• OPEN ACCESS

Progress in the Correlation Between Inflammasome NLRP3 and Liver Fibrosis

  • Meihua Sun1 ,
  • Yanqing Zhang1 ,
  • Anbing Guo2,
  • Zongting Xia1 and
  • Lijun Peng2,* 
 Author information
Journal of Clinical and Translational Hepatology 2024;12(2):191-200

DOI: 10.14218/JCTH.2023.00231

Abstract

Liver fibrosis is a reversible condition that occurs in the early stages of chronic liver disease. To develop effective treatments for liver fibrosis, understanding the underlying mechanism is crucial. The NOD-like receptor protein 3 (NLRP3) inflammasome, which is a part of the innate immune system, plays a crucial role in the progression of various inflammatory diseases. NLRP3 activation is also important in the development of various liver diseases, including viral hepatitis, alcoholic or nonalcoholic liver disease, and autoimmune liver disease. This review discusses the role of NLRP3 and its associated molecules in the development of liver fibrosis. It also highlights the signal pathways involved in NLRP3 activation, their downstream effects on liver disease progression, and potential therapeutic targets in liver fibrosis. Further research is encouraged to develop effective treatments for liver fibrosis.

Keywords

NLRP3, Liver disease, Liver fibrosis, Hepatic stellate cells, Macrophages

Introduction

Liver disease is a serious public health problem, accounting for about 2 million deaths worldwide each year.1 Chronic liver injuries are caused by a range of stimuli including viral hepatitis, alcoholic and nonalcoholic liver disease, and autoimmune liver disease. These conditions lead to liver inflammation and fibrosis, ultimately progressing to cirrhosis. In China, liver cirrhosis accounts for 11% of all the deaths from liver diseases worldwide.2 Constant or repeated inflammation and necrosis of liver cells lead to an enhanced repair response, triggering massive production of fibrous substances such as collagen, proteoglycans, etc. Insufficient degradation of fibrous substances results in the formation of liver fibrosis. If timely interventions are taken, the possibility of liver fibrosis evolving into cirrhosis, liver failure, and liver cancer can be reduced.

Inflammasomes comprise a variety of protein complexes assembled with the involvement of cytoplasmic pattern recognition receptors, and are a key component of the innate immune system.3 Inflammasome components are found in various cells, including immune and nonimmune cells, such as macrophages, neutrophils, monocytes, hepatic stellate cells (HSCs), and fibroblasts/myofibroblasts. Those components are expressed in multiple intracellular locations including mitochondria, Golgi apparatus, and nucleus.4,5 Inflammasomes recognize damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), and subsequently activate caspase-1. This triggers the release of interleukin (IL)-18 and IL-1β, which contributes to the progression of fibrosis. To date, several types of inflammasomes have been revealed, including NOD-like receptor protein 1 (NLRP1), NLRP3, and NOD-like receptor C4. Among them, NLRP3 has been studied extensively and is known to play a crucial role in antibacterial immunological responses.6,7 Abnormal activation of NLRP3 has been linked to various diseases, including Alzheimer’s disease, arthritis, atherosclerosis, and cancer.8 Importantly, several studies have shown that NLRP3 participates in the development of liver fibrosis.9–11 This review discusses current research on the role of NLRP3 in liver fibrosis.

Activation of NLRP3

NLRP3 is a typical NLR protein that contains the innate immune receptor NLRP3, caspase-1, and apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC). In the activation of NLRP3 (Fig. 1), the first step is the initiation of NLRP3, involving the upregulation of NLRP3, IL-18, and IL-1β. PAMPs bind to Toll-like receptors (TLRs) and activate the transcription factor nuclear factor-kappa B (NF-κB), which subsequently mediates the transcription of NLRP3, IL-1β precursor (pro-IL-1β) and IL-18 precursor (pro-IL-18). Meanwhile, damaged cell DAMP signals such as uric acid crystals, cholesterol crystals, reactive oxygen species (ROS), and oxidized mitochondrial (mt)DNA activate and oligomerize NLRP3.12,13

Classical activation of NLRP3.
Fig. 1  Classical activation of NLRP3.

PAMPs bind to TLR and activate NF-κB, participating in NLRP3 activation. DAMPs, K+ efflux, and the contents released from damaged lysosomes activate and oligomerize NLRP3. The activated NLRP3 releases caspase-1, which promotes the maturation of pro-IL-1β and pro-IL-18. ATP, adenosine triphosphate; DAMP, damage-associated molecular pattern; IL-1β, interleukin-1β; IL-18, interleukin-18; NF-κB, transcription factor nuclear factor-kappa B; NLRP3, NOD-like receptor protein 3; ox-mtDNA, oxidized mitochondrial DNA; PAMP, pathogen-associated molecular pattern; pro-IL-1β, IL-1β precursor; pro-IL-18, IL-18 precursor; PRR, pattern recognition receptor; P2X7R, P2X purine receptor 7 channel; ROS, reactive oxygen species; TWIK2, two-pore domain weakly inward rectifying potassium channel 2; TLR, Toll-like receptor.

After NLRP3 inflammasomes are triggered, the components are recruited and assembled, promoting the cleavage of procaspase-1 into active caspase-1. This process facilitates the maturation of IL-18 and IL-1β. Additionally, activated caspase-1 cleaves gasdermin D and release its N-terminal domain, which induces pyroptosis and the subsequent release of cellular contents.8 Caspase-11 activates NLRP3 inflammasomes through pyroptosis.14 In addition, NLRP3 can be activated by a noncanonical activation pathway that involves the activation of caspase-4 and caspase-5 in humans or caspase-11 in mice. The interaction between caspase-4/5/11 and lipopolysaccharide (LPS) along with lipid A results in their transformation into an active form. Activated caspase-4/5/11 further triggers the activation of NLRP3 by K+ efflux and pyroptosis.14–16 Recent research suggests that the orphan receptor Nur77 combines with mtDNA and LPS to mediate nontypical activation of NLRP3. However, Nur77 association with intracellular LPS does not depend on caspase-11 or gasdermin D.16

Activation mechanism of NLRP3

K+ outflow

K+ efflux is one of the upstream signals that activates NLRP3. For example, extracellular ATP triggers K+ efflux through the ATP-gated P2X purinoceptor 7 channel and the two-pore domain weakly inward rectifying potassium channel 2, which then triggers the activation of NLRP3.17,18 Moreover, particles like calcium pyrophosphate crystals, cholesterol crystals, and silica can also induce potassium efflux, activating NLRP3.19 A study has shown that NLRP3 is activated when the K+ content of cells drops below 80%.20 Moreover, caspase-11 triggers the noncanonical inflammasome pathway and involves the activation of the pannexin-1 channel and leads to K+ efflux and NLRP3 activation.21

Lysosome rupture

Under some pathological conditions, such as the phagocytosis of particulate matter, lysosome damage can activate the NLRP3 inflammasome. Phagocytosed crystals lead to lysosome acidification, swelling, and loss of lysosomal membrane integrity over time. Upon damage, lysosomal contents leak into the cytoplasm and trigger NLRP3.19,22 Release of lysosome contents into the cytoplasm is also related to the activation of caspase-1.23

ROS and mitochondria

Cells under harmful stimuli produce ROS and reactive nitrogen species that cause physiological and pathological responses in cells and tissues. Excess ROS can result in oxidative stress. Oxidative stress can increase liver inflammation and activate HSC, thereby enhancing the production of extracellular matrix (ECM), ultimately leading to fibrosis.24 Damaged hepatocytes caused by various factors such as alcohol abuse, hepatitis virus infection, and chronic cholestasis may generate ROS and participate in the assembly and activation of NLRP3. ROS is one of many important NLRP3 inflammasome activators.25 Conversely, ROS inhibitors (e.g., diphenyl iodine, and n-acetyl-l-cysteine) can suppress NLRP3 transcription.5 In the early stage of an inflammatory response, ROS activate the NF-κB pathway. ROS causes conformational change and activation of NLRP3 by promoting the transcription of NF-κB.26–28 A study reported that the activation of NLRP3 occurred via the ROS-TXNIP axis.29 Furthermore, both O2− and H2O2 in some cells have been shown to participate in NLRP3 activation.29,30

