BPG is committed to discovery and dissemination of knowledge
Editorial Open Access
Copyright ©The Author(s) 2026. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jan 21, 2026; 32(3): 114229
Published online Jan 21, 2026. doi: 10.3748/wjg.v32.i3.114229
Beyond anti-inflammatory strategies: Epigenetic targets as emerging therapeutic frontiers in acute pancreatitis
Li-Ping Liang, Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, Guangzhou 510180, Guangdong Province, China
Le Zhang, Dan-Dan Jin, Shao-Heng Zhang, Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
Le Liu, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, Guangdong Province, China
ORCID number: Shao-Heng Zhang (0000-0002-8849-4724); Le Liu (0000-0002-9270-9951).
Co-first authors: Li-Ping Liang and Le Zhang.
Author contributions: Liang LP and Zhang L contributed equally as co-first authors; Liu L served as the corresponding author and provided overall project supervision; Liang LP and Zhang L drafted the manuscript; Jin DD, Zhang SH, and Liu L subsequent revisions incorporating input; all authors participated in manuscript refinement and approved the final version for submission.
Supported by Shenzhen Medical Research Fund, No. A2403044; National Natural Science Foundation of China, No. 82200612; and Guangdong Basic and Applied Basic Research Foundation, No. 2023A1515111183.
Conflict-of-interest statement: The authors declare that they have no conflict of interest.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Le Liu, MD, Principal Investigator, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, No. 1333 Xinhu Road, Shenzhen 518000, Guangdong Province, China. 1402744723@smu.edu.cn
Received: September 15, 2025
Revised: November 14, 2025
Accepted: December 4, 2025
Published online: January 21, 2026
Processing time: 123 Days and 20.1 Hours

Abstract

Acute pancreatitis (AP) remains a challenging clinical condition with limited therapeutic options and high mortality rates in severe cases. Traditional anti-inflammatory approaches have shown disappointing results in clinical trials, highlighting the urgent need for novel therapeutic strategies targeting the underlying pathophysiological mechanisms. The study by Jia et al presents compelling evidence for a previously unrecognized mechanism through which rutaecarpine, a bioactive alkaloid from traditional Chinese medicine, exerts protective effects against AP. This research demonstrates that rutaecarpine alleviates AP by targeting the epigenetic machinery, specifically through enhancer of EZH2-mediated suppression of FBXW11. The authors employed both in vitro cerulein-induced AR42J cell models and in vivo sodium taurocholate-induced rat models to establish the therapeutic efficacy of rutaecarpine and elucidate its molecular mechanisms. Their findings reveal that rutaecarpine upregulates EZH2 expression, leading to increased histone H3 methylation at the FBXW11 promoter region, thereby suppressing FBXW11 expression and consequently reducing inflammatory infiltration and oxidative stress. The significance of this work extends beyond demonstrating rutaecarpine’s protective effects. It identifies FBXW11 as a novel therapeutic target in AP and provides the first evidence that traditional Chinese medicine compounds can modulate epigenetic reprogramming in pancreatic inflammation. Recent studies have confirmed FBXW11’s role as an inflammatory biomarker in pancreatitis, supporting the clinical relevance of this pathway. The study’s comprehensive approach, combining molecular docking, cellular thermal shift assays, and co-immunoprecipitation studies, strengthens the mechanistic insights. These findings open new avenues for AP treatment by targeting epigenetic regulators rather than relying solely on conventional anti-inflammatory strategies, potentially leading to more effective therapeutic interventions for this devastating condition.

Key Words: Acute pancreatitis; Epigenetic regulation; Rutaecarpine; FBXW11; EZH2

Core Tip: Acute pancreatitis (AP) remains without effective mechanism-based therapy beyond supportive care. This editorial emphasizes epigenetic regulation as an emerging therapeutic frontier, highlighting how the traditional Chinese medicine alkaloid rutaecarpine protects against AP via enhancer of EZH2-mediated repression of FBXW11. By integrating evidence on FBXW11 as an inflammatory biomarker, the pleiotropic actions of rutaecarpine, and the clinical availability of EZH2 inhibitors, this article outlines an epigenetic, multi-target framework for future AP interventions.



INTRODUCTION

Acute pancreatitis (AP) is a common hospitalization-requiring gastrointestinal disease characterized by a local and systemic inflammatory response and usually manifested by severe upper abdominal pain[1]. Representing one of the most challenging conditions in gastroenterology, AP is associated with significant morbidity and mortality. Mortality for pancreatitis is approximately 1% overall[2,3]; however, among hospitalized patients with pancreatitis and organ failure or pancreatic necrosis, mortality may be as high as 30%-40%[4]. The pathogenesis of AP is complex, but inflammation and tissue necrosis are considered the main initiating pathological factors[5]. Despite decades of research into the inflammatory cascades underlying this condition, therapeutic interventions remain largely supportive, and specific targeted therapies have consistently failed to translate from promising preclinical studies to clinical success. Recent guidelines from the International Association of Pancreatology (2025) continue to emphasize the urgent need for mechanism-based therapeutic approaches[6]. The work by Jia et al[7] published in this issue offers a refreshing perspective by identifying a novel epigenetic mechanism through which rutaecarpine, a traditional Chinese medicine compound, exerts protective effects against AP.

