Back to Journals » Journal of Inflammation Research » Volume 18
From Inflammasomes to Pyroptosis: Molecular Mechanisms in Chronic Intestinal Diseases — Opportunity or Challenge?
Authors Fang J , Zhu W, Yu D, Zhu L , Zha H, Tang J, Li Y , Zhu X , Zhao T, Zhang W
Received 8 October 2024
Accepted for publication 28 February 2025
Published 7 March 2025 Volume 2025:18 Pages 3349—3360
DOI https://doi.org/10.2147/JIR.S498703
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Professor Ning Quan
Jintao Fang,1,* Weihan Zhu,1,* Dian Yu,1 Lujian Zhu,2 Haorui Zha,1 Jingyi Tang,3 Yujia Li,1 Xiaxin Zhu,1 Ting Zhao,1 Wei Zhang1,4
1The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China; 2Department of Infectious Diseases, Jinhua Municipal Central Hospital, Jinhua, 321000, People’s Republic of China; 3Lanxi Hospital of Traditional Chinese Medicine, Jinhua, Zhejiang, 321100, People’s Republic of China; 4Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Wei Zhang, Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China, Email [email protected]
Abstract: Pyroptosis is a unique form of programmed cell death characterized by intense inflammation. It involves the activation of Gasdermin proteins, which form membrane pores, leading to rapid cell rupture and the release of inflammatory molecules. Unlike other types of cell death, pyroptosis has distinct activation mechanisms and plays a complex role in chronic intestinal diseases, including inflammatory bowel disease, intestinal fibrosis, chronic infectious enteritis, and colorectal cancer. This review comprehensively examines how pyroptosis influences disease development and progression while exploring the therapeutic potential of targeting pyroptosis-related pathways. Moreover, the complex interplay between gut microbiota and pyroptosis is summarized, highlighting its critical role in the pathogenesis of chronic intestinal disorders. A deeper understanding of pyroptosis-related mechanisms in these diseases may provide valuable insights for future research and contribute to the development of innovative therapeutic strategies in gastroenterology.
Keywords: pyroptosis, chronic intestinal diseases, inflammasomes, gasdermin proteins, cell death
Introduction
Pyroptosis is a form of programmed necrotic cell death characterized by intense inflammatory responses.1–4 It is triggered by intracellular infections caused by bacteria, viruses, fungi, and protozoa in response to pathogen-associated or damage-associated molecular patterns (PAMPs or DAMPs).5,6 While pyroptosis plays a key role in host defense by eliminating infected cells, excessive inflammatory responses can contribute to severe pathological conditions, such as multi-organ dysfunction.7 Pyroptosis can be activated via both inflammasome-dependent and non-inflammasome-dependent pathways, with the former being more extensively studied. The classic inflammasome-dependent pathway begins with recognizing pathogenic microorganisms or intracellular damage signals by pattern-recognition receptors (PRRs) (Figure 1). Upon activation, PRRs interact with pro-caspase-1 and the adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD), forming a multiprotein inflammasome complex.8 This inflammasome plays a central role in pyroptosis, facilitating caspase-1 activation.9 Once activated, caspase-1 undergoes auto-cleavage into its CARD domain and P20/P10 subunits, which oligomerize into a tetramer. This tetramer cleaves Gasdermin D (GSDMD) and processes pro-interleukin (IL)-1β and pro-IL-18 into their active forms, IL-1β and IL-18. GSDMD serves as a key mediator of pyroptosis.10 Upon cleavage, its N-terminal domain forms pores in the plasma membrane, leading to membrane rupture and the release of intracellular contents along with pro-inflammatory cytokines.11,12 While this mechanism effectively combats infections, it can also exacerbate inflammatory damage in various disease contexts.13 On the other hand, the non-classical pyroptosis pathway involves caspase-4 and 5 in humans or caspase-11 in mice, which directly detect intracellular signals, bypassing the inflammasome complex.8,14 Unlike caspase-1, caspase-11 does not process pro-IL-1β or pro-IL-18 into their mature forms.15
![]() |
Figure 1 The classic activation mechanism of pyroptosis: caspase-1-dependent. By Figdraw. |
Recent research has highlighted the significant roles of Gasdermin protein isoforms in chronic intestinal diseases, particularly Gasdermin B (GSDMB) and Gasdermin C (GSDMC). GSDMB exists in multiple isoforms with distinct functions, and certain variants can induce pyroptosis when cleaved by proteases such as Granzyme A. This cleavage releases N-terminal fragments that promote membrane lysis and drive inflammatory responses, associating GSDMB with inflammatory bowel disease (IBD) and colorectal cancer (CRC).16 Meanwhile, GSDMC, expressed in intestinal epithelial cells, is regulated by type 2 cytokines such as IL-4 and IL-13. Targeting Gasdermin isoforms presents promising therapeutic strategies for chronic intestinal diseases: enhancing pro-pyroptotic GSDMB isoforms may strengthen immune responses against pathogens while reducing CRC tumorigenesis, whereas inhibiting non-pyroptotic variants could mitigate excessive inflammation in IBD. Understanding post-translational modifications and cleavage mechanisms of these proteins may lead to novel interventions that selectively regulate their activity.17
Inflammasomes are essential cytoplasmic multiprotein complexes that detect host threats by recognizing PAMPs, DAMPs, homeostasis-altering molecular processes (HAMPs), or effector-triggered immunity (ETI).18 They play a pivotal role in pyroptosis by managing inflammatory responses to infections and tissue damage, promoting the release of IL-1β and IL-18.18 Typical inflammasomes consist of three main components: 1) PRRs, which serve as the sensors of PAMPs and DAMPs and are essential for inflammasome assembly. 2) ASC, which is activated and undergoes oligomerization and recruitment upon detecting inflammatory ligands, serving as a bridge between sensor proteins and effector proteins, forming the scaffold for inflammasome assembly.19 3) Effector proteins, primarily represented by inflammatory caspases such as Caspase-1, essential for inflammasome function.20 Pyroptosis is one of several regulated cell death mechanisms, alongside apoptosis and necroptosis, each with distinct pathways, outcomes, and inflammatory implications. Pyroptosis is a rapid inflammatory response to infection or cellular stress, contrasting with apoptosis, which is a controlled, non-inflammatory process. Necroptosis acts as an alternative pathway when apoptosis fails, sharing some characteristics with pyroptosis but distinct in its regulatory mechanisms and triggers. Necroptosis is often a backup mechanism activated when apoptosis is inhibited and is mediated by receptor-interacting protein kinases (RIPK1 and RIPK3), leading to MLKL activation.21,22
Recent research has highlighted pyroptosis as a key regulator of intestinal homeostasis and inflammation in chronic diseases.23 Chronic intestinal diseases, such as IBD, irritable bowel syndrome (IBS),24 celiac disease (CeD),25 chronic infectious enteritis, and intestinal fibrosis,26 are increasingly recognized as significant global public health concerns due to their rising prevalence and associated mortality rates.27 Given its crucial role in both immune defense and pathological inflammation, this review aims to provide a comprehensive overview of the molecular mechanisms underlying pyroptosis in chronic intestinal diseases (Figure 2). By examining its dual roles in both defense and pathology, this review sheds light on the multifaceted nature of pyroptosis and its implications for future research and clinical advancements in gastroenterology.
