Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Mol. Biosci., 16 July 2025

Sec. Cellular Biochemistry

Volume 12 - 2025 | https://doi.org/10.3389/fmolb.2025.1634132

An overview of current research on the modulation of NLRP3 inflammasome by traditional Chinese medicine to combat acute pancreatitis

Xiongjie HeXiongjie He1Jia XiaJia Xia1Qi ChenQi Chen1Zhaozhao HuangZhaozhao Huang1Juanjuan LuJuanjuan Lu2Yisong Ren
Yisong Ren1*
  • 1Department of Critical Care Medicine, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan, China
  • 2Department of Endocrinology, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan, China

Acute pancreatitis (AP), a life-threatening gastrointestinal emergency, is characterized by acute onset, rapid clinical deterioration, and high mortality rates, imposing profound long-term health burdens and socioeconomic costs on patients and healthcare systems. Current therapeutic strategies focus on supportive care, as no curative therapies exist to halt AP progression. Traditional Chinese medicine (TCM), with its multi-target, multi-component, and multi-pathway pharmacological properties, has emerged as a promising therapeutic drug against inflammation-driven pathologies, including AP. This review systematically discussed the assembly, activation, and pathogenic contributions of the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome in AP pathogenesis. Mechanistically, NLRP3 activation exacerbated pancreatic injury through caspase-1-dependent maturation of interleukin-1β (IL-1β) and gasdermin D (GSDMD)-mediated pyroptosis, perpetuating systemic inflammation. We systematically summarized the research progress of TCM in the treatment of AP by reducing pancreatic necrosis, neutrophil infiltration, and intestinal barrier dysfunction through targeting NLRP3 inflammasome, as well as its clinical evidence. Collectively, this review highlights the translational potential of TCM as an adjunctive therapy for AP through NLRP3 inflammasome inhibition, offering mechanistic insights and evidence-based support for its integration into integrative medicine strategies.

GRAPHICAL ABSTRACT
www.frontiersin.org

GRAPHICAL ABSTRACT | The functional role and mechanisms of TCM in the prevention and treatment of AP by inactivation of NLRP3 inflammasome. TCM, traditional Chinese medicine; OS, oxidative stress; TLR4, toll-like receptor 4; MAPK, mitogen-activated protein kinase; JNK, c-Jun N-terminal kinase; ERK, extracellular signal-regulated kinase; Nrf2, nuclear factor-erythroid factor 2-related factor 2; ARE, antioxidant response element; Keap1, Kelch-like ECH-associated protein 1; AP, acute pancreatitis.

1 Introduction

Acute pancreatitis (AP) is a life-threatening inflammatory disorder characterized by premature activation of pancreatic enzymes, leading to autodigestion, edema, hemorrhage, and necrosis of pancreatic tissues (Mederos et al., 2021). Epidemiological data indicated that approximately 20% of patients with AP developed severe AP, which was characterized by dysregulated cytokine storms that exacerbate pancreatic damage and precipitate multi-organ failure (e.g., pulmonary insufficiency, intestinal barrier dysfunction) (Garg and Singh, 2019). Global health statistics revealed that the global incidence and death of AP increased by 2.75 million and 122,416 in 2021 (Iannuzzi et al., 2022), which undoubtedly imposed substantial socioeconomic burdens on patients, families, and healthcare systems. Current management paradigms remain predominantly supportive, prioritizing aggressive fluid resuscitation, opioid-sparing analgesia, and enteral nutrition to mitigate systemic complications (Petrov and Yadav, 2019). Invasive procedures were reserved exclusively for managing refractory complications, such as infected necrotizing pancreatitis or persistent pseudocyst-related symptoms (Gardner et al., 2020). These clinical challenges underscored the urgent need to unravel AP pathogenesis and developed effective preventive and therapeutic strategies.

A critical driver of AP pathogenesis was the dysregulated activation of the nucleotide oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a cytosolic multiprotein complex that orchestrates caspase-1-mediated maturation of pro-inflammatory cytokines interleukin-1β (IL-1β) and IL-18, as well as gasdermin D (GSDMD)-dependent pyroptotic cell death (Ferrero-Andrés et al., 2020). NLRP3 inflammasome was a group of multi-protein complexes that participate in innate immunity processes through the activation of pro-inflammatory caspases (Martinon et al., 2002). In AP, damage-associated molecular patterns (DAMPs) released from necrotic acinar cells, such as mitochondrial DNA, extracellular ATP, and reactive oxygen species (ROS), which activated NLRP3 inflammasome and resulted in perpetuating a vicious cycle of inflammation, pancreatic necrosis, and systemic inflammatory response syndrome (Papantoniou et al., 2024). This inflammatory cascade was further amplified by infiltrating immune cells, including macrophages and neutrophils, which secreted additional inflammatory cytokines and chemokines, exacerbating tissue injury (Wan et al., 2020). Numerous studies have proved that activation of the NLRP3 inflammasome was associated with the severity of AP (Jia et al., 2020; Sendler et al., 2020). Preclinical studies have demonstrated that targeted inactivation of NLRP3 inflammasome [such as indomethacin (Lu G. et al., 2017), INT-777 (Li B. et al., 2018), apocynin (Jin et al., 2019), T-614 (Hou et al., 2019), and MCC950 (Sendler et al., 2020)] alleviated AP progression and associated organ injury by suppressing inflammation and pancreatic acinar cell apoptosis. Meanwhile, the inflammatory cytokines IL-18 and IL-1β may serve as the markers of the severity of AP patients (Janiak et al., 2015). Current therapeutic strategies, however, remain largely palliative, underscoring the urgent need for mechanistically targeted interventions to disrupt NLRP3-driven AP progression.

Traditional Chinese Medicine (TCM) is one of the ancient and most accepted alternative medicinal systems in the world for the treatment of health ailments, especially when Western medicine is not very effective (Luo et al., 2019). For hundreds of years, medicinal herbs have been used with apparent safety and efficacy for alleviating and treating AP in China. TCM, with its multi-component, multi-target, and multi-pathway pharmacological profiles, has emerged as a promising approach to modulating NLRP3 inflammasome activity in inflammation-associated diseases (Xue et al., 2023). Unlike synthetic inhibitors such as MCC950, a potent NLRP3 antagonist with clinical limitations due to off-target effects and pharmacokinetic challenges, TCM exerts pleiotropic pharmacological effects with low toxicity, including anti-inflammatory, antioxidant, antitumor, and immunomodulatory properties (Li and Zhang, 2013). For instance, Chaihuang Qingfu pill prevented severe AP-induced lung injury by inhibiting NLRP3-mediated macrophage pyroptosis (Xiao et al., 2025). Chlorogenic acid attenuated the development of severe AP by inhibiting NLRP3 inflammation activation and activating the Nrf2/HO-1 pathway (Ye et al., 2025). Psidium guajava flavonoids exerted a protective role in severe AP by inactivation of NLRP3 inflammasome (Zhang G. et al., 2021). Of note, clinical studies have also confirmed that TCM formulas were given to slow down the progression of AP (Zhang et al., 2008), as well as improve immune function (Jiang et al., 2016) and gastrointestinal function (Miao et al., 2018). Moreover, combined treatment of TCM and Western medicine contributed to reducing inflammation and improving immune dysfunction compared with Western medicine alone for patients with AP (Liu et al., 2011; Chen et al., 2021). Deng et al. (2024) showed that integrated TCM and Western medicine treatment reduced the risks of mortality and organ failure and achieved better economic effectiveness in patients with AP than Western medicine alone treatment. These results indicated that TCM prescriptions, monomers, and extracts possessed an inhibitory effect on NLRP3 inflammation, which could benefit the treatment of AP. However, there is still a lack of comprehensive review on TCM regulation of NLRP3 inflammasome-associated pathways in the treatment of AP.

Herein, we discussed the NLRP3 and its functional role in the development and progression of AP. Moreover, we summarized the therapeutic effect of TCM (prescriptions, extracts, and monomer compounds) on AP by targeting NLRP3 inflammasome-associated pathways. Furthermore, we analyzed the efficacy and safety of TCM for the treatment of AP in clinical trials, and discussed their challenges and future development directions.

2 Research methodology

This review article was conducted using electronic databases such as PubMed, Google Scholar, Springer Link, Science Direct, Cochrane Library, Embase, Web of Science, and Scopus. All published data till the year 2025 have been taken into consideration. The following search keywords were used in the search of materials for this study: “acute pancreatitis”, “NLRP3 inflammasome”, “inflammasome”, “medicinal plants”, “TCM prescription/decoction/formula”, “herbal extract”, “TCM extract”, “bioactive compounds”, “active ingredients”, “polyphenols”, “flavonoids”, “alkaloids”, “terpenes”, “anthraquinones”, “shikonin”, “polysaccharide”, “biological activity”, “pharmacological activities”, and other similar keywords in combination with words such as traditional Chinese medicine, Clinical trials, botanical description, toxicity, human health, and nutritional composition. All articles addressing these principal keywords were considered when available in the English language, and in peer-reviewed journals, whether published as review or research articles. Papers were reviewed in their entirety if their abstract mentioned that the article presented any potential relevance to the inclusion criteria. Articles were excluded based on title, abstract, or full text because of their lack of pertinence to the issue concerned. Articles were excluded if they were letters, comments, or not available for access to full article.

3 Overview of NLRP3 inflammasome

Innate immunity serves as the host’s primary defense barrier, wherein pattern recognition receptors (PRRs) on immune cells detect pathogen-associated molecular patterns (PAMPs), such as viral nucleic acids, bacterial lipopolysaccharides, and flagellin, or endogenous DAMPs released from damaged or dying cells. This recognition initiates innate immune responses and activates downstream inflammatory pathways to eliminate microbial infections and promote tissue repair (Kelley et al., 2019; Fu and Wu, 2023). In 2002, a novel class of PRR termed the inflammasome was identified as a critical signal transduction platform in innate immunity (Martinon et al., 2002). Certain members of the NOD-like receptor (NLR) family assemble into multiprotein complexes, forming a subset of inflammasomes (Toldo et al., 2022). To date, at least 22 inflammasome subtypes have been characterized, with the NLRP3 inflammasome being the most extensively studied (Zhuang et al., 2021). Under physiological conditions, NLRP3 inflammasome activation is essential for host defense against pathogens and homeostatic maintenance. However, its dysregulated activation drives excessive inflammatory responses and host tissue damage, contributing to autoimmune disorders such as AP (Papantoniou et al., 2024).