NOVEL MECHANISTIC INSIGHTS: FBXW11 AS AN INFLAMMATORY BIOMARKER

The identification of FBXW11 as a key mediator in AP pathogenesis represents a significant advancement in understanding this complex disease. Recent breakthrough research by Tan et al[8] established FBXW11 as a novel inflammatory biomarker, directly linking its expression to immune infiltration and nuclear factor kappa-B (NF-κB) pathway activation in both pancreatitis and pancreatic cancer. Notably, FBXW11 expression exhibits a progressive pattern across the disease spectrum, from normal pancreatic tissue through AP to pancreatic ductal adenocarcinoma, thereby establishing its clinical relevance throughout disease progression. FBXW11 is an F-box protein that functions as a subunit of the SKP1-cullin-F-box ubiquitin E3 Ligase complex, serving as a fundamental regulator of cell-cycle progression and tumorigenesis[9]. While FBXW11 dysregulation has been implicated in Alzheimer’s disease[10], developmental disorders[11], and various cancers[12-14], its inflammatory role in pancreatic disease was previously unexplored. Jia et al[7] demonstrated that FBXW11 expression is significantly elevated in AP models, with overexpression exacerbating cellular damage and knockdown conferring protection against cerulein-induced injury. This observation aligns with emerging pan-cancer analyses revealing FBXW11’s prognostic and immunological significance across multiple tumor types[15]. Of particular significance, the discovery that rutaecarpine targets the histone methyltransferase enhancer of EZH2 to suppress FBXW11 expression through epigenetic modification represents a conceptual breakthrough in elucidating traditional medicine’s molecular mechanisms. The EZH2-H3 methylation-FBXW11 axis provides a unifying molecular framework linking epigenetic regulation to inflammatory control, thereby offering mechanistic insights into the sophisticated pharmacological properties of traditional therapeutic compounds.

EZH2: A CENTRAL HUB FOR INFLAMMATORY REGULATION

EZH2 is a member of the polycomb group gene family, a large class of epigenetic regulators that suppress transcription. Polycomb repressive complex 2 (PRC2), one of the two major polycomb complexes, primarily functions by altering chromatin architecture to enforce gene repression[16]. As the catalytic component of PRC2, EZH2 catalyzes the trimethylation of lysine 27 on histone 3 (H3K27me3), a modification that regulates gene expression[17]. Accumulating evidence demonstrates EZH2’s involvement across diverse pathological contexts. Li et al[18] demonstrated that EZH2 suppresses angiotensin-converting enzyme 2 expression through histone H3 methylation, with therapeutic implications for coronavirus disease. In atrial fibrosis models, EZH2 was shown to repress the transcription of CDKN2a (p16, p19) and Timp4 by establishing canonical H3K27me3 marks at their promoters[19]. Additionally, EZH2 promotes pancreatic tissue regeneration in cerulein-induced pancreatitis by silencing nuclear factor of activated T cells cytoplasmic 1[20]. EZH2’s multifaceted role in inflammatory disease regulation provides a compelling mechanistic foundation for the proposed pathway. Recent high-impact studies have elucidated EZH2’s critical regulatory function in pancreatic inflammatory conditions. Yuan et al[21] revealed that EZH2 competes with p53 to modulate inflammasome activation through long noncoding RNA (lncRNA) Neat1-mediated transcription, thereby directly regulating the inflammatory cascade. Notably, their findings showed that EZH2’s SANT2 domain maintains histone H3 lysine 27 acetylation at inflammatory gene promoters, promoting chromatin accessibility necessary for inflammatory responses. Complementing these findings, Chibaya et al[22] demonstrated that EZH2 inhibition remodels pancreatic cancer inflammation by suppressing pro-inflammatory senescence-associated secretory phenotype factors through H3K27me3-mediated epigenetic silencing. Collectively, this evidence substantiates the therapeutic potential of EZH2 as a target in pancreatic inflammatory conditions.

