Back to Journals » Drug Design, Development and Therapy » Volume 17

The Role of AMPK Signaling in Ulcerative Colitis

Authors Yuan Y, Wang F, Liu X, Shuai B, Fan H 

Received 18 October 2023

Accepted for publication 22 December 2023

Published 29 December 2023 Volume 2023:17 Pages 3855—3875

DOI https://doi.org/10.2147/DDDT.S442154

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Anastasios Lymperopoulos



Yuyi Yuan,1,* Fang Wang,2,* Xingxing Liu,1,* Bo Shuai,1 Heng Fan1

1Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China; 2Department of Rehabilitation Medicine, Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Jingshan, Hubei, 431800, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Bo Shuai; Heng Fan, Email [email protected]; [email protected]

Abstract: Ulcerative colitis (UC) is a chronic non-specific inflammatory bowel disease characterized by inflammation and ulcer formation of the intestinal mucosa. Due to its high recurrence rate, prolonged course, limited curative options, and significant impact on patients’ quality of life, along with a notable potential for malignant transformation, UC is designated as a refractory global health challenge by the World Health Organization (WHO). The elucidation of the pathogenesis and therapeutic strategies for UC requires further in-depth investigation. AMP-activated protein kinase (AMPK) serves as a central regulator of cellular energy metabolic homeostasis. Emerging evidence indicates that interventions involving traditional Chinese medicine (TCM) components, as well as other pharmacological measures, exert beneficial effects on the intestinal mucosal inflammation and epithelial barrier dysfunction in UC by modulating AMPK signaling, thereby influencing biological processes such as cellular autophagy, apoptosis, inflammatory responses, macrophage polarization, and NLRP3 inflammasome-mediated pyroptosis. The role of AMPK in UC is of significant importance. This manuscript provides a comprehensive overview of the mechanisms through which AMPK is involved in UC, as well as a compilation of pharmacological agents capable of activating the AMPK signaling pathway within the context of UC. The primary objective is to facilitate a deeper comprehension of the pivotal role of AMPK in UC among researchers and clinical practitioners, thereby advancing the identification of novel therapeutic targets for interventions in UC.

Keywords: ulcerative colitis, AMPK, colitis-associated biological processes, traditional Chinese medicine monomers

Introduction

Ulcerative colitis (UC) is a chronic non-specific inflammatory bowel disease (IBD) characterized by chronic inflammation and ulcer formation of the intestinal mucosa. It presents clinically with symptoms such as abdominal pain, diarrhea, and mucous bloody stools. The disease course typically exhibits a chronic, persistent, and recurrent nature.1 With changes in lifestyle and dietary patterns, the incidence of UC among Asian populations has been steadily increasing.2 It is generally believed that its pathogenesis is associated with genetic predisposition, environmental factors, luminal factors, and mucosal immune dysregulation.3 The therapeutic drugs for UC mainly include aminosalicylates, corticosteroids, biologics (such as infliximab), JAK inhibitors, and calcineurin inhibitors. Surgical intervention may be considered for refractory cases of the disease.4 Nevertheless, the clinical efficacy of these treatments is currently unsatisfactory, with potential long-term side effects, and a propensity for relapse upon discontinuation. Therefore, in-depth research into the pathogenesis, therapeutic methods, and novel treatment drugs and strategies for UC from an integrative perspective of traditional Chinese and Western medicine is highly necessary.

Adenosine monophosphate-activated protein kinase, AMPK, is a pivotal energy sensor and regulator within cells. It functions to perceive alterations in cellular energy status and elicits responses to maintain a balance between energy generation and expenditure by regulating various metabolic and immune-related activities. Its substrates encompass multiple key enzymes and transcriptional regulatory factors involved in cellular growth, proliferation, metabolism, and immune modulation processes.5 Evidence suggests that aberrant expression of AMPK plays a crucial role in the occurrence and progression of UC. For instance, the modulation of AMPK has the potential to influence cellular autophagy, thereby ameliorating intestinal mucosal immunity, inflammation, oxidative stress, and microbiota homeostasis. This modulation contributes to the preservation of the normal function of the intestinal mucosal barrier, thus emerging as a pivotal approach for effectively preventing and treating UC.6 This process is regulated by a variety of proteins and signaling pathways. When the organism is stimulated by factors such as infection, inflammation, and endoplasmic reticulum stress, AMPK is activated and mammalian target of rapamycin complex 1 (mTORC1) is inhibited, facilitating the initiation of autophagy through the activation of the Unc-51-like kinase (ULK) complex.7 Studies have indicated that Huangqin decoction improves ulcerative colitis in mice by regulating fatty acid metabolism through AMPK, thereby mediating macrophage polarization.8 Fermented Platycodon grandiflorum root extract modulates M1/M2 macrophage polarization through the AMPK/NFκB/NLRP3 pathway, enhancing the mRNA expression of intestinal epithelial cell tight junction proteins, and mitigating inflammation in a murine model of IBD.9 Research indicates that Schisandrin B regulates cell pyroptosis and reduces intestinal epithelial damage in colitis by modulating the AMPK/NRF2/NLRP3 inflammasome pathway.10 In a study by Su et al,11 it was found that berberine can enhance the expression levels of LC3II and LC3I proteins in colonic tissues of UC model mice, upregulate the colonic tissue cell P-AMPK/AMPK protein ratio, downregulate intestinal inflammatory responses, restore gut microbiota equilibrium, and mitigate intestinal mucosal barrier damage.12 Decades of research have demonstrated that dysfunctional autophagy results in disrupted intestinal epithelial function, dysbiosis of the gut microbiota, impaired Paneth cell antimicrobial peptide secretion, endoplasmic reticulum stress response, and aberrant immune responses to pathogens in ulcerative colitis UC;13 The dysregulated balance of macrophage polarization constitutes a significant factor in the pathogenesis of UC and serves as a therapeutic target;14,15 Cell pyroptosis can induce potent inflammatory responses to protect the host against microbial infections; however, excessive pyroptosis can lead to autoimmune diseases such as UC. Schisandrin B can mitigate intestinal epithelial cell damage in colitis by modulating cell pyroptosis through the AMPK/Nrf2/NLRP3 inflammasome pathway.10 AMPK plays a significant role in various biological mechanisms implicated in the pathogenesis of UC. Consequently, the activation of AMPK carries noteworthy therapeutic implications for UC treatment and merits in-depth exploration. This review aims to delve into the correlation between AMPK and regulatory mechanisms, including programmed cell death, mucosal immunity, and intestinal epithelial barrier function in UC. Furthermore, we will investigate the potential of specific Chinese herbal monomers or drugs to modulate these mechanisms through AMPK activation, thereby contributing to the advancement of UC treatment strategies.

The Concept of AMPK Activity Regulation Mechanisms

Introduction to AMPK Molecule

The history of AMPK (Figure 1A) can be traced back to 1973 when two different laboratories discovered the existence of an enzyme in the rat liver that could phosphorylate and inactivate acetyl-CoA carboxylase (ACC)16 and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGR),17 These two enzymes are key enzymes involved in fatty acid and cholesterol synthesis. In 1980, Yeh et al demonstrated that “ACC kinase” can be activated through 5’-AMP isomerization and proposed a pioneering hypothesis: this mechanism could serve to inhibit fatty acid synthesis (ATP consumption) in situations of compromised cellular energy status.18 In 1987, Carling et al confirmed that the activities of ACC and HMGR kinases are functions of the same protein kinase. This kinase can be activated both by AMP and through phosphorylation.19 In 1988, this kinase was renamed AMPK, reflecting the realization that AMPK has a minimum of two downstream targets (a review article listed no fewer than 60 targets).20 Subsequently, the critical phosphorylation activation site was identified as Thr172, located within the catalytic subunit of rat AMPK.21 The primary kinase responsible for phosphorylating Thr172 was eventually discovered in 2003 and definitively identified as the tumor suppressor LKB1.22

Figure 1 (A) A brief history of AMPK; (B) The AMPK molecule and its upstream and downstream mechanism.

AMPK is a serine/threonine kinase that is ubiquitously present in the form of a heterotrimeric complex composed of a catalytic α subunit and regulatory β and γ subunits. The α subunit encompasses the catalytic domain of the serine/threonine protein kinase and a pivotal residue, Thr172, which can be phosphorylated by upstream kinases. The β subunit includes a carbohydrate-binding module allowing AMPK to interact with glycogen, while the C-terminal domain of the β subunit stabilizes the interaction between the α and γ subunits. The γ subunit comprises four consecutive cystathionine-β-synthase (CBS) domains, binding adenine nucleotides, thereby ensuring the monitoring and regulation of ATP/AMP levels by AMPK.23 The γ subunit of AMPK binds to AMP or, to a lesser extent, ADP, thereby stimulating the activity of AMPK24 (as shown in Figure 1B). Genes encoding these subunits are found in the genomes of essentially all eukaryotic organisms, indicating that AMPK heterotrimeric complexes emerged early in the evolution of eukaryotes.25 A significant clue to the evolution of eukaryotic cells is the engulfment of aerobic bacteria, which eventually gave rise to mitochondria. Following this engulfment event, the host and mitochondria gradually developed the ability to exchange ADP and ATP. It can be anticipated that the host would require a regulatory mechanism to monitor and control this process.26 Given its capacity to monitor intracellular adenine nucleotide levels and its engagement in contemporary processes like mitochondrial biogenesis, mitosis, and mitochondrial fission, it is reasonable to conjecture that the functional role of AMPK may have emerged through this mechanism during evolution. Remarkably, mitochondrial impairment is recognized as a hallmark of pathological injury in ulcerative colitis,13 the pivotal significance of AMPK activation in addressing mitochondrial damage within UC intestinal epithelial cells is conspicuous. AMPK activation leads to a reduction in anabolic metabolism, consequently decreasing ATP consumption, while simultaneously bolstering catabolic metabolism to enhance ATP production, thereby upholding energy equilibrium. As a paramount controller of energy metabolism, AMPK’s functionality is intrinsically linked to cellular energy levels, particularly the concentrations of adenine nucleotides (AMP, ADP, and ATP).27 The interconversion between ATP and ADP constitutes a fundamental mechanism of energy cycling within most cells. Catabolic metabolism transforms ADP into ATP, generating a substantial reserve of easily accessible energy for cellular utilization. Conversely, the conversion of ATP to ADP liberates a noteworthy quantity of energy to drive diverse cellular processes.28