3.1 Structure of NLRP3 inflammasome

The NLRP3 inflammasome is composed of a sensor (NLRP3), an adaptor (ASC; also known as PYCARD), and an effector (Caspase-1) (Huang Y. et al., 2021). NLRP3, a trimeric protein, encompasses three functional domains: (1) the C-terminal leucine-rich repeat (LRR), which is crucial for ligand sensing; (2) the central nucleotide-binding and oligomerization (NACHT) domain, and (3) the amino-terminal pyrin domain (PYD) that mediates protein-protein interactions (Unterberger et al., 2021). Upon stimulation by PAMPs or DAMPs, NLRP3 oligomerizes homotypically through its NACHT domain. The oligomerized NLRP3 then recruits the apoptosis-associated speck-like protein containing a CARD (ASC) via PYD-PYD interactions. Subsequently, ASC facilitates the assembly of the NLRP3-ASC-pro-caspase-1 complex (the canonical NLRP3 inflammasome) by binding pro-caspase-1 through CARD-CARD interactions. Activation of the NLRP3 inflammasome triggers autoproteolytic cleavage of pro-caspase-1 into enzymatically active caspase-1, which catalyzes the maturation of pro-inflammatory cytokines IL-1β and IL-18 (Lechtenberg et al., 2014). These cytokines, in turn, amplify immune responses through downstream signaling cascades. Concurrently, active caspase-1 cleaves GSDMD to generate N-terminal fragments that form plasma membrane pores, inducing a lytic programmed cell death pathway associated with inflammatory cytokine release (Li L. et al., 2024).

3.2 Mechanism of NLRP3 inflammasome activation

Currently, NLRP3 inflammasome activation pathways are broadly categorized into canonical, noncanonical, and alternative pathways (Figure 1), with classification criteria predicated on stimulus specificity (e.g., PAMP vs. damage-associated signals) and cell type (e.g., immune vs. non-immune cell types) (Seoane et al., 2020). The canonical NLRP3 inflammasome activation requires two sequential priming and activation steps. The priming phase is initiated when DAMPs or PAMPs engage receptors such as toll-like receptors (TLRs) or cytokine receptors, triggering the NF-κB pathway to increase mRNA levels of NLRP3, pro-IL-1β, and pro-IL-18 (Zahid et al., 2019). Beyond transcriptional priming, this phase also orchestrates NLRP3 post-translational modifications (PTMs), including phosphorylation, ubiquitination, and SUMOylation, which play an important role in NLRP3 activation (Paik et al., 2021; Xu et al., 2022). The activation phase, the second step in the canonical pathway, is driven by diverse stimuli such as K+ efflux, ROS overproduction, lysosomal rupture, mitochondrial DNA (mtDNA) leakage, and organelle dysfunction (e.g., mitochondria, Golgi apparatus, endoplasmic reticulum) (Akbal et al., 2022). These events promoted NLRP3 inflammasome activation. In the noncanonical pathway, NLRP3 inflammasome activation is induced by lipopolysaccharide (LPS) internalization into the cytosol by transfection or infection (Santos et al., 2018), which can be recognized by caspase-11 (the mouse homolog of human caspase-4/5). Then, activated caspase 4/5/11 causes pyroptosis by cleaving GSDMD, and also triggers the assembly of NLRP3 inflammasome. Cross-talk between non-canonical and canonical inflammasome activation pathways, such as the activation of pannexin-1 by caspase-11 and subsequent release of ATP and activation of P2X7R to induce K+ efflux and thus canonical NLRP3 assembly, has also been suggested (Downs et al., 2020). Distinct from the above pathways, the alternative pathway of NLRP3 inflammasome activation bypasses pyroptosis, ASC polymerization, and K+ efflux, which has been exclusively characterized in primary human and porcine monocytes. Mechanistically, LPS directly activated NLRP3 inflammasome through the TLR4-TRIF-MyD88-RIPK1-FADD-CASP8 axis without eliciting pyroptotic cell death (He et al., 2016).

Figure 1
www.frontiersin.org

Figure 1. Mechanisms of NLRP3 inflammasome activation pathways. PAMPs, pathogen-associated molecular patterns; DAMPs, damage-associated molecular patterns; NF-κB, nuclear factor kappa-B; NLRP3, NOD-like receptor family pyrin domain-containing 3; ASC, apoptosis-associated speck-like protein containing a caspase recruitment domain; ROS, reactive oxygen species; mtDNA, mitochondrial DNA; IL, interleukin; GSDMD, gasdermin D; LPS, lipopolysaccharide; TRIF, TIR-domain-containing adapter-inducing interferon-β; RIPK1, receptor-interacting protein kinase 1; FADD, FAS-associated death domain; CASP8, caspase-8.

4 NLRP3 inflammasome in the pathogenesis of AP

AP is an inflammatory disorder characterized by excessive activation of pancreatic enzymes due to diverse etiological factors, leading to autodigestion, edema, hemorrhage, and necrosis of pancreatic parenchyma and adjacent tissues. A clinical study has confirmed elevated expression of NLRP3 inflammasome components in serum samples from pancreatitis patients compared to healthy controls, with NLRP3 activation correlating positively with disease severity (Algaba-Chueca et al., 2017). Sendler et al. (2020) reported that increased levels of proinflammatory cytokines (IL-1β and IL-18) and ASC were detected in serum samples from patients with severe AP. Hoque et al. (2011) further identified that NLRP3 inflammasome activation was a driver of disease progression through amplification of initial inflammatory cascades. Functionally, NLRP3 inflammasome were commonly activated by DAMPs (e.g., HMGB1 and HSP70) or PAMPs stimulation, which further promoted pancreatic inflammation and tissue injury, eventually advancing AP (Hoque et al., 2011). Meanwhile, LPS-stimulated-caspase-4/5/11 further triggers GSDMD driven pancreatic cell death and tissue injury, leading to the course of the pathogenesis of AP (Yi, 2020). In addition, ROS-mediated TXNIP and the activation of P2X7 trigger the NLRP3 inflammasome (Zhang et al., 2017). In mouse models of AP, NLRP3 deficiency ameliorated pancreatic inflammation and associated complications by reducing neutrophil infiltration (Fu et al., 2018). Another study showed that NLRP3 inflammasome activation promoted lung dysfunction by triggering alveolar macrophage pyroptosis in pancreatitis progression (Wu et al., 2020). In a recent study, pharmacological inhibition of NLRP3 inflammasome by MCC950 improved pathological damage and reduced inflammatory response in experimental pancreatitis (Shen et al., 2022). Mechanistically, the inflammation mediated by the Caspase-1/NLRP3, TLR4/NLRP3, MAPK/NF-κB/NLRP3, and Nrf2/HO-1/NLRP3 pathways have been implicated in the pathogenesis and progression of AP (Papantoniou et al., 2024), highlighting their potential as therapeutic targets. Collectively, these findings indicated that inhibition of NLRP3 inflammation may be a promising therapeutic strategy for AP management.

5 TCM used to relieve AP by targeting NLRP3

In Chinese medicine (CM), AP is classified under abdominal pain (Futong), spleen-heart pain (Pi Xintong), and pancreatic inflammation (Yi Dan) (Li et al., 2019a). Etiological factors in CM theory include gallstone obstruction, dietary irregularities (e.g., excessive greasy food), emotional disturbances (Gan Yu), trauma, and invasion of exogenous pathogens (Liu Yin) (Wu, 2002), all of which may exacerbate the progression of AP. Pathophysiologically, it manifests as root deficiency (Ben Xu) with branch excess (Biao Shi), predominantly presenting as interior excess-heat syndrome (Li Shi Re Zheng) (Mao et al., 2003). CM therapeutic strategies emphasize heat-clearing and bowel-unblocking methods (Qing Re Tong Fu Fa), integrating root-cause treatment (Zhi Ben) with symptomatic management (Zhi Biao) (Li et al., 2019a). Emerging pharmacological evidence has demonstrated that TCM prescription, extract, and active ingredient mitigated AP progression by suppressing NLRP3 inflammasome activation and downstream pyroptosis pathways (Jiang et al., 2021; Zeng et al., 2024; An et al., 2025). Moreover, the functional role of TCM in AP by regulating NLRP3 inflammation is summarized in Table 1 and 2.

Table 1
www.frontiersin.org

Table 1. Summary of TCM prescriptions and extracts in the prevention and treatment of AP by modulating inflammation in the last 5 years.

Table 2
www.frontiersin.org

Table 2. Summary of active components of TCM in the prevention and treatment of AP in the last 5 years.

5.1 TCM prescriptions for AP treatment

TCM prescriptions have been utilized in China for preventing and managing AP for a long time. Preclinical studies have identified many TCM formulations with therapeutic efficacy against AP, including Chaiqin chengqi decoction, Dahuangfuzi decoction, Chengqi-series decoction, Qingyi decoction, Qingxiajieyi formula, Dachengqi decoction, Dachaihu decoction, and Chaiqinchengqi decoction (Yang et al., 2021; Lin et al., 2023; Han et al., 2024; Wen et al., 2024). Moreover, previous studies have demonstrated that AP-associated multi-organ dysfunction (e.g., lung, kidney, liver) can be improved by TCM prescription treatment, such as Dachengqi decoction (Liu et al., 2023), Yinchenhao decoction (Zhao et al., 2024), and Chaiqin Chengqi decoction (Yang X. et al., 2020). Functionally, the main compounds (emodin, rhein, baicalin, and chrysin) from Chaiqin chengqi decoction diminished pancreatic acinar cell necrosis and systemic inflammation by inhibiting the TLR4/NLRP3 pathway (Wen et al., 2020). Qingyi decoction and its active ingredients (e.g., Wogonoside) alleviated pancreatic injury and systemic inflammation in AP by inactivation of the NF-κB/NLRP3/Caspase-1 pathway (An et al., 2025). Another study by Zhang R. et al. (2024) reported that Rhizoma Alismatis decoction alleviated AP by restoration of autophagy flux and mitochondrial homeostasis, leading to downregulation of NLRP3 and IL-1β in the pancreas. Other studies have proved that Chaiqin Chengqi decoction (Cao et al., 2024), Chaihuang Qingfu pill (Xiao et al., 2025), and Qingjie Huagong decoction (Feng et al., 2024) inhibited NLRP3 inflammasome and GSDMD activation-mediated pyroptosis and systemic inflammation in AP models. The above preclinical studies have confirmed the efficacy of TCM prescriptions in treating AP, but are hindered by methodological limitations (e.g., small sample sizes, non-standardized animal models, and lack of randomized controls) and insufficient mechanistic insights (e.g., unclear active ingredients, incomplete safety evaluations, and absence of long-term follow-up data), thus impeding translational validation and clinical implementation.

5.2 Herbal extracts in the treatment of AP

In addition to the aforementioned TCM formulas, herbal extracts have shown therapeutic efficacy against AP. For example, free total rhubarb anthraquinones ameliorated intestinal and pancreatic damage in AP rats while reducing inflammation and pyroptosis by inactivation of the NLRP3/caspase-1/GSDMD pathway (Zeng et al., 2024). Meanwhile, rhubarb anthraquinones treatment enhanced intestinal immunity by modulating Treg/Th17 balance (Chen et al., 2016). Xiong et al. (2019) showed that Lycium barbarum polysaccharides exhibited anti-inflammatory and antioxidant effects in cerulein-induced AP mice, as evidenced by reduced levels of serum amylase, TNF-α, and IL-1β. Another study reported that P. guajava flavonoids reduced pancreatic inflammation and fibrosis by inhibition of NLRP3 inflammasome activation in AP (Zhang G. et al., 2021). Moreover, Lonicera japonica flower extracts (Ruan et al., 2019) and Salvia miltiorrhiza extracts (Yang et al., 2019) ameliorated AP progression by suppressing the ROS-NLRP3 inflammasome axis and reducing the expression of IL-1β and IL-18. Collectively, these findings highlight the potential of herbal extracts to mitigate AP pathogenesis by targeting the NLRP3 inflammasome and modulating inflammatory cascades.