Pyroptosis in IBD
IBD, which includes Crohn’s disease (CD) and ulcerative colitis (UC),28,29 is a chronic, non-specific inflammatory disorder of the gastrointestinal tract.30,31 The etiology of IBD is multifactorial and not yet fully understood. However, it is widely recognized that environmental factors, genetic predisposition, and gut microbiome dysbiosis interact to disrupt intestinal immunity, compromise the mucosal barrier, and trigger chronic inflammation and tissue damage.32,33 Pyroptosis plays a key role in IBD progression, with excessive activation observed in the intestinal tissues of IBD patients.
This dysregulated pyroptotic response contributes to chronic inflammation by amplifying damage signals and intensifying local immune responses, perpetuating a cycle of persistent inflammation that exacerbates the disease.34 Emerging evidence suggests that pyroptosis is not only a major driver of intestinal injury but also a key factor in tissue repair, fibrosis, and long-term complications such as CRC.23
Several studies have reported elevated expression of inflammasomes in the intestines of IBD patients, leading to excessive pyroptosis and exacerbating intestinal damage.35 Research has demonstrated that NOD-like receptor protein 3 (NLRP3)-deficient mice show significantly reduced pyroptosis in colonic tissues, alleviating IBD severity and delaying disease onset.36 This finding highlights the NLRP3 inflammasome as a crucial mediator in both the development and potential treatment of colitis. Furthermore, Gasdermin family proteins, such as GSDMB, Gasdermin D (GADMD), and Gasdermin E (GSDME), which are critical in executing pyroptosis, strongly correlate with IBD severity, making them promising therapeutic targets.37 Recent advances in therapeutic strategies include the development of oral pyroptosis inhibitors, which specifically target intestinal lesions.34,38 These inhibitors hold great potential for reducing inflammation, preventing excessive pyroptosis, and preserving intestinal integrity, offering new avenues for IBD management.
Pyroptosis in Intestinal Fibrosis
Intestinal fibrosis is a severe complication of chronic gastrointestinal diseases, including IBD, which encompasses CD and UC, as well as conditions such as ulcerative jejunitis and radiation enteritis.39 This pathological condition results in bowel narrowing, structural alterations, and functional impairment, significantly reducing patients’ quality of life.40 Intestinal fibrosis is closely associated with chronic inflammation and is influenced by factors such as cellular damage, recruitment of inflammatory cells, and the production of transforming growth factor-beta (TGF-β).41 Recent studies have revealed a positive correlation between fibrosis, pyroptosis, and inflammasomes.42,43 Key upstream activators of the TGF-β/Sma-Mad related protein (SMAD) pathway, such as caspase-1, NLRP3, and nuclear factor kappa B (NF-κB), have been identified, with TGFβ emerging as a potential anti-fibrotic target. Activation of inflammasomes, including NLRP3 and absent in melanoma 2 (AIM2), has been implicated in fibrosis progression.44 The NLRP3 inhibitor MCC950 has demonstrated significant anti-fibrotic effects in preclinical studies.45 During chronic inflammation, macrophages and lymphocytes are recruited to affected tissues, where they undergo pyroptosis. This process releases inflammatory factors, activates the TGF-β signaling pathway, increases extracellular matrix (ECM) synthesis, and indirectly promotes intestinal fibrosis.46 GSDMD-dependent macrophage pyroptosis has been shown to intensify the inflammatory response in the intestinal mucosa, contributing to local cell death and fibrosis progression.47,48 Furthermore, GSDME has emerged as a novel therapeutic target for fibrosis prevention and treatment.49 In vitro studies using intestinal epithelial cell models have highlighted a strong association between pyroptosis and intestinal fibrosis. As the understanding of pyroptosis mechanisms deepens, researchers are actively exploring anti-fibrotic strategies targeting pyroptosis pathways, including inhibitors of NLRP3 inflammasomes and caspase-1, which have demonstrated promise in preclinical models.50 While studies on pyroptosis and fibrosis have focused on other organs, such as the liver,51 kidneys,52 and lungs,53 further investigation is necessary to fully elucidate its role in intestinal fibrosis. Targeting pyroptosis could offer novel therapeutic opportunities for managing intestinal fibrosis and improving outcomes for patients with chronic gastrointestinal diseases.
Pyroptosis in CeD
CeD is a chronic autoimmune disorder of the small intestine triggered by gluten ingestion in genetically susceptible individuals.54 This condition affects approximately 1% of the global population.55 In CeD, DAMPs released during inflammatory cell death are thought to initiate mucosal damage and contribute to the persistence of chronic disease. Pyroptosis, a pro-inflammatory form of programmed cell death, is believed to play a significant role in the pathophysiology of CeD by driving inflammation and tissue damage. Elevated caspase-1, IL-1β, and GSDMD levels have been observed in the duodenal tissues of CeD patients, suggesting active pyroptosis.56 In genetically predisposed individuals, gluten consumption induces an influx of γδT cells into the duodenal mucosa. These cells produce high levels of interferon-γ, which upregulates interferon regulatory factor 1 (IRF1) expression and enhances GSDMD expression in intestinal epithelial cells, priming them for a “pre-pyroptotic” state. These cells undergo pyroptosis following gluten exposure, leading to villous atrophy in the duodenum and subsequent nutrient malabsorption.57 During pyroptosis, the release of intracellular contents into the extracellular space can be misinterpreted by the immune system as foreign antigens, thereby triggering an autoimmune response. This process is important in the pathogenesis of CeD58 and may also contribute to further damage in the small intestine. Furthermore, CeD is frequently associated with other autoimmune disorders, such as thyroid diseases and type 1 diabetes, where pyroptosis is also thought to play a significant role.59 The association between pyroptosis and CeD highlights its broader relevance in autoimmune diseases. Future research should aim to elucidate the precise molecular mechanisms underlying pyroptosis in CeD and explore targeted therapeutic strategies to mitigate inflammation and intestinal damage.
