BPG is committed to discovery and dissemination of knowledge
Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Dec 28, 2025; 31(48): 113840
Published online Dec 28, 2025. doi: 10.3748/wjg.v31.i48.113840
Mitochondrial dysfunction as a bridge to pathology in acute pancreatitis: From molecular insights to novel therapeutic strategies
Chuan-Chao Xia, Yue Xu, Guo-Qiang Xu, Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
Zhen-Huan Wang, Department of Radiology, The Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
ORCID number: Chuan-Chao Xia (0000-0001-7486-773X); Yue Xu (0000-0002-4992-6651); Zhen-Huan Wang (0009-0007-0747-5534); Guo-Qiang Xu (0000-0003-1337-9120).
Co-first authors: Chuan-Chao Xia and Yue Xu.
Co-corresponding authors: Zhen-Huan Wang and Guo-Qiang Xu.
Author contributions: Xia CC and Xu Y wrote the manuscript as co-first authors; Wang ZH contributed to searching the literature; Xia CC and Wang ZH revised the manuscript; Xu GQ reviewed and supervised the project; Xu GQ and Wang ZH made equal contributions as co-corresponding authors. All authors have read and approved the final manuscript.
Supported by National Natural Science Foundation of China, No. 8217030254.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
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: Guo-Qiang Xu, MD, Professor, Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou 310003, Zhejiang Province, China. 1193065@zju.edu.cn
Received: September 5, 2025
Revised: October 28, 2025
Accepted: November 14, 2025
Published online: December 28, 2025
Processing time: 113 Days and 18.5 Hours

Abstract

Acute pancreatitis (AP) is a life-threatening inflammatory condition triggered by the premature activation of trypsin. The limited understanding of its underlying pathophysiology remains a key obstacle to the development of targeted therapies. Mounting evidence now underscores mitochondrial dysfunction as a critical pathogenic driver in AP. Cellular mitochondrial dysfunction often precedes both cytokine release and trypsin activation, potentially serving as a primary initiator in the development and advancement of AP. Mitochondrial dysfunction is associated with calcium overload, inflammatory reactions, mitochondrial permeability transition pore opening, mitophagy damage, and other potential pathogenesis of pancreatic cell injury. Elucidating the impact of mitochondrial injury in AP may facilitate the development of innovative treatment approaches. This review provides a comprehensive and systematic analysis of the pivotal role of mitochondria in regulating pancreatic homeostasis, while evaluating emerging therapeutic strategies aimed at mitigating mitochondrial dysfunction. By integrating cutting-edge research findings, this work highlights the translational potential of these advancements in redefining diagnostic frameworks and optimizing therapeutic approaches for the management of AP.

Key Words: Acute pancreatitis; Mitochondria; Mitochondrial dysfunction; Molecular mechanism; Treatment

Core Tip: Mitochondrial dysfunction is increasingly recognized as a pivotal initiating factor in acute pancreatitis (AP), preceding trypsin activation and inflammatory amplification. Key mechanisms include calcium overload, oxidative stress, mitochondrial permeability transition pore opening, and impaired mitophagy. This review synthesizes current evidence on mitochondrial dysregulation in AP and highlights emerging therapeutic strategies targeting mitochondrial pathways, offering new avenues for transitioning from supportive care to mechanism-driven precision medicine in AP management.



INTRODUCTION

Acute pancreatitis (AP), a potentially fatal inflammatory disorder of the pancreas, arises from premature intra-acinar trypsin activation triggered by multifactorial etiologies including biliary obstruction, alcohol metabolites, hypertriglyceridemia, and genetic variants in cationic trypsinogen 1, or serine peptidase inhibitor Kazal type 1[1]. The pathophysiological cascade of AP is initiated by dysregulated intra-pancreatic trypsinogen activation, which precipitates acinar cell autodigestion and subsequent tissue injury. This process elicits a robust inflammatory cascade, culminating in hallmark pathological manifestations such as pancreatic edema, parenchymal hemorrhage, and focal or diffuse necrosis. Clinically, the disease spectrum is categorized into three distinct severity classifications: Mild AP, moderately severe AP, and severe AP (SAP), based on systemic complications and organ failure duration. Epidemiological studies reveal that approximately 80% of AP cases are self-limiting (mild AP), whereas 20% progress to SAP, a life-threatening condition associated with mortality rates exceeding 30% due to multi-organ dysfunction syndrome[2]. Elevated serum concentrations of pancreatic enzymes, particularly amylase and lipase, constitute biochemical hallmarks of AP, though their diagnostic specificity remains limited. The pathophysiological cascade involves premature activation of trypsinogen, a zymogen synthesized exclusively by pancreatic acinar cells. Under physiological conditions, this protease precursor undergoes site-specific activation through enteropeptidase-mediated cleavage in the duodenal lumen[3]. In AP pathogenesis, however, the aberrant intra-acinar conversion of trypsinogen to active trypsin represents a critical initiating event. This mechanistic paradigm has been rigorously validated across multiple experimental models, including caerulein-induced AP in mice and rats, demonstrating conserved activation pathways across species[4].

Despite substantial progress in elucidating AP pathophysiology, key mechanistic gaps remain in our understanding of the molecular pathways that orchestrate inflammatory activation and cellular injury, significantly impeding the development of targeted therapies. Although pancreatic autodigestion is widely recognized as a central pathogenic driver, recent studies highlight the critical involvement of gut-derived inflammation, microcirculatory disturbances, and dysregulated immunomodulatory networks in disease exacerbation and systemic complications[5,6]. Recent studies have increasingly highlighted the critical role of pancreatic acinar cell organelle disruption in AP pathogenesis[7]. The organelles of pancreatic cells, including mitochondria are destroyed and trigger the formation and development of AP[8]. Mitochondria are important organelles present in eukaryotes, known as energy factories of cells, which can produce energy required by the body[9]. At the same time, mitochondria are also involved in various signaling pathways and inflammation in cells. Increasing studies have been conducted on mitochondrial dysfunction and AP in recent years[10,11].

Mitochondrial dysfunction represents a pivotal pathogenic axis in AP, driving both disease onset and progression through multiple interconnected mechanisms. Under physiological conditions, mitochondria-derived reactive oxygen species (ROS), primarily generated via electron transport chain (ETC) activity during oxidative phosphorylation, serve as critical regulators of redox signaling and cellular homeostasis[12].

Pathological ROS overproduction triggers inflammatory cascades via activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and NLR family pyrin domain-containing protein 3 (NLRP3) inflammasome, driving transcriptional upregulation of pro-inflammatory cytokines, chemokines, and adhesion molecules that potentiate pancreatic inflammation[13]. Activation of the NF-κB pathway induces the expression of multiple inflammatory mediators, including cytokines, chemokines, immune receptors, and focal adhesion molecules, thereby triggering a sterile inflammatory response and propagating a cascade of pancreatic injury[14,15]. Acute cellular injury can cause important mitochondrial activities to be lost or deteriorate, which is linked to a reduction in adenosine triphosphate (ATP) synthesis and bioenergetic impairment[16]. Therefore, ATP has a dual function in the physiology and pathophysiology of the pancreas since it is essential for cell activity and, like many other systems, serves as a crucial messenger between cells[17]. Emerging research indicates that estrogen-related receptor γ, a key regulator of pancreatic acinar cell metabolism, can modulate cellular biology and energy homeostasis by disrupting mitochondrial function, subsequently inducing oxidative stress, and autophagic dysfunction[18]. In addition, Damage-associated molecular patterns (DAMPs) drive aseptic inflammation, which has been demonstrated in numerous studies to be a crucial process mediating subsequent pancreatic injury, downstream organ damage, and disease resolution[19]. DAMPs cannot activate pattern recognition receptors to cause inflammation in normal circumstances. Changes in membrane permeability during inflammation enable these chemicals to trigger pattern recognition receptors and propel the inflammatory process[20]. Several studies have demonstrated that mitochondrial damage can lead to energy generation disorders, Ca2+ homeostasis disorders, impaired autophagy, affecting cell function and even leading to cell death[3,10,11,21].

This review systematically elucidates the pivotal role of mitochondria in maintaining pancreatic homeostasis, while delineating their pathological contributions to AP via dysregulated calcium flux, impaired autophagy, oxidative stress-mediated damage, mitochondrial DNA (mtDNA) dysfunction and compromised membrane integrity. Furthermore, we evaluate emerging evidence on mitochondrial-centric signaling cascades implicated in AP progression and highlight novel therapeutic interventions targeting mitochondrial dysfunction, offering translational potential for refining diagnostic paradigms and treatment strategies in AP management.

A comprehensive systematic literature search was conducted across PubMed and Web of Science databases following PRISMA guidelines. The search strategy employed Boolean logic to combine Medical Subject Headings terms “acute pancreatitis” or “mild acute pancreatitis” or “severe acute pancreatitis” or “pancreatic acinar cells” and “mitochondria”. Our search encompassed articles published from January 1, 2000 to September 30, 2025, and Only publications in English language were included.

MITOCHONDRIAL STRUCTURE AND FUCTION IN PANCREATIC ACINAR CELLS

Mitochondria, double-membrane-bound organelles ubiquitous in eukaryotic cells, serve as the primary site for energy production through intracellular aerobic respiration, earning their designation as the “cellular powerhouses” (Figure 1)[22]. Beyond their role in energy metabolism, these organelles play crucial roles in cellular stress adaptation and programmed cell death regulation[23]. The mitochondrial inner membrane (IMM) exhibits a characteristic invaginated structure, forming cristae that house the respiratory chain complexes and essential enzymatic systems[24]. In contrast, the mitochondrial outer membrane (OMM) functions as both a selective barrier and molecular exchange interface. This membrane system serves a dual protective function: (1) Acting as a selective barrier that limits passive small-molecule diffusion; and (2) Maintaining cellular integrity by sequestering cytotoxic mitochondrial metabolites, including ROS and apoptotic signaling molecules[25].

Figure 1
Figure 1 Mitochondrial structure and function in pancreatic acinar cells. Cyp D: Cyclophilin D; ANT: Adenine nucleotide translocation; mtDNA: Mitochondrial deoxyribonucleic acid; SERCA: Sarco/endoplasmic reticulum Ca2+-ATPases; PMCA: Plasma membrane Ca2+-ATPases.

