• 제목/요약/키워드: NLRP3 inflammasome activation

검색결과 49건 처리시간 0.024초

Mitophagy: a balance regulator of NLRP3 inflammasome activation

  • Kim, Min-Ji;Yoon, Joo-Heon;Ryu, Ji-Hwan
    • BMB Reports
    • /
    • 제49권10호
    • /
    • pp.529-535
    • /
    • 2016
  • The NLRP3 inflammasome is activated by a variety of external or host-derived stimuli and its activation initiates an inflammatory response through caspase-1 activation, resulting in inflammatory cytokine IL-1β maturation and secretion. The NLRP3 inflammasome activation is a kind of innate immune response, most likely mediated by myeloid cells acting as a host defense mechanism. However, if this activation is not properly regulated, excessive inflammation induced by overactivated NLRP3 inflammasome can be detrimental to the host, causing tissue damage and organ dysfunction, eventually causing several diseases. Previous studies have suggested that mitochondrial damage may be a cause of NLRP3 inflammasome activation and autophagy, which is a conserved self-degradation process that negatively regulates NLRP3 inflammasome activation. Recently, mitochondria-selective autophagy, termed mitophagy, has emerged as a central player for maintaining mitochondrial homeostasis through the elimination of damaged mitochondria, leading to the prevention of hyperinflammation triggered by NLRP3 inflammasome activation. In this review, we will first focus on the molecular mechanisms of NLRP3 inflammasome activation and NLRP3 inflammasome-related diseases. We will then discuss autophagy, especially mitophagy, as a negative regulator of NLPP3 inflammasome activation by examining recent advances in research.

RAW 264.7 대식세포에서 마늘 유래 황 함유 화합물에 의한 요산 유도 inflammasome 활성화의 억제는 ROS 생성 차단과 연관성이 있음 (Suppression of Monosodium Urate-induced NLRP3 Inflammasome Activation by Garlic-derived Sulfur-containing Phytochemicals is Associated with Blocking ROS Generation in RAW 264.7 Macrophages)

  • 김민영;최영현
    • 생명과학회지
    • /
    • 제33권4호
    • /
    • pp.349-356
    • /
    • 2023
  • 만성 염증성 관절염 질환인 통풍은 고요산혈증을 특징으로 하며 NLRP3 inflammasome의 활성화에 따른 IL-1β와 같은 염증성 cytokine 방출과 연관된 MSU에 대한 염증 반응에 의해 유발될 수 있다. 마늘에 함유된 황 함유 화합물은 다양한 질병에 대한 잠재적인 유익한 약리학적 효능을 가지지만, NLRP3 inflammasome 활성화와 연관된 통풍 억제에 대한 효능은 현재까지 입증되지 않았다. 본 연구에서는 대표적인 마늘 유래 황화합물인 DADS와 DATS가 MSU에 의한 NLRP3 inflammasome 활성을 억제할 수 있는지를 조사하였다. 본 연구의 결과에 의하면, 비세포 독성 조건에서 DADS와 DATS는 LPS가 전처리된 RAW 264.7 대식세포에서 MSU에 대한 반응으로 증가된 NO의 생성과 IL-1β 유리를 유의적으로 차단하였다. DADS와 DATS는 또한 증가된 NLRP3, ASC, caspase-1 p20 및 IL-1β의 발현을 감소시켰으며, 이는 MSU로 유도된 LRP3 inflammasome 활성화가 DADS와 DATS에 의해 억제되었음을 의미한다. 아울러, DADS와 DATS는 NLRP3 inflammasome 활성화에 상위 신호로 작용하는 산화적 스트레스를 차단했으며 ROS 생성을 제거한다는 사실에서 입증되었다. 결론적으로, 본 연구의 결과는 DADS와 DATS가 ROS/NLRP3 경로를 억제하여 inflammasome 활성화를 차단함으로서 NLRP3 의존성 통풍성 관절염 치료를 위한 잠재력이 우수함을 의미한다.