Upstream Activation Pathways of AMPK

The classical theory proposes that AMP plays a pivotal role as a signal for AMPK activation. It activates AMPK through three complementary mechanisms, all of which are countered by ATP: (1) promoting the phosphorylation of Thr172; (2) inhibiting the dephosphorylation of Thr172; and (3) inducing allosteric activation. These mechanisms confer onto mammalian AMPK a high sensitivity to alterations in the intracellular AMP/ATP ratio.29 Mammalian cells house a range of AMPK subtypes, comprising two α subunits, two β subunits, and three γ subunits, which can come together to generate up to 12 diverse heterotrimeric complexes. While the precise functions of these distinct isoform combinations in vivo remain partially elucidated, substantial evidence indicates potential discrepancies in tissue distribution, subcellular localization, and functionality. Notably, the γ2 and γ3 subunits, characterized by unique variable-length N-terminal regions, have been associated with the subcellular localization of AMPK.30 The most prominent upstream kinase of AMPK is the tumor suppressor protein LKB1 (also referred to as Stk11). Investigations involving mice with a knocked-out LKB1 gene have unveiled LKB1’s role in triggering AMPK activation during energy stress. Research performed on cells devoid of LKB1 has further demonstrated its role in mediating AMPK activation in response to mitochondrial damage and low-energy conditions.31,32

In relation to the latest advancements in the field of AMPK research, a non-classical pathway of AMPK activation has come to light, termed the lysosomal pathway. Recent studies have revealed that neither an elevation in AMP levels nor glucose deprivation is observable under glucose-free cell culture conditions or in animals experiencing low blood sugar due to starvation. This compellingly suggests the existence of an unexplored pathway within the body, capable of independently sensing glucose levels apart from AMP. This pathway, coined the “Lin pathway”, was unveiled by a research team led by Academician Lin Shengcai. Initially, Professor Lin’s research group embarked on investigating the novel physiological roles of Axin, initially recognized as a constituent of the Wnt signaling pathway, and its deficiency’s association with developmental irregularities. As their research progressed, the ensuing experimental outcomes yielded even more astonishing insights. Building upon prior experiments demonstrating the essentiality of N-myristoylation modification on the β1 and β2 subunits of AMPK for its activation through glucose deprivation, Lin Shengcai’s team formulated a hypothesis that AMPK might necessitate pre-association with lipid-containing membrane constituents under physiological conditions, thereby priming it for activation upon AMP engagement. In subsequent experiments, the research team employed yeast two-hybrid technology, ultimately discovering an interacting protein named p18/LAMTOR1, which interacts with the AXIN protein on the lysosomal membrane. This breakthrough adds a new layer of understanding to AMPK activation and its intricate regulatory mechanisms, unveiling the previously unexplored lysosomal pathway as an essential contributor to glucose-sensing mechanisms in the body.25 These findings establish a significant link between AMPK activation and the lysosome, unveiling the pivotal regulatory factor within this pathway: the crucial glycolytic enzyme, aldolase. As glucose becomes scarce, the substrate of aldolase, fructose-1,6-bisphosphate (FBP), experiences a rapid reduction. During this phase, aldolase becomes unbound from its substrate, facilitating dynamic interactions among aldolase, v-ATPase, Ragulator, and the AMPK-AXIN-LKB1 complex. Consequently, this cascade triggers the activation of the AMPK pathway, thereby contributing to the intricate regulation of cellular metabolism.25,33,34 The “anchor” of AMPK within lysosomes constitutes a protein complex that orchestrates both catabolic and anabolic metabolic processes, thereby illuminating the mechanisms governing the transition between catabolic and anabolic metabolism.33

The two pathways of AMPK activation mentioned above are mutually complementary (Figure 1B), functioning independently yet synergistically reinforcing each other. Together, they play a collaborative role in the intricate regulation of AMPK activity.

Downstream Pathways of AMPK

The downstream substrate proteins within the AMPK pathway exhibit significant diversity (Figure 1B), with almost 60 recognized substrates governed by AMPK. AMPK has the capability to phosphorylate and activate cytoplasmic acetyl-CoA carboxylase (ACC1), thus impeding fatty acid synthesis. Concurrently, AMPK governs ACC2 within mitochondria to enhance the oxidative degradation of fatty acids.35,36 AMPK directly activates ULK1 and Beclin-1 proteins, responsible for autophagy initiation,37,38 and by regulating the activity of upstream key proteins on the mTORC1 pathway, it activates the autophagy pathway.39 Furthermore, AMPK reacts to declining glucose levels by assembling a supercomplex on the lysosomal membrane, facilitating the separation of mTORC1, Ragulator, and v-ATPase, leading to the activation of the autophagy pathway. AMPK also directly modulates the mitochondrial fission protein MFF, promoting mitochondrial autophagy.40 Both cellular autophagy and mitochondrial autophagy are crucial aspects in the context of ulcerative colitis. Additionally, it has been observed that AMPK localizes within the nucleus and establishes direct interactions with TET2.41

AMPK-Related Technologies

UC is characterized by disruptions in glucose, energy, and amino acid metabolism. The functions of AMPK, such as inhibiting fatty acid synthesis and promoting fatty acid breakdown, hold the potential to restore the metabolic equilibrium in ulcerative colitis.On October 10, 2022, Academician Lin Shengcai from Xiamen University’s School of Life Sciences, in collaboration with Professor Deng Xianming, published a research paper titled “The aldolase inhibitor aldometanib mimics glucose starvation to activate lysosomal AMPK” in the journal Nature Metabolism. This study integrates fasting, calorie restriction, and other physiological conditions based on a novel AMPK activation pathway developed by Academician Lin Shengcai’s research team in recent years. Their work led to the creation of an AMPK activator named “Aldometanib”, which diverges from the conventional approaches pursued by many pharmaceutical companies. In Chinese, Aldometanib is translated as “Bìgǔjīng”, signifying “Fasting Essence”.42 By formulating AMPK activators based on the “Lin pathway”, a prevalent mode of AMPK activation during glucose-deprived circumstances, the potential arises to replicate physiological conditions where AMPK is mobilized, eliciting an array of advantageous effects. This innovative approach tackles challenges that conventional methods fail to address. In essence, the “Fasting Essence” operates as a mediator, targeting aldolase within the Lin pathway, which functions as a glucose sensor. As a result, the undesirable effects commonly linked to traditional AMPK activation are circumvented. This approach engenders AMPK activation in a manner that aligns with nature’s principles, mimicking a “natural” process that upholds the organism’s health and longevity.

AMPK Gets Involved in UC

Programmed Cell Death

Autophagy

Human clinical studies as well as in vitro and in vivo models have consistently underscored the pivotal role of autophagy in upholding intestinal homeostasis, governing gut microbiota, coordinating precise intestinal immune reactions, and furnishing antimicrobial defense. The derangement of autophagy can culminate in the disturbance of intestinal epithelial function, imbalanced gut microbiota, compromised secretion of antimicrobial peptides by Paneth cells, endoplasmic reticulum stress responses, and atypical immune reactions to pathogenic bacteria.7 The interplay between autophagy and endoplasmic reticulum (ER) stress is broadly acknowledged as a plausible mechanism contributing to the compromise of the epithelial barrier in IBD. Intestinal epithelial cells afflicted with autophagy impairments exhibit heightened vulnerability to damage.43,44 Autophagy deficiency emerges as a potential pivotal factor driving cellular paracellular permeability in intestinal disorders.45 Research on AMPK regulation of autophagy for treating UC has become highly advanced. One classic example is the “yin-yang” relationship with downstream mTOR, such as the use of resveratrol A1 to alleviate DSS-induced ulcerative colitis by modulating autophagy mediated by the AMPK/mTOR/p70S6K pathway.46 Berberine promotes autophagy by regulating the AMPK-mTOR signaling pathway, thereby reducing intestinal damage caused by UC.11 Foerster et al found that AMPK positively regulates autophagy initiation by activating the ULK1 complex and inhibiting mTORC1. This leads to increased availability of nutrients through the catabolic breakdown activity of autophagy.13 Sup et al uncovered that estrogen-related receptor alpha (ESRRA) exhibits significant expression in intestinal tissue and plays a pivotal role in maintaining intestinal homeostasis. The absence of ESRRA results in compromised AMPK phosphorylation, diminished transcription factor EB (TFEB) expression, escalated inflammation, compromised mitochondrial function within intestinal epithelial cells, and notably, anomalies in genes associated with autophagy.47 Studies have found that heat shock protein 90 (HSP90) and NLRP3 inflammasome are both associated with AMPK-mediated autophagy. Additionally, HSP90 and NLRP3 are inflammatory markers in UC, indicating the therapeutic potential of AMPK.48 SIRT1 deacetylates proteins, a reaction that requires NAD+ as a co-factor. Activation of AMPK increases cellular NAD+ levels, thereby enhancing sirtuin activity. SIRT1 increases p-AMPK levels through the deacetylation of LKB-1. Both SIRT1 and p-AMPK negatively regulate mTOR signaling to induce autophagy, thereby alleviating colitis.49 Metrnl, also recognized as Cometin or Subfatin, emerges as a recently identified secreted protein with abundant expression in the intestinal epithelium. Investigations have unveiled that Metrnl deficiency intensifies ulcerative colitis, at least in part, by impeding autophagy via the AMPK-mTOR-p70S6K pathway.50 Triggering AMPK-ULK1-mediated autophagy could potentially enhance the management of DSS-induced colitis by facilitating P62-mediated mitophagy to suppress the formation of the NLRP3 inflammasome.51 Subsequent investigations have suggested that the Alpha7 nicotinic acetylcholine receptor mitigates inflammatory bowel disease by inducing autophagy through the AMPK-mTOR-p70S6K pathway.52 AMPK and mTOR are canonical pathways implicated in autophagy. Research has revealed that the activation of the AMPK/mTOR pathway can mitigate Th17/Treg imbalance, subsequently ameliorating DSS-induced colitis—a characteristic immune dysregulation often observed in UC.53,54 It is evident that the role of AMPK in UC extends beyond its impact on cellular autophagy for improving colitis. AMPK is implicated in various other mechanisms as well. However, further research is needed to elucidate the precise mechanisms underlying the beneficial effects of AMPK activation on autophagy and UC.