5.3 Monomer components isolated from TCM for the treatment of AP

A wide range of bioactive compounds, including polyphenols, flavonoids, alkaloids, and terpenes, have been isolated and purified from TCM. Pharmacological studies have demonstrated that these TCM-derived compounds exhibited therapeutic potential against AP by suppressing inflammatory response (Lu et al., 2021). The functional roles of these bioactive components in the treatment of AP by targeting NLRP3 are summarized in Table 2.

5.3.1 Polyphenols

Polyphenols, recognized for their anti-inflammatory and antioxidant properties, have shown efficacy in mitigating chronic diseases due to their versatile biological activities (e.g., anti-inflammatory and anti-oxidation) (Vivarelli et al., 2023), which contributed to the treatment of AP. Meanwhile, targeting the inflammasome pathway by polyphenols may be an effective therapeutic strategy for AP (Nani and Tehami, 2023). Resveratrol, a natural polyphenolic compound, has been proven to improve the pathophysiology of AP by reducing inflammation, cell apoptosis, pancreatic damage, blocking calcium overload, and improving microcirculation (Agah et al., 2021; Cai et al., 2025). Recently, resveratrol (Wu S. K. et al., 2024) and epigallocatechin-3-gallate (Luo et al., 2021) exhibited protective effects on severe AP by inactivation of NLRP3 inflammasome. Similarly, paeonol ameliorated AP by promoting M2 macrophage polarization through inactivation of NLRP3 inflammasome (Yuan et al., 2022). Another study found that rosmarinic acid has been shown to reduce inflammation by inhibition of the NF-κB pathway in the murine model of AP (Fan et al., 2015). In addition, scopoletin attenuated AP-induced organ injury (lung and intestine) by blocking the TLR4/NF-κB/NLRP3 pathway (Leema and Tamizhselvi, 2018). Further study highlights the protective effects of urolithin A against AP through suppression of apoptosis and mitochondrial dysfunction (Yang Y. et al., 2023).

5.3.2 Flavonoids

Flavonoids have attracted increasing attention as promising candidates for the modulation of inflammation due to their dual anti-inflammatory and immunomodulatory properties (Al-Khayri et al., 2022). Intriguingly, flavonoids exerted protective effects against AP by targeting key pathogenic processes, including NLRP3 inflammasome activation, oxidative stress, and cytokine storm. For example, baicalein (Wang et al., 2021), rutin (Aruna et al., 2014), naringenin (Li Y. et al., 2018), and luteolin (Rajapriya and Geetha, 2021) have been shown to inhibit the assembly of NLRP3 inflammasome complexes, thereby reducing caspase-1 activation and subsequent IL-1β maturation in AP models. Other studies have proved that proanthocyanidins (Sheng et al., 2023) and baicalein (Wang X. et al., 2025) ameliorated AP by promoting macrophage M2 polarization through suppressing NLRP3 inflammasome activation. Moreover, naringenin improved AP-associated intestinal injury by inhibiting NLRP3 inflammasome activation (Yan et al., 2023). Other flavonoids such as apigenin (Charalabopoulos et al., 2019), biochanin A (Pan et al., 2023), and luteolin (Xiong et al., 2017) alleviated AP by inhibition of the TLR4/NF-κB pathway-mediated inflammation. Zhou et al. (2024) showed that administration of tectoridin inhibited pancreatic injury in AP by triggering macrophage M2 polarization. Another study showed that isorhamnetin alleviated mitochondrial injury and inhibited ROS generation in severe AP (Li X. et al., 2024).

5.3.3 Alkaloids

Alkaloids have emerged as pivotal therapeutic agents in modern medicine due to their broad-spectrum anti-inflammatory and antibacterial properties (Bai et al., 2021). Berberine (BBR), a natural alkaloid extracted from medicinal plants, exhibited multifunctional pharmacological activities (Zhang Y. et al., 2021), including anti-inflammatory, anti-tumor, lipid-lowering, hypoglycemic, and anti-osteoarthritic activities in preclinical studies. Numerous studies have demonstrated that BBR attenuated AP by inhibition of AMPK-mediated inflammation and M2 macrophage polarization (Bansod et al., 2020; Bansod et al., 2025). Meanwhile, BBR treatment improved histological damage to the pancreas, lungs, and intestinal by blocking the NF-κB pathway (Liang et al., 2014; Choi et al., 2017). Li Z. et al. (2020) reported that anisodamine pretreatment mitigated lipopolysaccharide-induced apoptosis and inflammation of pancreatic acinar cells by inactivating the NLRP3 inflammasome and blocking the NF-κB pathway. Moreover, other alkaloids [e.g., castanospermine (Hong et al., 2016), ellipticine (Li X. et al., 2020), rutaecarpine (Huang H. et al., 2021), colchicine (Zhang et al., 2022), matrine (Jin et al., 2023), oxymatrine (Lu M. et al., 2017), nicotine (Zheng et al., 2015)] have been shown to combat AP and AP-induced organ injury by inhibiting the inflammatory response. A recent study showed that galantamine exhibited an anti-inflammatory effect against AP (Thompson et al., 2023), which was an FDA-approved acetylcholinesterase inhibitor for Alzheimer’s disease in clinical trials.

5.3.4 Terpenes

Terpenes represent a structurally diverse class of natural compounds with potent anti-inflammatory and immunomodulatory activities, holding promise for treating inflammation-associated diseases (Chang and Xiong, 2020). Currently, many terpenes (e.g., micheliolide (Wu C. Y. et al., 2024), artesunate (Liu et al., 2025), nimbolide (Bansod and Godugu, 2021), betulinic acid (Zhou et al., 2021), triptolide (Yang et al., 2022), irisin (Han et al., 2023), etc.) attenuated AP progression by reducing inflammatory response and inhibiting neutrophil extracellular traps formation. Mechanistically, treatment with ganoderic acid A (Zhang et al., 2025), DGA (Yue et al., 2024), and pachymic acid (Li et al., 2022) improved intestinal dysfunction, macrophage pyroptosis, and pancreatic fibrosis in AP by repressing NLRP3 inflammasome activation. Moreover, other terpenes were found to effectively inhibit necroptosis/apoptosis/ferroptosis and conferr protection against AP, such as celastrol (Liang et al., 2023), crocetin (Zhu and He, 2022), and glycyrrhizin (Cui et al., 2024). Of note, both terpenes [limonin (Xia et al., 2021) and astaxanthin (Kwak et al., 2021)] exert pancreatic protection by suppressing JAK2/STAT3 hyperactivation, thereby reducing pro-inflammatory cytokine production.

5.3.5 Others

Beyond the aforementioned phytochemicals, additional medicinal plant-derived compounds exhibit targeted therapeutic potential against AP. Cordycepin, a nucleoside derivative extracted from Cordyceps militaris, inhibited pancreatic inflammation and injury by blocking the NF-κB/NLRP3 inflammasome pathway (Yang J. et al., 2020). Anthraquinones (e.g., emodin) exerted pleiotropic effects on inflammation and pancreatic tissue repair via the inactivation of NLRP3 inflammasome (Zhang et al., 2019). Shikonin (Xiong et al., 2013) and Astragalus polysaccharides (Wang Q. et al., 2025) mitigated AP progression by inhibiting NF-κB pathway-mediated inflammation. Moreover, the protective effect of notoginsenoside R1 (He et al., 2022) and protocatechuic acid (Abdelmageed et al., 2021) on AP-induced lung injury by blocking the HMGB1/TLR4/NF-κB pathway. Collectively, these findings underscored that bioactive compounds derived from TCM counteract AP pathogenesis by targeting inflammatory pathways, including NF-κB, MAPK, and NLRP3 inflammasome.

6 Clinical study of TCM for the prevention and treatment of AP

Preclinical studies have confirmed that TCM possesses significant therapeutic potential against AP. Notably, randomized controlled trials have revealed that TCM interventions reduced mortality rates, shortened hospitalization duration, and mitigated postoperative complications in AP patients compared to conventional therapies (Qiong et al., 2005). For instance, among the included 248 patients with AP (124 patients in each group), Chaiqinchengqi decoction treatment reduced the duration of 28-day respiratory failure (median: 1.0 days, 95% confidence interval: −2.0 to 0.0) and improve 6-month clinical outcomes in AP patients compared with the placebo (Deng et al., 2025). Similarly, Dachengqi decoction, as adjunctive therapy, reduced multiple organ dysfunction syndrome incidence by 40% and pancreatic infection risk by 5% in severe AP patients (Chen et al., 2010). Tongfu powder treatment alleviated gastrointestinal dysfunction in AP cohorts (Miao et al., 2018). Moreover, integrative approaches combining TCM with Western medicine exhibited synergistic benefits, including decreased organ failure risk (4.1% vs. 5.9%), reduced hospitalization costs ($2,157/patient vs. $2,895/patient) and overall mortality rate (1.7% vs. 3.4%) (Deng et al., 2024). Notably, Guo qing yi tang decoction treatment enhanced intestinal barrier integrity (serum DAO and MFG-8↓, p < 0.05) and reduced inflammatory cytokines (TNF-α, IL-6, and IL-8↓, p < 0.05), APACHE II scores (7.84, p < 0.001), and hospital stay after 1 week in a total of 38 AP patients compared with the control group (cluster therapy alone, 70 patients) (Chen et al., 2021). These findings contrast with the limitations of conventional Western medications, including high costs and adverse effects. Herein, ongoing national clinical trials evaluating TCM safety and efficacy in AP are summarized in Table 3. Meanwhile, some preclinical and clinical studies have proved that no significant adverse effects were observed during the TCM treatment period (Wan et al., 2011; Lu et al., 2014). However, TCM formulas may cause mild gastrointestinal discomfort, such as nausea or diarrhea (Tan et al., 2020). Other TCM herbs may interact with antiplatelet or anticoagulant drugs, increasing the risk of bleeding (Li et al., 2019b).

Table 3
www.frontiersin.org

Table 3. Clinical trials of TCM in AP.

Despite these promising results, translating preclinical TCM research into clinical practice faces multifaceted challenges. A primary hurdle involves securing regulatory approval for commercialization due to resource limitations and difficulties in semi-synthetic production or medicinal plant engineering of bioactive compounds. This dependency on natural resources raises ethical concerns, particularly given reports of over 20,000 medicinal plant species at risk of extinction. Additionally, critical gaps remain in mechanistic understanding and robust clinical data. Other challenges include poor solubility/absorption profiles, intellectual property issues, and limitations in drug-likeness and purity of TCM-derived compounds. Of note, integrating TCM into standard care was constrained by methodological limitations (e.g., small sample sizes, non-RCT designs), mechanistic ambiguity, and quality control issues, necessitating large-scale randomized trials, ingredient standardization, and safety evaluations to facilitate evidence-based implementation.