Nonsaponin fractions of Korean Red Ginseng extracts prime activation of NLRP3 inflammasome

  • Han, Byung-Cheol;Ahn, Huijeong;Lee, Jiseon;Jeon, Eunsaem;Seo, Sanghoon;Jang, Kyoung Hwa;Lee, Seung-Ho;Kim, Cheon Ho;Lee, Geun-Shik
    • Journal of Ginseng Research
    • /
    • 제41권4호
    • /
    • pp.513-523
    • /
    • 2017
  • Background: Korean Red Ginseng extracts (RGE) have been suggested as effective immune modulators, and we reported that ginsenosides possess anti-inflammasome properties. However, the properties of nonsaponin components of RGE have not been well studied. Methods: To assess the roles of nonsaponin fractions (NS) in NLRP3 inflammasome activation, we treated murine macrophages with or without first or second inflammasome activation signals with RGE, NS, or saponin fractions (SF). The first signal was nuclear factor kappa-light-chain-enhancer of activated B cells (NF-${\kappa}B$)-mediated transcription of pro-interleukin (IL)-$1{\beta}$ and NLRP3 while the second signal triggered assembly of inflammasome components, leading to IL-$1{\beta}$ maturation. In addition, we examined the role of NS in IL-6 production and IL-$1{\beta}$ maturation in mice. Results: NS induced IL-$1{\beta}$ and NLRP3 transcription via toll-like receptor 4 signaling, whereas SF blocked expression. During the second signal, SF attenuated NLRP3 inflammasome activation while NS did not. Further, NS-injected mice presented increased IL-$1{\beta}$ maturation and IL-6 production. Conclusion: SF and NS of RGE play differential roles in the NLRP3 inflammasome activation. Hence, RGE can be suggested as an NLRP3 inflammasome modulator.

Loganin Prevents Hepatic Steatosis by Blocking NLRP3 Inflammasome Activation

  • Joo Hyeon Jang;Gabsik Yang;Jin Kyung Seok;Han Chang Kang;Yong-Yeon Cho;Hye Suk Lee;Joo Young Lee
    • Biomolecules & Therapeutics
    • /
    • 제31권1호
    • /
    • pp.40-47
    • /
    • 2023
  • Activation of the NLRP3 inflammasome is a necessary process to induce fibrosis in nonalcoholic fatty liver disease (NAFLD). Nonalcoholic steatohepatitis (NASH) is a kind of NAFLD that encompasses the spectrum of liver disease. It is characterized by inflammation and ballooning of hepatocytes during steatosis. We tested whether inhibiting the NLRP3 inflammasome could prevent the development and pathology of NASH. We identified loganin as an inhibitor of the NLRP3 inflammasome and investigated whether in vivo administration of loganin prevented NASH symptoms using a methionine-choline deficient (MCD) diet model in mice. We found that loganin inhibited the NLRP3 inflammasome activation triggered by ATP or nigericin, as shown by suppression of the production of interleukin (IL)-1β and caspase-1 (p10) in mouse primary macrophages. The speck formation of apoptosisassociated speck-like protein containing a caspase recruitment domain (ASC) was blocked by loganin, showing that the assembly of the NLRP3 inflammasome complex was impaired by loganin. Administration of loganin reduced the clinical signs of NASH in mice fed the MCD diet, including hepatic inflammation, fat accumulation, and fibrosis. In addition, loganin reduced the expression of NLRP3 inflammasome components in the liver. Our findings indicate that loganin alleviates the inflammatory symptoms associated with NASH, presumably by inhibiting NLRP3 inflammasome activation. In summary, these findings imply that loganin may be a novel nutritional and therapeutic treatment for NASH-related inflammation.

Inflammasomes: Molecular Regulation and Implications for Metabolic and Cognitive Diseases

  • Choi, Alexander J.S.;Ryter, Stefan W.
    • Molecules and Cells
    • /
    • 제37권6호
    • /
    • pp.441-448
    • /
    • 2014
  • Inflammasomes are specialized signaling platforms critical for the regulation of innate immune and inflammatory responses. Various NLR family members (i.e., NLRP1, NLRP3, and IPAF) as well as the PYHIN family member AIM2 can form inflammasome complexes. These multiprotein complexes activate inflammatory caspases (i.e., caspase-1) which in turn catalyze the maturation of select pro-inflammatory cytokines, including interleukin (IL)-$1{\beta}$ and IL-18. Activation of the NLRP3 inflammasome typically requires two initiating signals. Toll-like receptor (TLR) and NOD-like receptor (NLR) agonists activate the transcription of pro-inflammatory cytokine genes through an NF-${\kappa}B$-dependent priming signal. Following exposure to extracellular ATP, stimulation of the P2X purinoreceptor-7 ($P2X_7R$), which results in $K^+$ efflux, is required as a second signal for NLRP3 inflammasome formation. Alternative models for NLRP3 activation involve lysosomal destabilization and phagocytic NADPH oxidase and /or mitochondria-dependent reactive oxygen species (ROS) production. In this review we examine regulatory mechanisms that activate the NLRP3 inflammasome pathway. Furthermore, we discuss the potential roles of NLRP3 in metabolic and cognitive diseases, including obesity, type 2 diabetes mellitus, Alzheimer's disease, and major depressive disorder. Novel therapeutics involving inflammasome activation may result in possible clinical applications in the near future.