Pancreatic acinar cells exhibit a characteristic polarized architecture, compartmentalized into three distinct regions: The apical, basal, and mitochondrial (buffer) domains[26]. Physiologically, these cells orchestrate a compartmentalized mitochondrial architecture comprising three topographically segregated pools strategically positioned to regulate secretion-ready vesicles, membrane trafficking nodes, and nuclear-cytoplasmic signaling[27,28]. Mitochondria exhibit a polarized distribution, localized at the critical interface between the apical granule-rich domain and the basolateral compartment, where they execute indispensable physiological functions. This unique spatial organization facilitates essential cellular processes, including the establishment of a Ca2+ buffering barrier and the efficient synthesis of ATP[29].

Under physiological conditions, Ca2+ is released from the endoplasmic reticulum (ER), resulting in transient elevation of cytoplasmic Ca2+ levels[30]. During each cytosolic Ca2+ wave, calcium-induced calcium release mechanisms facilitate mitochondrial Ca2+ uptake. Subsequently, the Na+/Ca2+ exchanger gradually releases Ca2+, thereby modulating intracellular Ca2+ concentrations ([Ca2+]i). Importantly, mitochondria maintain physical connections with specialized ER subdomains, which serve as conduits for Ca2+ transfer to mitochondria, establishing dynamic Ca2+ exchange between these organelles. Upon reaching peak Ca2+ levels during ER depletion, store-operated calcium channels become activated, initiating Ca2+ replenishment[31]. This elevated Ca2+ level stimulates mitochondrial ATP production through activation of oxidative phosphorylation. The generated ATP subsequently energizes sarco/ER Ca2+-ATPases and plasma membrane Ca2+-ATPases, enabling Ca2+ sequestration into the ER lumen or extrusion across the plasma membrane. This regulatory mechanism prevents the accumulation of potentially cytotoxic Ca2+ concentrations within the cytoplasm[1].

Mitochondrial permeability transition pore (MPTP) is a group of protein complexes located between IMM and OMM. Most scholars believe that it consists of voltage-dependent anion channels in the OMM and adenine nucleotide translocation proteins and cyclophilin D (CypD) in the IMM[32]. The opening of MPTP underlies many forms of AP, and various factors, such as mitochondrial Ca2+ overload and overproduction of ROS, could lead to MPTP opening, which would activate pathological proenzymes within the acinar cells, and mediate impaired ATP production and necrosis[33,34]. Mitochondria regulate mitochondrial number and energy metabolism to maintain cellular homeostasis by producing ROS and causing cell death through autophagy or mitochondrial damage. As an MPTP-regulated degradation process, mitophagy modulates mitochondrial dynamics and suppresses ROS generation[30].

MOLECULAR MECHANISMS UNDERLYING MITOCHONDRIAL DYSFUNCTION IN AP

Mitochondria serve dual essential roles as both the powerhouse of cellular energy metabolism and key regulators of fundamental cellular processes, including calcium homeostasis. In pancreatic acinar cells, mitochondria demonstrate heightened susceptibility to inflammatory insults, with mitochondrial impairment being recognized as a pivotal etiological factor in the pathogenesis of pancreatitis. The underlying mechanisms involve pathological calcium overload, defective mitophagy, oxidative stress accumulation, loss of mitochondrial membrane potential, and mtDNA leakage. These interconnected perturbations synergistically exacerbate disease progression by disrupting cellular metabolic equilibrium and amplifying pro-inflammatory pathways (Figure 2).

Figure 2
Figure 2 The molecular mechanisms underlying mitochondrial dysfunction in acute pancreatitis. SOCE: Store-operated calcium entry; Cyp D: Cyclophilin D; ROS: Reactive oxygen species; MPTP: Mitochondrial permeability transition pore; IP3Rs: Inositol triphosphate receptors; RyRs: Ryanodine receptors; STIM1: Stromal interaction molecule 1; SERCA: Sarco/endoplasmic reticulum Ca2+-ATPases; OXPHOS: Oxidative phosphorylation; LC3: Microtubule-associated protein light chain 3; LAMP-2: Lysosome-associated membrane protein-2; PINK1: PTEN-induced putative kinase 1; FUNDC1: FUN14 domain containing 1; mtROS: Mitochondrial reactive oxygen species; mtDNA: Mitochondrial DNA; cGAS: Cyclic GMP-AMP synthase; STING: Stimulator of interferon genes; IRF7: Interferon regulatory factor 7; IRF3: Interferon regulatory factor 3.
Calcium overload signals in AP

Ca2+ signaling research has primarily centered on pancreatic acinar cells, which play a pivotal role in secreting stored digestive enzymes essential for intestinal food digestion[35]. The secretion of these digestive enzymes is regulated through G-protein coupled receptors located on the basolateral membrane, which acetylcholine and cholecystokinin (CCK) act as key mediators. This process is modulated by an increase in intracellular free calcium concentration ([Ca2+]i), a response triggered by secretagogues that activate these receptors, which is crucial in both physiological and pathological conditions, particularly pancreatitis[17]. Pathological calcium signaling is predominantly characterized by a sustained, whole-cell, peak-plateau-type elevation in [Ca2+]i, often referred to as calcium overload. Scientific investigations dating back to 1995 have implicated calcium overload in pancreatic acinar cells as a critical factor in the initiation and progression of AP[36]. Among cellular organelles, mitochondria emerge as key regulators of Ca2+ homeostasis. While physiological Ca2+ signals induced by stimulation are transient and predominantly localized to the granule-containing apical pole, persistent global increases in cytosolic Ca2+ concentrations can be cytotoxic. Such Ca2+ overload triggers inappropriate intracellular trypsin activation, cellular vacuolization, and necrosis, leading to exacerbated cell damage and frequently fatal outcomes in cases of human AP[37,38]. The mitochondria act as a buffer for Ca2+ in the cell due to their quick calcium absorption. Mitochondria maintain calcium homeostasis in acinar cells through Ca2+ storage and coordination with ER and extracellular components[39]. In the granular region of pancreatic acinar cells, it has been observed that mitochondrial Ca2+ uptake begins as soon as the cytosolic Ca2+ concentration in the granular area increases following stimulation[40]. An aberrant Ca2+ concentration in the mitochondria can disrupt energy metabolism, resulting in excessive ROS buildup, mitochondrial damage, and either autophagy or death. Early and crucial signs of almost all types of pancreatitis include a persistent increase in the Ca2+ concentration and mitochondrial permeability[40].

Intracellular Ca2+ signaling in pancreatic acinar cells is precisely orchestrated through the coordinated action of multiple regulatory components, including Ca2+ release channels, endocytosis channels, efflux proteins, and mitochondria-mediated ATP synthesis. Among these, inositol triphosphate receptors and ryanodine receptors function as principal Ca2+ release channels, exhibiting polarized distribution patterns in both murine and human pancreatic acinar cells[41,42]. The activation of phospholipase C triggers the hydrolysis of phosphatidylinositol bisphosphate, generating inositol triphosphate that subsequently binds to inositol triphosphate receptors. This interaction induces Ca2+ release from ER stores into the cytoplasm, thereby initiating diverse Ca2+ dependent signaling cascades[43]. A critical regulator of sustained intracellular Ca2+ elevation is stromal interaction molecule 1, whose activation, oligomerization, and membrane translocation are essential for maintaining Ca2+ homeostasis[44]. Pathological conditions, particularly those induced by ethanol and its metabolites, demonstrate that aberrant Ca2+ influx mediated by efflux proteins contributes to trypsinogen activation and necrotic pathway induction in acinar cells[45]. Mitochondrial Ca2+ handling plays a dual role in cellular physiology, not only modulating Ca2+ signaling but also synchronizing energy production with secretory demands. The activation of secretion by acetylcholine or CCK-elicited Ca2+ signals necessitates a rapid upregulation of ATP synthesis. Glucose metabolism serves as the primary energy source, with hexokinases catalyzing the conversion to glucose-6-phosphate, initiating glycolysis and ultimately generating pyruvate to fuel mitochondrial ATP synthesis. During physiological stimulation of pancreatic acinar cells with acetylcholine or CCK, mitochondria maintain local Ca2+ oscillations in the apical region through the mitochondrial calcium uniporter, which facilitates Ca2+ transport across the inner mitochondrial membrane into the mitochondrial matrix[39].

Autophagy and mitophagy in AP

Autophagy represents a fundamental cellular process that facilitates the transport and degradation of surplus or dysfunctional cytoplasmic components and organelles within lysosomes, maintaining cellular homeostasis and organelle renewal through the regulation of autophagy-related genes (ATGs)[46]. Emerging evidence implicates defective autophagy mechanisms in the pathogenesis of AP, particularly through their contribution to acinar cell vacuolization and inappropriate zymogen activation[47]. During AP progression, pancreatic acinar cells exhibit distinct autophagy dysregulation, manifesting as enhanced autophagosome biogenesis coupled with impaired lysosomal degradation capacity. This perturbation results in inflammatory cell infiltration and exacerbates acinar cell necrosis and apoptosis[11]. The lysosome-associated membrane protein-2 (LAMP-2), a crucial mediator of autophagosome-lysosome fusion events, is abundantly expressed in pancreatic tissue[48]. Experimental evidence from rat models of acute necrotizing pancreatitis demonstrates that LAMP-2 deficiency leads to intracellular accumulation of undegraded substrates within abnormally enlarged vacuoles, indicative of autophagy impairment[49]. Genetic ablation of LAMP-2 exacerbates AP severity by disrupting autophagosome-lysosome complex formation, resulting in autophagosome accumulation, impaired zymogen granule degradation, and aberrant trypsinogen activation[50]. Recent investigations have highlighted the role of sequestosome 1 (SQSTM1/p62), a multifunctional protein involved in substrate recognition during macroautophagy. Notably, extracellular SQSTM1 derived from monocytes and macrophages has been implicated in AP pathogenesis by promoting autophagy-dependent ferroptotic cell death during bacterial infections[51]. Quantitative assessment of pancreatic autophagy markers reveals that elevated levels of ubiquitinated proteins, microtubule-associated protein light chain 3 (LC3)-II, and SQSTM1/p62 serve as specific indicators of impaired autophagic flux in AP[52]. Therapeutic strategies targeting the modulation of LC3 and SQSTM1/p62 expression demonstrate promising potential in restoring autophagic function and ameliorating pancreatic tissue damage in AP models[53].

