Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML

  • Supplementary Files
    • Supplementary_Data.pdf
Review Open Access

Dysregulated programmed cell death of intestinal epithelial cells in ulcerative colitis: Molecular mechanisms and novel therapeutic interventions (Review)

  • Authors:
    • Bihua Wu
    • Shuangyan Su
    • Yuwei Li
    • Le Guo
  • View Affiliations / Copyright

    Affiliations: Department of Medical Microbiology and Immunology, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671000, P.R. China
    Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 230
    |
    Published online on: October 22, 2025
       https://doi.org/10.3892/ijmm.2025.5671
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Ulcerative colitis (UC), a chronic idiopathic inflammatory bowel disease affecting the colonic mucosa, has seen a rising global incidence and poses a growing public health challenge. The clinical presentation typically includes recurrent abdominal pain, hematochezia, weight loss and structural damage to the colonic epithelium, substantially impacting patients' quality of life. Recent research has underscored the role of intestinal barrier dysfunction as a fundamental driver of persistent inflammation and disease progression in UC. Intestinal epithelial cells (IECs) form a monolayer through tight intercellular junctions and constitute the primary defense against luminal pathogens. Dysregulation of programmed cell death pathways in IECs such as apoptosis, necroptosis, pyroptosis, ferroptosis and autophagy‑related cell death compromises epithelial integrity and exacerbates inflammation. The present review systematically examines how these death pathways contribute to UC pathogenesis, highlighting the molecular mechanisms through which natural bioactive compounds and nanoparticle‑based drug delivery systems modulate them. Key signaling targets include the NF‑κB pathway, MAPK cascade, NLR family pyrin domain containing 3 inflammasome and autophagy‑related networks. By integrating advances in target identification, structure‑activity relationship optimization and mechanistic insights, the present review provides a comprehensive framework for understanding UC and facilitates the development of innovative therapeutic approaches aimed at restoring barrier function and regulating epithelial cell death.

Introduction

Ulcerative colitis (UC), a prototypical clinical subtype of inflammatory bowel disease (IBD), is characterized by chronic and recurrent non-specific inflammation primarily localized within the mucosal and submucosal layers of the rectum and colon. Surveillance data have highlighted a concerning rise in the global incidence of UC (1,2), a trend that not only severely compromises the quality of life of patients but also significantly elevates the risk of colorectal carcinogenesis (3,4). At present, conventional therapeutic agents such as 5-aminosalicylic acid, glucocorticoids and biologics (such as anti-TNF and anti-integrin agents) are commonly employed. Nevertheless, these pharmacotherapeutic interventions are hampered by notable limitations in terms of efficacy, adverse effects, individual responsiveness and a high relapse rate (5). Moreover, the economic burden and psychological impact of long-term treatment further exacerbate patient stress, underscoring the necessity for more effective and individualized therapeutic approaches. Despite extensive research efforts, the precise etiological determinants and comprehensive pathophysiological mechanisms underlying UC remain incompletely elucidated. Growing evidence accentuates the pivotal role of intestinal barrier integrity (6,7), a sophisticated, multi-layered defense system encompassing the mucus layer, epithelial cells and intercellular junction complexes (8), in both the initiation and progression of UC. Mechanistic investigations have demonstrated that UC pathogenesis involves intricate interactions among immune dysregulation, disruption of intestinal epithelial homeostasis, dysbiosis-driven bacterial colonization and compromised epithelial barrier function (9).

Pathological alterations in tight junction (TJ) proteins, such as ZO-1 and occludin, result in abnormal intestinal permeability, facilitating the translocation of luminal bacteria, endotoxins and undigested dietary antigens into the lamina propria (10). These microbial components and metabolites directly activate lamina propria immune cells, triggering the robust release of pro-inflammatory cytokines (11). This chemotactic signaling recruits additional leukocytes to the inflammatory sites, initiating a cascade that results in characteristic pathological manifestations, including tissue erythema, exudation and mucosal erosion. Notably, this self-perpetuating inflammatory milieu exacerbates the degradation of TJ proteins (12,13), establishing a detrimental cycle that perpetuates mucosal injury.

Intestinal epithelial cells (IECs) constitute the primary defense of the intestinal mucosal barrier, playing a pivotal role in maintaining mucosal homeostasis through dynamic cell turnover and selective permeability regulation. Recent investigations have identified dysregulated IEC death as a key factor contributing to the perpetuation of colonic inflammation and the resistance to therapy in UC pathogenesis (14-16). Under physiological conditions, apoptosis maintains intestinal epithelial homeostasis through programmed cell turnover. However, an excess of apoptotic activity exceeding regenerative capacity can lead to extensive depletion of IECs, causing structural denudation and compromising barrier integrity (17). In addition to apoptosis, other cell death mechanisms, including necroptosis, PANoptosis, pyroptosis, ferroptosis and autophagy, have been implicated in the pathogenesis of UC (18-22).

Therefore, elucidating the role of IEC death in UC pathogenesis and systematically consolidating current research achievements on modulating excessive IEC death through natural compounds and nanoparticle-mediated drug delivery systems to alleviate UC progression (particularly by integrating recent advances in mechanistic pathways, bioactive constituents and molecular targets) may offer novel perspectives for unraveling UC etiology. This strategy could facilitate the precise identification of potential therapeutic targets, thereby paving the way for innovative methods for clinical UC management. Future research endeavors should concentrate on developing targeted therapies that address the multifaceted nature of IEC death and its downstream inflammatory repercussions, ultimately disrupting the cycle of mucosal damage and the inflammation characteristic of UC.

Major forms of IEC death

IEC apoptosis

IECs are characterized by rapid and continuous cellular renewal. Apoptosis, a key programmed cell death mechanism, serves as an essential pathway for the physiological elimination of aged IECs at the end of their life cycle and plays a critical role in sustaining intestinal homeostasis (23). However, excessive activation of apoptosis during UC progression may amplify inflammatory responses via enhanced cytokine signaling (24). Upon exposure to stress stimuli, such as the inflammatory microenvironment of UC, pro-apoptotic proteins Bax and Bak undergo oligomerization and translocate to the mitochondrial outer membrane, forming transmembrane pores (25). This event triggers mitochondrial swelling and facilitates the efflux of cytochrome c from the intermembrane space into the cytosol (26). Released cytochrome c subsequently binds to apoptotic protease-activating factor-1, assembling the apoptosome complex that activates caspase-9 through proteolytic cleavage (27). This initiates a downstream caspase cascade culminating in the execution phase of apoptosis via caspase-3 activation and eventual cellular dismantling (28).

In the dextran sulfate sodium (DSS)-induced mouse model of UC, intestinal tissues display a pronounced state of oxidative stress, accompanied by upregulated Bax protein expression, decreased mitochondrial membrane potential, increased cytochrome c release, markedly elevated apoptosis of IECs and impaired intestinal barrier function, which manifests as increased intestinal permeability and frequent bacterial translocation (29,30). Clinical investigations have demonstrated that patients with UC exhibit significantly elevated expression levels of Fas and Fas ligand (FasL) in intestinal mucosal tissues compared with healthy controls, with these levels positively correlating with clinical severity indices (31,32). These findings collectively suggest that the Fas/FasL-mediated extrinsic apoptosis pathway is aberrantly activated during UC pathogenesis, driving excessive IEC apoptosis and thereby compromising barrier integrity. Within the colonic tissues of patients with UC, the tumor suppressor p53 functions as a pivotal transcriptional regulator orchestrating multiple apoptosis-related genes (33). Notably, enhanced phosphorylation of p53 has been shown to be positively correlated with disease severity (34). Under inflammatory conditions, interleukin (IL)-1β released from activated immune cells potentiates p53-mediated apoptotic signaling, thereby exacerbating epithelial apoptosis and amplifying mucosal inflammation (35). An experimental study employing TNF-α-induced murine models revealed augmented IEC apoptosis in both acute and chronic inflammation settings (36). This apoptotic imbalance disrupts epithelial homeostasis, thereby facilitating UC progression. Moreover, the upregulated expression of METTL3 and long non-coding RNAs such as MALAT1 under inflammatory conditions has been mechanistically linked to suppressed cellular viability and enhanced IEC apoptosis, unveiling novel molecular pathways in UC pathogenesis (37,38).

Following IEC apoptosis, multiple chemokines are released to recruit immune cells to intestinal inflammatory sites, including monocyte chemoattractant protein-1 and IL-8, a process that plays a central role in the pathogenesis of UC. During disease progression, persistent inflammatory stimuli and sustained cell death signaling maintain macrophage activation, thereby establishing a self-amplifying pathological loop (39,40). In DSS-induced murine UC models, a rapid elevation of IL-8 levels has been observed in intestinal tissues post-induction, concomitant with robust neutrophil infiltration. This pathological progression exhibits temporal synchronization with IEC apoptosis and necrosis, and these three pathological events mutually reinforce one another through feed-forward mechanisms. Collectively, this triad drives the chronicity and perpetuation of intestinal inflammation (41).

Conventional therapies have been predominantly aimed at suppressing excessive immune responses but have largely neglected emerging pathological aspects such as the restoration of intestinal mucosal barrier integrity and the correction of dysregulated IEC apoptosis, resulting in suboptimal efficacy and frequent relapses in certain patient populations (42). These limitations have catalyzed the exploration of natural compounds and novel targeted therapeutic. Ginkgetin (GK), a natural compound derived from Ginkgo biloba with multi-protective properties, has been shown to ameliorate DSS-induced experimental colitis in murine models. Mechanistic investigations reveal that GK administration attenuates IEC apoptosis through inhibition of the EGFR/PI3K/AKT signaling pathway, accompanied by upregulation of the anti-apoptotic protein Bcl-2 and downregulation of pro-apoptotic proteins Bax and caspase-3, thereby restoring intestinal barrier integrity (43). Similarly, in acetic acid-induced rat models of UC, Centella asiatica treatment effectively normalizes dysregulated apoptosis markers, including elevated Bcl-2 alongside reduced Bax and caspase-3, while mitigating oxidative stress and attenuating inflammatory responses (44). Despite consistent modulation of key biomarkers, studies on Centella asiatica often lack rigorous dose-response evaluation and thorough long-term toxicity assessments, which are essential for translational applicability. Daphnetin exerts protective effects against IEC apoptosis by inhibiting the regenerating islet-derived protein 3α-dependent Janus kinase 2 (JAK2)/STAT3 signaling pathway, resulting in reduced expression of pro-apoptotic proteins and decreased levels of pro-inflammatory cytokines (45). Both lipopolysaccharide (LPS)-treated human colon adenocarcinoma Caco-2 cell inflammatory models and DSS-induced UC rat models have demonstrated that the natural bioactive compound paeoniflorin alleviates UC by modulating serum metabolites and suppressing the CDC42/JNK signaling pathway, thereby inhibiting IEC apoptosis (46).

Nonetheless, current investigations primarily emphasize acute therapeutic efficacy, with limited assessment of remission durability or relapse prevention in chronic UC contexts. Emerging as a promising therapeutic approach, exosome-based interventions have gained attention due to their low immunogenicity and superior circulatory stability. In murine UC models, caprine milk-derived exosomes have been shown to attenuate oxidative stress, suppress apoptosis, restore intestinal barrier integrity and modulate gut microbiota composition (47). Mechanistically, oxidative stress acts as a key driver of IEC apoptosis and recent evidence indicates that mesenchymal stem cell-derived exosomes confer protective effects by mitigating ROS accumulation in IECs, thereby alleviating UC-associated tissue damage (48). While exosome therapies offer multi-modal benefits, current preclinical studies face challenges including heterogeneity in exosome isolation techniques, lack of standardized dosing regimens and limited investigation of potential off-target effects. Furthermore, the precise mechanisms underlying exosome-mediated immunomodulation, barrier restoration and concurrent suppression of inflammatory signaling and apoptotic pathways remain to be fully elucidated.

While active exploration into natural compounds and biological vectors such as exosomes continues, advances in pharmaceutical engineering and drug repurposing have concurrently unveiled innovative avenues for UC therapy. Prior investigations have substantiated that berberine (BBR), a bioactive constituent of traditional Chinese medicine, effectively mitigates DSS-induced colonic inflammation; however, its clinical translation has been hampered by inherent limitations including poor aqueous solubility and short half-life (49,50). In recent years, the advent of nanotechnology-engineered poly (lactic-co-glycolic acid) nanoparticles encapsulating BBR has markedly enhanced drug encapsulation efficiency, aqueous solubility and bioactivity, thereby conferring superior therapeutic outcomes in UC experimental models (51). Donepezil, originally developed for Alzheimer's disease management, is renowned for its neuroprotective, anti-inflammatory and antioxidant attributes (52,53). Evidence has further elucidated its capacity to upregulate low-density lipoprotein receptor-related protein 1 expression, activate AMPK signaling and suppress the NF-κB inflammatory cascade, thereby attenuating intestinal inflammation and epithelial apoptosis (54). Analogously, the small-molecule inhibitor ruxolitinib ameliorates UC pathological progression by targeting the JAK/STAT3 pathway, effectively suppressing NF-κB activation, curtailing apoptosis and fostering barrier repair, thus presenting a promising targeted therapeutic strategy (55). Notwithstanding these advancements, emerging modalities continue to confront key challenges, including unresolved optimal dosing regimens, tissue-specific targeting and sustained long-term efficacy within the intestinal milieu.