Pyroptosis in Diverticulitis
Diverticulitis is a chronic inflammatory condition of the colon associated with diverticular disease.60 It is characterized by inflammation or infection of diverticula—pouches formed within the wall of the large intestine. The recurrence of diverticulitis is unpredictable and often results in persistent gastrointestinal symptoms.61 The pathogenesis of diverticulitis is closely associated with an imbalance in the gut microbiota and inflammation of the intestinal wall. A complex positive feedback loop exists between the inflammatory response in diverticulitis and pyroptosis. Research suggests that the composition of the gut microbiota significantly influences both the formation of diverticula and the subsequent inflammatory response.62 During episodes of diverticulitis, infection by gut microbes can trigger pyroptosis, a pro-inflammatory form of programmed cell death. This process leads to cellular destruction and the release of cytokines, which amplify the immune response. The resulting inflammatory cascade exacerbates intestinal inflammation and contributes to disease progression.63
Some studies suggest that acute diverticulitis may lead to subsequent IBS-like symptoms, although the mechanisms underlying this transition remain poorly understood. Furthermore, the role of pyroptosis in diverticulitis has not been extensively investigated. Further research is needed to address this gap and determine how pyroptosis influences disease progression, recurrence, and symptomatology.
Pyroptosis in IBS
IBS is a common chronic functional gastrointestinal disorder characterized by recurrent abdominal pain or discomfort and altered bowel habits.64 It affects approximately 5% to 10% of the global population.24,65 Although the exact causes and pathophysiological mechanisms of IBS remain unclear, it is widely believed that a combination of factors, such as gut microbiota, immune responses, gastrointestinal motility, and psychological influences, contribute to its development. Psychological factors play an important role in IBS, with evidence indicating that conditions such as anxiety may exacerbate pyroptosis, potentially worsening IBS symptoms.66,67 Excessive pyroptosis has been implicated in altering the gut microbiota composition, leading to microbial dysbiosis. This dysbiosis can activate inflammasome, further driving pyroptosis and creating a feedback loop that intensifies IBS symptoms.68,69 Moreover, dysregulation of inflammasomes may contribute to abnormal immune responses and gut inflammation in IBS. Studies have shown that pyroptosis can also compromise the integrity of intestinal epithelial cells by increasing intestinal permeability, allowing harmful substances to enter the gut and trigger inflammation. The release of cytokines such as IL-1β and IL-18 during pyroptosis has been associated with intestinal discomfort and pain.64 Moreover, infectious gastroenteritis is recognized as a significant risk factor for the onset of IBS.70 Such infections can induce pyroptosis, increasing inflammatory responses and subsequently disrupting gut function.71,72 Current research has provided preliminary insights into the association between pyroptosis and IBS. However, inconsistencies remain regarding the specific role of pyroptosis in IBS pathogenesis. This highlights the need for further targeted studies to clarify the relationship between pyroptosis, gut microbiota, and IBS. Investigating this association could pave the way for innovative therapeutic approaches. Modulating pyroptosis pathways may help alleviate IBS symptoms, and interventions targeting pyroptosis—such as anti-inflammatory drugs or agents that regulate gut microbiota—offer promising new perspectives for IBS treatment.
Pyroptosis in Chronic Infectious Enteritis
Pyroptosis plays a significant role in the progression of chronic infectious enteritis.73 Research indicates that patients with chronic infectious enteritis show elevated inflammasome activity in the gut, leading to increased pyroptosis. This process serves both as a defense mechanism against persistent infection and an adaptive response to gut microbial dysbiosis. Pyroptosis may affect the progression of chronic infectious enteritis through several mechanisms: 1) Inhibition of Pathogen Replication: Infected intestinal epithelial cells undergoing pyroptosis can release inflammatory factors that improve local immune responses, directly eliminate infected cells, and inhibit bacterial replication and spread within the gut. This mechanism helps to control infection and modulate inflammation.74 2) Modulation of Gut Microbiota: Pyroptosis can alter the composition of the gut microbiota, which in turn affects immune responses.1 A balanced microbiota competes with pathogens, reducing inflammation and preventing persistent infections.3 3) Pyroptosis contributes to gut self-repair mechanisms. The clearance of damaged cells creates space for new cell regeneration, facilitating tissue healing. However, excessive pyroptosis can lead to excessive inflammation, worsening enteritis symptoms.75 However, excessive pyroptosis can exacerbate inflammation, worsening enteritis symptoms.76 Studies have shown that gut microbial infections can induce pyroptosis, which clears infected cells and intensifies local inflammation.77 The interaction between microbial infection and pyroptosis is considered a key factor in the pathogenesis of chronic infectious enteritis.23,73 Some bacteria, such as Helicobacter pylori,78 can evade host immune surveillance by inducing pyroptosis. This allows them to maintain chronic inflammation within the host,79 leading to gut tissue damage and gastrointestinal dysfunction.80 The Gasdermin family, particularly GSDMD, plays a key role in defending intestinal epithelial cells against bacterial infections and regulating intestinal inflammation.
The colonic mucus layer serves as both a habitat for symbiotic bacteria and a physical barrier against pathogens such as E. coli. In intestinal epithelial cells, GSDMD deficiency leads to reduced mucus secretion, loss of the mucus layer, and significant changes in the spatial distribution and composition of the gut microbiota.81 GSDMD-deficient mice are more susceptible to gut pathogen infections, highlighting the importance of GSDMD in maintaining gut integrity. In chronic infectious enteritis, pyroptosis triggers local inflammatory responses and may impact systemic immune status. Caspase-driven damage signals activate various inflammatory pathways, contributing to chronic infectious enteritis.23 Persistent pathogen stimulation and immune activation make pyroptosis a defining feature of the disease. Chronic inflammation, coupled with ongoing tissue regeneration, can impair gut barrier function and promote microbial dysbiosis. This dysbiosis, in turn, exacerbates inflammation and may lead to severe complications such as intestinal perforation and sepsis. Thus, understanding the mechanisms of pyroptosis offers valuable insights into the pathogenesis of chronic infectious enteritis and related inflammatory conditions.
