- Review
- Open access
- Published:
Metabolic reprogramming of peritoneal mesothelial cells in peritoneal dialysis–associated fibrosis: therapeutic targets and strategies
Cell Communication and Signaling volume 23, Article number: 114 (2025)
Abstract
Peritoneal dialysis (PD) is considered a life-saving treatment for end-stage renal disease. However, prolonged PD use can lead to the development of peritoneal fibrosis (PF), diminishing its efficacy. Peritoneal mesothelial cells (PMCs) are key initiators of PF when they become damaged. Exposure to high glucose‑based peritoneal dialysis fluids (PDFs) contributes to PF development by directly affecting highly metabolically active PMCs. Recent research indicates that PMCs undergo metabolic reprogramming when exposed to high-glucose PDFs, including enhanced glycolysis, impaired oxidative phosphorylation, abnormal lipid metabolism, and mitochondrial dysfunction. Although this metabolic transition temporarily compensates for the cellular damage and maintains energy levels, its long-term impact on peritoneal tissue is concerning. Multiple studies have identified a close association between this shift in energy metabolism and PF, and may promote the progression of PF through various molecular mechanisms. This review explores recent findings regarding the role and mechanism of PMC metabolic reprogramming in PF progression. Moreover, it provides a summary of potential therapeutic strategies aimed at various metabolic processes, including glucose metabolism, lipid metabolism, and mitochondrial function. The review establishes that targeting metabolic reprogramming in PMCs may be a novel strategy for preventing and treating PD-associated fibrosis.
Graphical Abstract
Overview of the metabolic reprogramming of PMCs in PF associated with PD and therapeutic implications. Under physiological conditions, PMCs primarily produce ATP through OXPHOS and FAO to maintain cellular functions. High-glucose PDFs can induce metabolic reprogramming of PMCs, characterized by increased glycolysis, the polyol pathway, and the PPP, inhibited OXPHOS, impaired FAO, lipid exocytosis and deposition, and mitochondrial dysfunction. These metabolic changes lead to peritoneal damage and fibrosis through multiple metabolic pathways. Potential therapeutic strategies target glucose absorption, glycolysis, the polyol pathway, FAO restoration, lipid deposition, and mitochondrial dysfunction. FAO, fatty acid oxidation; OXPHOS, oxidative phosphorylation; PD, peritoneal dialysis, PDF, peritoneal dialysis fluid; PMC, peritoneal mesothelial cell; PPP, pentose phosphate pathway.

Introduction
Currently, there are 697.5 million patients with chronic kidney disease worldwide [1], with over 2.5 million receiving renal replacement therapy [2]. Globally, peritoneal dialysis (PD) is the primary treatment for end-stage renal disease (ESRD), constituting approximately 11% of all dialysis and 9% of kidney replacement therapies [3, 4]. Nonetheless, peritoneal fibrosis (PF) poses a formidable challenge for the widespread clinical application of PD. PF is one of the pathological changes occurring gradually in the peritoneum due to chronic inflammation and infection, and it is also one of the primary causes of PD technique failure, being responsible for 30% of such failures [5,6,7]. The 3-year PD technique survival rates range from 29 to 91% worldwide [4, 8, 9]. During long-term PD treatment, the progression of peritoneal fibrosis is closely linked to PD technique failure, reduced patient survival, and the development of other complications [10, 11]. Effective interventions to prevent PF progression are lacking, highlighting the necessity to investigate the mechanisms underlying PF.
Glucose containing peritoneal dialysis fluid (PDF)-mediated PF is a key factor limiting PD efficacy [12,13,14]. High-glucose PDFs create an osmotic gradient that facilitates water and solute removal from the peritoneal cavity. The biological incompatibility of high-glucose PDFs involves factors, such as elevated osmolarity (10–50 times greater than serum) [7], glucose degradation product formation during heat sterilization [15], and advanced glycation end product (AGE) production in the peritoneal cavity [7, 16]. High-glucose-induced PF is commonly associated with metabolic abnormalities, inducing a diabetes-like condition within the peritoneal cavity [17]. However, the metabolic mechanisms underlying PF remain unclear.
Peritoneal mesothelial cells (PMCs), the first line of defense on the peritoneum surface, are crucial for peritoneum defense and function during PD, thereby maintaining homeostasis, managing fluid transport, being involved in tissue repair, and responding to immune challenges [6, 18,19,20]. Highly metabolically active PMCs undergo considerable metabolic alterations during prolonged PD to maintain peritoneal homeostasis [21]. Prolonged PD exposure to high-glucose solutions alters PMC metabolism, characterized by hyperglycolysis, ultimately resulting in PF [22]. This exposure also causes PMC injury and mesothelial-mesenchymal transition (MMT) by triggering mitochondrial dysfunction and oxidative stress [23, 24]. Such changes compromise peritoneal integrity and function, making PMC metabolic reprogramming a central factor in PD-related fibrosis.
Metabolic alterations in PMCs are pivotal for determining the prognosis of patients undergoing PD. The structural and functional modifications induced by long-term PD are intricately associated with the metabolic activities of PMCs [25]. High - glucose PDFs can cause mesothelial cell dysfunction and induce MMT, initiate and accelerate PF, and affect the patient’s technical survival rate and long - term prognosis [26]. Moreover, the metabolic activities of PMCs are linked to the peritoneal inflammatory response [27, 28]. This inflammatory response not only compromises peritoneal integrity but also affects systemic inflammation and patient prognosis [29]. Additionally, high-glucose PDFs can lead to abnormal lipid metabolism, further impacting PMC function and the patient’s overall metabolic state [30]. In conclusion, a comprehensive understanding of the mechanisms underlying metabolic changes in PMCs is crucial for the development of new treatment strategies.
Recent studies have focused on the pathological and physiological mechanisms of metabolic reprogramming in PMCs and their role in promoting PD-related fibrosis. This review highlights the potential of targeting metabolic reprogramming as a novel approach to delay PF progression, offering new therapeutic strategies for preventing and treating PD-related fibrosis (Table 1).
Metabolic reprogramming and PF progression
Metabolic reprogramming, a mechanism wherein cells alter their metabolic patterns to support proliferation, growth, and energy demands, is a hallmark of cancer [31, 32]. While most cells generate energy primarily through oxidative phosphorylation (OXPHOS), tumor cells preferentially utilize glycolysis to produce adenosine triphosphate (ATP) even in oxygen-rich environments, a phenomenon known as the Warburg effect [33]. However, metabolic reprogramming occurs in non-tumor cells following genetic and environmental changes [34, 35]. Substantial alterations in the intracellular metabolic pool often accompany metabolic reprogramming [36]. This cellular adaptation helps cells resist external stress and acquire new functions. Moreover, it is intricately linked to the onset and progression of diseases.
A recent randomized controlled trial found that patients with ESRD undergoing PD exhibited substantial metabolic disturbances, which were associated with adverse clinical outcomes [37]. PD techniques have faced considerable limitations owing to the development of high-glucose PDFs-mediated PF and ultrafiltration failure. Exposure to high-glucose PDFs is closely linked to metabolic disorders [38, 39], inflammatory responses [6], activation of the renin-angiotensin system (RAS) [40], and angiogenesis in the peritoneum [41, 42]. These factors contribute to PMC damage, death, apoptosis, and MMT, resulting in the promotion of PF and even adverse outcomes [13, 17]. Thus, glucose-containing PDFs-induced metabolic reprogramming may substantially affect peritoneal pathophysiology.
Two key mechanisms underlie high-glucose PDFs-induced fibrosis: the production of AGE, accompanied by a decrease in the ultrafiltration osmotic pressure gradient due to heightened expression of glucose transporter type 1 (GLUT-1), and an increase in the nicotinamide adenine dinucleotide (NADH)/nicotinamide adenine dinucleotide (NAD+) ratio (NADH/NAD+), causing a pseudohypoxia state in peritoneal tissues and cells [22, 43]. These conditions can lead to peritoneal damage and subsequent fibrosis. High-glucose PDF-induced PF is closely associated with metabolic disorders, including abnormal glycolysis, mitochondrial dysfunction, impaired OXPHOS, and disrupted lipid homeostasis [22, 39, 44, 45]. In summary, prolonged exposure to high-glucose PDFs induces metabolic reprogramming and promotes PF progression, highlighting the important role of metabolism in PD-related fibrosis (Fig. 1).
Metabolic reprogramming in PMCs during PD. Long-term PD induces metabolic reprogramming as an adaptive response to high-glucose PDFs. This involves alterations in glucose metabolism, characterized by enhanced activity of glycolytic, polyol, pentose phosphate pathways, alongside OXPHOS inhibition. Subsequently, upregulation of intracellular damage factors and a considerable increase in the NADH/NAD + ratio exacerbate cellular pseudohypoxia, injury, and death. Concurrently, lipid metabolism reprogramming occurs due to impaired efflux, leading to lipid deposition and lipotoxicity injury. Impaired FAO affects mitochondrial function and cellular energy metabolism balance. FAO, fatty acid oxidation; OXPHOS, oxidative phosphorylation; PD, peritoneal dialysis, PDF, peritoneal dialysis fluid
Metabolism reprogramming in PMCs during PD
Glucose metabolism
During long-term PD treatment, PMCs undergo substantial alterations [46], such as metabolic reprogramming in response to stimulation by glucose-based PDFs, characterized by increased activation of glycolytic and polyol pathways, alongside OXPHOS inhibition [21, 24, 47, 48]. Prolonged maintenance of PMCs in this abnormal metabolic state increases energy demand and drives abnormal biological responses, including changes in cell phenotype, proliferation, apoptosis, and senescence [21, 48]. Thus, the metabolic reprogramming of PMCs is an important cellular process that mediates PD-related fibrosis.
Glycolysis
Glycolysis, a relatively inefficient anaerobic pathway used for the production of ATP, converts glucose into various products such as pyruvate and lactate. During long-term PD treatment, PMCs undergo a marked shift in glucose metabolism toward glycolysis [21, 48, 49]. Increased glycolysis in PMCs, stimulated by high glucose levels, elevates the NADH/NAD + ratio and lactate levels, resulting in the formation of a hypoxic intracellular environment [13, 47]. This pseudohypoxic environment induces pathological changes in PMCs including cell damage, apoptosis, and senescence [7, 22, 50]. The pseudohypoxia hypothesis is based on two primary factors. First, the excessive intracellular lactate concentrations impair lactate dehydrogenase function, disrupting normal intracellular and NADH/NAD + compensatory mechanisms for hypoxia in the tricarboxylic acid (TCA) cycle [51]. Second, activation of the intracellular expression of hypoxia-inducible factor-1 (HIF-1) induces chronic injury in PMCs [43], leading to the secretion of various pro-fibrotic and angiogenic factors that impair organ function [43, 52, 53].