IEC necroptosis

Necroptosis, a distinct form of programmed necrotic cell death, exerts a pivotal influence on intestinal inflammation. The core molecular machinery involves sequential activation and signaling transduction of receptor-interacting protein kinase (RIPK) 1, RIPK3 and mixed lineage kinase domain-like protein (MLKL) (56). Under homeostatic conditions, RIPK1 serves as a central signaling hub that coordinates cellular stress responses and preserves epithelial integrity (57). During intestinal inflammation triggered by pathogenic stimuli or excessive TNF-α release, TNF-α engagement with tumor necrosis factor receptor 1 initiates RIPK1 oligomerization via death domain interactions, culminating in kinase activation (58). RIPK1 is subsequently incorporated into a complex containing Fas-associated death domain (FADD) and caspase-8 (complex IIa), in which caspase-8 acts as a critical molecular switch. Upon activation, caspase-8 cleaves RIPK1 and RIPK3, thereby promoting apoptotic cell death while concurrently suppressing necroptosis initiation (59). By contrast, when caspase-8 activity is compromised (for example, by genetic deletion or pharmacological inhibition) RIPK1 interacts with RIPK3 to facilitate formation of the RIPK1-RIPK3 complex, known as the necrosome (60-62). Activated RIPK3 phosphorylates MLKL, converting it from an inactive monomer to a functional oligomeric state (63). The phosphorylated MLKL oligomers acquire membrane-targeting capability through conformational changes and subsequently translocate to the plasma membrane, ultimately leading to cellular swelling, membrane rupture and necroptosis. The resultant released of damage-associated molecular patterns (DAMPs), including bioactive molecules such as high mobility group box 1 (HMGB1) and ATP, have been demonstrated to exert immunomodulatory effects (64). Studies have demonstrated that selective deficiency or functional impairment of caspase-8 in the intestinal epithelium markedly increases susceptibility to necroptosis, exacerbates experimental colitis and further underscores its protective role in the pathogenesis of UC (65,66).

Studies encompassing patients with UC and animal models have substantiated that hyperactivation of the necroptosis pathway represents a pivotal pathogenic determinant, contributing to both intestinal barrier disruption and aberrant inflammatory cascades (67,68). Histopathological analyses have revealed markedly elevated expression levels of RIPK1, RIPK3 and MLKL in IECs, accompanied by enhanced phosphorylation status, within the inflammatory lesions of patients with UC and experimental models (22). Accumulating evidence indicates that additional regulatory molecules, including purinergic receptor P2Y14 (P2Y14), retinoic acid-inducible gene I (RIG-I) and macrophage migration inhibitory factor (MIF), may participate in the fine-tuned regulation of this process. For instance, the purinergic receptor P2Y14 is upregulated in the inflamed colonic mucosa of patients with UC, where it facilitates RIPK1 transcription via the cAMP/protein kinase A/cAMP response element-binding protein signaling pathway, thereby aggravating IEC necroptosis and amplifying intestinal inflammation (69). Furthermore, upon ligand engagement, the cytosolic RNA sensor RIG-I upregulates MLKL expression via interferon signaling, directly facilitating pore formation (70). Alternatively, RIG-I may indirectly amplify RIPK3-dependent necroptosis by eliciting cytokine production, including type I interferons and TNF (71). Under pathological stress conditions such as ischemia and inflammation, MIF actively fosters RIPK1-mediated necroptosis, thereby intensifying tissue injury (72,73). Collectively, these findings underscore that necroptosis in UC is governed by a sophisticated, multifaceted regulatory network. Furthermore, investigations using the DSS-induced murine colitis model have demonstrated significantly upregulated expression of A20-binding inhibitor of NF-κB activation 1, RIPK1, RIPK3 and MLKL in colonic tissues. Notably, pharmacological blockade with Nec-1s potently inhibits RIPK1 kinase activity, markedly suppressing IEC necroptosis and mitigating colonic inflammatory responses (74). Subsequent analyses of clinical specimens have corroborated a statistically significant positive correlation between RIPK3 expression in the colonic tissues of patients with UC and disease severity indices. Concurrently, genetic ablation of the RIPK3 gene in mice confers notable protection against IEC apoptosis through blockade of the Toll-like receptor 4 (TLR4)/myeloid differentiation primary response protein 88 (MyD88)/NF-κB pathway, effectively alleviating experimental colitis (75).

Contemporary research endeavors are centered on the development of targeted inhibitors against pivotal necroptosis regulators, such as RIPK1, RIPK3 and MLKL, with accumulating evidence demonstrating that pharmacologically modulating these targets in IEC can effectively ameliorate UC. For instance, the citrus flavonoid naringenin and curcumin have been shown to suppress IEC necroptosis by downregulating the mRNA expression of RIPK3 and MLKL, thereby preserving intestinal barrier integrity and markedly ameliorating colitis pathology (76,77). A natural chalcone derivative, ermanin from the genus Leptinella, has exhibited dual inhibition of RIPK1/3 kinases in the DSS-induced colitis model. Mechanistic elucidation revealed that it attenuates intestinal barrier impairment via blockade of MLKL phosphorylation, demonstrating promising therapeutic potential for UC (78). Further investigations demonstrated that polysaccharides from pine pollen and their sulfated derivatives markedly attenuate the inflammatory index in the colitis model, diminish IEC necroptosis incidence and enhance mucosal barrier function through augmented mucin 2 secretion, thereby furnishing robust experimental substantiation for clinical translation (79). The traditional Chinese herbal remedy Sargentodoxa cuneata ameliorates disease manifestations in murine colitis models by curtailing IEC necroptosis (80).

In addition to the aforementioned natural products, chemically modified compounds and targeted chemical agents have likewise unveiled therapeutic utility. For instance, the myricetin-3-O-β-d-lactose sodium salt derivative M10, engineered via incorporation of a hydrophilic glycosyl moiety, displays full aqueous solubility and high stability. A study has established that oral administration of M10 inhibits necroptosis in inflamed colonic epithelium by suppressing TNF-α signaling, exhibiting superior efficacy relative to mesalazine in averting chronic colitis (81). Indole-3-carbinol, a naturally occurring dietary agonist of the aryl hydrocarbon receptor, upon receptor engagement, impedes RIPK1 activation and necrosome assembly in a temporally regulated manner, thereby reducing IEC apoptosis and ameliorating intestinal inflammation (82). As a linchpin modulator of necroptosis and inflammatory cascades, RIPK1 inhibitors potently disrupt inflammatory signaling, attenuate IEC injury and curtail leukocyte infiltration. Multiple studies have corroborated that RIPK1 inhibitors such as SZ-15, HtrA2 and LY3009120 markedly attenuate colonic inflammation, foster tissue regeneration and ameliorate UC progression, thereby illuminating the compelling therapeutic prospects of RIPK1-targeted interventions in UC management (83-85).

Dynamic regulatory nodes orchestrate the delicate equilibrium between necroptosis and apoptosis, ensuring adaptive responses to inflammatory stimuli. In the nascent phases of inflammation, where cellular insult remains modest, the organism preferentially engages programmed apoptotic pathways. This non-inflammatory clearance modality safeguards tissue homeostasis and mitigates excessive immune activation (86). This phase is characterized by selective engagement of apoptotic cascades, where the extrinsic apoptotic pathway mediated by the Fas/FasL pathway and the mitochondrial-dependent intrinsic apoptotic pathway are preferentially activated to facilitate the orderly clearance of damaged cells (87). As inflammatory escalation ensues, perturbations in intracellular homeostasis, such as caspase inhibition or aberrant upregulation of anti-apoptotic effectors, precipitate a phenotypic shift in cell death modality toward necroptosis (88). Notably, DAMPs released during necroptosis engage pattern recognition receptors on adjacent cells via paracrine signaling, thereby upregulating death receptor expression and fostering an apoptosis-prone milieu (89). This ultimately leads to a positive feedback loop of cell death and inflammation, significantly exacerbating intestinal epithelial barrier dysfunction and the spread of inflammation (90). Precise modulation of apoptosis-associated gene expression is expected to fundamentally restore the homeostasis of cell death and promote the repair of intestinal epithelial barrier function. This paradigm unveils a novel molecular intervention strategy for personalized UC management, holding notable clinical translational promise. Nonetheless, effectively implementing these conceptual advances into practical therapeutics mandates surmounting critical challenges, including target specificity, efficacious delivery platforms and inter-individual heterogeneity in gene expression signatures.

IEC pyroptosis

Pyroptosis, a distinct programmed inflammatory cell death modality, has a molecular foundation based on inflammasome-dependent activation of select caspase family members, notably caspase-1 and the caspase-4/5/11 isoforms (91,92). In the pathological evolution of UC, the NLR family pyrin domain containing 3 (NLRP3) inflammasome has emerged as a pivotal regulatory hub mediating inflammatory cascades within the intestinal mucosa. As an intracellular multiprotein complex, the canonical NLRP3 inflammasome incorporates three foundational constituents: The pattern recognition receptor NLRP3, the adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) and the procaspase-1 precursor (93). Under homeostatic conditions, NLRP3 adopts an auto-inhibited conformation with its activation meticulously governed by an array of modulatory factors, thereby safeguarding equilibrium in IECs. Upon perturbation of the intestinal milieu by pathological cues, pathological stimuli including pathogen-associated molecular patterns (PAMPs), DAMPs or metabolic stress, the NLRP3 inflammasome initiates its assembly process (94). In patients with UC, intestinal dysbiosis results in a marked elevation of LPS derived from Gram-negative bacteria. Functioning as a canonical PAMP, LPS initiates nuclear translocation of NF-κB through engagement with TLR4. Activated NF-κB upregulates NLRP3 gene expression and fosters synthesis of pro-inflammatory cytokines (95,96). Consequent to secondary signal stimulation, intracellular potassium efflux, ROS accumulation or lysosomal destabilization, the nascent NLRP3 undergoes NACHT domain-mediated oligomerization, recruiting the adaptor protein ASC to form multiprotein inflammasome complexes. ASC, through PYD-CARD domain interplay, facilitates autoproteolytic maturation of procaspase-1, yielding enzymatically proficient caspase-1 heterotetramers (97). Activated caspase-1 exerts dichotomous biological effects: i) Orchestrates extracellular release of mature IL-1β and IL-18 through cleavage of their pro-forms; and ii) specifically cleaves gasdermin D (GSDMD) to generate the pore-forming N-terminal domain (GSDMD-N). GSDMD-N subsequently oligomerizes to form transmembrane pores on the plasma membrane, triggering cellular osmotic lysis (98). This lytic event facilitates a large release of DAMPs and HMGB1, thereby engendering a self-amplifying feedback loop that intensifies intestinal inflammatory amplification (99).

Pyroptosis in IECs triggers a cascade of immune responses that disrupt the intestinal immune perturbations that destabilize the intestinal immune milieu, a phenomenon intimately linked to UC pathogenesis. In DSS-induced colitis models, transgenic overexpression of lipocalin-2 in wild-type mice markedly exacerbates colonic inflammation and epithelial injury, concomitant with elevated pyroptosis biomarkers specifically localized to the IECs of colonic mucosa derived from patients with UC (100). Type III interferons (IFN-λ) have been demonstrated to augment IEC pyroptosis, thereby undermining mucosal wound repair and curtailing regenerative potential (101). Danger signals released during pyroptosis foster dendritic cell maturation and enhance their antigen-presenting capacity, driving naïve T cell differentiation toward pro-inflammatory Th1 and Th17 subsets (102). Concurrently, these signals attenuate regulatory T cell functionality, engendering an immunosuppressive imbalance that perpetuates uncontrolled inflammatory responses (103). Collectively, these observations delineate IEC pyroptosis as a perpetuator of intestinal inflammation in UC via a self-sustaining inflammatory circuit, which fundamentally underpins the therapeutic refractoriness encountered in clinical practice.

While pyroptosis predominantly exerts pro-inflammatory and tissue-damaging effects in the pathogenesis of UC, elucidation of its molecular underpinnings furnishes a foundational rationale for devising targeted therapeutic interventions. Phytosterols, a class of plant-derived bioactive sterols, exhibit therapeutic potential through their prototypical constituent sitosterol (SIT). Mechanistically, SIT modulates the NLRP3/caspase-1/GSDMD signaling axis to significantly attenuate IEC pyroptosis and pro-inflammatory cytokine release, while concurrently augmenting TJ protein expression to bolster mucosal barrier integrity (104). However, most evidence remains preclinical, and the pharmacokinetics, bioavailability and safety of SIT in chronic UC contexts remain inadequately characterized. The extract from Astragalus membranaceus Bunge has been shown to ameliorate UC by curtailing IEC pyroptosis via upregulation of phospholipase C-β2 (105). The pathological characteristics of refractory UC progression primarily encompass a self-perpetuating cycle linking mucosal barrier disruption and unrelenting inflammatory amplification. Recent evidence substantiates that nanocarrier-mediated targeted delivery of 4-octyl itaconate to IECs potently suppresses GSDME-mediated pyroptosis (14). Further investigation has revealed that the artemisinin derivative SM934 exhibits dual modulatory effects in experimental colitis models; it concurrently inhibits programmed cell death modalities and abrogates caspase-1-dependent pyroptotic cascades, thereby markedly ameliorating epithelial barrier impairment (106). Nevertheless, the long-term efficacy, toxicity and dosing regimens for SM934 in chronic models warrant comprehensive evaluation to surmount translational hurdles. Schisandrin B attenuates NLRP3 inflammasome activation-mediated IL-1β secretion and IEC pyroptosis in colitis models by activating AMPK/Nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent signaling to mitigate ROS-induced mitochondrial damage, suggesting its potential as a therapeutic strategy for acute colitis (107). In LPS/ATP-induced in vitro models of IEC pyroptosis and inflammation utilizing HT29 human colonic carcinoma cells, resveratrol forestalls pyroptosis onset by impeding NF-κB pathway activation (108). Beyond phytogenic agents, mesenchymal stem cells (MSCs) and their secreted exosomes have recently garnered notable attention for therapeutic applications. Hair follicle-derived MSC exosomes convey differentially expressed microRNAs (miRNAs) that concurrently suppress both tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling and IFN-γ inflammatory pathways, thereby effectively inhibiting IEC pyroptosis (109). Notably, bone marrow-derived MSC exosomes selectively suppress NLRP3/caspase-1 pathway activation via miR-539-5p shuttling, thereby modulating the pyroptosis-associated molecular network and ultimately attenuating UC progression (110).