Pyroptosis

Cellular pyroptosis is a type of cell death characterized by cellular swelling and the formation of prominent bubbles on the cell membrane.55 Research has demonstrated that mitigating cellular pyroptosis can attenuate experimental colitis. Schisandrin B, a compound derived from Schisandra chinensis, mitigates epithelial cell damage in colitis by modulating the AMPK/Nrf2/NLRP3 inflammasome-mediated cellular pyroptosis pathway.10 Deng et al revealed that the Gly-Pro-Ala (GPA) peptide can mitigate cellular pyroptosis and improve DSS-induced colitis in mice. This effect is achieved by inhibiting the assembly of the NLRP3 inflammasome and cleavage of GSDMD through the AMPK pathway.56 Evidence suggests that resveratrol has the capacity to reduce the expression of inflammation-induced miR-155, a key regulator of innate immune responses in macrophages, dendritic cells, and epithelial cells. In the context of UC, miR-155 holds prominence due to its heightened expression. Consequently, the mitigation of UC through resveratrol’s downregulation of miR-155 becomes evident. Recent research highlights that the disruption of the AMPK and Sirt1 positive feedback loop effectively counteracts the inhibitory impact of resveratrol on miR-155 expression. This disruption leads to heightened miR-155 expression and instigates cellular pyroptosis, driven by NLRP3 inflammasome activation.57 Therefore, it can be inferred that activation of AMPK-related pathways can alleviate cellular pyroptosis and ameliorate UC.58

Inflammatory Response

UC initiates a cascade of inflammatory responses, characterized by the overexpression of pro-inflammatory cytokines including TNF-α, IFN-γ, and IL-1β. These cytokines contribute to a sequential escalation of inflammation, culminating in intestinal tissue damage and the formation of ulcerative lesions.59 Increasing evidence suggests that AMPK plays a crucial role in UC pathogenesis. AMPK can regulate the signaling pathways of NF-κB and MAPK, as well as the expression of inflammatory genes, thereby exerting anti-inflammatory effects.60 NLRP3 facilitates the intricate assembly of the inflammasome complex in response to microbial ligands, triggering the activation of caspase-1 and subsequent secretion of IL-1β and IL-18. Both IL-1β and IL-18 play pivotal roles in the pathogenesis of IBD.61 AMPK has the ability to enhance the expression of SIRT1 by mitigating the inhibitory impact of DBC1 on SIRT1 activity. Consequently, SIRT1 works to diminish the activity of transcription factors through deacetylating NF-κB, AP-1, and histones. This process helps reinstate a condensed chromatin structure, ultimately restraining the transcription of genes related to inflammation.62 Studies have revealed that activation of the AMPK/SIRT1 pathway can inhibit the formation of NLRP3 inflammasomes in macrophages, thereby alleviating inflammatory responses in colitis.63 Activation of AMPK exhibits the capacity to impede the activation of the IKK/NF-κB pathway provoked by TNF-α, as well as the JAK/STAT3 pathway prompted by IL-6. Furthermore, it can suppress NADPH oxidase activity, thus mitigating intestinal inflammatory responses.63 Hydroxycinnamic acid fosters the expression of PPARγ-responsive genes CD36 and LPL, triggers the translocation of PPARγ from the cytoplasm to the nucleus, and facilitates the association of PPARγ with reporter genes. This activation of PPARγ consequently induces the AMPK/ACC1-mediated shift from Th17 to Treg cells, thereby restoring the balance between Th17 and Treg cells and effectively mitigating intestinal inflammatory responses in cases of colitis.64 The knockout of P2Y1R, as one of the G protein-coupled receptors, can inhibit Th17 differentiation and alleviate intestinal inflammation through an AMPK-dependent mechanism.65 Activation of AMPK diminishes the activation of p38 MAPK, subsequently curtailing the expression of IL-6 and effectively mitigating pathological inflammation in the intestinal region.66 Based on the above information, it is evident that AMPK plays a crucial role in suppressing inflammatory responses in intestinal tissues.

Intestinal Epithelial Barrier

A reduction in the phosphorylation of AMPKαThr172 has been noted in the intestines of individuals with IBD, evident in both the epithelial and lamina propria compartments. This observation underscores the plausible correlation between AMPK and the functionality of the intestinal barrier.66 The caudal type homeobox 2 (CDX2) serves as a pivotal transcription factor responsible for committing cells to the intestinal epithelial lineage. Deficiency of CDX2 negates the intestinal differentiation facilitated by AMPK activation. In vivo studies reveal that epithelial AMPK knockout results in diminished CDX2 expression, thereby compromising the integrity of the intestinal barrier, including tight junction integrity and ultrastructure. This situation further contributes to epithelial cell migration, intensifies intestinal proliferation, and worsens colitis induced by dextran sulfate sodium.67 Metformin increases the expression of tight junction proteins ZO-1, Claudin-1, and Occludin in the intestinal epithelium of mice treated with DSS by activating AMPK.68 Astragaloside IV restores intestinal mucosal barrier by promoting AMPK activation and driving the expression of Muc-2.69 The compromised integrity of the intestinal barrier results in heightened intestinal permeability, manifesting as increased serum recovery of FD-4 and elevated expression of MLCK protein. Geniposide intervenes by reducing the expression of MLCK protein, thus mitigating LPS-induced in vitro barrier dysfunction through the activation of AMPK phosphorylation.70 Sinensetin facilitates epithelial autophagy through the activation of the AMPK/ULK1 pathway. Notably, the promotion of epithelial autophagy has been shown to counteract epithelial cell apoptosis while enhancing the degradation of Claudin-2. Given that Claudin-2 expression correlates with epithelial permeability, the notion that AMPK safeguards intestinal epithelial barrier integrity by facilitating Claudin-2 degradation is substantiated.71 Based on this information, it can be concluded that the activation of AMPK is essential for the proper functioning of the intestinal epithelial barrier.

Macrophage Polarization

The AMPK signaling pathway is one of the classical pathways involved in macrophage polarization.72 In typical conditions, intestinal macrophages are crucial for preserving intestinal integrity and averting inflammation. Nonetheless, genetic and environmental factors can disrupt the equilibrium between M1 and M2 macrophage polarization within the intestines. This disruption can convert regular inflammatory responses into pathological intestinal harm. In the case of ulcerative colitis (UC) patients, the disturbance of intestinal inflammation is intricately connected to an uneven distribution of M1 and M2 macrophage polarization. Consequently, rectifying the M1/M2 macrophage polarization balance could hold substantial therapeutic potential for addressing UC.14 Evidence suggests that the traditional Chinese medicine formula “Xianhecao-Changyan Fang” effectively diminishes the expression of M1 macrophage markers (CD11c, IL-6, and IL-1β) and enhances the expression of M2 macrophage markers (CD206) in colitis-afflicted mice. This observation implies that the formula might modulate macrophage polarization, possibly by means of AMPK-mediated metabolic reprogramming. Consequently, this could enhance the anti-inflammatory activity of macrophages.73 Natural flavonoids have demonstrated a substantial capacity to ameliorate symptoms and pathological alterations in colitis-affected mice. Remarkably, when mice were treated with compound C, a pharmacological inhibitor of AMPK, the therapeutic efficacy of Eup was noticeably diminished. Furthermore, Eup exhibited a noteworthy ability to effectively restrain the inflammatory response induced by LPS in macrophages.74 These findings suggest that AMPK is involved in the inflammatory response of macrophages in UC. Quercetin, a significant constituent within traditional Chinese medicine formulations aimed at addressing ulcerative colitis (UC), holds noteworthy implications. Studies indicate that quercetin possesses anti-inflammatory properties by suppressing M1 macrophage polarization while concurrently bolstering M2 macrophage polarization and endogenous antioxidant expression within both macrophages and microglial cells. This elevation of M2 markers and inherent antioxidants is attributed to the activation of AMP-activated protein kinase (AMPK) and Akt signaling pathways, orchestrated by quercetin.75 As per reports, the activation of AMPK through phosphorylation can facilitate the expression and translocation of Nrf2 into the nucleus. Consequently, this prompts the upregulation of downstream gene HO-1. This intricate mechanism contributes to the modulation of M1 to M2 macrophage polarization in cases of colitis.76 Numerous studies have demonstrated that AMPK mediates macrophage polarization through the regulation of the NF-κB pathway.77 Transgenic mice overexpressing SIRT1 in macrophages exhibit enhanced polarization towards M2 macrophages. Activated SIRT1 suppresses LPS-induced NF-κB activity by inhibiting p65 acetylation and the release of M1-related cytokines. SIRT1 also forms a positive feedback regulation with AMPK, further promoting the shift from M1 to M2 polarization.8,78 XHCF, a traditional Chinese medicine formula documented in the Chinese Pharmacopoeia (2015 edition), has been shown to inhibit M1 macrophage polarization and promote M2 macrophage polarization to ameliorate DSS-induced colitis in mice. This process is likely mediated by the activation of AMPK phosphorylation.73 Activation of AMPK diminishes the degradation of IκB, enhances Akt activation, and suppresses the NF-κB pathway. These effects contribute to the transition of M1 macrophages to the M2 phenotype, consequently alleviating colitis.79 Thus, it can be inferred that both macrophage polarization and macrophage inflammatory status in UC can be modulated by AMPK.

The aforementioned involvement of AMPK in various pathophysiological mechanisms of UC is reviewed in Figure 2.

Figure 2 Possible Biological Mechanisms of AMPK in Ulcerative Colitis.

Potential Drugs Targeting AMPK-Regulated Pathway in UC

Potential Drug Inclusion Methods

There are three criteria for potential drugs to be selected: (1) The potential pharmaceutical agent demonstrates a conspicuous therapeutic impact on UC, ameliorating associated phenotypes. (2) A substantial volume of in vivo or in vitro experimentation substantiates that the potential pharmaceutical agent can directly modulate AMPK molecules. (3) The regulatory influence of this potential pharmaceutical agent on AMPK can elicit alterations in downstream pathways or biological processes. (4) The implicated AMPK-related pathways and biological processes are closely associated with UC.