Mitophagy, a highly selective form of autophagy, serves as a critical quality control mechanism that orchestrates the targeted degradation of impaired mitochondria through lysosomal pathways. This evolutionarily conserved process is dynamically regulated in response to diverse cellular stressors, including oxidative damage, senescence-associated signals, and inflammatory cascades, thereby maintaining mitochondrial homeostasis and cellular integrity[46]. This essential cellular mechanism serves as a critical stress response mechanism, maintaining mitochondrial network integrity and functionality. The timely removal of compromised mitochondria through various mitophagy pathways is fundamental for cellular survival and function[54]. In the context of AP, the activation of mitophagy becomes crucial in mitigating cellular damage by preventing excessive inflammatory cell infiltration and ROS accumulation, thereby reducing cellular necrosis and apoptosis[11] (Figure 2). Aberrant mitophagy flux disrupts mitochondrial quality control, leading to pathological accumulation of depolarized mitochondria with impaired oxidative phosphorylation capacity. This dysfunction fundamentally alters the cellular death paradigm by skewing the necrosis-to-apoptosis equilibrium, a molecular switch that directly governs pancreatic acinar cell fate determination and ultimately dictates the clinical trajectory of pancreatitis progression[7]. The cellular machinery employs multiple molecular pathways to execute mitophagy, with three primary mechanisms being particularly well-characterized: PTEN-induced putative kinase 1 (PINK1)/Parkin RBR E3 ubiquitin-protein ligase (Parkin) axis, the Bnip3 L/Nix pathway, and the FUN14 domain containing 1 (FUNDC1)-mediated mechanism[55]. The PINK1/Parkin pathway initiates when mitochondrial membrane potential collapse leads to PINK1 accumulation on the outer mitochondrial membrane, subsequently recruiting Parkin, an E3 ubiquitin ligase. Parkin then ubiquitinates mitochondrial surface proteins, marking them for autophagosomal degradation[55]. The FUNDC1-mediated pathway, particularly responsive to hypoxic conditions, represents another crucial mitophagy mechanism. FUNDC1, an outer mitochondrial membrane protein, interacts with LC3 to facilitate the removal of damaged mitochondria. Hypoxia-induced FUNDC1 dephosphorylation enhances its binding affinity with LC3, promoting mitophagy activation and cellular protection during pathological conditions such as AP[56]. These coordinated molecular pathways collectively maintain mitochondrial quality control and cellular homeostasis under stress conditions[57].

As a fundamental mitochondrial quality control mechanism, mitophagy selectively eliminates dysfunctional, impaired, or redundant mitochondria to maintain cellular homeostasis. This evolutionarily conserved process represents a critical biological safeguard for preserving mitochondrial integrity and functional stability. Mounting evidence indicates that precise regulation of autophagosome biogenesis can effectively inhibit pancreatic zymogen activation, mitigate autophagy-associated cellular injury, and attenuate the inflammatory cascade in AP[58]. In subsequent sections, we will systematically elucidate the regulatory mechanisms of autophagy-associated pathways and their therapeutic implications in AP management. Future investigations should prioritize the comprehensive characterization of autophagy dynamics in AP pathogenesis, with particular emphasis on developing targeted strategies to restore normal autophagic flux in pancreatic acinar cells.

Oxidative stress in AP

Oxidative stress arises from disrupted equilibrium between ROS production and cellular antioxidant capacity[59]. Mitochondria constitute the predominant intracellular source of ROS, which at physiological levels act as crucial secondary messengers regulating redox-sensitive signaling cascades. Under pathological inflammatory states, however, uncontrolled ROS production exceeds endogenous antioxidant defenses, precipitating mitochondrial dysfunction through oxidative to ETC components, ultimately triggering programmed cell death pathways[12] (Figure 2). Substantial clinical and experimental evidence demonstrates that oxidative stress manifests in the initial phase of AP, with pancreatic acinar cells and tissue releasing copious oxygen free radicals that exacerbate disease progression[60].

The role of ROS in AP extends beyond cellular damage, as it significantly modulates inflammatory responses. The persistent release of inflammatory mediators in AP subjects mitochondria to sustained stress, compromising respiratory chain function and contributing to pathological ROS accumulation, a critical determinant of disease progression[61]. Experimental studies utilizing the oxidant menadione have established a direct correlation between escalating ROS levels and increased apoptotic cell death, highlighting the detrimental impact of oxidative stress on pancreatic acinar cell viability[62].

Mitochondrial dysfunction significantly impairs ATP synthesis, a critical process for maintaining essential cellular functions. In pancreatitis pathogenesis, excessive mitochondrial ROS (mitoROS) plays a pivotal role by activating multiple inflammatory signaling cascades and promoting cytokine production[12]. Experimental evidence reveals that mitoROS serves as a signaling molecule, inducing pro-inflammatory cytokine release through both inflammasome-dependent and independent mechanisms[63]. Specifically, mitoROS reduction has been shown to inhibit lipopolysaccharide-induced MAP kinase pathway activation and subsequent tumor necrosis factor-α and interleukin-6 production via an inflammasome-independent process[64]. Notably, mtDNA release, which amplifies inflammatory responses, requires coordinated activation of the NLRP inflammasome and mitoROS generation[65]. Inhibition of autophagy or mitophagy pathways leads to the accumulation of dysfunctional, ROS-generating mitochondria and cytoplasmic mtDNA release, subsequently triggering caspase-1 activation and NLRP3 inflammasome assembly[66]. Pancreatitis-induced mitochondrial dysfunction elevates intracellular oxidative stress, increasing mitoROS production that directly damages cellular components including proteins, nucleic acids, and membrane structures, ultimately culminating in necrotic and apoptotic cell death[67].

MPTP dysfunction in acute pancreatitis

The MPTP is a non-selective channel facilitating the passage of molecules under 1.5 kD across the inner mitochondrial membrane, and its opening would cause the loss of mitochondrial membrane potential, which is necessary for the synthesis of ATP[34]. Structural analyses reveal that MPTP comprises three core components: CypD in the inner membrane, adenine nucleotide translocator proteins, and voltage-dependent anion channels in the outer membrane. While transient MPTP opening facilitates physiological signal transduction, including Ca2+ and ROS exchange between the mitochondrial matrix and cytoplasm, persistent activation induces mitochondrial swelling, ultimately triggering apoptotic cell death[68]. In AP, MPTP activation mediates key pathological processes, including intra-acinar zymogen activation, ATP depletion, inflammatory cascades, and necrotic cell death, representing molecular, cellular, and systemic manifestations of the disease[33]. CypD, a member of the peptidyl-prolyl cis-trans isomerase family, catalyzes proline imidic peptide bond isomerization and facilitates protein folding[69]. Under conditions of elevated Ca2+ concentrations, CypD translocates to the inner mitochondrial membrane, where it interacts with adenine nucleotide translocator to induce MPTP opening while inhibiting ATP/adenosine diphosphate binding[33]. Genetic ablation of CypD in murine models demonstrates its critical role in AP pathogenesis, as CypD deficiency attenuates mitochondrial damage, preserves pancreatic ATP production, and mitigates key pathological features of arginine-induced AP, including inflammatory responses, necrotic/apoptotic cell death, trypsinogen activation, and acinar cell vacuolization[11]. These findings imply that blocking CypD as a novel strategy for AP management.

MtDNA integrity and dysfunction in AP

MtDNA, a critical DAMP, is typically sequestered within the mitochondrial matrix, shielded from innate immune recognition[70]. However, cellular stress conditions disrupt mitochondrial integrity, leading to mtDNA leakage into the cytosol, where it activates multiple signaling cascades and initiates immune responses[71]. MtDNA exhibits dual roles, functioning both as a pathophysiological mediator and a potential biomarker for disease severity assessment in pancreatitis pathogenesis. Clinical researches have established circulating mtDNA levels as a sensitive early indicator of AP severity, demonstrating strong correlations with clinical manifestations[72]. The association between mtDNA dysfunction and pancreatitis is further evidenced by recurrent AP cases in patients with Kearns-Sayre syndrome, a mitochondrial disorder linked to mtDNA abnormalities[73]. Experimental studies using premature aging models reveal that mtDNA mutations, particularly those affecting mitochondrial complex I, induce pancreatic α-cell proliferation, suggesting both detrimental and adaptive roles in pancreatic tissue homeostasis[45].

Mechanistically, mtDNA drives pancreatitis progression through multifaceted pathways. It coordinates mitochondrial dynamics by inducing fission and facilitating vacuole membrane protein 1-mediated selective mitophagy, thereby exerting cytoprotective effects during AP[74]. Furthermore, mtDNA regulates cell fate decisions by modulating the apoptosis-necrosis balance, a critical determinant of disease severity[7]. which is supported by the research that mtDNA plays a crucial part in the pathophysiology of pancreatitis by causing apoptosis in acinar cells and contributing to mitochondrial malfunction[75]. In pancreatic β-cells, mtDNA-mediated mitochondrial fusion events trigger lipid peroxidation and mitochondrial dysfunction, while cytosolic mtDNA fragments activate the stimulator of interferon genes (STING) pathway, culminating in apoptotic cell death[76]. Single-cell sequencing and experimental studies have elucidated that mtDNA leakage perpetuates pancreatic acinar cell damage through the cyclic GMP-AMP synthase-STING-NF-κB signaling axis[77]. In SAP cases, mtDNA-driven cyclic GMP-AMP synthase-STING signaling exacerbates lung injury by promoting macrophage pyroptosis through interferon regulatory factor 7/interferon regulatory factor 3 activation[78].

The impact of specific mtDNA mutations on pancreatitis susceptibility has been demonstrated through genetic studies. Notably, the A3243G mutation in the mtDNA has been associated with increased diabetes prevalence and recurrent pancreatitis in specific familial cohorts[79]. These findings collectively underscore the critical role of mtDNA integrity in pancreatic pathophysiology and highlight the complex interplay between mitochondrial function and pancreatic disease progression.