In recent years, contemporary drug discovery paradigms targeting pyroptosis have diversified, with numerous chemical and nucleic acid therapeutics evincing robust efficacy. For instance, miR-141-3p effectively inhibits LPS-induced IEC pyroptosis by targeting the HSP90 molecular chaperone, while concurrently alleviating inflammation in DSS-induced murine colitis, highlighting its promise as a nucleic acid-based therapeutic approach for UC (111). The broad-spectrum antiviral nucleotide remdesivir likewise ameliorates IEC pyroptosis and gut inflammatory cascades by inhibiting the NLRP3 inflammasome and downstream caspase-1/GSDMD signaling (112). Meanwhile, ruscogenin, a steroidal sapogenin from Ophiopogon japonicus, has demonstrated potential in mitigating inflammatory processes by suppressing NLRP3 inflammasome activation and caspase-1-dependent canonical pyroptosis (113). Moreover, in acute severe UC, the methyl-donor betaine effectively inhibits oxidative stress-induced inflammatory pyroptosis, thereby expanding the therapeutic repertoire for UC (114). These findings suggest that precision regulation strategies targeting aberrant pyroptosis in IECs may disrupt the 'pyroptosis-inflammation-barrier disruption' positive feedback loop, thereby establishing an innovative therapeutic paradigm for UC pathological intervention.

IEC ferroptosis

Ferroptosis, a unique modality of programmed cell death, was initially characterized and designated in 2012 by Dixon et al (115) through systematic investigation. Distinct from other forms of regulated cell death, ferroptosis is pathologically defined by the intricate interplay between iron ion homeostasis dysregulation and lipid peroxidation accumulation (116). IECs predominantly internalize iron via transferrin receptor 1-dependent endocytosis (117). Upon endocytosis, Fe3+ is enzymatically reduced to biologically active Fe2+ species via metalloreductases such as Six-transmembrane epithelial antigen of prostate 3 (118,119). During the pathogenesis of UC, IECs are subjected to a sustained inflammatory milieu, which elicits dysregulated mobilization of excess Fe2+ from the labile iron pool and thereby instigates ROS generation through Fenton reaction cascades (19). The Fenton reaction employs Fe2+-mediated catalytic cycles to decompose hydrogen peroxide into highly reactive hydroxyl radicals. These radicals preferentially attack membrane phospholipids containing polyunsaturated fatty acids (120), thereby initiating a self-amplifying chain reaction of lipid peroxidation. Glutathione peroxidase 4 (GPX4), a pivotal regulator of lipid redox balance, forms the cornerstone of ferroptosis suppression. Under homeostatic conditions, GPX4 utilizes reduced glutathione (GSH) to transmute deleterious lipid hydroperoxides into innocuous lipid alcohols, effectively suppressing the cascade amplification of lipid peroxidation (121). Notably, in UC progression, multiple pathogenic factors synergistically inhibit GPX4 enzymatic activity, thereby impairing its detoxification of peroxidation intermediates. Upon decompensation of the antioxidant defense system, lipid peroxidation products surpass cellular homeostatic thresholds, ultimately triggering ferroptosis in IECs through the disruption of membrane structural integrity (122,123).

As the principal mucosal interface organ, the intestinal tract exhibits a characteristic oxidative stress microenvironment due to persistent exposure to exogenous stimuli including microbiota, metabolites and food-derived antigens (124). Investigations have revealed a reciprocal pathophysiological interplay between ferroptosis in IECs and gut oxidative stress status. In murine colitis models, inflammation-orchestrated oxidative perturbations markedly elevate ferroptosis effectors [cyclooxygenase-2 and acyl-CoA synthetase long-chain family member 4 (ACSL4)] in parallel with diminished protein abundance of GPX4 and ferritin heavy chain 1. These molecular signatures demonstrate positive correlation with the extent of IEC injury (125). Pharmacological preconditioning with ferroptosis inhibitors has been demonstrated to markedly attenuate above-mentioned histopathological manifestations in DSS-induced colitis. Integrative multi-omics profiling has unveiled signature expression patterns of ferroptosis-linked genes in tissues derived from patients with UC (126,127). Mechanistic investigations have demonstrated that ACSL4, a master regulator of ferroptosis, accelerates IEC ferroptosis by activating the NF-κB signaling pathway, thereby contributing to UC pathogenesis (128).

There have been notable advancements in therapeutic modalities targeting ferroptosis regulation in IECs, with innovative interventions conferring antioxidant cytoprotection and inflammatory attenuation via selective ferroptosis pathway blockade. Astragalus polysaccharide (APS), the predominant bioactive polysaccharide from the traditional Chinese herb A. membranaceus, has demonstrated anti-ferroptotic efficacy across multiple experimental models. Experimental evidence demonstrated that APS significantly suppresses ferroptosis progression and sustains cellular homeostasis in DSS-induced murine colitis models and as well as RSL3-treated human IECs in vitro (129). Multi-omics analyses has unveiled that vanillic acid (VA) modulates the ferroptosis axis through targeted ligation to carbonic anhydrase IX and stromal interaction molecule 1, effectively restoring intestinal epithelial barrier integrity and emerging as a novel therapeutic candidate for UC (130). Nevertheless, the target fidelity and off-target liabilities of VA within intricate human milieus necessitate rigorous delineation. Emerging data implicate that disrupted iron homeostasis exacerbates colitis progression via dual mechanisms involving ferroptosis activation and gut microbiota dysbiosis (131,132). Pharmacological investigations confirm that palmatine, a natural isoquinoline alkaloid, markedly diminishes colonic iron deposition and ameliorates experimental UC pathology via multi-target modulation, concurrently suppressing NF-κB inflammatory signaling, ROS generation and ferroptosis signal cascades (133). Notably, the oral iron chelator deferasirox represses IEC ferroptosis, reprograms gut microbiota architecture and augments short-chain fatty acid (SCFA) biosynthesis, thereby ameliorating DSS-induced UC inflammation through multifaceted mechanisms and evincing clinical promise (134). However, the use of iron chelators still requires cautious monitoring of dosage and safety to avoid potential adverse effects (135,136).

Beyond directly targeting the ferroptosis pathway, certain nutritional factors and metabolites also demonstrate therapeutic value. Butyrate is a microbiota-derived SCFA depleted in colitis-afflicted murine cohorts. A study has revealed that sodium butyrate treatment activates the Nrf2/GPX4 signaling pathway, thereby inhibiting ferroptosis, alleviating oxidative stress and inflammatory responses and restoring intestinal barrier function (137). Furthermore, vitamin D has been demonstrated to attenuate ferroptosis in DSS-induced murine models and LPS-stimulated HCT116 cells through ACSL4 repression, consequently tempering UC severity (138). Additionally, supplementation with the essential trace element selenium, particularly in the form of sodium selenite, effectively reduces IEC mortality, intracellular iron content, lipid ROS and mitochondrial membrane damage, ultimately attenuating DSS-induced colitis (139). Nevertheless, these benefits, optimal dosing paradigms, delivery modalities and protracted efficacy demand validation via robust clinical trials.

Oxymatrine (OY), the primary bioactive compound derived from Sophora flavescens, is routinely employed in Chinese integrative UC management. Through integrated approaches combining bioinformatics, molecular biology and animal experimentation, OY has been demonstrated to alleviate UC symptoms via mechanisms involving the regulation of Fe2+ and GSH levels, as well as anti-inflammatory pathways (140). Lizhong decoction (LZD) potently represses IEC ferroptosis by reducing iron overload and elevating recombinant solute carrier family 7 member 11 and GPX4 expression in colonic mucosa (141). However, the polypharmacology of multi-herbal decoctions such as LZD complicates mechanistic attribution, underscoring the imperative for active constituent isolation to facilitate clinical advancement.

While breakthrough advances have been achieved in natural compound research, exosomes (critical mediators of intercellular communication) have emerged as possessing unique biomedical potential in the therapeutics of UC. Human umbilical cord MSC-derived exosomes can deliver miR-129-5p to specifically suppress the expression of ACSL4, dually regulating the progression of lipid peroxidation and the function of the GSH-GPX4 antioxidant axis (142). A recent study has demonstrated that endometrial regenerative cell-derived exosomes enhance GSH biosynthesis and GPX4 enzymatic activity while coordinately attenuating tissue iron accumulation, malondialdehyde levels and ACSL4 protein expression, demonstrating marked efficacy in ameliorating both histopathological damage and clinical manifestations of colitis (143). An in-depth analysis of the molecular regulatory network underlying abnormal ferroptosis in IECs would not only yield precise therapeutic targets for UC but also promises the advent of innovative pharmacotherapeutics through targeted modulation of key ferroptosis pathways, thereby improving intestinal barrier function.

IEC autophagy

Autophagy, an evolutionarily conserved intracellular degradation system, orchestrates cellular homeostasis and stress adaptation via regulated material recycling. Based on substrate transport mechanisms, autophagy is categorized into three subtypes: Macroautophagy, microautophagy and chaperone-mediated autophagy (144). Among these, macroautophagy predominates as the primary regulatory mechanism, forming the cornerstone of autophagy research. Upon microenvironmental stimuli such as nutrient deprivation or oxidative stress (145,146), endoplasmic reticulum-resident unfolded protein response sensors synergize with other stress detectors to trigger a transcriptional cascade of autophagy-related genes via regulators including transcription factor EB (147). This process initiates with the formation of a double-membrane structure termed the phagophore in the cytoplasm (148), which achieves quality control by selectively enveloping targeted substrates including damaged organelles and misfolded proteins. Following subsequent membrane extension and closure, the phagophore matures into an autophagosome that fuses with lysosomal membranes to form an autolysosome (149). Within the autolysosomal lumen, acid hydrolases degrade the enclosed cargo into recyclable metabolites, such as amino acids and free fatty acids (150), that are effluxed into the cytoplasm through lysosomal transporters. These metabolites are released into the cytoplasm via lysosomal membrane transporters, where they re-enter biosynthetic pathways or fuel the tricarboxylic acid cycle, thereby completing the closed-loop regulation of intracellular material recycling (151).

In recent years, the regulatory role of autophagy in UC pathogenesis has garnered substantial attention in gastroenterology research, with its dual regulatory properties exhibiting complex biological effects during disease progression. Pathologically, UC is characterized by dysregulated dynamic equilibrium of pro-/anti-inflammatory cytokine networks, coupled with unrelenting inflammatory cascades, aberrant intestinal barrier permeability and perturbed expression of TJ proteins, collectively driving disease chronicity (152). Mechanistic investigations have elucidated that TNF-α-driven inflammatory microenvironments impair intestinal epithelial barrier integrity via impaired autophagic flux, manifesting as aberrant claudin-2 expression and TJ disruption, a process mechanistically linked to autolysosomal system dysfunction (153). Analyses of clinical specimens from patients with active UC reveal notable downregulation of autophagy regulator activating transcription factor 4 in intestinal mucosa, implicating diminished autophagic capacity in disease exacerbation (154). Pharmacological autophagy activation in LPS-stimulated Caco-2 cell models and experimental colitis animals significantly reduces pro-inflammatory cytokines and ameliorates oxidative stress indices, underscoring the therapeutic potential of autophagy modulation in IBD (155,156). In DSS-induced colitis models, autophagy impairment intensifies intestinal inflammation through hyperactivation of the NLRP3 inflammasome, thereby promoting caspase-1 cleavage and maturation of IL-1β/IL-18 (157,158). Crucially, excessive autophagy activation may induce type II programmed cell death, underscoring the importance of precise autophagic activity regulation given this dual-edged effect. In Erbin knockout murine models, autophagy inhibitor chloroquine mitigates DSS-induced hyperinflammation by blocking autophagosome-lysosome fusion, with mechanisms involving downregulation of cell death-associated proteins, thereby illustrating the context-specific utility of autophagy suppression (159). Collectively, current evidence establishes autophagy as a homeostatic modulator in UC, with its pro-survival and pro-death duality being precisely controlled by microenvironmental signaling networks.

Pharmacological investigations on the classical formula Baitouweng decoction have demonstrated that it augments autophagic flux via AMPKα phosphorylation activation and mTORC1 complex inhibition, thereby restoring intestinal epithelial barrier integrity and attenuating disease activity index scores in DSS-induced murine colitis models (160). Mechanistic investigations reveal that procyanidin A1 augments autophagic activity through the AMPK/mTOR/p70S6K signaling axis in LPS-stimulated IEC inflammation models, markedly suppressing pro-inflammatory cytokine secretion (161). Notably, the probiotic strain Lactobacillus plantarum OLL2712 activates protective autophagy in IECs via the MyD88-dependent pathway, thereby reorganizing TJ proteins to fortify intestinal barrier mechanics (162). Although probiotic-mediated autophagy modulation represents a burgeoning therapeutic avenue, elucidation of strain specificity, dosage regimens and host microbiome interplay remains imperative. Compound sophora decoction significantly alleviates DSS-induced intestinal inflammation by fostering autophagy through suppression of PI3K/AKT pathway activation (163). Moreover, the Jianpi Qingchang (JPQC) decoction, composed of nine traditional Chinese medicinal herbs, alleviates colitis progression by suppressing endoplasmic reticulum stress-associated excessive autophagy in IECs within a DSS-induced model (164) Notably, JPQC underscores the contextual therapeutic merits of both autophagy activation and inhibition, albeit the dose-dependent dichotomous effects warrant deeper scrutiny.