MECHANISTIC RIGOR AND ALIGNMENT WITH PRECISION MEDICINE IN AP

The methodological strengths of the study lie in its comprehensive and multilayered experimental design, which integrates in vitro cerulein-induced AR42J cellular models with in vivo sodium taurocholate-induced rat models to provide robust evidence for rutaecarpine’s therapeutic efficacy across diverse experimental settings[7]. This rigor is further reinforced by systematic molecular validation: Molecular docking followed by cellular thermal shift assays demonstrates a direct interaction between rutaecarpine and EZH2, while co-immunoprecipitation confirms the functional consequences of this binding. The strategic application of gain- and loss-of-function manipulations for both EZH2 and FBXW11 substantially strengthens causal inference, and the finding that FBXW11 overexpression abolishes rutaecarpine’s protective effects provides compelling evidence for pathway specificity, thereby underscoring the centrality of the EZH2-FBXW11 axis. Importantly, this mechanistic framework is highly congruent with current research directions in AP. Emerging studies have highlighted the relevance of PRC2-mediated epigenetic pathways in pancreatic inflammation. Zhao et al[23], for instance, identified the lncRNA FENDRR as a modulator of autophagy via interaction with the PRC2 complex, which includes EZH2, supporting the broader physiological significance of EZH2-dependent regulatory circuits. In parallel, increasing emphasis on biomarker-guided and mechanism-based therapies, including cytokine-driven stratification strategies, reflects a shift toward precision medicine in AP care. This trend is further reinforced by the 2024 American College of Gastroenterology guidelines[24], which highlight individualized, mechanism-targeted approaches, providing an ideal translational landscape for therapeutic strategies centered on the EZH2-FBXW11 axis.

RUTAECARPINE’S MULTI-TARGET ANTI-INFLAMMATORY PROFILE

Rutaecarpine, a major quinazolinocarboline alkaloid isolated from the traditional Chinese herbal medicine Wu-Chu-Yu (Evodia rutaecarpa), has been utilized in pharmaceutical preparations for hypertension management in China for centuries[25]. Accumulating evidence demonstrates that rutaecarpine exhibits potent anti-inflammatory and immunomodulatory effects through multiple molecular targets[26-28]. The mitogen-activated protein kinase (MAPK) and NF-κB signaling cascades constitute the primary pathways regulating inflammatory mediator transcription and biosynthesis in AP[29]. Within the MAPK family, p38 and extracellular regulated protein kinases (ERK) 1/2 play pivotal roles in cytokine expression during AP pathogenesis. Notably, p38 MAPK signaling modulates NF-κB activation, which represents a critical node in the inflammatory cascade. Activation of p38 MAPK triggers downstream protein kinase phosphorylation[30], ultimately stimulating NF-κB and amplifying the inflammatory response. By targeting these pathways, rutaecarpine significantly reduces nitric oxide production and suppresses the expression of inducible nitric oxide synthase, cyclooxygenase-2, and interleukin-1β in lipopolysaccharide- and lipoteichoic acid-stimulated macrophages[31,32]. These anti-inflammatory properties extend beyond pancreatitis; rutaecarpine attenuates inflammation and cartilage degradation in osteoarthritis by inhibiting the phosphatidylinositol 3-kinase/protein kinase B/NF-κB and MAPK pathways, potentially through integrin αVβ3 activation[33]. Furthermore, rutaecarpine exerts antioxidative effects by activating the nuclear factor erythroid-2-related factor 2 (NRF2) pathway across diverse pathological conditions, including acute liver injury[34], colitis[35], traumatic brain injury[36], and migraine[37]. In the specific context of AP, studies demonstrated that rutaecarpine mitigates cerulein-induced inflammation in both rodent models and AR42J cells by upregulating calcitonin gene-related peptide, subsequently suppressing MAPK and NF-κB signaling[38,39].

The EZH2-FBXW11 axis identified by Jia et al[7] likely functions synergistically with these established mechanisms rather than operating in isolation. Critically, FBXW11’s documented role in NF-κB activation through inhibitor of NF-κB alpha (IκBα) ubiquitination suggests substantial crosstalk between epigenetic regulatory circuits and canonical inflammatory signaling pathways. Rutaecarpine’s suppression of FBXW11 via EZH2-mediated epigenetic silencing may therefore synergize with its direct inhibitory effects on NF-κB and MAPK pathways, culminating in enhanced anti-inflammatory efficacy. Recent mechanistic investigations have further elucidated rutaecarpine’s sophisticated anti-inflammatory profile and its potential interactions with the EZH2-FBXW11 network. Jayakumar et al[32] demonstrated that rutaecarpine inhibits NF-κB and ERK/p38 MAPK pathways by blocking IκBα and NF-κB p65 phosphorylation, thereby preventing their nuclear translocation. This mechanism directly correlates with FBXW11’s established role in NF-κB activation as identified by Tan et al[8]. Intriguingly, novel epigenetic connections have emerged from the work of Xu et al[36], who discovered that rutaecarpine promotes PGK1 ubiquitination to activate the NRF2 antioxidant pathway. This finding reveals direct engagement with protein degradation machinery analogous to FBXW11’s E3 ubiquitin ligase function, indicating that rutaecarpine may modulate FBXW11-mediated protein targeting through multiple convergent mechanisms.