Pyroptosis in CRC
CRC is a complex disease with high incidence and mortality rates, influenced by genetic, environmental, and immune factors. It is the third most common cancer globally, driven by aging populations and deteriorating environmental conditions.82 CRC often develops in conjunction with other inflammatory intestinal disorders.83 Recent studies have shown that pyroptosis plays a dual role in CRC, inhibiting tumor cell growth and metastasis and potentially promoting tumor progression through chronic inflammation.84,85 In the tumor microenvironment (TME), pyroptosis significantly impacts immune regulation by releasing IL-1β and IL-18, which increase the interaction between innate and adaptive immunity.86 This process recruits immune cells, such as natural killer (NK) cells and cytotoxic T lymphocytes (CTLs), to the tumor site, thereby boosting tumor immune surveillance.87 Moreover, tumor-associated macrophages (TAMs) can also induce pyroptosis in CRC cells via the secretion of pro-inflammatory factors, though, in some cases, this may paradoxically support tumor survival and proliferation.88 Moreover, in CRC patients, pyroptosis-related genes (PRGs) are strongly correlated with disease prognosis.85,88 One key pyroptosis protein in CRC is GSDME. The methylation status of GSDME is emerging as a potential biomarker for CRC, offering opportunities for early diagnosis and disease monitoring.84 Genetic, mutational, and environmental factors influence the expression of human inflammasome signaling and pyroptosis-related genes, which in turn regulate inflammasome activation. In CRC cells, caspase-3 activation cleaves GSDME, releasing an N-terminal fragment that forms membrane pores, triggering pyroptosis. While GSDME is typically expressed in various cell types, it is often silenced in tumor cells. However, GSDME expression can shift tumor cells from apoptosis induced by tumor necrosis factor-alpha (TNF-α) and cycloheximide to pyroptosis.89 The absence of pyroptosis-related proteins in tumor cells, such as GSDME, is associated with decreased levels of perforin and granzyme B, as well as reduced infiltration of NK cells and CTLs in the TME. This suggests that a lack of pyroptosis can change the immune landscape and impact tumor progression.8 Furthermore, single nucleotide polymorphisms in the NLRP3 gene have been associated with decreased survival rates in invasive CRC patients.90 Research highlights the role of pyroptosis-related genes, including NLRP3, AIM2, and Gasdermin M5 (GSDM5), in the pyroptosis signaling pathways of various cancers, such as gastric cancer, colitis-associated CRC, and esophageal cancer. Inflammasomes, key regulators of mucosal innate immune responses, influence intestinal pathogen infections and inflammation-driven tumorigenesis.91 Deficiency in NLRP3 inflammasomes can impair NK cell IL-18 signaling pathways, promoting CRC cell growth and metastasis. Mice lacking NLRP3 or caspase-1 show increased susceptibility to inflammation-induced colon tumors.92 Similarly, mice deficient in Asc and caspase-1 are more prone to colitis and colitis-associated cancer,93 demonstrating the protective role of inflammasome in CRC-related inflammation models. From a therapeutic perspective, certain Food and Drug Administration-approved chemotherapeutic drugs and natural compounds have been shown to induce pyroptosis in tumor cells. For instance, 5-fluorouracil (5-FU)94 and programmed death-ligand 1 (PD-L1)2 inhibitors enhance drug efficacy and reduce resistance by promoting pyroptosis by activating caspase-3 and caspase-1. Moreover, translocator protein (TSPO)-targeted photodynamic therapy (PDT) has shown promise in inhibiting CRC progression by inducing pyroptosis via photosensitizers that generate reactive oxygen species under specific light wavelengths.95 Emerging advancements in nanotechnology96 and gene editing97,98 are being tailored to patient-specific genetic profiles and tumor characteristics to improve treatment outcomes. Overall, pyroptosis is critical in the onset and progression of CRC, representing a promising therapeutic target. Further research into the mechanisms of pyroptosis and its clinical applications is expected to improve CRC prognosis and provide novel treatment strategies.
Connection Between Gut Microbiota and Pyroptosis
The gut microbiota, a diverse microbial community within the gastrointestinal tract, plays a crucial role in host health and disease. Its composition is intricately associated with pyroptosis. Dysbiosis, or an imbalance in gut microbiota, can lead to the production of abnormal metabolic products that modulate inflammation and pyroptosis.99 Certain beneficial microbes, such as specific strains of Lactobacilli and Bifidobacteria, can inhibit inflammasome activation and reduce pyroptosis by producing metabolites like short-chain fatty acids.100 However, some pathogenic bacteria, such as toxin-producing Clostridium perfringens, can trigger pyroptosis by activating the NLRP3 inflammasome, compromising the integrity of the gut barrier and exacerbating inflammation.101,102 Pyroptosis plays a direct role in shaping the diversity and composition of the gut microbiota and is closely associated with inflammasomes in gut pathology. For example, the nucleotide-binding domain and leucine-rich repeat protein 6 (NLRP6) inflammasome significantly influence gut microbiota composition, epithelial cell function, and susceptibility to gastrointestinal inflammation, infections, and tumors.103 The NLRP3 inflammasome has been shown to impact oxidative stress by altering the gut microbiota, specifically targeting bacteria such as Bacteroidetes and Lactobacillus species, including Lactobacillus reuteri.104 Similarly, the AIM2 inflammasome helps regulate the gut microbiota and prevent dysbiosis and inflammation by managing the IL-18/IL-22BP/IL-22 and signal transducer and activator of transcription 3 (STAT3) pathways, along with the expression of specific antimicrobial peptides.105 This complex interplay between pyroptosis and gut microbiota is especially relevant in chronic intestinal diseases such as IBD and CRC.37 In IBD, pyroptosis is significantly elevated, leading to gut microbiota imbalances, extensive epithelial cell death, gut barrier destruction, and a perpetuating cycle of inflammation. Studies in animal models have shown that the nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing protein 1 (NLRP1) inflammasome exacerbates colitis through interactions with commensal microbes, reducing populations of butyrate-producing bacteria via IL-18-induced antimicrobial peptides. This disruption further compromises gut barrier function and perpetuates inflammation.106 In CRC, pyroptosis plays a dual role in both anti-tumor immunity and tumor progression. On the one hand, inducing pyroptosis in CRC cells can inhibit tumor growth by increasing immune surveillance. On the other hand, chronic inflammatory responses within the TME may support tumor progression. Fusobacterium nucleatum, a bacterium associated with CRC, has been shown to affect CASP6 expression, which is linked to resistance to 5-FU chemotherapy. This suggests that microbial interactions may affect CRC progression through mechanisms involving pyroptosis. Therefore, understanding the complex feedback loops between pyroptosis and the gut microbiota is essential for elucidating the pathogenesis of chronic intestinal diseases.
Conclusion and Future Research Perspectives
As research on pyroptosis in chronic intestinal diseases advances, the development of diagnostic and therapeutic strategies targeting pyroptosis has become a growing priority. This review provides a comprehensive analysis of pyroptosis across multiple chronic intestinal diseases, emphasizing its dual role in both host defense and inflammation. This review incorporates the latest findings on pyroptosis mechanisms, particularly the roles of inflammasomes and Gasdermin proteins, exploring the complex interaction between pyroptosis and gut microbiota and highlighting how dysbiosis can modulate inflammation. Moreover, it discussed the potential of targeting pyroptosis as a therapeutic strategy and recent advancements in pyroptosis inhibitors, which could revolutionize disease management.
Recent research has focused on various preclinical intervention strategies to modulate pyroptosis as a therapeutic target in chronic intestinal diseases, such as small-molecule inhibitors targeting components of the NLRP3 inflammasome, herbal medicine inhibiting NLRP3 activation, and targeting Gasdermin proteins.14 While preclinical studies have shown promise, clinical trials specifically investigating these interventions in chronic intestinal conditions are still emerging. Ongoing studies on NLRP3 inflammasome inhibitors may provide insights into their efficacy in treating IBD and related disorders.107 However, comprehensive clinical data remain limited, requiring further research.