TCM Monomer

Monomers of traditional Chinese medicine refer to purified substances extracted and isolated from herbal medicines, which are purer compared to compound herbal medicines. This high purity facilitates the accurate determination of their chemical constituents and pharmacological effects, making it conducive to the study of their therapeutic efficacy and safety. The following several monomers of traditional Chinese medicine exert a protective effect in colitis by activating AMPK and promoting its phosphorylation, ultimately repairing the intestinal epithelial barrier function. Mogrol, an aglycone of mogrosides, which protects intestinal epithelial cells by promoting AMPK activation and inhibiting the formation of NLRP3 inflammasome.12 Phellodendrine, a component of phellodendrone, potentially exerts a protective effect on the intestinal epithelial cells in ulcerative colitis (UC) by activating the AMPK-mTOR signaling pathway to promote autophagy.11 Schisandrin B, ingredient in Schisandra chinensis, regulates pyroptosis through the AMPK/Nrf2/NLRP3 inflammasome pathway, alleviating epithelial cell damage in colitis.10 Kai Zhou et al found that Eupatilin significantly suppressed NFκB activation and ROS production in activated macrophages, and fortified epithelial tight junctions (TJs) via an AMPK-dependent mechanism.74 Andrographolide (Andro) is a naturally occurring compound isolated from Andrographis paniculata (Burm. f.) Nees, which has a long history of traditional use in China, India, and other parts of Asia for managing inflammation-related ailments. In various studies, Andro has demonstrated its ability to suppress inflammatory responses in lipopolysaccharide (LPS)-stimulated macrophages and alleviate murine acute colitis by activating AMPK.80 Heterophyllin B exhibited significant protection against DSS-damaged intestinal epithelium physical barrier to limit access of enteric microbes through promoting AMPK activation.69 Wedelolactone treatment effectively alleviated DSS-induced mouse colitis by inhibiting NLRP3-mediated inflammatory responses and restoring epithelial barrier function via AMPK activation.81 Sinensetin acts by enhancing AMPK phosphorylation to promote autophagy in epithelial cells, thereby further inhibiting cellular apoptosis, facilitating the degradation of claudin-2, and significantly alleviating intestinal barrier dysfunction in patients with colitis.71 Picroside III can improve DSS-induced colitis in mice by promoting intestinal mucosal wound healing and restoration of epithelial barrier function through activation of AMPK.82 There are also several monomers that can alleviate UC by modulating intestinal mucosal immunity and mucosal inflammation through activation of the AMPK pathway. Atractylenolide III prevents mitochondrial dysfunction and improves the development of colitis by activating the AMPK/SIRT1/PGC-1α signaling pathway.83 Cinnamtannin D1 prevents Th17/Treg imbalance and improves DSS-induced colitis by activating the AMPK/mTOR pathway.53 Geniposide inhibits NLRP3 inflammasome in macrophages through AMPK/SIRT1-dependent signaling, leading to improvement of inflammation response in colitis.63 Additionally, studies have shown that Geniposide can reduce inflammation and regulate the disrupted epithelial barrier function by activating the AMPK signaling pathway, thereby ameliorating TNBS-induced experimental colitis in rats.70 Forskolin inhibits the inflammatory response in LPS-stimulated macrophages and acute colitis in mice by activating AMPKα2.80 Ginsenoside Rd inhibits sodium dextran sulfate-induced experimental colitis in mice by promoting AMPK-ULK1-p62 axis-driven mitophagy-mediated inactivation of NLRP3 inflammasomes, resulting in reduced secretion of IL-1β in macrophages.51 Madecassic acid restores Th1/Treg balance via the PPARγ/AMPK/ACC17 pathway, exerting anti-colitis effects.64 Dipotassium glycyrrhizate via HMGB1 or AMPK signaling suppresses oxidative stress during intestinal inflammation.84 Platycodin D activates AMPK, which in turn activates the PI3K/Akt pathway and inhibits the NF-κB pathway, thereby participating in macrophage polarization and suppressing intestinal inflammation associated with colitis.79 Xiaofan Xu et al found that BBR (berberine) can promote autophagy through the AMPK/MTOR pathway, leading to the inhibition of lysozyme expression and secretion. In contrast to the Unc-51-like kinase 1 (ULK1) pathway, BBR promotes lysosome maturation and expression. Therefore, BBR has the potential to be a candidate drug for the treatment of UC (ulcerative colitis) patients in the future.85 The aforementioned studies indicate that the activation of AMPK is indispensable for the efficacy of monotherapy with traditional Chinese medicine monomer in the treatment of UC.

Traditional Chinese Prescription

Traditional Chinese prescription, composed of multiple Chinese medicinal herbs, is a primary application in clinical practice. Numerous studies have shown through experimental research that Chinese herbal formulations have a multi-target comprehensive effect in treating UC (ulcerative colitis), and many of them have been successfully applied in clinical settings with favorable outcomes. Traditional Chinese medicine prescriptions function by suppressing the release of inflammatory factors and regulating intestinal immune response.86 Baitouweng decoction (BD), consisting of four traditional Chinese medicines - Pulsatilla chinensis (Bunge) Regel (Bai Tou Weng), Phellodendron chinense C.K.Schneid. (Huang Bo), Coptis chinensis Franch. (Huang Lian), and Cortex Fraxini (Qin pi), has a long-standing history in the treatment of intestinal diseases, including UC. The research findings indicate that BD promotes autophagy and facilitates the restoration of the intestinal epithelial barrier in dextran sulfate sodium (DSS)-induced colitis mice by activating AMPK phosphorylation and suppressing mTOR expression. Enrichment analysis results provide evidence that BD can modulate the autophagy, AMPK, and mTOR signaling pathways.87 The Xian-He-Cao-Chang-Yan formula (XHCF) consists of six crude drugs: Agrimoniae Herba (AH, the aerial part of Agrimonia pilosa Ledeb.), Coptidis Rhizoma (CR, the rhizome of Coptis chinensis Franch.), Aucklandiae Radix (AR, the root of Aucklandia lappa Decne.), Cicadae Periostracum (CP, the exuviae of Cryptotympana pustulata Fabricius.), Acori Tatarinowii Rhizoma (ATR, the rhizome of Acorus tatarinowii Schott.), and Platycodonis Radix (PR, the root of Platycodon grandiflorum (Jacq) A.DC.). These ingredients are combined in a ratio of 5:1.5:1.5:1.5:1.5:1. XHCF has the potential to enhance the activation of AMPK, which consequently regulates macrophage polarization. The therapeutic benefits of XHCF on UC may be attributed to the synergistic actions of its multiple ingredients.73 Huangqin decoction (HQD) is a traditional Chinese medicine (TCM) formula derived from Shang Han Lun. It consists of a combination of four herbs: the root of Scutellaria baicalensis Georgi. (skullcap), Glycyrrhiza uralensis Fisch. (liquorice), Paeonia lactiflora Pall. (peony), and the fruit of Ziziphus jujuba Mill. (jujube). HQD ameliorated UC by regulating fatty acid metabolism to mediate macrophage polarization via activating FFAR4-AMPK-PPARα pathway.8 Sishen Pill (SSP) is composed of Euodia rutaecarpa (Juss.) Benth., Schisandra chinensis (Turcz.) Baill, Psoralea corylifolia L., Myristica fragrans Houtt., Ziziphus jujuba Mill. and Zingiber officinale Rosc, which has been treating chronic enteritis in China for thousands of years. The research findings indicate that SSP effectively regulates peripheral blood effector memory T cells, thereby alleviating DSS-induced experimental colitis. This phenomenon may be associated with the inhibition of the PI3K/Akt signaling pathway and the activation of AMPK.88

Traditional Chinese prescriptions exhibit a multi-target comprehensive effect in the treatment of UC, and most of them have been clinically utilized with favorable outcomes. However, the specific active components of these prescriptions are intricate, necessitating a more scientific theoretical framework to elucidate their therapeutic principles.86

Diet

Dietary factors can be directly associated with the pathogenesis or progression of UC through their impact on the host or indirectly through the modulation of the composition or function of the gut microbiota.89 The administration of nanoparticles derived from human-consumed broccoli exhibits a suppressive effect on murine colitis through the activation of AMPK in dendritic cells.90 The activity of AMPK is notably augmented in mice treated with DSS and supplemented with dietary red raspberries, leading to a subsequent decrease in intestinal mucosal and epithelial damage.91 The potential application of chitosan oligosaccharide as a dietary supplement or nutritional health product has been investigated. It activates AMPK in intestinal epithelial cells to improve gastrointestinal disorders.92 Creatine levels are maintained through dietary intake and endogenous synthesis of arginine and glycine. The preservation of creatine maintains intestinal homeostasis and helps prevent colitis, partially relying on the AMPK pathway. However, further research is needed to elucidate the precise connection between creatine and AMPK signaling in the context of colitis.93 Plant-based foods rich in anthocyanins and anthocyanin formulations play a crucial role in the prevention of IBD and its associated colorectal cancer (CRC). The AMPK cascade serves as the primary pathway through which anthocyanins exert their protective effects against colitis.94 Le Carbone, a dietary supplement (a charcoal supplement), regulates DSS-induced acute colitis in mice by activating AMPKα and downregulating the STAT3 and cystathionine gamma-lyase-dependent apoptotic pathways.95 Due to its high nutritional value, jujube fruit is highly regarded. Research has found that polysaccharides extracted from wild jujube enhance intestinal barrier function through the AMPK pathway, thereby preventing experimental inflammatory bowel disease.96

These findings will contribute to the formulation of increasingly health-conscious dietary recommendations, specifically targeting the prevention of colitis, while simultaneously mitigating potential negative reactions that may arise from the influence of AMPK on metabolic energy.

Compounds Found in Plants

The chemical constituents extracted from numerous plants exhibit therapeutic effects on UC. Proanthocyanidins are phenolic compounds extracted from various plants. Proanthocyanidin A1 (PCA1) is a type of proanthocyanidin. Studies have revealed that PCA1 alleviates colitis damage by inhibiting the mTOR and p70S6K signaling pathways through the activation of AMPK in DSS-induced UC mice.46 Alginate is an acidic polysaccharide extracted from marine brown algae. Alginate can be degraded into alginate oligosaccharides (AOS) with lower molecular weight, lower viscosity, improved solubility, and enhanced bioavailability. AOS inhibits LPS-mediated inflammatory responses and alleviates DSS-induced colitis through the activation of AMPK signaling and suppression of NF-κB activation.97 Dioscin, which is a form of plant steroidal saponin abundant in some medical plants, alleviates DSS-induced colitis by inhibiting the mTOR pathway through the promotion of AMPK phosphorylation.98 As the precursor of the phytochemical sulforaphane (SFN), glucoraphanin (GRP) is highly concentrated in cruciferous vegetables such as bok choy, broccoli, and cabbage. GRP exhibits its therapeutic effects by activating the AMPK/PGC1α/NRF2 pathway, thus ameliorating dextran-sulphate-sodium (DSS)-induced colitis in mice.99 Resveratrol (RES), a natural polyphenol, is found in several plants and fruits such as grapes, blueberries, and peanuts. Its microbial metabolite, 4HPP, is involved in the potential mechanism of maintaining intestinal barrier function. This mechanism includes the regulation of TJ expression through AMPK-activated CDX2 expression and the inhibition of pro-inflammatory cytokines through the AMPK-mediated SIRT1/NF-κB pathway.100 Platycodon grandiflorum (PG), a perennial herb of the Campanulaceae family, exerts anti-inflammatory effects in a murine IBD model through the AMPK/NF-κB/NLRP3 pathway upon administration of PG root fermentation broth (PGRFB).9