Mitochondria are the main source of ROS. Therefore, mitochondrial dysfunction can trigger inflammatory responses through the inflammasome signaling pathway. The production of mtROS during mitochondrial injury is a known activator of NLRP3.31 A study showed that excessive free fatty acids in the livers of high-fat/calorie diet mice led to mitochondrial damage, leading to ROS generation and NLRP3 activation.32 Under long-term ethanol stimulation, mouse macrophages, or human peripheral blood mononuclear cells were shown to induce the release of mtROS, activating NLRP3.33 The 66 kDa isoform of Shc, a redox enzyme can mediate the generation of mitochondrial ROS and activate NLRP3 inflammasome, hence promoting HSC activation. ROS can also induce the oxidation of mtDNA.34 Oxidized mtDNA is capable of binding and directly activating NLRP3, which triggers caspase-1 activation, and promotes the release of IL-18 and IL-1β. In addition, mtDNA amplifies the activation of NLRP3.35 Notably, most NLRP3 agonists lead to mitochondrial malfunction, ROS generation, and mtDNA oxidation, all of which encourage NLRP3 activation.36 Similarly, the activation of NLRP3 also leads to mitochondrial damage and mtROS production. Recent evidence suggests that mitochondrial homeostasis largely depends on the removal of damaged mitochondria.37 Inhibition of mitochondrial autophagy can increase the accumulation of ROS, thus activating NLRP3 inflammasomes.38

Activation of NLRP3 involves complex and diverse mechanisms, such as K+ efflux, lysosome rupture, oxidative stress, etc. K+ efflux functions with many NLRP3 activators but is not necessary for NLRP3 inflammasome activation. For instance, CL097 and imiquimod directly target mitochondria without involving K+ efflux to induce NLRP3 inflammasome activation.39 An ethanolic extract of Artemisia anomala has a lysosome protective function by inhibiting the TAK1-JNK pathway, thus preventing activation of NLRP3.40 However, it neither inhibits mitochondrial damage nor affects the efflux of K+ and chloride ions.40 In addition, multiple cell signaling events sometimes overlap and function with each other. For instance, lysosome damage and K+ efflux together participate in NLRP3 inflammasome activation driven by polybrominated diphenyl ethers.41 Similarly, K+ efflux-induced mtDNA release activates NLRP3 inflammasomes.42 Apilimod relies on lysosomal mediated mitochondrial damage and ROS production to activate NLRP3.43 Overall, the activation signals can act independently or in together. Such complexities make the activation mechanism of NLRP3 inflammasome more multifaceted and diversified. Therefore, a precise NLRP3 activation mechanism under specific conditions remains unknown.

Fibrosis of liver

Under various chronic stimuli, chronic inflammation, and necrosis of hepatocytes trigger an enhanced repair response, resulting in massive proliferation and insufficient degradation of fibers. This causes a massive deposition of fibrous materials in the liver tissue, i.e. liver fibrosis. Numerous cellular pathways participate in fibrosis, and HSCs play a significant role. Many stimuli act on HSCs to promote their activation, resulting in a significant buildup of ECM progressing to fibrous scar tissue. Systemic inflammation driven by immune cells is another key factor in the progression of cirrhosis. Macrophages ensure immune balance in the liver and also participate in inflammation. Inflammatory responses in the liver mediate hepatocyte damage, cause cell differentiation and proliferation, perpetuate chronic liver inflammation, promote fibrous tissue growth, and worsen liver fibrosis. Both Kupffer cells (KCs) and HSCs have high levels of NLRP3 inflammasome activation, which is critical in the development of liver fibrosis.44

HSCs and NLRP3

In healthy livers, about 15% of resident cells are HSCs, which is about one-third of the population of nonparenchymal cells. After activation, HSCs can transform into myofibroblasts, which secrete ECM, generate fibrous scars, and participate in the process of liver fibrosis. HSC is the main source of myofibroblasts, but other sources include resident liver cells, portal vein fibroblasts, and bone marrow-derived cells.45,46 Under normal conditions, HSCs are quiescent. When the liver is damaged, HSCs are activated by inflammatory mediators or other stimulatory factors. Activated HSCs proliferate and move toward the injured liver tissue. Apart from producing alpha smooth muscle actin (α-SMA), activated HSCs produce tissue inhibitors of metalloproteinases (TIMPs) that inhibit the activity of matrix metalloproteinases. This reduces ECM degradation, causing excessive deposition of ECM and thereby the formation of fibrotic scars.45,47

A wide range of factors are involved in HSC activation, such as platelet-derived growth factor, transforming growth factor beta (TGF-β), IL6, IL8, and inflammasomes (NLRP1, NLRP3, etc.). NLRP3 is closely related to hepatic fibrosis and acts on HSC to promote liver fibrosis. NLRP3 along with the main proinflammatory factor NF-κB promotes profibrosis molecules (IL-1β and IL-18) to activate HSCs.26 However, NLRP3 can be directly expressed and triggered in HSCs, causing hepatic fibrosis (Fig. 2).4 All components of NLRP3 exist in HSCs and regulate their various functions, including the transition of quiescent HSCs to a collagen-producing myoblast state.48–50 The NLRP3 inflammasome is a downstream effect factor of DAMPs, and it has been reported that DAMPs released from dead hepatocytes may directly or indirectly promote HSC activation and fibrosis (Fig. 2).51,52 Notably, NLRP3 mutant mice had significantly higher expression of connective tissue growth factor and TIMP 1 than wild-type mice. That implies that NLRP3 inflammation can induce HSC activation and collagen deposition.51

Hepatic macrophages, HSCs, and NLRP3.
Fig. 2  Hepatic macrophages, HSCs, and NLRP3.

Chronic stimuli such as alcohol, viruses, cholestasis, and lipid accumulation damage hepatocytes. PAMPs and DAMPs activate NLRP3 in macrophages. This activation triggers the release of proinflammatory factors (IL-1β, IL-18, IL-6, TNF-α) and TGF-β. Proinflammatory factors and TGF-β then promote the proliferation and differentiation of HSCs into myofibroblasts. Furthermore, HSCs can also directly express and activate NLRP3. α-SMA, alpha smooth muscle actin; col-1, type I collagen; DAMP, damage-associated molecular pattern; ECM, extracellular matrix; HSC, human hepatic stellate cell; IL-1β, interleukin 1β; IL-18, interleukin 18; IL-6, interleukin 6; NLRP3, NOD-like receptor protein 3; NF-κB, transcription factor nuclear factor-kappa B; PAMP, pathogen-associated molecular pattern; PRR, pattern recognition receptor; ROS, reactive oxygen species; TGF-β, transforming growth factor beta; TLR, Toll-like receptor; TNF-α, tumor necrosis factor alpha.

Macrophages and NLRP3

Hepatic macrophages mainly include resident macrophages (KCs) and monocyte-derived macrophages, all of which ensure immunological homeostasis in the liver. In the steady state, resident macrophages derived from the yolk sac predominate. Under injury stimulation, monocyte-derived macrophages are recruited, which differentiate from circulating monocytes in the liver.53 Macrophages can be grouped into M1 macrophages and M2 macrophages. M1 macrophages produce inflammatory cytokines with a proinflammatory role. M2 macrophages have healing and anti-inflammatory functions that regulate inflammation. The balance of M1 and M2 macrophages may mediate the advancement and regression of liver fibrosis.47,54 Upon liver damage, a large number of bone marrow-derived monocytes aggregate in the liver and differentiate into macrophages to produce proinflammatory and profibrotic cytokines that promote inflammatory responses and HSC activation. Activated HSCs express α-SMA and collagen I, which promotes ECM deposition and progression of liver fibrosis.47,52 Studies have shown that when HSCs are cocultured with KCs, KCs promote the proliferation and activation of HSCs. Furthermore, HSCs cocultured with KCs secrete more intracellular and extracellular collagen I, as well as TIMP 1.55

NLRP3 is mainly expressed in macrophages.56 In contrast to HSCs, KCs were shown to express higher levels of NLRP3, NLRP1, and Absent In Melanoma 2 in a mouse model of hepatic fibrosis.13 After binding to the membrane receptors on KCs, PAMP activated NLRP3 in KCs through the NF-κB signaling pathway (Fig. 2), causing the production of its related components (NLRP3, caspase-1, and IL-1β).4 In addition, the macrophage X-box binding protein-1 (XBP1) gene can induce M1 macrophage polarization and activate macrophage NLRP3.57 Activation of macrophage NLRP3 has a significant impact on liver fibrosis. A study suggested that the activation of macrophage NLRP3 can promote disease progression in cholestasis causing liver damage.58 Zhang et al.44 reported that NLRP3 inflammasomes play a vital role in S. japonicum-induced liver fibrosis through the NF-κB signaling pathway. They also revealed that NLRP3 inflammasomes in both KCs and HSCs contributed to the development of liver fibrosis in S. japonicum-infected mice, and NLRP3 activation was mainly caused by KCs. In addition, s100a8-mediated NLRP3-dependent macrophage pyroptosis was shown to promote the activation of human HSCs.59 NLRP3 in mouse macrophages participated in ECM deposition by activating HSCs (Fig. 2).60