ADVANCES IN THERAPEUTIC STRATEGIES TARGETING MITOCHONDRIAL DYSFUNCTION IN AP

Despite the current lack of specific therapeutics for AP, emerging research has elucidated the relationship between mitochondrial dysfunction and AP pathogenesis, revealing promising therapeutic targets (Table 1). Current strategies primarily target core pathological mechanisms underlying mitochondrial dysfunction, including calcium dysregulation, mitochondrial membrane disruption, oxidative stress, and impaired mitophagy flux.

Table 1 Studies about therapeutic strategies targeting mitochondrial dysfunction in acute pancreatitis.
Ref.
Journal
Evidence level
Treatment
Model
Target
Gerasimenko et al[45], 2013Proc Natl Acad Sci U S AIn vitroGSK-7975APancreatic acinar cellsCa2+ channels
Wen et al[81], 2015GastroenterologyIn vivoGSK-7975A, CM_128TLCS-AP in mice, CER-AP in mice, FAEE-AP in miceORAI1 channel
Muili et al[82], 2013J Biol ChemIn vivoFK506, cyclosporine A, calcineurin inhibitory peptideTLCS-AP in miceCa2+ channels
Farooq et al[83], 2022Am J Physiol Gastrointest Liver PhysiolIn vivoPhosphate supplementationCER-AP in miceATP synthesis
Farooq et al[84], 2021GastroenterologyIn vivoPhosphate supplementationAlcohol-induced AP in miceATP synthesis
Javed et al[85], 2018PancreasIn vivoTRO40303FAEE-AP in miceCypD channels
Tóth et al[88], 2019J PhysiolIn vivoNIM811FAEE-AP in miceMPTP
Márta et al[89], 2017BMJ OpenClinical trial1ATP supplementationPatients with APEnergy nutritional support
Hu et al[90], 2023Adv Sci (Weinh)In vitroHypoxia-treated functional mitochondriaPancreatic acinar cellsMPTP
Zhang et al[91], 2024PhytomedicineIn vivoRhizoma Alismatis decoctionCER-AP in mice, HFD-AP in miceMitophagy
Yuan et al[92], 2021Oxid Med Cell LongevIn vivoVitamin B123% NaT induced AP in miceCBS/SIRT1
Lee et al[57], 2021Int J Mol SciIn vivoLycopenePancreatic acinar cellsROS
Piplani et al[94], 2019Biochim Biophys Acta Mol Basis DisIn vivoSimvastatinCER-AP in miceMitophagy
Shen et al[95], 2018Br J PharmacolIn vivoDihydrodiosgeninTauro-induced AP in rats, CER-AP in mice, FAEE-AP in micePI3Kγ/Akt
Li et al[96], 2022Free Radic ResIn vivoDeoxyarbutin3.5% NaT induced AP in miceHtrA2/PGC-1α
Li et al[97], 2024Int J Mol SciIn vivoIsorhamnetin3.5% NaT induced AP in miceHtrA2/GSK-3β/PGC-1α
Hoque et al[99], 2011GastroenterologyIn vivoTLR9 antagonistCER-AP in miceDAMP

A pathogenic hallmark shared across diverse AP etiologies is the dysregulation of calcium homeostasis, manifesting as sustained intracellular Ca2+ overload and mitochondrial permeability alterations. Recent studies indicate that pharmacologically inhibiting store-operated calcium entry or calcium release-activated calcium channels attenuates ER Ca2+ depletion-induced cytosolic Ca2+ accumulation, thereby effectively preventing trypsinogen activation and acinar cell necrosis in alcohol-related AP models[45]. However, the potential adverse effects associated with Ca2+ influx inhibition necessitate the exploration of alternative strategies, such as modulation of acinar cell Ca2+ channels, which may offer safer therapeutic approaches[80].

Accumulating mechanistic evidence indicates that mitochondrial stabilization via calcium homeostasis modulation represents a promising therapeutic strategy to ameliorate AP severity. Notably, calcium release-activated calcium modulator calcium channel inhibitors, which block pathological Ca2+ influx into acinar cells, demonstrate consistent protective effects in both preclinical models and primary human acinar cells, attenuating pancreatic injury and systemic complications[45,81]. Additionally, BAPTA, a membrane-permeable calcium chelator, has been shown to prevent bile acid-induced mitochondrial damage and CCK-stimulated vacuolization in pancreatic acinar cells[82]. Recent investigations have further revealed that phosphate administration improves mitochondrial activity and attenuates pathological Ca2+ elevation, providing protection against experimental pancreatitis[83,84].

The opening of the MPTP has been implicated in the pathogenesis of various forms of AP. Cyclosporine A, currently the only clinically approved experimental inhibitor of MPTP formation, exerts its therapeutic effect through specific inhibition of CypD[85]. As a member of the peptidyl prolyl cis-trans isomerase family, CypD plays a pivotal role in MPTP regulation, and its inhibition through genetic, molecular, or pharmacological approaches effectively prevents MPTP opening, thereby mitigating mitochondrial dysfunction and cellular necrosis in AP. Experimental evidence demonstrates that autophagy blockade through genetic deletion of ATG5, ATG7, or NF-κB kinase subunit inhibitors induces ER stress and promotes the accumulation of dysfunctional mitochondria, leading to ATP depletion. Conversely, CypD gene ablation has been shown to restore mitochondrial function and alleviate ER stress[86,87]. In the search for novel therapeutic agents, TRO40303 has emerged as a promising MPTP inhibitor. This compound interacts with the exo-mitochondrial translocation protein, delaying MPTP opening through a CypD-independent mechanism, preserving mitochondrial membrane potential, and reducing acinar cell necrosis, while demonstrating systemic protective effects in AP models[85]. Recent advancements in drug development have identified N-methyl-4-isoleucine cyclosporin, a cyclosporin A derivative, as a potent MPTP inhibitor. This compound not only rescues calcium overload-induced mitochondrial dysfunction in both pancreatic acinar and ductal cells but also dose-dependently attenuates pancreatic injury across diverse experimental AP models without observable toxicity at relevant preclinical doses[88].

Various pathological stimuli, including bile acids, ethanol, fatty acids, and their metabolites (fatty acid ethyl esters), induce mitochondrial dysfunction and ATP depletion in both pancreatic acinar and ductal cells, ultimately leading to cellular death and pancreatic necrosis. Experimental evidence demonstrates that ATP restoration in these cell types not only prevent cell death but also partially rescues cellular function. This therapeutic potential is currently being evaluated in a multicenter clinical trial investigating the efficacy of ATP supplementation combined with caloric support in AP management[89].

Hypoxic preconditioning of mitochondria has emerged as a promising therapeutic strategy, as preconditioned mitochondria can be internalized by pancreatic acinar cells, enhancing their resistance to toxic stimuli through metabolic stabilization and sustained energy supply. This protective mechanism involves hypoxia-mediated suppression of mitochondrial superoxide accumulation and upregulation of membrane potential, resulting in metabolic reprogramming[90]. In the realm of traditional medicine, Rhizoma Alismatis decoction (RAD), a two-component formulation comprising Rhizoma Alismatis and Atractylodes macrocephala rhizoma, has demonstrated significant therapeutic potential. RAD, recognized for its anti-inflammatory and lipid-modulating effects, significantly improves mitochondrial membrane potential and oxidative phosphorylation function. Mechanistic studies reveal that RAD modulates key autophagy markers, reducing beclin-1 and LC3-II expression while upregulating LAMP-1 and the Parkin-PINK pathway, thereby ameliorating hyperlipidemic AP-induced mitochondrial dysfunction through suppression of oxidative damage and enhancement of mitophagic flux[91].

Vitamins have antioxidant activity and have also been shown to negatively correlate with AP[92]. Vitamin B12 has been shown to enhance mitochondrial function repair through mitophagy activation, subsequently attenuating pancreatic edema, inflammatory responses, and necrotic damage[93]. Lycopene, a potent antioxidant, exhibits protective effects in AP by reducing ROS generation and modulating inflammatory pathways. Experimental studies demonstrate that lycopene effectively prevents palmitoleic acid-induced mitochondrial membrane potential collapse and ATP depletion in pancreatic acinar cells, suggesting that dietary lycopene supplementation may mitigate alcoholic pancreatitis progression[57].

The PINK1/Parkin2-mediated mitophagy pathway plays a crucial regulatory role in pancreatitis pathogenesis. Activation of this pathway has been shown to alleviate pancreatic inflammation through modulation of NLRP3 inflammasome activity[93]. Simvastatin, a well-known statin, demonstrates therapeutic potential in AP by reducing pancreatic edema, cellular apoptosis, and trypsin activation through Parkin-dependent mechanisms. Furthermore, simvastatin maintains autophagic flux and promotes autophagosome-lysosome fusion, thereby preventing mtDNA release[94].

Recent pharmacological research has identified dihydrodiosgenin, a diosgenin derivative, as a promising mitochondrial protectant. This compound prevents mitochondrial depolarization, ATP depletion, ROS overproduction, and excessive inflammatory responses, offering protection against pancreatitis-associated acute lung injury[95]. Deoxyarbutin has also shown therapeutic efficacy by restoring mitochondrial function and enhancing autophagic processes in pancreatic injury models[96]. Isorhamnetin, a naturally occurring flavonoid with potent antioxidant and anti-inflammatory properties, exerts protective effects in SAP through lysine (K)-specific demethylase 5B/HtrA serine peptidase 2 signaling pathway regulation. This mechanism maintains mitochondrial membrane potential, sustains ATP production, and prevents oxidative damage and mtDNA release, thereby reducing mitochondrial dysfunction in SAP[97].

Emerging evidence highlights the immunostimulatory properties of mtDNA, which contains abundant unmethylated cytosine-phosphate-guanine dinucleotide sequences capable of activating Toll-like receptor 9 (TLR9) and triggering subsequent inflammatory cascades[98,99]. Experimental studies demonstrate that both TLR9 gene silencing and pharmacological inhibition significantly attenuate pancreatic edema and inflammatory cell infiltration: TLR9 antagonists have shown efficacy in reducing pancreatic necrosis and pulmonary inflammation in taurolithocholic acid 3-sulfate-induced AP models, positioning TLR9 inhibition as a promising therapeutic strategy for AP management[94]. Mounting evidence underscores the translational value of mitochondrial biomarkers in AP. Macrophage-derived MAP kinase 14 modulates potentially regulate inflammation in SAP[100], while cytochrome C release from the inner mitochondrial membrane, may serve as an independent prognostic indicator for SAP[101].