PMCs undergo glucose metabolic reprogramming to reduce oxidative metabolism, promote ATP production, and enhance cell survival [48]. This reprogramming involves inhibiting OXPHOS via the GLUT-1 pathway and enhancing glycolysis by upregulating essential glycolysis genes, corresponding enzymes, and glucose transporter proteins [13, 48]. Excessive glycolysis caused by high glucose levels can damage PMCs through several alternative pathways. For example, hyperglycolysis in PMCs activates the NLRP3 inflammasome, leading to PF development [ 54]. Moreover, inhibiting glycolysis in the context of high-glucose PDFs reduces transforming growth factor beta 1 (TGF-β1)-induced cellular MMT and PF in mice [48]. While brief periods of mild hypoxia may enhance cell survival and recovery, prolonged exposure to hypoxia can result in cell damage and death [44]. Thus, the compensatory reprogramming of glucose metabolism in PMCs due to long-term exposure to high-glucose PDFs exacerbates cellular hypoxia and damage, ultimately resulting in PF and ultrafiltration failure.
OXPHOS
Under physiological conditions, glucose undergoes aerobic degradation processes, including the TCA and OXPHOS, ultimately converting it into H2O and CO2. Mitochondrial OXPHOS drives ATP production through the respiratory electron transport chain [ 55]. However, high-glucose environments inhibit mitochondrial OXPHOS, increase glucose consumption, enhance glycolysis, and disrupt lipid metabolism, resulting in a substantial increase in the cellular NADH/NAD + ratio [47]. Similarly, cellular pseudohypoxia induced by high-glucose PDFs leads to the shutdown of mitochondrial OXPHOS. Thus, high-glucose PDFs impair normal mitochondrial OXPHOS and induce the metabolic reprogramming of PMCs.
Impaired mitochondrial OXPHOS primarily arises from high-glucose PDF-induced mitochondrial dysfunction, leading to PMC injury and PF [24, 56, 57]. High-glucose PDFs causes pathological damage to mitochondria of PMC via various mechanisms, including TCA cycle enzyme deficiency, mitochondrial DNA damage, mitochondrial reactive oxygen species (mtROS) accumulation, and mitochondrial membrane damage, thereby inhibiting mitochondrial respiration and OXPHOS [23, 24, 48]. Clinical peritoneal samples from patients on continuous ambulatory peritoneal dialysis further support this, demonstrating that high-glucose PDFs may promote toxic free radical production during mitochondrial metabolism in PMCs, resulting in cumulative mitochondrial DNA damage [58, 59]. Moreover, mitochondrial dysfunction plays a considerable role in PF progression through tumor necrosis factor-α-induced cyclooxygenase-2 expression, prostaglandin E2 production, and activation of NLRP3 inflammasomes in PD-related PF [23, 60]. Mitochondrial dysfunction and impaired OXPHOS represent the vital causes of high-glucose PDF-induced PMC injury, highlighting their critical role as potential targets for PF treatment.
Polyol pathway
The polyol glucose metabolic pathway also exerts potentially deleterious effects on the peritoneum during long-term PD [61, 62]. The polyol pathway, a high-glucose-driven pathway, involves the reduction of NADPH-dependent glucose to sorbitol by aldose reductase (AR), followed by NAD+-dependent oxidation to fructose by sorbitol dehydrogenase [63, 64]. Under physiological conditions, most glucose is phosphorylated to glucose-6-phosphate (G6P) by hexokinase (HK), with a minor portion of non-phosphorylated glucose entering the polyol pathway, which has been attributed to the lower affinity of AR for glucose compared to that of HK [65]. However, a substantial increase in glucose concentration enhances polyol pathway activation to increase intracellular glucose metabolism [22]. The polyol pathway increases the intracellular NADH/NAD + ratio, exacerbating cellular hypoxia and enhancing the accumulation of intracellular ROS, ultimately leading to high-glucose-induced oxidative stress [22, 64]. Additionally, the polyol pathway induces the intracellular accumulation of poorly permeable sorbitol, increasing cell osmolarity and hypertonicity and resulting in cellular edema [65].
Moreover, the enhanced polyol pathway exacerbates PMC injury and enhances PF development through various downstream molecular mechanisms. In vitro studies have demonstrated that exposure to high-glucose PDFs leads to the activation of the polyol pathway in PMCs, thereby impairing cell function and causing injury via the upregulation of TGF-β1 and MCP-1 synthesis [62]. Conversely, studies conducted in rats have revealed that inhibition of the polyol pathway using AR activity inhibitors attenuates PF and angiogenesis [61]. Overall, abnormal activation of the polyol pathway represents a potential risk factor for PF in patients undergoing PD.
Lipid metabolism
Recent reports have implicated lipid metabolism in PF [39, 66]. Long-term exposure to high-glucose PDFs disrupts normal lipid metabolism in PMCs, with impaired fatty acid oxidation (FAO) and abnormal lipid deposition leading to imbalances in cellular energy metabolism [39, 66]. Triglycerides mainly consist of free fatty acids (FAs). Notably, FA homeostasis is maintained through their synthesis, transport, and metabolism. FA influx is mediated by increased expression of CD36, known as the scavenger receptor B2 [67]. Enhanced de novo FA synthesis is mediated by SREBP-1 C, FASN, and SCD-1 [68]; and β-oxidation of FAs is mediated by the rate-limiting enzyme carnitine palmitoyltransferase 1 A (CPT1A) [69]. Alternatively, cholesterol synthesis is regulated by HMGCR, while its efflux is mediated by ABCA1 and ABCG1 [70]. Abnormalities in these processes can result in the accumulation of intracellular lipids, leading to lipotoxic injury [13, 71]. Thus, disruption of lipid homeostasis represents an important mechanism involved in the development of high-glucose PDF–induced fibrosis.
High-glucose PDFs may promote PF by dysregulating lipid metabolism through various molecular pathways [13, 38, 39]. FAO, a major source of ATP and NADPH predominantly observed in mitochondria, is impaired in PMCs, promoting the progression of PF [45, 72]. Nonetheless, restoration of mesothelial FAO in PD animal models increases ATP and NADPH production, reverses mitochondrial superoxide production, maintains mitochondrial homeostasis, and preserves peritoneal structure during PD [45, 73]. In a peritonitis mouse model, the upregulation of FAO during inflammation promoted the resolution of inflammation by providing sufficient energy [74]. Thus, FAO plays an important functional role in PD-associated PF and may be a promising therapeutic target. High glucose levels induce the abnormal expression of angiotensin II type 2 receptor (AT2R), mTOR complex 1 (mTORC1), and Klotho, interfering with lipid metabolism. This leads to apoptosis, inflammation, and oxidative stress, which, in turn, activate the downstream TGF-β1 signaling pathway and MMT, ultimately leading to fibrosis [13, 75]. Moreover, high-glucose stimulation disrupts lipid metabolism through the activation of the sterol-regulatory element-binding protein-2/cleavage-activating protein pathway (SCAP/SREBP-2) and increased expression of HIF-1α, thereby promoting angiogenesis and PF [13, 66]. Therefore, restoring normal lipid metabolism in PMCs may serve as a novel therapeutic strategy for improving peritoneal injury.
Abnormal lipid deposition can cause lipotoxic damage and, thus, serves as an important contributor to PD-related fibrosis. High-glucose-induced lipotoxicity can directly exacerbate the expression of extracellular matrix (ECM) components, leading to the loss of epithelial characteristics in human peritoneal mesothelial cells (HPMCs), ultimately resulting in fibrotic changes [76]. In vitro and in vivo studies have revealed that Klotho, a key gene involved in lipid metabolism, promotes MMT and PF induced by high glucose PD [39]. Moreover, the histone lysine methyltransferase, enhancer of zeste homolog 2 (EZH2) can directly regulate Klotho expression through epigenetic modifications; thus, inhibiting EZH2 expression attenuates Klotho-mediated lipid deposition and PF [39]. Additionally, in vitro studies revealed that inhibition of AT2R promotes lipid disorder-mediated ECM deposition by upregulating lectin-like oxidized lipoprotein receptor-1 (LOX-1), resulting in intracellular deposition of lipid droplets in HPMCs [76]. Furthermore, in vitro and in vivo studies have demonstrated that high-glucose PDFs reduces ABCA1 expression through inhibition of peroxisome proliferator-activated receptor γ (PPARγ) activity, impairing lipid efflux and promoting lipid deposition [39]. Alternatively, CD36 acts as a metabolic regulator by mediating lipid sensing and transport. Notably, high glucose-induced upregulation of CD36 may lead to metabolic disorders by downregulating the PPARγ pathway; however, there are limited studies investigating these interactions in PD models.
The intricate and close relationship between lipid metabolism and mitochondria adversely affects mitochondrial function when lipid disorders occur [77, 78]. Notably, a high-glucose environment induces dysregulation of mitochondrial oxidative metabolism and increases mtROS generation, impairing mitochondrial lipid utilization and promoting lipid accumulation [79]. Lipotoxicity induces mitochondrial metabolic stress and dysfunction in diabetic mouse models, triggering the release of mtDNA and activation of the cGAS-STING pathway, ultimately promoting inflammation and apoptosis [80]. Disturbances in lipid metabolism may also potentially impede mitochondrial dynamics, leading to reduced mitochondrial function in PMCs.
Overall, lipid metabolism disorders induced by high-glucose PDFs contribute to PF progression, involving impaired lipid metabolism processes such as FAO, lipid efflux, and lipid deposition in PMCs.
Other metabolic alterations
In addition to glucose and lipid metabolism, other metabolic pathways such as amino acid metabolism, ketone body (KB) metabolism, and pentose phosphate pathway (PPP) play important roles in metabolic reprogramming [81,82,83] However, there has been limited research in the field of PD. A thorough investigation into the role of these metabolic processes can provide a better understanding of the pathophysiological process of PF and offer more effective solutions for clinical treatment.