Gut microflora and NLRP3

Due to the existence of the gut-liver axis, risk factors originating from the intestine have become one of the contributing factors in the development of liver diseases. The gut microbiota is a group of microorganisms that are present in the human intestine and affect health. In addition to participating in digestion and absorption, it also has a role in immune regulation. NLRP3 is widely distributed in epithelial cells and immune cells. In the intestine, PAMPs bind to pattern recognition receptors to activate NLRP3, triggering an inflammatory response to maintain intestinal immune homeostasis.61 External stimuli such as infection, trauma, drugs, poor diet, etc., can disrupt the gut microbiota, increasing the proportion of harmful bacteria. Metabolites and toxins secreted by harmful bacteria can cause intestinal inflammation and intestinal barrier damage. Damage of the intestinal barrier allows intestinal LPS entry into the liver through the portal vein, where LPS binds to TLR and activates NLRP3, causing liver inflammation (Fig. 3).62 This contributes to the progression of various chronic liver diseases such as nonalcoholic liver disease (NAFLD), alcohol-related liver disease (ALD), viral hepatitis, and autoimmune liver diseases (Table 1).63–66 Recently, it was shown that ursolic acid inhibited the NOX4/NLRP3 inflammasome signaling pathway, reduced the abundance of harmful gut bacteria, and increased the abundance of beneficial gut bacteria, all of which helped to reverse liver fibrosis.67Tylophora yunnanensis Schltr can regulate the gut microbiota by inhibiting the activation of NLRP3 to improve nonalcoholic steatohepatitis (NASH).68 Astragaloside IV can regulate gut microbiota imbalance, improve intestinal barrier function, inhibit the NLRP3/Caspase-1 inflammatory signaling pathway, and alleviate alcohol-induced liver inflammation.69 Additionally, probiotics can enhance the intestinal mucus barrier by increasing the secretion of specific mucins. Probiotic intervention can help rebalance the gut microbiota and regulate intestinal barrier, thereby alleviating the liver damage.64,70,71

Activation of NLRP3 inflammasome in chronic liver disease.
Fig. 3  Activation of NLRP3 inflammasome in chronic liver disease.

High-fat diet and long-term alcohol consumption damage hepatocytes, leading to the accumulation of lipid in hepatocytes and causing an inflammatory response. Lipid-accumulated liver cells are prone to lipid peroxidation and oxidative damage, activating NLRP3. Viral proteins activate NLRP3, resulting in inflammatory reactions and activating HSCs. Abnormal expression of autoantigens may lead to an abnormal immune response in the liver, causing the activation of NLRP3. Immune-mediated bile duct injury results in intrahepatic bile duct narrowing and bile stasis. Bile stasis, in turn, activates NLRP3 through the NF-κB pathway. High-fat diet, alcohol, virus infection, and cholestasis lead to dysbiosis of the gut microbiota and increase intestinal permeability. Gut-derived PAMPs enter the liver and then activate NLRP3. ECM, extracellular matrix; HSCs, human hepatic stellate cells; HBV, hepatitis B virus; HCV, hepatitis C virus; IL-1β, interleukin 1β; IL-18, interleukin 18; LPS, lipopolysaccharide; NLRP3, NOD-like receptor protein 3; NF-κB, transcription factor nuclear factor-kappa B; ROS, reactive oxygen species.

Table 1

Summary and comparison of NLRP3 activation in chronic liver disease

Liver diseaseEtiologyActivation of NLRP3
NAFLDLipid toxicity; chronic inflammation; oxidative stress; insulin resistanceLipid toxicity, mitochondrial dysfunction, and excessive ROS are involved in the activation of NLRP3; Activation of NLRP3 by regulating the NF-κB pathwayHigh-fat diet, alcohol, virus infection, and cholestasis lead to dysbiosis of the gut microbiota, increasing intestinal permeability. Gut-derived PAMPs enter the liver and then activate NLRP3.
ALDLong-term ethanol toxicity (damage to intestinal mucosa, direct damage to liver)Ethanol exposure increases recruitment of inflammatory cells and activates NLRP3 by regulating the NF-κB pathway; Alcohol metabolism exacerbates oxidative stress and induces the production of ROS to activate NLRP3
Viral hepatitisVirus infection; immune response caused by virus-related componentsHBV and HCV infection activate NLRP3 by promoting the production of ROS and the oxidative stress response; HBV and HCV infection activate NLRP3 by regulating the NF-κB pathway
Autoimmune liver disease (AIH,PBC,PSC)Autoimmune reaction; immune-mediated bile duct injury; cholestasisAbnormal expression of autoantigens may lead to an abnormal immune response in the liver, activating NLRP3; Oxidative stress and mitochondrial damage are involved in the activation of NLRP3; Cholestasis triggers TLR/NF-κB signaling and activates NLRP3

NLRP3 downstream molecules

IL-1β is a key inflammatory cell factor. It is an active version of IL-1 that is mainly produced by macrophages.72 PAMPs and DAMPs participate in the release of mature IL-1β and IL-18 by triggering NLRP3. IL-1β and IL-18 have biological activity and participate in fibrosis.4,72 IL-1β can regulate the expression of TIMPs and matrix metalloproteinases, which have an impact on fibrosis and tissue regeneration.73 The NLRP3/IL-1β secretory axis is also present in the HSCs.72In vitro studies have demonstrated that IL-1β can directly activate HSCs, promoting their proliferation and differentiation into myofibroblasts. The myofibroblasts increase the release of fibrosis markers such as collagen and TGF-β.13 IL-1β promotes fibrous tissue development by binding to cell surface IL-1β receptors.74 Endogenous inhibitors of IL-1β receptors were shown to improve liver fibrosis in a mouse model of alcoholic hepatitis.75

Multifunctional cytokine IL-18 has proinflammatory and fibrosis-promoting activity. IL-18 has previously been linked to the progression of fibrosis in the lungs, heart, and kidneys.76 It also has a key role in the progression of liver injury and liver fibrosis. Significant increase of IL-18 plasma level has been observed in chronic liver disease and hepatosclerosis.77 Increased IL-18 expression was found in the livers of NASH patients, and involvement in liver fibrosis.78 IL-18 can activate HSCs promoting their differentiation into myofibroblasts, upregulating the expression of collagen genes, and the production of connective tissue growth factor and α-SMA.76 As liver cells do not have IL-18 receptors, IL-18 cannot directly act on the hepatic cells. However, IL-18 can activate CD4+ T cells. The CD4+ T cells secrete various cell factors that exacerbate liver inflammation, progressing to liver fibrosis. In conjunction with this, anti-IL-18 therapy can reduce liver inflammation and noticeably delay liver fibrosis.79

Chronic liver disease and NLRP3

NAFLD

NAFLD includes a range of liver changes, starting with nonalcoholic fatty liver potentially progressing to NASH. In advanced cases, NASH can lead to cirrhosis, liver failure, and liver cancer.80 The occurrence and progression of NAFLD supposedly involve multiple parallel attacks involving different events such as lipid toxicity, chronic inflammation, and oxidative stress that simultaneously participate in the development of NAFLD (Table 1).81 Abnormal activation of NLRP3 is a major driver of liver injury, steatosis, inflammation, and fibrosis (Fig. 3).82,83 The role of abnormal activation of NLRP3 in NALFD has been extensively studied (Table 1). In NAFLD patients and NASH mouse models, activation of NLRP3 exacerbates liver inflammation and progression of liver fibrosis.9,82 In NASH patients, XBP1 promotes lipid accumulation and expression of proinflammatory factors in hepatocytes by activating NLRP3 in macrophages, thereby exacerbating the progression of steatohepatitis. On the contrary, XBP1 knockout in macrophages inhibited the expression of TGF-β and HSCs activation.57 Mitochondria-derived risk signals (ROS and mitochondrial dysfunction) promote expression of inflammatory factors and activate HSCs (Fig. 3), driving liver fibrosis in mice and NASH patients.84,85 Disturbed mitophagy was shown to activate NLRP3 inflammasomes, which was associated with the progression of nonalcoholic steatosis to nonalcoholic steatohepatitis.32 The above examples demonstrate the close relationship of NLRP3 with NAFLD. Many studies have suggested that inhibiting NLRP3 reduced liver inflammation and fibrosis. For instance, blocking NLRP3 inflammasome activation with echinatin can improve NASH and lessen liver inflammation and fibrosis.86 The NLRP3 inhibitor MCC950 was shown to reduce the severity of liver inflammation.9 Although MCC950 is an effective inhibitor of NLRP3, it was found to be hepatotoxic in phase II clinical trials of rheumatoid arthritis, which prevented further evaluation.87 Some traditional Chinese medicines and extracts, such as rhubarb-free anthraquinones, danshen, cryptotanshinone, etc., regulate the activation of NLRP3, thereby improving liver inflammation in NAFLD and NASH.88–91 Although targeting the inflammasome pathway can inhibit the development of NAFLD, the studies are still at an early stage, which limits clinical application.

