scholarly journals Lactic Acid Fermentation Is Required for NLRP3 Inflammasome Activation

2021 ◽  
Vol 12 ◽  
Author(s):  
Hsin-Chung Lin ◽  
Yu-Jen Chen ◽  
Yau-Huei Wei ◽  
Hsin-An Lin ◽  
Chien-Chou Chen ◽  
...  

Activation of the Nod-like receptor 3 (NLRP3) inflammasome is important for activation of innate immune responses, but improper and excessive activation can cause inflammatory disease. We previously showed that glycolysis, a metabolic pathway that converts glucose into pyruvate, is essential for NLRP3 inflammasome activation in macrophages. Here, we investigated the role of metabolic pathways downstream glycolysis – lactic acid fermentation and pyruvate oxidation—in activation of the NLRP3 inflammasome. Using pharmacological or genetic approaches, we show that decreasing lactic acid fermentation by inhibiting lactate dehydrogenase reduced caspase-1 activation and IL-1β maturation in response to various NLRP3 inflammasome agonists such as nigericin, ATP, monosodium urate (MSU) crystals, or alum, indicating that lactic acid fermentation is required for NLRP3 inflammasome activation. Inhibition of lactate dehydrogenase with GSK2837808A reduced lactate production and activity of the NLRP3 inflammasome regulator, phosphorylated protein kinase R (PKR), but did not reduce the common trigger of NLRP3 inflammasome, potassium efflux, or reactive oxygen species (ROS) production. By contrast, decreasing the activity of pyruvate oxidation by depletion of either mitochondrial pyruvate carrier 2 (MPC2) or pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1) enhanced NLRP3 inflammasome activation, suggesting that inhibition of mitochondrial pyruvate transport enhanced lactic acid fermentation. Moreover, treatment with GSK2837808A reduced MSU-mediated peritonitis in mice, a disease model used for studying the consequences of NLRP3 inflammasome activation. Our results suggest that lactic acid fermentation is important for NLRP3 inflammasome activation, while pyruvate oxidation is not. Thus, reprograming pyruvate metabolism in mitochondria and in the cytoplasm should be considered as a novel strategy for the treatment of NLRP3 inflammasome-associated diseases.

2009 ◽  
Vol 75 (15) ◽  
pp. 5175-5178 ◽  
Author(s):  
Kenji Okano ◽  
Shogo Yoshida ◽  
Tsutomu Tanaka ◽  
Chiaki Ogino ◽  
Hideki Fukuda ◽  
...  

ABSTRACT Optically pure d-lactic acid fermentation from arabinose was achieved by using the Lactobacillus plantarum NCIMB 8826 strain whose l-lactate dehydrogenase gene was deficient and whose phosphoketolase gene was substituted with a heterologous transketolase gene. After 27 h of fermentation, 38.6 g/liter of d-lactic acid was produced from 50 g/liter of arabinose.


2020 ◽  
Vol 11 ◽  
Author(s):  
Ziwei Xu ◽  
Zi-mo Chen ◽  
Xiaoyan Wu ◽  
Linjie Zhang ◽  
Ying Cao ◽  
...  

The NLRP3 inflammasome is a core component of innate immunity, and dysregulation of NLRP3 inflammasome involves developing autoimmune, metabolic, and neurodegenerative diseases. Potassium efflux has been reported to be essential for NLRP3 inflammasome activation by structurally diverse pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs). Thus, the molecular mechanisms underlying potassium efflux to activate NLRP3 inflammasome are under extensive investigation. Here, we review current knowledge about the distinction channels or pore-forming proteins underlying potassium efflux for NLRP3 inflammasome activation with canonical/non-canonical signaling or following caspase-8 induced pyroptosis. Ion channels and pore-forming proteins, including P2X7 receptor, Gasdermin D, pannexin-1, and K2P channels involved present viable therapeutic targets for NLRP3 inflammasome related diseases.


2008 ◽  
Vol 75 (2) ◽  
pp. 462-467 ◽  
Author(s):  
Kenji Okano ◽  
Qiao Zhang ◽  
Satoru Shinkawa ◽  
Shogo Yoshida ◽  
Tsutomu Tanaka ◽  
...  

ABSTRACT In order to achieve direct and efficient fermentation of optically pure d-lactic acid from raw corn starch, we constructed l-lactate dehydrogenase gene (ldhL1)-deficient Lactobacillus plantarum and introduced a plasmid encoding Streptococcus bovis 148 α-amylase (AmyA). The resulting strain produced only d-lactic acid from glucose and successfully expressed amyA. With the aid of secreting AmyA, direct d-lactic acid fermentation from raw corn starch was accomplished. After 48 h of fermentation, 73.2 g/liter of lactic acid was produced with a high yield (0.85 g per g of consumed sugar) and an optical purity of 99.6%. Moreover, a strain replacing the ldhL1 gene with an amyA-secreting expression cassette was constructed. Using this strain, direct d-lactic acid fermentation from raw corn starch was accomplished in the absence of selective pressure by antibiotics. This is the first report of direct d-lactic acid fermentation from raw starch.