Accumulating evidence indicates that disrupting autophagic homeostasis is a pivotal node in UC pathogenesis, with pharmacological restoration of autophagic dynamic equilibrium emerging as a novel therapeutic target. Beyond the aforementioned Chinese herbal and natural products, diverse active compounds and biomacromolecules have shown therapeutic promise in UC management via autophagy modulation. For instance, epimedium polysaccharide (EPS), a bioactive compound, has been shown to engender autophagy augmentation via the AMPK/mTOR pathway amid colonic inflammation, exerting a protective effect in DSS-induced UC models, which suggests EPS as a potential therapeutic target (165). Upregulation of circular RNA HECTD1 has been shown to orchestrate HuR-dependent autophagy in IECs via miR-182-5p sequestration, consequently ameliorating UC histopathology (166) The Slit family of glycoproteins (Slit1-3), canonically expressed in neural and immune compartments, exert regulatory oversight on inflammatory cascades (167,168). Overexpression of Slit2 has been shown to sustain intestinal stem cell proliferation and normal autophagic flux in mice following DSS challenge, thereby mitigating colonic inflammation and curtailing pro-inflammatory cytokine secretion (169). These emerging strategies provide innovative perspectives for UC intervention through the modulation of autophagy; however, the majority of current studies remain focused on mechanistic exploration and are still considerably distant from clinical application. Future research should focus on enhancing target specificity, optimizing delivery systems and clarifying the pharmacodynamic-to-toxicological equilibrium, thereby facilitating their translation into clinical practice.

Discussion

Disruption of intestinal barrier integrity is firmly established as a central pathological feature of UC, driven by intricate molecular mechanisms involving a dynamic interplay between IEC homeostatic imbalance and dysregulation of the immune microenvironment. The present review summarizes the major programmed cell death modalities in IECs in UC: Autophagy, ferroptosis, pyroptosis, apoptosis and necroptosis (Fig. 1). As the principal components of the intestinal mechanical barrier, dysregulated IEC death directly impairs TJ complexes, leading to epithelial barrier breakdown and subsequent hyperactivation of pattern recognition receptor-mediated innate immune responses. This series of events leads to a self-sustaining cycle of persistent inflammation and mucosal injury. Recent progress in single-cell RNA sequencing and intestinal organoid technologies has yielded unprecedented insights into the spatiotemporal dynamics of IEC death modalities and their association with UC pathological phenotypes, providing a molecular framework for understanding disease heterogeneity and patient-specific variations (14,17,100,170-172).

Mechanism of programmed cell death in
IECs. During the development of UC, various forms of programmed
cell death mechanisms occur in IECs, including apoptosis,
necroptosis, pyroptosis, ferroptosis and autophagy. IECs,
intestinal epithelial cells; UC, ulcerative colitis; Bak, Bcl-2
homologous antagonist/killer; FasL, Fas ligand; TNFR1, tumor
necrosis factor receptor 1; TLR4, Toll-like receptor 4; FADD,
Fas-associated protein with death domain; RIPK1/3,
receptor-interacting serine/threonine-protein kinase 1/3; MLKL,
mixed lineage kinase domain-like protein; NLRP3, NOD-, LRR- and
pyrin domain-containing protein 3; IL, interleukin; GSDMD-N,
N-terminal domain of Gasdermin D; GSH, glutathione; GPX-4,
glutathione peroxidase 4; ROS, reactive oxygen species; ER,
endoplasmic reticulum; ATG, autophagy-related gene.

Figure 1

Mechanism of programmed cell death in IECs. During the development of UC, various forms of programmed cell death mechanisms occur in IECs, including apoptosis, necroptosis, pyroptosis, ferroptosis and autophagy. IECs, intestinal epithelial cells; UC, ulcerative colitis; Bak, Bcl-2 homologous antagonist/killer; FasL, Fas ligand; TNFR1, tumor necrosis factor receptor 1; TLR4, Toll-like receptor 4; FADD, Fas-associated protein with death domain; RIPK1/3, receptor-interacting serine/threonine-protein kinase 1/3; MLKL, mixed lineage kinase domain-like protein; NLRP3, NOD-, LRR- and pyrin domain-containing protein 3; IL, interleukin; GSDMD-N, N-terminal domain of Gasdermin D; GSH, glutathione; GPX-4, glutathione peroxidase 4; ROS, reactive oxygen species; ER, endoplasmic reticulum; ATG, autophagy-related gene.

Recent studies have unveiled a novel programmed cell death pathway known as PANoptosis, which merges key molecular characteristics of pyroptosis, apoptosis and necroptosis to form a multifaceted death complex (173,174). The regulatory framework of PANoptosis entails the integration and interplay of multiple signaling pathways. Research has highlighted the crucial roles of key regulatory molecules such as Z-DNA binding protein 1 (ZBP1), RIPK1 and RIPK3 in the PANoptosis pathway (175). The nucleic acid sensor ZBP1, activated during pathogen infection or cellular stress by recognizing viral nucleic acids or endogenous DAMPs, undergoes conformational changes to recruit and phosphorylate RIPK3 (176). Notably, upon activation by death receptor ligands or PAMPs, RIPK1 initiates PANoptosis signaling transduction by engaging downstream molecules through its death domain. Additionally, RIPK1 forms a functional complex with RIPK3, jointly driving the phosphorylation cascade of MLKL and ultimately inducing cells to enter the PANoptosis program (177,178). Within IECs, a complex molecular interplay exists among apoptosis, necroptosis, pyroptosis and the resulting PANoptosis, all of which play critical roles in the pathogenesis of IBD. Caspase-8 and its adaptor protein FADD serve as central molecules interconnecting these three programmed cell death pathways (59). Upon excessive stimulation by TNF or TLR signaling, caspase-8 is activated and regulates cell fate by initiating apoptosis through the cleavage of downstream caspase-3/7, while also modulating RIPK1 and RIPK3 to inhibit necroptosis (179). Concurrently, caspase-8 can interact with ASC to activate caspase-1 and cleave GSDMD, thereby triggering pyroptosis (180). When caspase-8 function is compromised or inhibited by pathogens, both apoptotic and pyroptotic pathways are impaired, leading cells to undergo RIPK3-mediated necroptosis, which exacerbates intestinal barrier disruption and inflammatory responses. Furthermore, in the absence of caspase-1 in IECs, inflammasome sensors such as NLRP1b and NLRC4 can still initiate apoptosis through ASC-dependent caspase-8 activation (181). The necroptosis effector MLKL can also promote ASC oligomerization and caspase-1 activation, thereby linking necroptosis with pyroptosis (182). These intricate molecular interactions illustrate that apoptosis, necroptosis and pyroptosis are interconnected through shared molecular components, collectively driving epithelial cell death, barrier dysfunction and exacerbated intestinal inflammation in UC. Emerging evidence has demonstrated that PANoptosis plays a crucial regulatory role in the pathogenesis of IBD, particularly UC. In the DSS-induced murine colitis model, IECs exhibited concurrent phenotypes of Dynamin related protein 1 mediated mitochondrial fission and ZBP1-dependent PANoptosis. Notably, analysis of clinical specimens revealed a significant positive correlation between the activation levels of PANoptosis in IECs and the clinical activity index of patients with UC (21,183).

Despite notable advancements in recent years in elucidating the mechanisms of IEC programmed death in UC, several crucial issues remain unresolved. For instance, the intricate interplay between various programmed death pathways, including apoptosis, necroptosis, pyroptosis and ferroptosis, requires further elucidation. For instance, the novel multimodal cell death pathway PANoptosis, which integrates key molecular features of apoptosis, necroptosis and pyroptosis, necessitates in-depth exploration of its specific regulatory mechanisms in UC and its synergistic or antagonistic interactions with other death pathways. Additionally, the dynamic interactions between programmed death pathways and the intestinal microenvironment, such as dysbiosis and metabolic products, demand thorough investigation to unravel the complex networks in the pathological progression of UC. Given the intricate interconnections and mutual regulation among these programmed cell death pathways, combination therapies that target multiple key nodes of apoptosis, necroptosis and pyroptosis concurrently may enable precise modulation of the cell death network, leading to more comprehensive intestinal protection and inflammation control. Furthermore, emerging strategies such as the utilization of gene-editing technologies (such as CRISPR/Cas9) to regulate the expression of critical genes including caspase-8 and RIPK3, or the application of stem cell and genetically engineered cell-based therapies to restore compromised epithelial function, offer promising avenues for future therapeutics. Collectively, interventions that target programmed cell death in IECs at multiple levels and from various angles hold significant promise for mitigating epithelial damage and inflammation in UC, thereby advancing therapeutic strategies for this disease.

Natural bioactive compounds and nanoparticle-mediated drug delivery systems have gained significant attention in biomedical research due to their notable translational potential, especially in the management of metabolic disorders and tumor immunomodulation. Their therapeutic efficacy stems from their unique ability to simultaneously modulate multiple molecular targets, while maintaining favorable biosafety profiles, offering innovative and multifaceted strategies for UC treatment. Notably, an increasing body of evidence highlights the distinct potential of natural bioactive compounds and nanoparticle-mediated drug delivery systems in UC therapy, particularly in their capability to preserve colonic epithelial barrier integrity and mitigate pro-inflammatory cytokine cascades by precisely regulating programmed cell death pathways. These compounds and nanoparticles exert their protective effects by targeting key molecular mechanisms underlying epithelial dysfunction and immune dysregulation, thereby addressing the dual pathological axes of UC.

The present review provides a comprehensive analysis of the central role of IEC death in the pathogenesis of UC-associated chronic inflammation and mucosal injury. Furthermore, it consolidates recent progress in the utilization of natural small-molecule compounds and nanoparticle-mediated drug delivery systems that selectively target abnormal IEC death to mitigate barrier dysfunction, as summarized in Table SI. These compounds, sourced from a variety of natural origins, have exhibited effectiveness in modulating crucial signaling pathways, presenting promising therapeutic opportunities for UC management. In terms of clinical translation, while natural compounds and nanomedicines have demonstrated significant potential in modulating programmed death pathways, their targeting and bioavailability encounter challenges. For instance, the delivery efficiency of nanomedicines is constrained by the intricate intestinal milieu, and enhancing their targeting, such as through surface ligand modification or utilization of intestinal-specific receptors, represents a current research priority. Additionally, despite the advantages of the multi-target properties of natural compounds in treating UC, their potential for unforeseen side effects necessitates further structural refinement and pharmacological assessment. Future research should integrate single-cell omics, organoid models and systems pharmacology techniques to precisely decipher the molecular mechanisms of programmed death pathways and develop more efficient and safer targeted therapeutic strategies to surmount existing treatment bottlenecks and improve the clinical prognosis of patients with UC.

Looking forward, transformative breakthroughs in UC therapy will necessitate the development of integrated therapeutic strategies that concurrently target IEC death pathways and their downstream hyperinflammatory responses. These strategies aim to disrupt the self-sustaining cycle of 'barrier damage-inflammation amplification' that underlies the core of UC pathogenesis. One approach involves the development of therapeutic agents capable of synergistically modulating multiple targets within the regulated cell death network. Given the intricate molecular crosstalk among different forms of programmed cell death, involving key molecules such as caspase-8, RIPK3 and GSDMD, inhibitors targeting a single pathway may be insufficient to fully prevent the dysregulated death of IECs. The design of small molecules or biologics that concurrently regulate multiple critical nodes holds potential for more effectively restoring IEC homeostasis, thereby achieving comprehensive protection of the intestinal barrier and improved control of inflammation. Additionally, enhancing targeting specificity while minimizing systemic side effects is crucial for successful clinical translation. Employing nanotechnology, antibody-drug conjugates or oral targeted delivery systems to selectively accumulate therapeutics within inflamed intestinal tissues or IECs can enhance local drug concentrations and markedly reduce off-target effects in other organs. Novel therapeutic approaches involving gene editing and cell therapy also warrant exploration. While still in the early stages of investigation, in vivo or ex vivo editing of dysregulated genes in IEC (such as caspase-8 or RIPK3) using technologies such as CRISPR/Cas9, or the administration of genetically engineered stem cells or organoids to repair damaged epithelium, presents a highly promising next-generation treatment strategy. These interventions may provide novel options for patients with refractory UC who are intolerant to conventional pharmacological or biological therapies. In summary, future drug development for UC should transcend the traditional 'single target, single disease' paradigm and adopt integrated, multi-targeted, multimodal and precision-focused strategies with an emphasis on local intestinal targeting. By addressing these unresolved mechanisms and clinical translation challenges, the present review not only establishes a new theoretical framework for understanding the pathomechanism of UC but also establishes a robust basis for the advancement of innovative therapeutic strategies, offering significant scientific and clinical implications.

Supplementary Data

Availability of data and materials

Not applicable.

Authors' contributions

BW drafted and revised the manuscript. BW and SS conceptualized the review and contributed to the writing. YL participated in literature collation and manuscript editing. LG reviewed and revised the manuscript. All authors contributed to the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

This work was supported by the Research Foundation of the Department of Education of Yunnan Province (grant no. 2025Y1195) and Yunnan Fundamental Research Projects (grant no. 202501AT070405).