While this editorial emphasizes the novel EZH2-FBXW11 epigenetic mechanism, it is essential to recognize that rutaecarpine’s overall therapeutic benefit likely derives from the integrated effects of multiple pathways operating in concert. The relative contribution of each molecular pathway may vary depending on AP etiology, disease severity, and timing of therapeutic intervention. This polypharmacological profile may actually confer therapeutic advantages for treating complex inflammatory conditions like AP, where single-target therapies have historically demonstrated limited clinical success. The multi-target nature of rutaecarpine potentially addresses the multifactorial pathogenesis of AP more comprehensively than conventional mono-targeted approaches, offering a rationale for its promising preclinical efficacy.

CLINICAL IMPLICATIONS AND THERAPEUTIC POTENTIAL

The therapeutic implications of these findings extend well beyond the immediate application of rutaecarpine in AP treatment. The identification of FBXW11 as a druggable target establishes novel therapeutic opportunities, including the development of selective FBXW11 inhibitors and modulators of its upstream regulators. Furthermore, the elucidation of EZH2-mediated epigenetic mechanisms suggests that other epigenetic modulators may similarly confer therapeutic benefit in AP, thereby broadening the therapeutic armamentarium for this condition. Recent clinical developments strongly support this therapeutic strategy. Multiple EZH2 inhibitors (tazemetostat, valemetostat) and clinical-stage compounds offer well-established pharmacological pathways for targeting this mechanism. Ongoing clinical trials investigating EZH2 inhibitors in solid tumors, including pancreatic cancer (No. NCT04705818), have established the translational infrastructure necessary for potential expansion to AP applications. More importantly, this work provides a robust scientific rationale for clinical investigation of rutaecarpine in AP patients. The demonstrated efficacy in reducing inflammatory markers, oxidative stress, and tissue damage in preclinical models, combined with the well-documented safety profile of rutaecarpine in traditional medicine, substantiates the clinical trial rationale. Recent real-world analyses demonstrate the efficacy of integrating traditional Chinese medicine with conventional medical approaches in managing mild-to-moderate AP[40].

BROADER IMPACT ON TRADITIONAL MEDICINE RESEARCH

This study exemplifies how modern molecular techniques can systematically elucidate the mechanisms underlying traditional medicine practices, potentially validating centuries-old therapeutic approaches through rigorous contemporary scientific frameworks. The demonstration that rutaecarpine can modulate epigenetic machinery highlights the inherent biological sophistication of natural compounds and reveals that traditional medicines may operate through mechanisms substantially more complex than previously recognized. Rather than crude, non-specific effects, this work demonstrates that natural alkaloids can engage sophisticated regulatory circuits comparable to rationally designed therapeutics. The epigenetic modulation mechanism identified here likely extends beyond AP to other inflammatory conditions where FBXW11 or similar epigenetic pathways play crucial roles. This interpretation is supported by recent evidence demonstrating that traditional Chinese medicine compounds modulate epigenetic regulation in inflammatory bowel disease and related gastrointestinal disorders[41,42], suggesting convergence of mechanistic principles across disease contexts. Recent bibliometric analyses of traditional Chinese medicine research in AP (2007-2023) reveal a pronounced shift toward mechanism-based investigations and molecular pathway elucidation[43], positioning this work within a broader disciplinary trend toward evidence-based traditional medicine research. This paradigmatic evolution reflects the field’s maturation from empirical observation toward systematic molecular validation, underscoring the scientific legitimacy of investigating traditional therapeutics through contemporary frameworks.

LIMITATIONS AND FUTURE DIRECTIONS

While the Jia et al’s study[7] provides compelling mechanistic insights, several important limitations merit careful consideration. The reliance on animal models, sodium taurocholate-induced rats and cerulein-stimulated AR42J cells, while necessary for proof-of-concept investigations, may not fully recapitulate the heterogeneity of human AP pathophysiology. In clinical practice, AP presents with diverse etiologies (gallstone-induced, alcohol-related, hypertriglyceridemia-associated) and severity classifications (mild, moderately severe, severe), each potentially influencing therapeutic response. The current models primarily represent bile acid-induced and hyperstimulation-induced AP; validation in human pancreatic tissue from patients with varying etiologies would be essential to establish the clinical relevance and generalizability of the EZH2-FBXW11 axis.

