• 제목/요약/키워드: nlrp3

검색결과 79건 처리시간 0.025초

Polymorphism of NLRP3 Gene and Association with Susceptibility to Digestive Disorders in Rabbit

  • Yang, Yu;Zhang, Gong-Wei;Chen, Shi-Yi;Peng, Jin;Lai, Song-Jia
    • Asian-Australasian Journal of Animal Sciences
    • /
    • 제26권4호
    • /
    • pp.455-462
    • /
    • 2013
  • NLR family pyrin domain containing 3 (NLRP3) is a key component of the inflammasome, whose assembly is a crucial part of the innate immune response. The aim of the present study was to evaluate the association between exon 3 polymorphisms of NLRP3 and the susceptibility to digestive disorders in rabbits. In total, five coding single-nucleotide polymorphisms (cSNPs) were identified; all of which are synonymous. Among them, c.456 C> and c.594 G> were further genotyped for association analysis based on case-control design (n =162 vs n =102). Meanwhile, growing rabbits were experimentally induced to digestive disorders by feeding a fiber-deficient diet, subsequently they were subjected to mRNA expression analysis. Association analysis revealed that haplotype H1 (the two cSNPs: GT) played a potential protective role against digestive disorders (p<0.001). The expression of NLRP3 in the group $H1HX_1$ ($H1HX_1$ is composed of H1H1, H1H3 and H1H4) was the lowest among four groups which were classified by different types of diplotypes. Those results suggested that the NLRP3 gene was significantly associated with susceptibility to digestive disorders in rabbit.

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.

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.

비만 및 제2형 당뇨병 쥐 모델에서 과당과 포도당의 섭취가 지방간과 NLRP3 염증조절결합체에 미치는 영향 (Effects of Dietary Fructose and Glucose on Hepatic Steatosis and NLRP3 Inflammasome in a Rodent Model of Obesity and Type 2 Diabetes)

  • 이희재;양수진
    • 한국식품영양과학회지
    • /
    • 제42권10호
    • /
    • pp.1576-1584
    • /
    • 2013
  • 이상의 결과를 종합해 볼 때, 높은 수준의 포도당 섭취는 공복혈당과 공복혈당면적을 높이고 혈중 인슐린 농도와 아티포넥틴의 수준을 낮추어 혈당조절 능력을 억제시켰다. 반면, 높은 수준의 과당 섭취는 인슐린을 요구하지 않는 과당대사의 특이성으로 인해 혈당조절에는 효과적으로 보인다. 그러나 고과당 섭취는 간 조직 및 혈중 중성지방의 농도를 높이고 염증조절복합체 구성단백질의 발현을 조절하여 전염증인자의 발현을 증가시켰다. 이는 간조직에 있어 과당이 포도당보다 높은 수준의 염증반응을 유도하여 NAFLD의 발병과 진행에 보다 유의적인 영향을 준다는 것을 보여준다. 본 연구의 제한점은 일상에서 과당이나 포도당을 단독으로 섭취하는 경우가 드물다는 점과 3주라는 짧은 중재기간에 의한 실험 결과라는 것이다. 앞으로의 연구는 단순당을 장기간 섭취했을 때 혈청과 간 조직을 포함한 다른 대사 관련조직에서 나타나는 변화에 초점을 맞출 필요가 있으며, 염증조절복합체가 염증인자의 발현을 증가시키는 기전을 구체화하는 것이 요구된다.

Production of IL-1β and Inflammasome with Up-Regulated Expressions of NOD-Like Receptor Related Genes in Toxoplasma gondii-Infected THP-1 Macrophages

  • Chu, Jia-Qi;Shi, Ge;Fan, Yi-Ming;Choi, In-Wook;Cha, Guang-Ho;Zhou, Yu;Lee, Young-Ha;Quan, Juan-Hua
    • Parasites, Hosts and Diseases
    • /
    • 제54권6호
    • /
    • pp.711-717
    • /
    • 2016
  • Toxoplasma gondii is an obligate intracellular parasite that stimulates production of high levels of proinflammatory cytokines, which are important for innate immunity. NLRs, i.e., nucleotide-binding oligomerization domain (NOD)-like receptors, play a crucial role as innate immune sensors and form multiprotein complexes called inflammasomes, which mediate caspase-1-dependent processing of $pro-IL-1{\beta}$. To elucidate the role of inflammasome components in T. gondiiinfected THP-1 macrophages, we examined inflammasome-related gene expression and mechanisms of inflammasome-regulated cytokine $IL-1{\beta}$ secretion. The results revealed a significant upregulation of $IL-1{\beta}$ after T. gondii infection. T. gondii infection also upregulated the expression of inflammasome sensors, including NLRP1, NLRP3, NLRC4, NLRP6, NLRP8, NLRP13, AIM2, and NAIP, in a time-dependent manner. The infection also upregulated inflammasome adaptor protein ASC and caspase-1 mRNA levels. From this study, we newly found that T. gondii infection regulates NLRC4, NLRP6, NLRP8, NLRP13, AIM2, and neuronal apoptosis inhibitor protein (NAIP) gene expressions in THP-1 macrophages and that the role of the inflammasome-related genes may be critical for mediating the innate immune responses to T. gondii infection.