ALD

ALD, which ranges from early steatosis to alcoholic fatty liver, cirrhosis, and liver cancer, is the result of liver damage brought on by long-term ethanol toxicity and a complex immunological reaction.23 Long-term ethanol consumption activates the innate immune system, producing proinflammatory and anti-inflammatory cytokines. It induces an inflammatory cascade in the liver and in the whole body.23 Long-term exposure to ethanol increases neutrophil and macrophage recruitment, which promotes the activation of NLRP3/caspase-1/ASC inflammasome and the release of pro-inflammatory cytokines (Table 1, Fig. 3). Mice lacking caspase-1, ASC, and IL-1 receptors had a reduction in ethanol-induced hepatic steatosis and inflammation.75,92 This suggests that NLRP3 activation in ALD is closely related to inflammatory response and liver injury. Correspondingly, inhibiting the activation of NLRP3 can improve the prognosis of alcoholic liver disease. For instance, diallyl disulfide was shown to inhibit the activation of ethanol-induced mouse liver NF-κB signals and NLRP3, slowing disease progression.93 Zeaxanthin dipalmitate inhibited hepatic inflammatory infiltration and fat droplet accumulation in a rat ALD model by restoring mitophagy that was impaired due to ethanol poisoning and suppressed NLRP3.94 A traditional Chinese medicine magnolol extract can inhibit NLRP3 preventing alcohol-induced liver injury.95

Ethanol inhibits the breakdown of fatty acids, which promotes fat accumulation in liver cells, which makes them prone to lipid peroxidation and oxidative damage (Fig. 3). ROS production by dysfunctional mitochondrial and oxidative stress are key causes of ALD. Oxidative metabolism of alcohol damages mitochondria, which produce ROS and activate NLRP3, causing inflammatory responses in the liver (Table 1).80 Ginsenoside Rg1 was shown to suppress NLRP3 activation by preventing oxidative stress, which alleviated pathological changes in the liver tissue of mice and rats on alcohol.96 Oroxylin A can reduce the accumulation of mitochondrial superoxide and intracellular ROS in hepatocytes induced by ethanol, thus mediating the inactivation of NLRP3.97 The inhibition of NLRP3 signaling can restrain the oxidative stress response in ALD, thus improving ALD. Traditional Chinese medicine extracts astragaloside IV was shown to inhibit the NLRP3/Caspase-1 inflammatory signaling pathway, alleviating alcohol-induced liver inflammation and oxidative stress in the liver.69 Moreover, hepatocytic pyroptosis is closely associated with NLRP3 activation in the pathogenesis of ALD. Diallyl trisulfide alleviates alcohol-induced hepatocyte apoptosis by downregulating the accumulation of intracellular ROS and inhibiting NLRP3.98 In conclusion, NLRP3 plays a pivotal role in the pathogenesis and progression of ALD, and suppression of NLRP3 activation can ameliorate the prognosis of alcoholic liver disease.

Viral hepatitis

Viral hepatitis is an infectious disease mainly caused by multiple hepatitis viruses (hepatitis A, B, C, D, and E viruses). The most common are hepatitis B and C. Viral infection activates the host immune response system, causing inflammatory responses activating NLRP3 (Table 1, Fig. 3). An excessive and ongoing inflammatory response causes chronic inflammatory disorders that lead to liver fibrosis. The expression levels of NLRP3, ASC, and IL-1β in the cytoplasm of hepatitis B virus (HBV)-negative patients are lower, while the same increase in HBV-positive patients.99 The severity of HBV-induced liver inflammation is proportional to the expression levels of NLRP3, gastric dermal protein D, caspase-1, IL-1β, and IL-18.100 Therefore, therapeutic targeting of NLRP3 can potentially suppress excessive inflammatory responses and alleviate inflammatory damage caused by viral hepatitis. HBV infection induces hepatic injury through the actions of HBV-associated proteins. Hepatitis B core antigen upregulates NLRP3 by promoting the phosphorylation of NF-κB thereby promoting liver injury.101 Hepatitis B virus X protein activates NLRP3 under oxidative stress, enhancing NLRP3 inflammasome-mediated inflammation and pyroptosis by enhancing the generation of mtROS in liver cells.99 Investigating the activation mechanisms of NLRP3 in hepatitis B virus infection can aid the development of NLRP3-directed antiviral therapies.

Hepatitis C virus (HCV) infection can activate NLRP3 inflammasomes, thus increasing the expression of NLRP3-related components in HCV-infected liver cells.12,102 NLRP3 can influence macrophage activation and promote the regulation of the immune response. HCV activates NLRP3 in liver macrophages or KCs, driving liver inflammation. HCV core protein activates NLRP3, promoting the production and release of IL-1β by macrophages.103 HCV infection activates NLRP3 in KCs by inducing potassium efflux, resulting in production of IL-1β. The secretion of IL-1β drives chemokines, proinflammatory cytokines, and immunoregulatory genes that are associated with the severity of HCV disease.104 NLRP3 is activated in HCV infection through the NF-κB signaling pathway. In addition, HCV infection can induce endoplasmic reticulum stress that increases the release of intracellular ROS and subsequently activates NLRP3.102 Although the aberrant activation of NLRP3 is important in viral hepatitis pathogenesis, the specific regulatory mechanism remains needs to be further explored.105

Autoimmune liver disease

Autoimmune hepatitis

Autoimmune hepatitis (AIH) is an autoimmune inflammation reaction in liver tissue, involving the action of innate immune cells, such as macrophages, T cells, and natural killer T cells (Fig. 3).106,107 NLRP3 is a component of the innate immune system that the occurrence and development of AIH. In the pathogenesis of AIH, T helper (Th)0 lymphocytes differentiate into Th1 and Th2 cells. Th1 can activate macrophages by secreting IL2 and interferon gamma, thereby releasing IL-1.106 TLRs 2, 4, and 9 can mediate the activation of inflammasomes in AIH and de novo autoimmune hepatitis, suggesting that the inflammasome activation has a role in the pathogenesis of AIH.108 NLRP3 inflammasomes are known to contribute to concanavalin A (Con A)-induced hepatitis (AIH model). NLRP3 and ASC expression levels are upregulated in Con A-induced hepatitis. NLRP3 inflammasome activation, IL-1β production, and pyroptosis were significantly increased in Con A-induced AIH mice.10 Recombinant human IL-1 receptor antagonists can inhibit NLRP3 in AIH by inhibiting ROS production and mitochondrial dysfunction in liver tissue.10 The activation of NLRP3 may involve the NF-κB (Table 1) and the protein kinase A (PKA) signaling pathways. Formononetin inhibits NLRP3 activation by inhibiting the NF-κB pathway and protects the liver against Con A-induced liver injury in mice.109 Dimethyl fumarate can inhibit the activation of the NLRP3 inflammasome by regulating the PKA signaling pathway, and prevent Con A-induced hepatitis.110 The regulatory mechanism of NLRP3 is extensive, and its relationship with the PKA and NF-κB signaling pathways in the pathogenesis of AIH should be intensely studied.

Primary biliary cholangitis

Primary biliary cholangitis (PBC) is a chronic inflammatory autoimmune cholestasis liver disease that is characterized by immune-mediated bile duct injury and is accompanied by chronic cholestasis.111,112 However, the specific pathogenesis of PBC is still unclear. Bile stasis can trigger TLR 4 signaling and enhance NF-κB activation, activating NLRP3 and thereby aggravating liver fibrosis (Table 1, Fig. 3).113 NLRP3 is involved in liver inflammation and fibrosis. It is not only expressed in immune cells but also in liver cells and bile duct cells.114 NLRP3 expression is significantly increased in the livers of PBC patients and mice, as shown by studies.115 Moreover, in a mouse PBC model, galectin-3 directly stimulated the activation of NLRP3, causing autoimmune cholangitis and fibrosis.116 MCC950, an NLRP3 inhibitor, can dramatically lessen bile duct ligation-induced liver injury by inhibiting the activation of NLRP3.117 Paeoniflorin can reduce the degree of liver injury and liver fibrosis in PBC mice by inhibiting NLRP3 and related cascade inflammatory pathways.115 Therefore, inhibiting NLRP3 and related cascading inflammatory pathways may be a new approach to the prevention and treatment of PBC.