Moreover, the role of mitochondrial metabolic regulation is also receiving increasing attention. Emerging evidence implicates the sirtuin 5-succinyl-CoA ligase ADP-forming subunit beta axis in mitochondrial dysfunction during AP, where cytochrome C1 succinylation modulates ETC complex III activity. Experimental manipulation of protein succinylation through sirtuin 5 overexpression or succinyl-CoA ligase ADP-forming subunit beta desuccinylation reduced macrophage activation and inflammation, highlighting its therapeutic potential[102]. Research demonstrates that pharmacological activation of histidine triad nucleotide-binding protein 2 alleviates inflammatory responses and pancreatic necrosis in AP by restoring mitochondrial oxidative phosphorylation via AMP-activated protein kinase/peroxisome proliferator-activated receptor gamma coactivator-1α signaling axis[103]. Furthermore, mechanistic investigations revealed that chromatin remodeler mortality factor 4-like protein 1 exacerbates apoptosis in lipidemic AP by suppressing FUNDC1-dependent mitophagy, thereby disrupting mitochondrial quality control and amplifying caspase-3 activation[104].

While targeted therapies modulating Ca2+, ROS, or MPTP show therapeutic promise, their translational potential requires careful risk-benefit analysis. As fundamental regulators of cellular homeostasis, global inhibition or activation may induce off-target effects in non-diseased tissues. Compared to systemic administration, tissue-specific delivery systems could enhance specificity[105-108]. Notably, interspecies differences in drug metabolism may render compounds safe in animal models yet clinically hazardous. More evidence is needed.

LIMITATIONS

Our study has several limitations that warrant consideration. First, despite employing systematic search strategies, the included literature may not be exhaustive. This is particularly relevant for studies investigating mitochondrial involvement in AP pathogenesis but lacking explicit terminology in titles or keywords. Additionally, potential publication bias may have led to underrepresentation of negative or null results in the available literature. Second, substantial heterogeneity exists among the included AP research models, which may compromise cross-study comparability and generalizability of findings. Third, the translational implications of our findings are constrained by the predominance of preclinical evidence, as the majority of included studies utilized animal models or in vitro systems, with limited representation of clinical trials or human cohort studies. Finally, while this work specifically examines mitochondrial dysfunction and related therapeutic targets, we acknowledge that AP involves complex interplay among multiple pathways, including calcium dysregulation, ER stress, and etc., which were not the primary focus of this investigation but may functionally interact with mitochondrial mechanisms.

Future research recommendations

Despite compelling preclinical evidence linking mitochondrial dysfunction to AP, including impaired oxidative phosphorylation, ROS overproduction, and MPTP opening, no clinically validated mitochondrial biomarkers currently exist for AP severity stratification. The development of such translational biomarkers may revolutionize AP management by enabling early risk prediction and personalized therapeutic interventions. Furthermore, AP pathogenesis involves a complex interplay of multiple pathological mechanisms, notably calcium overload-induced acinar cell injury, ER stress-mediated apoptosis, and mitochondrial bioenergetic failure. This multi-hit pathophysiology suggests that single-target therapies may be insufficient, whereas multi-target strategies could offer synergistic benefits. Such precision medicine approaches may improve AP outcomes by addressing its multidimensional pathophysiology, ultimately reducing disease progression and organ failure. Inflammatory diseases may share the common feature of mitochondrial dysfunction. Specifically, hepatitis or myocarditis shared mechanisms including MPTP dysregulation, mitochondrial redox imbalance, DAMP release, and etc. This mechanistic overlap suggests therapeutic cross-reactivity: Mitochondrial-protective agents effective in hepatitis or myocarditis may demonstrate efficacy in AP. It is important to emphasize that current evidence remains confined to preclinical studies, with a translational gap persisting between bench-scale discoveries and clinical applications.

CONCLUSION

Mitochondrial dysfunction emerges as a pivotal factor in the pathophysiological cascade of AP, a severe inflammatory disorder of the pancreatic tissue. The onset and progression of AP are fundamentally associated with impaired mitochondrial performance, characterized by diminished ATP synthesis, elevated oxidative stress markers, and disruption of mitochondrial membrane integrity. These alterations lead to excessive generation of mitoROS, thereby amplifying cellular injury and inflammatory responses. The compromised mitochondrial function initiates programmed cell death pathways through the cytoplasmic release of pro-apoptotic factors, particularly cytochrome C. Additionally, AP-induced oxidative modifications and genetic alterations in mtDNA further deteriorate mitochondrial bioenergetics and cellular metabolic homeostasis. These molecular insights underscore the critical role of mitochondria in AP pathogenesis and highlight their potential as therapeutic targets.

Footnotes

Provenance and peer review: Unsolicited 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, Grade B, Grade C

Novelty: Grade A, Grade B, Grade B, Grade B

Creativity or Innovation: Grade B, Grade B, Grade B, Grade B

Scientific Significance: Grade A, Grade A, Grade B, Grade B

P-Reviewer: Valencia ED, Professor, Colombia; Zhang XB, China; Zhao GD, PhD, Associate Professor, China S-Editor: Wu S L-Editor: A P-Editor: Lei YY