The PPP, branching off from glycolysis, is considered the first step in glucose metabolism, serves as a major source of NADPH, and is a vital component in cell biosynthesis, metabolism, and cellular redox homeostasis [84]. Notably, high glucose levels can directly stimulate increased cellular glucose uptake, leading to an increase in the metabolic substrate G6P, thereby increasing its flow into the PPP [85]. Previous studies have confirmed that PPP plays a crucial role in combating oxidative stress and maintaining metabolic and redox homeostasis by reducing NADP to NADPH [86]. Therefore, PPP activity may rise in response to prolonged exposure to high-glucose PDFs as a means to regulate oxidative stress. However, comprehensive studies investigating PPP in the context of PD are limited; therefore, further studies are needed to confirm its involvement in PF.
Research into the mechanisms underlying amino acid metabolism in PMCs is lacking. A widespread loss of peritoneal proteins and amino acids has been observed in patients undergoing PD, potentially due to the use of high-glucose-based PD strategies [87]. Amino acid-based PD techniques appear to better preserve the ultrastructure, viability, and protein biosynthesis of HPMCs than conventional glucose-based PD [88, 89]. Notably, glutamine metabolism plays a potential protective role against PF [90, 91]. Glutamine, the most abundant free amino acid in the human body, contributes to DNA, RNA, and protein synthesis. Moreover, it promotes ATP production through OXPHOS [92]. A human clinical trial has revealed that glutamine deficiency during PD is associated with peritoneal pathological mechanisms, including impaired stress response and impaired host defense [91]. However, the precise mechanism underlying the protective effects of amino acid metabolism remains unclear and requires further investigation.
Recent advancements in our understanding of KB metabolism have revealed that KBs serve as alternative sources of ATP and regulate protein post-translational modifications, modulation of inflammation, and regulation of oxidative stress [93, 94]. Furthermore, KBs serve as important regulators of mitochondrial and nuclear metabolism and engage in complex competitive interactions with other OXPHOS substrates including glucose and FAs [95]. Given the pleiotropic effects of KBs, we postulate that KB metabolism may play an important role in the metabolic reprogramming of PMCs. However, evidence to support this hypothesis is currently lacking.
Therapeutic targets
The role of high-glucose PDFs in PF development is evident; however, effective interventions are still lacking. Nonetheless, glucose-based PDFs remains the most widely used type of PD, primarily due to cost limitations, challenges in promoting the use of biocompatible PDFs, and issues with the clinical translation of existing drugs. Metabolic abnormalities in PMCs can induce pathological changes, such as inflammation, oxidative stress, lipid deposition, and fibrosis. Thus, early interventions targeting glucose metabolism, mitochondrial function, and lipid metabolism may prove more efficacious than addressing individual downstream events. Various drugs and compounds aimed at modulating glycolysis, lipid metabolism, and other metabolic processes have shown promise in ameliorating PMC damage and inhibiting PF progression (Table 1).
Targeting glucose metabolism
Glucose metabolism plays a crucial role in the pathogenesis of PF. Recent studies have explored strategies to impede PF progression by reducing glucose absorption, inhibiting glycolysis, and decreasing the NADH/NAD + ratio. Numerous studies have highlighted the potential of targeting glucose metabolism to inhibit PF progression, with various metabolism-targeting drugs and compounds showing promise in the mitigation of peritoneal injury and fibrosis. Nonetheless, further investigation is needed before using these drugs as novel therapeutic strategies for PF (Fig. 2).
Targeting glucose metabolism in PMCs. Potential therapeutic strategies include targeting inhibiting glucose uptake using SGLT-2, GLUT-1, DPP-4, or GLP-1R inhibitors, hyperglycolysis inhibition using 2-DG, curcumin, or specific microRNAs (microRNA-26a and microRNA-200a), polyol pathway inhibition, and sorbitol clearance using zopolrestat (an AR inhibitor), sorbinil, or sodium pyruvate. 2-DG, 2-deoxyglucose; AR, aldose reductase; PMC, peritoneal mesothelial cell; SGLT-2, sodium glucose cotransporter-2; GLUT-1, glucose transporter type 1; DPP-4, Dipeptidyl peptidase-4; GLP-1R, Glucagon-like peptide 1 receptor
Targeting glucose absorption
Several anti-diabetic agents that reduce peritoneal glucose absorption exert protective effects against PF. The expression of glucose transporters in PMCs is influenced by glucose concentration, affecting the peritoneal response to inflammation, ECM production, and PF progression [96, 97]. Inhibitors of GLUT-1 and sodium glucose cotransporter-2 (SGLT-2) are effective inhibitors of glucose absorptions. In preclinical studies, GLUT-1 inhibitors have demonstrated efficacy in targeting glycolysis and inhibiting glucose uptake [98]. However, their clinical application requires further development of these drugs and compounds.
Conversely, SGLT-2 inhibitors, such as empagliflozin, dapagliflozin, and canagliflozin, have been widely used in clinical practice. SGLT-2 inhibitors hold the potential to reduce glucose absorption during PD, thereby attenuating MMT, fibrosis, and ultrafiltration failure through various mechanisms, including inhibiting SGLT-2 activity [99], alongside Nrf2/HO-1 [100], HIF [52], and TGF-β/Smad signaling [101]. However, the peritoneal protective effect of SGLT-2 inhibitors remains debatable. A study conducted on a rat model of acute PD determined that SGLT-2 inhibitors failed to reduce glucose uptake or increase ultrafiltration [102]; conversely, other studies using animal models of chronic PD have demonstrated that SGLT-2 inhibitors improve PF and function [17, 99, 101]. Therefore, further research is needed to clarify the peritoneal protective mechanisms of SGLT-2 inhibitors and determine whether these discrepancies in acute or chronic peritoneal injury responses are accurate. Recent studies have shown that SGLT − 2 inhibitors not only affect glucose metabolism but also have a significant impact on lipid and other related metabolic pathways. They positively influence lipid metabolism through multiple mechanisms, including lowering blood lipids, improving the ratio of low-density lipoprotein (LDL) particles, regulating lipid synthesis and transport, promoting fatty acid oxidation, and ketone body production [103,104,105]. They also enhance mitochondrial function, reduce oxidative stress, and increase mitochondrial synthesis [106, 107]. Moreover, recent research has reported that they can improve glycine metabolism [108]. The findings indicate that the effects of SGLT-2 inhibitors on peritoneal metabolism may extend beyond the scope of glucose metabolism. However, research on their impact in patients undergoing PD is still preliminary, necessitating further investigation.
In addition to glucose transporter proteins, novel antidiabetic medications, such as dipeptidyl peptidase-4 (DPP-4) inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists, may improve the pathophysiology of PD-associated fibrosis. DPP-4, a type II integral membrane glycoprotein with serine peptidase activity, primarily targets the enteric insulin GLP-1 [109]. DPP4 inhibitors, such as sitagliptin and linagliptin, exhibit hypoglycemic effects and demonstrate cytoprotective pleiotropic effects, including anti-inflammatory and anti-fibrotic actions [110, 111]. Previous studies have identified DPP4 as a substantial factor associated with PF [112]. Notably, in vitro and in vivo experiments have revealed that DPP4 inhibitors and exendin-4 can mitigate PD dysfunction and exhaustion by suppressing DPP-4 activity, the TGF/Smad3 pathway, the NF-κB pathway, and MMT [112]. Therefore, approaches targeting DPP-4 are potential therapeutic strategies for PF.
Targeting the polyol pathway
The polyol pathway plays a deleterious role in the peritoneum during long-term PD. Studies have indicated that the use of polyol pathway inhibitors can help reduce glucose-mediated peritoneal damage and enhance the long-term survival of PMCs [61, 62]. For example, zopolrestat, an AR inhibitor, alters the NADH/NAD + ratio by inhibiting AR activity in the polyol pathway, thereby impeding the conversion of glucose to sorbitol. Zopolrestat reportedly reduces peritoneal angiogenesis and fibrosis in a rat model of PF induced by high-glucose dialysate [61]. However, zopolrestat is not currently available for human use owing to its side effects. Nonetheless, other drugs have also been found to target the polyol pathway to delay PF progression, including sorbinil and sodium pyruvate [62]. In vitro studies have demonstrated that sorbinil and sodium pyruvate can alleviate PMC damage by inhibiting high-glucose–induced intracellular TGF-β1 and MCP-1 synthesis, as well as by enhancing intracellular sorbitol clearance [62].
Pyruvate contributes to improved sorbitol clearance, therefore, some studies have investigated methods for delaying PF progression based on pyruvate metabolism. Pyruvate dehydrogenase (PDH), a key enzyme that couples glycolysis with the Krebs cycle, has emerged as an important target for this therapeutic approach, with its anti-fibrotic effects being closely linked to its activity [113, 114]. Dichloroacetic acid (DCA), a potent inhibitor of PDH kinase, modulates the transfer of pyruvate to the Krebs cycle, thereby maintaining PDH activity and increasing intracellular energy [114]. Notably, DCA has demonstrated high efficacy in inhibiting fibrosis [113, 115]. Additionally, L-carnitine exerts anti-fibrotic effects by maintaining PDH activity through the reduction of PDH kinase activity and mitochondrial acetyl-CoA production [7, 116]. However, the efficacy of these drugs requires further validation.
Targeting glycolysis
Targeting glycolysis has also emerged as a promising therapeutic strategy for the treatment of PF. Notably, 2-deoxyglucose (2-DG), a glucose derivative that inhibits glycolysis [117], directly blocks hyperglycolysis by inhibiting HK activity, thereby suppressing the TGF-β1–induced fibrotic cell phenotype and subsequent PF in mice [48]. Additionally, curcumin has also been identified as a drug that targets glycolysis [118, 119]. Curcumin, a diketone derived from plant rhizomes, possesses anti-inflammatory, antimicrobial, and anticancer properties [119, 120]. Curcumin reportedly downregulates the expression and activity of HK, phosphofructokinase-2, glucose transporter type 4 (Glut4), and monocarboxylate transporter 4 (MCT4) in hepatic stellate cells [121]. Moreover, prior studies have confirmed the substantial protective effects of curcumin against glucose PD effluent–induced MMT and PF [122, 123]. These findings suggest that curcumin inhibits several steps in the glycolytic pathway [121]. Furthermore, certain microRNAs found in the peritoneum, such as microRNA-26a and microRNA-200a, target hyperglycolysis and fibrotic signaling in PMCs [48]. However, the effectiveness and safety of these methods require further investigations.