Primary sclerosing cholangitis

Primary sclerosing cholangitis (PSC) is also a chronic cholestatic liver disease. Inflammation and fibrosis result in multifocal biliary strictures and end-stage liver disease. The etiology of PSC is not clear, and so far, there is no specific and effective treatment. Elevated markers of NLRP3 inflammasome activation have been detected in liver biopsies of PSC patients.118 NLRP3 immunostaining had positive expression of reactive bile duct cells in the livers of PSC patients and mouse PSC models, suggesting that activation of NLRP3 may have a role.119 Although NLRP3 does not affect the proliferation of bile duct cells, it can destroy the integrity of bile duct epithelium, increasing epithelial permeability.114,120 It has been established that primary pathogenesis of PSC in Mdr2−/− mice (a common PSC mouse model) includes loss of the integrity of the bile duct epithelial cell layer.120 Furthermore, NLRP3 was found significantly activated in human PSC and Mdr2−/− livers. The extent of liver fibrosis in PSC patients positively correlates with the levels of NLRP3 and IL-1β.65 Therefore, targeting NLRP3 is a new direction in the treatment of PSC.

Conclusion

This article provides a comprehensive review of the relationship between NLRP3 and liver fibrosis. The process of liver fibrosis involves the interaction of multiple cells and molecules. The mechanisms of these interactions need to be further studied in the future. Several NLRP3 inflammasome-related molecular inhibitors have been studied in liver diseases and have shown good results in reducing inflammation, fibrosis, and other tissue damage. Many traditional Chinese medicines have lipid-metabolism regulating, anti-inflammatory, and antioxidant effects, and alleviate hepatitis and liver fibrosis by inhibiting the NLRP3 inflammatory pathway. Furthermore, traditional Chinese medicines can slow down the progression of chronic liver disease by regulating the gut microbiome.68,69 These experimental studies provide a preliminary foundation for clinical practice and new strategies for the development of drugs or treatments targeting NLRP3. In the future, the pharmacological effects of NLRP3-related molecular inhibitors and the synergistic effects of other drugs, especially traditional Chinese medicine preparations, are worthy of further exploration. Although some preliminary clinical trials and animal studies have shown the potential efficacy of targeted NLRP3 therapy, it has not yet been widely applied in clinical practice. The activation mechanism of the NLRP3 pathway is not fully understood and therefore targeted NLRP3 therapy needs deeper evaluations.

Abbreviations

AIH: 

autoimmune hepatitis

ALD: 

alcohol-related liver disease

ASC: 

apoptosis-associated speck-like protein containing a caspase-recruitment domain

Con A: 

concanavalin A

DAMPs: 

damage-associated molecular patterns

ECM: 

extracellular matrix

HBV: 

hepatitis B virus

HCV: 

hepatitis C virus

HSCs: 

hepatic stellate cells

IL: 

interleukin

KCs: 

Kupffer cells

LPS: 

lipopolysaccharide

Mt: 

mitochondrial

NAFLD: 

nonalcoholic liver disease

NASH: 

nonalcoholic steatohepatitis

NF-κB: 

transcription factor nuclear factor-kappa B

NLRP1: 

NOD-like receptor protein 1

NLRP3: 

NOD-like receptor protein 3

PAMPs: 

pathogen-associated molecular patterns

PBC: 

Primary biliary cholangitis

PKA: 

protein kinas A

pro-IL-1β: 

IL-1β precursor

pro-IL-18: 

IL-18 precursor

PSC: 

Primary sclerosing cholangitis

ROS: 

reactive oxygen species

TGF-β: 

transforming growth factor beta

TIMPs: 

tissue inhibitors of metalloproteinases

TLRs: 

Toll-like receptors

Th: 

T helper

XBP1

X-box binding protein-1

α-SMA: 

alpha smooth muscle actin

Declarations

Funding

The work was supported in part by Shandong Provincial Natural Science Foundation (ZR2023MH295, LJP) and Linyi People’s Hospital Postgraduate Fund Project (YJS2023002, MHS).

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

Literature review and manuscript writing (MS), giving suggestions and revising the manuscript (YZ, AG), drafting figures (ZX), and topic conception and critical revision (LP). All authors made significant contributions to this study and have approved the final manuscript.