Anti-Diabetic Compounds

AMPK, being a pivotal molecule in energy metabolism, plays a critical role as a target for anti-diabetic medications. Research findings have demonstrated that specific anti-diabetic drugs possess the ability to partially ameliorate UC. To date, metformin has been demonstrated to possess multiple effects beyond its anti-diabetic properties. It can promote tight junction expression, enhance mucosal integrity, and regulate mucosal immunity to alleviate inflammatory responses through both AMPK-dependent and AMPK-independent mechanisms. Importantly, metformin exhibits greater efficacy in alleviating DSS-induced colitis compared to mesalazine, rendering it more appealing for the treatment of UC.66,68,101–105 Dapagliflozin has emerged as a promising selective SGLT2 inhibitor for the treatment of type 2 diabetes, offering a reduced risk of hypoglycemia. It exerts its effects by targeting key pathways including AMPK/mTOR, HMGB1/RAGE, and Nrf2/HO-1, which activate autophagy and inhibit apoptosis. This mechanism of action has shown potential in alleviating symptoms associated with experimental IBD in rats.106 Linagliptin, which is a potent inhibitor of dipeptidyl peptidase-IV (D-IV) and used for the treatment of type 2 diabetes mellitus (T2DM), has been found to improve acetic acid-induced colitis in rats by modulating the AMPK/SIRT1/PGC-1α and JAK2/STAT3 signaling pathways.107

AMPK Activator

Several studies have demonstrated the therapeutic effects of various AMPK activators in colitis, as they directly activate AMPK to exert their functions. In a study conducted by Suhrid Banskota et al, it was discovered that the administration of VAS2870, a pan-Nox inhibitor, effectively alleviated colitis induced by dextran sodium sulfate (DSS) in mice with macrophage-specific AMPKβ1 deficiency (AMPKβ1LysM).108 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR), which is an intermediate in the production of monophosphoryl adenosine, acts as an activator of AMPK. By modulating intestinal mucosal immunity, it can exert a protective effect on the intestinal mucosa of experimental colitis mice.109,110 Studies have demonstrated that the small molecule GL-V9 (5-hydroxy-8-methoxy-2-phenyl-7-(4-(pyrrolidin-1-yl)butoxy)4 H-chromen-4-one), an AMPK activator, exhibits a protective effect against DSS-induced colitis by attenuating oxidative stress. This effect is achieved through the up-regulation of (Thioredoxin-1) Trx-1 via the activation of the AMPK/FOXO3 pathway. Moreover, additional investigations have revealed that GL-V9 induces autophagy in macrophages by activating AMPK and also leads to the degradation of NLRP3 inflammasome through autophagosome activation.111 The research conducted by Luca Antonioli et al indicates that the novel AMPK activator FA5 can serve as an innovative pharmaceutical agent for clinical treatment of intestinal inflammation.112

Others

Certain specific drugs, such as bioactive substances, can also exert their effects in UC by activating AMPK. GPA peptides isolated from fish skin collagen hydrolysate can improve DSS-induced colitis by specifically inhibiting NLRP3 inflammasome-mediated pyroptosis. Additionally, GPA peptides can increase AMPK phosphorylation to suppress ROS production and impede NLRP3 mitochondrial localization, thereby inhibiting NLRP3 inflammasome activation.56 BJ-3105, a 6-alkoxy-pyridin-3-ol derivative, has previously been shown to inhibit T cell differentiation and autoimmune encephalomyelitis by blocking cytokine-induced JAK phosphorylation.113 Pallavi Gurung et al discovered that BJ-3105 improves DSS-induced colitis and azoxymethane/DSS-induced colitis-associated tumorigenesis in mice through the AMPK-NOX axis.114 The P2Y1 receptor, a G protein-coupled receptor, plays a crucial role in the immune response of inflammatory bowel disease. Studies have shown that the absence of P2Y1R can inhibit Th17 cell differentiation in an AMPK-dependent manner, leading to improvements in colitis. Therefore, P2Y1R can serve as an important modulator of Th17 cell differentiation to control intestinal inflammation.65 Coenzyme Q10 (CoQ10), also known as ubiquinone, is a lipophilic antioxidant. CoQ10 treatment has been shown to increase the expression of p-AMPK and FOXP3 while inhibiting the expression of p-STAT3 and IL-17, thereby reducing the severity of colitis. These findings suggest that CoQ10 holds potential as a targeted therapeutic approach for treating UC.115 A novel bioactive mitochondrial-derived peptide encoded in mitochondrial 12S rRNA, referred to as MOTS-c, has been shown to alleviate inflammatory responses, enhance anti-apoptotic capacity, upregulate p-AMPK expression, and inhibit the activation of the ERK/JNK signaling pathway, thereby exerting anti-colitis effects.116 Cannabinoid receptor 2 (CB2R) belongs to the G-protein-coupled receptor (GPCR) family and possesses 7 transmembrane α-helices, a glycosylated extracellular N-terminus, and an intracellular carboxyl terminus. Activation of CB2R has been demonstrated to ameliorate DSS-induced colitis by inhibiting the NLRP3 inflammasome in macrophages.117

The detailed information for the aforementioned drugs involved in the activation of AMPK for the treatment of UC can be found in Table 1.

Table 1 AMPK-Targeting Drugs for UC Treatment

Discussion

AMPK is responsible for maintaining the equilibrium between energy production and expenditure through the regulation of a multitude of metabolic activities. Among its targets are pivotal enzymes and transcriptional regulatory factors that play roles in cell growth, proliferation, and metabolic pathways. Consequently, disruptions in AMPK function are intricately linked to metabolic disorders, including diabetes, obesity, and the onset of cancer. Nonetheless, UC does not conform to the conventional framework of metabolic disorders, which might introduce certain biases when exploring AMPK as a mechanism and potential therapeutic target for UC. Nevertheless, I hold the belief that organisms are the remarkable outcomes of evolution, endowed with intricate mechanisms that surpass our imagination in tackling diverse challenges. Moreover, a multitude of studies have underscored the dysregulation of AMPK in UC, and the activation of AMPK through diverse pharmaceutical agents or biological processes has demonstrated substantial mitigation of colitis. However, further research is imperative to delve into the intricate biological underpinnings associated with the pathogenesis and treatment of UC.119

The intricate integration of diverse upstream and downstream pathways at the cellular level by AMPK remains an enigma within the realm of AMPK research. AMPK is found across various intracellular locales, including the lysosomal membrane, cytosol, mitochondria, and the cell nucleus. The manner in which AMPK orchestrates and governs these distinct compartments, as well as the potential existence of distinct downstream regulatory genes, remains shrouded in uncertainty. The predominant focus of AMPK research in the context of UC pertains to its involvement in cellular autophagy. While grasping the significance of AMPK in regulating autophagy offers insights into its mechanism, present conclusions are circumscribed. There exists a dearth of studies that bridge the crucial role of AMPK in autophagy regulation with clinical applications to benefit UC patients. The translation of foundational research into tangible clinical interventions stands as a pressing matter that requires urgent attention.

Recent research has unveiled the advantageous therapeutic outcomes associated with AMPK activators across a range of conditions, including autosomal dominant polycystic kidney disease, inflammatory bowel disease, muscular atrophy, neuronal loss, and X-linked adrenoleukodystrophy. Notably, compounds known for their anti-aging properties, such as dasatinib and quercetin, which extend the lifespan of mice, also function as AMPK activators. It is paramount to recognize, however, that prolonged AMPK activation could potentially entail detrimental consequences, including the risk of cardiac and renal hypertrophy, along with the potential association with Alzheimer’s disease.120 Hence, a targeted and transient approach to AMPK activation, akin to the effects of regular exercise, could prove pivotal in the management of ulcerative colitis and other closely linked disorders.

Conclusion and Perspectives

Disruption of intestinal mucosal barrier function, heightened mucosal permeability, immune-inflammatory infiltration within the mucosa, imbalanced Th17/Treg ratio, skewed macrophage polarization, inflammatory conditions, mitochondrial impairment, mitophagy, cellular apoptosis, autophagy, and necrosis collectively contribute to the progression, recurrence, chronicity, and potential carcinogenesis of ulcerative colitis. Taking into account the aforementioned examination, AMPK is implicated to varying extents in these pathological pathways, assuming a significant yet enigmatic role. Research on the involvement of AMPK in macrophage polarization and cell necrosis in ulcerative colitis remains relatively sparse. In my view, forthcoming investigations should allocate greater attention to exploring these two dimensions. The foundational principles underpinning the majority of these mechanisms continue to center on safeguarding the adenine nucleotide reservoir and, in a broader context, upholding energy equilibrium. Considering drug development, the diverse nature of AMPK has given rise to concerns regarding potential unintended outcomes when targeting it. While potent pan-β activators have been linked to cardiac hypertrophy in non-human primates,121 clinical investigations centered on type 2 diabetes and/or cardiovascular disease have indicated that judicious use of AMPK activators proves both safe and efficacious.

Undoubtedly, in the future, we need to examine and study the complex and profound relationship between energy, substance metabolism, and the coordinated, interactive, and regulatory aspects of biological stress responses with a broader perspective and comprehensive knowledge.

Acknowledgments

This research was supported by grants from the research cooperation project of Huazhong University of Science and Technology Union Jingshan Hospital (2023-XHJS-008).

Yuyi Yuan, Fang Wang and Xingxing Liu have contributed equally to this work and share the first authorship.

Author Contributions

All authors have made substantial contributions to the work presented in the report, whether in conceptualization, research design, execution, data acquisition, analysis and interpretation, or in all of these domains. They participated in drafting, revising, or critically reviewing the manuscript, ultimately approving the final published version. Consensus was reached on the submission of the manuscript to the journal, and all authors have agreed to take responsibility for various aspects of the work.

Disclosure

The authors declare no conflict of interest.