Despite the comprehensive overview of pyroptosis and its role in chronic intestinal diseases, several challenges remain. First, much of the current understanding is based on preclinical studies with limited clinical validation. More extensive clinical research is needed to fully elucidate the therapeutic potential of targeting pyroptosis. Second, the interaction between pyroptosis and gut microbiota is highly complex, and the precise contributions of specific microbial species to pyroptosis-driven pathology require further investigation. Lastly, while the manuscript provides insights into potential therapeutic strategies, the feasibility, specificity, and long-term safety of these interventions in clinical settings have not been fully evaluated.
Future research should focus on developing novel treatments that target inflammasomes or the Gasdermin family of proteins to reduce inflammation in chronic intestinal conditions. For instance, selectively inducing pyroptosis in CRC cells while sparing normal cells could enhance anti-tumor immune responses and improve the efficacy of existing cancer therapies.23 However, the potential limitations of targeting pyroptosis must be carefully considered. The role of pyroptosis in chronic intestinal diseases is intertwined with immune responses and gut microbiota, and its impact extends across various cell types and molecular pathways, making it challenging to target specifically without impacting other physiological processes. Targeting pyroptosis non-specifically could inadvertently impair immune defense mechanisms against infections or cancer. Furthermore, therapeutic responses may vary due to genetic factors and individual microbiome compositions, requiring personalized treatment approaches. Therefore, a deeper understanding of pyroptosis is important for developing innovative diagnostic tools and therapeutic strategies that minimize adverse effects while maximizing clinical benefits. Continued efforts in this field hold the potential to open new therapeutic avenues, ultimately improving the management of chronic intestinal diseases and CRC.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This research was supported by a grant from National Natural Science Foundation of China (No. 82274475), and 2024 Zhejiang Chinese Medicine University Postgraduate Scientific Research Fund Project (No. 2024YKJ12), and Open Fund of Jinhua Municipal Science and Technology Bureau Key Project (No. 2023-3-091), and the Postdoctoral Fellowship Program of CPSF (GZB20230665), the China Postdoctoral Science Foundation (2024M752911).
Disclosure
The authors declare that they have no competing interests in this work.
References
1. Yu P, Zhang X, Liu N, Tang L, Peng C, Chen X. Pyroptosis: mechanisms and diseases. Signal Transduct Target Ther. 2021;6(1):128. doi:10.1038/s41392-021-00507-5
2. Lu L, Zhang Y, Tan X, et al. Emerging mechanisms of pyroptosis and its therapeutic strategy in cancer. Cell Death Discov. 2022;8(1):338. doi:10.1038/s41420-022-01101-6
3. Rao Z, Zhu Y, Yang P, et al. Pyroptosis in inflammatory diseases and cancer. Theranostics. 2022;12(9):4310–4329. doi:10.7150/thno.71086
4. Elias EE, Lyons B, Muruve DA. Gasdermins and pyroptosis in the kidney. Nat Rev Nephrol. 2023;19(5):337–350. doi:10.1038/s41581-022-00662-0
5. Vande Walle L, Lamkanfi M. Drugging the NLRP3 inflammasome: from signalling mechanisms to therapeutic targets. Nat Rev Drug Discov. 2024;23(1):43–66. doi:10.1038/s41573-023-00822-2
6. Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25(3):486–541. doi:10.1038/s41418-017-0012-4
7. Hou J, Hsu JM, Hung MC. Molecular mechanisms and functions of pyroptosis in inflammation and antitumor immunity. Mol Cell. 2021;81(22):4579–4590. doi:10.1016/j.molcel.2021.09.003
8. Hsu SK, Li CY, Lin IL, et al. Inflammation-related pyroptosis, a novel programmed cell death pathway, and its crosstalk with immune therapy in cancer treatment. Theranostics. 2021;11(18):8813–8835. doi:10.7150/thno.62521
9. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821–832. doi:10.1016/j.cell.2010.01.040
10. Anderson MJ, den Hartigh AB, Fink SL. Molecular mechanisms of pyroptosis. Methods mol Biol. 2023;2641:1–16.
11. Yuan J, Ofengeim D. A guide to cell death pathways. Nat Rev mol Cell Biol. 2024;25(5):379–395. doi:10.1038/s41580-023-00689-6
12. den Hartigh AB, Fink SL. Pyroptosis induction and detection. Curr Protoc Immunol. 2018;122(1):e52. doi:10.1002/cpim.52
13. Chen M, Rong R, Xia X. Spotlight on pyroptosis: role in pathogenesis and therapeutic potential of ocular diseases. J Neuroinflammation. 2022;19(1):183. doi:10.1186/s12974-022-02547-2
14. Wei S, Feng M, Zhang S. Molecular characteristics of cell pyroptosis and its inhibitors: a review of activation, regulation, and inhibitors. Int J mol Sci. 2022;23(24):16115. doi:10.3390/ijms232416115
15. Yuan J, Najafov A, Py BF. Roles of Caspases in Necrotic Cell Death. Cell. 2016;167(7):1693–1704. doi:10.1016/j.cell.2016.11.047
16. Gong W, Yang K, Zhao W, et al. Intestinal Gasdermins for regulation of inflammation and tumorigenesis. Front Immunol. 2022;13:1052111. doi:10.3389/fimmu.2022.1052111
17. Privitera G, Rana N, Armuzzi A, Pizarro TT. The gasdermin protein family: emerging roles in gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol. 2023;20(6):366–387. doi:10.1038/s41575-023-00743-w
18. Barnett KC, Li S, Liang K, Ting JP. A 360 degrees view of the inflammasome: mechanisms of activation, cell death, and diseases. Cell. 2023;186(11):2288–2312. doi:10.1016/j.cell.2023.04.025
19. Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling. Nat Rev Immunol. 2016;16(7):407–420. doi:10.1038/nri.2016.58
20. Liu T, Zhang L, Joo D, Sun SC. NF-kappaB signaling in inflammation. Signal Transduct Target Ther. 2017;2(17023):17023. doi:10.1038/sigtrans.2017.23
