Next Article in Journal
Functionalisation of Inorganic Material Surfaces with Staphylococcus Protein A: A Molecular Dynamics Study
Next Article in Special Issue
Proteomics and Extracellular Vesicles as Novel Biomarker Sources in Peritoneal Dialysis in Children
Previous Article in Journal
Orofacial Pain: Molecular Mechanisms, Diagnosis, and Treatment 2021
Previous Article in Special Issue
Volume-Independent Sodium Toxicity in Peritoneal Dialysis: New Insights from Bench to Bed
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fibrosis of Peritoneal Membrane as Target of New Therapies in Peritoneal Dialysis

1
Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
2
Department of Medicine, Section of Nephrology and Dialysis, G. d’Annunzio University, 66013 Chieti, Italy
3
Unit of Nephrology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza Miraglia, 80138 Naples, Italy
4
Unit of Nephrology, Santa Maria Hospital, 05100 Terni, Italy
5
Division of Nephrology and Dialysis, Department of Medicine, Piazzale A. Stefani 1, 37126 Verona, Italy
6
Unit of Nephrology, Sant’Eugenio Hospital, 00144 Rome, Italy
7
R&D Department, Iperboreal Pharma, 65122 Pescara, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(9), 4831; https://doi.org/10.3390/ijms23094831
Submission received: 6 April 2022 / Revised: 25 April 2022 / Accepted: 25 April 2022 / Published: 27 April 2022
(This article belongs to the Special Issue Application of Bio-Molecular Sciences in Peritoneal Dialysis)

Abstract

:
Peritoneal dialysis (PD) is an efficient renal replacement therapy for patients with end-stage renal disease. Even if it ensures an outcome equivalent to hemodialysis and a better quality of life, in the long-term, PD is associated with the development of peritoneal fibrosis and the consequents patient morbidity and PD technique failure. This unfavorable effect is mostly due to the bio-incompatibility of PD solution (mainly based on high glucose concentration). In the present review, we described the mechanisms and the signaling pathway that governs peritoneal fibrosis, epithelial to mesenchymal transition of mesothelial cells, and angiogenesis. Lastly, we summarize the present and future strategies for developing more biocompatible PD solutions.

1. Introduction

All over the world, it is estimated that 2 million people suffer from end-stage renal disease (ESRD), and this number continues to increase every year, representing an important economic problem [1]. The ideal treatment for ESRD would be kidney transplantation, but, in the absence of this availability, most patients undergo dialysis. Peritoneal dialysis (PD) is a well-established renal replacement treatment that several studies have shown to be safe and as efficacious as hemodialysis [2,3,4].
With respect to hemodialysis, PD has a series of advantages: it is home-based and thus cost-saving [5], allowing a superior quality of life, it preserves better the residual renal function, while at the same time it produces a gradual and continuous solute and fluid exchange with minimal cardiac stress [6,7,8].
Although PD has a strong potential, the proportion of ESRD patients treated with this technique in developed countries is consistently lower compared to hemodialysis (about 13% in Europe and 10% in the USA) [6,9,10], well below the optimal estimated utilization rate of 25–30% [11]. On the whole, the sub-optimal utilization of PD might be due to financial and economic reasons that favor hemodialysis, lack of patient information about this renal replacement therapy option, or fear of complications and side effects [12]. Another reason is the concern about the durability of the technique as it may be limited by peritoneal membrane integrity and capacity to sustain the treatment over time. It has been proved that peritoneal membrane dysfunction is responsible for about 30% of technique failure [13], and clinical studies showed that peritoneal ultrafiltration (UF) gradually declines 2–4 years after the initiation of PD [14,15]. In the short and medium period, the main causes of PD failure are infections (mainly peritonitis) and issues with the catheter [16,17], whereas, in the long period, the principal problem is the bio-incompatibility of PD solutions which do not preserve the integrity and functionality of peritoneal membrane [18]. Consequently, novel strategies to slow peritoneal membrane deterioration are desirable to allow a significant diffusion of PD, considering its higher economic and environmental sustainability than HD [6].