Targeting lipid metabolism
Recent studies have established that lipotoxicity mediates the progression of PF [39, 124]. Consequently, targeting lipid metabolism has become a key focus of current research. Medications aimed at improving lipid metabolism and identifying novel targets may represent new therapeutic strategies to slow the progression of PD-related fibrosis (Fig. 3).
Targeting lipid metabolism in PMCs. Potential therapeutic strategies include restoring and enhancing FAO using CPT1A agonists (C75), inhibiting lipid synthesis and uptake, and promoting cholesterol efflux using agents, such as rapamycin, rosiglitazone (a PPAR-γ agonist), and RAS blockers. FAO, fatty acid oxidation; PMC, peritoneal mesothelial cell; RAS, renin-angiotensin system
Targeting lipid deposition
Statins are lipid-lowering drugs that inhibit cholesterol synthesis by competitively inhibiting 3-hydroxy-3-methyl-glutaryl CoA (HMG-CoA) reductase, thereby blocking the conversion of HMG-CoA to methylglutarate [125]. Several statins, including atorvastatin, pitavastatin, and simvastatin, exhibit anti-fibrotic effects [126,127,128]. In both in vivo and in vitro PD models, statins inhibit MMT via the mevalonate pathway, thereby preserving peritoneal integrity and inhibiting PF [124]. Additionally, statins may alleviate peritoneal inflammation, fibrosis, and angiogenesis by modulating glutathione reductase activity and the TGF-β pathway [129]. However, prolonged use of statins may increase insulin resistance, disrupt lipid metabolism, and exacerbate inflammation and fibrosis [130, 131]. Moreover, simvastatin did not attenuate PF in a rat model of PD [132]. Thus, the anti-fibrotic effects of statins remain controversial, and further studies are required to determine their potential as therapeutic strategies in PD.
Rapamycin, a commonly used immunosuppressant targeting mTOR, has demonstrated anti-fibrotic effects and improves peritoneal membrane transport function in PD models [133, 134]. Recent studies have revealed its ability to inhibit intracellular lipid accumulation by blocking mTORC1 activity, thereby regulating lipid homeostasis and reversing low-density lipoprotein receptor (LDLr) dysfunction. Consequently, rapamycin exhibits a clear protective effect against lipid disorder-mediated PF [13, 135]. Furthermore, rapamycin can enhance intracellular lipid homeostasis by inhibiting cellular lipid uptake and increasing cholesterol efflux, thereby exerting a substantial protective effect against high-glucose PDF-induced fibrosis [66]. However, the safety and efficacy of rapamycin remain unclear.
PPARγ, an important nuclear receptor that controls the transcription of specific genes, is crucial in the interaction between lipid and glucose metabolism [136, 137]. Inhibiting PPARγ activity disrupts intracellular lipid efflux, leading to lipid droplet deposition and subsequent lipid disorder–mediated peritoneal injury and fibrosis [66]. Several studies using animal models of PD have revealed that the PPAR-γ agonist, rosiglitazone, protects peritoneal integrity and reduces PF by mitigating the accumulation of AGEs and inflammation [138, 139]. Additionally, PPARγ can inhibit the expression of GLUT1, thereby maintaining metabolic homeostasis and alleviating high-glucose PDF-induced PF [140, 141]. Given its dual role in both glucose and lipid metabolism, PPARγ stands out as a potentially ideal target for treating high-glucose-induced PF, providing further elucidation of its mechanism of action.
Abnormal activation of RAS has been implicated in lipid disorder-mediated PF [40, 76]. Specifically, RAS activation is crucial in regulating the LDLr pathway [40]. In vitro studies have shown that increased intracellular RAS activity impairs lipid homeostasis by disrupting the LDLr pathway and promoting ECM accumulation in HPMCs [40]. Moreover, AT2Rs ameliorate lipid disturbances and attenuate ECM accumulation in HPMCs by suppressing lectin-like oxidized LOX-1 [76]. Therefore, RAS blockers may maintain lipid homeostasis and protect against peritoneal injury.
EZH2 and Klotho also represent potential targets for modulating lipid metabolism in the treatment of PF [39, 142,143,144]. Specifically, EZH2 promotes lipid deposition and fibrosis, whereas Klotho is involved in lipid metabolism and possesses anti-fibrotic properties [39, 143, 144]. EZH2 inhibitors, such as GSK343 and 3-deazaneplanocin A, attenuate high-glucose-induced lipid deposition, MMT, angiogenesis, and PF [142, 145]. Furthermore, EZH2 inhibition mitigates lipid deposition and PF by suppressing EZH2 expression and restoring Klotho expression [39]. Klotho protects the peritoneum from PF by down-regulating the Wnt/β-catenin signaling pathway in a PD mouse model [144].
Targeting FAO restoration
Dysfunctional FAO in mesothelial cells promotes PD-associated PF; thus, enhancing FAO may be a therapeutic approach for PF [45]. The rate-limiting step in FAO is the translocation of long-chain FAs into mitochondria via CPT1A [69]. Notably, reduced expression of CPT1A has been associated with fibrosis [146, 147]. A recent study demonstrated that treatment of PD mice with a CPT1A activator (C75) restored FAO, reversed the pro-fibrotic phenotype in mesothelial cells, and reduced PF by upregulating CPT1A expression [45]. Consequently, CPT1A has emerged as a promising target for PF treatment. Alternatively, adipsin, stilbenes, and resveratrol metabolites are compounds known to enhance mitochondrial energy metabolism and increase FAO, potentially serving as therapeutic agents for PF [148,149,150]. However, further research is needed to validate their efficacy as PF treatments.
Targeting mitochondrial dysfunction
Preserving mitochondrial integrity may be a novel therapeutic approach for protecting the peritoneum from PD-induced fibrosis [23, 24]. For example, mitoTEMPO (an mtROS scavenger), BAY-117,085 (an NF-κB inhibitor), and resveratrol (an anti-inflammatory antioxidant) alleviate mitochondria-induced inflammation in mesothelial cells by protecting mitochondrial function [23]. Moreover, Wu et al. [151] found that proliferator-activated receptor-γ coactivator (PGC)-1α overexpression or metformin treatment can attenuate PF by preserving mitochondrial morphology and preventing damage to mitochondrial structure through activation of the AMPK-PGC-1α pathway. Astragalus total saponins (ATS) alleviate PF by promoting mitochondrial synthesis and inhibiting PMC apoptosis [152]. Alternatively, astaxanthin can eliminate glucose-induced mtROS in PMCs and inhibit MMT during PD, thereby demonstrating antioxidant and anti-inflammatory activities [153]. Furthermore, mitochonic acid-5 (MA-5) restored mitochondrial function, by inhibiting macrophage infiltration and oxidative stress, thus relieving PF in mice [154]. Ultimately, these findings suggest that targeting the mitochondria is a promising therapeutic approach for PF (Fig. 4).
Targeting mitochondrial function in PMCs. Strategies include using mitoTEMPO (a mitochondrial ROS scavenger), BAY-117,085 (an NF-κB inhibitor), and resveratrol (the natural anti-inflammatory antioxidant) to protect and reduce mitochondrial damage. Alternatively, ATS promotes mitochondrial synthesis, while AST and MA-5 inhibit mitochondrial oxidative stress, protecting mitochondrial function. AST, astaxanthin; ATS, astragalus total saponins
Discussion
The metabolic reprogramming of PMCs is a crucial event in the progression of PD-associated PF. Initially serving as an adaptive response to counteract damaging effects of high-glucose PDFs, metabolic pattern shifts caused by high glucose can lead to metabolic disorders, resulting in cellular damage, inflammation, and eventual cell death, forming the so-called “honey trap.” Consequently, long-term exposure to hyperglycemic environments induces sustained metabolic reprogramming, trapping PMCs in the “honey trap,” impairing the cells’ ability to adapt to physiological conditions and ultimately resulting in chronic peritoneal inflammation and fibrosis.
Despite increasing understanding, substantial breakthroughs in the prevention and treatment of PF remain elusive. Recent studies have highlighted the correlation between PMC metabolic reprogramming and PD-related fibrosis, offering a novel direction for the treatment of PF. Nonetheless, several challenges remain in the development of effective therapeutic strategies for PF. A primary concern is the predominant focus on PMCs in current research, leaving uncertainties regarding potential metabolic alterations in other peritoneal cells types during PD, such as fibroblasts, macrophages, endothelial cells, T cells, and adipocytes. Therefore, exploring the role and mechanisms of metabolic dysfunction in the entire peritoneum in future clinical and translational research, including investigating the role of metabolic reprogramming in other peritoneal cells during PD treatment, is crucial.
Moreover, metabolic reprogramming is a complex process that involves multiple metabolic pathways. While existing studies predominantly investigating PF have focused on glucose and lipid metabolism, the mechanisms and regulation of other metabolic processes, such as amino acid metabolism, ketone body metabolism, and the PPP, remain unclear. Clarifying the specific interactions between multiple these metabolic processes also require further elucidation. Finally, while interventions targeting metabolism have shown promise in animal and cellular experiments, clinical translation of these strategies remains limited. Furthermore, some of the existing clinical drugs possess potential adverse and off-target effects. Therefore, the clinical application of drugs and compounds that target metabolism requires further investigation and validation to ensure their safety and efficacy in PF management.