7 Conclusion and perspectives

Recent advances in elucidating the pathogenesis of AP have coincided with the growing recognition of TCM as a promising therapeutic strategy. Accumulating evidence highlights TCM’s remarkable therapeutic efficacy in AP management through multi-target modulation, particularly its capacity to regulate NLRP3 inflammasome activation and downstream inflammatory cascades. This review systematically summarized current knowledge on TCM-derived compounds targeting NLRP3-mediated pathways in AP, while critically addressing persisting challenges in translational applications. Key limitations hindering clinical translation include: (1) Formula standardization gaps: Current research predominantly focuses on empirical or self-formulated TCM prescriptions, with insufficient validation of classical prescriptions through randomized controlled trials; (2) Bioavailability challenges: Many active ingredients of TCM exhibited suboptimal pharmacokinetic profiles due to structural instability, rapid oxidation, and poor membrane permeability, advanced delivery systems (e.g., lipid-based encapsulation, nanoparticle carriers) can be considered; (3) Safety and metabolism uncertainties: Comprehensive characterization of TCM pharmacokinetics, tissue distribution, and long-term toxicity in AP-specific contexts remains imperative; (4) Mechanistic complexity: The polypharmacological nature of TCM necessitates integrative multi-omics approaches, including metabolomics, network pharmacology, proteomics, immunomics, and gut metagenomics, to decode its regulatory effects on NLRP3 inflammasome; (5) Standardized protocols in TCM research are essential to ensure reproducibility, validate therapeutic efficacy, and facilitate regulatory acceptance for broader clinical implementation.

In summary, NLRP3 inflammasome inhibition represented a strategic diagnostic and therapeutic nexus in AP. TCM served as a reservoir of NLRP3 inflammasome-modulating agents with the potential to restrict AP progression. Of note, this review provided a framework for evidence-based optimization of TCM to combat AP, advocating for the integration of systems biology and advanced drug delivery platforms to bridge traditional knowledge with modern precision medicine paradigms.

Author contributions

XH: Writing – original draft, Data curation, Investigation, Writing – review and editing, Formal Analysis, Visualization. JX: Investigation, Data curation, Writing – review and editing. QC: Data curation, Writing – review and editing, Formal Analysis. ZH: Investigation, Writing – review and editing. JL: Data curation, Writing – review and editing. YR: Investigation, Supervision, Writing – review and editing, Resources.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abdelmageed, M. E., Nader, M. A., and Zaghloul, M. S. (2021). Targeting HMGB1/TLR4/NF-κB signaling pathway by protocatechuic acid protects against l-arginine induced acute pancreatitis and multiple organs injury in rats. Eur. J. Pharmacol. 906, 174279. doi:10.1016/j.ejphar.2021.174279

PubMed Abstract | CrossRef Full Text | Google Scholar

Agah, S., Akbari, A., Sadeghi, E., Morvaridzadeh, M., Basharat, Z., Palmowski, A., et al. (2021). Resveratrol supplementation and acute pancreatitis: a comprehensive review. Biomed. Pharmacother. 137, 111268. doi:10.1016/j.biopha.2021.111268

PubMed Abstract | CrossRef Full Text | Google Scholar

Akbal, A., Dernst, A., Lovotti, M., Mangan, M. S. J., McManus, R. M., and Latz, E. (2022). How location and cellular signaling combine to activate the NLRP3 inflammasome. Cell Mol. Immunol. 19 (11), 1201–1214. doi:10.1038/s41423-022-00922-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Algaba-Chueca, F., de-Madaria, E., Lozano-Ruiz, B., Martínez-Cardona, C., Quesada-Vázquez, N., Bachiller, V., et al. (2017). The expression and activation of the AIM2 inflammasome correlates with inflammation and disease severity in patients with acute pancreatitis. Pancreatology 17 (3), 364–371. doi:10.1016/j.pan.2017.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Al-Khayri, J. M., Sahana, G. R., Nagella, P., Joseph, B. V., Alessa, F. M., and Al-Mssallem, M. Q. (2022). Flavonoids as potential anti-inflammatory molecules: a review. Molecules 27 (9), 2901. doi:10.3390/molecules27092901

PubMed Abstract | CrossRef Full Text | Google Scholar

An, Y., Tu, Z., Wang, A., Gou, W., Yu, H., Wang, X., et al. (2025). Qingyi decoction and its active ingredients ameliorate acute pancreatitis by regulating acinar cells and macrophages via NF-κB/NLRP3/Caspase-1 pathways. Phytomedicine 139, 156424. doi:10.1016/j.phymed.2025.156424

PubMed Abstract | CrossRef Full Text | Google Scholar

Aruna, R., Geetha, A., and Suguna, P. (2014). Rutin modulates ASC expression in NLRP3 inflammasome: a study in alcohol and cerulein-induced rat model of pancreatitis. Mol. Cell Biochem. 396 (1-2), 269–280. doi:10.1007/s11010-014-2162-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Bai, R., Yao, C., Zhong, Z., Ge, J., Bai, Z., Ye, X., et al. (2021). Discovery of natural anti-inflammatory alkaloids: potential leads for the drug discovery for the treatment of inflammation. Eur. J. Med. Chem. 213, 113165. doi:10.1016/j.ejmech.2021.113165

PubMed Abstract | CrossRef Full Text | Google Scholar

Bansod, S., Chilvery, S., Saifi, M. A., Das, T. J., Tag, H., and Godugu, C. (2021). Borneol protects against cerulein-induced oxidative stress and inflammation in acute pancreatitis mice model. Environ. Toxicol. 36 (4), 530–539. doi:10.1002/tox.23058

PubMed Abstract | CrossRef Full Text | Google Scholar

Bansod, S., Doijad, N., and Godugu, C. (2020). Berberine attenuates severity of chronic pancreatitis and fibrosis via AMPK-Mediated inhibition of TGF-β1/Smad signaling and M2 polarization. Toxicol. Appl. Pharmacol. 403, 115162. doi:10.1016/j.taap.2020.115162

PubMed Abstract | CrossRef Full Text | Google Scholar

Bansod, S., and Godugu, C. (2021). Nimbolide ameliorates pancreatic inflammation and apoptosis by modulating NF-κB/SIRT1 and apoptosis signaling in acute pancreatitis model. Int. Immunopharmacol. 90, 107246. doi:10.1016/j.intimp.2020.107246

PubMed Abstract | CrossRef Full Text | Google Scholar

Bansod, S. P., Saifi, M. A., Chilvery, S., Doijad, N., and Godugu, C. (2025). Berberine attenuates cerulein-induced acute pancreatitis by modulating Nrf2/NOX2 signaling pathway via AMPK activation. Environ. Toxicol. 40 (5), 764–773. doi:10.1002/tox.24468

PubMed Abstract | CrossRef Full Text | Google Scholar

Cai, W., Li, Z., Wang, W., Liu, S., Li, Y., Sun, X., et al. (2025). Resveratrol in animal models of pancreatitis and pancreatic cancer: a systematic review with machine learning. Phytomedicine 139, 156538. doi:10.1016/j.phymed.2025.156538

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, F., Xiang, J., Wang, Y., Chen, X., Lu, X., Xu, X., et al. (2024). Chaiqin chengqi decoction alleviates acute pancreatitis by targeting gasdermin D-mediated pyroptosis. J. Ethnopharmacol. 318 (Pt A), 116920. doi:10.1016/j.jep.2023.116920

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, M. X., and Xiong, F. (2020). Astaxanthin and its effects in inflammatory responses and inflammation-associated diseases: recent advances and future directions. Molecules 25 (22), 5342. doi:10.3390/molecules25225342

PubMed Abstract | CrossRef Full Text | Google Scholar

Charalabopoulos, A., Davakis, S., Lambropoulou, M., Papalois, A., Simopoulos, C., and Tsaroucha, A. (2019). Apigenin exerts anti-inflammatory effects in an experimental model of acute pancreatitis by down-regulating TNF-α. Vivo 33 (4), 1133–1141. doi:10.21873/invivo.11583

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, H., Li, F., Jia, J. G., Diao, Y. P., Li, Z. X., and Sun, J. B. (2010). Effects of traditional Chinese medicine on intestinal mucosal permeability in early phase of severe acute pancreatitis. Chin. Med. J. Engl. 123 (12), 1537–1542. doi:10.3760/cma.j.issn.0366-6999.2010.12.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, H., Lu, X., Xu, B., Cheng, G., Li, Y., and Xie, D. (2024). Saikosaponin d protects pancreatic acinar cells against cerulein-induced pyroptosis through alleviating mitochondrial damage and inhibiting cGAS-STING pathway. J. Appl. Toxicol. 44 (7), 1005–1013. doi:10.1002/jat.4594

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J., Li, F., Luo, W. S., Zhu, M. F., Zhao, N. J., Zhang, Z. H., et al. (2025). Therapeutic potential of Da Cheng Qi Decoction and its ingredients in regulating ferroptosis via the NOX2-GPX4 signaling pathway to alleviate and predict severe acute pancreatitis. Cell Signal 131, 111733. doi:10.1016/j.cellsig.2025.111733

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, L., Fan, L., Tan, X. Y., Wang, L. L., Liu, Y. Q., and Xiong, Y. X. (2016). Effects of free anthraquinone of rhubarb on immune function of mesenteric lymph nodes in rats with severe acute pancreatitis. Shandong Med. J. 56 (24), 13–16. doi:10.3969/j.issn.1002-266X.2016.24.004

CrossRef Full Text | Google Scholar

Chen, Y. J., Chen, M. Z., Zhang, H. W., Wu, G. S., and Guo, S. R. (2021). Effect of guo qing yi tang combined with Western medicine cluster therapy on acute pancreatitis. Am. J. Emerg. Med. 50, 66–70. doi:10.1016/j.ajem.2021.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi, S. B., Bae, G. S., Jo, I. J., Song, H. J., and Park, S. J. (2017). Effects of berberine on acute necrotizing pancreatitis and associated lung injury. Pancreas 46 (8), 1046–1055. doi:10.1097/mpa.0000000000000877

PubMed Abstract | CrossRef Full Text | Google Scholar

Cui, Q., Wang, W., Shi, J., Lai, F., Luo, S., Du, Y., et al. (2024). Glycyrrhizin ameliorates cardiac injury in rats with severe acute pancreatitis by inhibiting ferroptosis via the Keap1/Nrf2/HO-1 pathway. Dig. Dis. Sci. 69 (7), 2477–2487. doi:10.1007/s10620-024-08398-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Deng, L., Chen, Z., Jin, T., Cai, F., He, Y., Shen, Y., et al. (2025). Traditional Chinese medicine chaiqinchengqi decoction for patients with acute pancreatitis: a randomized clinical trial. Phytomedicine 138, 156393. doi:10.1016/j.phymed.2025.156393

PubMed Abstract | CrossRef Full Text | Google Scholar

Deng, L., Chen, Z., Zhu, P., Hu, C., Jin, T., Wang, X., et al. (2024). Effects of integrated traditional Chinese and Western medicine for acute pancreatitis: a real-world study in a tertiary teaching hospital. J. Evid. Based Med. 17 (3), 575–587. doi:10.1111/jebm.12635