References

1. Feuerstein JD, Moss AC, Farraye FA. Ulcerative Colitis. Mayo Clin Proc. 2019;94(7):1357–1373. doi:10.1016/j.mayocp.2019.01.018

2. Klionsky DJ, Abdelmohsen K, Abe A, et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy. 2016;12(1):1–222. doi:10.1080/15548627.2015.1100356

3. Kobayashi T, Siegmund B, Le Berre C, et al. Ulcerative colitis. Nat Rev Dis Primers. 2020;6(1):74. doi:10.1038/s41572-020-0205-x

4. Kucharzik T, Koletzko S, Kannengiesser K, et al. Ulcerative Colitis-Diagnostic and Therapeutic Algorithms. Dtsch Arztebl Int. 2020;117(33–34):564–574. doi:10.3238/arztebl.2020.0564

5. Yan Y, Mukherjee S, Harikumar KG, et al. Structure of an AMPK complex in an inactive, ATP-bound state. Science. 2021;373(6553):413–419. doi:10.1126/science.abe7565

6. Ramos GP, Papadakis KA. Mechanisms of Disease: inflammatory Bowel Diseases. Mayo Clin Proc. 2019;94(1):155–165. doi:10.1016/j.mayocp.2018.09.013

7. Larabi A, Barnich N, Nguyen HTT. New insights into the interplay between autophagy, gut microbiota and inflammatory responses in IBD. Autophagy. 2020;16(1):38–51. doi:10.1080/15548627.2019.1635384

8. Li MY, Wu Y-Z, Qiu J-G, et al. Huangqin Decoction ameliorates ulcerative colitis by regulating fatty acid metabolism to mediate macrophage polarization via activating FFAR4-AMPK-PPARalpha pathway. J Ethnopharmacol. 2023;311:116430. doi:10.1016/j.jep.2023.116430

9. Wang Z, Li C, He X, et al. Platycodon grandiflorum root fermentation broth reduces inflammation in a mouse IBD model through the AMPK/NF-κB/NLRP3 pathway. Food Funct. 2022;13(7):3946–3956. doi:10.1039/D1FO03969E

10. Zhang W, Wang W, Shen C, et al. Network pharmacology for systematic understanding of Schisandrin B reduces the epithelial cells injury of colitis through regulating pyroptosis by AMPK/Nrf2/NLRP3 inflammasome. Aging. 2021;13(19):23193–23209. doi:10.18632/aging.203611

11. Su S, Wang X, Xi X, et al. Phellodendrine promotes autophagy by regulating the AMPK/mTOR pathway and treats ulcerative colitis. J Cell Mol Med. 2021;25(12):5707–5720. doi:10.1111/jcmm.16587

12. Liang H, Cheng R, Wang J, et al. Mogrol, an aglycone of mogrosides, attenuates ulcerative colitis by promoting AMPK activation. Phytomedicine. 2021;81:153427. doi:10.1016/j.phymed.2020.153427

13. Foerster EG, Mukherjee T, Cabral-Fernandes L, et al. How autophagy controls the intestinal epithelial barrier. Autophagy. 2022;18(1):86–103. doi:10.1080/15548627.2021.1909406

14. Yang Z, Lin S, Feng W, et al. A potential therapeutic target in traditional Chinese medicine for ulcerative colitis: macrophage polarization. Front Pharmacol. 2022;13:999179. doi:10.3389/fphar.2022.999179

15. Wang K, Mao T, Lu X, et al. A potential therapeutic approach for ulcerative colitis: targeted regulation of macrophage polarization through phytochemicals. Front Immunol. 2023;14:1155077. doi:10.3389/fimmu.2023.1155077

16. Carlson CA, Kim KH. Regulation of hepatic acetyl coenzyme A carboxylase by phosphorylation and dephosphorylation. J Biol Chem. 1973;248(1):378–380. doi:10.1016/S0021-9258(19)44486-4

17. Beg ZH, Allmann DW, Gibson DM. Modulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity with cAMP and with protein fractions of rat liver cytosol. Biochem Biophys Res Commun. 1973;54(4):1362–1369. doi:10.1016/0006-291X(73)91137-6

18. Yeh LA, Lee KH, Kim KH. Regulation of rat liver acetyl-CoA carboxylase. Regulation of phosphorylation and inactivation of acetyl-CoA carboxylase by the adenylate energy charge. J Biol Chem. 1980;255(6):2308–2314. doi:10.1016/S0021-9258(19)85891-X

19. Carling D, Zammit VA, Hardie DG. A common bicyclic protein kinase cascade inactivates the regulatory enzymes of fatty acid and cholesterol biosynthesis. FEBS Lett. 1987;223(2):217–222. doi:10.1016/0014-5793(87)80292-2

20. Hardie DG, Schaffer BE, Brunet A. AMPK: an Energy-Sensing Pathway with Multiple Inputs and Outputs. Trends Cell Biol. 2016;26(3):190–201. doi:10.1016/j.tcb.2015.10.013

21. Hawley SA, Davison M, Woods A, et al. Characterization of the AMP-activated protein kinase kinase from rat liver and identification of threonine 172 as the major site at which it phosphorylates AMP-activated protein kinase. J Biol Chem. 1996;271(44):27879–27887. doi:10.1074/jbc.271.44.27879

22. Woods A, Johnstone SR, Dickerson K, et al. LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr Biol. 2003;13(22):2004–2008. doi:10.1016/j.cub.2003.10.031

23. Wang S, Li H, Yuan M, et al. Role of AMPK in autophagy. Front Physiol. 2022;13:1015500. doi:10.3389/fphys.2022.1015500

24. Herzig S, Shaw RJ. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 2018;19(2):121–135. doi:10.1038/nrm.2017.95

25. Lin SC, Hardie DG. AMPK: sensing Glucose as well as Cellular Energy Status. Cell Metab. 2018;27(2):299–313. doi:10.1016/j.cmet.2017.10.009

26. Sagan L. On the origin of mitosing cells. 1967. J NIH Res. 1993;5(3):65–72.

27. Garcia D, Shaw RJ. AMPK: mechanisms of Cellular Energy Sensing and Restoration of Metabolic Balance. Mol Cell. 2017;66(6):789–800. doi:10.1016/j.molcel.2017.05.032

28. Hardie DG, Hawley SA. AMP-activated protein kinase: the energy charge hypothesis revisited. Bioessays. 2001;23(12):1112–1119. doi:10.1002/bies.10009

29. Oakhill JS, Steel R, Chen Z-P, et al. AMPK is a direct adenylate charge-regulated protein kinase. Science. 2011;332(6036):1433–1435. doi:10.1126/science.1200094

30. Myers RW, Guan H-P, Ehrhart J, et al. Systemic pan-AMPK activator MK-8722 improves glucose homeostasis but induces cardiac hypertrophy. Science. 2017;357(6350):507–511. doi:10.1126/science.aah5582

31. Shackelford DB, Shaw RJ. The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer. 2009;9(8):563–575. doi:10.1038/nrc2676

32. Shaw RJ, Lamia KA, Vasquez D, et al. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science. 2005;310(5754):1642–1646. doi:10.1126/science.1120781

33. Zhang CS, Jiang B, Li M, et al. The lysosomal v-ATPase-Ragulator complex is a common activator for AMPK and mTORC1, acting as a switch between catabolism and anabolism. Cell Metab. 2014;20(3):526–540. doi:10.1016/j.cmet.2014.06.014

34. Zhang CS, Hawley SA, Zong Y, et al. Fructose-1,6-bisphosphate and aldolase mediate glucose sensing by AMPK. Nature. 2017;548(7665):112–116. doi:10.1038/nature23275

35. Davies SP, Sim ATR, Hardie DG. Location and function of three sites phosphorylated on rat acetyl-CoA carboxylase by the AMP-activated protein kinase. Eur J Biochem. 1990;187(1):183–190. doi:10.1111/j.1432-1033.1990.tb15293.x

36. Fediuc S, Gaidhu MP, Ceddia RB. Regulation of AMP-activated protein kinase and acetyl-CoA carboxylase phosphorylation by palmitate in skeletal muscle cells. J Lipid Res. 2006;47(2):412–420. doi:10.1194/jlr.M500438-JLR200

37. Egan DF, Shackelford DB, Mihaylova MM, et al. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science. 2011;331(6016):456–461. doi:10.1126/science.1196371

38. Kim J, Kim Y, Fang C, et al. Differential regulation of distinct Vps34 complexes by AMPK in nutrient stress and autophagy. Cell. 2013;152(1–2):290–303. doi:10.1016/j.cell.2012.12.016

39. Inoki K, Zhu T, Guan KL. TSC2 mediates cellular energy response to control cell growth and survival. Cell. 2003;115(5):577–590. doi:10.1016/S0092-8674(03)00929-2

40. Toyama EQ, Herzig S, Courchet J, et al. Metabolism. AMP-activated protein kinase mediates mitochondrial fission in response to energy stress. Science. 2016;351(6270):275–281. doi:10.1126/science.aab4138

41. Wu D, Hu D, Chen H, et al. Glucose-regulated phosphorylation of TET2 by AMPK reveals a pathway linking diabetes to cancer. Nature. 2018;559(7715):637–641. doi:10.1038/s41586-018-0350-5

42. Zhang CS, Li M, Wang Y, et al. The aldolase inhibitor aldometanib mimics glucose starvation to activate lysosomal AMPK. Nat Metab. 2022;4(10):1369–1401. doi:10.1038/s42255-022-00640-7

43. Lassen KG, Kuballa P, Conway KL, et al. Atg16L1 T300A variant decreases selective autophagy resulting in altered cytokine signaling and decreased antibacterial defense. Proc Natl Acad Sci U S A. 2014;111(21):7741–7746. doi:10.1073/pnas.1407001111

44. Asano J, Sato T, Ichinose S, et al. Intrinsic Autophagy Is Required for the Maintenance of Intestinal Stem Cells and for Irradiation-Induced Intestinal Regeneration. Cell Rep. 2017;20(5):1050–1060. doi:10.1016/j.celrep.2017.07.019

45. Strisciuglio C, Duijvestein M, Verhaar AP, et al. Impaired autophagy leads to abnormal dendritic cell-epithelial cell interactions. J Crohns Colitis. 2013;7(7):534–541. doi:10.1016/j.crohns.2012.08.009

46. Zhang H, Lang W, Liu X, et al. Procyanidin A1 alleviates DSS-induced ulcerative colitis via regulating AMPK/mTOR/p70S6K-mediated autophagy. J Physiol Biochem. 2022;78(1):213–227. doi:10.1007/s13105-021-00854-5

47. Kim S, Lee J-Y, Shin SG, et al. ESRRA (estrogen related receptor alpha) is a critical regulator of intestinal homeostasis through activation of autophagic flux via gut microbiota. Autophagy. 2021;17(10):2856–2875. doi:10.1080/15548627.2020.1847460