Conclusions and perspectives
The metabolic pathways discussed in this paper represent novel endogenous targets closely related to PMC homeostasis and PD-associated fibrosis. While previous research predominantly focused on cellular MMT, apoptosis, and death, recent studies have shifted toward investigating the metabolic mechanisms of PMCs. Notably, the balance between glucose metabolism, lipid metabolism, and mitochondrial respiration is increasingly recognized as crucial for cellular energy production and homeostasis maintenance [53]. Notably, preclinical studies investigating several anti-fibrotic drugs targeting metabolic pathways are currently underway [155], sparking a growing interest in exploring the restoration of metabolic homeostasis in mesothelial cells as a potential therapeutic approach for PD-associated fibrosis. Although recent studies have identified a strong relationship between PMC metabolism and PF, research on metabolic reprogramming in PMCs is still in its early stages. In conclusion, ongoing research into metabolic reprogramming offers a promising avenue for enhancing our understanding of the mechanisms driving PD-related fibrosis and may ultimately lead to the development of novel therapeutic strategies aimed at treating PF.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- PD:
-
Peritoneal dialysis
- SRD:
-
End-stage renal disease
- PMCs:
-
Peritoneal mesothelial cells
- PDFs:
-
Peritoneal dialysis fluids
- AGE:
-
Advanced glycation end products
- MMT:
-
Mesothelial–mesenchymal transition
- OXPHOS:
-
Oxidative phosphorylation
- RAS:
-
Renin-angiotensin system
- GLUT-1:
-
Glucose transporter type 1
- NADH:
-
Nicotinamide adenine dinucleotide
- NAD+:
-
Nicotinamide adenine dinucleotide
- NADH/NAD+:
-
Nicotinamide adenine dinucleotide and nicotinamide adenine dinucleotide ratio
- HIF-1:
-
Hypoxia-inducible factor-1
- TCA:
-
Tricarboxylic acid
- mtROS:
-
mitochondrial reactive oxygen species
- AR:
-
Aldose reductase
- G6P:
-
Glucose-6-phosphate
- HK:
-
Hexokinase
- ROS:
-
Reactive oxygen species
- FFAs:
-
Free fatty acids
- FAs:
-
Fatty acids
- AT2R:
-
Angiotensin II type 2 receptor
- mTORC1:
-
mTOR complex 1
- SCAP/SREBP-2:
-
Sterol-regulatory element-binding protein-2/cleavage-activating protein pathway
- HPMCs:
-
Human peritoneal mesothelial cells
- EZH2:
-
Enhancer of zeste homolog 2
- LOX-1:
-
Lectin-like oxidized lipoprotein receptor-1
- PPARγ:
-
Peroxisome proliferator-activated receptor γ
- KB:
-
Ketone body
- PPP:
-
Pentose phosphate pathway
- SGLT-2:
-
Sodium glucose cotransporter-2
- DPP-4:
-
Dipeptidyl peptidase-4
- GLP-1:
-
Glucagon-like peptide-1
- PDH:
-
Pyruvate dehydrogenase
- DCA:
-
Dichloroacetic acid
- 2-DG:
-
2-Deoxyglucose
- CG:
-
Chlorhexidine gluconate
- Glut4:
-
Glucose transporter type 4
- MCT4:
-
Monocarboxylate transporter 4
- HMG-CoA:
-
3-hydroxy-3-methyl-glutaryl CoA
- LDL:
-
Low-density lipoprotein
- LDLr:
-
Low-density lipoprotein receptor
- ATS:
-
Astragalus total saponins
- AST:
-
Astaxanthin
- MA-5:
-
Mitochonic acid-5
- TGF-β1:
-
Transforming growth factor beta 1
- PGC:
-
Peroxisome proliferator-activated receptor-γ coactivator
- ECM:
-
Extracellular matrix
- FAO:
-
Fatty acid oxidation
References
Collaboration GBDCKD. Global, regional, and national burden of chronic kidney disease, 1990–2017: a systematic analysis for the global burden of Disease Study 2017. Lancet. 2020;395:709–33.
Liyanage T, Ninomiya T, Jha V, Neal B, Patrice HM, Okpechi I, et al. Worldwide access to treatment for end-stage kidney disease: a systematic review. Lancet. 2015;385:1975–82.
Cho Y, Bello AK, Levin A, Lunney M, Osman MA, Ye F, et al. Peritoneal Dialysis use and practice patterns: an International Survey Study. Am J Kidney Dis. 2021;77:315–25.
Bello AK, Okpechi IG, Osman MA, Cho Y, Cullis B, Htay H, et al. Epidemiology of peritoneal dialysis outcomes. Nat Rev Nephrol. 2022;18:779–93.
Wang Y, Zhang Y, Ma M, Zhuang X, Lu Y, Miao L, et al. Mechanisms underlying the involvement of peritoneal macrophages in the pathogenesis and novel therapeutic strategies for dialysis-induced peritoneal fibrosis. Front Immunol. 2024;15:1507265.
Ito Y, Sun T, Tawada M, Kinashi H, Yamaguchi M, Katsuno T et al. Pathophysiological mechanisms of peritoneal fibrosis and peritoneal membrane dysfunction in peritoneal Dialysis. Int J Mol Sci. 2024; 25.
Masola V, Bonomini M, Borrelli S, Di Liberato L, Vecchi L, Onisto M et al. Fibrosis of Peritoneal Membrane as target of New therapies in Peritoneal Dialysis. Int J Mol Sci. 2022; 23.
Jose MD, Johnson DW, Mudge DW, Tranaeus A, Voss D, Walker R, et al. Peritoneal dialysis practice in Australia and New Zealand: a call to action. Nephrol (Carlton). 2011;16:19–29.
Wu C, Chen X, Ying Wang A, Chen J, Gao H, Li G, et al. Peritoneal dialysis in Sichuan Province of China - report from the Chinese National Renal Data System. Ren Fail. 2018;40:577–82.
Li J, Liu Y, Liu J. A review of research progress on mechanisms of peritoneal fibrosis related to peritoneal dialysis. Front Physiol. 2023;14:1220450.
Suryantoro SD, Thaha M, Sutanto H, Firdausa S. Current insights into Cellular determinants of Peritoneal Fibrosis in Peritoneal Dialysis: a narrative review. J Clin Med. 2023; 12.
Song Q, Wang P, Wang H, Pan M, Li X, Yao Z, et al. Integrative analysis of chromatin accessibility and transcriptome landscapes in the induction of peritoneal fibrosis by high glucose. J Transl Med. 2024;22:243.
Zhao H, Zhang HL, Jia L. High glucose dialysate-induced peritoneal fibrosis: pathophysiology, underlying mechanisms and potential therapeutic strategies. Biomed Pharmacother. 2023;165:115246.
Krediet RT, Parikova A. Glucose-induced pseudohypoxia and advanced glycosylation end products explain peritoneal damage in long-term peritoneal dialysis. Perit Dial Int. 2024;44:6–15.
Gensberger-Reigl S, Weigel I, Stutzer J, Auditore A, Nikolaus T, Pischetsrieder M. Degradation and de novo formation of nine major glucose degradation products during storage of peritoneal dialysis fluids. Sci Rep. 2022;12:4268.
Lui SL, Yung S, Yim A, Wong KM, Tong KL, Wong KS, et al. A combination of biocompatible peritoneal dialysis solutions and residual renal function, peritoneal transport, and inflammation markers: a randomized clinical trial. Am J Kidney Dis. 2012;60:966–75.
Balzer MS, Rong S, Nordlohne J, Zemtsovski JD, Schmidt S, Stapel B et al. SGLT2 inhibition by Intraperitoneal Dapagliflozin mitigates peritoneal fibrosis and Ultrafiltration failure in a mouse model of chronic peritoneal exposure to High-Glucose Dialysate. Biomolecules. 2020; 10.
Kastelein AW, Vos LMC, de Jong KH, van Baal J, Nieuwland R, van Noorden CJF, et al. Embryology, anatomy, physiology and pathophysiology of the peritoneum and the peritoneal vasculature. Semin Cell Dev Biol. 2019;92:27–36.
Capobianco A, Cottone L, Monno A, Manfredi AA, Rovere-Querini P. The peritoneum: healing, immunity, and diseases. J Pathol. 2017;243:137–47.
Su H, Zou R, Su J, Chen X, Yang H, An N, et al. Sterile inflammation of peritoneal membrane caused by peritoneal dialysis: focus on the communication between immune cells and peritoneal stroma. Front Immunol. 2024;15:1387292.
Zhang J, Chen Y, Chen T, Miao B, Tang Z, Hu X, et al. Single-cell transcriptomics provides new insights into the role of fibroblasts during peritoneal fibrosis. Clin Transl Med. 2021;11:e321.
Krediet RT. Aging of the peritoneal Dialysis membrane. Front Physiol. 2022;13:885802.
Ramil-Gomez O, Lopez-Pardo M, Fernandez-Rodriguez JA, Rodriguez-Carmona A, Perez-Lopez T, Vaamonde-Garcia C et al. Involvement of mitochondrial dysfunction in the inflammatory response in human mesothelial cells from peritoneal Dialysis Effluent. Antioxidants (Basel). 2022; 11.
Ramil-Gomez O, Rodriguez-Carmona A, Fernandez-Rodriguez JA, Perez-Fontan M, Ferreiro-Hermida T, Lopez-Pardo M et al. Mitochondrial dysfunction plays a relevant role in pathophysiology of peritoneal membrane damage Induced by Peritoneal Dialysis. Antioxid (Basel). 2021; 10.
Krediet RT. Physiology of peritoneal dialysis; pathophysiology in long-term patients. Front Physiol. 2024;15:1322493.
Xie S, Xu F, Lu Y, Zhang Y, Li X, Yu M, et al. Elabela attenuates the TGF-beta1-Induced epithelial-mesenchymal transition of peritoneal mesothelial cells in patients receiving peritoneal Dialysis. Front Pharmacol. 2022;13:890881.
Bonomini M, Masola V, Procino G, Zammit V, Divino-Filho JC, Arduini A et al. How to Improve the Biocompatibility of Peritoneal Dialysis Solutions (without Jeopardizing the Patient’s Health). Int J Mol Sci. 2021; 22.
Diao X, Zhan C, Ye H, Wu H, Yi C, Lin J, et al. Single-cell transcriptomic reveals the peritoneal microenvironmental change in long-term peritoneal dialysis patients with ultrafiltration failure. iScience. 2024;27:111383.
Yu Z, Lambie M, Chess J, Williams A, Do JY, Topley N, et al. Peritoneal protein clearance is a function of local inflammation and membrane area whereas systemic inflammation and Comorbidity Predict Survival of Incident Peritoneal Dialysis patients. Front Physiol. 2019;10:105.
Law S, Davenport A. The effect of glucose absorption from peritoneal dialysates on changes in lipid profiles in prevalent peritoneal dialysis patients. Perit Dial Int. 2021;41:115–7.
DelNero P, Hopkins BD, Cantley LC, Fischbach C. Cancer metabolism gets physical. Sci Transl Med. 2018; 10.
Martinez-Reyes I, Chandel NS. Cancer metabolism: looking forward. Nat Rev Cancer. 2021;21:669–80.