PubMed Abstract | CrossRef Full Text | Google Scholar

Downs, K. P., Nguyen, H., Dorfleutner, A., and Stehlik, C. (2020). An overview of the non-canonical inflammasome. Mol. Asp. Med. 76, 100924. doi:10.1016/j.mam.2020.100924

PubMed Abstract | CrossRef Full Text | Google Scholar

Fan, J., Duan, L., Wu, N., Xu, X., Xin, J., Jiang, S., et al. (2020). Baicalin ameliorates pancreatic fibrosis by inhibiting the activation of pancreatic stellate cells in mice with chronic pancreatitis. Front. Pharmacol. 11, 607133. doi:10.3389/fphar.2020.607133

PubMed Abstract | CrossRef Full Text | Google Scholar

Fan, Y. T., Yin, G. J., Xiao, W. Q., Qiu, L., Yu, G., Hu, Y. L., et al. (2015). Rosmarinic acid attenuates sodium taurocholate-induced acute pancreatitis in rats by inhibiting nuclear Factor-κB activation. Am. J. Chin. Med. 43 (6), 1117–1135. doi:10.1142/s0192415x15500640

PubMed Abstract | CrossRef Full Text | Google Scholar

Feng, M., Qin, B., Luo, F., Zhu, X., Liu, K., Li, K., et al. (2024). Qingjie huagong decoction inhibits pancreatic acinar cell pyroptosis by regulating circHipk3/miR-193a-5p/NLRP3 pathway. Phytomedicine 126, 155265. doi:10.1016/j.phymed.2023.155265

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferrero-Andrés, A., Panisello-Roselló, A., Roselló-Catafau, J., and Folch-Puy, E. (2020). NLRP3 inflammasome-mediated inflammation in acute pancreatitis. Int. J. Mol. Sci. 21 (15), 5386. doi:10.3390/ijms21155386

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, J., and Wu, H. (2023). Structural mechanisms of NLRP3 inflammasome assembly and activation. Annu. Rev. Immunol. 41, 301–316. doi:10.1146/annurev-immunol-081022-021207

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, Q., Zhai, Z., Wang, Y., Xu, L., Jia, P., Xia, P., et al. (2018). NLRP3 deficiency alleviates severe acute pancreatitis and pancreatitis-associated lung injury in a mouse model. Biomed. Res. Int. 2018, 1294951. doi:10.1155/2018/1294951

PubMed Abstract | CrossRef Full Text | Google Scholar

Gardner, T. B., Adler, D. G., Forsmark, C. E., Sauer, B. G., Taylor, J. R., and Whitcomb, D. C. (2020). ACG clinical guideline: chronic pancreatitis. Am. J. Gastroenterol. 115 (3), 322–339. doi:10.14309/ajg.0000000000000535

PubMed Abstract | CrossRef Full Text | Google Scholar

Garg, P. K., and Singh, V. P. (2019). Organ failure due to systemic injury in acute pancreatitis. Gastroenterology 156 (7), 2008–2023. doi:10.1053/j.gastro.2018.12.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, F., Ding, Z. F., Shi, X. L., Zhu, Q. T., Shen, Q. H., Xu, X. M., et al. (2023). Irisin inhibits neutrophil extracellular traps formation and protects against acute pancreatitis in mice. Redox Biol. 64, 102787. doi:10.1016/j.redox.2023.102787

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, X., Bao, J., Ni, J., Li, B., Song, P., Wan, R., et al. (2024). Qing xia jie yi formula granules alleviated acute pancreatitis through inhibition of M1 macrophage polarization by suppressing glycolysis. J. Ethnopharmacol. 325, 117750. doi:10.1016/j.jep.2024.117750

PubMed Abstract | CrossRef Full Text | Google Scholar

He, J., Liu, M. W., Wang, Z. Y., and Shi, R. J. (2022). Protective effects of the notoginsenoside R1 on acute lung injury by regulating the miR-128-2-5p/Tollip signaling pathway in rats with severe acute pancreatitis. Innate Immun. 28 (1), 19–36. doi:10.1177/17534259211068744

PubMed Abstract | CrossRef Full Text | Google Scholar

He, Y., Hara, H., and Núñez, G. (2016). Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem. Sci. 41 (12), 1012–1021. doi:10.1016/j.tibs.2016.09.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Hong, Y. P., Chen, C., Guo, W. Y., Zhao, L., Mei, F. C., Xiang, M. W., et al. (2016). Effects of castanospermine on inflammatory response in a rat model of experimental severe acute pancreatitis. Arch. Med. Res. 47 (6), 436–445. doi:10.1016/j.arcmed.2016.11.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoque, R., Sohail, M., Malik, A., Sarwar, S., Luo, Y., Shah, A., et al. (2011). TLR9 and the NLRP3 inflammasome link acinar cell death with inflammation in acute pancreatitis. Gastroenterology 141 (1), 358–369. doi:10.1053/j.gastro.2011.03.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Hou, C., Zhu, X., Shi, C., Peng, Y., Huang, D., Li, Q., et al. (2019). Iguratimod (T-614) attenuates severe acute pancreatitis by inhibiting the NLRP3 inflammasome and NF-κB pathway. Biomed. Pharmacother. 119, 109455. doi:10.1016/j.biopha.2019.109455

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, J., Zhang, Y. M., Miao, Y. F., Zhu, L., Yi, X. L., Chen, H., et al. (2020). Effects of yue-bi-tang on water metabolism in severe acute pancreatitis rats with acute lung-kidney injury. World J. Gastroenterol. 26 (43), 6810–6821. doi:10.3748/wjg.v26.i43.6810

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, Q., Yao, J., Wu, X., Li, J., Li, G., Tang, W., et al. (2022). Emodin attenuates severe acute pancreatitis-associated acute lung injury by suppressing pancreatic exosome-mediated alveolar macrophage activation. Acta Pharm. Sin. B 12 (10), 3986–4003. doi:10.1016/j.apsb.2021.10.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, H., Wang, M., Guo, Z., Wu, D., Wang, H., Jia, Y., et al. (2021a). Rutaecarpine alleviates acute pancreatitis in mice and AR42J cells by suppressing the MAPK and NF-κB signaling pathways via calcitonin gene-related peptide. Phytother. Res. 35 (11), 6472–6485. doi:10.1002/ptr.7301

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, Y., Xu, W., and Zhou, R. (2021b). NLRP3 inflammasome activation and cell death. Cell Mol. Immunol. 18 (9), 2114–2127. doi:10.1038/s41423-021-00740-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Iannuzzi, J. P., King, J. A., Leong, J. H., Quan, J., Windsor, J. W., Tanyingoh, D., et al. (2022). Global incidence of acute pancreatitis is increasing over time: a systematic review and meta-analysis. Gastroenterology 162 (1), 122–134. doi:10.1053/j.gastro.2021.09.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Janiak, A., Leśniowski, B., Jasińska, A., Pietruczuk, M., and Małecka-Panas, E. (2015). Interleukin 18 as an early marker or prognostic factor in acute pancreatitis. Prz. Gastroenterol. 10 (4), 203–207. doi:10.5114/pg.2015.50993

PubMed Abstract | CrossRef Full Text | Google Scholar

Jia, L., Chen, H., Yang, J., Fang, X., Niu, W., Zhang, M., et al. (2020). Combinatory antibiotic treatment protects against experimental acute pancreatitis by suppressing gut bacterial translocation to pancreas and inhibiting NLRP3 inflammasome pathway. Innate Immun. 26 (1), 48–61. doi:10.1177/1753425919881502

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, D. L., Yang, J., Jiang, S. Y., Yuan, F. L., Gu, Y. L., Li, J. P., et al. (2016). Modified Da Chengqi granules improvement in immune function in early severe acute pancreatitis patients. Genet. Mol. Res. 15 (2), 15028787. doi:10.4238/gmr.15028787

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, N., Li, Z., Luo, Y., Jiang, L., Zhang, G., Yang, Q., et al. (2021). Emodin ameliorates acute pancreatitis-induced lung injury by suppressing NLRP3 inflammasome-mediated neutrophil recruitment. Exp. Ther. Med. 22 (2), 857. doi:10.3892/etm.2021.10289

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, H., Zhao, K., Li, J., Xu, Z., Liao, S., and Sun, S. (2023). Matrine alleviates oxidative stress and ferroptosis in severe acute pancreatitis-induced acute lung injury by activating the UCP2/SIRT3/PGC1α pathway. Int. Immunopharmacol. 117, 109981. doi:10.1016/j.intimp.2023.109981

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, H. Z., Yang, X. J., Zhao, K. L., Mei, F. C., Zhou, Y., You, Y. D., et al. (2019). Apocynin alleviates lung injury by suppressing NLRP3 inflammasome activation and NF-κB signaling in acute pancreatitis. Int. Immunopharmacol. 75, 105821. doi:10.1016/j.intimp.2019.105821

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, H., Hu, Q., Yang, Y., Huang, G., Li, J., Zhao, X., et al. (2024). Urolithin a's role in alleviating severe acute pancreatitis via endoplasmic reticulum-mitochondrial calcium channel modulation. ACS Nano 18 (21), 13885–13898. doi:10.1021/acsnano.4c03044

PubMed Abstract | CrossRef Full Text | Google Scholar

Kelley, N., Jeltema, D., Duan, Y., and He, Y. (2019). The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int. J. Mol. Sci. 20 (13), 3328. doi:10.3390/ijms20133328

PubMed Abstract | CrossRef Full Text | Google Scholar

Kwak, M. S., Lim, J. W., and Kim, H. (2021). Astaxanthin inhibits Interleukin-6 expression in Cerulein/resistin-stimulated pancreatic acinar cells. Mediat. Inflamm. 2021, 5587297. doi:10.1155/2021/5587297

PubMed Abstract | CrossRef Full Text | Google Scholar

Lechtenberg, B. C., Mace, P. D., and Riedl, S. J. (2014). Structural mechanisms in NLR inflammasome signaling. Curr. Opin. Struct. Biol. 29, 17–25. doi:10.1016/j.sbi.2014.08.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Leema, G., and Tamizhselvi, R. (2018). Protective effect of scopoletin against cerulein-induced acute pancreatitis and associated lung injury in mice. Pancreas 47 (5), 577–585. doi:10.1097/mpa.0000000000001034

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, B., Yang, N., Li, C., Li, C., Gao, K., Xie, X., et al. (2018a). INT-777, a bile acid receptor agonist, extenuates pancreatic acinar cells necrosis in a mouse model of acute pancreatitis. Biochem. Biophys. Res. Commun. 503 (1), 38–44. doi:10.1016/j.bbrc.2018.05.120

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, F., Chen, M., Ji, J., Tang, R., Huang, J., Zhang, X., et al. (2022). Pachymic acid alleviates experimental pancreatic fibrosis through repressing NLRP3 inflammasome activation. Biosci. Biotechnol. Biochem. 86 (11), 1497–1505. doi:10.1093/bbb/zbac114

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Chen, J., and Tang, W. (2019a). The consensus of integrative diagnosis and treatment of acute pancreatitis-2017. J. Evid. Based Med. 12 (1), 76–88. doi:10.1111/jebm.12342