48. Saber S, El-Kader EMA. Novel complementary coloprotective effects of metformin and MCC950 by modulating HSP90/NLRP3 interaction and inducing autophagy in rats. Inflammopharmacology. 2021;29(1):237–251. doi:10.1007/s10787-020-00730-6

49. Talero E, Alcaide A, Ávila-Román J, et al. Expression patterns of sirtuin 1-AMPK-autophagy pathway in chronic colitis and inflammation-associated colon neoplasia in IL-10-deficient mice. Int Immunopharmacol. 2016;35:248–256. doi:10.1016/j.intimp.2016.03.046

50. Zhang SL, Li Z-Y, Wang D-S, et al. Aggravated ulcerative colitis caused by intestinal Metrnl deficiency is associated with reduced autophagy in epithelial cells. Acta Pharmacol Sin. 2020;41(6):763–770. doi:10.1038/s41401-019-0343-4

51. Liu C, Wang J, Yang Y, et al. Ginsenoside Rd ameliorates colitis by inducing p62-driven mitophagy-mediated NLRP3 inflammasome inactivation in mice. Biochem Pharmacol. 2018;155:366–379. doi:10.1016/j.bcp.2018.07.010

52. Shao BZ, Wang S-L, Fang J, et al. Alpha7 Nicotinic Acetylcholine Receptor Alleviates Inflammatory Bowel Disease Through Induction of AMPK-mTOR-p70S6K-Mediated Autophagy. Inflammation. 2019;42(5):1666–1679. doi:10.1007/s10753-019-01027-9

53. Yang YX, Yuan Y, Xia B. Cinnamtannin D1 ameliorates DSS-induced colitis by preventing Th17/Treg imbalance through activation of the AMPK/mTOR pathway. Allergol Immunopathol. 2022;50(5):153–161. doi:10.15586/aei.v50i5.654

54. Leng XY, Yang J, Fan H, et al. JMJD3/H3K27me3 epigenetic modification regulates Th17/Treg cell differentiation in ulcerative colitis. Int Immunopharmacol. 2022;110:109000. doi:10.1016/j.intimp.2022.109000

55. Chao L, Li Z, Zhou J, et al. Shen-Ling-Bai-Zhu-San Improves Dextran Sodium Sulfate-Induced Colitis by Inhibiting Caspase-1/Caspase-11-Mediated Pyroptosis. Front Pharmacol. 2020;11:814. doi:10.3389/fphar.2020.00814

56. Deng Z, Ni J, Wu X, et al. GPA peptide inhibits NLRP3 inflammasome activation to ameliorate colitis through AMPK pathway. Aging. 2020;12(18):18522–18544. doi:10.18632/aging.103825

57. Guo J, Liao M, Wang J. TLR4 signaling in the development of colitis-associated cancer and its possible interplay with microRNA-155. Cell Commun Signal. 2021;19(1):90. doi:10.1186/s12964-021-00771-6

58. Tufekci KU, Eltutan BI, Isci KB, et al. Resveratrol Inhibits NLRP3 Inflammasome-Induced Pyroptosis and miR-155 Expression in Microglia Through Sirt1/AMPK Pathway. Neurotox Res. 2021;39(6):1812–1829. doi:10.1007/s12640-021-00435-w

59. Yao X. Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol Ther. 2014;141(2):125–139. doi:10.1016/j.pharmthera.2013.09.004

60. Deng J, Zeng L, Lai X, et al. Metformin protects against intestinal barrier dysfunction via AMPKα1-dependent inhibition of JNK signalling activation. J Cell Mol Med. 2018;22(1):546–557. doi:10.1111/jcmm.13342

61. Perera AP, Fernando R, Shinde T, et al. MCC950, a specific small molecule inhibitor of NLRP3 inflammasome attenuates colonic inflammation in spontaneous colitis mice. Sci Rep. 2018;8(1):8618. doi:10.1038/s41598-018-26775-w

62. Keshk WA, Soliman NA, Ali DA, et al. Mechanistic evaluation of AMPK/SIRT1/FXR signaling axis, inflammation, and redox status in thioacetamide-induced liver cirrhosis: the role of Cichorium intybus linn (chicory)-supplemented diet. J Food Biochem. 2019;43(8):e12938. doi:10.1111/jfbc.12938

63. Pu Z, Liu Y, Li C, et al. Using Network Pharmacology for Systematic Understanding of Geniposide in Ameliorating Inflammatory Responses in Colitis Through Suppression of NLRP3 Inflammasome in Macrophage by AMPK/Sirt1 Dependent Signaling. Am J Chin Med. 2020;48(7):1693–1713. doi:10.1142/S0192415X20500846

64. Xu X, Wang Y, Wei Z, et al. Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARgamma/AMPK/ACC1 pathway. Cell Death Dis. 2017;8(3):e2723. doi:10.1038/cddis.2017.150

65. Chang YY, Huan Q-C, Peng J, et al. P2Y1R Ligation Suppresses Th17 Cell Differentiation and Alleviates Colonic Inflammation in an AMPK-Dependent Manner. Front Immunol. 2022;13:820524. doi:10.3389/fimmu.2022.820524

66. Di Fusco D, Dinallo V, Monteleone I, et al. Metformin inhibits inflammatory signals in the gut by controlling AMPK and p38 MAP kinase activation. Clin Sci. 2018;132(11):1155–1168. doi:10.1042/CS20180167

67. Sun X, Yang Q, Rogers CJ, et al. AMPK improves gut epithelial differentiation and barrier function via regulating Cdx2 expression. Cell Death Differ. 2017;24(5):819–831. doi:10.1038/cdd.2017.14

68. Chen L, Wang J, You Q, et al. Activating AMPK to Restore Tight Junction Assembly in Intestinal Epithelium and to Attenuate Experimental Colitis by Metformin. Front Pharmacol. 2018;9:761. doi:10.3389/fphar.2018.00761

69. Chen C, Liang H, Wang J, et al. Heterophyllin B an Active Cyclopeptide Alleviates Dextran Sulfate Sodium-Induced Colitis by Modulating Gut Microbiota and Repairing Intestinal Mucosal Barrier via AMPK Activation. Mol Nutr Food Res. 2022;66(17):e2101169. doi:10.1002/mnfr.202101169

70. Xu B, Li Y-L, Xu M, et al. Geniposide ameliorates TNBS-induced experimental colitis in rats via reducing inflammatory cytokine release and restoring impaired intestinal barrier function. Acta Pharmacol Sin. 2017;38(5):688–698. doi:10.1038/aps.2016.168

71. Xiong YJ, Deng Z-B, Liu J-N, et al. Enhancement of epithelial cell autophagy induced by sinensetin alleviates epithelial barrier dysfunction in colitis. Pharmacol Res. 2019;148:104461. doi:10.1016/j.phrs.2019.104461

72. Zhou D, Huang C, Lin Z, et al. Macrophage polarization and function with emphasis on the evolving roles of coordinated regulation of cellular signaling pathways. Cell Signal. 2014;26(2):192–197. doi:10.1016/j.cellsig.2013.11.004

73. Li J, Li M, Ye K, et al. Chemical profile of Xian-He-Cao-Chang-Yan formula and its effects on ulcerative colitis. J Ethnopharmacol. 2021;267:113517. doi:10.1016/j.jep.2020.113517

74. Zhou K, Cheng R, Liu B, et al. Eupatilin ameliorates dextran sulphate sodium-induced colitis in mice partly through promoting AMPK activation. Phytomedicine. 2018;46:46–56. doi:10.1016/j.phymed.2018.04.033

75. Tsai CF, Chen G-W, Chen Y-C, et al. Regulatory Effects of Quercetin on M1/M2 Macrophage Polarization and Oxidative/Antioxidative Balance. Nutrients. 2021;14(1):67. doi:10.3390/nu14010067

76. Wu YX. Dehydrocostus Lactone Attenuates Methicillin-Resistant Staphylococcus aureus-Induced Inflammation and Acute Lung Injury via Modulating Macrophage Polarization. Int J Mol Sci. 2021;22(18):567.

77. Ji G, Zhang Y, Yang Q, et al. Genistein suppresses LPS-induced inflammatory response through inhibiting NF-kappaB following AMP kinase activation in RAW 264.7 macrophages. PLoS One. 2012;7(12):e53101. doi:10.1371/journal.pone.0053101

78. Duan WJ, Li Y-F, Liu F-L, et al. A SIRT3/AMPK/autophagy network orchestrates the protective effects of trans-resveratrol in stressed peritoneal macrophages and RAW 264.7 macrophages. Free Radic Biol Med. 2016;95:230–242. doi:10.1016/j.freeradbiomed.2016.03.022

79. Guo R, Meng Q, Wang B, et al. Anti-inflammatory effects of Platycodin D on dextran sulfate sodium (DSS) induced colitis and E. coli Lipopolysaccharide (LPS) induced inflammation. Int Immunopharmacol. 2021;94:107474. doi:10.1016/j.intimp.2021.107474

80. Kim N, Lertnimitphun P, Jiang Y, et al. Andrographolide inhibits inflammatory responses in LPS-stimulated macrophages and murine acute colitis through activating AMPK. Biochem Pharmacol. 2019;170:113646. doi:10.1016/j.bcp.2019.113646

81. Wei W, Ding M, Zhou K, et al. Protective effects of wedelolactone on dextran sodium sulfate induced murine colitis partly through inhibiting the NLRP3 inflammasome activation via AMPK signaling. Biomed Pharmacother. 2017;94:27–36. doi:10.1016/j.biopha.2017.06.071

82. Huan Q, Gao Z, Wu P, et al. Picroside III, an Active Ingredient of Picrorhiza scrophulariiflora, Ameliorates Experimental Colitis by Protecting Intestinal Barrier Integrity. Chem Biodivers. 2023;20(6):e202300572. doi:10.1002/cbdv.202300572

83. Han J, Hardie DG, et al. Atractylenolide III Improves Mitochondrial Function and Protects Against Ulcerative Colitis by Activating AMPK/SIRT1/PGC-1alpha. Mediators Inflamm. 2022;2022:9129984. doi:10.1155/2022/9129984

84. Vitali R, Palone F, Pierdomenico M, et al. Dipotassium glycyrrhizate via HMGB1 or AMPK signaling suppresses oxidative stress during intestinal inflammation. Biochem Pharmacol. 2015;97(3):292–299. doi:10.1016/j.bcp.2015.07.039