References

1 

Buie MJ, Quan J, Windsor JW, Coward S, Hansen TM, King JA, Kotze PG, Gearry RB, Ng SC, Mak JWY, et al: Global hospitalization trends for Crohn's disease and ulcerative colitis in the 21st century: A systematic review with temporal analyses. Clin Gastroenterol Hepatol. 21:2211–2221. 2023. View Article : Google Scholar

2 

Ordás I, Eckmann L, Talamini M, Baumgart DC and Sandborn WJ: Ulcerative colitis. Lancet. 380:1606–1619. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Leppkes M and Neurath MF: Cytokines in inflammatory bowel diseases - Update 2020. Pharmacol Res. 158:1048352020. View Article : Google Scholar : PubMed/NCBI

4 

Le Berre C, Loeuille D and Peyrin-Biroulet L: Combination therapy with vedolizumab and tofacitinib in a patient with ulcerative colitis and spondyloarthropathy. Clin Gastroenterol Hepatol. 17:794–796. 2019. View Article : Google Scholar

5 

Abdalla MI and Levesque BG: Progress in corticosteroid use in the Era of biologics with room for improvement. Am J Gastroenterol. 116:1187–1188. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Quansah E, Gardey E, Ramoji A, Meyer-Zedler T, Goehrig B, Heutelbeck A, Hoeppener S, Schmitt M, Waldner M, Stallmach A and Popp J: Intestinal epithelial barrier integrity investigated by label-free techniques in ulcerative colitis patients. Sci Rep. 13:26812023. View Article : Google Scholar : PubMed/NCBI

7 

Chen Q, Chen T, Xiao H, Wang F, Li C, Hu N, Bao L, Tong X, Feng Y, Xu Y, et al: APEX1 in intestinal epithelium triggers neutrophil infiltration and intestinal barrier damage in ulcerative colitis. Free Radic Biol Med. 225:359–373. 2024. View Article : Google Scholar : PubMed/NCBI

8 

Peterson LW and Artis D: Intestinal epithelial cells: Regulators of barrier function and immune homeostasis. Nat Rev Immunol. 14:141–153. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Jiminez JA, Uwiera TC, Douglas Inglis G and Uwiera RR: Animal models to study acute and chronic intestinal inflammation in mammals. Gut Pathog. 7:292015. View Article : Google Scholar : PubMed/NCBI

10 

Yan H and Ajuwon KM: Butyrate modifies intestinal barrier function in IPEC-J2 cells through a selective upregulation of tight junction proteins and activation of the Akt signaling pathway. PLoS One. 12:e01795862017. View Article : Google Scholar : PubMed/NCBI

11 

Mehandru S and Colombel JF: The intestinal barrier, an arbitrator turned provocateur in IBD. Nat Rev Gastroenterol Hepatol. 18:83–84. 2021. View Article : Google Scholar

12 

Su L, Nalle SC, Shen L, Turner ES, Singh G, Breskin LA, Khramtsova EA, Khramtsova G, Tsai PY, Fu YX, et al: TNFR2 activates MLCK-dependent tight junction dysregulation to cause apoptosis-mediated barrier loss and experimental colitis. Gastroenterology. 145:407–415. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Fukuda T, Majumder K, Zhang H, Turner PV, Matsui T and Mine Y: Adenine inhibits TNF-α signaling in intestinal epithelial cells and reduces mucosal inflammation in a dextran sodium sulfate-induced colitis mouse model. J Agric Food Chem. 64:4227–4234. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Li W, Chen D, Zhu Y, Ye Q, Hua Y, Jiang P, Xiang Y, Xu Y, Pan Y, Yang H, et al: Alleviating pyroptosis of intestinal epithelial cells to restore mucosal integrity in ulcerative colitis by targeting delivery of 4-Octyl-itaconate. ACS Nano. 18:16658–16673. 2024. View Article : Google Scholar : PubMed/NCBI

15 

Chi F, Zhang G, Ren N, Zhang J, Du F, Zheng X, Zhang C, Lin Z, Li R, Shi X and Zhu Y: The anti-alcoholism drug disulfiram effectively ameliorates ulcerative colitis through suppressing oxidative stresses-associated pyroptotic cell death and cellular inflammation in colonic cells. Int Immunopharmacol. 111:1091172022. View Article : Google Scholar : PubMed/NCBI

16 

Chen Y, Yan W, Chen Y, Zhu J, Wang J, Jin H, Wu H, Zhang G, Zhan S, Xi Q, et al: SLC6A14 facilitates epithelial cell ferroptosis via the C/EBPβ-PAK6 axis in ulcerative colitis. Cell Mol Life Sci. 79:5632022. View Article : Google Scholar

17 

Zhang J, Cen L, Zhang X, Tang C, Chen Y, Zhang Y, Yu M, Lu C, Li M, Li S, et al: MPST deficiency promotes intestinal epithelial cell apoptosis and aggravates inflammatory bowel disease via AKT. Redox Biol. 56:1024692022. View Article : Google Scholar : PubMed/NCBI

18 

Foerster EG, Mukherjee T, Cabral-Fernandes L, Rocha JDB, Girardin SE and Philpott DJ: How autophagy controls the intestinal epithelial barrier. Autophagy. 18:86–103. 2022. View Article : Google Scholar :

19 

Xu M, Tao J, Yang Y, Tan S, Liu H, Jiang J, Zheng F and Wu B: Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis. Cell Death Dis. 11:862020. View Article : Google Scholar : PubMed/NCBI

20 

Ma ZR, Li ZL, Zhang N, Lu B, Li XW, Huang YH, Nouhoum D, Liu XS, Xiao KC, Cai LT, et al: Inhibition of GSDMD-mediated pyroptosis triggered by Trichinella spiralis intervention contributes to the alleviation of DSS-induced ulcerative colitis in mice. Parasit Vectors. 16:2802023. View Article : Google Scholar : PubMed/NCBI

21 

Wang JM, Yang J, Xia WY, Wang YM, Zhu YB, Huang Q, Feng T, Xie LS, Li SH, Liu SQ, et al: Comprehensive analysis of PANoptosis-related gene signature of ulcerative colitis. Int J Mol Sci. 25:3482023. View Article : Google Scholar

22 

Akanyibah FA, Zhu Y, Jin T, Ocansey DKW, Mao F and Qiu W: The function of necroptosis and its treatment target in IBD. Mediators Inflamm. 2024:72753092024. View Article : Google Scholar : PubMed/NCBI

23 

Iwanaga T and Takahashi-Iwanaga H: Disposal of intestinal apoptotic epithelial cells and their fate via divergent routes. Biomed Res. 43:59–72. 2022. View Article : Google Scholar : PubMed/NCBI

24 

Soroosh A, Fang K, Hoffman JM, Law IKM, Videlock E, Lokhandwala ZA, Zhao JJ, Hamidi S, Padua DM, Frey MR, et al: Loss of miR-24-3p promotes epithelial cell apoptosis and impairs the recovery from intestinal inflammation. Cell Death Dis. 13:82021. View Article : Google Scholar : PubMed/NCBI

25 

Wolf P, Schoeniger A and Edlich F: Pro-apoptotic complexes of BAX and BAK on the outer mitochondrial membrane. Biochim Biophys Acta Mol Cell Res. 1869:1193172022. View Article : Google Scholar : PubMed/NCBI

26 

Sheridan C, Delivani P, Cullen SP and Martin SJ: Bax- or Bak-induced mitochondrial fission can be uncoupled from cytochrome C release. Mol Cell. 31:570–585. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Zhou M, Li Y, Hu Q, Bai XC, Huang W, Yan C, Scheres SH and Shi Y: Atomic structure of the apoptosome: mechanism of cytochrome c- and dATP-mediated activation of Apaf-1. Genes Dev. 29:2349–2361. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Albalawi GA, Albalawi MZ, Alsubaie KT, Albalawi AZ, Elewa MAF, Hashem KS and Al-Gayyar MMH: Curative effects of crocin in ulcerative colitis via modulating apoptosis and inflammation. Int Immunopharmacol. 118:1101382023. View Article : Google Scholar : PubMed/NCBI

29 

Lin S, Zhang X, Zhu X, Jiao J, Wu Y, Li Y and Zhao L: Fusobacterium nucleatum aggravates ulcerative colitis through promoting gut microbiota dysbiosis and dysmetabolism. J Periodontol. 94:405–418. 2023. View Article : Google Scholar

30 

Li Y, Ma M, Wang X, Li J, Fang Z, Li J, Yang B, Lu Y, Xu X and Li Y: Celecoxib alleviates the DSS-induced ulcerative colitis in mice by enhancing intestinal barrier function, inhibiting ferroptosis and suppressing apoptosis. Immunopharmacol Immunotoxicol. 46:240–254. 2024. View Article : Google Scholar : PubMed/NCBI

31 

Iwamoto M, Makiyama K, Koji T, Kohno S and Nakane PK: Expression of Fas and Fas-ligand in epithelium of ulcerative colitis. Nihon Rinsho. 54:1970–1974. 1996.In Japanese. PubMed/NCBI

32 

Souza HS, Tortori CJ, Castelo-Branco MT, Carvalho AT, Margallo VS, Delgado CF, Dines I and Elia CC: Apoptosis in the intestinal mucosa of patients with inflammatory bowel disease: Evidence of altered expression of FasL and perforin cytotoxic pathways. Int J Colorectal Dis. 20:277–286. 2005. View Article : Google Scholar

33 

Tang R, Jiang L, Ji Q, Kang P, Liu Y, Miao P, Xu X and Tang M: Resveratrol targeting MDM2/P53/PUMA axis to inhibit colonocyte apoptosis in DSS-induced ulcerative colitis mice. Front Pharmacol. 16:15729062025. View Article : Google Scholar : PubMed/NCBI

34 

Chen J: The Cell-Cycle Arrest and Apoptotic Functions of p53 in Tumor Initiation and Progression. Cold Spring Harb Perspect Med. 6:a0261042016. View Article : Google Scholar : PubMed/NCBI

35 

Eissa N, Hussein H, Diarra A, Elgazzar O, Gounni AS, Bernstein CN and Ghia JE: Semaphorin 3E regulates apoptosis in the intestinal epithelium during the development of colitis. Biochem Pharmacol. 166:264–273. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Parker A, Vaux L, Patterson AM, Modasia A, Muraro D, Fletcher AG, Byrne HM, Maini PK, Watson AJM and Pin C: Elevated apoptosis impairs epithelial cell turnover and shortens villi in TNF-driven intestinal inflammation. Cell Death Dis. 10:1082019. View Article : Google Scholar : PubMed/NCBI

37 

Yan R, Liang X and Hu J: MALAT1 promotes colonic epithelial cell apoptosis and pyroptosis by sponging miR-22-3p to enhance NLRP3 expression. PeerJ. 12:e184492024. View Article : Google Scholar : PubMed/NCBI

38 

Yang L, Wu G, Wu Q, Peng L and Yuan L: METTL3 overexpression aggravates LPS-induced cellular inflammation in mouse intestinal epithelial cells and DSS-induced IBD in mice. Cell Death Discov. 8:622022. View Article : Google Scholar : PubMed/NCBI

39 

Wu MM, Wang QM, Huang BY, Mai CT, Wang CL, Wang TT and Zhang XJ: Dioscin ameliorates murine ulcerative colitis by regulating macrophage polarization. Pharmacol Res. 172:1057962021. View Article : Google Scholar : PubMed/NCBI

40 

Palmela C, Chevarin C, Xu Z, Torres J, Sevrin G, Hirten R, Barnich N, Ng SC and Colombel JF: Adherent-invasive Escherichia coli in inflammatory bowel disease. Gut. 67:574–587. 2018. View Article : Google Scholar

41 

Chen B, Wang Y, Niu Y and Li S: Acalypha australis L. Extract attenuates DSS-induced ulcerative colitis in mice by regulating inflammatory factor release and blocking NF-κB activation. J Med Food. 26:663–671. 2023. View Article : Google Scholar : PubMed/NCBI

42 

Lasa JS, Olivera PA, Danese S and Peyrin-Biroulet L: Efficacy and safety of biologics and small molecule drugs for patients with moderate-to-severe ulcerative colitis: A systematic review and network meta-analysis. Lancet Gastroenterol Hepatol. 7:161–170. 2022. View Article : Google Scholar

43 

Geng Z, Zuo L, Li J, Yin L, Yang J, Duan T, Wang L, Zhang X, Song X, Wang Y and Hu J: Ginkgetin improved experimental colitis by inhibiting intestinal epithelial cell apoptosis through EGFR/PI3K/AKT signaling. FASEB J. 38:e238172024. View Article : Google Scholar : PubMed/NCBI

44 

Lokman MS, Kassab RB, Salem FAM, Elshopakey GE, Hussein A, Aldarmahi AA, Theyab A, Alzahrani KJ, Hassan KE, Alsharif KF, et al: Asiatic acid rescues intestinal tissue by suppressing molecular, biochemical, and histopathological changes associated with the development of ulcerative colitis. Biosci Rep. 44:BSR202320042024. View Article : Google Scholar : PubMed/NCBI

45 

He Z, Liu J and Liu Y: Daphnetin attenuates intestinal inflammation, oxidative stress, and apoptosis in ulcerative colitis via inhibiting REG3A-dependent JAK2/STAT3 signaling pathway. Environ Toxicol. 38:2132–2142. 2023. View Article : Google Scholar : PubMed/NCBI

46 

Hu Q, Xie J, Jiang T, Gao P, Chen Y, Zhang W, Yan J, Zeng J, Ma X and Zhao Y: Paeoniflorin alleviates DSS-induced ulcerative colitis by suppressing inflammation, oxidative stress, and apoptosis via regulating serum metabolites and inhibiting CDC42/JNK signaling pathway. Int Immunopharmacol. 142(Pt A): 1130392024. View Article : Google Scholar : PubMed/NCBI

47 

Gao F, Wu S, Zhang K, Xu Z, Zhang X, Zhu Z and Quan F: Goat milk exosomes ameliorate ulcerative colitis in mice through modulation of the intestinal barrier, gut microbiota, and metabolites. J Agric Food Chem. 72:23196–23210. 2024. View Article : Google Scholar : PubMed/NCBI

48 

Zhu F, Wei C, Wu H, Shuai B, Yu T, Gao F, Yuan Y, Zuo D, Liu X, Zhang L and Fan H: Hypoxic mesenchymal stem cell-derived exosomes alleviate ulcerative colitis injury by limiting intestinal epithelial cells reactive oxygen species accumulation and DNA damage through HIF-1α. Int Immunopharmacol. 113(Pt A): 1094262022. View Article : Google Scholar : PubMed/NCBI