Interruption of Helicobacter pylori-Induced NLRP3 Inflammasome Activation by Chalcone Derivatives

  • Choi, Hye Ri;Lim, Hyun;Lee, Ju Hee;Park, Haeil;Kim, Hyun Pyo
    • Biomolecules & Therapeutics
    • /
    • 제29권4호
    • /
    • pp.410-418
    • /
    • 2021
  • Helicobacter pylori causes chronic gastritis through cag pathogenicity island (cagPAI), vacuolating cytotoxin A (VacA), lipopolysaccharides (LPS), and flagellin as pathogen-related molecular patterns (PAMPs), which, in combination with the pattern recognition receptors (PRRs) of host cells promotes the expression and secretion of inflammation-causing cytokines and activates innate immune responses such as inflammasomes. To identify useful compounds against H. pylori-associated gastric disorders, the effect of chalcone derivatives to activate the nucleotide-binding oligomerization domain (NOD)-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome was examined in an H. pylori-infected human monocytic THP-1 cell line in this study. Among the five synthetic structurally-related chalcone derivatives examined, 2'-hydroxy-4',6'-dimethoxychalcone (8) and 2'-hydroxy-3,4,5-trimethoxychalcone (12) strongly blocked the NLRP3 inflammasome in H. pylori-infected THP-1 cells. At 10 μM, these compounds inhibited the production of active IL-1β, IL-18, and caspase-1, and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) oligomerization, but did not affect the expression levels of NLRP3, ASC, and pro-caspase-1. The interruption of NLRP3 inflammasome activation by these compounds was found to be mediated via the inhibition of the interleukin-1 receptor-associated kinase 4 (IRAK4)/IκBα/NF-κB signaling pathway. These compounds also inhibited caspase-4 production associated with non-canonical NLRP3 inflammasome activation. These results show for the first time that certain chalcones could interrupt the activation of the NLRP3 inflammasome in H. pylori-infected THP-1 cells. Therefore, these chalcones may be helpful in alleviating H. pylori-related inflammatory disorders including chronic gastritis.

Fructose-arginine, a non-saponin molecule of Korean Red Ginseng, attenuates AIM2 inflammasome activation

  • Ahn, Huijeong;Han, Byung-Cheol;Lee, Seung-Ho;Lee, Geun-Shik
    • Journal of Ginseng Research
    • /
    • 제44권6호
    • /
    • pp.808-814
    • /
    • 2020
  • Background: Korean Red Ginseng extract (RGE) has been reported to act as an inflammasome modulator. Ginsenosides, saponin molecules of RGE, selectively inhibit activation of NLRP3 and AIM2 inflammasomes, while non-saponin molecules of RGE upregulate inflammasome components associated with the initiation of NLRP3 inflammasome activation. In this study, we investigated the effect of non-saponin components of RGE on AIM2 inflammasome activation. Methods: The role of non-saponins of RGE on AIM2 inflammasomes was tested in mouse bone marrow-derived macrophages, a human monocyte-like cell line, and a mouse animal model. Cells or mice were transfected with dsDNA or inoculated with Listeria monocytogenes to activate AIM2 inflammasomes. Several indices of inflammasome activation were examined via immunoblot or ELISA analysis. Results: The non-saponin fraction and saponin-eliminating fraction (SEF) of RGE selectively attenuated the activation of AIM2 inflammasomes, but not that of NLRP3 or NLRC4 inflammasomes. Fructose-arginine, an amino-sugar, was shown to be effective against AIM2 inflammasome activation. Conclusion: Non-saponins of RGE, such as fructose-arginine, might be effective in regulating infectious and autoimmune diseases resulting from AIM2 inflammasome activation.