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Liang, Q., and Sun, G. (2019b). Interaction between traditional Chinese medicine and anticoagulant/antiplatelet drugs. Curr. Drug Metab. 20 (9), 701–713. doi:10.2174/1389200220666190827160212

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, L. R., Chen, L., and Sun, Z. J. (2024a). Igniting hope: harnessing NLRP3 inflammasome-GSDMD-mediated pyroptosis for cancer immunotherapy. Life Sci. 354, 122951. doi:10.1016/j.lfs.2024.122951

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, S., and Zhang, B. (2013). Traditional Chinese medicine network pharmacology: theory, methodology and application. Chin. J. Nat. Med. 11 (2), 110–120. doi:10.1016/s1875-5364(13)60037-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Wang, T., Zhou, Q., Li, F., Liu, T., Zhang, K., et al. (2024b). Isorhamnetin alleviates mitochondrial injury in severe acute pancreatitis via modulation of KDM5B/HtrA2 signaling pathway. Int. J. Mol. Sci. 25 (7), 3784. doi:10.3390/ijms25073784

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Yan, Z., Cao, X., Chen, X., Guan, Z., Tang, S., et al. (2025). Dachaihu decoction alleviates chronic pancreatitis by regulating MAPK signaling pathway: insights from network pharmacology and experimental validation. J. Ethnopharmacol. 337 (Pt 1), 118833. doi:10.1016/j.jep.2024.118833

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Ye, C., Mulati, M., Sun, L., and Qian, F. (2020a). Ellipticine blocks synergistic effects of IL-17A and TNF-α in epithelial cells and alleviates severe acute pancreatitis-associated acute lung injury. Biochem. Pharmacol. 177, 113992. doi:10.1016/j.bcp.2020.113992

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., Pan, Y., Gao, L., Zhang, J., Xie, X., Tong, Z., et al. (2018b). Naringenin protects against acute pancreatitis in two experimental models in mice by NLRP3 and Nrf2/HO-1 pathways. Mediat. Inflamm. 2018, 3232491. doi:10.1155/2018/3232491

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Z., Xu, C., Tao, Y., Liang, Y., Liang, Q., Li, J., et al. (2020b). Anisodamine alleviates lipopolysaccharide-induced pancreatic acinar cell injury through NLRP3 inflammasome and NF-κB signaling pathway. J. Recept Signal Transduct. Res. 40 (1), 58–66. doi:10.1080/10799893.2020.1713808

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, H. Y., Chen, T., Yan, H. T., Huang, Z., and Tang, L. J. (2014). Berberine ameliorates severe acute pancreatitis-induced intestinal barrier dysfunction via a myosin light chain phosphorylation-dependent pathway. Mol. Med. Rep. 9 (5), 1827–1833. doi:10.3892/mmr.2014.1996

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, Q. Q., Shi, Z. J., Yuan, T., Chen, S. Y., Li, Y. P., Zhang, H. R., et al. (2023). Celastrol inhibits necroptosis by attenuating the RIPK1/RIPK3/MLKL pathway and confers protection against acute pancreatitis in mice. Int. Immunopharmacol. 117, 109974. doi:10.1016/j.intimp.2023.109974

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, J., Han, C., Dai, N., Bi, S., Du, D., and Xia, Q. (2023). Effectiveness of Chengqi-series decoctions in treating severe acute pancreatitis: a systematic review and meta-analysis. Phytomedicine 113, 154727. doi:10.1016/j.phymed.2023.154727

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, D., Liu, C., Deng, F., Ouyang, F., Qin, R., Zhai, Z., et al. (2025). Artesunate protects against a mouse model of cerulein and lipopolysaccharide-induced acute pancreatitis by inhibiting TLR4-dependent autophagy. Int. J. Mol. Med. 55 (2), 25. doi:10.3892/ijmm.2024.5466

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X., Yuan, L., Tang, Y., Wu, Y., Kong, J., Zhou, B., et al. (2023). Da-cheng-qi decoction improves severe acute pancreatitis-associated acute lung injury by interfering with intestinal lymphatic pathway and reducing HMGB1-induced inflammatory response in rats. Pharm. Biol. 61 (1), 144–154. doi:10.1080/13880209.2022.2160768

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Z., Shen, Y., Cui, N., and Yang, J. (2011). Clinical observation of immunity for severe acute pancreatitis. Inflammation 34 (5), 426–431. doi:10.1007/s10753-010-9249-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, G., Pan, Y., Kayoumu, A., Zhang, L., Yin, T., Tong, Z., et al. (2017a). Indomethacin inhabits the NLRP3 inflammasome pathway and protects severe acute pancreatitis in mice. Biochem. Biophys. Res. Commun. 493 (1), 827–832. doi:10.1016/j.bbrc.2017.08.060

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, M., Zhang, Q., Chen, K., Xu, W., Xiang, X., and Xia, S. (2017b). The regulatory effect of oxymatrine on the TLR4/MyD88/NF-κB signaling pathway in lipopolysaccharide-induced MS1 cells. Phytomedicine 36, 153–159. doi:10.1016/j.phymed.2017.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, W. W., Chen, X., Ni, J. L., Cai, W. J., Zhu, S. L., Fei, A. H., et al. (2021). Study on the medication rule of traditional Chinese medicine in the treatment of acute pancreatitis based on machine learning technology. Ann. Palliat. Med. 10 (10), 10616–10625. doi:10.21037/apm-21-2505

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, X., Xiao, W., Kang, X., Yu, J., and Fan, Z. (2014). The effect of Chinese herbal medicine on non-biliogenic severe acute pancreatitis: a systematic review and meta-analysis. J. Ethnopharmacol. 155 (1), 21–29. doi:10.1016/j.jep.2014.05.040

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, Y., Wang, C. Z., Hesse-Fong, J., Lin, J. G., and Yuan, C. S. (2019). Application of Chinese medicine in acute and critical medical conditions. Am. J. Chin. Med. 47 (6), 1223–1235. doi:10.1142/s0192415x19500629

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, Z. L., Sun, H. Y., Wu, X. B., Cheng, L., and Ren, J. D. (2021). Epigallocatechin-3-gallate attenuates acute pancreatitis induced lung injury by targeting mitochondrial reactive oxygen species triggered NLRP3 inflammasome activation. Food Funct. 12 (12), 5658–5667. doi:10.1039/d1fo01154e

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, X., Jin, T., Han, C., Shi, N., Liang, G., Wen, Y., et al. (2020). Aqueous extraction from dachengqi formula granules reduces the severity of mouse acute pancreatitis via inhibition of pancreatic pro-inflammatory signalling pathways. J. Ethnopharmacol. 257, 112861. doi:10.1016/j.jep.2020.112861

PubMed Abstract | CrossRef Full Text | Google Scholar

Mao, E. Q., Tang, Y. Q., and Zhang, S. D. (2003). Formalized therapeutic guideline for hyperlipidemic severe acute pancreatitis. World J. Gastroenterol. 9 (11), 2622–2626. doi:10.3748/wjg.v9.i11.2622

PubMed Abstract | CrossRef Full Text | Google Scholar

Martinon, F., Burns, K., and Tschopp, J. (2002). The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol. Cell 10 (2), 417–426. doi:10.1016/s1097-2765(02)00599-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Mederos, M. A., Reber, H. A., and Girgis, M. D. (2021). Acute pancreatitis: a review. Jama 325 (4), 382–390. doi:10.1001/jama.2020.20317

PubMed Abstract | CrossRef Full Text | Google Scholar

Miao, B., Li, F. W., Zhang, S. W., Wang, H., Qi, W. J., and Wang, C. (2018). Efficacy and safety of tongfu powder in acute pancreatitis patients with gastrointestinal dysfunction: a clinical trial. Drug Des. Devel Ther. 12, 3665–3673. doi:10.2147/dddt.S163645

PubMed Abstract | CrossRef Full Text | Google Scholar

Nani, A., and Tehami, W. (2023). Targeting inflammasome pathway by polyphenols as a strategy for pancreatitis, gastrointestinal and liver diseases management: an updated review. Front. Nutr. 10, 1157572. doi:10.3389/fnut.2023.1157572

PubMed Abstract | CrossRef Full Text | Google Scholar

Niu, X., Sun, W., Tang, X., Chen, J., Zheng, H., Yang, G., et al. (2024). Bufalin alleviates inflammatory response and oxidative stress in experimental severe acute pancreatitis through activating Keap1-Nrf2/HO-1 and inhibiting NF-κB pathways. Int. Immunopharmacol. 142 (Pt A), 113113. doi:10.1016/j.intimp.2024.113113

PubMed Abstract | CrossRef Full Text | Google Scholar

Paik, S., Kim, J. K., Silwal, P., Sasakawa, C., and Jo, E. K. (2021). An update on the regulatory mechanisms of NLRP3 inflammasome activation. Cell Mol. Immunol. 18 (5), 1141–1160. doi:10.1038/s41423-021-00670-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, X., Ye, L., Ren, Z., Li, J., Li, B., Pan, L. L., et al. (2023). Biochanin A ameliorates caerulein-induced acute pancreatitis and associated intestinal injury in mice by inhibiting TLR4 signaling. J. Nutr. Biochem. 113, 109229. doi:10.1016/j.jnutbio.2022.109229

PubMed Abstract | CrossRef Full Text | Google Scholar

Papantoniou, K., Aggeletopoulou, I., Michailides, C., Pastras, P., and Triantos, C. (2024). Understanding the role of NLRP3 inflammasome in acute pancreatitis. Biol. (Basel). 13 (11), 945. doi:10.3390/biology13110945

PubMed Abstract | CrossRef Full Text | Google Scholar

Petrov, M. S., and Yadav, D. (2019). Global epidemiology and holistic prevention of pancreatitis. Nat. Rev. Gastroenterol. Hepatol. 16 (3), 175–184. doi:10.1038/s41575-018-0087-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Qiong, W., Yiping, W., Jinlin, Y., Tao, G., Zhen, G., and Pengcheng, Z. (2005). Chinese medicinal herbs for acute pancreatitis. Cochrane Database Syst. Rev. 2005 (1), Cd003631. doi:10.1002/14651858.CD003631.pub2

PubMed Abstract | CrossRef Full Text | Google Scholar

Rajapriya, S., and Geetha, A. (2021). Effect of luteolin on the gene level expression of apoptosis-associated speck-like protein containing a caspase recruitment domain of NLRP3 inflammasome and NF-κB in rats subjected to experimental pancreatitis - influence of HSP70. J. Basic Clin. Physiol. Pharmacol. 33 (4), 477–486. doi:10.1515/jbcpp-2020-0255

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruan, H. H., Wei, W., and Xu, Y. F. (2019). Effects of honeysuckle extract on ROS-NLRP3 inflammasome signaling pathway in lung injury of severe acute pancreatitis rats. J. Emerg. Tradit. Chin. Med. 28 (08), 1344–1348+1353. doi:10.3969/j.issn.1004-745X.2019.08.008