2. PD Technique

In PD, the peritoneum, the membrane covering the entire peritoneal cavity, is used as a dialysis membrane because it is highly vascularized ad has a large surface area. The parietal peritoneum comprises a single layer of mesothelial cells and a sub-mesothelial area. The mesothelial cells line the peritoneal cavity. Below, the sub-mesothelial zone contains the interstitium, which is a gel-like matrix containing fibroblasts, mast cells, collagen, and other extracellular matrix material. The third layer contains a network of capillary endothelium, endothelial basement membrane, and a capillary fluid film overlying the endothelium [19,20].
PD removes excess water by osmosis and electrolytes as well as metabolic waste products by diffusion across a concentration gradient between the capillary blood and the PD fluid infused into the peritoneal cavity via an implanted intra-abdominal catheter. Usually, two liters of PD solutions are infused into the peritoneal cavity and the effluent is drained after some hours (4 to 8 h are typical dwell times). This procedure is then repeated manually about four times daily (continuous ambulatory PD; CAPD) or using a cycler during the night (automated PD; APD). The solute and water transport across the peritoneal membrane is explained via the three-pore model [21]. In brief, solute and water transport occurs across the vascular endothelium through three pores of varying sizes: large, small, and ultra-small. The large pores (1%) are formed by inter-endothelial cell gaps and are the main site for macromolecule and protein transport. Moreover, the small pores are formed by inter-endothelial gaps, but they account for over 95% of both solutes and water solute removal. Ultra-small pores, made up of aquaporin-1, have been described in endothelial cells of the peritoneal membrane and function as transcellular channels that, under the influence of an osmotic gradient, allow the movement of water only [21,22].
PD solution generally contains the physiological amount of electrolytes (chloride, calcium, sodium, and magnesium), a buffer to correct uremic acidosis (bicarbonate and lactate), and an osmotic agent to induce peritoneal ultrafiltration (UF). Glucose is the main used osmotic agent because it is highly effective, has a low cost, and has a satisfactory safety profile. However, to create an osmotic gradient for the removal of electrolytes and toxins in convection with water, glucose is used 10- to 50-folds higher than serum concentration and this constitutes the principal issue of PD solution bio-incompatibility.
PD removes excess water by osmosis and electrolytes as well as metabolic waste products by diffusion across a concentration gradient between the capillary blood and the PD solution infused into the peritoneal cavity via an implanted intra-abdominal catheter. PD solution contains physiological amounts of electrolytes, a buffer to correct uremic acidosis (bicarbonate or lactate), and an osmotic agent to induce peritoneal UF. Glucose is the main used osmotic agent because it is highly effective, has a low cost, and has a satisfactory safety profile. However, to create an effective osmotic gradient, glucose is used 10- to 50- folds higher than serum concentration and this constitutes the principal issue of PD solution bio-incompatibility.
A multiplicity of studies has proved that long-term exposure to the peritoneum with an un-physiological PD solution activates a series of pathological events such as changes in peritoneal vasculature solutes (neoangiogenesis) with a consequently increased transport of small changes in the interstitium (fibrosis) with a consequent reduction of osmotic conductance as well as the recruitment of inflammatory cells and increased production of inflammatory cytokines which worsen and fuel the un-physiological situation [13,23]. Consequently, the consequent loss of peritoneal integrity reduces UF capacity and PD drop-out [24].