Korean Red Ginseng, a regulator of NLRP3 inflammasome, in the COVID-19 pandemic

  • Jung, Eui-Man;Lee, Geun-Shik
    • Journal of Ginseng Research
    • /
    • 제46권3호
    • /
    • pp.331-336
    • /
    • 2022
  • Coronavirus disease 2019 (COVID-19) exhibits various symptoms, ranging from asymptomatic to severe pneumonia or death. The major features of patients in severe COVID-19 are the dysregulation of cytokine secretion, pneumonia, and acute lung injury. Consequently, it leads to acute respiratory distress syndrome, disseminated intravascular coagulation, multiple organ failure, and death. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative virus of COVID-19, influences nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3 (NLRP3), the sensor of inflammasomes, directly or indirectly, culminating in the assembly of NLRP3 inflammasome and activation of inflammatory caspases, which induce the inflammatory disruption in severe COVID-19. Accordingly, the target therapeutics for inflammasome has attracted attention as a treatment for COVID-19. Korean Red Ginseng (KRG) inhibits several inflammatory responses, including the NLRP3 inflammasome signaling. This review discusses the role of KRG in the treatment and prevention of COVID-19 based on its anti-NLRP3 inflammasome efficacy.

Repurposing Auranofin, an Anti-Rheumatic Gold Compound, to Treat Acne Vulgaris by Targeting the NLRP3 Inflammasome

  • Yang, Gabsik;Lee, Seon Joo;Kang, Han Chang;Cho, Yong-Yeon;Lee, Hye Suk;Zouboulis, Christos C.;Han, Sin-Hee;Ma, Kyung-Ho;Jang, Jae-Ki;Lee, Joo Young
    • Biomolecules & Therapeutics
    • /
    • 제28권5호
    • /
    • pp.437-442
    • /
    • 2020
  • Activation of the NLRP3 inflammasome is critical for host defense as well as the progression of inflammatory diseases through the production of the proinflammatory cytokine IL-1β, which is cleaved by active caspase-1. It has been reported that overactivation of the NLRP3 inflammasome contributes to the development and pathology of acne vulgaris. Therefore, inhibiting activation of the NLRP3 inflammasome may provide a new therapeutic strategy for acne vulgaris. In this study, we investigated whether auranofin, an anti-rheumatoid arthritis agent, inhibited NLRP3 inflammasome activation, thereby effectively treating acne vulgaris. Auranofin suppressed NLRP3 inflammasome activation induced by Propionibacterium acnes, reducing the production of IL-1β in primary mouse macrophages and human sebocytes. In a P. acnes-induced acne mouse model, injection of P. acnes into the ears of mice induced acne symptoms such as redness, swelling, and neutrophil infiltration. Topical application of auranofin (0.5 or 1%) to mouse ears significantly reduced the inflammatory symptoms of acne vulgaris induced by P. acnes injection. Topical application of auranofin led to the downregulation of the NLRP3 inflammasome activated by P. acnes in mouse ear skin. These results show that auranofin inhibits the NLRP3 inflammasome, the activation of which is associated with acne symptoms. The results further suggest that topical application of auranofin could be a new therapeutic strategy for treating acne vulgaris by targeting the NLRP3 inflammasome.

Panax ginseng as a potential therapeutic for neurological disorders associated with COVID-19; Toward targeting inflammasome

  • Seo Won Shin;Ik Hyun Cho
    • Journal of Ginseng Research
    • /
    • 제47권1호
    • /
    • pp.23-32
    • /
    • 2023
  • Coronavirus disease 2019 (COVID-19) is a highly infectious respiratory disease caused by a severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). SARS-CoV-2 infection may cause clinical manifestations of multiple organ damage, including various neurological syndromes. There are currently two oral antiviral drugs-Paxlovid and molnupiravir-that are recognized to treat COVID-19, but there are still no drugs that can specifically fight the challenges of SARS-CoV-2 variants. Nucleotide-binding oligomerization domain-like receptor pyrin domain-containing-3 (NLRP3) inflammasome is a multimolecular complex that can sense heterogeneous pathogen-associated molecular patterns associated with neurological disorders. The NLRP3 activation stimulates the production of caspase-1-mediated interleukin (IL)-1β, IL-18, and other cytokines in immune cells. Panax (P.) ginseng is a medicinal plant that has traditionally been widely used to boost immunity and treat various pathological conditions in the nervous system due to its safety and anti-inflammatory/oxidant/viral activities. Several recent reports have indicated that P. ginseng and its active ingredients may regulate NLRP3 inflammasome activation in the nervous system. Therefore, this review article discusses the current knowledge regarding the pathogenesis of neurological disorders related to COVID-19 and NLRP3 inflammasome activation and the possibility of using P. ginseng in a strategy targeting this pathway to treat neurological disorders.