References
1.  Lee PJ, Papachristou GI. New insights into acute pancreatitis. Nat Rev Gastroenterol Hepatol. 2019;16:479-496.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 218]  [Cited by in RCA: 558]  [Article Influence: 93.0]  [Reference Citation Analysis (0)]
2.  Garg PK, Singh VP. Organ Failure Due to Systemic Injury in Acute Pancreatitis. Gastroenterology. 2019;156:2008-2023.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 166]  [Cited by in RCA: 412]  [Article Influence: 68.7]  [Reference Citation Analysis (0)]
3.  Voronina S, Chvanov M, De Faveri F, Mayer U, Wileman T, Criddle D, Tepikin A. Autophagy, Acute Pancreatitis and the Metamorphoses of a Trypsinogen-Activating Organelle. Cells. 2022;11:2514.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 23]  [Reference Citation Analysis (0)]
4.  Saluja A, Dudeja V, Dawra R, Sah RP. Early Intra-Acinar Events in Pathogenesis of Pancreatitis. Gastroenterology. 2019;156:1979-1993.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 105]  [Cited by in RCA: 198]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
5.  Pasca di Magliano M, Forsmark C, Freedman S, Hebrok M, Pasricha PJ, Saluja A, Stanger BZ, Holt J, Serrano J, James SP, Rustgi AK. Advances in acute and chronic pancreatitis: from development to inflammation and repair. Gastroenterology. 2013;144:e1-e4.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
6.  Ge P, Luo Y, Okoye CS, Chen H, Liu J, Zhang G, Xu C, Chen H. Intestinal barrier damage, systemic inflammatory response syndrome, and acute lung injury: A troublesome trio for acute pancreatitis. Biomed Pharmacother. 2020;132:110770.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 22]  [Cited by in RCA: 139]  [Article Influence: 27.8]  [Reference Citation Analysis (0)]
7.  Gukovsky I, Pandol SJ, Gukovskaya AS. Organellar dysfunction in the pathogenesis of pancreatitis. Antioxid Redox Signal. 2011;15:2699-2710.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 53]  [Cited by in RCA: 70]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
8.  Habtezion A, Gukovskaya AS, Pandol SJ. Acute Pancreatitis: A Multifaceted Set of Organelle and Cellular Interactions. Gastroenterology. 2019;156:1941-1950.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 91]  [Cited by in RCA: 209]  [Article Influence: 34.8]  [Reference Citation Analysis (1)]
9.  Harrington JS, Ryter SW, Plataki M, Price DR, Choi AMK. Mitochondria in health, disease, and aging. Physiol Rev. 2023;103:2349-2422.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 146]  [Cited by in RCA: 352]  [Article Influence: 176.0]  [Reference Citation Analysis (0)]
10.  Chen F, Xu K, Han Y, Ding J, Ren J, Wang Y, Ma Z, Cao F. Mitochondrial dysfunction in pancreatic acinar cells: mechanisms and therapeutic strategies in acute pancreatitis. Front Immunol. 2024;15:1503087.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
11.  Biczo G, Vegh ET, Shalbueva N, Mareninova OA, Elperin J, Lotshaw E, Gretler S, Lugea A, Malla SR, Dawson D, Ruchala P, Whitelegge J, French SW, Wen L, Husain SZ, Gorelick FS, Hegyi P, Rakonczay Z Jr, Gukovsky I, Gukovskaya AS. Mitochondrial Dysfunction, Through Impaired Autophagy, Leads to Endoplasmic Reticulum Stress, Deregulated Lipid Metabolism, and Pancreatitis in Animal Models. Gastroenterology. 2018;154:689-703.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 294]  [Cited by in RCA: 295]  [Article Influence: 42.1]  [Reference Citation Analysis (0)]
12.  Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol. 2020;21:363-383.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1094]  [Cited by in RCA: 3123]  [Article Influence: 624.6]  [Reference Citation Analysis (0)]
13.  Lingappan K. NF-κB in Oxidative Stress. Curr Opin Toxicol. 2018;7:81-86.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 511]  [Cited by in RCA: 703]  [Article Influence: 100.4]  [Reference Citation Analysis (0)]
14.  Huang H, Liu Y, Daniluk J, Gaiser S, Chu J, Wang H, Li ZS, Logsdon CD, Ji B. Activation of nuclear factor-κB in acinar cells increases the severity of pancreatitis in mice. Gastroenterology. 2013;144:202-210.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 180]  [Cited by in RCA: 187]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
15.  Muili KA, Jin S, Orabi AI, Eisses JF, Javed TA, Le T, Bottino R, Jayaraman T, Husain SZ. Pancreatic acinar cell nuclear factor κB activation because of bile acid exposure is dependent on calcineurin. J Biol Chem. 2013;288:21065-21073.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 23]  [Cited by in RCA: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
16.  Brand MD, Nicholls DG. Assessing mitochondrial dysfunction in cells. Biochem J. 2011;435:297-312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1580]  [Cited by in RCA: 1953]  [Article Influence: 139.5]  [Reference Citation Analysis (0)]
17.  Pandol SJ, Gottlieb RA. Calcium, mitochondria and the initiation of acute pancreatitis. Pancreatology. 2022;22:838-845.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 16]  [Reference Citation Analysis (0)]
18.  Choi J, Oh TG, Jung HW, Park KY, Shin H, Jo T, Kang DS, Chanda D, Hong S, Kim J, Hwang H, Ji M, Jung M, Shoji T, Matsushima A, Kim P, Mun JY, Paik MJ, Cho SJ, Lee IK, Whitcomb DC, Greer P, Blobner B, Goodarzi MO, Pandol SJ, Rotter JI; North American Pancreatitis Study 2 (NAPS2) Consortium, Fan W, Bapat SP, Zheng Y, Liddle C, Yu RT, Atkins AR, Downes M, Yoshihara E, Evans RM, Suh JM. Estrogen-Related Receptor γ Maintains Pancreatic Acinar Cell Function and Identity by Regulating Cellular Metabolism. Gastroenterology. 2022;163:239-256.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
19.  Zhou X, Jin S, Pan J, Lin Q, Yang S, Ambe PC, Basharat Z, Zimmer V, Wang W, Hong W. Damage associated molecular patterns and neutrophil extracellular traps in acute pancreatitis. Front Cell Infect Microbiol. 2022;12:927193.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 36]  [Reference Citation Analysis (0)]
20.  Vanpouille-Box C, Hoffmann JA, Galluzzi L. Pharmacological modulation of nucleic acid sensors - therapeutic potential and persisting obstacles. Nat Rev Drug Discov. 2019;18:845-867.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 84]  [Cited by in RCA: 153]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
21.  Shalbueva N, Mareninova OA, Gerloff A, Yuan J, Waldron RT, Pandol SJ, Gukovskaya AS. Effects of oxidative alcohol metabolism on the mitochondrial permeability transition pore and necrosis in a mouse model of alcoholic pancreatitis. Gastroenterology. 2013;144:437-446.e6.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 81]  [Cited by in RCA: 105]  [Article Influence: 8.8]  [Reference Citation Analysis (2)]
22.  Maléth J, Rakonczay Z Jr, Venglovecz V, Dolman NJ, Hegyi P. Central role of mitochondrial injury in the pathogenesis of acute pancreatitis. Acta Physiol (Oxf). 2013;207:226-235.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 48]  [Cited by in RCA: 49]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
23.  Vakifahmetoglu-Norberg H, Ouchida AT, Norberg E. The role of mitochondria in metabolism and cell death. Biochem Biophys Res Commun. 2017;482:426-431.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 297]  [Cited by in RCA: 505]  [Article Influence: 63.1]  [Reference Citation Analysis (0)]
24.  Guo R, Gu J, Zong S, Wu M, Yang M. Structure and mechanism of mitochondrial electron transport chain. Biomed J. 2018;41:9-20.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 89]  [Cited by in RCA: 150]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
25.  Checchetto V, Szabo I. Novel Channels of the Outer Membrane of Mitochondria: Recent Discoveries Change Our View. Bioessays. 2018;40:e1700232.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
26.  Petersen OH. Ca2+ signalling and Ca2+-activated ion channels in exocrine acinar cells. Cell Calcium. 2005;38:171-200.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 81]  [Cited by in RCA: 75]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
27.  Johnson PR, Dolman NJ, Pope M, Vaillant C, Petersen OH, Tepikin AV, Erdemli G. Non-uniform distribution of mitochondria in pancreatic acinar cells. Cell Tissue Res. 2003;313:37-45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 46]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
28.  Park MK, Ashby MC, Erdemli G, Petersen OH, Tepikin AV. Perinuclear, perigranular and sub-plasmalemmal mitochondria have distinct functions in the regulation of cellular calcium transport. EMBO J. 2001;20:1863-1874.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 238]  [Cited by in RCA: 252]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
29.  Petersen OH. Specific mitochondrial functions in separate sub-cellular domains of pancreatic acinar cells. Pflugers Arch. 2012;464:77-87.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
30.  Criddle DN, McLaughlin E, Murphy JA, Petersen OH, Sutton R. The pancreas misled: signals to pancreatitis. Pancreatology. 2007;7:436-446.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 41]  [Cited by in RCA: 47]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
31.  Manhas N. Computational Model of Complex Calcium Dynamics: Store Operated Ca(2+) Channels and Mitochondrial Associated Membranes in Pancreatic Acinar Cells. Cell Biochem Biophys. 2025;83:519-535.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
32.  Guo L. Mitochondria and the permeability transition pore in cancer metabolic reprogramming. Biochem Pharmacol. 2021;188:114537.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
33.  Lerch MM, Halangk W, Mayerle J. Preventing pancreatitis by protecting the mitochondrial permeability transition pore. Gastroenterology. 2013;144:265-269.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
34.  Halestrap AP, Richardson AP. The mitochondrial permeability transition: a current perspective on its identity and role in ischaemia/reperfusion injury. J Mol Cell Cardiol. 2015;78:129-141.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 299]  [Cited by in RCA: 334]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
35.  Petersen OH, Gerasimenko JV, Gerasimenko OV, Gryshchenko O, Peng S. The roles of calcium and ATP in the physiology and pathology of the exocrine pancreas. Physiol Rev. 2021;101:1691-1744.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 28]  [Cited by in RCA: 102]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
36.  Ward JB, Petersen OH, Jenkins SA, Sutton R. Is an elevated concentration of acinar cytosolic free ionised calcium the trigger for acute pancreatitis? Lancet. 1995;346:1016-1019.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 131]  [Cited by in RCA: 115]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
37.  Reed AM, Husain SZ, Thrower E, Alexandre M, Shah A, Gorelick FS, Nathanson MH. Low extracellular pH induces damage in the pancreatic acinar cell by enhancing calcium signaling. J Biol Chem. 2011;286:1919-1926.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 20]  [Cited by in RCA: 21]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
38.  Voronina S, Sherwood M, Barrow S, Dolman N, Conant A, Tepikin A. Downstream from calcium signalling: mitochondria, vacuoles and pancreatic acinar cell damage. Acta Physiol (Oxf). 2009;195:161-169.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 6]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