85. Xu X, Li W, Yu Z, et al. Berberine Ameliorates Dextran Sulfate Sodium-Induced Ulcerative Colitis and Inhibits the Secretion of Gut Lysozyme via Promoting Autophagy. Metabolites. 2022;12(8):676. doi:10.3390/metabo12080676

86. Liu Y, Li B-G, Su Y-H, et al. Potential activity of Traditional Chinese Medicine against Ulcerative colitis: a review. J Ethnopharmacol. 2022;289:115084. doi:10.1016/j.jep.2022.115084

87. Pan SM, Wang C-L, Hu Z-F, et al. Baitouweng decoction repairs the intestinal barrier in DSS-induced colitis mice via regulation of AMPK/mTOR-mediated autophagy. J Ethnopharmacol. 2023;318(Pt A):116888. doi:10.1016/j.jep.2023.116888

88. Ge W, Wang H-Y, Zhao H-M, et al. Effect of Sishen Pill on Memory T Cells From Experimental Colitis Induced by Dextran Sulfate Sodium. Front Pharmacol. 2020;11:908. doi:10.3389/fphar.2020.00908

89. Keshteli AH, Madsen KL, Dieleman LA. Diet in the Pathogenesis and Management of Ulcerative Colitis; A Review of Randomized Controlled Dietary Interventions. Nutrients. 2019;11(7):1498. doi:10.3390/nu11071498

90. Deng Z, Rong Y, Teng Y, et al. Broccoli-Derived Nanoparticle Inhibits Mouse Colitis by Activating Dendritic Cell AMP-Activated Protein Kinase. Mol Ther. 2017;25(7):1641–1654. doi:10.1016/j.ymthe.2017.01.025

91. Bibi S, Kang Y, Du M, et al. Dietary red raspberries attenuate dextran sulfate sodium-induced acute colitis. J Nutr Biochem. 2018;51:40–46. doi:10.1016/j.jnutbio.2017.08.017

92. Muanprasat C, Wongkrasant P, Satitsri S, et al. Activation of AMPK by chitosan oligosaccharide in intestinal epithelial cells: mechanism of action and potential applications in intestinal disorders. Biochem Pharmacol. 2015;96(3):225–236. doi:10.1016/j.bcp.2015.05.016

93. Turer E, McAlpine W, Wang K-W, et al. Creatine maintains intestinal homeostasis and protects against colitis. Proc Natl Acad Sci U S A. 2017;114(7):E1273–E1281. doi:10.1073/pnas.1621400114

94. Farzaei MH, Gao Z, Wu P, et al. An update on dietary consideration in inflammatory bowel disease: anthocyanins and more. Expert Rev Gastroenterol Hepatol. 2018;12(10):1007–1024. doi:10.1080/17474124.2018.1513322

95. Afrin MR, Arumugam S, Rahman MA, et al. Le Carbone, a charcoal supplement, modulates DSS-induced acute colitis in mice through activation of AMPKalpha and downregulation of STAT3 and caspase 3 dependent apoptotic pathways. Int Immunopharmacol. 2017;43:70–78. doi:10.1016/j.intimp.2016.10.023

96. Yue Y, Wu S, Li Z, et al. Wild jujube polysaccharides protect against experimental inflammatory bowel disease by enabling enhanced intestinal barrier function. Food Funct. 2015;6(8):2568–2577. doi:10.1039/C5FO00378D

97. Zhang Y, Guo C, Li Y, et al. Alginate Oligosaccharides Ameliorate DSS-Induced Colitis through Modulation of AMPK/NF-kappaB Pathway and Intestinal Microbiota. Nutrients. 2022;14(14).

98. Li H, Pang B, Nie B, et al. Dioscin promotes autophagy by regulating the AMPK-mTOR pathway in ulcerative colitis. Immunopharmacol Immunotoxicol. 2022;44(2):238–246. doi:10.1080/08923973.2022.2037632

99. Tian Q, Xu Z, Sun Q, et al. Broccoli-Derived Glucoraphanin Activates AMPK/PGC1alpha/NRF2 Pathway and Ameliorates Dextran-Sulphate-Sodium-Induced Colitis in Mice. Antioxidants (Basel). 2022;11(12). doi:10.3390/antiox11122404

100. Zhang B, Zhang Y, Liu X, et al. Differential Protective Effect of Resveratrol and Its Microbial Metabolites on Intestinal Barrier Dysfunction is Mediated by the AMPK Pathway. J Agric Food Chem. 2022;70(36):11301–11313. doi:10.1021/acs.jafc.2c04101

101. Olivier S, Diounou H, Pochard C, et al. Intestinal Epithelial AMPK Deficiency Causes Delayed Colonic Epithelial Repair in DSS-Induced Colitis. Cells. 2022;11(4):590. doi:10.3390/cells11040590

102. Takahara M, Takaki A, Hiraoka S, et al. Metformin ameliorates chronic colitis in a mouse model by regulating interferon-γ-producing lamina propria CD4 + T cells through AMPK activation. FASEB J. 2022;36(2):e22139. doi:10.1096/fj.202100831RR

103. Youssef ME, Abd El-Fattah EE, Abdelhamid AM, et al. Interference With the AMPKalpha/mTOR/NLRP3 Signaling and the IL-23/IL-17 Axis Effectively Protects Against the Dextran Sulfate Sodium Intoxication in Rats: a New Paradigm in Empagliflozin and Metformin Reprofiling for the Management of Ulcerative Colitis. Front Pharmacol. 2021;12:719984. doi:10.3389/fphar.2021.719984

104. Koh SJ, Kim JM, Kim I-K, et al. Anti-inflammatory mechanism of metformin and its effects in intestinal inflammation and colitis-associated colon cancer. J Gastroenterol Hepatol. 2014;29(3):502–510. doi:10.1111/jgh.12435

105. Park H, Kim W, Kim D, et al. Mesalazine Activates Adenosine Monophosphate-activated Protein Kinase: implication in the Anti-inflammatory Activity of this Anti-colitic Drug. Curr Mol Pharmacol. 2019;12(4):272–280. doi:10.2174/1874467212666190308103448

106. Arab HH, Al-Shorbagy MY, Saad MA. Activation of autophagy and suppression of apoptosis by dapagliflozin attenuates experimental inflammatory bowel disease in rats: targeting AMPK/mTOR, HMGB1/RAGE and Nrf2/HO-1 pathways. Chem Biol Interact. 2021;335:109368. doi:10.1016/j.cbi.2021.109368

107. El-Ghannam MS, Saad MA, Nassar NN, et al. Linagliptin ameliorates acetic acid-induced colitis via modulating AMPK/SIRT1/PGC-1alpha and JAK2/STAT3 signaling pathway in rats. Toxicol Appl Pharmacol. 2022;438:115906. doi:10.1016/j.taap.2022.115906

108. Banskota S, Wang H, Kwon YH, et al. Inhibition of NADPH Oxidase (NOX) 2 Mitigates Colitis in Mice with Impaired Macrophage AMPK Function. Biomedicines. 2023;11(5):1443. doi:10.3390/biomedicines11051443

109. Bai A, Yong M, Ma AG, et al. Novel anti-inflammatory action of 5-aminoimidazole-4-carboxamide ribonucleoside with protective effect in dextran sulfate sodium-induced acute and chronic colitis. J Pharmacol Exp Ther. 2010;333(3):717–725. doi:10.1124/jpet.109.164954

110. Bai A, Ma AG, Yong M, et al. AMPK agonist downregulates innate and adaptive immune responses in TNBS-induced murine acute and relapsing colitis. Biochem Pharmacol. 2010;80(11):1708–1717. doi:10.1016/j.bcp.2010.08.009

111. Zhao Y, Guo Q, Zhao K, et al. Small molecule GL-V9 protects against colitis-associated colorectal cancer by limiting NLRP3 inflammasome through autophagy. Oncoimmunology. 2017;7(1):e1375640. doi:10.1080/2162402X.2017.1375640

112. Antonioli L, Pellegrini C, Fornai M, et al. Preclinical Development of FA5, a Novel AMP-Activated Protein Kinase (AMPK) Activator as an Innovative Drug for the Management of Bowel Inflammation. Int J Mol Sci. 2021;22(12):6325. doi:10.3390/ijms22126325

113. Timilshina M, Kang Y, Dahal I, et al. BJ-3105, a 6-Alkoxypyridin-3-ol Analog, Impairs T Cell Differentiation and Prevents Experimental Autoimmune Encephalomyelitis Disease Progression. PLoS One. 2017;12(1):e0168942. doi:10.1371/journal.pone.0168942

114. Gurung P, Dahal S, Chaudhary P, et al. Potent Inhibitory Effect of BJ-3105, a 6-Alkoxypyridin-3-ol Derivative, on Murine Colitis Is Mediated by Activating AMPK and Inhibiting NOX. Int J Mol Sci. 2020;21(9):3145. doi:10.3390/ijms21093145

115. Lee SY, Lee SH, Yang E-J, et al. Coenzyme Q10 Inhibits Th17 and STAT3 Signaling Pathways to Ameliorate Colitis in Mice. J Med Food. 2017;20(9):821–829. doi:10.1089/jmf.2016.3859

116. Jiang J, Chang X, Nie Y, et al. Orally administered MOTS-c analogue ameliorates dextran sulfate sodium-induced colitis by inhibiting inflammation and apoptosis. Eur J Pharmacol. 2023;939:175469. doi:10.1016/j.ejphar.2022.175469

117. Ke P, Shao B-Z, Xu Z-Q, et al. Activation of Cannabinoid Receptor 2 Ameliorates DSS-Induced Colitis through Inhibiting NLRP3 Inflammasome in Macrophages. PLoS One. 2016;11(9):e0155076. doi:10.1371/journal.pone.0155076

118. Banskota S, Wang H, Kwon YH, et al. Salicylates Ameliorate Intestinal Inflammation by Activating Macrophage AMPK. Inflamm Bowel Dis. 2021;27(6):914–926. doi:10.1093/ibd/izaa305

119. Ma T, Tian X, Zhang B, et al. Low-dose metformin targets the lysosomal AMPK pathway through PEN2. Nature. 2022;603(7899):159–165. doi:10.1038/s41586-022-04431-8

120. Steinberg GR, Hardie DG. New insights into activation and function of the AMPK. Nat Rev Mol Cell Biol. 2022.

121. Bu Y, Peng M, Tang X, et al. Protective effects of metformin in various cardiovascular diseases: clinical evidence and AMPK -dependent mechanisms. J Cell Mol Med. 2022;26(19):4886–4903. doi:10.1111/jcmm.17519

Creative Commons License © 2023 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.