CrossRef Full Text | Google Scholar

Santos, J. C., Dick, M. S., Lagrange, B., Degrandi, D., Pfeffer, K., Yamamoto, M., et al. (2018). LPS targets host guanylate-binding proteins to the bacterial outer membrane for non-canonical inflammasome activation. Embo J. 37 (6), e98089. doi:10.15252/embj.201798089

PubMed Abstract | CrossRef Full Text | Google Scholar

Sendler, M., van den Brandt, C., Glaubitz, J., Wilden, A., Golchert, J., Weiss, F. U., et al. (2020). NLRP3 inflammasome regulates development of systemic inflammatory response and compensatory anti-inflammatory response syndromes in mice with acute pancreatitis. Gastroenterology 158 (1), 253–269. doi:10.1053/j.gastro.2019.09.040

PubMed Abstract | CrossRef Full Text | Google Scholar

Seoane, P. I., Lee, B., Hoyle, C., Yu, S., Lopez-Castejon, G., Lowe, M., et al. (2020). The NLRP3-inflammasome as a sensor of organelle dysfunction. J. Cell Biol. 219 (12), e202006194. doi:10.1083/jcb.202006194

PubMed Abstract | CrossRef Full Text | Google Scholar

Shen, Y., Yang, H., Wu, D., Yang, H., and Hong, D. (2022). NLRP3 inflammasome inhibitor MCC950 can reduce the damage of pancreatic and intestinal barrier function in mice with acute pancreatitis. Acta Cir. Bras. 37 (7), e370706. doi:10.1590/acb370706

PubMed Abstract | CrossRef Full Text | Google Scholar

Sheng, B., Zhao, L., Zang, X., Zhen, J., Liu, Y., Bian, W., et al. (2021). Quercetin inhibits caerulein-induced acute pancreatitis through regulating miR-216b by targeting MAP2K6 and NEAT1. Inflammopharmacology 29 (2), 549–559. doi:10.1007/s10787-020-00767-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Sheng, L. P., Han, C. Q., Ling, X., Guo, X. W., Lin, R., and Ding, Z. (2023). Proanthocyanidins suppress NLRP3 inflammasome and M1 macrophage polarization to alleviate severe acute pancreatitis in mice. J. Biochem. Mol. Toxicol. 37 (2), e23242. doi:10.1002/jbt.23242

PubMed Abstract | CrossRef Full Text | Google Scholar

Tan, N., Gwee, K. A., Tack, J., Zhang, M., Li, Y., Chen, M., et al. (2020). Herbal medicine in the treatment of functional gastrointestinal disorders: a systematic review with meta-analysis. J. Gastroenterol. Hepatol. 35 (4), 544–556. doi:10.1111/jgh.14905

PubMed Abstract | CrossRef Full Text | Google Scholar

Thompson, D. A., Tsaava, T., Rishi, A., George, S. J., Hepler, T. D., Hide, D., et al. (2023). Galantamine ameliorates experimental pancreatitis. Mol. Med. 29 (1), 149. doi:10.1186/s10020-023-00746-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Toldo, S., Mezzaroma, E., Buckley, L. F., Potere, N., Di Nisio, M., Biondi-Zoccai, G., et al. (2022). Targeting the NLRP3 inflammasome in cardiovascular diseases. Pharmacol. Ther. 236, 108053. doi:10.1016/j.pharmthera.2021.108053

PubMed Abstract | CrossRef Full Text | Google Scholar

Unterberger, S., Davies, K. A., Rambhatla, S. B., and Sacre, S. (2021). Contribution of toll-like receptors and the NLRP3 inflammasome in rheumatoid arthritis pathophysiology. Immunotargets Ther. 10, 285–298. doi:10.2147/itt.S288547

PubMed Abstract | CrossRef Full Text | Google Scholar

Vivarelli, S., Costa, C., Teodoro, M., Giambò, F., Tsatsakis, A. M., and Fenga, C. (2023). Polyphenols: a route from bioavailability to bioactivity addressing potential health benefits to tackle human chronic diseases. Arch. Toxicol. 97 (1), 3–38. doi:10.1007/s00204-022-03391-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Wan, J., Ren, Y., Yang, X., Li, X., Xia, L., and Lu, N. (2020). The role of neutrophils and neutrophil extracellular traps in acute pancreatitis. Front. Cell Dev. Biol. 8, 565758. doi:10.3389/fcell.2020.565758

PubMed Abstract | CrossRef Full Text | Google Scholar

Wan, M. H., Li, J., Gong, H. L., Xue, P., Zhu, L., Chen, G. Y., et al. (2011). Clinical observation on the effect of dexamethasone and Chinese herbal decoction for purgation in severe acute pancreatitis patients. Chin. J. Integr. Med. 17 (2), 141–145. doi:10.1007/s11655-011-0630-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Q., Liu, X., Du, Z., Zheng, Y., Meng, Z., Lv, Z., et al. (2025a). Astragalus polysaccharide reduces the severity of acute pancreatitis under a high-fat diet through enriching L. reuteri and propionate. Int. J. Biol. Macromol. 298, 140021. doi:10.1016/j.ijbiomac.2025.140021

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, R., Jiang, J., Song, P., Peng, Q., Jin, X., Li, B., et al. (2025b). Kinsenoside alleviates experimental acute pancreatitis by suppressing M1 macrophage polarization via the TLR4/STAT1 signaling pathway. J. Ethnopharmacol. 345, 119551. doi:10.1016/j.jep.2025.119551

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X., Cai, H., Chen, Z., Zhang, Y., Wu, M., Xu, X., et al. (2021). Baicalein alleviates pyroptosis and inflammation in hyperlipidemic pancreatitis by inhibiting NLRP3/Caspase-1 pathway through the miR-192-5p/TXNIP axis. Int. Immunopharmacol. 101 (Pt B), 108315. doi:10.1016/j.intimp.2021.108315

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X., Zhou, Y., Liu, Y., Mo, T., Chen, Z., Zhang, Y., et al. (2025c). Baicalein reduces pyroptosis of acinar cells in hyperlipidemic acute pancreatitis by inhibiting M1 polarization of macrophages via the HMGB1/TLR4/NLRP3 pathway. Inflammation. doi:10.1007/s10753-025-02254-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Z., Liu, J., Li, F., Ma, S., Zhao, L., Ge, P., et al. (2023). Mechanisms of qingyi decoction in severe acute pancreatitis-associated acute lung injury via gut microbiota: targeting the short-chain fatty acids-mediated AMPK/NF-κB/NLRP3 pathway. Microbiol. Spectr. 11 (4), e0366422. doi:10.1128/spectrum.03664-22

PubMed Abstract | CrossRef Full Text | Google Scholar

Wen, Y., Han, C., Liu, T., Wang, R., Cai, W., Yang, J., et al. (2020). Chaiqin chengqi decoction alleviates severity of acute pancreatitis via inhibition of TLR4 and NLRP3 inflammasome: identification of bioactive ingredients via pharmacological sub-network analysis and experimental validation. Phytomedicine 79, 153328. doi:10.1016/j.phymed.2020.153328

PubMed Abstract | CrossRef Full Text | Google Scholar

Wen, Y., Li, Y., Liu, T., Huang, L., Yao, L., Deng, D., et al. (2024). Chaiqin chengqi decoction treatment mitigates hypertriglyceridemia-associated acute pancreatitis by modulating liver-mediated glycerophospholipid metabolism. Phytomedicine 134, 155968. doi:10.1016/j.phymed.2024.155968

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, C. Y., Wang, K. Q., Qin, Y. Y., Wang, H. W., Wu, M. M., Zhu, X. D., et al. (2024a). Micheliolide ameliorates severe acute pancreatitis in mice through potentiating Nrf2-mediated anti-inflammation and anti-oxidation effects. Int. Immunopharmacol. 143 (Pt 2), 113490. doi:10.1016/j.intimp.2024.113490

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, S. K., Wang, L., Wang, F., and Zhang, J. (2024b). Resveratrol improved mitochondrial biogenesis by activating SIRT1/PGC-1α signal pathway in SAP. Sci. Rep. 14 (1), 26216. doi:10.1038/s41598-024-76825-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X., Yao, J., Hu, Q., Kang, H., Miao, Y., Zhu, L., et al. (2022). Emodin ameliorates acute pancreatitis-associated lung injury through inhibiting the alveolar macrophages pyroptosis. Front. Pharmacol. 13, 873053. doi:10.3389/fphar.2022.873053

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X. B., Sun, H. Y., Luo, Z. L., Cheng, L., Duan, X. M., and Ren, J. D. (2020). Plasma-derived exosomes contribute to pancreatitis-associated lung injury by triggering NLRP3-dependent pyroptosis in alveolar macrophages. Biochim. Biophys. Acta Mol. Basis Dis. 1866 (5), 165685. doi:10.1016/j.bbadis.2020.165685

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X. N. (2002). Guidelines for treatment of severe acute pancreatitis. Hepatobiliary Pancreat. Dis. Int. 1 (3), 446–451.

PubMed Abstract | Google Scholar

Xia, T., Gu, Y., Shen, J., Zheng, L., and Xu, C. (2021). Limonin ameliorates acute pancreatitis by suppressing JAK2/STAT3 signaling pathway. Environ. Toxicol. 36 (12), 2392–2403. doi:10.1002/tox.23352

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, J., Huang, K., Lin, H., Xia, Z., Zhang, J., Li, D., et al. (2020). Mogroside II(E) inhibits digestive enzymes via suppression of interleukin 9/Interleukin 9 receptor signalling in acute pancreatitis. Front. Pharmacol. 11, 859. doi:10.3389/fphar.2020.00859

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, W., Shi, H., Tian, Y., Chen, F., Xie, Y., Han, X., et al. (2025). Chaihuang qingfu pills protect against acute pancreatitis-associated acute lung injury through MMP9-NLRP3-Pyroptosis pathway. J. Inflamm. Res. 18, 2317–2338. doi:10.2147/jir.S501531

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiong, G. F., Li, D. W., Zheng, M. B., and Liu, S. C. (2019). The effects of lycium barbarum polysaccharide (LBP) in a mouse model of cerulein-induced acute pancreatitis. Med. Sci. Monit. 25, 3880–3886. doi:10.12659/msm.913820

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiong, J., Ni, J., Hu, G., Shen, J., Zhao, Y., Yang, L., et al. (2013). Shikonin ameliorates cerulein-induced acute pancreatitis in mice. J. Ethnopharmacol. 145 (2), 573–580. doi:10.1016/j.jep.2012.11.032

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiong, J., Wang, K., Yuan, C., Xing, R., Ni, J., Hu, G., et al. (2017). Luteolin protects mice from severe acute pancreatitis by exerting HO-1-mediated anti-inflammatory and antioxidant effects. Int. J. Mol. Med. 39 (1), 113–125. doi:10.3892/ijmm.2016.2809

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, T., Yu, W., Fang, H., Wang, Z., Chi, Z., Guo, X., et al. (2022). Ubiquitination of NLRP3 by gp78/Insig-1 restrains NLRP3 inflammasome activation. Cell Death Differ. 29 (8), 1582–1595. doi:10.1038/s41418-022-00947-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Xue, J. C., Yuan, S., Hou, X. T., Meng, H., Liu, B. H., Cheng, W. W., et al. (2023). Natural products modulate NLRP3 in ulcerative colitis. Front. Pharmacol. 14, 1265825. doi:10.3389/fphar.2023.1265825