BV-2 미세아교세포에서 메트포르민 또는 알파-리포산의 염증반응과 NLRP3 인플라마솜 약화에 관한 연구 (Metformin or α-Lipoic Acid Attenuate Inflammatory Response and NLRP3 Inflammasome in BV-2 Microglial Cells)

  • 최혜림;하지선;김인식;양승주
    • 대한임상검사과학회지
    • /
    • 제52권3호
    • /
    • pp.253-260
    • /
    • 2020
  • 알츠하이머 병은 인지 기능 저하로 인한 치매 발생으로 설명할 수 있는 만성 및 진행성 신경 퇴행성 질환이다. 알츠하이머 병의 특징은 세포 외 및 세포 내 아밀로이드 플라크의 형성이다. 아밀로이드 베타는 알츠하이머 병의 특징이며 미세아교세포는 아밀로이드 베타의 존재하에 활성화될 수 있다. 활성화된 미세아교세포는 전 염증성 사이토카인을 분비한다. 게다가, S100A9는 염증의 중요한 선천성 전 염증 기여자이며 알츠하이머 병에 잠재적인 기여자로 알려져 있다. 이 연구는 아밀로이드 베타 및 S100A9이 처리된 BV-2 세포에서 염증반응 및 NLRP3 인플라마솜 활성화에 대한 메트포르민 및 알파리포산의 효과를 조사했다. 메트포르민과 알파-리포산은 종양 괴사 인자-알파 및 일터루킨-6와 같은 염증성 사이토카인을 약화시킨다. 또한 메트포르민과 알파-리포산은 JNK, ERK, p38의 인산화를 억제하고, NF-kB 경로 및 NLRP3 인플라마솜의 활성화를 억제했다. 또한 메트포르민과 알파-리포산은 M1 표현형인 ICAM1의 수준을 감소시킨 반면 M2 표현형인 ARG1은 증가시켰다. 이러한 발견은 메트포르민과 알파-리포산이 아밀로이드베타 및 S100A9에 의한 신경 염증 반응에 대한 치료제가 될 수 있음을 시사한다.

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.

Pneumococcal Δpep27 Immunization Attenuates TLRs and NLRP3 Expression and Relieves Murine Ovalbumin-Induced Allergic Rhinitis

  • Yu, Jae Ik;Kim, Ji-Hoon;Nam, Ki-El;Lee, Wonsik;Rhee, Dong-Kwon
    • Journal of Microbiology and Biotechnology
    • /
    • 제32권6호
    • /
    • pp.709-717
    • /
    • 2022
  • Allergic rhinitis (AR), one of the most common inflammatory diseases, is caused by immunoglobulin E (IgE)-mediated reactions against inhaled allergens. AR involves mucosal inflammation driven by type 2 helper T (Th2) cells. Previously, it was shown that the Streptococcus pneumoniae pep27 mutant (Δpep27) could prevent and treat allergic asthma by reducing Th2 responses. However, the underlying mechanism of Δpep27 immunization in AR remains undetermined. Here, we investigated the role of Δpep27 immunization in the development and progression of AR and elucidated potential mechanisms. In an ovalbumin (OVA)-induced AR mice model, Δpep27 alleviated allergic symptoms (frequency of sneezing and rubbing) and reduced TLR2 and TLR4 expression, Th2 cytokines, and eosinophil infiltration in the nasal mucosa. Mechanistically, Δpep27 reduced the activation of the NLRP3 inflammasome in the nasal mucosa by down-regulating the Toll-like receptor signaling pathway. In conclusion, Δpep27 seems to alleviate TLR signaling and NLRP3 inflammasome activation to subsequently prevent AR.

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.

Diosmetin Alleviates Lipopolysaccharide-Induced Acute Lung Injury through Activating the Nrf2 Pathway and Inhibiting the NLRP3 Inflammasome

  • Liu, Qinmei;Ci, Xinxin;Wen, Zhongmei;Peng, Liping
    • Biomolecules & Therapeutics
    • /
    • 제26권2호
    • /
    • pp.157-166
    • /
    • 2018
  • Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a common clinical syndrome of diffuse lung inflammation with high mortality rates and limited therapeutic methods. Diosmetin, an active component from Chinese herbs, has long been noticed because of its antioxidant and anti-inflammatory activities. The aim of this study was to evaluate the effects of diosmetin on LPS-induced ALI model and unveil the possible mechanisms. Our results revealed that pretreatment with diosmetin effectively alleviated lung histopathological changes, which were further evaluated by lung injury scores. Diosmetin also decreased lung wet/dry ratios, as well as total protein levels, inflammatory cell infiltration and proinflammatory cytokine (eg. $TNF-{\alpha}$, $IL-1{\beta}$ and IL-6) overproduction in bronchoalveolar lavage fluid (BALF). Additionally, increased MPO, MDA and ROS levels induced by LPS were also markly suppressed by diosmetin. Furthermore, diosmetin significantly increased the expression of Nrf2 along with its target gene HO-1 and blocked the activation of NLRP3 inflammasome in the lung tissues, which might be central to the protective effects of diosmetin. Further supporting these results, in vitro experiments also showed that diosmetin activated Nrf2 and HO-1, as well as inhibited the NLRP3 inflammasome in both RAW264.7 and A549 cells. The present study highlights the protective effects of diosmetin on LPS-induced ALI via activation of Nrf2 and inhibition of NLRP3 inflammasome, bringing up the hope of its application as a therapeutic drug towards LPS-induced ALI.