3. Pathophysiology of Peritoneal Membrane Failure (Fibrosis, EMT, Angiogenesis)

The unphysiological characteristics of PD solutions and the uremic status are considered as main factors leading to the functional decline of the peritoneal membrane [25]. These factors induce a chronic peritoneal inflammation that can be worsened by episodes of peritonitis. Structural changes of the peritoneal membrane, including loss of mesothelial cells monolayer, sub-mesothelial fibrosis, angiogenesis, and hyalinizing vasculopathy, are the consequence of reparative processes to inflammatory insults [26,27].
Indeed, inflammation induces neoangiogenesis that increases the surface area available for solute diffusion, and on the other hand, fibrotic thickening of the peritoneum increases flux resistance and consequently reduces water flow. Therefore, initial UF decline is related to increased solute transport and consequent dissipation of the osmotic gradient. Moreover, the onset of fibrosis and neovascularization contribute to increased small-solute transport and UF failure [28].
Peritoneal fibrosis is a slow process, but functional alterations are detectable well before structural changes [29]; moreover, some authors reported that sub-mesothelial thickening and vascular changes could be present even without signs of mesothelial cell layer loss [26].
The first features of peritoneal fibrosis in PD patients were described in the 1980s [28] and subsequently, it has been proved that uremic condition and PD duration are responsible for the development of peritoneal deterioration [26,30]. Macroscopically, the peritoneum exposed to dialysate is brownish or tanned, and it displays texture alterations such as the loss of surface moisture [31]. Histologically, the first alterations occur in the mesothelial layer with distinctive cytoplasmic inclusion and signs of focal defoliation [32]. The subsequent alteration involves the sub-mesothelial compartment. Importantly, sub-mesothelial thickness and vascular alteration are associated with the duration of PD and UF failure [26].
Over the last twenty years, it has been proved that fibrosis, inflammation, angiogenesis, and epithelial-to-mesenchymal transition (EMT) are tightly interconnected in the pathogenesis of UF failure [23,33]. EMT is a common process during physiological situations such as development and wound healing but also in pathological events such as cancer and organ fibrosis [34]. In the peritoneum, the correct definition of EMT is a mesothelial-to-mesenchymal transition (MMT).
MMT represents a complex phenomenon of cellular trans-differentiation that converts the mesothelial phenotype into a mesenchymal one, with the loss of epithelial characteristics and the acquisition of mesenchymal features [35]. MMT was initially thought of as irreversible, but several studies have shown that it is potentially reversible [36]. During MMT, mesothelial cells lose cell polarization, undergo the disassembly of cellular contacts such as adherent and tight junctions, and, at the same time, acquire a fibroblastic shape characterized by higher motility and the capacity to produce and secrete extracellular matrix (ECM). Given these characteristics, mesothelial cells that underwent MMT can migrate to the sub-mesothelial zone and secrete ECM, thus contributing to fibrosis [35]. However, there is still an ongoing debate about the individual contribution of MMT-derived fibroblasts to the pool of sub-mesothelial activated fibroblasts with respect to the activated resident stromal fibroblasts [23]. Moreover, also endothelial-to-mesenchymal transition (EndoMT) could contribute to the pool of activated sub-mesothelial fibroblasts [37], as it occurs in the onset of fibrosis in other districts [38].
The earliest event in MMT involves the loss of cell-to-cell contact, which is associated with the downregulation of epithelial markers such as E-cadherin, cytokeratin, and zonula-occludens-1 (ZO-1) [39,40]. Tight junction proteins such as claudins and occludins are para-cellular components which regulate the transport in the peritoneal mesothelium and their expression and localization are altered in PD patients [41]. In addition, loss of mesothelial layer integrity induces sub-mesothelial tissue to come into contact with bio-incompatible PD solutions as well as inflammatory cytokines [42]. However, it must be kept in mind that as mesothelial cells are of mesodermal origin, they co-express in basal conditions with both epithelial and mesenchymal markers. This may explicate their higher plasticity. Regarding epithelial markers, these cells express a high amount of epithelial cytokeratins, such as cytokeratin 8–18, and proteins of tight and adherens junctions, such as junctional adhesion molecule 1 (JAM1) and ZO-1. E-cadherin is expressed on the membrane and cytoplasm of mesothelial cells [43]. Like mesenchymal cells, mesothelial cells express the intermediate filaments vimentin and desmin constitutively [35,44]. E-cadherin downregulation is due to the induction of Snail, a master factor of EMT, directly inhibiting the E-cadherin transcription [35].
Other possible causes of bio-incompatibility of PD solutions are hypertonicity for the generation of crystalloid osmosis [45], glucose degradation products (GDPs) formed during heat sterilization [46,47], as well as advanced glycation end products (AGEs) formed in the peritoneal cavity [48,49].
UF failure is associated with the increased vascular surface area due to neo-angiogenesis. Vascular wall thickening and augmented permeability increase small solute permeability [50,51,52]. Experimental studies proved increased VEGF production was associated with the use of standard PD solutions and the time of dialysis vintage [53]. Interestingly, VEGF levels decreased when patients were switched from a glucose-based to a glucose-free PD solution (icodextrin, glycerol, and amino acid-based dialysis solutions), suggesting a central role of high glucose concentration in the upregulation of peritoneal VEGF production [54].
The connection between angiogenesis and EMT is well recognized. EMT in mesothelial cells is associated with increased levels of peritoneal VEGF [55,56,57]. Expression of VEGF is firmly controlled at several steps: transcription, mRNA stabilization, alternative splicing, and translation [58,59]; in addition, different factors and cytokines are usually upregulated during PD (IL-1b, IL-6, IL-17, oxidative stress) can regulate its production [60,61]. In particular, TGF-β, a master supervisor of EMT, increases VEGF expression in mesothelial and fibroblasts cells. Moreover, TGF-β inhibition decreased peritoneal fibrosis and VEGF production in a murine model [62]. VEGF signaling is also regulated by the expression of VEGF receptors and co-receptors [58] and they are modulated in mesothelial cells EMT [61].

4. Molecular Pathway of Fibrosis

4.1. TGF-Beta/Smad/Non-Smad/Glucose

TGF-β is part of a superfamily that includes different signaling proteins such as bone morphogenic proteins, activins, and TGF-β isoforms [62], which are involved in several physiological and pathological processes, including proliferation, apoptosis, embryonic development, and organ fibrosis [63]. TGF-βsignaling represents a common mediator of peritoneal fibrogenesis induced by glucose, GDPs, and AGEs in bioincompatible PD solutions [64]. Exposure of mesothelial cells to a high glucose dialysate is associated with a higher synthesis of TGF-β [65]. Moreover, TGF-β signaling is amplified after glucose exposure due to the up-regulation of TGF-β receptor types I and II (TGFR1, TGFR2) in mesothelial cells [66]. Protein kinase C-α (PKC-α) is the common signaling pathway driving TGF-β upregulation in mesothelial cells) [67].
GDPs have also been implied in altering mesothelial cell function and proliferation [68], increasing TGF-β expression and extracellular matrix deposition in the peritoneal wall [69]. Clinical studies reported that TGF-β production correlates with PD vintage [70,71], and in-vivo studies prove that exogenous TGF-β overexpression induces peritoneal fibrosis, increases vessel density, and deteriorates solute transport as well as for UF capacity [72,73].
TGF-β1 can transduce signals through Smad-dependent and Smad-independent pathways, even though most profibrotic actions of TGF-b1 run via Smad signaling. In the classical pathway, Smad2/3 are phosphorylated by PKC, activated by TGFR1, and activin receptor I-β (ACTR1B). Subsequently, they are released from the receptor complex to form a heterotrimeric complex with Smad4 and translocate into the nucleus. Here, they regulate the transcription of target genes in collaboration with various coactivators and corepressors [74,75].
Smad7 is a type of inhibitory Smad, which inhibits Smad2/3 phosphorylation by blocking access to TGFRs. Some works highlighted the positive role of Smad7 on peritoneal failures, such as attenuation of PD-induced peritoneal fibrosis, angiogenesis, and inflammation [76,77,78]. On the other side, BMP-7 exerts antagonistic effects on TGF-β as in PD fluid-instilled rats and co-administration of BMP-7 ameliorated peritoneal fibrosis and increased capillary density [79]. Besides, Smad3 inhibition in uremic-PD rat models treated with recombinant BMP7 decreased peritoneal fibrosis, sub-mesothelial capillary density, and increased UF capacity [80,81]. It has also been proved that mesothelial cells constitutively express BMP-7 and that BMP-7-dependent Smads1/5/8 are reduced in response to conventional PD solutions [79].
Non-Smad signaling pathways are characterized by the activation of protein kinase C, extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and phosphatidylinositol-3-kinase activating the serine-/threonine-specific protein kinase [23].
Several data indicate that high glucose mediates the phosphorylation of PKC [67] and MAPK [82]; furthermore, TGF-β exposure up-regulates Akt (also known as protein kinase B), a phosphatidylinositol-3 kinase (PI3K) target indicating the implication of non-Smad signaling in peritoneal EMT and fibrosis [83]. In-vivo models also confirmed that inhibition of JNK and p38 MAPK counteract TGF-β-induced peritoneal fibrosis [84,85,86].
Moreover, NF-κB inhibition has been linked to TGF-β signaling inhibition [87].
Finally, high glucose concentration in PD solutions is tightly connected with TGF-β signaling and UF failure. It has been proposed that the degradation of up-taken glucose induces changes in the intracellular NADH/NAD+ ratio, like hypoxia. Exposure to high levels of glucose stimulates the formation of mediators such as TGF-β and plasminogen activator inhibitor-1. This effect is also associated with a higher expression of glucose transporter 1 (GLUT-1). The increased amount of GLUT1 further enhances intracellular glucose uptake and thereby stimulates the vicious loop, including dialysate glucose exposure, peritoneal fibrosis, and UF failure [88].