The pharmacokinetic properties of rutaecarpine present substantial clinical translation challenges: Low oral bioavailability, rapid first-pass metabolism yielding hydroxylated derivatives, and short plasma half-life collectively constrain systemic drug exposure. However, recent advances offer promising solutions. Structural modification strategies, including N-substituted rutaecarpine analogs, have demonstrated improved metabolic stability and enhanced anti-inflammatory efficacy[44,45]. Additionally, nano-delivery platforms, lipid-based nanoparticles or polymeric nanocarriers, could substantially enhance intestinal absorption while protecting against first-pass metabolism. Pancreas-targeted delivery systems using tissue-specific ligands represent another promising approach to maximize local drug accumulation while minimizing systemic toxicity[46].

Several additional mechanistic investigations warrant attention. The relative contribution of the EZH2-FBXW11 pathway likely varies across different AP etiologies; comparative studies across multiple disease models would clarify whether this pathway represents a universal mechanistic principle or exhibits context-specific importance. The dose-response relationship remains incompletely characterized, while the original study employed 300 μg/kg based on prior optimization, optimal dosing for different AP severities, the therapeutic window, and potential dose-dependent toxicity require systematic evaluation. Furthermore, the study’s focus on a single time point (24 hours post-induction) may not capture full temporal dynamics; time-course studies across multiple intervals (6, 12, 24, 48, 72 hours) would define the optimal intervention window and distinguish primary therapeutic effects from secondary compensatory responses.

Multi-center preclinical validation studies are essential for establishing experimental reproducibility and identifying potential confounding factors related to laboratory protocols, animal strains, or reagent variability. Human tissue validation through endoscopic ultrasound-guided fine needle aspiration or surgical specimens should accompany well-designed clinical trials stratified by AP etiology and severity. Finally, investigating whether other traditional medicine alkaloids, such as berberine, with its documented histone deacetylase modulation, operate through similar epigenetic mechanisms could reveal additional therapeutic opportunities and accelerate translation of traditional medicine wisdom into precision therapeutics[47].

RESEARCH GAPS AND NOVEL CONTRIBUTIONS

While extensive literature supports individual components of this mechanistic framework, no current investigations directly explore the rutaecarpine-EZH2-FBXW11 signaling axis in AP, rendering this work’s contribution genuinely novel and intellectually significant. The proposed mechanism is robustly grounded in converging lines of evidence. FBXW11’s well-established role as an inflammatory biomarker in pancreatitis demonstrates direct correlation with NF-κB pathway activation, while EZH2’s proven capacity to regulate inflammatory responses across diverse disease contexts, coupled with the availability of Food and Drug Administration (FDA)-approved therapeutic inhibitors, facilitates clinical translation. Rutaecarpine’s documented multi-target profile encompasses protein ubiquitination, transcriptional regulation, and NRF2 antioxidant activation, extending beyond the proposed epigenetic axis. Furthermore, established precedents exist for epigenetic modulation by structurally related alkaloids such as berberine, substantiating the plausibility of alkaloid-mediated histone methylation. Collectively, these converging elements position the proposed mechanism not as speculative hypothesis, but as mechanistically coherent and experimentally testable within the contemporary framework of epigenetic pharmacology.

CONCLUSION

The work by Jia et al[7] represents a significant advancement in AP research, providing novel mechanistic insights that bridge traditional medicine and modern epigenetics. The identification of the EZH2-FBXW11 axis as a therapeutic target offers concrete hope for developing effective treatments. Recent confirmation of FBXW11 as an inflammatory biomarker and EZH2’s established role in inflammatory regulation provide strong support for this mechanism. This study exemplifies how rigorous molecular investigation can validate traditional therapeutic approaches within modern pharmacological science. The integration of traditional wisdom with cutting-edge epigenetic research may herald a paradigm shift in drug discovery, where ancient remedies inform identification of molecular targets through sophisticated mechanistic understanding. The convergence of FDA-approved EZH2 inhibitors, validated FBXW11 biomarkers, and rutaecarpine’s demonstrated safety profile creates an optimal environment for clinical translation. Beyond AP, this mechanistic framework applies to numerous conditions where traditional medicines have shown efficacy but lacked mechanistic understanding, potentially revolutionizing integrative medicine in the era of precision therapeutics.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A, Grade A

Novelty: Grade A, Grade A

Creativity or Innovation: Grade A, Grade A

Scientific Significance: Grade A, Grade C

P-Reviewer: Zhao JN, MD, Post Doctoral Researcher, United States S-Editor: Fan M L-Editor: A P-Editor: Lei YY