21. Frank D, Vince JE. Pyroptosis versus necroptosis: similarities, differences, and crosstalk. Cell Death Differ. 2019;26(1):99–114. doi:10.1038/s41418-018-0212-6
22. Choudhury SM, Sarkar R, Karki R, Kanneganti TD. A comparative study of apoptosis, pyroptosis, necroptosis, and PANoptosis components in mouse and human cells. PLoS One. 2024;19(2):e0299577. doi:10.1371/journal.pone.0299577
23. Zhang S, Liang Y, Yao J, Li DF, Wang LS. Role of pyroptosis in inflammatory Bowel Disease (IBD): from Gasdermins to DAMPs. Front Pharmacol. 2022;13:833588. doi:10.3389/fphar.2022.833588
24. Chey WD, Kurlander J, Eswaran S. Irritable bowel syndrome: a clinical review. JAMA. 2015;313(9):949–958. doi:10.1001/jama.2015.0954
25. Holtmeier W, Caspary WF. Celiac disease. Orphanet J Rare Dis. 2006;1:3. doi:10.1186/1750-1172-1-3
26. Lenti MV, Di Sabatino A. Intestinal fibrosis. mol Aspects Med. 2019;65:100–109. doi:10.1016/j.mam.2018.10.003
27. Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat Rev Gastroenterol Hepatol. 2015;12(12):720–727. doi:10.1038/nrgastro.2015.150
28. Ananthakrishnan AN, Kaplan GG, Ng SC. Changing global epidemiology of inflammatory bowel diseases: sustaining health care delivery into the 21st Century. Clin Gastroenterol Hepatol. 2020;18(6):1252–1260. doi:10.1016/j.cgh.2020.01.028
29. Munyaka PM, Sepehri S, Ghia JE, Khafipour E. Carrageenan Gum and Adherent Invasive Escherichia coli in a Piglet Model of inflammatory bowel disease: impact on Intestinal mucosa-associated microbiota. Front Microbiol. 2016;7:462. doi:10.3389/fmicb.2016.00462
30. Wang HH, He Y, Wang HX, et al. Comparison of TPMT and NUDT15 polymorphisms in Chinese patients with inflammatory bowel disease. World J Gastroenterol. 2018;24(8):941–948. doi:10.3748/wjg.v24.i8.941
31. Yu Y, Yang W, Li Y, Cong Y. Enteroendocrine cells: sensing gut microbiota and regulating inflammatory Bowel Diseases. Inflamm Bowel Dis. 2020;26(1):11–20. doi:10.1093/ibd/izz217
32. Galipeau HJ, Caminero A, Turpin W, et al. Novel fecal biomarkers that precede clinical diagnosis of ulcerative colitis. Gastroenterology. 2021;160(5):1532–1545. doi:10.1053/j.gastro.2020.12.004
33. Negroni A, Colantoni E, Cucchiara S, Stronati L. Necroptosis in intestinal inflammation and cancer: new concepts and therapeutic perspectives. Biomolecules. 2020;10(10):1431. doi:10.3390/biom10101431
34. Li Z, Zhu Z, Zhou D, et al. Inhibition of transmural inflammation in Crohn’s disease by orally administered tumor necrosis factor-alpha deoxyribozymes-loaded pyroptosis nanoinhibitors. ACS Appl Mater Interfaces. 2024;16(31):40499–40514. doi:10.1021/acsami.4c05921
35. Bertheloot D, Latz E, Franklin BS. Necroptosis, pyroptosis and apoptosis: an intricate game of cell death. Cell mol Immunol. 2021;18(5):1106–1121.
36. Bauer C, Duewell P, Lehr HA, Endres S, Schnurr M. Protective and aggravating effects of Nlrp3 inflammasome activation in IBD models: influence of genetic and environmental factors. Dig Dis. 2012;30(Suppl 1):82–90. doi:10.1159/000341681
37. Bao Z, Deng Y, Qian Z, Zhuang Y. Novel insights into the interaction between Enteropathogenic Bacteria, pyroptosis and IBD. Front Biosci. 2024;29(7):254. doi:10.31083/j.fbl2907254
38. Yuan YY, Xie KX, Wang SL, Yuan LW. Inflammatory caspase-related pyroptosis: mechanism, regulation and therapeutic potential for inflammatory bowel disease. Gastroenterol Rep. 2018;6(3):167–176. doi:10.1093/gastro/goy011
39. Rieder F, Mukherjee PK, Massey WJ, Wang Y, Fiocchi C. Fibrosis in IBD: from pathogenesis to therapeutic targets. Gut. 2024;73(5):854–866. doi:10.1136/gutjnl-2023-329963
40. Wang Y, Huang B, Jin T, Ocansey DKW, Jiang J, Mao F. Intestinal fibrosis in inflammatory bowel disease and the prospects of mesenchymal stem cell therapy. Front Immunol. 2022;13:835005. doi:10.3389/fimmu.2022.835005
41. Santacroce G, Lenti MV, Di Sabatino A. Therapeutic targeting of intestinal fibrosis in Crohn’s disease. Cells. 2022;11(3):429. doi:10.3390/cells11030429
42. Suetomi T, Willeford A, Brand CS, et al. Inflammation and NLRP3 inflammasome activation initiated in response to pressure overload by Ca(2+)/Calmodulin-Dependent Protein Kinase II delta Signaling in Cardiomyocytes Are Essential for Adverse Cardiac Remodeling. Circulation. 2018;138(22):2530–2544. doi:10.1161/CIRCULATIONAHA.118.034621
43. Byrne NJ, Soni S, Takahara S, et al. Chronically elevating circulating ketones can reduce cardiac inflammation and blunt the development of heart failure. Circ Heart Fail. 2020;13(6):e006573. doi:10.1161/CIRCHEARTFAILURE.119.006573
44. Cai SM, Yang RQ, Li Y, et al. Angiotensin-(1-7) improves liver fibrosis by regulating the NLRP3 inflammasome via redox balance modulation. Antioxid Redox Signal. 2016;24(14):795–812. doi:10.1089/ars.2015.6498
45. Anders HJ, Suarez-Alvarez B, Grigorescu M, et al. The macrophage phenotype and inflammasome component NLRP3 contributes to nephrocalcinosis-related chronic kidney disease independent from IL-1-mediated tissue injury. Kidney Int. 2018;93(3):656–669. doi:10.1016/j.kint.2017.09.022
46. Liu Y, Zhang T, Pan K, Wei H. Mechanisms and therapeutic research progress in intestinal fibrosis. Front Med. 2024;11:1368977. doi:10.3389/fmed.2024.1368977
47. Song Z, Gong Q, Guo J. Pyroptosis: mechanisms and Links with Fibrosis. Cells. 2021;10(12):3509. doi:10.3390/cells10123509
48. Chai R, Li Y, Shui L, Ni L, Zhang A. The role of pyroptosis in inflammatory diseases. Front Cell Dev Biol. 2023;11:1173235. doi:10.3389/fcell.2023.1173235
49. Li Y, Yuan Y, Huang ZX, et al. GSDME-mediated pyroptosis promotes inflammation and fibrosis in obstructive nephropathy. Cell Death Differ. 2021;28(8):2333–2350. doi:10.1038/s41418-021-00755-6