4.2. Other Signaling Pathway: CTGF, NLRP3/IL-1b, and Cytokines

Connective tissue growth factor (CTGF) is a downstream mediator of TGF-β [89] and induces similar effects: ECM production, cell proliferation, adhesion, and migration [90]. In detail, CTGF expression is activated by TGF-β via a responsive element in the promoter region of the CTGF gene [91] and mediated by Smad3 and Smad4 [92]. Its profibrotic properties have been shown in multiple mesenchymal cells, in which CTGF is a downstream effector of TGF-β [93].
Clinical data demonstrate that CTGF is upregulated in PD patients with UF failure [94,95]; its expression is regulated by glucose [96] and correlates with peritoneal membrane thickness in PD patients with and without EPS [97].
Studies in mouse models proved that also AGEs and GDPs act via CTGF in peritoneal fibrosis, angiogenesis, and inflammation [96,98,99]. Although CTGF is involved in peritoneal fibrosis, additional studies will be necessary to characterize its potential as a pharmacological target as it lacks a specific receptor, it has several isoforms, and it interacts with multiple factors (bone morphogenic factors, VEGF, Wnt, integrins, heparan sulfate proteoglycans, and epidermal growth factor receptor) [100].
Recent data suggest that the NOD-like receptor protein 3 (NLRP3) inflammasome is involved in peritoneal inflammation and consecutive fibrosis. NLRP3 intracellular complex is a component of the innate immune system that mediates caspase-1 activation and regulates the release of pro-inflammatory cytokines IL-1β and IL-18 in response to microbial infection and cellular damage [101]. It has been shown that high glucose-based PD solution activates NLRP3/IL-1β peritoneal mesothelial cells [102,103] and that genetic deficiency of NLRP3 complex or IL-1β reduces inflammatory and peritoneal fibrosis model in mice [104].
Peritoneal injury causes the activation of macrophages, neutrophils, endothelial cells (ECs), and MCs, which are the main sources of proinflammatory cytokines and fibrotic mediators in response to external signals [105,106]. Once activated, they release numerous inflammatory cytokines, including IL-6, IL-1β, IL-8, TNF-α, monocyte chemoattractant protein-1 (MCP-1), and macrophage inflammatory protein 2 [107,108,109].
IL-6 is a crucial actor in modulating peritoneal inflammation. Intraperitoneal IL-6 is associated with increasing peritoneal solute transport rate [110] and intraperitoneal IL-6 production is proportional to dialysate glucose concentration [111]. IL-6 and soluble IL-6 receptors induce the synthesis and secretion of MCP-1, which attracts monocytes and lymphocytes [112]. A recent study proved that IL-6 leads to peritoneal inflammation and fibrosis development via a STAT3-dependent pathway [113]. IL-6 inhibition ameliorated EMT in human peritoneal mesothelial cells in vitro and ameliorated high glucose-mediated peritoneal fibrosis development in vivo via inhibiting STAT3 phosphorylation [113].
Another cytokine involved in the peritoneal inflammation is IL-17, which strongly affects mesothelial cell cytokine production such as CXCL1 [114]. Moreover, IL-17 is present in the peritoneum of PD patients and correlates with both the duration of PD and the extent of peritoneal inflammation and fibrosis [115]. Recent surveys showed that treatment with alanyl-glutamine on rats and mice exposed to PD fluids resulted in a reduction of peritoneal fibrosis associated with reduced peritoneal IL-17 expression [116].