References

  1. Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E, Kamath PS. Global burden of liver disease: 2023 update. J Hepatol 2023;79(2):516-537 View Article PubMed/NCBI
  2. Seki E, Schwabe RF. Hepatic inflammation and fibrosis: functional links and key pathways. Hepatology 2015;61(3):1066-1079 View Article PubMed/NCBI
  3. Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int J Mol Sci 2019;20(13):3328 View Article PubMed/NCBI
  4. Duspara K, Bojanic K, Pejic JI, Kuna L, Kolaric TO, Nincevic V, et al. Targeting the Wnt Signaling Pathway in Liver Fibrosis for Drug Options: An Update. J Clin Transl Hepatol 2021;9(6):960-971 View Article PubMed/NCBI
  5. Pandey A, Shen C, Feng S, Man SM. Cell biology of inflammasome activation. Trends Cell Biol 2021;31(11):924-939 View Article PubMed/NCBI
  6. Kong R, Sun L, Li H, Wang D. The role of NLRP3 inflammasome in the pathogenesis of rheumatic disease. Autoimmunity 2022;55(1):1-7 View Article PubMed/NCBI
  7. Li Z, Guo J, Bi L. Role of the NLRP3 inflammasome in autoimmune diseases. Biomed Pharmacother 2020;130:110542 View Article PubMed/NCBI
  8. Huang Y, Xu W, Zhou R. NLRP3 inflammasome activation and cell death. Cell Mol Immunol 2021;18(9):2114-2127 View Article PubMed/NCBI
  9. Mridha AR, Wree A, Robertson AAB, Yeh MM, Johnson CD, Van Rooyen DM, et al. NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J Hepatol 2017;66(5):1037-1046 View Article PubMed/NCBI
  10. Luan J, Zhang X, Wang S, Li Y, Fan J, Chen W, et al. NOD-Like Receptor Protein 3 Inflammasome-Dependent IL-1beta Accelerated ConA-Induced Hepatitis. Front Immunol 2018;9:758 View Article PubMed/NCBI
  11. Gieling RG, Wallace K, Han YP. Interleukin-1 participates in the progression from liver injury to fibrosis. Am J Physiol Gastrointest Liver Physiol 2009;296(6):G1324-1331 View Article PubMed/NCBI
  12. Szabo G, Petrasek J. Inflammasome activation and function in liver disease. Nat Rev Gastroenterol Hepatol 2015;12(7):387-400 View Article PubMed/NCBI
  13. de Carvalho Ribeiro M, Szabo G. Role of the Inflammasome in Liver Disease. Annu Rev Pathol 2022;17:345-365 View Article PubMed/NCBI
  14. Downs KP, Nguyen H, Dorfleutner A, Stehlik C. An overview of the non-canonical inflammasome. Mol Aspects Med 2020;76:100924 View Article PubMed/NCBI
  15. Kayagaki N, Warming S, Lamkanfi M, Vande Walle L, Louie S, Dong J, et al. Non-canonical inflammasome activation targets caspase-11. Nature 2011;479(7371):117-121 View Article PubMed/NCBI
  16. Zhu F, Ma J, Li W, Liu Q, Qin X, Qian Y, et al. The orphan receptor Nur77 binds cytoplasmic LPS to activate the non-canonical NLRP3 inflammasome. Immunity 2023;56(4):753-767.e758 View Article PubMed/NCBI
  17. Di A, Xiong S, Ye Z, Malireddi RKS, Kometani S, Zhong M, et al. The TWIK2 Potassium Efflux Channel in Macrophages Mediates NLRP3 Inflammasome-Induced Inflammation. Immunity 2018;49(1):56-65.e54 View Article PubMed/NCBI
  18. Surprenant A, Rassendren F, Kawashima E, North RA, Buell G. The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7). Science 1996;272(5262):735-738 View Article PubMed/NCBI
  19. Akbal A, Dernst A, Lovotti M, Mangan MSJ, McManus RM, Latz E. How location and cellular signaling combine to activate the NLRP3 inflammasome. Cell Mol Immunol 2022;19(11):1201-1214 View Article PubMed/NCBI
  20. Munoz-Planillo R, Kuffa P, Martinez-Colon G, Smith BL, Rajendiran TM, Nunez G. K(+) efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity 2013;38(6):1142-1153 View Article PubMed/NCBI
  21. Yang D, He Y, Munoz-Planillo R, Liu Q, Nunez G. Caspase-11 Requires the Pannexin-1 Channel and the Purinergic P2X7 Pore to Mediate Pyroptosis and Endotoxic Shock. Immunity 2015;43(5):923-932 View Article PubMed/NCBI
  22. Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol 2008;9(8):847-856 View Article PubMed/NCBI
  23. Le Dare B, Ferron PJ, Gicquel T. The Purinergic P2X7 Receptor-NLRP3 Inflammasome Pathway: A New Target in Alcoholic Liver Disease?. Int J Mol Sci 2021;22(4):2139 View Article PubMed/NCBI
  24. Mohammed S, Nicklas EH, Thadathil N, Selvarani R, Royce GH, Kinter M, et al. Role of necroptosis in chronic hepatic inflammation and fibrosis in a mouse model of increased oxidative stress. Free Radic Biol Med 2021;164:315-328 View Article PubMed/NCBI
  25. Zhan SS, Jiang JX, Wu J, Halsted C, Friedman SL, Zern MA, et al. Phagocytosis of apoptotic bodies by hepatic stellate cells induces NADPH oxidase and is associated with liver fibrosis in vivo. Hepatology 2006;43(3):435-443 View Article PubMed/NCBI
  26. Ramos-Tovar E, Muriel P. Molecular Mechanisms That Link Oxidative Stress, Inflammation, and Fibrosis in the Liver. Antioxidants (Basel) 2020;9(12):1279 View Article PubMed/NCBI
  27. Pang Y, Wu D, Ma Y, Cao Y, Liu Q, Tang M, et al. Reactive oxygen species trigger NF-kappaB-mediated NLRP3 inflammasome activation involvement in low-dose CdTe QDs exposure-induced hepatotoxicity. Redox Biol 2021;47:102157 View Article PubMed/NCBI
  28. Abais JM, Xia M, Zhang Y, Boini KM, Li PL. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector?. Antioxid Redox Signal 2015;22(13):1111-1129 View Article PubMed/NCBI
  29. Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol 2010;11(2):136-140 View Article PubMed/NCBI
  30. Dominic A, Le NT, Takahashi M. Loop Between NLRP3 Inflammasome and Reactive Oxygen Species. Antioxid Redox Signal 2022;36(10-12):784-796 View Article PubMed/NCBI
  31. Zhong Z, Liang S, Sanchez-Lopez E, He F, Shalapour S, Lin XJ, et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature 2018;560(7717):198-203 View Article PubMed/NCBI
  32. Zhang NP, Liu XJ, Xie L, Shen XZ, Wu J. Impaired mitophagy triggers NLRP3 inflammasome activation during the progression from nonalcoholic fatty liver to nonalcoholic steatohepatitis. Lab Invest 2019;99(6):749-763 View Article PubMed/NCBI
  33. Hoyt LR, Randall MJ, Ather JL, DePuccio DP, Landry CC, Qian X, et al. Mitochondrial ROS induced by chronic ethanol exposure promote hyper-activation of the NLRP3 inflammasome. Redox Biol 2017;12:883-896 View Article PubMed/NCBI
  34. Zhao Y, Wang Z, Feng D, Zhao H, Lin M, Hu Y, et al. p66Shc Contributes to Liver Fibrosis through the Regulation of Mitochondrial Reactive Oxygen Species. Theranostics 2019;9(5):1510-1522 View Article PubMed/NCBI
  35. Mills EL, Kelly B, O’Neill LAJ. Mitochondria are the powerhouses of immunity. Nat Immunol 2017;18(5):488-498 View Article PubMed/NCBI
  36. Wang Y, Shi P, Chen Q, Huang Z, Zou D, Zhang J, et al. Mitochondrial ROS promote macrophage pyroptosis by inducing GSDMD oxidation. J Mol Cell Biol 2019;11(12):1069-1082 View Article PubMed/NCBI
  37. Xu Y, Tang Y, Lu J, Zhang W, Zhu Y, Zhang S, et al. PINK1-mediated mitophagy protects against hepatic ischemia/reperfusion injury by restraining NLRP3 inflammasome activation. Free Radic Biol Med 2020;160:871-886 View Article PubMed/NCBI
  38. Yang G, Lee HE, Lee JY. A pharmacological inhibitor of NLRP3 inflammasome prevents non-alcoholic fatty liver disease in a mouse model induced by high fat diet. Sci Rep 2016;6:24399 View Article PubMed/NCBI
  39. Gross CJ, Mishra R, Schneider KS, Medard G, Wettmarshausen J, Dittlein DC, et al. K(+) Efflux-Independent NLRP3 Inflammasome Activation by Small Molecules Targeting Mitochondria. Immunity 2016;45(4):761-773 View Article PubMed/NCBI
  40. Hong F, Zhao M, Xue LL, Ma X, Liu L, Cai XY, et al. The ethanolic extract of Artemisia anomala exerts anti-inflammatory effects via inhibition of NLRP3 inflammasome. Phytomedicine 2022;102:154163 View Article PubMed/NCBI
  41. Yang B, Wang Y, Fang C, Song E, Song Y. Polybrominated diphenyl ether quinone exposure leads to ROS-driven lysosomal damage, mitochondrial dysfunction and NLRP3 inflammasome activation. Environ Pollut 2022;311:119846 View Article PubMed/NCBI
  42. Zhang T, Zhao J, Liu T, Cheng W, Wang Y, Ding S, et al. A novel mechanism for NLRP3 inflammasome activation. Metabol Open 2022;13:100166 View Article PubMed/NCBI
  43. Hou Y, He H, Ma M, Zhou R. Apilimod activates the NLRP3 inflammasome through lysosome-mediated mitochondrial damage. Front Immunol 2023;14:1128700 View Article PubMed/NCBI
  44. Zhang WJ, Fang ZM, Liu WQ. NLRP3 inflammasome activation from Kupffer cells is involved in liver fibrosis of Schistosoma japonicum-infected mice via NF-kappaB. Parasit Vectors 2019;12(1):29 View Article PubMed/NCBI