50. Lyu T, Yin Q. Research progress on pyroptosis in hematological malignancies. Curr Treat Options Oncol. 2023;24(10):1439–1450. doi:10.1007/s11864-023-01119-7
51. Koh EH, Yoon JE, Ko MS, et al. Sphingomyelin synthase 1 mediates hepatocyte pyroptosis to trigger non-alcoholic steatohepatitis. Gut. 2021;70(10):1954–1964. doi:10.1136/gutjnl-2020-322509
52. Komada T, Muruve DA. The role of inflammasomes in kidney disease. Nat Rev Nephrol. 2019;15(8):501–520. doi:10.1038/s41581-019-0158-z
53. Sun B, Wang X, Liao YP, et al. Repetitive dosing of fumed silica leads to profibrogenic effects through unique structure-activity relationships and biopersistence in the lung. ACS Nano. 2016;10(8):8054–8066. doi:10.1021/acsnano.6b04143
54. Catassi C, Verdu EF, Bai JC, Lionetti E. Coeliac disease. Lancet. 2022;399(10344):2413–2426. doi:10.1016/S0140-6736(22)00794-2
55. Lebwohl B, Sanders DS, Green PHR. Coeliac disease. Lancet. 2018;391(10115):70–81. doi:10.1016/S0140-6736(17)31796-8
56. Ruera CN, Perez F, Iribarren ML, et al. Coexistence of apoptosis, pyroptosis, and necroptosis pathways in celiac disease. Clin Exp Immunol. 2023;214(3):328–340. doi:10.1093/cei/uxad082
57. Chen S, Liu X, Wang Z, et al. Transcriptome profile and immune infiltrated landscape revealed a novel role of gammadeltaT cells in mediating pyroptosis in celiac disease. J Transl Med. 2023;21(1):497. doi:10.1186/s12967-023-04359-1
58. Perez F, Ruera CN, Miculan E, Carasi P, Chirdo FG. Programmed cell death in the small intestine: implications for the pathogenesis of celiac disease. Int J mol Sci. 2021;22(14):7426. doi:10.3390/ijms22147426
59. Choung RS, Larson SA, Khaleghi S, et al. Prevalence and morbidity of undiagnosed celiac disease from a community-based study. Gastroenterology. 2017;152(4):830–839e835. doi:10.1053/j.gastro.2016.11.043
60. Piscopo N, Ellul P. Diverticular disease: a review on pathophysiology and recent evidence. Ulster Med J. 2020;89(2):83–88. doi:10.1136/bmj.d4131
61. Peery AF, Shaukat A, Strate LL. AGA clinical practice update on medical management of colonic diverticulitis: expert review. Gastroenterology. 2021;160(3):906–911e901. doi:10.1053/j.gastro.2020.09.059
62. Piccioni A, Franza L, Brigida M, et al. Gut microbiota and acute diverticulitis: role of probiotics in management of this delicate pathophysiological balance. J Pers Med. 2021;11(4):298. doi:10.3390/jpm11040298
63. Chao L, Zhang W, Feng Y, Gao P, Ma J. Pyroptosis: a new insight into intestinal inflammation and cancer. Front Immunol. 2024;15:1364911. doi:10.3389/fimmu.2024.1364911
64. Ford AC, Sperber AD, Corsetti M, Camilleri M. Irritable bowel syndrome. Lancet. 2020;396(10263):1675–1688. doi:10.1016/S0140-6736(20)31548-8
65. Canavan C, West J, Card T. The epidemiology of irritable bowel syndrome. Clin Epidemiol. 2014;6:71–80. doi:10.2147/CLEP.S40245
66. Huang KY, Wang FY, Lv M, Ma XX, Tang XD, Lv L. Irritable bowel syndrome: epidemiology, overlap disorders, pathophysiology and treatment. World J Gastroenterol. 2023;29(26):4120–4135. doi:10.3748/wjg.v29.i26.4120
67. Bonetto S, Fagoonee S, Battaglia E, Grassini M, Saracco GM, Pellicano R. Recent advances in the treatment of irritable bowel syndrome. Pol Arch Intern Med. 2021;131(7–8):709–715. doi:10.20452/pamw.16067
68. Dothel G, Barbaro MR, Di Vito A, et al. New insights into irritable bowel syndrome pathophysiological mechanisms: contribution of epigenetics. J Gastroenterol. 2023;58(7):605–621. doi:10.1007/s00535-023-01997-6
69. Shrestha B, Patel D, Shah H, et al. The role of gut-microbiota in the pathophysiology and therapy of irritable bowel syndrome: a systematic review. Cureus. 2022;14(8):e28064. doi:10.7759/cureus.28064
70. Kanazawa M, Fukudo S. Relationship between infectious gastroenteritis and irritable bowel syndrome. Clin J Gastroenterol. 2014;7(1):14–18. doi:10.1007/s12328-013-0444-4
71. Barbara G, Cremon C, Carini G, et al. The immune system in irritable bowel syndrome. J Neurogastroenterol Motil. 2011;17(4):349–359. doi:10.5056/jnm.2011.17.4.349
72. Mayer EA, Ryu HJ, Bhatt RR. The neurobiology of irritable bowel syndrome. mol Psychiatry. 2023;28(4):1451–1465. doi:10.1038/s41380-023-01972-w
73. Xiao C, Cao S, Li Y, et al. Pyroptosis in microbial infectious diseases. Mol Biol Rep. 2023;51(1):42. doi:10.1007/s11033-023-09078-w
74. Li ZH, Wang Y, Yu XY. Exploring the role of pyroptosis and immune infiltration in sepsis based on bioinformatic analysis. Immunobiology. 2024;229(5):152826. doi:10.1016/j.imbio.2024.152826
75. Huang H, Weng Y, Tian W, Lin X, Chen J, Luo L. Molecular mechanisms of pyroptosis and its role in anti-tumor immunity. Int J Biol Sci. 2023;19(13):4166–4180. doi:10.7150/ijbs.86855
76. Loveless R, Bloomquist R, Teng Y. Pyroptosis at the forefront of anticancer immunity. J Exp Clin Cancer Res. 2021;40(1):264. doi:10.1186/s13046-021-02065-8
77. Guan Q. A comprehensive review and update on the pathogenesis of inflammatory bowel disease. J Immunol Res. 2019;2019:7247238. doi:10.1155/2019/7247238
78. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7(2):99–109. doi:10.1038/nrmicro2070
79. Wu Y, Zhang J, Yu S, et al. Cell pyroptosis in health and inflammatory diseases. Cell Death Discov. 2022;8(1):191. doi:10.1038/s41420-022-00998-3
80. Wei Y, Yang L, Pandeya A, Cui J, Zhang Y, Li Z. Pyroptosis-induced inflammation and tissue damage. J mol Biol. 2022;434(4):167301. doi:10.1016/j.jmb.2021.167301
81. Zhang J, Yu Q, Jiang D, et al. Epithelial Gasdermin D shapes the host-microbial interface by driving mucus layer formation. Sci Immunol. 2022;7(68):eabk2092. doi:10.1126/sciimmunol.abk2092
82. Marmol I, Sanchez-de-Diego C, Pradilla Dieste A, Cerrada E, Rodriguez Yoldi MJ. Colorectal carcinoma: a general overview and future perspectives in colorectal cancer. Int J mol Sci. 2017;18(1):197.