References
1.  Boxhoorn L, Voermans RP, Bouwense SA, Bruno MJ, Verdonk RC, Boermeester MA, van Santvoort HC, Besselink MG. Acute pancreatitis. Lancet. 2020;396:726-734.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 276]  [Cited by in RCA: 656]  [Article Influence: 109.3]  [Reference Citation Analysis (0)]
2.  Gapp J, Hall AG, Walters RW, Jahann D, Kassim T, Reddymasu S. Trends and Outcomes of Hospitalizations Related to Acute Pancreatitis: Epidemiology From 2001 to 2014 in the United States. Pancreas. 2019;48:548-554.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 42]  [Cited by in RCA: 87]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
3.  Wadhwa V, Patwardhan S, Garg SK, Jobanputra Y, Lopez R, Sanaka MR. Health Care Utilization and Costs Associated With Acute Pancreatitis. Pancreas. 2017;46:410-415.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 27]  [Cited by in RCA: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
4.  Petrov MS, Shanbhag S, Chakraborty M, Phillips AR, Windsor JA. Organ failure and infection of pancreatic necrosis as determinants of mortality in patients with acute pancreatitis. Gastroenterology. 2010;139:813-820.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 517]  [Cited by in RCA: 584]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
5.  Escobar J, Pereda J, Arduini A, Sandoval J, Sabater L, Aparisi L, López-Rodas G, Sastre J. Cross-talk between oxidative stress and pro-inflammatory cytokines in acute pancreatitis: a key role for protein phosphatases. Curr Pharm Des. 2009;15:3027-3042.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 68]  [Cited by in RCA: 79]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
6.  IAP/APA/EPC/IPC/JPS Working Group; IAP/APA/EPC/IPC/JPS Working Group. International Association of Pancreatology Revised Guidelines on Acute Pancreatitis 2025: Supported and Endorsed by the American Pancreatic Association, European Pancreatic Club, Indian Pancreas Club, and Japan Pancreas Society. Pancreatology. 2025;25:770-814.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 17]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
7.  Jia Y, Shi YX, Gu H, Liu Y, Peng J, Yan L. Rutaecarpine targets F-box and WD repeat domain containing 11 to inhibit inflammatory infiltration and alleviate acute pancreatitis. World J Gastroenterol. 2025;31:109486.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
8.  Tan P, Cai S, Huang Z, Li M, Liu S, Chen J, Fu W, Zhao L. E3 ubiquitin ligase FBXW11 as a novel inflammatory biomarker is associated with immune infiltration and NF-κB pathway activation in pancreatitis and pancreatic cancer. Cell Signal. 2024;116:111033.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
9.  Shi L, Du D, Peng Y, Liu J, Long J. The functional analysis of Cullin 7 E3 ubiquitin ligases in cancer. Oncogenesis. 2020;9:98.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
10.  Sun J, Qin X, Zhang X, Wang Q, Zhang W, Wang M. FBXW11 deletion alleviates Alzheimer's disease by reducing neuroinflammation and amyloid-β plaque formation via repression of ASK1 signaling. Biochem Biophys Res Commun. 2021;548:104-111.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
11.  Holt RJ, Young RM, Crespo B, Ceroni F, Curry CJ, Bellacchio E, Bax DA, Ciolfi A, Simon M, Fagerberg CR, van Binsbergen E, De Luca A, Memo L, Dobyns WB, Mohammed AA, Clokie SJH, Zazo Seco C, Jiang YH, Sørensen KP, Andersen H, Sullivan J, Powis Z, Chassevent A, Smith-Hicks C, Petrovski S, Antoniadi T, Shashi V, Gelb BD, Wilson SW, Gerrelli D, Tartaglia M, Chassaing N, Calvas P, Ragge NK. De Novo Missense Variants in FBXW11 Cause Diverse Developmental Phenotypes Including Brain, Eye, and Digit Anomalies. Am J Hum Genet. 2019;105:640-657.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 21]  [Cited by in RCA: 37]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
12.  Chen C, Zhou H, Zhang X, Liu Z, Ma X. Association of FBXW11 levels with tumor development and prognosis in chondrosarcoma. Cancer Biomark. 2022;35:429-437.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
13.  Yao J, Yang J, Yang Z, Wang XP, Yang T, Ji B, Zhang ZY. FBXW11 contributes to stem-cell-like features and liver metastasis through regulating HIC1-mediated SIRT1 transcription in colorectal cancer. Cell Death Dis. 2021;12:930.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
14.  Zhang Q, Yin X, Zhang Y. MicroRNA-221 Promotes Cell Proliferation and Inhibits Apoptosis in Osteosarcoma Cells by Directly Targeting FBXW11 and Regulating Wnt Signaling. Arch Med Res. 2021;52:191-199.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 19]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