4.3. The Role of Metabolism

Glycolysis, glutaminolysis, and fatty acid oxidation are metabolic processes that supervise the deposition and breakdown of collagen and other ECM components, which may result in fibrosis [117]. Hyperglycemia upregulates TGF-β and hypoxia-inducible factor 1 subunit alpha (HIF1α) expression [118] by increasing the glycolytic rate and inhibiting pyruvate dehydrogenase complex (PDH), promotes the production of lactic acid [119]. Glycolytic intermediates are important in the synthesis of amino acid substrates for collagen synthesis [120] and lactic acid promotes lactylation of lysine residues in extracellular proteins, favoring the conversion of macrophages to an inflammatory phenotype [121]. In summary, hyperglycemia increases TGF-β and HIF1α expression, which in turn elevates rates of glycolysis and lactic acid production with the consequently increased collagen synthesis, acidification, expansion, and lower degradation of ECM. In essence, a pathway promoting and sustaining fibrosis. The substrate glutamine also has a role in EMT. This amino acid is important for collagen synthesis, but it can be converted to glutamate and then to ketoglutarate, which provides a substrate for the generation of NADH and FADH2 and consequently ATP through oxidative phosphorylation [122,123,124] (Figure 1).

5. Effluent Biomarkers to Monitor PD Efficiency

Prognostic biomarkers have been proposed in PD patients to evaluate the peritoneal membrane deterioration. The ideal PD biomarker could be directly accessible in PD effluents to allow one to identify PD patients who are at a high risk of complications. The main biomarkers currently used in PD are IL-6, a marker of chronic peritoneal inflammation, and cancer antigen-125 (CA-125), which is an expression of mesothelial cell mass [125].
IL-6 increases in the effluent of patients with acute bacterial peritonitis and may be used to evaluate the bacterial clearance during the infection. Furthermore, IL-6 in PD effluent correlates with subclinical infections (e.g., biofilms on PD catheter) [126]. Notably, experimental studies suggest persistent peritoneal IL-6 is associated with membrane change/fibrosis and angiogenesis. Other interleukins (IL-8, IL-17) are investigated to evaluate a potential role as an inflammatory marker [125].
The peritoneal membrane undergoes progressive remodeling over PD time resulting in the accumulation of extracellular matrix and fibrosis, included in a complex process called Peritoneal MMT in which mesothelial cells are transformed into fibroblast-like cells leading to inflammation, fibrosis, and angiogenesis. Peritoneal levels of CA-125 have been proposed to estimate mesothelial cell mass as a surrogate parameter for the peritoneal membrane status. The change over time in CA-125 has been proposed as a marker of MMT though the findings are not conclusive [127].
Micro RNAs M (MiR) are small non-coding RNA molecules (18–24 nucleotides), which work as a post-transcriptional regulator in several cellular processes. In PD, microRNA-21 and microRNA-31 were recently proposed to evaluate MMT, but their role is still debated [128].
Recently, PD effluent biomarkers, identified by “omics” technologies, especially proteomics and metabolomics, could predict the onset of peritoneal membrane dysfunction. The metabolic profile in PD effluent might be the expression of a healthy membrane and its change over time predict technique survival [129].
Interestingly, more recently, water channel Aquaporin 1 (AQP1) excreted by the mesothelium has been studied as a biomarker in PD effluent. AQP-1 levels in the effluent correlate with ultrafiltration and free water transport (sodium sieving) evaluated by the peritoneal equilibration test [130].
However, there is no evidence of association of any PD biomarkers with relevant clinical outcomes and their use in the clinical practice is modest.

6. Strategies to Reduce Fibrosis

6.1. Low GDPs and Neutral pH

New glucose-based solutions with a neutral- or physiological-pH and low-GDP content (using multi-chamber bags) have been developed to increase the biocompatibility of PD dialysate [131]. The use of lactate or bicarbonate as a pH buffer has significantly reduced systemic GDPs and AGEs. However, the clinical superiority of neutral-pH, low-GDP PD solutions has been questioned [132,133]. In detail, neutral-pH, low-GDP PD solutions seem to be better at preserving the peritoneal endothelial glycocalyx compared to conventional acidic solutions during prolonged PD [134]. However, biopsies in children showed early peritoneal inflammation, hypervascularization, fibroblast activation, and epithelial–mesenchymal transition, which affected PD membrane-transport function [135].

6.2. Glucose Sparing

The high glucose content of the PD solution is the main culprit for peritoneal damage over time. In addition, exposure to high glucose concentration leads to systemic adverse effects such as hyperglycemia, insulin resistance, diabetes, and cardiovascular diseases [136,137]. Numerous compounds have been tested as alternatives to glucose, but only two osmotic agents are currently available in clinical practice: icodextrin and amino acids. Unfortunately, these compounds can only be used in a single daily peritoneal exchange [138,139], reducing daily glucose load by only 30–50% [140]
Icodextrin is a water-soluble glucose polymer derived from starch. The use of icodextrin-containing PD solution is associated with improved peritoneal UF and fewer episodes of fluid overload [141]. However, the low pH of the icodextrin solution may induce an increased local and systemic inflammation and activation of the EMT process [142].
PD solutions containing amino acids (e.g., Nutrineal®) have a pH of 6.7 and are free of GDPs. This PD solution may improve the nutritional status of some malnourished PD patients by increasing muscle amino acid uptake [143]. Peritoneal ultrafiltration rate and small solute clearance over a 6-h dwell did not show any main difference between amino acid-based PD fluid and equimolar glucose-based solutions [144,145]. However, the biocompatibility of these PD solutions, which influences the peritoneal function over time, is debated. Indeed, while some experimental studies showed a better biocompatible profile compared to standard glucose-based PD solutions, others reported increased generation of nitric oxide in human mesothelial cells cultured with a PD solution containing amino acids, a finding that may have pathophysiological relevance [146].
Other compounds that have been tested for a potential use in the PD solution as osmotic agents to replace glucose include taurine and hyperbranched polyglycerol, but they are under experimental development.