49 

Li H, Fan C, Lu H, Feng C, He P, Yang X, Xiang C, Zuo J and Tang W: Protective role of berberine on ulcerative colitis through modulating enteric glial cells-intestinal epithelial cells-immune cells interactions. Acta Pharm Sin B. 10:447–461. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Gao C, Liu L, Zhou Y, Bian Z, Wang S and Wang Y: Novel drug delivery systems of Chinese medicine for the treatment of inflammatory bowel disease. Chin Med. 14:232019. View Article : Google Scholar : PubMed/NCBI

51 

Liu C, Gong Q, Liu W, Zhao Y, Yan X and Yang T: Berberine-loaded PLGA nanoparticles alleviate ulcerative colitis by targeting IL-6/IL-6R axis. J Transl Med. 22:9632024. View Article : Google Scholar : PubMed/NCBI

52 

Buck A, Rezaei K, Quazi A, Goldmeier G, Silverglate B and Grossberg GT: The donepezil transdermal system for the treatment of patients with mild, moderate, or severe Alzheimer's disease: A critical review. Expert Rev Neurother. 24:607–614. 2024. View Article : Google Scholar : PubMed/NCBI

53 

Gwon HJ, Cho W, Choi SW, Lim DS, Tanriverdi EÇ, Abd El-Aty AM, Jeong JH and Jung TW: Donepezil improves skeletal muscle insulin resistance in obese mice via the AMPK/FGF21-mediated suppression of inflammation and ferroptosis. Arch Pharm Res. 47:940–953. 2024. View Article : Google Scholar : PubMed/NCBI

54 

Li A, Zhang J, Chen K, Wang J, Xu A and Wang Z: Donepezil attenuates inflammation and apoptosis in ulcerative colitis via regulating LRP1/AMPK/NF-κB signaling. Pathol Int. 73:549–559. 2023. View Article : Google Scholar : PubMed/NCBI

55 

Li C, Xu Y, Gao T, Zhang S, Lin Z, Gu S, Fang Y, Yuan X, Yu S, Jiang Q, et al: Ruxolitinib alleviates inflammation, apoptosis, and intestinal barrier leakage in ulcerative colitis via STAT3. Inflamm Bowel Dis. 29:1191–1201. 2023. View Article : Google Scholar : PubMed/NCBI

56 

Tang R, Xu J, Zhang B, Liu J, Liang C, Hua J, Meng Q, Yu X and Shi S: Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J Hematol Oncol. 13:1102020. View Article : Google Scholar : PubMed/NCBI

57 

Newton K: RIPK1 and RIPK3: Critical regulators of inflammation and cell death. Trends Cell Biol. 25:347–353. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Degterev A, Ofengeim D and Yuan J: Targeting RIPK1 for the treatment of human diseases. Proc Natl Acad Sci USA. 116:9714–9722. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Zhang W, Zhu C, Liao Y, Zhou M, Xu W and Zou Z: Caspase-8 in inflammatory diseases: A potential therapeutic target. Cell Mol Biol Lett. 29:1302024. View Article : Google Scholar : PubMed/NCBI

60 

Mifflin L, Ofengeim D and Yuan J: Receptor-interacting protein kinase 1 (RIPK1) as a therapeutic target. Nat Rev Drug Discov. 19:553–571. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Zhou Y, Xiang Y, Liu S, Li C, Dong J, Kong X, Ji X, Cheng X and Zhang L: RIPK3 signaling and its role in regulated cell death and diseases. Cell Death Discov. 10:2002024. View Article : Google Scholar : PubMed/NCBI

62 

Yuan J, Amin P and Ofengeim D: Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat Rev Neurosci. 20:19–33. 2019. View Article : Google Scholar :

63 

Karlowitz R and van Wijk SJL: Surviving death: emerging concepts of RIPK3 and MLKL ubiquitination in the regulation of necroptosis. FEBS J. 290:37–54. 2023. View Article : Google Scholar

64 

Mohammed S, Thadathil N, Selvarani R, Nicklas EH, Wang D, Miller BF, Richardson A and Deepa SS: Necroptosis contributes to chronic inflammation and fibrosis in aging liver. Aging Cell. 20:e135122021. View Article : Google Scholar : PubMed/NCBI

65 

Jia Z, Xu C, Shen J, Xia T, Yang J and He Y: The natural compound celastrol inhibits necroptosis and alleviates ulcerative colitis in mice. Int Immunopharmacol. 29:552–559. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Lehle AS, Farin HF, Marquardt B, Michels BE, Magg T, Li Y, Liu Y, Ghalandary M, Lammens K, Hollizeck S, et al: Intestinal inflammation and dysregulated immunity in patients with inherited caspase-8 deficiency. Gastroenterology. 156:275–278. 2019. View Article : Google Scholar

67 

Pierdomenico M, Negroni A, Stronati L, Vitali R, Prete E, Bertin J, Gough PJ, Aloi M and Cucchiara S: Necroptosis is active in children with inflammatory bowel disease and contributes to heighten intestinal inflammation. Am J Gastroenterol. 109:279–287. 2014. View Article : Google Scholar

68 

Liu L, Liang L, Yang C, Zhou Y and Chen Y: Extracellular vesicles of Fusobacterium nucleatum compromise intestinal barrier through targeting RIPK1-mediated cell death pathway. Gut Microbes. 13:1–20. 2021. View Article : Google Scholar

69 

Liu C, Wang H, Han L, Zhu Y, Ni S, Zhi J, Yang X, Zhi J, Sheng T, Li H and Hu Q: Targeting P2Y(14)R protects against necroptosis of intestinal epithelial cells through PKA/CREB/RIPK1 axis in ulcerative colitis. Nat Commun. 15:20832024. View Article : Google Scholar : PubMed/NCBI

70 

Dunker W, Ye X, Zhao Y, Liu L, Richardson A and Karijolich J: TDP-43 prevents endogenous RNAs from triggering a lethal RIG-I-dependent interferon response. Cell Rep. 35:1089762021. View Article : Google Scholar : PubMed/NCBI

71 

Maelfait J, Liverpool L and Rehwinkel J: Nucleic acid sensors and programmed cell death. J Mol Biol. 432:552–568. 2020. View Article : Google Scholar :

72 

Li Y, Zou C, Chen C, Li S, Zhu Z, Fan Q, Pang R, Li F, Chen Z, Wang Z, et al: Myeloid-derived MIF drives RIPK1-mediated cerebromicrovascular endothelial cell death to exacerbate ischemic brain injury. Proc Natl Acad Sci USA. 120:e22190911202023. View Article : Google Scholar : PubMed/NCBI

73 

Soppert J, Kraemer S, Beckers C, Averdunk L, Möllmann J, Denecke B, Goetzenich A, Marx G, Bernhagen J and Stoppe C: Soluble CD74 reroutes MIF/CXCR4/AKT-mediated survival of cardiac myofibroblasts to necroptosis. J Am Heart Assoc. 7:e0093842018. View Article : Google Scholar : PubMed/NCBI

74 

Bao J, Ye B and Ren Y: ABIN1 inhibits inflammation through necroptosis-dependent pathway in ulcerative colitis. Genet Res (Camb). 2022:93135592022. View Article : Google Scholar : PubMed/NCBI

75 

Duan C, Xu X, Lu X, Wang L and Lu Z: RIP3 knockdown inhibits necroptosis of human intestinal epithelial cells via TLR4/MyD88/NF-κB signaling and ameliorates murine colitis. BMC Gastroenterol. 22:1372022. View Article : Google Scholar

76 

Zhong Y, Tu Y, Ma Q, Chen L, Zhang W, Lu X, Yang S, Wang Z and Zhang L: Curcumin alleviates experimental colitis in mice by suppressing necroptosis of intestinal epithelial cells. Front Pharmacol. 14:11706372023. View Article : Google Scholar : PubMed/NCBI

77 

Zhang J, Lei H, Hu X and Dong W: Hesperetin ameliorates DSS-induced colitis by maintaining the epithelial barrier via blocking RIPK3/MLKL necroptosis signaling. Eur J Pharmacol. 873:1729922020. View Article : Google Scholar : PubMed/NCBI

78 

Shen X, Chen H, Wen T, Liu L, Yang Y, Xie F and Wang L: A natural chalcone cardamonin inhibits necroptosis and ameliorates dextran sulfate sodium (DSS)-induced colitis by targeting RIPK1/3 kinases. Eur J Pharmacol. 954:1758402023. View Article : Google Scholar : PubMed/NCBI

79 

Li Z, Wang H, Wang Z and Geng Y: Pine pollen polysaccharides' and sulfated polysaccharides' effects on UC mice through modulation of cell tight junctions and RIPK3-dependent necroptosis pathways. Molecules. 27:76822022. View Article : Google Scholar : PubMed/NCBI

80 

Wang Y, Zhang B, Liu S, Xu E and Wang Z: The traditional herb Sargentodoxa cuneata alleviates DSS-induced colitis by attenuating epithelial barrier damage via blocking necroptotic signaling. J Ethnopharmacol. 319(Pt 3): 1173732024. View Article : Google Scholar

81 

Zhou XL, Yang J, Qu XJ, Meng J, Miao RR and Cui SX: M10, a Myricetin-3-O-b-D-lactose sodium salt, prevents ulcerative colitis through inhibiting necroptosis in mice. Front Pharmacol. 11:5573122020. View Article : Google Scholar : PubMed/NCBI

82 

Peng C, Wu C, Xu X, Pan L, Lou Z, Zhao Y, Jiang H, He Z and Ruan B: Indole-3-carbinol ameliorates necroptosis and inflammation of intestinal epithelial cells in mice with ulcerative colitis by activating aryl hydrocarbon receptor. Exp Cell Res. 404:1126382021. View Article : Google Scholar : PubMed/NCBI

83 

Zeng YS, Peng J, Gao XF, Tian D, Zhan W, Liu J, Hu XJ, Huang S, Tian ST, Qiu L, et al: A novel gut-restricted RIPK1 inhibitor, SZ-15, ameliorates DSS-induced ulcerative colitis. Eur J Pharmacol. 937:1753812022. View Article : Google Scholar : PubMed/NCBI

84 

Zhang C, He A, Liu S, He Q, Luo Y, He Z, Chen Y, Tao A and Yan J: Inhibition of HtrA2 alleviated dextran sulfate sodium (DSS)-induced colitis by preventing necroptosis of intestinal epithelial cells. Cell Death Dis. 10:3442019. View Article : Google Scholar : PubMed/NCBI

85 

Zhang C, Luo Y, He Q, Liu S, He A and Yan J: A pan-RAF inhibitor LY3009120 inhibits necroptosis by preventing phosphorylation of RIPK1 and alleviates dextran sulfate sodium-induced colitis. Clin Sci (Lond). 133:919–932. 2019. View Article : Google Scholar : PubMed/NCBI

86 

Kayagaki N, Webster JD and Newton K: Control of cell death in health and disease. Annu Rev Pathol. 19:157–180. 2024. View Article : Google Scholar

87 

Krzyzowska M, Shestakov A, Eriksson K and Chiodi F: Role of Fas/FasL in regulation of inflammation in vaginal tissue during HSV-2 infection. Cell Death Dis. 2:e1322011. View Article : Google Scholar : PubMed/NCBI

88 

Ma D, Wang X, Liu J, Cui Y, Luo S and Wang F: The development of necroptosis: What we can learn. Cell Stress Chaperones. 28:969–987. 2023. View Article : Google Scholar : PubMed/NCBI

89 

Nakano H, Murai S and Moriwaki K: Regulation of the release of damage-associated molecular patterns from necroptotic cells. Biochem J. 479:677–685. 2022. View Article : Google Scholar : PubMed/NCBI

90 

Ma M, Jiang W and Zhou R: DAMPs and DAMP-sensing receptors in inflammation and diseases. Immunity. 57:752–771. 2024. View Article : Google Scholar : PubMed/NCBI

91 

Chen M, Rong R and Xia X: Spotlight on pyroptosis: role in pathogenesis and therapeutic potential of ocular diseases. J Neuroinflammation. 19:1832022. View Article : Google Scholar : PubMed/NCBI

92 

Wei X, Xie F, Zhou X, Wu Y, Yan H, Liu T, Huang J, Wang F, Zhou F and Zhang L: Role of pyroptosis in inflammation and cancer. Cell Mol Immunol. 19:971–992. 2022. View Article : Google Scholar : PubMed/NCBI

93 

Fu J and Wu H: Structural mechanisms of NLRP3 inflammasome assembly and activation. Annu Rev Immunol. 41:301–316. 2023. View Article : Google Scholar : PubMed/NCBI

94 

Biasizzo M and Kopitar-Jerala N: Interplay Between NLRP3 Inflammasome and Autophagy. Front Immunol. 11:5918032020. View Article : Google Scholar : PubMed/NCBI

95 

Que X, Zheng S, Song Q, Pei H and Zhang P: Fantastic voyage: The journey of NLRP3 inflammasome activation. Genes Dis. 11:819–829. 2024. View Article : Google Scholar

96 

Nabavi-Rad A, Sadeghi A, Asadzadeh Aghdaei H, Yadegar A, Smith SM and Zali MR: The double-edged sword of probiotic supplementation on gut microbiota structure in Helicobacter pylori management. Gut Microbes. 14:21086552022. View Article : Google Scholar : PubMed/NCBI

97 

Wang Y, Yuan H, Shen D, Liu S, Kong W, Zheng K, Yang J and Ge L: Artemisinin attenuated ischemic stroke induced pyroptosis by inhibiting ROS/TXNIP/NLRP3/Caspase-1 signaling pathway. Biomed Pharmacother. 177:1168942024. View Article : Google Scholar : PubMed/NCBI

98 

Zhang MY, Jiang YX, Yang YC, Liu JY, Huo C, Ji XL and Qu YQ: Cigarette smoke extract induces pyroptosis in human bronchial epithelial cells through the ROS/NLRP3/caspase-1 pathway. Life Sci. 269:1190902021. View Article : Google Scholar : PubMed/NCBI