  45. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021;18(3):151-166 View Article PubMed/NCBI
  46. Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev 2017;121:27-42 View Article PubMed/NCBI
  47. Zhang CY, Yuan WG, He P, Lei JH, Wang CX. Liver fibrosis and hepatic stellate cells: Etiology, pathological hallmarks and therapeutic targets. World J Gastroenterol 2016;22(48):10512-10522 View Article PubMed/NCBI
  48. Tao Y, Wang N, Qiu T, Sun X. The Role of Autophagy and NLRP3 Inflammasome in Liver Fibrosis. Biomed Res Int 2020;2020:7269150 View Article PubMed/NCBI
  49. Watanabe A, Sohail MA, Gomes DA, Hashmi A, Nagata J, Sutterwala FS, et al. Inflammasome-mediated regulation of hepatic stellate cells. Am J Physiol Gastrointest Liver Physiol 2009;296(6):G1248-1257 View Article PubMed/NCBI
  50. Inzaugarat ME, Johnson CD, Holtmann TM, McGeough MD, Trautwein C, Papouchado BG, et al. NLR Family Pyrin Domain-Containing 3 Inflammasome Activation in Hepatic Stellate Cells Induces Liver Fibrosis in Mice. Hepatology 2019;69(2):845-859 View Article PubMed/NCBI
  51. Wree A, Eguchi A, McGeough MD, Pena CA, Johnson CD, Canbay A, et al. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology 2014;59(3):898-910 View Article PubMed/NCBI
  52. Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol 2017;14(7):397-411 View Article PubMed/NCBI
  53. Guilliams M, Scott CL. Liver macrophages in health and disease. Immunity 2022;55(9):1515-1529 View Article PubMed/NCBI
  54. Wan J, Benkdane M, Teixeira-Clerc F, Bonnafous S, Louvet A, Lafdil F, et al. M2 Kupffer cells promote M1 Kupffer cell apoptosis: a protective mechanism against alcoholic and nonalcoholic fatty liver disease. Hepatology 2014;59(1):130-142 View Article PubMed/NCBI
  55. Nieto N. Oxidative-stress and IL-6 mediate the fibrogenic effects of [corrected] Kupffer cells on stellate cells. Hepatology 2006;44(6):1487-1501 View Article PubMed/NCBI
  56. Hou L, Yang L, Chang N, Zhao X, Zhou X, Dong C, et al. Macrophage Sphingosine 1-Phosphate Receptor 2 Blockade Attenuates Liver Inflammation and Fibrogenesis Triggered by NLRP3 Inflammasome. Front Immunol 2020;11:1149 View Article PubMed/NCBI
  57. Wang Q, Zhou H, Bu Q, Wei S, Li L, Zhou J, et al. Role of XBP1 in regulating the progression of non-alcoholic steatohepatitis. J Hepatol 2022;77(2):312-325 View Article PubMed/NCBI
  58. Hou L, Zhang Z, Yang L, Chang N, Zhao X, Zhou X, et al. NLRP3 inflammasome priming and activation in cholestatic liver injury via the sphingosine 1-phosphate/S1P receptor 2/Galpha(12/13)/MAPK signaling pathway. J Mol Med (Berl) 2021;99(2):273-288 View Article PubMed/NCBI
  59. Liu Y, Kong X, You Y, Xiang L, Zhang Y, Wu R, et al. S100A8-Mediated NLRP3 Inflammasome-Dependent Pyroptosis in Macrophages Facilitates Liver Fibrosis Progression. Cells 2022;11(22):3579 View Article PubMed/NCBI
  60. Jiang S, Zhang Y, Zheng JH, Li X, Yao YL, Wu YL, et al. Potentiation of hepatic stellate cell activation by extracellular ATP is dependent on P2X7R-mediated NLRP3 inflammasome activation. Pharmacol Res 2017;117:82-93 View Article PubMed/NCBI
  61. Pan H, Jian Y, Wang F, Yu S, Guo J, Kan J, et al. NLRP3 and Gut Microbiota Homeostasis: Progress in Research. Cells 2022;11(23):3758 View Article PubMed/NCBI
  62. Bawa M, Saraswat VA. Gut-liver axis: role of inflammasomes. J Clin Exp Hepatol 2013;3(2):141-149 View Article PubMed/NCBI
  63. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012;482(7384):179-185 View Article PubMed/NCBI
  64. Plaza-Diaz J, Solis-Urra P, Rodriguez-Rodriguez F, Olivares-Arancibia J, Navarro-Oliveros M, Abadia-Molina F, et al. The Gut Barrier, Intestinal Microbiota, and Liver Disease: Molecular Mechanisms and Strategies to Manage. Int J Mol Sci 2020;21(21):8351 View Article PubMed/NCBI
  65. Liao L, Schneider KM, Galvez EJC, Frissen M, Marschall HU, Su H, et al. Intestinal dysbiosis augments liver disease progression via NLRP3 in a murine model of primary sclerosing cholangitis. Gut 2019;68(8):1477-1492 View Article PubMed/NCBI
  66. Cheng Z, Yang L, Chu H. The Gut Microbiota: A Novel Player in Autoimmune Hepatitis. Front Cell Infect Microbiol 2022;12:947382 View Article PubMed/NCBI
  67. Nie Y, Liu Q, Zhang W, Wan Y, Huang C, Zhu X. Ursolic acid reverses liver fibrosis by inhibiting NOX4/NLRP3 inflammasome pathways and bacterial dysbiosis. Gut Microbes 2021;13(1):1972746 View Article PubMed/NCBI
  68. Lin YP, Fang QL, Xue YM, Fu SN, Hu CY, Huang F, et al. Effects of Tylophora yunnanensis Schltr on regulating the gut microbiota and its metabolites in non-alcoholic steatohepatitis rats by inhibiting the activation of NOD-like receptor protein 3. J Ethnopharmacol 2023;305:116145 View Article PubMed/NCBI
  69. Wu S, Wen F, Zhong X, Du W, Chen M, Wang J. Astragaloside IV ameliorate acute alcohol-induced liver injury in mice via modulating gut microbiota and regulating NLRP3/caspase-1 signaling pathway. Ann Med 2023;55(1):2216942 View Article PubMed/NCBI
  70. Ding Q, Cao F, Lai S, Zhuge H, Chang K, Valencak TG, et al. Lactobacillus plantarum ZY08 relieves chronic alcohol-induced hepatic steatosis and liver injury in mice via restoring intestinal flora homeostasis. Food Res Int 2022;157:111259 View Article PubMed/NCBI
  71. Wang W, Xu AL, Li ZC, Li Y, Xu SF, Sang HC, et al. Combination of Probiotics and Salvia miltiorrhiza Polysaccharide Alleviates Hepatic Steatosis via Gut Microbiota Modulation and Insulin Resistance Improvement in High Fat-Induced NAFLD Mice. Diabetes Metab J 2020;44(2):336-348 View Article PubMed/NCBI
  72. Szabo G, Csak T. Inflammasomes in liver diseases. J Hepatol 2012;57(3):642-654 View Article PubMed/NCBI
  73. Robert S, Gicquel T, Bodin A, Fautrel A, Barreto E, Victoni T, et al. Influence of inflammasome pathway activation in macrophages on the matrix metalloproteinase expression of human hepatic stellate cells. Int Immunopharmacol 2019;72:12-20 View Article PubMed/NCBI
  74. Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 2009;27:519-550 View Article PubMed/NCBI
  75. Petrasek J, Bala S, Csak T, Lippai D, Kodys K, Menashy V, et al. IL-1 receptor antagonist ameliorates inflammasome-dependent alcoholic steatohepatitis in mice. J Clin Invest 2012;122(10):3476-3489 View Article PubMed/NCBI
  76. Knorr J, Kaufmann B, Inzaugarat ME, Holtmann TM, Geisler L, Hundertmark J, et al. Interleukin-18 signaling promotes activation of hepatic stellate cells in mouse liver fibrosis. Hepatology 2023;77(6):1968-1982 View Article PubMed/NCBI
  77. Ludwiczek O, Kaser A, Novick D, Dinarello CA, Rubinstein M, Vogel W, et al. Plasma levels of interleukin-18 and interleukin-18 binding protein are elevated in patients with chronic liver disease. Journal of clinical immunology 2002;22(6):331-337 View Article PubMed/NCBI
  78. Wree A, McGeough MD, Peña CA, Schlattjan M, Li H, Inzaugarat ME, et al. NLRP3 inflammasome activation is required for fibrosis development in NAFLD. J Mol Med (Berl) 2014;92(10):1069-1082 View Article PubMed/NCBI
  79. Zhang Y, Li P, Li G, Huang X, Meng Q, Lau WY, et al. The mechanism of how anti-IL-18 prevents concanavalin-A-induced hepatic fibrosis on a mouse model. J Surg Res 2007;142(1):175-183 View Article PubMed/NCBI
  80. Torres S, Segales P, Garcia-Ruiz C, Fernandez-Checa JC. Mitochondria and the NLRP3 Inflammasome in Alcoholic and Nonalcoholic Steatohepatitis. Cells 2022;11(9):1475 View Article PubMed/NCBI
  81. Hu J, Wang H, Li X, Liu Y, Mi Y, Kong H, et al. Fibrinogen-like protein 2 aggravates nonalcoholic steatohepatitis via interaction with TLR4, eliciting inflammation in macrophages and inducing hepatic lipid metabolism disorder. Theranostics 2020;10(21):9702-9720 View Article PubMed/NCBI
  82. Unamuno X, Gomez-Ambrosi J, Ramirez B, Rodriguez A, Becerril S, Valenti V, et al. NLRP3 inflammasome blockade reduces adipose tissue inflammation and extracellular matrix remodeling. Cell Mol Immunol 2021;18(4):1045-1057 View Article PubMed/NCBI
  83. Yu L, Hong W, Lu S, Li Y, Guan Y, Weng X, et al. The NLRP3 Inflammasome in Non-Alcoholic Fatty Liver Disease and Steatohepatitis: Therapeutic Targets and Treatment. Front Pharmacol 2022;13:780496 View Article PubMed/NCBI
  84. Cortez-Pinto H, de Moura MC, Day CP. Non-alcoholic steatohepatitis: from cell biology to clinical practice. J Hepatol 2006;44(1):197-208 View Article PubMed/NCBI