6.3. Use of Metabolically Active Osmolytes

The osmo-metabolic approach uses osmolytes in the PD solution, which may offer a bioactive glucose-sparing by reducing intraperitoneal glucose load without compromising UF and mitigating the underlying systemic negative metabolic effects caused by the glucose load.
L-carnitine (LC) and xylitol may be used as osmo-metabolic agents in PD dialysate. LC is a naturally occurring compound involved in fatty acid oxidation [147]. The mode of action of LC relates to its ability to modulate intra-mitochondrial acetyl-CoA levels, a key metabolic intermediate able to affect both muscle glucose disposal and liver glucose production [148]. Xylitol, a five-carbon sugar alcohol, is a physiologic metabolic intermediate of the glucuronate-xylulose cycle, a pathway very active in the liver and intimately interconnected with the pentose monophosphate shunt at the level of D-xylulose-5-phosphate [149,150]. Interestingly, xylitol is a very poor insulin secretagogue compared to glucose [151]. A key attribute of xylitol is that it does not undergo a Maillard reaction as it usually happens between traditional reducing sugars (i.e., glucose) and amino acids/proteins, a reaction also commonly responsible for the formation of AGEs [152].
LC and xylitol are characterized by molecular weight similar to glucose, high water solubility, and osmotic properties [148]. The good biocompatibility of LC- and xylitol-containing solutions has been demonstrated in several in-vitro and in-vivo models [153,154] In addition, clinical studies have demonstrated excellent tolerability and feasibility of xylitol [155] and L-carnitine solutions [156], as well as a better preservation of urine volume, compared to controls (treated with standard glucose-based PD fluids) over a 4-month period [157]. Clinical use of xylitol- or LC-containing dialysate in CAPD patients was associated with positive metabolic effects such as improving glycemic control.
PD solution containing LC, xylitol, and low glucose, has been designed to achieve a favorable synergistic combination of the two osmo-metabolic agents. In-vitro studies provide further evidence that this novel formulation of PD solutions better preserves the integrity of the mesothelial cell layer compared to conventional PD solutions reducing fibrogenic features and inflammation [158,159].
Preliminary results obtained from a phase II, prospective, open, multicenter study to investigate the tolerability and the efficacy of osmo-metabolic agent-based PD solutions in CAPD patients (NCT04001036) confirmed that these novel solutions are well tolerated and no serious adverse reactions were reported. Non-inferiority of the osmo-metabolic agent-based PD solutions compared to standard solutions in terms of peritoneal transport and adequacy also was demonstrated as targets [160].

6.4. Use of Pharmacological Agents Added to Conventional PD Solutions

To counteract the adverse effects of conventional PD solutions, several compounds have been tested in-vitro and in-vivo. Unfractionated heparin, low-molecular-weight heparins, and sulodexide showed a different response in clinical trials, probably due to their different capacity to inhibit complement [161,162,163,164]. With the same objective to inhibit complement, sodium citrate has been tested in association with heparin [165].
A recently tested strategy is the addition of pharmacological doses of alanyl-glutamine (Ala-Gln) to glucose-based PD solutions and phase II clinical trials indicate that Ala-Gln supplementation in PD solution improves biomarkers of peritoneal membrane integrity, immune competence, and systemic inflammation when compared to non-supplemented PD solution with neutral pH and low-glucose degradation probably via an antioxidant mechanism [166,167,168]. However, use in clinical practice remains still debated.
Another element that has been tested as a possible pharmacological agent to add to the DP solution is molecular hydrogen (H2) [169]. Its antioxidant and anti-inflammatory properties have been tested in various animal models [170]. Molecular hydrogen, added to a standard PD solution, has also been tested in humans (6 patients), confirming a reduction in oxidative stress at both the peritoneal and systemic levels in the absence of adverse events [171]. In addition, recent in-vivo studies indicate that molecular hydrogen could preserve mesothelial integrity and reduce the progression of glucose-induced fibrosis [172,173]; thus, future clinical studies will be necessary to evaluate the efficacy and safety of this therapeutic solution.
Finally, a recent study proposes the addition of lithium chloride to conventional PD solutions for preserving peritoneal membrane integrity [174]. In detail, lithium chloride could reduce apoptosis, peritoneal membrane fibrosis, and angiogenesis by regulating the activity of some kinases, such as glycogen synthase kinase 3 and protein kinase 2. Although these findings may be hopeful, the real benefits will have to be demonstrated, considering that lithium chloride is an antidepressant and potentially nephrotoxic agent.
Even if the addition of pharmacological agents can improve the characteristics of traditional dialysis solutions, the glucose concentration remains very high, and consequently, the harmful effects associated with it would remain present [132,146]