2021 ◽  
Vol 17 (7) ◽  
pp. e1009712
Author(s):  
Shivangi Rastogi ◽  
Sarah Ellinwood ◽  
Jacques Augenstreich ◽  
Katrin D. Mayer-Barber ◽  
Volker Briken

Mycobacterium tuberculosis (Mtb) has evolved to evade host innate immunity by interfering with macrophage functions. Interleukin-1β (IL-1β) is secreted by macrophages after the activation of the inflammasome complex and is crucial for host defense against Mtb infections. We have previously shown that Mtb is able to inhibit activation of the AIM2 inflammasome and subsequent pyroptosis. Here we show that Mtb is also able to inhibit host cell NLRP3 inflammasome activation and pyroptosis. We identified the serine/threonine kinase PknF as one protein of Mtb involved in the NLRP3 inflammasome inhibition, since the pknF deletion mutant of Mtb induces increased production of IL-1β in bone marrow-derived macrophages (BMDMs). The increased production of IL-1β was dependent on NLRP3, the adaptor protein ASC and the protease caspase-1, as revealed by studies performed in gene-deficient BMDMs. Additionally, infection of BMDMs with the pknF deletion mutant resulted in increased pyroptosis, while the IL-6 production remained unchanged compared to Mtb-infected cells, suggesting that the mutant did not affect the priming step of inflammasome activation. In contrast, the activation step was affected since potassium efflux, chloride efflux and the generation of reactive oxygen species played a significant role in inflammasome activation and subsequent pyroptosis mediated by the Mtb pknF mutant strain. In conclusion, we reveal here that the serine/threonine kinase PknF of Mtb plays an important role in innate immune evasion through inhibition of the NLRP3 inflammasome.


2008 ◽  
Vol 35 (2) ◽  
pp. 131 ◽  
Author(s):  
Rudy Dolferus ◽  
Mark Wolansky ◽  
Rebecka Carroll ◽  
Yo Miyashita ◽  
Kathleen Ismond ◽  
...  

During waterlogging conditions plants switch from aerobic respiration to anaerobic fermentation to cope with the lack of available oxygen. Plants have two main fermentation pathways: ethanol and lactic acid fermentation. In this paper we carry out a functional analysis of the Arabidopsis lactate dehydrogenase gene, LDH1. Our results indicate that LDH1, like some other anaerobic genes, is expressed in a root-specific manner and is affected by a variety of abiotic stresses (hypoxia, drought, cold) and mechanical wounding. Functional analysis of LDH1 was carried out using transgenic Arabidopsis overexpressing the gene (35S promoter) and a T-DNA knockout line. Overexpression of LDH1 resulted in improved survival of low oxygen stress conditions in roots but not in shoots. Increased lactic acid fermentation also resulted in significantly higher activities of pyruvate decarboxylase (PDC). Knockout mutants of LDH1 showed reduced survival under low oxygen conditions and PDC activity levels were not changed compared with the wild type. Our data suggest that there is an interdependency between the lactic and ethanol fermentation pathways and that lactic acid fermentation may play a role in stimulating ethanol fermentation and improving plant survival. We show also that Arabidopsis plants are able to exude lactate efficiently into the medium, preventing it accumulating to toxic levels in the cells.


2021 ◽  
Author(s):  
Romeo Ricci ◽  
Zhirong Zhang ◽  
Li Ran ◽  
Rossella Venditti ◽  
Zengzhen Liu ◽  
...  

Abstract Inflammasome complexes are pivotal in the innate immune response to pathogens and other danger signals. The NLRP3 inflammasome is activated in response to a broad variety of cellular stressors. Most of the stimuli act in a potassium efflux-dependent manner but a primary and converging sensing mechanism by the NLRP3 receptor initiating inflammasome assembly remains ill-defined. Here we show that NLRP3 activators disrupt endosome-TGN retrograde transport (ETRT) and lead to localization of NLRP3 to endosomal vesicles. Genetic and pharmacologic perturbation of ETRT leads to accumulation of phosphoinositol-4-phosphate (PI4P) in endosomes to which NLRP3 is recruited. Disruption of ETRT potentiates NLRP3 inflammasome activation in murine and human macrophages in vitro. Mice with defects in ETRT in the myeloid compartment are more susceptible to LPS-induced sepsis showing enhanced mortality and IL-1β serum levels as compared to control animals. Our study thus uncovers that changes in endocytic trafficking mediate NLRP3-dependent inflammatory responses.


2021 ◽  
Author(s):  
Zhirong ZHANG ◽  
Li RAN ◽  
Rossella Venditti ◽  
Zengzhen Liu ◽  
Annette Schurmann ◽  
...  