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, X., Lin, T., Zhu, Q., Zhang, Y., Song, Z., and Pan, X. (2023). Naringenin protects against acute pancreatitis-associated intestinal injury by inhibiting NLRP3 inflammasome activation via AhR signaling. Front. Pharmacol. 14, 1090261. doi:10.3389/fphar.2023.1090261

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, C., Wang, T., Chen, J., He, J., Li, Y., Chen, C., et al. (2021). Traditional Chinese medicine formulas alleviate acute pancreatitis: pharmacological activities and mechanisms. Pancreas 50 (10), 1348–1356. doi:10.1097/mpa.0000000000001931

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, J., Tang, X., Ke, X., Dai, Y., and Shi, J. (2022). Triptolide suppresses NF-κB-Mediated inflammatory responses and activates expression of Nrf2-Mediated antioxidant genes to alleviate caerulein-induced acute pancreatitis. Int. J. Mol. Sci. 23 (3), 1252. doi:10.3390/ijms23031252

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, J., Zhou, Y., and Shi, J. (2020a). Cordycepin protects against acute pancreatitis by modulating NF-κB and NLRP3 inflammasome activation via AMPK. Life Sci. 251, 117645. doi:10.1016/j.lfs.2020.117645

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Q., Yue, C., Huang, X., Wang, Z., Li, Z., Hu, W., et al. (2024). Functional mechanism of baicalein in alleviating severe acute pancreatitis-acute lung injury by blocking the TLR4/MyD88/TRIF signaling pathway. Histol. Histopathol. 39 (10), 1381–1394. doi:10.14670/hh-18-733

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, X., Liu, Y., Zhong, C., Hu, J., Xu, S., Zhang, P., et al. (2023a). Total flavonoids of Chrysanthemum indicum L inhibit acute pancreatitis through suppressing apoptosis and inflammation. BMC Complement. Med. Ther. 23 (1), 23. doi:10.1186/s12906-023-03851-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, X., Zhang, X., Lin, Z., Guo, J., Yang, X., Yao, L., et al. (2020b). Chaiqin chengqi decoction alleviates severe acute pancreatitis associated acute kidney injury by inhibiting endoplasmic reticulum stress and subsequent apoptosis. Biomed. Pharmacother. 125, 110024. doi:10.1016/j.biopha.2020.110024

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Y., Hu, Q., Kang, H., Li, J., Zhao, X., Zhu, L., et al. (2023b). Urolithin A protects severe acute pancreatitis-associated acute cardiac injury by regulating mitochondrial fatty acid oxidative metabolism in cardiomyocytes. MedComm (2020) 4 (6), e459. doi:10.1002/mco2.459

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Y., Yin, H. Z., and Hu, Y. F. (2019). Mechanism in the improvement of ROS-NLRP3 inflammasome signaling pathway by Salvia miltiorrhiza extract in rats with severe acute pancreatitis and lung injury. Chin. Pharm. 22 (10), 1795–1800.

Google Scholar

Ye, Z., Cheng, L., Xuan, Y., Yu, K., Li, J., and Gu, H. (2025). Chlorogenic acid alleviates the development of severe acute pancreatitis by inhibiting NLPR3 inflammasome activation via Nrf2/HO-1 signaling. Int. Immunopharmacol. 151, 114335. doi:10.1016/j.intimp.2025.114335

PubMed Abstract | CrossRef Full Text | Google Scholar

Yi, Y. S. (2020). Caspase-11 non-canonical inflammasome: emerging activator and regulator of infection-mediated inflammatory responses. Int. J. Mol. Sci. 21 (8), 2736. doi:10.3390/ijms21082736

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, C., Xu, X., Wang, N., Zhu, Q., Zhang, J., Gong, W., et al. (2022). Paeonol protects against acute pancreatitis by inhibiting M1 macrophage polarization via the NLRP3 inflammasomes pathway. Biochem. Biophys. Res. Commun. 600, 35–43. doi:10.1016/j.bbrc.2022.02.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Yue, X., Lai, L., Wang, R., Tan, L., Wang, Y., Xie, Q., et al. (2024). DGA ameliorates severe acute pancreatitis through modulating macrophage pyroptosis. Inflamm. Res. 73 (10), 1803–1817. doi:10.1007/s00011-024-01931-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Zahid, A., Li, B., Kombe, A. J. K., Jin, T., and Tao, J. (2019). Pharmacological inhibitors of the NLRP3 inflammasome. Front. Immunol. 10, 2538. doi:10.3389/fimmu.2019.02538

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, Y., Liu, X., Yi, Q., Qiao, G., Wang, L., Chen, L., et al. (2024). Free total rhubarb anthraquinones protect intestinal mucosal barrier of SAP rats via inhibiting the NLRP3/caspase-1/GSDMD pyroptotic pathway. J. Ethnopharmacol. 326, 117873. doi:10.1016/j.jep.2024.117873

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhan, L., Pu, J., Hu, Y., Xu, P., Liang, W., and Ji, C. (2021). Uncovering the pharmacology of xiaochaihu decoction in the treatment of acute pancreatitis based on the network pharmacology. Biomed. Res. Int. 2021, 6621682. doi:10.1155/2021/6621682

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, D., Li, L., Li, J., Wei, Y., Tang, J., Man, X., et al. (2022). Colchicine improves severe acute pancreatitis-induced acute lung injury by suppressing inflammation, apoptosis and oxidative stress in rats. Biomed. Pharmacother. 153, 113461. doi:10.1016/j.biopha.2022.113461

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, G., Tang, L., Liu, H., Liu, D., Wang, M., Cai, J., et al. (2021a). Psidium guajava flavonoids prevent NLRP3 inflammasome activation and alleviate the pancreatic fibrosis in a chronic pancreatitis mouse model. Am. J. Chin. Med. 49 (8), 2001–2015. doi:10.1142/s0192415x21500944

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, G. X., Wang, M. X., Nie, W., Liu, D. W., Zhang, Y., and Liu, H. B. (2017). P2X7R blockade prevents NLRP3 inflammasome activation and pancreatic fibrosis in a mouse model of chronic pancreatitis. Pancreas 46 (10), 1327–1335. doi:10.1097/mpa.0000000000000928

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, J., Wu, Y. L., Xu, H. X., Zhang, Y. B., Ren, P. Y., Xian, Y. F., et al. (2024a). Brusatol alleviates pancreatic carcinogenesis via targeting NLRP3 in transgenic Krastm4Tyj Trp53tm1Brn Tg (Pdx1-cre/Esr1*) #dam mice. Biomed. Pharmacother. 177, 116977. doi:10.1016/j.biopha.2024.116977

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, L., Wang, K., Huang, L., Deng, B., Chen, C., Zhao, K., et al. (2025). Ganoderic acid A alleviates severe acute pancreatitis by modulating gut homeostasis and inhibiting TLR4-NLRP3 signaling. J. Agric. Food Chem. 73 (2), 1563–1579. doi:10.1021/acs.jafc.4c07635

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, M. J., Zhang, G. L., Yuan, W. B., Ni, J., and Huang, L. F. (2008). Treatment of abdominal compartment syndrome in severe acute pancreatitis patients with traditional Chinese medicine. World J. Gastroenterol. 14 (22), 3574–3578. doi:10.3748/wjg.14.3574

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Q., Tao, X., Xia, S., Qu, J., Song, H., Liu, J., et al. (2019). Emodin attenuated severe acute pancreatitis via the P2X ligand-gated ion channel 7/NOD-like receptor protein 3 signaling pathway. Oncol. Rep. 41 (1), 270–278. doi:10.3892/or.2018.6844

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, R., Zhu, Z., Ma, Y., Tang, T., Wu, J., Huang, F., et al. (2024b). Rhizoma alismatis decoction improved mitochondrial dysfunction to alleviate SASP by enhancing autophagy flux and apoptosis in hyperlipidemia acute pancreatitis. Phytomedicine 129, 155629. doi:10.1016/j.phymed.2024.155629

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Ma, J., and Zhang, W. (2021b). Berberine for bone regeneration: therapeutic potential and molecular mechanisms. J. Ethnopharmacol. 277, 114249. doi:10.1016/j.jep.2021.114249

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, X., Wu, X., Hu, Q., Yao, J., Yang, Y., Wan, M., et al. (2024). Yinchenhao decoction protects against acute liver injury in mice with biliary acute pancreatitis by regulating the gut microflora-bile acids-liver axis. Gastroenterol. Res. Pract. 2024, 8882667. doi:10.1155/2024/8882667

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, Y. S., Wu, Z. S., Zhang, L. Y., Ke, L., Li, W. Q., Li, N., et al. (2015). Nicotine ameliorates experimental severe acute pancreatitis via enhancing immunoregulation of CD4+ CD25+ regulatory T cells. Pancreas 44 (3), 500–506. doi:10.1097/mpa.0000000000000294

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, L., Yu, J., Wang, S., Ma, Y., Liu, X., Zhang, X., et al. (2024). Tectoridin alleviates caerulein-induced severe acute pancreatitis by targeting ERK2 to promote macrophage M2 polarization. Arch. Biochem. Biophys. 752, 109873. doi:10.1016/j.abb.2023.109873

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Z., Choi, J. W., Shin, J. Y., Kim, D. U., Kweon, B., Oh, H., et al. (2021). Betulinic acid ameliorates the severity of acute pancreatitis via inhibition of the NF-κB signaling pathway in mice. Int. J. Mol. Sci. 22 (13), 6871. doi:10.3390/ijms22136871

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, L., and He, C. (2022). Crocetin alleviates the caerulein-induced apoptosis and inflammation in AR42J cells by activating SIRT1 via NF-κB. J. Nat. Med. 76 (2), 410–418. doi:10.1007/s11418-021-01597-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhuang, Y., Wang, H., Su, J., Zhang, J., Wang, Q., Chen, Y., et al. (2021). A narrative review of positive regulation of NLRP3 inflammasome in rheumatoid arthritis. Ann. Palliat. Med. 10 (12), 12877–12885. doi:10.21037/apm-21-3472

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: acute pancreatitis, traditional Chinese medicine, NLRP3, inflammation, antiinflammatory

Citation: He X, Xia J, Chen Q, Huang Z, Lu J and Ren Y (2025) An overview of current research on the modulation of NLRP3 inflammasome by traditional Chinese medicine to combat acute pancreatitis. Front. Mol. Biosci. 12:1634132. doi: 10.3389/fmolb.2025.1634132

Received: 23 May 2025; Accepted: 07 July 2025;
Published: 16 July 2025.

Edited by:

Lynda Bourebaba, Wroclaw University of Environmental and Life Sciences, Poland

Reviewed by:

Junpu Wang, Central South University, China
Jia Zhao, North Sichuan Medical College, China

Copyright © 2025 He, Xia, Chen, Huang, Lu and Ren. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Yisong Ren, cGRxenl5eWljdUAxNjMuY29t

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.