6.5. Glycolytic and Pyruvate Metabolism as Targets to Control Peritoneal Fibrosis

An alternative pharmacological strategy to control fibrosis could be based on a fatty acid or pyruvate oxidation or even by inhibiting glycolysis. For instance, an increase of PDH, a key enzyme in coupling glycolysis with the Krebs cycle, can be achieved through the inhibition of pyruvate dehydrogenase activity kinase (PDHK), a potent inhibitor of PDH, using dichloroacetate (DCA) [175]. DCA has been shown to be very effective in inhibiting fibrosis in various experimental models [175,176,177,178,179]. Another means to keep more active PDH is by reducing the intramitochondrial pool of acetyl-CoA, a potent allosteric activator of PDHK, by supraphysiological concentrations of L-carnitine [148]. The latter mechanism involves the freely reversible reaction catalyzed by carnitine acetyltransferase in transferring the acetyl-residue esterified to Coenzyme A to L-carnitine to form acetyl-carnitine. Indeed, as this enzymatic reaction is very sensitive to the mass action effect of L-carnitine, the intramitochondrial concentration of acetyl-CoA will be significantly reduced, translating into a less active PDK1 and, hence, a more active PDH [180]. L-Carnitine administration has been shown to mitigate the induction of fibrosis in various experimental models. A third option may be the inhibiting glycolysis by 2-deoxyglucose, a glucose derivative that acts as an inhibitor of hexokinase 2 and hence of glycolysis [181]. As TGF-β1 is a key facilitator of the EMT transition by switching cellular energy provision from oxidative phosphorylation to substrate-level phosphorylation through aerobic glycolysis [182], the reduction of high glycolytic fluxes with 2-deoxyglucose could reduce peritoneal fibrosis [183,184]. However, according to the mode of action of DCA and L-carnitine, their anti-fibrotic effects may not necessarily require a reduction of glycolytic flux but rather an efficient coupling of such flux with an active PDH. In addition, it remains to be established whether the inhibition of glycolysis is a safer strategy compared to the diversion of pyruvate metabolism towards oxidative phosphorylation [185] (Figure 2).

Author Contributions

Conceptualization, V.M., M.B. and A.A.; writing—review and editing, V.M., M.B., S.B., L.D.L., L.V., M.O., G.G., R.P., A.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

A.A. is an employee of Iperboreal Pharma srl, Italy. The rest of the authors declare no conflict of interest.

