mesenchymal stem cells in a murine colitis model

  • Ahn, Ji-Su;Seo, Yoojin;Oh, Su-Jeong;Yang, Ji Won;Shin, Ye Young;Lee, Byung-Chul;Kang, Kyung-Sun;Sung, Eui-Suk;Lee, Byung-Joo;Mohammadpour, Hemn;Hur, Jin;Shin, Tae-Hoon;Kim, Hyung-Sik
    • BMB Reports
    • /
    • 제53권6호
    • /
    • pp.329-334
    • /
    • 2020
  • Inflammasomes are cytosolic, multiprotein complexes that act at the frontline of the immune responses by recognizing pathogen- or danger-associated molecular patterns or abnormal host molecules. Mesenchymal stem cells (MSCs) have been reported to possess multipotency to differentiate into various cell types and immunoregulatory effects. In this study, we investigated the expression and functional regulation of NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome in human umbilical cord blood-derived MSCs (hUCB-MSCs). hUCB-MSCs expressed inflammasome components that are necessary for its complex assembly. Interestingly, NLRP3 inflammasome activation suppressed the differentiation of hUCB-MSCs into osteoblasts, which was restored when the expression of adaptor proteins for inflammasome assembly was inhibited. Moreover, the suppressive effects of MSCs on T cell responses and the macrophage activation were augmented in response to NLRP3 activation. In vivo studies using colitic mice revealed that the protective abilities of hUCB-MSCs increased after NLRP3 stimulation. In conclusion, our findings suggest that the NLRP3 inflammasome components are expressed in hUCB-MSCs and its activation can regulate the differentiation capability and the immunomodulatory effects of hUCB-MSCs.

라스베라트롤 투여가 고지방식이 비만쥐의 지방조직에서의 inflammasome과 대식세포 마커에 미치는 영향 (Resveratrol Ameliorates High-fat-induced Metabolic Complications by Changing the Expression of Inflammasome Markers and Macrophage M1 and M2 Markers in Obese Mice)

  • 이영란;피핏 피트리아니;박희근;이왕록
    • 생명과학회지
    • /
    • 제27권12호
    • /
    • pp.1462-1469
    • /
    • 2017
  • 본 연구 목적은 고지방식이 유도 비만 쥐의 피하지방조직에서 라스베라트롤 투여가 대식세포 침윤관련 염증인자에 미치는 영향을 규명하고자 하였다. 본 연구를 위해 정상식이군, 고지방식이군, 고지방식이+라스베라트롤 투여군으로 분류한 후, 라스베라트롤 투여군은 15주간 25 mg/kg 농도로 Dimethyl Sulfoxide에 용해하여 투여하였으며, 비교군은 Dimethyl Sulfoxide 용액만을 투여하였다. 연구결과 고지방식이군은 정상식이군에 비하여 체중이 유의하게 증가하였고, 라스베라트롤 투여군에서 고지방식이 군보다 NLRP3. ASC, Casepase1 mRNA 발현이 감소하였다. 또한 염증마커로 알려진 IL-18 mRNA 발현이 라스베라트롤 투여군에서 정상식이군과 고지방식이군보다 낮게 나타났다. 대식세포 침윤 마커인 F480, CD86 mRNA 발현에서도 라스베라트롤 투여군에서 고지방식이 군보다 유의한 감소를 보였다. 따라서 라스베라트롤 투여는 고지방식이 유도 비만 상황에서 대식세포 침윤 염증과 inflammasome에 긍정적인 영향을 미치는 것으로 보여진다.

식물 자원을 활용한 염증반응 조절 (Modulation of Inflammation by Plant Resources)

  • 이하늘;성수희;김보람;김진호;서찬;임수아;김정은;정지민;정진우
    • 한국자원식물학회:학술대회논문집
    • /
    • 한국자원식물학회 2023년도 임시총회 및 춘계학술대회
    • /
    • pp.17-17
    • /
    • 2023
  • Chrysanthemum zawadskii (C. zawadskii) is used in traditional East Asian medicine for the treatment of various diseases, including inflammatory disease. However, it has remained unclear whether extracts of C. zawadskii inhibit inflammasome activation in macrophages. The present study assessed the inhibitory effect of an ethanol extract of C. zawadskii (CZE) on the activation of the inflammasome in macrophages and the underlying mechanism. Bone marrow[-derived macrophages (BMDMs) were obtained from wild-type C57BL/6 mice. The release of IL-1β and lactate dehydrogenase in response to nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome activators, such as ATP, nigericin and monosodium urate (MSU) crystals, was significantly decreased by CZE in lipopolysaccharide(LPS)-primed BMDMs. Western blotting revealed that CZE inhibited ATP-induced caspase-1 cleavage and IL-1β maturation. To investigate whether CZE inhibits the priming step of the NLRP3 inflammasome, we confirmed the role of CZE at the gene level using RT-qPCR. CZE also downregulated the gene expression of NLRP3 and pro-IL-1β as well as NF-κB activation in BMDMs in response to LPS. Apoptosis associated speck-like protein containing a caspase-recruitment domain (CARD) oligomerization and speck formation by NLRP3 inflammasome activators were suppressed by CZE. By contrast, CZE did not affect NLR family CARD domain containing protein 4 (NLRC4) or absent in melanoma 2 (AIM2) inflammasome activation in response to Salmonella typhimurium and poly(dA:dT) in LPS-primed BMDMs, respectively. The results revealed that three key components of CZE, namely linarin, 3,5-dicaffeoylquinic acid and chlorogenic acid, decreased IL-1β secretion in response to ATP, nigericin and MSU. These findings suggest that CZE effectively inhibited activation of the NLRP3 inflammasome.

  • PDF

Prolonged Exposure to Lipopolysaccharide Induces NLRP3-Independent Maturation and Secretion of Interleukin (IL)-1β in Macrophages

  • Hong, Sujeong;Yu, Je-Wook
    • Journal of Microbiology and Biotechnology
    • /
    • 제28권1호
    • /
    • pp.115-121
    • /
    • 2018
  • Upon sensing of microbial infections or endogenous danger signals in macrophages, inflammasome signaling plays a significant role in triggering inflammatory responses via producing interleukin (IL)-$1{\beta}$. Recent studies revealed that active caspase-1, a product of the inflammasome complex, causes maturation of inactive pro-IL-$1{\beta}$ into the active form. However, the underlying mechanism by which this leaderless cytokine is secreted into the extracellular space remains to be elucidated. In this study, we demonstrated that prolonged lipopolysaccharide (LPS) treatment to macrophages could trigger the unexpected maturation and extracellular release of IL-$1{\beta}$ through a nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 (NLRP3)-independent manner. Short-term treatment (less than 6 h) of LPS induced robust production of the IL-$1{\beta}$ precursor form inside cells but did not promote the maturation and secretion of IL-$1{\beta}$ in bone marrow-derived macrophages or peritoneal macrophages. Instead, prolonged LPS treatment (more than 12 h) led to a significant release of matured IL-$1{\beta}$ with no robust indication of caspase-1 activation. Intriguingly, this LPS-triggered secretion of IL-$1{\beta}$ was also observed in NLRP3-deficient macrophages. In addition, this unexpected IL-$1{\beta}$ release was only partially impaired by a caspase-1 and NLRP3 inflammasome inhibitor. Collectively, our results propose that prolonged exposure to LPS is able to drive the maturation and secretion of IL-$1{\beta}$ in an NLRP3 inflammasome-independent manner.