Urbano AM. Otto Warburg: the journey towards the seminal discovery of tumor cell bioenergetic reprogramming. Biochim Biophys Acta Mol Basis Dis. 2021;1867:165965.
Li Z, Lu S, Li X. The role of metabolic reprogramming in tubular epithelial cells during the progression of acute kidney injury. Cell Mol Life Sci. 2021;78:5731–41.
Zhu Z, Hu J, Chen Z, Feng J, Yang X, Liang W, et al. Transition of acute kidney injury to chronic kidney disease: role of metabolic reprogramming. Metabolism. 2022;131:155194.
Ge T, Gu X, Jia R, Ge S, Chai P, Zhuang A, et al. Crosstalk between metabolic reprogramming and epigenetics in cancer: updates on mechanisms and therapeutic opportunities. Cancer Commun (Lond). 2022;42:1049–82.
Yang T, Wei B, Liu J, Si X, Wang L, Jiang C. A landscape of metabolic variation among clinical outcomes of peritoneal dialysis in end-stage renal disease. Clin Chim Acta. 2024;555:117826.
Liu J, Feng Y, Sun C, Zhu W, Zhang QY, Jin B, et al. Valsartan ameliorates high glucose-induced peritoneal fi brosis by blocking mTORC1 signaling. Exp Biol Med (Maywood). 2020;245:983–93.
Wang Q, Sun J, Wang R, Sun J. Inhibition of EZH2 mitigates peritoneal fibrosis and lipid precipitation in peritoneal mesothelial cells mediated by klotho. Ren Fail. 2023;45:2149411.
Liu J, Feng Y, Li N, Shao QY, Zhang QY, Sun C, et al. Activation of the RAS contributes to peritoneal fibrosis via dysregulation of low-density lipoprotein receptor. Am J Physiol Ren Physiol. 2021;320:F273–84.
Kariya T, Nishimura H, Mizuno M, Suzuki Y, Matsukawa Y, Sakata F, et al. TGF-beta1-VEGF-A pathway induces neoangiogenesis with peritoneal fibrosis in patients undergoing peritoneal dialysis. Am J Physiol Ren Physiol. 2018;314:F167–80.
Zhang Z, Jiang N, Ni Z. Strategies for preventing peritoneal fibrosis in peritoneal dialysis patients: new insights based on peritoneal inflammation and angiogenesis. Front Med. 2017;11:349–58.
Krediet RT, Parikova A. Relative contributions of Pseudohypoxia and inflammation to peritoneal alterations with long-term peritoneal Dialysis patients. Clin J Am Soc Nephrol. 2022;17:1259–66.
Krediet RT. Acquired decline in Ultrafiltration in Peritoneal Dialysis: the role of glucose. J Am Soc Nephrol. 2021;32:2408–15.
Su W, Hu Z, Zhong X, Cong A, Zhang Y, Zhou Z, et al. Restoration of CPT1A-mediated fatty acid oxidation in mesothelial cells protects against peritoneal fibrosis. Theranostics. 2023;13:4482–96.
Mutsaers SE, Prele CM, Pengelly S, Herrick SE. Mesothelial cells and peritoneal homeostasis. Fertil Steril. 2016;106:1018–24.
Wiley CD, Campisi J. The metabolic roots of senescence: mechanisms and opportunities for intervention. Nat Metab. 2021;3:1290–301.
Si M, Wang Q, Li Y, Lin H, Luo D, Zhao W et al. Inhibition of hyperglycolysis in mesothelial cells prevents peritoneal fibrosis. Sci Transl Med. 2019; 11.
Hu W, Li G, Dong W, He P, Liu W, Wu Y, et al. Single-cell sequencing reveals peritoneal environment and insights into fibrosis in CAPD patients. iScience. 2023;26:106336.
Wang L, Balzer MS, Rong S, Menne J, von Vietinghoff S, Dong L, et al. Protein kinase C alpha inhibition prevents peritoneal damage in a mouse model of chronic peritoneal exposure to high-glucose dialysate. Kidney Int. 2016;89:1253–67.
Song J, Yang X, Yan LJ. Role of pseudohypoxia in the pathogenesis of type 2 diabetes. Hypoxia (Auckl). 2019;7:33–40.
Wang J, Lv X, AS AN-W, Tian SS, Wang JM, Liu HY, et al. Canagliflozin alleviates high glucose-induced peritoneal fibrosis via HIF-1alpha inhibition. Front Pharmacol. 2023;14:1152611.
Kierans SJ, Taylor CT. Regulation of glycolysis by the hypoxia-inducible factor (HIF): implications for cellular physiology. J Physiol. 2021;599:23–37.
Balzer MS. Molecular pathways in peritoneal fibrosis. Cell Signal. 2020;75:109778.
Vercellino I, Sazanov LA. The assembly, regulation and function of the mitochondrial respiratory chain. Nat Rev Mol Cell Biol. 2022;23:141–61.
Chan DC. Mitochondrial dynamics and its involvement in Disease. Annu Rev Pathol. 2020;15:235–59.
MacVicar T, Ohba Y, Nolte H, Mayer FC, Tatsuta T, Sprenger HG, et al. Lipid signalling drives proteolytic rewiring of mitochondria by YME1L. Nature. 2019;575:361–5.
Chen JB, Lin TK, Liou CW, Liao SC, Lee LC, Wang PW, et al. Correlation of oxidative stress biomarkers and peritoneal urea clearance with mitochondrial DNA copy number in continuous ambulatory peritoneal dialysis patients. Am J Nephrol. 2008;28:853–9.
Oberacker T, Fritz P, Schanz M, Alscher MD, Ketteler M, Schricker S. Enhanced oxidative DNA-Damage in peritoneal Dialysis patients via the TXNIP/TRX Axis. Antioxid (Basel). 2022; 11.
Hung KY, Liu SY, Yang TC, Liao TL, Kao SH. High-dialysate-glucose-induced oxidative stress and mitochondrial-mediated apoptosis in human peritoneal mesothelial cells. Oxid Med Cell Longev. 2014; 2014: 642793.
van Westrhenen R, Aten J, Aberra M, Dragt CA, Deira G, Krediet RT. Effects of inhibition of the polyol pathway during chronic peritoneal exposure to a dialysis solution. Perit Dial Int. 2005;25(Suppl 3):S18–21.
Wong TY, Phillips AO, Witowski J, Topley N. Glucose-mediated induction of TGF-beta 1 and MCP-1 in mesothelial cells in vitro is osmolality and polyol pathway dependent. Kidney Int. 2003;63:1404–16.
Sano H, Nakamura A, Yamane M, Niwa H, Nishimura T, Araki K, et al. The polyol pathway is an evolutionarily conserved system for sensing glucose uptake. PLoS Biol. 2022;20:e3001678.
Garg SS, Gupta J. Polyol pathway and redox balance in diabetes. Pharmacol Res. 2022;182:106326.
Bacha MM, Nadeem H, Zaib S, Sarwar S, Imran A, Rahman SU, et al. Rhodanine-3-acetamide derivatives as aldose and aldehyde reductase inhibitors to treat diabetic complications: synthesis, biological evaluation, molecular docking and simulation studies. BMC Chem. 2021;15:28.
Liu J, Jiang CM, Feng Y, Zhu W, Jin B, Xia YY, et al. Rapamycin inhibits peritoneal fibrosis by modifying lipid homeostasis in the peritoneum. Am J Transl Res. 2019;11:1473–85.
Li Y, Huang X, Yang G, Xu K, Yin Y, Brecchia G, et al. CD36 favours fat sensing and transport to govern lipid metabolism. Prog Lipid Res. 2022;88:101193.
Kawamura S, Matsushita Y, Kurosaki S, Tange M, Fujiwara N, Hayata Y et al. Inhibiting SCAP/SREBP exacerbates liver injury and carcinogenesis in murine nonalcoholic steatohepatitis. J Clin Invest. 2022; 132.
Schlaepfer IR, Joshi M. CPT1A-mediated Fat Oxidation, mechanisms, and therapeutic potential. Endocrinology. 2020; 161.
Luo J, Yang H, Song BL. Mechanisms and regulation of cholesterol homeostasis. Nat Rev Mol Cell Biol. 2020;21:225–45.
Feng L, Gu C, Li Y, Huang J. High glucose promotes CD36 expression by upregulating peroxisome proliferator-activated receptor gamma levels to exacerbate lipid deposition in renal tubular cells. Biomed Res Int. 2017;2017:1414070.
Adeva-Andany MM, Carneiro-Freire N, Seco-Filgueira M, Fernandez-Fernandez C, Mourino-Bayolo D. Mitochondrial beta-oxidation of saturated fatty acids in humans. Mitochondrion. 2019;46:73–90.
Zhao M, Wang Y, Li L, Liu S, Wang C, Yuan Y, et al. Mitochondrial ROS promote mitochondrial dysfunction and inflammation in ischemic acute kidney injury by disrupting TFAM-mediated mtDNA maintenance. Theranostics. 2021;11:1845–63.
Fujieda Y, Manno A, Hayashi Y, Rhodes N, Guo L, Arita M, et al. Inflammation and resolution are associated with upregulation of fatty acid beta-oxidation in Zymosan-induced peritonitis. PLoS ONE. 2013;8:e66270.
Liu Q, Deng C, Xing X, Hu Y, Wang Z, Liang Y. Silencing RIPK1/mTORC1 signalling attenuated the inflammation and oxidative stress in diabetic cardiomyopathy. Exp Cell Res. 2023;422:113417.
Liu J, Jin B, Lu J, Feng Y, Li N, Wan C, et al. Angiotensin II type 2 receptor prevents extracellular matrix accumulation in human peritoneal mesothelial cell by ameliorating lipid disorder via LOX-1 suppression. Ren Fail. 2022;44:1687–97.
Benador IY, Veliova M, Liesa M, Shirihai OS. Mitochondria bound to lipid droplets: where mitochondrial dynamics regulate lipid storage and utilization. Cell Metab. 2019;29:827–35.
Cavaliere G, Cimmino F, Trinchese G, Catapano A, Petrella L, D’Angelo M et al. From obesity-Induced Low-Grade inflammation to Lipotoxicity and mitochondrial dysfunction: altered multi-crosstalk between adipose tissue and metabolically active organs. Antioxidants (Basel). 2023; 12.