15.  Huang T, OuYang X, Li J, Shi B, Shan Z, Shi Z, Yang Z. Pan-cancer analysis of FBXW family with potential implications in prognosis and immune infiltration. Front Immunol. 2022;13:1084339.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
16.  Pasini D, Di Croce L. Emerging roles for Polycomb proteins in cancer. Curr Opin Genet Dev. 2016;36:50-58.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 70]  [Cited by in RCA: 98]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
17.  Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res. 2008;647:21-29.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 600]  [Cited by in RCA: 683]  [Article Influence: 37.9]  [Reference Citation Analysis (0)]
18.  Li Y, Li H, Zhou L. EZH2-mediated H3K27me3 inhibits ACE2 expression. Biochem Biophys Res Commun. 2020;526:947-952.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 45]  [Cited by in RCA: 50]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
19.  Li Y, Fang G, Cao W, Yuan J, Song S, Peng H, Wang Y, Wang Q. Ezh2 Inhibits Replicative Senescence of Atrial Fibroblasts Through Promotion of H3K27me3 in the Promoter Regions of CDKN2a and Timp4 Genes. J Inflamm Res. 2022;15:4693-4708.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 16]  [Reference Citation Analysis (0)]
20.  Chen NM, Neesse A, Dyck ML, Steuber B, Koenig AO, Lubeseder-Martellato C, Winter T, Forster T, Bohnenberger H, Kitz J, Reuter-Jessen K, Griesmann H, Gaedcke J, Grade M, Zhang JS, Tsai WC, Siveke J, Schildhaus HU, Ströbel P, Johnsen SA, Ellenrieder V, Hessmann E. Context-Dependent Epigenetic Regulation of Nuclear Factor of Activated T Cells 1 in Pancreatic Plasticity. Gastroenterology. 2017;152:1507-1520.e15.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 24]  [Cited by in RCA: 38]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
21.  Yuan J, Zhu Q, Zhang X, Wen Z, Zhang G, Li N, Pei Y, Wang Y, Pei S, Xu J, Jia P, Peng C, Lu W, Qin J, Cao Q, Xiao Y. Ezh2 competes with p53 to license lncRNA Neat1 transcription for inflammasome activation. Cell Death Differ. 2022;29:2009-2023.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
22.  Chibaya L, Murphy KC, DeMarco KD, Gopalan S, Liu H, Parikh CN, Lopez-Diaz Y, Faulkner M, Li J, Morris JP 4th, Ho YJ, Chana SK, Simon J, Luan W, Kulick A, de Stanchina E, Simin K, Zhu LJ, Fazzio TG, Lowe SW, Ruscetti M. EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance. Nat Cancer. 2023;4:872-892.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 22]  [Cited by in RCA: 82]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
23.  Zhao SP, Yu C, Yang MS, Liu ZL, Yang BC, Xiao XF. Long Non-coding RNA FENDRR Modulates Autophagy Through Epigenetic Suppression of ATG7 via Binding PRC2 in Acute Pancreatitis. Inflammation. 2021;44:999-1013.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
24.  Tenner S, Vege SS, Sheth SG, Sauer B, Yang A, Conwell DL, Yadlapati RH, Gardner TB. American College of Gastroenterology Guidelines: Management of Acute Pancreatitis. Am J Gastroenterol. 2024;119:419-437.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 64]  [Cited by in RCA: 169]  [Article Influence: 84.5]  [Reference Citation Analysis (0)]
25.  Hu CP, Xiao L, Deng HW, Li YJ. The depressor and vasodilator effects of rutaecarpine are mediated by calcitonin gene-related peptide. Planta Med. 2003;69:125-129.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 42]  [Cited by in RCA: 44]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
26.  Chen J, Hu ZY, Ma Y, Jiang S, Yin JY, Wang YK, Wu YG, Liu XQ. Rutaecarpine alleviates inflammation and fibrosis by targeting CK2α in diabetic nephropathy. Biomed Pharmacother. 2024;180:117499.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
27.  Li Y, Zhang G, Chen M, Tong M, Zhao M, Tang F, Xiao R, Wen H. Rutaecarpine inhibited imiquimod-induced psoriasis-like dermatitis via inhibiting the NF-κB and TLR7 pathways in mice. Biomed Pharmacother. 2019;109:1876-1883.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 33]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
28.  Luo DN, Li FJ, Zou YY. [Therapeutic effects of rutaecarpine on dextran sodium sulfate-induced experimental colitis in mice]. Zhonghua Yi Xue Za Zhi. 2018;98:533-538.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
29.  Liu HS, Pan CE, Liu QG, Yang W, Liu XM. Effect of NF-kappaB and p38 MAPK in activated monocytes/macrophages on pro-inflammatory cytokines of rats with acute pancreatitis. World J Gastroenterol. 2003;9:2513-2518.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 61]  [Cited by in RCA: 62]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