39.  Li J, Zhou R, Zhang J, Li ZF. Calcium signaling of pancreatic acinar cells in the pathogenesis of pancreatitis. World J Gastroenterol. 2014;20:16146-16152.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 32]  [Cited by in RCA: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
40.  Giorgi C, Marchi S, Pinton P. The machineries, regulation and cellular functions of mitochondrial calcium. Nat Rev Mol Cell Biol. 2018;19:713-730.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 331]  [Cited by in RCA: 623]  [Article Influence: 89.0]  [Reference Citation Analysis (0)]
41.  Lewarchik CM, Orabi AI, Jin S, Wang D, Muili KA, Shah AU, Eisses JF, Malik A, Bottino R, Jayaraman T, Husain SZ. The ryanodine receptor is expressed in human pancreatic acinar cells and contributes to acinar cell injury. Am J Physiol Gastrointest Liver Physiol. 2014;307:G574-G581.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 13]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
42.  Futatsugi A, Nakamura T, Yamada MK, Ebisui E, Nakamura K, Uchida K, Kitaguchi T, Takahashi-Iwanaga H, Noda T, Aruga J, Mikoshiba K. IP3 receptor types 2 and 3 mediate exocrine secretion underlying energy metabolism. Science. 2005;309:2232-2234.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 248]  [Cited by in RCA: 264]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
43.  Gerasimenko JV, Lur G, Sherwood MW, Ebisui E, Tepikin AV, Mikoshiba K, Gerasimenko OV, Petersen OH. Pancreatic protease activation by alcohol metabolite depends on Ca2+ release via acid store IP3 receptors. Proc Natl Acad Sci U S A. 2009;106:10758-10763.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 87]  [Cited by in RCA: 89]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
44.  Gerasimenko JV, Gerasimenko OV. The role of Ca(2+) signalling in the pathology of exocrine pancreas. Cell Calcium. 2023;112:102740.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
45.  Gerasimenko JV, Gryshchenko O, Ferdek PE, Stapleton E, Hébert TO, Bychkova S, Peng S, Begg M, Gerasimenko OV, Petersen OH. Ca2+ release-activated Ca2+ channel blockade as a potential tool in antipancreatitis therapy. Proc Natl Acad Sci U S A. 2013;110:13186-13191.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 127]  [Cited by in RCA: 157]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
46.  Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med. 2013;368:651-662.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1792]  [Cited by in RCA: 1937]  [Article Influence: 161.4]  [Reference Citation Analysis (0)]
47.  Mareninova OA, Hermann K, French SW, O'Konski MS, Pandol SJ, Webster P, Erickson AH, Katunuma N, Gorelick FS, Gukovsky I, Gukovskaya AS. Impaired autophagic flux mediates acinar cell vacuole formation and trypsinogen activation in rodent models of acute pancreatitis. J Clin Invest. 2009;119:3340-3355.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 186]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
48.  Huynh KK, Eskelinen EL, Scott CC, Malevanets A, Saftig P, Grinstein S. LAMP proteins are required for fusion of lysosomes with phagosomes. EMBO J. 2007;26:313-324.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 491]  [Cited by in RCA: 517]  [Article Influence: 28.7]  [Reference Citation Analysis (0)]
49.  Fortunato F, Bürgers H, Bergmann F, Rieger P, Büchler MW, Kroemer G, Werner J. Impaired autolysosome formation correlates with Lamp-2 depletion: role of apoptosis, autophagy, and necrosis in pancreatitis. Gastroenterology. 2009;137:350-360, 360.e1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 182]  [Cited by in RCA: 188]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
50.  Hirota M, Ohmuraya M, Hashimoto D, Suyama K, Sugita H, Ogawa M. Roles of Autophagy and Pancreatic Secretory Trypsin Inhibitor in Trypsinogen Activation in Acute Pancreatitis. Pancreas. 2020;49:493-497.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
51.  Yang L, Ye F, Liu J, Klionsky DJ, Tang D, Kang R. Extracellular SQSTM1 exacerbates acute pancreatitis by activating autophagy-dependent ferroptosis. Autophagy. 2023;19:1733-1744.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 30]  [Cited by in RCA: 64]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
52.  Gukovskaya AS, Gukovsky I. Autophagy and pancreatitis. Am J Physiol Gastrointest Liver Physiol. 2012;303:G993-G1003.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 97]  [Cited by in RCA: 115]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
53.  Mareninova OA, Jia W, Gretler SR, Holthaus CL, Thomas DDH, Pimienta M, Dillon DL, Gukovskaya AS, Gukovsky I, Groblewski GE. Transgenic expression of GFP-LC3 perturbs autophagy in exocrine pancreas and acute pancreatitis responses in mice. Autophagy. 2020;16:2084-2097.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 77]  [Cited by in RCA: 72]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
54.  Wang S, Long H, Hou L, Feng B, Ma Z, Wu Y, Zeng Y, Cai J, Zhang DW, Zhao G. The mitophagy pathway and its implications in human diseases. Signal Transduct Target Ther. 2023;8:304.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 406]  [Article Influence: 203.0]  [Reference Citation Analysis (0)]
55.  Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol. 2011;12:9-14.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2666]  [Cited by in RCA: 2590]  [Article Influence: 185.0]  [Reference Citation Analysis (0)]
56.  Liu L, Feng D, Chen G, Chen M, Zheng Q, Song P, Ma Q, Zhu C, Wang R, Qi W, Huang L, Xue P, Li B, Wang X, Jin H, Wang J, Yang F, Liu P, Zhu Y, Sui S, Chen Q. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat Cell Biol. 2012;14:177-185.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 960]  [Cited by in RCA: 1314]  [Article Influence: 101.1]  [Reference Citation Analysis (0)]
57.  Lee J, Lim JW, Kim H. Lycopene Inhibits Oxidative Stress-Mediated Inflammatory Responses in Ethanol/Palmitoleic Acid-Stimulated Pancreatic Acinar AR42J Cells. Int J Mol Sci. 2021;22:2101.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
58.  Yang S, Bing M, Chen F, Sun Y, Chen H, Chen W. Autophagy regulation by the nuclear factor κB signal axis in acute pancreatitis. Pancreas. 2012;41:367-373.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 12]  [Cited by in RCA: 16]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
59.  Sastre J, Pérez S, Sabater L, Rius-Pérez S. Redox signaling in the pancreas in health and disease. Physiol Rev. 2025;105:593-650.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 18]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
60.  Zheng X, Li L, Zhu Y, Huang X, Zhang Y, Yu B, He W, Lv N. Superoxide Dismutase Predicts Persistent Circulation Failure and Mortality in the Early Stage of Acute Pancreatitis. Dig Dis Sci. 2020;65:3551-3557.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
61.  Pérez S, Pereda J, Sabater L, Sastre J. Redox signaling in acute pancreatitis. Redox Biol. 2015;5:1-14.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 71]  [Cited by in RCA: 98]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
62.  Criddle DN, Gillies S, Baumgartner-Wilson HK, Jaffar M, Chinje EC, Passmore S, Chvanov M, Barrow S, Gerasimenko OV, Tepikin AV, Sutton R, Petersen OH. Menadione-induced reactive oxygen species generation via redox cycling promotes apoptosis of murine pancreatic acinar cells. J Biol Chem. 2006;281:40485-40492.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 305]  [Cited by in RCA: 285]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
63.  Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469:221-225.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3253]  [Cited by in RCA: 4397]  [Article Influence: 293.1]  [Reference Citation Analysis (0)]
64.  Anderson P. Post-transcriptional control of cytokine production. Nat Immunol. 2008;9:353-359.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 303]  [Cited by in RCA: 346]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
65.  Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA, Ryter SW, Choi AM. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;12:222-230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2499]  [Cited by in RCA: 2464]  [Article Influence: 176.0]  [Reference Citation Analysis (0)]
66.  Liu Q, Zhang D, Hu D, Zhou X, Zhou Y. The role of mitochondria in NLRP3 inflammasome activation. Mol Immunol. 2018;103:115-124.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 182]  [Cited by in RCA: 365]  [Article Influence: 52.1]  [Reference Citation Analysis (0)]
67.  Ryter SW, Kim HP, Hoetzel A, Park JW, Nakahira K, Wang X, Choi AM. Mechanisms of cell death in oxidative stress. Antioxid Redox Signal. 2007;9:49-89.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 876]  [Cited by in RCA: 937]  [Article Influence: 52.1]  [Reference Citation Analysis (0)]
68.  Elrod JW, Wong R, Mishra S, Vagnozzi RJ, Sakthievel B, Goonasekera SA, Karch J, Gabel S, Farber J, Force T, Brown JH, Murphy E, Molkentin JD. Cyclophilin D controls mitochondrial pore-dependent Ca(2+) exchange, metabolic flexibility, and propensity for heart failure in mice. J Clin Invest. 2010;120:3680-3687.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 311]  [Cited by in RCA: 316]  [Article Influence: 21.1]  [Reference Citation Analysis (0)]
69.  Bauer TM, Murphy E. Role of Mitochondrial Calcium and the Permeability Transition Pore in Regulating Cell Death. Circ Res. 2020;126:280-293.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 252]  [Cited by in RCA: 323]  [Article Influence: 64.6]  [Reference Citation Analysis (0)]
70.  West AP, Shadel GS. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat Rev Immunol. 2017;17:363-375.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 466]  [Cited by in RCA: 795]  [Article Influence: 99.4]  [Reference Citation Analysis (0)]
71.  Hu MM, Shu HB. Mitochondrial DNA-triggered innate immune response: mechanisms and diseases. Cell Mol Immunol. 2023;20:1403-1412.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 44]  [Cited by in RCA: 97]  [Article Influence: 48.5]  [Reference Citation Analysis (0)]
72.  Yakah W, Shah I, Skelton-Badlani D, Freedman SD, Popov YV, Sheth SG; BIDMC Acute Pancreatitis Working Group. Circulating Mitochondrial DNA as a Diagnostic Biomarker for Predicting Disease Severity in Patients With Acute Pancreatitis. Gastroenterology. 2023;164:1009-1011.e3.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
73.  Debray FG, Drouin E, Herzog D, Lortie A, Lambert M, Garel L, Mitchell GA, Michaud JL. Recurrent pancreatitis in mitochondrial cytopathy. Am J Med Genet A. 2006;140:2330-2335.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 13]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
74.  Vanasco V, Ropolo A, Grasso D, Ojeda DS, García MN, Vico TA, Orquera T, Quarleri J, Alvarez S, Vaccaro MI. Mitochondrial Dynamics and VMP1-Related Selective Mitophagy in Experimental Acute Pancreatitis. Front Cell Dev Biol. 2021;9:640094.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
75.  Singh L, Bakshi DK, Majumdar S, Arora SK, Vasishta RK, Wig JD. Mitochondrial dysfunction and apoptosis of acinar cells in chronic pancreatitis. J Gastroenterol. 2008;43:473-483.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11]  [Cited by in RCA: 16]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
76.  Gao Y, Mi N, Wu W, Zhao Y, Fan F, Liao W, Ming Y, Guan W, Bai C. Transfer of inflammatory mitochondria via extracellular vesicles from M1 macrophages induces ferroptosis of pancreatic beta cells in acute pancreatitis. J Extracell Vesicles. 2024;13:e12410.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 43]  [Article Influence: 43.0]  [Reference Citation Analysis (0)]