  85. Loureiro D, Tout I, Narguet S, Bed CM, Roinard M, Sleiman A, et al. Mitochondrial stress in advanced fibrosis and cirrhosis associated with chronic hepatitis B, chronic hepatitis C, or nonalcoholic steatohepatitis. Hepatology 2023;77(4):1348-1365 View Article PubMed/NCBI
  86. Xu G, Fu S, Zhan X, Wang Z, Zhang P, Shi W, et al. Echinatin effectively protects against NLRP3 inflammasome-driven diseases by targeting HSP90. JCI Insight 2021;6(2):e134601 View Article PubMed/NCBI
  87. Li H, Guan Y, Liang B, Ding P, Hou X, Wei W, et al. Therapeutic potential of MCC950, a specific inhibitor of NLRP3 inflammasome. Eur J Pharmacol 2022;928:175091 View Article PubMed/NCBI
  88. Liu T, Xu G, Liang L, Xiao X, Zhao Y, Bai Z. Pharmacological effects of Chinese medicine modulating NLRP3 inflammasomes in fatty liver treatment. Front Pharmacol 2022;13:967594 View Article PubMed/NCBI
  89. Wu C, Bian Y, Lu B, Wang D, Azami NLB, Wei G, et al. Rhubarb free anthraquinones improved mice nonalcoholic fatty liver disease by inhibiting NLRP3 inflammasome. J Transl Med 2022;20(1):294 View Article PubMed/NCBI
  90. Biao Y, Chen J, Liu C, Wang R, Han X, Li L, et al. Protective Effect of Danshen Zexie Decoction Against Non-Alcoholic Fatty Liver Disease Through Inhibition of ROS/NLRP3/IL-1beta Pathway by Nrf2 Signaling Activation. Front Pharmacol 2022;13:877924 View Article PubMed/NCBI
  91. Liu H, Zhan X, Xu G, Wang Z, Li R, Wang Y, et al. Cryptotanshinone specifically suppresses NLRP3 inflammasome activation and protects against inflammasome-mediated diseases. Pharmacol Res 2021;164:105384 View Article PubMed/NCBI
  92. Shang Y, Yang HX, Li X, Zhang Y, Chen N, Jiang XL, et al. Modulation of interleukin-36 based inflammatory feedback loop through the hepatocyte-derived IL-36R-P2X7R axis improves steatosis in alcoholic steatohepatitis. Br J Pharmacol 2022;179(17):4378-4399 View Article PubMed/NCBI
  93. Liu SX, Liu H, Wang S, Zhang CL, Guo FF, Zeng T. Diallyl disulfide ameliorates ethanol-induced liver steatosis and inflammation by maintaining the fatty acid catabolism and regulating the gut-liver axis. Food Chem Toxicol 2022;164:113108 View Article PubMed/NCBI
  94. Gao H, Lv Y, Liu Y, Li J, Wang X, Zhou Z, et al. Wolfberry-Derived Zeaxanthin Dipalmitate Attenuates Ethanol-Induced Hepatic Damage. Mol Nutr Food Res 2019;63(11):e1801339 View Article PubMed/NCBI
  95. Liu X, Wang Y, Wu D, Li S, Wang C, Han Z, et al. Magnolol Prevents Acute Alcoholic Liver Damage by Activating PI3K/Nrf2/PPARgamma and Inhibiting NLRP3 Signaling Pathway. Front Pharmacol 2019;10:1459 View Article PubMed/NCBI
  96. Yang C, He X, Zhao J, Huang W. Hepatoprotection by Ginsenoside Rg1 in alcoholic liver disease. Int Immunopharmacol 2021;92:107327 View Article PubMed/NCBI
  97. Kai J, Yang X, Wang Z, Wang F, Jia Y, Wang S, et al. Oroxylin a promotes PGC-1alpha/Mfn2 signaling to attenuate hepatocyte pyroptosis via blocking mitochondrial ROS in alcoholic liver disease. Free Radic Biol Med 2020;153:89-102 View Article PubMed/NCBI
  98. Zhu X, Lu R, Zhang G, Fan L, Zhan Y, Chen G, et al. Diallyl Trisulfide attenuates alcohol-induced hepatocyte pyroptosis via elevation of hydrogen sulfide. Biosci Biotechnol Biochem 2022;86(11):1552-1561 View Article PubMed/NCBI
  99. Xie WH, Ding J, Xie XX, Yang XH, Wu XF, Chen ZX, et al. Hepatitis B virus X protein promotes liver cell pyroptosis under oxidative stress through NLRP3 inflammasome activation. Inflamm Res 2020;69(7):683-696 View Article PubMed/NCBI
  100. Wang Y, Li X, Chen Q, Jiao F, Shi C, Pei M, et al. The relationship between liver pathological inflammation degree and pyroptosis in chronic hepatitis B patients. J Med Virol 2021;93(11):6229-6235 View Article PubMed/NCBI
  101. Ding X, Lei Q, Li T, Li L, Qin B. Hepatitis B core antigen can regulate NLRP3 inflammasome pathway in HepG2 cells. J Med Virol 2019;91(8):1528-1536 View Article PubMed/NCBI
  102. Ramachandran A, Kumar B, Waris G, Everly D. Deubiquitination and Activation of the NLRP3 Inflammasome by UCHL5 in HCV-Infected Cells. Microbiol Spectr 2021;9(1):e0075521 View Article PubMed/NCBI
  103. Negash AA, Olson RM, Griffin S, Gale M. Modulation of calcium signaling pathway by hepatitis C virus core protein stimulates NLRP3 inflammasome activation. PLoS Pathog 2019;15(2):e1007593 View Article PubMed/NCBI
  104. Negash AA, Ramos HJ, Crochet N, Lau DT, Doehle B, Papic N, et al. IL-1beta production through the NLRP3 inflammasome by hepatic macrophages links hepatitis C virus infection with liver inflammation and disease. PLoS Pathog 2013;9(4):e1003330 View Article PubMed/NCBI
  105. Tai DI, Tsai SL, Chen YM, Chuang YL, Peng CY, Sheen IS, et al. Activation of nuclear factor kappaB in hepatitis C virus infection: implications for pathogenesis and hepatocarcinogenesis. Hepatology 2000;31(3):656-664 View Article PubMed/NCBI
  106. Muratori L, Longhi MS. The interplay between regulatory and effector T cells in autoimmune hepatitis: Implications for innovative treatment strategies. J Autoimmun 2013;46:74-80 View Article PubMed/NCBI
  107. Wu YN, Zhang R, Song XC, Han XX, Zhang J, Li X. C6orf120 gene knockout in rats mitigates concanavalin A-induced autoimmune hepatitis via regulating NKT cells. Cell Immunol 2022;371:104467 View Article PubMed/NCBI
  108. Arterbery AS, Yao J, Ling A, Avitzur Y, Martinez M, Lobritto S, et al. Inflammasome Priming Mediated via Toll-Like Receptors 2 and 4, Induces Th1-Like Regulatory T Cells in De Novo Autoimmune Hepatitis. Front Immunol 2018;9:1612 View Article PubMed/NCBI
  109. Liu G, Zhao W, Bai J, Cui J, Liang H, Lu B. Formononetin protects against concanavalin-A-induced autoimmune hepatitis in mice through its anti-apoptotic and anti-inflammatory properties. Biochem Cell Biol 2021;99(2):231-240 View Article PubMed/NCBI
  110. Shi FL, Ni ST, Luo SQ, Hu B, Xu R, Liu SY, et al. Dimethyl fumarate ameliorates autoimmune hepatitis in mice by blocking NLRP3 inflammasome activation. Int Immunopharmacol 2022;108:108867 View Article PubMed/NCBI
  111. Sarcognato S, Sacchi D, Grillo F, Cazzagon N, Fabris L, Cadamuro M, et al. Autoimmune biliary diseases: primary biliary cholangitis and primary sclerosing cholangitis. Pathologica 2021;113(3):170-184 View Article PubMed/NCBI
  112. Lleo A, Leung PSC, Hirschfield GM, Gershwin EM. The Pathogenesis of Primary Biliary Cholangitis: A Comprehensive Review. Semin Liver Dis 2020;40(1):34-48 View Article PubMed/NCBI
  113. Li Z, Chen D, Jia Y, Feng Y, Wang C, Tong Y, et al. Methane-Rich Saline Counteracts Cholestasis-Induced Liver Damage via Regulating the TLR4/NF-kappaB/NLRP3 Inflammasome Pathway. Oxid Med Cell Longev 2019;2019:6565283 View Article PubMed/NCBI
  114. Maroni L, Ninfole E, Pinto C, Benedetti A, Marzioni M. Gut-Liver Axis and Inflammasome Activation in Cholangiocyte Pathophysiology. Cells 2020;9(3):736 View Article PubMed/NCBI
  115. Zhang Y, Zhang S, Luo X, Zhao H, Xiang X. Paeoniflorin mitigates PBC-induced liver fibrosis by repressing NLRP3 formation. Acta Cir Bras 2022;36(11):e361106 View Article PubMed/NCBI
  116. Tian J, Yang G, Chen HY, Hsu DK, Tomilov A, Olson KA, et al. Galectin-3 regulates inflammasome activation in cholestatic liver injury. FASEB J 2016;30(12):4202-4213 View Article PubMed/NCBI
  117. Qu J, Yuan Z, Wang G, Wang X, Li K. The selective NLRP3 inflammasome inhibitor MCC950 alleviates cholestatic liver injury and fibrosis in mice. Int Immunopharmacol 2019;70:147-155 View Article PubMed/NCBI
  118. Guan Y, Gu Y, Li H, Liang B, Han C, Zhang Y, et al. NLRP3 inflammasome activation mechanism and its role in autoimmune liver disease. Acta Biochim Biophys Sin (Shanghai) 2022;54(11):1577-1586 View Article PubMed/NCBI
  119. Maroni L, Agostinelli L, Saccomanno S, Pinto C, Giordano DM, Rychlicki C, et al. Nlrp3 Activation Induces Il-18 Synthesis and Affects the Epithelial Barrier Function in Reactive Cholangiocytes. Am J Pathol 2017;187(2):366-376 View Article PubMed/NCBI
  120. Fickert P, Fuchsbichler A, Wagner M, Zollner G, Kaser A, Tilg H, et al. Regurgitation of bile acids from leaky bile ducts causes sclerosing cholangitis in Mdr2 (Abcb4) knockout mice. Gastroenterology 2004;127(1):261-274 View Article PubMed/NCBI
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
  • Copyright © 2022 JCTH. All Rights Reserved.
  • Published by Xia & He Publishing Inc.
  • Address: 14090 Southwest Freeway, Suite 300, Sugar Land, Texas 77478, USA
  • Email: service@xiahepublishing.com