99 

Burdette BE, Esparza AN, Zhu H and Wang S: Gasdermin D in pyroptosis. Acta Pharm Sin B. 11:2768–2782. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Yang Y, Li S, Liu K, Zhang Y, Zhu F, Ben T, Chen Z and Zhi F: Lipocalin-2-mediated intestinal epithelial cells pyroptosis via NF-κB/NLRP3/GSDMD signaling axis adversely affects inflammation in colitis. Biochim Biophys Acta Mol Basis Dis. 1870:1672792024. View Article : Google Scholar

101 

Jena KK, Mambu J, Boehmer D, Sposito B, Millet V, de Sousa Casal J, Muendlein HI, Spreafico R, Fenouil R, Spinelli L, et al: Type III interferons induce pyroptosis in gut epithelial cells and impair mucosal repair. Cell. 187:7533–7550.e23. 2024. View Article : Google Scholar : PubMed/NCBI

102 

Liu X, Zhou M, Dai Z, Luo S, Shi Y, He Z and Chen Y: Salidroside alleviates ulcerative colitis via inhibiting macrophage pyroptosis and repairing the dysbacteriosis-associated Th17/Treg imbalance. Phytother Res. 37:367–382. 2023. View Article : Google Scholar

103 

Zhang S, Zhong R, Tang S, Chen L and Zhang H: Metabolic regulation of the Th17/Treg balance in inflammatory bowel disease. Pharmacol Res. 203:1071842024. View Article : Google Scholar : PubMed/NCBI

104 

Zhang D, Ge F, Ji J, Li YJ, Zhang FR, Wang SY, Zhang SJ, Zhang DM and Chen M: β-sitosterol alleviates dextran sulfate sodium-induced experimental colitis via inhibition of NLRP3/Caspase-1/GSDMD-mediated pyroptosis. Front Pharmacol. 14:12184772023. View Article : Google Scholar

105 

Shen J, Zhao Y and Cui W: Astragalus mongholicus Bunge extract improves ulcerative colitis by promoting PLCB2 to inhibit colonic epithelial cell pyroptosis. J Ethnopharmacol. 334:1185542024. View Article : Google Scholar : PubMed/NCBI

106 

Shao M, Yan Y, Zhu F, Yang X, Qi Q, Yang F, Hao T, Lin Z, He P, Zhou Y, et al: Artemisinin analog SM934 alleviates epithelial barrier dysfunction via inhibiting apoptosis and caspase-1-mediated pyroptosis in experimental colitis. Front Pharmacol. 13:8490142022. View Article : Google Scholar : PubMed/NCBI

107 

Zhang W, Wang W, Shen C, Wang X, Pu Z and Yin Q: Network pharmacology for systematic understanding of Schisandrin B reduces the epithelial cells injury of colitis through regulating pyroptosis by AMPK/Nrf2/NLRP3 inflammasome. Aging (Albany NY). 13:23193–23209. 2021. View Article : Google Scholar : PubMed/NCBI

108 

Zhao P, Ning J, Huang J and Huang X: Mechanism of resveratrol on LPS/ATP-induced pyroptosis and inflammatory response in HT29 cells. Autoimmunity. 57:24270942024. View Article : Google Scholar : PubMed/NCBI

109 

Chang Y, Zhang Y, Jiang Y, Zhao L, Lv C, Huang Q, Guan J and Jin S: From hair to colon: Hair follicle-derived MSCs alleviate pyroptosis in DSS-Induced ulcerative colitis by releasing exosomes in a paracrine manner. Oxid Med Cell Longev. 2022:90975302022. View Article : Google Scholar : PubMed/NCBI

110 

Wang D, Xue H, Tan J, Liu P, Qiao C, Pang C and Zhang L: Bone marrow mesenchymal stem cells-derived exosomes containing miR-539-5p inhibit pyroptosis through NLRP3/caspase-1 signalling to alleviate inflammatory bowel disease. Inflamm Res. 71:833–846. 2022. View Article : Google Scholar : PubMed/NCBI

111 

Yan R, Liang X and Hu J: miR-141-3p alleviates ulcerative colitis by targeting SUGT1 to inhibit colonic epithelial cell pyroptosis. Autoimmunity. 56:22209882023. View Article : Google Scholar : PubMed/NCBI

112 

Oraby MA, Abdel Mageed SS, Amr Raouf A, Abdelshafy DA, Ahmed EF, Khalil RT, Mangoura SA and Fadaly DS: Remdesivir ameliorates ulcerative colitis-propelled cell inflammation and pyroptosis in acetic acid rats by restoring SIRT6/FoxC1 pathway. Int Immunopharmacol. 137:1124652024. View Article : Google Scholar : PubMed/NCBI

113 

Li J, Wu H, Zhou J, Jiang R, Zhuo Z, Yang Q, Chen H and Sha W: Ruscogenin attenuates ulcerative colitis in mice by inhibiting caspase-1-dependent pyroptosis via the TLR4/NF-κB signaling pathway. Biomedicines. 12:9892024. View Article : Google Scholar

114 

Chen L, Liu D, Mao M, Liu W, Wang Y, Liang Y, Cao W and Zhong X: Betaine ameliorates acute sever ulcerative colitis by inhibiting oxidative stress induced inflammatory pyroptosis. Mol Nutr Food Res. 66:e22003412022. View Article : Google Scholar : PubMed/NCBI

115 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Mou Y, Wang J, Wu J, He D, Zhang C, Duan C and Li B: Ferroptosis, a new form of cell death: Opportunities and challenges in cancer. J Hematol Oncol. 12:342019. View Article : Google Scholar : PubMed/NCBI

117 

Chen AC, Donovan A, Ned-Sykes R and Andrews NC: Noncanonical role of transferrin receptor 1 is essential for intestinal homeostasis. Proc Natl Acad Sci USA. 112:11714–11719. 2015. View Article : Google Scholar : PubMed/NCBI

118 

Lomphithak T, Sae-Fung A, Sprio S, Tampieri A, Jitkaew S and Fadeel B: Exploiting the ferroaddiction of pancreatic cancer cells using Fe-doped nanoparticles. Nanomedicine. 55:1027142024. View Article : Google Scholar : PubMed/NCBI

119 

Cai H, Chen S, Zhu Y, Zhuang S, Wang J, Niu X, Cui T, Huang H, Ao R, Yu M, et al: A pH/STEAP cascade-responsive nanomedicine with self-supplied peroxide for precise chemodynamic therapy. Adv Healthc Mater. 14:e25007522025. View Article : Google Scholar : PubMed/NCBI

120 

Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS and Stockwell BR: Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 113:E4966–E4975. 2016. View Article : Google Scholar : PubMed/NCBI

121 

Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, et al: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. 2014. View Article : Google Scholar : PubMed/NCBI

122 

Sun SP, Lu YF, Li H, Weng CY, Chen JJ, Lou YJ, Lyu D and Lyu B: AMPK activation alleviated dextran sulfate sodium-induced colitis by inhibiting ferroptosis. J Dig Dis. 24:213–223. 2023. View Article : Google Scholar : PubMed/NCBI

123 

Guo M, Du X and Wang X: Inhibition of ferroptosis: A new direction in the treatment of ulcerative colitis by traditional Chinese medicine. J Ethnopharmacol. 324:1177872024. View Article : Google Scholar : PubMed/NCBI

124 

Huang F, Zhang S, Li X, Huang Y, He S and Luo L: STAT3-mediated ferroptosis is involved in ulcerative colitis. Free Radic Biol Med. 188:375–385. 2022. View Article : Google Scholar : PubMed/NCBI

125 

Chen Y, Zhang P, Chen W and Chen G: Ferroptosis mediated DSS-induced ulcerative colitis associated with Nrf2/HO-1 signaling pathway. Immunol Lett. 225:9–15. 2020. View Article : Google Scholar : PubMed/NCBI

126 

Zhu J, Wu Y, Ge X, Chen X and Mei Q: Discovery and validation of ferroptosis-associated genes of ulcerative colitis. J Inflamm Res. 17:4467–4482. 2024. View Article : Google Scholar : PubMed/NCBI

127 

Yokote A, Imazu N, Umeno J, Kawasaki K, Fujioka S, Fuyuno Y, Matsuno Y, Moriyama T, Miyawaki K, Akashi K, et al: Ferroptosis in the colon epithelial cells as a therapeutic target for ulcerative colitis. J Gastroenterol. 58:868–882. 2023. View Article : Google Scholar : PubMed/NCBI

128 

Lam IH, Chan CI, Han M, Li L and Yu HH: ACSL4 mediates inflammatory bowel disease and contributes to LPS-induced intestinal epithelial cell dysfunction by activating ferroptosis and inflammation. Open Med (Wars). 19:202409932024. View Article : Google Scholar : PubMed/NCBI

129 

Chen Y, Wang J, Li J, Zhu J, Wang R, Xi Q, Wu H, Shi T and Chen W: Astragalus polysaccharide prevents ferroptosis in a murine model of experimental colitis and human Caco-2 cells via inhibiting NRF2/HO-1 pathway. Eur J Pharmacol. 911:1745182021. View Article : Google Scholar

130 

Ni J, Zhang L, Feng G, Bao W, Wang Y, Huang Y, Chen T, Chen J, Cao X, You K, et al: Vanillic acid restores homeostasis of intestinal epithelium in colitis through inhibiting CA9/STIM1-mediated ferroptosis. Pharmacol Res. 202:1071282024. View Article : Google Scholar : PubMed/NCBI

131 

Long D, Mao C, Huang Y, Xu Y and Zhu Y: Ferroptosis in ulcerative colitis: Potential mechanisms and promising therapeutic targets. Biomed Pharmacother. 175:1167222024. View Article : Google Scholar : PubMed/NCBI

132 

Wu X, Zhao L, Yu Z and Zhang K: Buddlejasaponin IVb Alleviates DSS-Induced ulcerative colitis through the Nrf2/GPX4 pathway and gut microbiota modulation. J Agric Food Chem. 72:23183–23195. 2024. View Article : Google Scholar : PubMed/NCBI

133 

Ji W, Zhang Y, Qian X, Hu C and Huo Y: Palmatine alleviates inflammation and modulates ferroptosis against dextran sulfate sodium (DSS)-induced ulcerative colitis. Int Immunopharmacol. 143(Pt 2): 1133962024. View Article : Google Scholar : PubMed/NCBI

134 

Wu Y, Ran L, Yang Y, Gao X, Peng M, Liu S, Sun L, Wan J, Wang Y, Yang K, et al: Deferasirox alleviates DSS-induced ulcerative colitis in mice by inhibiting ferroptosis and improving intestinal microbiota. Life Sci. 314:1213122023. View Article : Google Scholar

135 

Porter JB: A risk-benefit assessment of iron-chelation therapy. Drug Saf. 17:407–421. 1997. View Article : Google Scholar

136 

Di Paola A, Tortora C, Argenziano M, Marrapodi MM and Rossi F: Emerging roles of the iron chelators in inflammation. Int J Mol Sci. 23:79772022. View Article : Google Scholar : PubMed/NCBI

137 

Chen H, Qian Y, Jiang C, Tang L, Yu J, Zhang L, Dai Y and Jiang G: Butyrate ameliorated ferroptosis in ulcerative colitis through modulating Nrf2/GPX4 signal pathway and improving intestinal barrier. Biochim Biophys Acta Mol Basis Dis. 1870:1669842024. View Article : Google Scholar

138 

Gao S, Sun C and Kong J: Vitamin D attenuates ulcerative colitis by inhibiting ACSL4-mediated ferroptosis. Nutrients. 15:48452023. View Article : Google Scholar : PubMed/NCBI

139 

Shi J, Ji S, Xu M, Wang Y and Shi H: Selenium inhibits ferroptosis in ulcerative colitis through the induction of Nrf2/Gpx4. Clin Res Hepatol Gastroenterol. 48:1024672024. View Article : Google Scholar : PubMed/NCBI

140 

Gao BB, Wang L, Li LZ, Fei ZQ, Wang YY, Zou XM, Huang MC, Lei SS and Li B: Beneficial effects of oxymatrine from Sophora flavescens on alleviating Ulcerative colitis by improving inflammation and ferroptosis. J Ethnopharmacol. 332:1183852024. View Article : Google Scholar : PubMed/NCBI

141 

Li W, Wang Y, Zhang Y, Fan Y, Liu J, Zhu K, Jiang S and Duan J: Lizhong decoction ameliorates ulcerative colitis by inhibiting ferroptosis of enterocytes via the Nrf2/SLC7A11/GPX4 pathway. J Ethnopharmacol. 326:1179662024. View Article : Google Scholar : PubMed/NCBI

142 

Wei Z, Hang S, Wiredu Ocansey DK, Zhang Z, Wang B, Zhang X and Mao F: Human umbilical cord mesenchymal stem cells derived exosome shuttling mir-129-5p attenuates inflammatory bowel disease by inhibiting ferroptosis. J Nanobiotechnology. 21:1882023. View Article : Google Scholar : PubMed/NCBI

143 

Zhu Y, Qin H, Sun C, Shao B, Li G, Qin Y, Kong D, Ren S, Wang H, Wang Z, et al: Endometrial regenerative cell-derived exosomes attenuate experimental colitis through downregulation of intestine ferroptosis. Stem Cells Int. 2022:30141232022. View Article : Google Scholar : PubMed/NCBI

144 

Takabatake Y, Kimura T, Takahashi A and Isaka Y: Autophagy and the kidney: health and disease. Nephrol Dial Transplant. 29:1639–1647. 2014. View Article : Google Scholar : PubMed/NCBI

145 

Anding AL and Baehrecke EH: Autophagy in cell life and cell death. Curr Top Dev Biol. 114:67–91. 2015. View Article : Google Scholar : PubMed/NCBI

146 

Mizushima N and Komatsu M: Autophagy: renovation of cells and tissues. Cell. 147:728–741. 2011. View Article : Google Scholar : PubMed/NCBI

147 

Klionsky DJ, Cregg JM, Dunn WA Jr, Emr SD, Sakai Y, Sandoval IV, Sibirny A, Subramani S, Thumm M, Veenhuis M and Ohsumi Y: A unified nomenclature for yeast autophagy-related genes. Dev Cell. 5:539–545. 2003. View Article : Google Scholar : PubMed/NCBI

148 

Mizushima N: A brief history of autophagy from cell biology to physiology and disease. Nat Cell Biol. 20:521–527. 2018. View Article : Google Scholar : PubMed/NCBI

149 

Yu L, Chen Y and Tooze SA: Autophagy pathway: Cellular and molecular mechanisms. Autophagy. 14:207–215. 2018. View Article : Google Scholar :

150 

Shen HM and Mizushima N: At the end of the autophagic road: An emerging understanding of lysosomal functions in autophagy. Trends Biochem Sci. 39:61–71. 2014. View Article : Google Scholar

151 

Takeshige K, Baba M, Tsuboi S, Noda T and Ohsumi Y: Autophagy in yeast demonstrated with proteinase-deficient mutants and conditions for its induction. J Cell Biol. 119:301–311. 1992. View Article : Google Scholar : PubMed/NCBI

152 

Liu Z and Wang H: Probiotics alleviate inflammatory bowel disease in mice by regulating intestinal microorganisms-bile acid-NLRP3 inflammasome pathway. Acta Biochim Pol. 68:687–693. 2021.PubMed/NCBI

153 

Zhang C, Yan J, Xiao Y, Shen Y, Wang J, Ge W and Chen Y: Inhibition of autophagic degradation process contributes to claudin-2 expression increase and epithelial tight junction dysfunction in TNF-α treated cell monolayers. Int J Mol Sci. 18:572017.