77.  Zhang D, Li J, Zhao L, Yang Z, Wu C, Liu Y, Li W, Jin Z, Ma J. Mitochondrial DNA Leakage Promotes Persistent Pancreatic Acinar Cell Injury in Acute Pancreatitis via the cGAS-STING-NF-κB Pathway. Inflammation. 2025;48:1420-1437.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
78.  Peng Y, Yang Y, Li Y, Shi T, Xu N, Liu R, Luan Y, Yao Y, Yin C. Mitochondrial (mt)DNA-cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling promotes pyroptosis of macrophages via interferon regulatory factor (IRF)7/IRF3 activation to aggravate lung injury during severe acute pancreatitis. Cell Mol Biol Lett. 2024;29:61.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 37]  [Reference Citation Analysis (0)]
79.  Verny C, Amati-Bonneau P, Letournel F, Person B, Dib N, Malinge MC, Slama A, Le Maréchal C, Ferec C, Procaccio V, Reynier P, Bonneau D. Mitochondrial DNA A3243G mutation involved in familial diabetes, chronic intestinal pseudo-obstruction and recurrent pancreatitis. Diabetes Metab. 2008;34:620-626.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 21]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
80.  Son A, Ahuja M, Schwartz DM, Varga A, Swaim W, Kang N, Maleth J, Shin DM, Muallem S. Ca(2+) Influx Channel Inhibitor SARAF Protects Mice From Acute Pancreatitis. Gastroenterology. 2019;157:1660-1672.e2.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 24]  [Cited by in RCA: 42]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
81.  Wen L, Voronina S, Javed MA, Awais M, Szatmary P, Latawiec D, Chvanov M, Collier D, Huang W, Barrett J, Begg M, Stauderman K, Roos J, Grigoryev S, Ramos S, Rogers E, Whitten J, Velicelebi G, Dunn M, Tepikin AV, Criddle DN, Sutton R. Inhibitors of ORAI1 Prevent Cytosolic Calcium-Associated Injury of Human Pancreatic Acinar Cells and Acute Pancreatitis in 3 Mouse Models. Gastroenterology. 2015;149:481-92.e7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 151]  [Cited by in RCA: 173]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
82.  Muili KA, Wang D, Orabi AI, Sarwar S, Luo Y, Javed TA, Eisses JF, Mahmood SM, Jin S, Singh VP, Ananthanaravanan M, Perides G, Williams JA, Molkentin JD, Husain SZ. Bile acids induce pancreatic acinar cell injury and pancreatitis by activating calcineurin. J Biol Chem. 2013;288:570-580.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 70]  [Cited by in RCA: 70]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
83.  Farooq A, Hernandez L, Swain SM, Shahid RA, Romac JM, Vigna SR, Liddle RA. Initiation and severity of experimental pancreatitis are modified by phosphate. Am J Physiol Gastrointest Liver Physiol. 2022;322:G561-G570.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
84.  Farooq A, Richman CM, Swain SM, Shahid RA, Vigna SR, Liddle RA. The Role of Phosphate in Alcohol-Induced Experimental Pancreatitis. Gastroenterology. 2021;161:982-995.e2.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
85.  Javed MA, Wen L, Awais M, Latawiec D, Huang W, Chvanov M, Schaller S, Bordet T, Michaud M, Pruss R, Tepikin A, Criddle D, Sutton R. TRO40303 Ameliorates Alcohol-Induced Pancreatitis Through Reduction of Fatty Acid Ethyl Ester-Induced Mitochondrial Injury and Necrotic Cell Death. Pancreas. 2018;47:18-24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 27]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
86.  Vege SS, DiMagno MJ, Forsmark CE, Martel M, Barkun AN. Initial Medical Treatment of Acute Pancreatitis: American Gastroenterological Association Institute Technical Review. Gastroenterology. 2018;154:1103-1139.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 142]  [Cited by in RCA: 179]  [Article Influence: 25.6]  [Reference Citation Analysis (1)]
87.  Pandol SJ, Saluja AK, Imrie CW, Banks PA. Acute pancreatitis: bench to the bedside. Gastroenterology. 2007;132:1127-1151.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 433]  [Cited by in RCA: 456]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
88.  Tóth E, Maléth J, Závogyán N, Fanczal J, Grassalkovich A, Erdős R, Pallagi P, Horváth G, Tretter L, Bálint ER, Rakonczay Z Jr, Venglovecz V, Hegyi P. Novel mitochondrial transition pore inhibitor N-methyl-4-isoleucine cyclosporin is a new therapeutic option in acute pancreatitis. J Physiol. 2019;597:5879-5898.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 29]  [Cited by in RCA: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
89.  Márta K, Szabó AN, Pécsi D, Varjú P, Bajor J, Gódi S, Sarlós P, Mikó A, Szemes K, Papp M, Tornai T, Vincze Á, Márton Z, Vincze PA, Lankó E, Szentesi A, Molnár T, Hágendorn R, Faluhelyi N, Battyáni I, Kelemen D, Papp R, Miseta A, Verzár Z, Lerch MM, Neoptolemos JP, Sahin-Tóth M, Petersen OH, Hegyi P; Hungarian Pancreatic Study Group. High versus low energy administration in the early phase of acute pancreatitis (GOULASH trial): protocol of a multicentre randomised double-blind clinical trial. BMJ Open. 2017;7:e015874.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 28]  [Cited by in RCA: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
90.  Hu Z, Wang D, Gong J, Li Y, Ma Z, Luo T, Jia X, Shi Y, Song Z. MSCs Deliver Hypoxia-Treated Mitochondria Reprogramming Acinar Metabolism to Alleviate Severe Acute Pancreatitis Injury. Adv Sci (Weinh). 2023;10:e2207691.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 31]  [Cited by in RCA: 36]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
91.  Zhang R, Zhu Z, Ma Y, Tang T, Wu J, Huang F, Xu L, Wang Y, Zhou J. Rhizoma Alismatis Decoction improved mitochondrial dysfunction to alleviate SASP by enhancing autophagy flux and apoptosis in hyperlipidemia acute pancreatitis. Phytomedicine. 2024;129:155629.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 15]  [Reference Citation Analysis (0)]
92.  Yuan J, Wei Z, Xin G, Liu X, Zhou Z, Zhang Y, Yu X, Wan C, Chen Q, Zhao W, Wang X, Dong Y, Chen Z, Chen X, Niu H, Huang W. Vitamin B(12) Attenuates Acute Pancreatitis by Suppressing Oxidative Stress and Improving Mitochondria Dysfunction via CBS/SIRT1 Pathway. Oxid Med Cell Longev. 2021;2021:7936316.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
93.  Zhang J, Huang W, He Q, Deng T, Wu B, Huang F, Bi J, Jin Y, Sun H, Zhang Q, Shi K. PINK1/PARK2 dependent mitophagy effectively suppresses NLRP3 inflammasome to alleviate acute pancreatitis. Free Radic Biol Med. 2021;166:147-164.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
94.  Piplani H, Marek-Iannucci S, Sin J, Hou J, Takahashi T, Sharma A, de Freitas Germano J, Waldron RT, Saadaeijahromi H, Song Y, Gulla A, Wu B, Lugea A, Andres AM, Gaisano HY, Gottlieb RA, Pandol SJ. Simvastatin induces autophagic flux to restore cerulein-impaired phagosome-lysosome fusion in acute pancreatitis. Biochim Biophys Acta Mol Basis Dis. 2019;1865:165530.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 26]  [Cited by in RCA: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
95.  Shen Y, Wen L, Zhang R, Wei Z, Shi N, Xiong Q, Xia Q, Xing Z, Zeng Z, Niu H, Huang W. Dihydrodiosgenin protects against experimental acute pancreatitis and associated lung injury through mitochondrial protection and PI3Kγ/Akt inhibition. Br J Pharmacol. 2018;175:1621-1636.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 30]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
96.  Li Y, Zhu Y, Li S, Dong Y, Wan C, Yu X, Xin G, Wei Z, Li F, Wang Y, Zhang K, Chen Q, Zhang C, Wen E, Niu H, Huang W. Deoxyarbutin attenuates severe acute pancreatitis via the HtrA2/PGC-1α pathway. Free Radic Res. 2022;56:651-665.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
97.  Li X, Wang T, Zhou Q, Li F, Liu T, Zhang K, Wen A, Feng L, Shu X, Tian S, Liu Y, Gao Y, Xia Q, Xin G, Huang W. Isorhamnetin Alleviates Mitochondrial Injury in Severe Acute Pancreatitis via Modulation of KDM5B/HtrA2 Signaling Pathway. Int J Mol Sci. 2024;25:3784.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
98.  Lam LKM, Murphy S, Kokkinaki D, Venosa A, Sherrill-Mix S, Casu C, Rivella S, Weiner A, Park J, Shin S, Vaughan AE, Hahn BH, Odom John AR, Meyer NJ, Hunter CA, Worthen GS, Mangalmurti NS. DNA binding to TLR9 expressed by red blood cells promotes innate immune activation and anemia. Sci Transl Med. 2021;13:eabj1008.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 36]  [Cited by in RCA: 148]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
99.  Hoque R, Sohail M, Malik A, Sarwar S, Luo Y, Shah A, Barrat F, Flavell R, Gorelick F, Husain S, Mehal W. TLR9 and the NLRP3 inflammasome link acinar cell death with inflammation in acute pancreatitis. Gastroenterology. 2011;141:358-369.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 245]  [Cited by in RCA: 241]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
100.  Xie X, Wang Z, Zhang H, Lu J, Cao F, Li F. Identification of mitophagy-related biomarkers in severe acute pancreatitis: integration of WGCNA, machine learning algorithms and scRNA-seq. Front Immunol. 2025;16:1594085.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
101.  Zhou Y, Huang X, Jin Y, Qiu M, Ambe PC, Basharat Z, Hong W. The role of mitochondrial damage-associated molecular patterns in acute pancreatitis. Biomed Pharmacother. 2024;175:116690.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
102.  Liu W, Wang X, Xu D, Gong F, Pei L, Yang S, Zhao S, Zheng X, Li R, Yang Z, Fei J, Mao E, Chen E, Chen Y. SIRT5 mediated succinylation of SUCLA2 regulates TCA cycle dysfunction and mitochondrial damage in pancreatic acinar cells in acute pancreatitis. Biochim Biophys Acta Mol Basis Dis. 2025;1871:167613.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
103.  Yao J, Jiang Y, Zhang P, Miao Y, Wu X, Lei H, Xie Z, Tian Y, Zhao X, Li J, Zhu L, Wan M, Tang W. Genetic and pharmacological targeting of HINT2 promotes OXPHOS to alleviate inflammatory responses and cell necrosis in acute pancreatitis. Pharmacol Res. 2025;212:107620.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
104.  Wang L, Gao Z, Tian M, Liu L, Xie J, Chen M, Huang Z, Dong B, Li W, Shi L, Tong Y, Xu H, Shen B, Cen D, Yu H, Yu X. A Nanosystem Alleviates Severe Acute Pancreatitis via Reactive Oxygen Species Scavenging and Enhancing Mitochondrial Autophagy. Nano Lett. 2025;25:8644-8654.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
105.  Yan J, Liu H, Xu Y, Sun X, Meng X, Wei S, Li Z, Jin X, Liu J, Wang X, Cui P, He Z. Tailored "Three-stage booster" nano-extinguisher for synergistic treatment of severe acute pancreatitis by rectifying mitochondrial dysfunction and inhibiting pancreatic autodigestion. Acta Biomater. 2025;200:569-590.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
106.  Wang D, Wang S, Liu J, Shi X, Xiong T, Li R, Wei W, Ji L, Huang Q, Gong X, Ai K. Nanomedicine Penetrating Blood-Pancreas Barrier for Effective Treatment of Acute Pancreatitis. Adv Sci (Weinh). 2025;12:e2413925.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
107.  Lou P, Zhou X, Zhang Y, Xie Y, Wang Y, Wang C, Liu S, Wan M, Lu Y, Liu J. Harnessing tissue-derived mitochondria-rich extracellular vesicles (Ti-mitoEVs) to boost mitochondrial biogenesis for regenerative medicine. Sci Adv. 2025;11:eadt1318.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
108.  Wu J, Huang H, Xu W, Cui B, Sun P, Hao X, Jiang S, Hou X, Qi X, Wei Z, Cheng Y, Zheng Y, Liu K, He J. Inflammation-driven biomimetic nano-polyphenol drug delivery system alleviates severe acute pancreatitis by inhibiting macrophage PANoptosis and pancreatic enzymes oversecretion. J Adv Res. 2025;S2090-1232(25)00225.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]