Ge M, Fontanesi F, Merscher S, Fornoni A. The vicious cycle of renal lipotoxicity and mitochondrial dysfunction. Front Physiol. 2020;11:732.
Ma XM, Geng K, Law BY, Wang P, Pu YL, Chen Q, et al. Lipotoxicity-induced mtDNA release promotes diabetic cardiomyopathy by activating the cGAS-STING pathway in obesity-related diabetes. Cell Biol Toxicol. 2023;39:277–99.
Zheng Y, Yao Y, Ge T, Ge S, Jia R, Song X, et al. Amino acid metabolism reprogramming: shedding new light on T cell anti-tumor immunity. J Exp Clin Cancer Res. 2023;42:291.
Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017;25:262–84.
Tang Y, Li W, Qiu L, Zhang X, Zhang L, Miyagishi M, et al. The p52-ZER6/G6PD axis alters aerobic glycolysis and promotes tumor progression by activating the pentose phosphate pathway. Oncogenesis. 2023;12:17.
TeSlaa T, Ralser M, Fan J, Rabinowitz JD. The pentose phosphate pathway in health and disease. Nat Metab. 2023;5:1275–89.
Peiro C, Romacho T, Azcutia V, Villalobos L, Fernandez E, Bolanos JP, et al. Inflammation, glucose, and vascular cell damage: the role of the pentose phosphate pathway. Cardiovasc Diabetol. 2016;15:82.
Chen L, Zhang Z, Hoshino A, Zheng HD, Morley M, Arany Z, et al. NADPH production by the oxidative pentose-phosphate pathway supports folate metabolism. Nat Metab. 2019;1:404–15.
Iyasere O, Nagar R, Jesus-Silva JA, Pepereke S, MacConaill K, Eid A, et al. The impact of amino acid dialysate on anthropometric measures in adult patients on peritoneal dialysis: a systematic review and meta-analysis. Perit Dial Int. 2022;42:314–23.
Chan TM, Leung JK, Sun Y, Lai KN, Tsang RC, Yung S. Different effects of amino acid-based and glucose-based dialysate from peritoneal dialysis patients on mesothelial cell ultrastructure and function. Nephrol Dial Transpl. 2003;18:1086–94.
Tjiong HL, Zijlstra FJ, Rietveld T, Wattimena JL, Huijmans JG, Swart GR et al. Peritoneal protein losses and cytokine generation in automated peritoneal dialysis with combined amino acids and glucose solutions. Mediators Inflamm. 2007; 2007: 97272.
Herzog R, Bartosova M, Tarantino S, Wagner A, Unterwurzacher M, Sacnun JM et al. Peritoneal Dialysis fluid supplementation with Alanyl-Glutamine attenuates conventional Dialysis fluid-mediated endothelial cell Injury by restoring perturbed cytoprotective responses. Biomolecules. 2020; 10.
Kratochwill K, Boehm M, Herzog R, Gruber K, Lichtenauer AM, Kuster L, et al. Addition of Alanyl-glutamine to Dialysis Fluid restores peritoneal Cellular stress responses - A First-In-Man Trial. PLoS ONE. 2016;11:e0165045.
Yoo HC, Yu YC, Sung Y, Han JM. Glutamine reliance in cell metabolism. Exp Mol Med. 2020;52:1496–516.
Nelson AB, Queathem ED, Puchalska P, Crawford PA. Metabolic messengers: ketone bodies. Nat Metab. 2023;5:2062–74.
Puchalska P, Crawford PA. Metabolic and Signaling Roles of Ketone Bodies in Health and Disease. Annu Rev Nutr. 2021;41:49–77.
Kadir AA, Stubbs BJ, Chong CR, Lee H, Cole M, Carr C, et al. On the interdependence of ketone body oxidation, glycogen content, glycolysis and energy metabolism in the heart. J Physiol. 2023;601:1207–24.
Debray-Garcia Y, Sanchez EI, Rodriguez-Munoz R, Venegas MA, Velazquez J, Reyes JL. Diabetes and exposure to peritoneal dialysis solutions alter tight junction proteins and glucose transporters of rat peritoneal mesothelial cells. Life Sci. 2016;161:78–89.
Schricker S, Oberacker T, Fritz P, Ketteler M, Alscher MD, Schanz M. Peritoneal expression of SGLT-2, GLUT1, and GLUT3 in peritoneal Dialysis patients. Kidney Blood Press Res. 2022;47:125–34.
Ceballos J, Schwalfenberg M, Karageorgis G, Reckzeh ES, Sievers S, Ostermann C, et al. Synthesis of Indomorphan Pseudo-natural product inhibitors of glucose transporters GLUT-1 and– 3. Angew Chem Int Ed Engl. 2019;58:17016–25.
Zhou Y, Fan J, Zheng C, Yin P, Wu H, Li X, et al. SGLT-2 inhibitors reduce glucose absorption from peritoneal dialysis solution by suppressing the activity of SGLT-2. Biomed Pharmacother. 2019;109:1327–38.
Shi P, Zhan Z, Ye X, Lu Y, Song K, Sheng F, et al. The antioxidative effects of empagliflozin on high glucose–induced epithelial-mesenchymal transition in peritoneal mesothelial cells via the Nrf2/HO-1 signaling. Ren Fail. 2022;44:1528–42.
Shentu Y, Li Y, Xie S, Jiang H, Sun S, Lin R, et al. Empagliflozin, a sodium glucose cotransporter-2 inhibitor, ameliorates peritoneal fibrosis via suppressing TGF-beta/Smad signaling. Int Immunopharmacol. 2021;93:107374.
Martus G, Bergling K, de Arteaga J, Oberg CM. SGLT2 inhibition does not reduce glucose absorption during experimental peritoneal dialysis. Perit Dial Int. 2021;41:373–80.
Yaribeygi H, Maleki M, Reiner Z, Jamialahmadi T, Sahebkar A. Mechanistic view on the effects of SGLT2 inhibitors on lipid metabolism in Diabetic Milieu. J Clin Med. 2022; 11.
Szekeres Z, Toth K, Szabados E. The Effects of SGLT2 Inhibitors on Lipid Metabolism. Metabolites. 2021; 11.
Yaribeygi H, Maleki M, Butler AE, Jamialahmadi T, Sahebkar A. New insights into cellular links between sodium-glucose cotransporter-2 inhibitors and ketogenesis. J Cell Biochem. 2022;123:1879–90.
Yaribeygi H, Maleki M, Butler AE, Jamialahmadi T, Sahebkar A. Sodium-glucose cotransporter 2 inhibitors and mitochondrial functions: state of the art. EXCLI J. 2023;22:53–66.
Mulder S, Hammarstedt A, Nagaraj SB, Nair V, Ju W, Hedberg J, et al. A metabolomics-based molecular pathway analysis of how the sodium-glucose co-transporter-2 inhibitor dapagliflozin may slow kidney function decline in patients with diabetes. Diabetes Obes Metab. 2020;22:1157–66.
Alosaimi ME, Alotaibi BS, Abduljabbar MH, Alnemari RM, Almalki AH, Serag A. Therapeutic implications of dapagliflozin on the metabolomics profile of diabetic rats: a GC-MS investigation coupled with multivariate analysis. J Pharm Biomed Anal. 2024;242:116018.
Love KM, Liu Z. DPP4 activity, Hyperinsulinemia, and atherosclerosis. J Clin Endocrinol Metab. 2021;106:1553–65.
Gallwitz B. Clinical use of DPP-4 inhibitors. Front Endocrinol (Lausanne). 2019;10:389.
Tomovic K, Lazarevic J, Kocic G, Deljanin-Ilic M, Anderluh M, Smelcerovic A. Mechanisms and pathways of anti-inflammatory activity of DPP-4 inhibitors in cardiovascular and renal protection. Med Res Rev. 2019;39:404–22.
Li YC, Sung PH, Yang YH, Chiang JY, Yip HK, Yang CC. Dipeptidyl peptidase 4 promotes peritoneal fibrosis and its inhibitions prevent failure of peritoneal dialysis. Commun Biol. 2021;4:144.
Goodwin J, Choi H, Hsieh MH, Neugent ML, Ahn JM, Hayenga HN, et al. Targeting hypoxia-inducible Factor-1alpha/Pyruvate dehydrogenase kinase 1 Axis by Dichloroacetate suppresses bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. 2018;58:216–31.
Tian L, Wu D, Dasgupta A, Chen KH, Mewburn J, Potus F, et al. Epigenetic metabolic reprogramming of right ventricular fibroblasts in pulmonary arterial hypertension: a pyruvate dehydrogenase kinase-dependent shift in mitochondrial metabolism promotes right ventricular fibrosis. Circ Res. 2020;126:1723–45.
Wei Q, Su J, Dong G, Zhang M, Huo Y, Dong Z. Glycolysis inhibitors suppress renal interstitial fibrosis via divergent effects on fibroblasts and tubular cells. Am J Physiol Ren Physiol. 2019;316:F1162–72.
Virmani MA, Cirulli M. The role of l-Carnitine in Mitochondria, Prevention of metabolic inflexibility and disease initiation. Int J Mol Sci. 2022; 23.
Pajak B, Siwiak E, Soltyka M, Priebe A, Zielinski R, Fokt I et al. 2-Deoxy-d-Glucose and its analogs: from Diagnostic to Therapeutic agents. Int J Mol Sci. 2019; 21.
Jo SL, Yang H, Lee SR, Heo JH, Lee HW, Hong EJ. Curcumae Radix Decreases Neurodegenerative Markers through Glycolysis Decrease and TCA Cycle Activation. Nutrients. 2022; 14.
Nelson KM, Dahlin JL, Bisson J, Graham J, Pauli GF, Walters MA. The essential Medicinal Chemistry of Curcumin. J Med Chem. 2017;60:1620–37.
Farzaei MH, Zobeiri M, Parvizi F, El-Senduny FF, Marmouzi I, Coy-Barrera E et al. Curcumin in Liver diseases: a systematic review of the Cellular mechanisms of oxidative stress and clinical perspective. Nutrients. 2018; 10.
Lian N, Jiang Y, Zhang F, Jin H, Lu C, Wu X, et al. Curcumin regulates cell fate and metabolism by inhibiting hedgehog signaling in hepatic stellate cells. Lab Invest. 2015;95:790–803.