83. Wu J, Li Q, Fu X. Fusobacterium nucleatum contributes to the carcinogenesis of colorectal cancer by inducing inflammation and suppressing host immunity. Transl Oncol. 2019;12(6):846–851. doi:10.1016/j.tranon.2019.03.003
84. You HM, Wang L, Meng HW, Huang C, Fang GY, Li J. Pyroptosis: shedding light on the mechanisms and links with cancers. Front Immunol. 2023;14:1290885. doi:10.3389/fimmu.2023.1290885
85. Lu H, Sun Y, Zhu Z, et al. Pyroptosis is related to immune infiltration and predictive for survival of colon adenocarcinoma patients. Sci Rep. 2022;12(1):9233. doi:10.1038/s41598-022-13212-2
86. Fang Y, Tian S, Pan Y, et al. Pyroptosis: a new frontier in cancer. Biomed Pharmacother. 2020;121:109595. doi:10.1016/j.biopha.2019.109595
87. Hu M, Deng F, Song X, Zhao H, Yan F. The crosstalk between immune cells and tumor pyroptosis: advancing cancer immunotherapy strategies. J Exp Clin Cancer Res. 2024;43(1):190. doi:10.1186/s13046-024-03115-7
88. Zhou J, Guo H, Liu L, Feng M, Yang X, Hao S. Pyroptosis patterns of colon cancer could aid to estimate prognosis, microenvironment and immunotherapy: evidence from multi-omics analysis. Aging. 2022;14(18):7547–7567. doi:10.18632/aging.204302
89. Wang Y, Gao W, Shi X, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 2017;547(7661):99–103. doi:10.1038/nature22393
90. Ungerback J, Belenki D, Jawad ul-Hassan A, et al. Genetic variation and alterations of genes involved in NFkappaB/TNFAIP3- and NLRP3-inflammasome signaling affect susceptibility and outcome of colorectal cancer. Carcinogenesis. 2012;33(11):2126–2134. doi:10.1093/carcin/bgs256
91. Zhou CB, Fang JY. The role of pyroptosis in gastrointestinal cancer and immune responses to intestinal microbial infection. Biochim Biophys Acta Rev Cancer. 2019;1872(1):1–10. doi:10.1016/j.bbcan.2019.05.001
92. Zaki MH, Vogel P, Body-Malapel M, Lamkanfi M, Kanneganti TD. IL-18 production downstream of the Nlrp3 inflammasome confers protection against colorectal tumor formation. J Immunol. 2010;185(8):4912–4920. doi:10.4049/jimmunol.1002046
93. Dupaul-Chicoine J, Yeretssian G, Doiron K, et al. Control of intestinal homeostasis, colitis, and colitis-associated colorectal cancer by the inflammatory caspases. Immunity. 2010;32(3):367–378. doi:10.1016/j.immuni.2010.02.012
94. Xia X, Wang X, Cheng Z, et al. The role of pyroptosis in cancer: pro-cancer or pro-“host”? Cell Death Dis. 2019;10(9):650. doi:10.1038/s41419-019-1883-8
95. Zhou Y, Zhang W, Wang B, et al. Mitochondria-targeted photodynamic therapy triggers GSDME-mediated pyroptosis and sensitizes anti-PD-1 therapy in colorectal cancer. J Immunother Cancer. 2024;12(3):e008054. doi:10.1136/jitc-2023-008054
96. Yin Q, Song SY, Bian Y, et al. Unlocking the potential of pyroptosis in tumor immunotherapy: a new horizon in cancer treatment. Front Immunol. 2024;15:1381778. doi:10.3389/fimmu.2024.1381778
97. Liu J, Chen T, Liu X, Li Z, Zhang Y. Engineering materials for pyroptosis induction in cancer treatment. Bioact Mater. 2024;33:30–45. doi:10.1016/j.bioactmat.2023.10.027
98. Liu SW, Song WJ, Ma GK, Wang H, Yang L. Pyroptosis and its role in cancer. World J Clin Cases. 2023;11(11):2386–2395. doi:10.12998/wjcc.v11.i11.2386
99. Ye L, Chen H, Wang J, et al. Aflatoxin B(1)-induced liver pyroptosis is mediated by disturbing the gut microbial metabolites: the roles of pipecolic acid and norepinephrine. J Hazard Mater. 2024;474:134822. doi:10.1016/j.jhazmat.2024.134822
100. Ji X, Chen M, Zhao M, Song Y, Zhao L. Effects of chitooligosaccharides on the rebalance of gut microorganisms and their metabolites in patients with nonalcoholic fatty liver disease. Journal of Functional Foods. 2021;77(4):104333. doi:10.1016/j.jff.2020.104333
101. Mathur A, Kay C, Xue Y, et al. Clostridium perfringens virulence factors are nonredundant activators of the NLRP3 inflammasome. EMBO Rep. 2023;24(6):e54600. doi:10.15252/embr.202254600
102. Liu Z, Mou S, Li L, et al. The barrier disruption and pyroptosis of intestinal epithelial cells caused by Perfringolysin O (PFO) from Clostridium perfringens. Cells. 2024;13(13):1140. doi:10.3390/cells13131140
103. Zheng D, Kern L, Elinav E. The NLRP6 inflammasome. Immunology. 2021;162(3):281–289. doi:10.1111/imm.13293
104. Li Z, Liu Y, Lin B, et al. Pyroptosis-related signature as potential biomarkers for predicting prognosis and therapy response in colorectal cancer patients. Front Genet. 2022;13:925338. doi:10.3389/fgene.2022.925338
105. Ratsimandresy RA, Indramohan M, Dorfleutner A, Stehlik C. The AIM2 inflammasome is a central regulator of intestinal homeostasis through the IL-18/IL-22/STAT3 pathway. Cell mol Immunol. 2017;14(1):127–142. doi:10.1038/cmi.2016.35
106. Watanabe D, Guo Y, Kamada N. Interaction between the inflammasome and commensal microorganisms in gastrointestinal health and disease. EMBO Mol Med. 2021;13(12):e13452. doi:10.15252/emmm.202013452
107. Coll RC, Schroder K, Pelegrin P. NLRP3 and pyroptosis blockers for treating inflammatory diseases. Trends Pharmacol Sci. 2022;43(8):653–668. doi:10.1016/j.tips.2022.04.003
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles
NcRNA Regulated Pyroptosis in Liver Diseases and Traditional Chinese Medicine Intervention: A Narrative Review
Deng J, Qin L, Qin S, Wu R, Huang G, Fang Y, Huang L, Zhou Z
Journal of Inflammation Research 2024, 17:2073-2088
Published Date: 3 April 2024
The Potential Therapeutic Prospect of PANoptosis in Heart Failure
Jia Y, Liu Y, Zuo Y, Zhang J, Li Y, Liu X, Lv S
Journal of Inflammation Research 2024, 17:9147-9168
Published Date: 19 November 2024