154 

Hu X, Deng J, Yu T, Chen S, Ge Y, Zhou Z, Guo Y, Ying H, Zhai Q, Chen Y, et al: ATF4 deficiency promotes intestinal inflammation in mice by reducing uptake of glutamine and expression of antimicrobial peptides. Gastroenterology. 156:1098–1111. 2019. View Article : Google Scholar

155 

Chen Z, Gu Q and Chen R: miR-146a-5p regulates autophagy and NLRP3 inflammasome activation in epithelial barrier damage in the in vitro cell model of ulcerative colitis through the RNF8/Notch1/mTORC1 pathway. Immunobiology. 228:1523862023. View Article : Google Scholar : PubMed/NCBI

156 

Zhou M, Xu W, Wang J, Yan J, Shi Y, Zhang C, Ge W, Wu J, Du P and Chen Y: Boosting mTOR-dependent autophagy via upstream TLR4-MyD88-MAPK signalling and downstream NF-κB pathway quenches intestinal inflammation and oxidative stress injury. EBioMedicine. 35:345–360. 2018. View Article : Google Scholar : PubMed/NCBI

157 

Martinon F, Burns K and Tschopp J: The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 10:417–426. 2002. View Article : Google Scholar : PubMed/NCBI

158 

Takahama M, Akira S and Saitoh T: Autophagy limits activation of the inflammasomes. Immunol Rev. 281:62–73. 2018. View Article : Google Scholar

159 

Shen T, Li S, Cai LD, Liu JL, Wang CY, Gan WJ, Li XM, Wang JR, Sun LN, Deng M, et al: Erbin exerts a protective effect against inflammatory bowel disease by suppressing autophagic cell death. Oncotarget. 9:12035–12049. 2018. View Article : Google Scholar : PubMed/NCBI

160 

Pan SM, Wang CL, Hu ZF, Zhang ML, Pan ZF, Zhou RY, Wang XJ, Huang SW, Li YY, Wang Q, et al: Baitouweng decoction repairs the intestinal barrier in DSS-induced colitis mice via regulation of AMPK/mTOR-mediated autophagy. J Ethnopharmacol. 318(Pt A): 1168882024. View Article : Google Scholar

161 

Zhang H, Lang W, Liu X, Bai J, Jia Q and Shi Q: Procyanidin A1 alleviates DSS-induced ulcerative colitis via regulating AMPK/mTOR/p70S6K-mediated autophagy. J Physiol Biochem. 78:213–227. 2022. View Article : Google Scholar : PubMed/NCBI

162 

Watanabe-Yasuoka Y, Gotou A, Shimizu S and Sashihara T: Lactiplantibacillus plantarum OLL2712 Induces Autophagy via MYD88 and strengthens tight junction integrity to promote the barrier function in intestinal epithelial cells. Nutrients. 15:26552023. View Article : Google Scholar : PubMed/NCBI

163 

Liu Y, Deng S, Sun L, He H, Zhou Q, Fan H, Yang C and Yang J: Compound sophorae decoction mitigates DSS-induced ulcerative colitis by activating autophagy through PI3K-AKT pathway: A integrative research combining network pharmacology and in vivo animal model validation. J Ethnopharmacol. 337:1188852025. View Article : Google Scholar

164 

Qiao D, Liu X, Zhang Y, Zhang Z, Tang Y, Chen Q, Shi Y, Chen Y, Tang Z and Dai Y: Jianpi-Qingchang decoction alleviates ulcerative colitis by modulating endoplasmic reticulum stress-related autophagy in intestinal epithelial cells. Biomed Pharmacother. 158:1141332023. View Article : Google Scholar

165 

Zhao L, Jiang T, Zhang Y and Shen Z: Epimedium polysaccharides ameliorate ulcerative colitis by inhibiting oxidative stress and regulating autophagy. J Sci Food Agric. 105:2655–2670. 2025. View Article : Google Scholar

166 

Xu Y, Tian Y, Li F, Wang Y, Yang J, Gong H, Wan X and Ouyang M: Circular RNA HECTD1 mitigates ulcerative colitis by promoting enterocyte autophagy Via miR-182-5p/HuR axis. Inflamm Bowel Dis. 28:273–288. 2022. View Article : Google Scholar

167 

Chen B, Carr L and Dun XP: Dynamic expression of Slit1-3 and Robo1-2 in the mouse peripheral nervous system after injury. Neural Regen Res. 15:948–958. 2020. View Article : Google Scholar

168 

Wang L, Zheng J, Pathak JL, Chen Y, Liang D, Yang L, Sun H, Zhong M, Wu L, Li L, et al: SLIT2 overexpression in periodontitis intensifies inflammation and alveolar bone loss, possibly via the activation of MAPK pathway. Front Cell Dev Biol. 8:5932020. View Article : Google Scholar : PubMed/NCBI

169 

Xie J, Li L, Deng S, Chen J, Gu Q, Su H, Wen L, Wang S, Lin C, Qi C, et al: Slit2/Robo1 mitigates dss-induced ulcerative colitis by activating autophagy in intestinal stem cell. Int J Biol Sci. 16:1876–1887. 2020. View Article : Google Scholar : PubMed/NCBI

170 

Wei S, Zhang J, Wu X, Chen M, Huang H, Zeng S, Xiang Z, Li X and Dong W: Fusobacterium nucleatum Extracellular Vesicles Promote Experimental Colitis by Modulating Autophagy via the miR-574-5p/CARD3 Axis. Inflamm Bowel Dis. 29:9–26. 2023. View Article : Google Scholar

171 

Wang XJ, Zhang D, Yang YT, Li XY, Li HN, Zhang XP, Long JY, Lu YQ, Liu L, Yang G, et al: Suppression of microRNA-222-3p ameliorates ulcerative colitis and colitis-associated colorectal cancer to protect against oxidative stress via targeting BRG1 to activate Nrf2/HO-1 signaling pathway. Front Immunol. 14:10898092023. View Article : Google Scholar : PubMed/NCBI

172 

Liang H, Zhang F, Wang W, Zhao W, Zhou J, Feng Y, Wu J, Li M, Bai X, Zeng Z, et al: Heat shock transcription factor 2 promotes mitophagy of intestinal epithelial cells through PARL/PINK1/Parkin pathway in ulcerative colitis. Front Pharmacol. 13:8934262022. View Article : Google Scholar : PubMed/NCBI

173 

Zhu P, Ke ZR, Chen JX, Li SJ, Ma TL and Fan XL: Advances in mechanism and regulation of PANoptosis: Prospects in disease treatment. Front Immunol. 14:11200342023. View Article : Google Scholar : PubMed/NCBI

174 

Qi Z, Zhu L, Wang K and Wang N: PANoptosis: Emerging mechanisms and disease implications. Life Sci. 333:1221582023. View Article : Google Scholar : PubMed/NCBI

175 

Sun X, Yang Y, Meng X, Li J, Liu X and Liu H: PANoptosis: Mechanisms, biology, and role in disease. Immunol Rev. 321:246–262. 2024. View Article : Google Scholar

176 

You YP, Yan L, Ke HY, Li YP, Shi ZJ, Zhou ZY, Yang HY, Yuan T, Gan YQ, Lu N, et al: Baicalin inhibits PANoptosis by blocking mitochondrial Z-DNA formation and ZBP1-PANoptosome assembly in macrophages. Acta Pharmacol Sin. 46:430–447. 2025. View Article : Google Scholar

177 

Shi C, Cao P, Wang Y, Zhang Q, Zhang D, Wang Y, Wang L and Gong Z: PANoptosis: A cell death characterized by pyroptosis, apoptosis, and necroptosis. J Inflamm Res. 16:1523–1532. 2023. View Article : Google Scholar : PubMed/NCBI

178 

Malireddi RKS, Kesavardhana S, Karki R, Kancharana B, Burton AR and Kanneganti TD: RIPK1 distinctly regulates yersinia-induced inflammatory cell death, PANoptosis. Immunohorizons. 4:789–796. 2020. View Article : Google Scholar : PubMed/NCBI

179 

Newton K, Wickliffe KE, Dugger DL, Maltzman A, Roose-Girma M, Dohse M, Kőműves L, Webster JD and Dixit VM: Cleavage of RIPK1 by caspase-8 is crucial for limiting apoptosis and necroptosis. Nature. 574:428–431. 2019. View Article : Google Scholar : PubMed/NCBI

180 

Zheng Z, Deng W, Bai Y, Miao R, Mei S, Zhang Z, Pan Y, Wang Y, Min R, Deng F, et al: The lysosomal rag-ragulator complex licenses RIPK1 and caspase-8-mediated pyroptosis by yersinia. Science. 372:eabg02692021. View Article : Google Scholar

181 

Van Opdenbosch N, Van Gorp H, Verdonckt M, Saavedra PHV, de Vasconcelos NM, Gonçalves A, Vande Walle L, Demon D, Matusiak M, Van Hauwermeiren F, et al: Caspase-1 engagement and TLR-Induced c-FLIP expression suppress ASC/Caspase-8-Dependent apoptosis by inflammasome sensors NLRP1b and NLRC4. Cell Rep. 21:3427–3444. 2017. View Article : Google Scholar : PubMed/NCBI

182 

Gutierrez KD, Davis MA, Daniels BP, Olsen TM, Ralli-Jain P, Tait SW, Gale M Jr and Oberst A: MLKL activation triggers NLRP3-mediated processing and release of IL-1β independently of gasdermin-D. J Immunol. 198:2156–2164. 2017. View Article : Google Scholar : PubMed/NCBI

183 

Ye Z, Deng M, Yang Y, Song Y, Weng L, Qi W, Ding P, Huang Y, Yu C, Wang Y, et al: Epithelial mitochondrial fission-mediated PANoptosis is crucial for ulcerative colitis and its inhibition by saquinavir through Drp1. Pharmacol Res. 210:1075382024. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Wu B, Su S, Li Y and Guo L: Dysregulated programmed cell death of intestinal epithelial cells in ulcerative colitis: Molecular mechanisms and novel therapeutic interventions (Review). Int J Mol Med 56: 230, 2025.
APA
Wu, B., Su, S., Li, Y., & Guo, L. (2025). Dysregulated programmed cell death of intestinal epithelial cells in ulcerative colitis: Molecular mechanisms and novel therapeutic interventions (Review). International Journal of Molecular Medicine, 56, 230. https://doi.org/10.3892/ijmm.2025.5671
MLA
Wu, B., Su, S., Li, Y., Guo, L."Dysregulated programmed cell death of intestinal epithelial cells in ulcerative colitis: Molecular mechanisms and novel therapeutic interventions (Review)". International Journal of Molecular Medicine 56.6 (2025): 230.
Chicago
Wu, B., Su, S., Li, Y., Guo, L."Dysregulated programmed cell death of intestinal epithelial cells in ulcerative colitis: Molecular mechanisms and novel therapeutic interventions (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 230. https://doi.org/10.3892/ijmm.2025.5671
Copy and paste a formatted citation
x
Spandidos Publications style
Wu B, Su S, Li Y and Guo L: Dysregulated programmed cell death of intestinal epithelial cells in ulcerative colitis: Molecular mechanisms and novel therapeutic interventions (Review). Int J Mol Med 56: 230, 2025.
APA
Wu, B., Su, S., Li, Y., & Guo, L. (2025). Dysregulated programmed cell death of intestinal epithelial cells in ulcerative colitis: Molecular mechanisms and novel therapeutic interventions (Review). International Journal of Molecular Medicine, 56, 230. https://doi.org/10.3892/ijmm.2025.5671
MLA
Wu, B., Su, S., Li, Y., Guo, L."Dysregulated programmed cell death of intestinal epithelial cells in ulcerative colitis: Molecular mechanisms and novel therapeutic interventions (Review)". International Journal of Molecular Medicine 56.6 (2025): 230.
Chicago
Wu, B., Su, S., Li, Y., Guo, L."Dysregulated programmed cell death of intestinal epithelial cells in ulcerative colitis: Molecular mechanisms and novel therapeutic interventions (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 230. https://doi.org/10.3892/ijmm.2025.5671
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team