Zhao JL, Guo MZ, Zhu JJ, Zhang T, Min DY. Curcumin suppresses epithelial-to-mesenchymal transition of peritoneal mesothelial cells (HMrSV5) through regulation of transforming growth factor-activated kinase 1 (TAK1). Cell Mol Biol Lett. 2019;24:32.
Zhao JL, Zhang T, Shao X, Zhu JJ, Guo MZ. Curcumin ameliorates peritoneal fibrosis via inhibition of transforming growth factor-activated kinase 1 (TAK1) pathway in a rat model of peritoneal dialysis. BMC Complement Altern Med. 2019;19:280.
Chang TI, Kang HY, Kim KS, Lee SH, Nam BY, Paeng J, et al. The effect of statin on epithelial-mesenchymal transition in peritoneal mesothelial cells. PLoS ONE. 2014;9:e109628.
Sirtori CR. The pharmacology of statins. Pharmacol Res. 2014;88:3–11.
Bosch J, Gracia-Sancho J, Abraldes JG. Cirrhosis as new indication for statins. Gut. 2020;69:953–62.
Kuo HF, Hsieh CC, Wang SC, Chang CY, Hung CH, Kuo PL et al. Simvastatin attenuates Cardiac Fibrosis via Regulation of Cardiomyocyte-Derived Exosome Secretion. J Clin Med. 2019; 8.
Elbaset MA, Mohamed B, Hessin A, Abd El-Rahman SS, Esatbeyoglu T, Afifi SM, et al. Nrf2/HO-1, NF-kappaB and PI3K/Akt signalling pathways decipher the therapeutic mechanism of pitavastatin in early phase liver fibrosis in rats. J Cell Mol Med. 2024;28:e18116.
Yenicerioglu Y, Uzelce O, Akar H, Kolatan E, Yilmaz O, Yenisey C, et al. Effects of atorvastatin on development of peritoneal fibrosis in rats on peritoneal dialysis. Ren Fail. 2010;32:1095–102.
Andreikos D, Karampitsakos T, Tzouvelekis A, Stratakos G. Statins’ still controversial role in pulmonary fibrosis: what does the evidence show? Pulm Pharmacol Ther. 2022;77:102168.
Huang TS, Wu T, Wu YD, Li XH, Tan J, Shen CH, et al. Long-term statins administration exacerbates diabetic nephropathy via ectopic fat deposition in diabetic mice. Nat Commun. 2023;14:390.
Baroni G, Schuinski AF, Berticelli PT, Silva MA, Gouveia DS, Pecoits Filho R, et al. The influence of simvastatin in induced peritoneal fibrosis in rats by peritoneal dialysis solution with glucosis 4.25%. Acta Cir Bras. 2012;27:350–6.
Xu T, Xie JY, Wang WM, Ren H, Chen N. Impact of rapamycin on peritoneal fibrosis and transport function. Blood Purif. 2012;34:48–57.
Xiang S, Li M, Xie X, Xie Z, Zhou Q, Tian Y, et al. Rapamycin inhibits epithelial-to-mesenchymal transition of peritoneal mesothelium cells through regulation of rho GTPases. FEBS J. 2016;283:2309–25.
Liu J, Zhu W, Jiang CM, Feng Y, Xia YY, Zhang QY, et al. Mammalian target of Rapamycin Complex 1 activation disrupts the low-density lipoprotein receptor pathway: a novel mechanism for Extracellular Matrix Accumulation in Human Peritoneal Mesothelial cells. Am J Nephrol. 2018;48:357–68.
Marion-Letellier R, Savoye G, Ghosh S. Fatty acids, eicosanoids and PPAR gamma. Eur J Pharmacol. 2016;785:44–9.
Hu W, Jiang C, Kim M, Xiao Y, Richter HJ, Guan D, et al. Isoform-specific functions of PPARgamma in gene regulation and metabolism. Genes Dev. 2022;36:300–12.
Zhang YF, Wang Q, Su YY, Wang JL, Hua BJ, Yang S, et al. PPAR–gamma agonist rosiglitazone protects rat peritoneal mesothelial cells against peritoneal dialysis solution–induced damage. Mol Med Rep. 2017;15:1786–92.
Zhang YF, Zou XL, Wu J, Yu XQ, Yang X. Rosiglitazone, a peroxisome proliferator-activated receptor (PPAR)-gamma agonist, attenuates inflammation Via NF-kappaB inhibition in Lipopolysaccharide-Induced Peritonitis. Inflammation. 2015;38:2105–15.
Lyu Z, Mao Z, Li Q, Xia Y, Liu Y, He Q, et al. PPARgamma maintains the metabolic heterogeneity and homeostasis of renal tubules. EBioMedicine. 2018;38:178–90.
Su X, Yu R, Yang X, Zhou G, Wang Y, Li L, et al. Telmisartan attenuates peritoneal fibrosis via peroxisome proliferator-activated receptor-gamma activation in rats. Clin Exp Pharmacol Physiol. 2015;42:671–9.
Wang Q, Xu L, Zhang X, Liu D, Wang R. GSK343, an inhibitor of EZH2, mitigates fibrosis and inflammation mediated by HIF-1alpha in human peritoneal mesothelial cells treated with high glucose. Eur J Pharmacol. 2020;880:173076.
Shi Y, Tao M, Wang Y, Zang X, Ma X, Qiu A, et al. Genetic or pharmacologic blockade of enhancer of zeste homolog 2 inhibits the progression of peritoneal fibrosis. J Pathol. 2020;250:79–94.
Kadoya H, Satoh M, Nishi Y, Kondo M, Wada Y, Sogawa Y, et al. Klotho is a novel therapeutic target in peritoneal fibrosis via wnt signaling inhibition. Nephrol Dial Transpl. 2020;35:773–81.
Shi Y, Li J, Chen H, Hu Y, Tang L, Wang Y, et al. Inhibition of EZH2 suppresses peritoneal angiogenesis by targeting a VEGFR2/ERK1/2/HIF-1alpha-dependent signaling pathway. J Pathol. 2022;258:164–78.
Fondevila MF, Fernandez U, Heras V, Parracho T, Gonzalez-Rellan MJ, Novoa E, et al. Inhibition of carnitine palmitoyltransferase 1A in hepatic stellate cells protects against fibrosis. J Hepatol. 2022;77:15–28.
Miguel V, Tituana J, Herrero JI, Herrero L, Serra D, Cuevas P et al. Renal tubule Cpt1a overexpression protects from kidney fibrosis by restoring mitochondrial homeostasis. J Clin Invest. 2021; 131.
Jiang MY, Man WR, Zhang XB, Zhang XH, Duan Y, Lin J, et al. Adipsin inhibits Irak2 mitochondrial translocation and improves fatty acid beta-oxidation to alleviate diabetic cardiomyopathy. Mil Med Res. 2023;10:63.
Aires V, Delmas D, Le Bachelier C, Latruffe N, Schlemmer D, Benoist JF, et al. Stilbenes and resveratrol metabolites improve mitochondrial fatty acid oxidation defects in human fibroblasts. Orphanet J Rare Dis. 2014;9:79.
Li Y, Zhang Y, Deng Q, Mao J, Jia Z, Tang M, et al. Resveratrol reverses palmitic acid-induced cow neutrophils apoptosis through shifting glucose metabolism into lipid metabolism via Cav-1/ CPT 1-mediated FAO enhancement. J Steroid Biochem Mol Biol. 2023;233:106363.
Wu J, Li J, Feng B, Bi Z, Zhu G, Zhang Y, et al. Activation of AMPK-PGC-1alpha pathway ameliorates peritoneal dialysis related peritoneal fibrosis in mice by enhancing mitochondrial biogenesis. Ren Fail. 2022;44:1545–57.
Li ZH, Xu R, Shi J, Yu MS, Zhong Y, He WM, et al. Astragalus Total Saponins ameliorate peritoneal fibrosis by promoting mitochondrial synthesis and inhibiting apoptosis. Am J Chin Med. 2022;50:261–74.
Hara K, Hamada C, Wakabayashi K, Kanda R, Kaneko K, Horikoshi S, et al. Scavenging of reactive oxygen species by astaxanthin inhibits epithelial-mesenchymal transition in high glucose-stimulated mesothelial cells. PLoS ONE. 2017;12:e0184332.
Inoue H, Torigoe K, Torigoe M, Muta K, Obata Y, Suzuki T, et al. Mitochonic acid-5 ameliorates chlorhexidine gluconate-induced peritoneal fibrosis in mice. Med Mol Morphol. 2022;55:27–40.
Selvarajah B, Azuelos I, Anastasiou D, Chambers RC. Fibrometabolism-An emerging therapeutic frontier in pulmonary fibrosis. Sci Signal. 2021; 14.
Bergling K, Martus G, Oberg CM. Phloretin improves Ultrafiltration and reduces glucose absorption during peritoneal Dialysis in rats. J Am Soc Nephrol. 2022;33:1857–63.
Zhang L, Liu J, Liu Y, Xu Y, Zhao X, Qian J, et al. Fluvastatin inhibits the expression of fibronectin in human peritoneal mesothelial cells induced by high-glucose peritoneal dialysis solution via SGK1 pathway. Clin Exp Nephrol. 2015;19:336–42.
Sandoval P, Loureiro J, Gonzalez-Mateo G, Perez-Lozano ML, Maldonado-Rodriguez A, Sanchez-Tomero JA, et al. PPAR-gamma agonist rosiglitazone protects peritoneal membrane from dialysis fluid-induced damage. Lab Invest. 2010;90:1517–32.
Acknowledgements
We thank FigDraw (https://www.figdraw.com/) for providing the original material for the figures. We thank the Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases platform for the technical and facility support.
Author information
Authors and Affiliations
Contributions
Fang Yu: Formal analysis, Investigation, Visualization, Resources, and Writing-original draft. Jia Chen: Validation and Supervision. Xiaoyue Wang: Investigation, and Visualization. Shihui Hou: Formal analysis and Investigation. Hong Li: Validation and Investigation. Yaru Yao: Validation and Visualization. Yani He: Conceptualization, Supervision, Resources, and Writing-review&editing. Kehong Chen: Project administration, Supervision, Resources, and Writing-review&editing.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Yu, F., Chen, J., Wang, X. et al. Metabolic reprogramming of peritoneal mesothelial cells in peritoneal dialysis–associated fibrosis: therapeutic targets and strategies. Cell Commun Signal 23, 114 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-025-02113-2
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-025-02113-2