Inflammasome complexes are pivotal in the innate immune response to pathogens and other danger signals. The NLRP3 inflammasome is activated in response to a broad variety of cellular stressors. Most of the stimuli act in a potassium efflux-dependent manner but a primary and converging sensing mechanism by the NLRP3 receptor initiating inflammasome assembly remains ill-defined. Here we show that NLRP3 activators disrupt endosome-TGN retrograde transport (ETRT) and lead to localization of NLRP3 to endosomal vesicles. Genetic and pharmacologic perturbation of ETRT leads to accumulation of phosphoinositol-4-phosphate (PI4P) in endosomes to which NLRP3 is recruited. Disruption of ETRT potentiates NLRP3 inflammasome activation in murine and human macrophages in vitro. Mice with defects in ETRT in the myeloid compartment are more susceptible to LPS-induced sepsis showing enhanced mortality and IL-1β serum levels as compared to control animals. Our study thus uncovers that changes in endocytic trafficking mediate NLRP3-dependent inflammatory responses.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2723-2723 ◽  
Author(s):  
Kyohei Nakamura ◽  
Tohru Fujiwara ◽  
Tomonori Ishii ◽  
Hideo Harigae ◽  
Kouetsu Ogasawara

Abstract Introduction. Monocytes/macrophages play an essential role in systemic iron homeostasis by their recycling and storage capacity of iron. Under chronic inflammatory condition, pro-inflammatory cytokines such as TNF-α and IL-1 stimulate production of hepcidin by liver cells, which in turn down-regulates iron exporter ferroportin expression on monocytes/macrophages. Thus, hepcidin/ferroportin axis contributes to the pathogenesis of anemia in chronic inflammation by restricting cellular iron export into plasma. Pro-inflammatory cytokine IL-1β is mainly produced by monocytes/macrophages througha molecular platform, called inflammasome. One of the best characterized members is NLRP3 inflammasome, which plays a central role in sterile inflammation in response to wide range of danger stimuli, including uric crystal, cholesterol crystal, silica and asbestos. Given that excess cellular iron increases the level of redox-active labile iron pool which is responsible for iron-mediated cytotoxicity, cellular labile iron might be sensed as a danger signal in monocytes/macrophages. In this study we addressed whether cellular labile iron activates NLRP3 inflammasome. Methods. Peripheral blood mononuclear cells (PBMCs) were isolated by density-gradient centrifugation from healthy donors. After 4 h priming with 10 ng/ml LPS, PBMCs were stimulated with various dose of ferric ammonium citrate (FAC) for 4 h. IL-1β production in culture supernatants was determined by ELISA. Cleaved caspase-1 and mature IL-1β was analyzed by western blot. Calcein-AM assay was used to determine cellular labile iron pool.To investigate the mechanism of NLRP3 inflammasome activation, total reactive oxygen species (ROS) detection assay and JC-10 mitochondrial membrane potential assay were performed. To evaluate lysosomal membrane permeabilization, PMA-differentiated THP-1 cells stained with Lyso Tracker were analyzed by fluorescence microscope. Results. FAC induced the concentration-dependent increase of the labile iron pool and secretion of IL-1β in LPS-primed human monocytes. Ferrous iron chelator, bipyridine significantly inhibited the IL-1β production, highlighting importance of cellular chelatable iron pool for IL-1β production. IL-1β production induced by FAC was abrogated by pan-caspase inhibitor as well as by caspase-1 specific inhibitor, suggesting that inflammasome-mediated caspase-1 activation is required for this process. Consistently, cleaved caspase-1 and mature IL-1β was confirmed by western blot analysis. We next addressed whether NLRP3 inflammasome is activated in response to cellular labile iron. NLRP3 inhibitor, glyburide significantly inhibited IL-1β production. Furthermore, FAC treatment induced IL-1β production in THP-1 cells, but not in NLRP3-deficient THP-1 cells, indicating that cellular iron activates NLRP3 inflammasome. Next, we addressed how cellular iron activates NLRP3 inflammasome. Potassium efflux is thought to be a common trigger of NLRP3 inflammasome activation. Consistently, IL-1β production was completely abrogated in high-potassium media, indicating that potassium efflux is required for iron-mediated NLRP3 inflammasome activation. Since cellular labile iron is involved in ROS generation through Fenton reactions, we next investigated the role of ROS in iron-mediated NLRP3 inflammasome activation. FAC treatment increased ROS levels of monocytes in concentration-dependent manner. Accordingly, the percentage of monocytes with decreased mitochondorial membrane potential was increased. N-acetyl cysteine inhibited FAC-induced IL-1β production, suggesting that ROS-mediated mitochondorial damage is involved in iron-mediated NLRP3 inflammasome activation. In addition, ROS-induced lysosomal membrane permeabilization was observed in FAC-treated THP-1 cells. A cathepsin B inhibitor, CA-074 methyl ester, inhibited IL-1β production, suggesting that ROS-mediated lysosomal damage is also involved in iron-mediated NLRP3 inflammasome activation. Conclusion. In this study, we found that cellular labile iron activates NLRP3 inflammasome through its redox activity. Because IL-1β has been reported to induce hepcidin synthesis, excess cellular iron in monocytes/macrophages might be implicated in a positive feedback loop for inflammation through NLRP3 inflammasome and hepcidin/ferroportin axis. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document