30.  Cao MH, Xu J, Cai HD, Lv ZW, Feng YJ, Li K, Chen CQ, Li YY. p38 MAPK inhibition alleviates experimental acute pancreatitis in mice. Hepatobiliary Pancreat Dis Int. 2015;14:101-106.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 22]  [Cited by in RCA: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
31.  Jayakumar T, Lin KC, Chang CC, Hsia CW, Manubolu M, Huang WC, Sheu JR, Hsia CH. Targeting MAPK/NF-κB Pathways in Anti-Inflammatory Potential of Rutaecarpine: Impact on Src/FAK-Mediated Macrophage Migration. Int J Mol Sci. 2021;23:92.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
32.  Jayakumar T, Yang CM, Yen TL, Hsu CY, Sheu JR, Hsia CW, Manubolu M, Huang WC, Hsieh CY, Hsia CH. Anti-Inflammatory Mechanism of An Alkaloid Rutaecarpine in LTA-Stimulated RAW 264.7 Cells: Pivotal Role on NF-κB and ERK/p38 Signaling Molecules. Int J Mol Sci. 2022;23:5889.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
33.  Wan J, Li M, Yuan X, Yu X, Chen A, Shao M, Kang H, Cheng P. Rutaecarpine ameliorates osteoarthritis by inhibiting PI3K/AKT/NF‑κB and MAPK signalling transduction through integrin αVβ3. Int J Mol Med. 2023;52:97.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
34.  Choi JH, Jin SW, Lee GH, Han EH, Hwang YP, Jeong HG. Rutaecarpine Protects against Acetaminophen-Induced Acute Liver Injury in Mice by Activating Antioxidant Enzymes. Antioxidants (Basel). 2021;10:86.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
35.  Zhang Y, Yan T, Sun D, Xie C, Wang T, Liu X, Wang J, Wang Q, Luo Y, Wang P, Yagai T, Krausz KW, Yang X, Gonzalez FJ. Rutaecarpine inhibits KEAP1-NRF2 interaction to activate NRF2 and ameliorate dextran sulfate sodium-induced colitis. Free Radic Biol Med. 2020;148:33-41.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 95]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
36.  Xu M, Li L, Liu H, Lu W, Ling X, Gong M. Rutaecarpine Attenuates Oxidative Stress-Induced Traumatic Brain Injury and Reduces Secondary Injury via the PGK1/KEAP1/NRF2 Signaling Pathway. Front Pharmacol. 2022;13:807125.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
37.  Xu M, Shi Z, He Z, Ling X, Wang W, Liu H, Gong M. Rutaecarpine alleviates migraine in nitroglycerin-induced mice by regulating PTEN/PGK1 signaling pathway to activate NRF2 antioxidant system. Biomed Pharmacother. 2023;166:115300.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
38.  Huang H, Wang M, Guo Z, Wu D, Wang H, Jia Y, Liu H, Ding J, Peng J. Rutaecarpine alleviates acute pancreatitis in mice and AR42J cells by suppressing the MAPK and NF-κB signaling pathways via calcitonin gene-related peptide. Phytother Res. 2021;35:6472-6485.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 18]  [Reference Citation Analysis (0)]
39.  Yan L, Li QF, Rong YT, Chen YH, Huang ZH, Wang ZZ, Peng J. The protective effects of rutaecarpine on acute pancreatitis. Oncol Lett. 2018;15:3121-3126.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
40.  Jia S, Chen Q, Liu X, Li Y, Wang L, Li X, Hu S. Efficacy of integrated traditional Chinese and western medicine in managing mild-moderate acute pancreatitis: a real-world clinical perspective analysis. Front Med (Lausanne). 2024;11:1429546.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
41.  Zhang C, Lio C, Li N, Huang C, Yao X, Luo J. Advances in Traditional Chinese Medicine for Modulating DNA Methylation in the Treatment of Inflammatory Diseases. Int J Mol Sci. 2025;26:6331.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
42.  Wu C. Epigenetics and traditional Chinese medicine: A noteworthy research area. Chin Herb Med. 2025;17:201-202.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
43.  Lan WP, Guo W, Zhou X, Li Z. Research trends on traditional Chinese medicine and acute pancreatitis: A bibliometric analysis from 2007 to mid-2023. Heliyon. 2024;10:e25659.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
44.  Li D, Huang Z, Xu X, Li Y. Promising derivatives of rutaecarpine with diverse pharmacological activities. Front Chem. 2023;11:1199799.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
45.  Son JK, Chang HW, Jahng Y. Progress in Studies on Rutaecarpine. II.--Synthesis and Structure-Biological Activity Relationships. Molecules. 2015;20:10800-10821.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 32]  [Cited by in RCA: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
46.  Demirtürk N, Bilensoy E. Nanocarriers targeting the diseases of the pancreas. Eur J Pharm Biopharm. 2022;170:10-23.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
47.  Wang Z, Liu Y, Xue Y, Hu H, Ye J, Li X, Lu Z, Meng F, Liang S. Berberine acts as a putative epigenetic modulator by affecting the histone code. Toxicol In Vitro. 2016;36:10-17.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 26]  [Cited by in RCA: 38]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]