References

  1. Howell, M.; Walker, R.C.; Howard, K. Cost Effectiveness of Dialysis Modalities: A Systematic Review of Economic Evaluations. Appl. Health Econ. Health Policy 2019, 17, 315–330. [Google Scholar] [CrossRef] [PubMed]
  2. Javaid, M.M.; Khan, B.A.; Subramanian, S. Peritoneal Dialysis as Initial Dialysis Modality: A Viable Option for Late-Presenting End-Stage Renal Disease. J. Nephrol. 2019, 32, 51–56. [Google Scholar] [CrossRef] [PubMed]
  3. Yeates, K.; Zhu, N.; Vonesh, E.; Trpeski, L.; Blake, P.; Fenton, S. Hemodialysis and Peritoneal Dialysis Are Associated with Similar Outcomes for End-Stage Renal Disease Treatment in Canada. Nephrol. Dial. Transplant. 2012, 27, 3568–3575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Mehrotra, R.; Chiu, Y.-W.; Kalantar-Zadeh, K.; Bargman, J.; Vonesh, E. Similar Outcomes with Hemodialysis and Peritoneal Dialysis in Patients with End-Stage Renal Disease. Arch. Intern. Med. 2011, 171, 110–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Karopadi, A.N.; Mason, G.; Rettore, E.; Ronco, C. Cost of Peritoneal Dialysis and Haemodialysis across the World. Nephrol. Dial. Transplant. 2013, 28, 2553–2569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Mehrotra, R.; Devuyst, O.; Davies, S.J.; Johnson, D.W. The Current State of Peritoneal Dialysis. J. Am. Soc. Nephrol. 2016, 27, 3238–3252. [Google Scholar] [CrossRef] [Green Version]
  7. Korevaar, J.C.; Jansen, M.A.; Merkus, M.P.; Dekker, F.W.; Boeschoten, E.W.; Krediet, R.T. Quality of Life in Predialysis End-Stage Renal Disease Patients at the Initiation of Dialysis Therapy. The NECOSAD Study Group. Perit. Dial. Int. 2000, 20, 69–75. [Google Scholar]
  8. Cameron, J.I.; Whiteside, C.; Katz, J.; Devins, G.M. Differences in Quality of Life across Renal Replacement Therapies: A Meta-Analytic Comparison. Am. J. Kidney Dis. 2000, 35, 629–637. [Google Scholar] [CrossRef] [Green Version]
  9. Li, P.K.-T.; Chow, K.M.; Van de Luijtgaarden, M.W.M.; Johnson, D.W.; Jager, K.J.; Mehrotra, R.; Naicker, S.; Pecoits-Filho, R.; Yu, X.Q.; Lameire, N. Changes in the Worldwide Epidemiology of Peritoneal Dialysis. Nat. Rev. Nephrol. 2017, 13, 90–103. [Google Scholar] [CrossRef]
  10. Kramer, A.; Pippias, M.; Noordzij, M.; Stel, V.S.; Andrusev, A.M.; Aparicio-Madre, M.I.; Arribas Monzón, F.E.; Åsberg, A.; Barbullushi, M.; Beltrán, P.; et al. The European Renal Association—European Dialysis and Transplant Association (ERA-EDTA) Registry Annual Report 2016: A Summary. Clin. Kidney J. 2019, 12, 702–720. [Google Scholar] [CrossRef] [Green Version]
  11. Lameire, N.; Van Biesen, W. Epidemiology of Peritoneal Dialysis: A Story of Believers and Nonbelievers. Nat. Rev. Nephrol. 2010, 6, 75–82. [Google Scholar] [CrossRef] [PubMed]
  12. Roumeliotis, S.; Dounousi, E.; Salmas, M.; Eleftheriadis, T.; Liakopoulos, V. Unfavorable Effects of Peritoneal Dialysis Solutions on the Peritoneal Membrane: The Role of Oxidative Stress. Biomolecules 2020, 10, 768. [Google Scholar] [CrossRef] [PubMed]
  13. Davies, S.J.; Mushahar, L.; Yu, Z.; Lambie, M. Determinants of Peritoneal Membrane Function over Time. Semin. Nephrol. 2011, 31, 172–182. [Google Scholar] [CrossRef] [PubMed]
  14. Davies, S.J.; Bryan, J.; Phillips, L.; Russell, G.I. Longitudinal Changes in Peritoneal Kinetics: The Effects of Peritoneal Dialysis and Peritonitis. Nephrol. Dial. Transplant. 1996, 11, 498–506. [Google Scholar] [CrossRef]
  15. Smit, W.; Schouten, N.; van den Berg, N.; Langedijk, M.J.; Struijk, D.G.; Krediet, R.T. Analysis of the Prevalence and Causes of Ultrafiltration Failure during Long-Term Peritoneal Dialysis: A Cross-Sectional Study. Perit. Dial. Int. 2004, 24, 562–570. [Google Scholar] [CrossRef]
  16. Li, P.K.-T.; Szeto, C.C.; Piraino, B.; de Arteaga, J.; Fan, S.; Figueiredo, A.E.; Fish, D.N.; Goffin, E.; Kim, Y.-L.; Salzer, W.; et al. ISPD Peritonitis Recommendations: 2016 Update on Prevention and Treatment. Perit. Dial. Int. 2016, 36, 481–508. [Google Scholar] [CrossRef] [Green Version]
  17. Hayat, A.; Collins, J.; Saweirs, W. Study of Early Complications Associated with Peritoneal Dialysis Catheters: An Analysis of the New Zealand Peritoneal Dialysis Registry Data. Int. Urol. Nephrol. 2021, 53, 1705–1711. [Google Scholar] [CrossRef]
  18. Bajo, M.A.; Del Peso, G.; Teitelbaum, I. Peritoneal Membrane Preservation. Semin. Nephrol. 2017, 37, 77–92. [Google Scholar] [CrossRef]
  19. Blackburn, S.C.; Stanton, M.P. Anatomy and Physiology of the Peritoneum. Semin. Pediatr. Surg. 2014, 23, 326–330. [Google Scholar] [CrossRef]
  20. Schaefer, B.; Bartosova, M.; Macher-Goeppinger, S.; Ujszaszi, A.; Wallwiener, M.; Nyarangi-Dix, J.; Sallay, P.; Burkhardt, D.; Querfeld, U.; Pfeifle, V.; et al. Quantitative Histomorphometry of the Healthy Peritoneum. Sci. Rep. 2016, 6, 21344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Rippe, B. A Three-Pore Model of Peritoneal Transport. Perit. Dial. Int. 1993, 13, S35–S38. [Google Scholar] [CrossRef] [PubMed]
  22. Devuyst, O.; Nielsen, S.; Cosyns, J.P.; Smith, B.L.; Agre, P.; Squifflet, J.P.; Pouthier, D.; Goffin, E. Aquaporin-1 and Endothelial Nitric Oxide Synthase Expression in Capillary Endothelia of Human Peritoneum. Am. J. Physiol. 1998, 275, H234–H242. [Google Scholar] [CrossRef] [PubMed]
  23. Balzer, M.S. Molecular Pathways in Peritoneal Fibrosis. Cell Signal. 2020, 75, 109778. [Google Scholar] [CrossRef]
  24. Davies, S.J.; Phillips, L.; Griffiths, A.M.; Russell, L.H.; Naish, P.F.; Russell, G.I. What Really Happens to People on Long-Term Peritoneal Dialysis? Kidney Int. 1998, 54, 2207–2217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Krediet, R.T.; Lindholm, B.; Rippe, B. Pathophysiology of Peritoneal Membrane Failure. Perit. Dial. Int. 2000, 20, S22–S42. [Google Scholar] [CrossRef] [PubMed]
  26. Williams, J.D.; Craig, K.J.; Topley, N.; Von Ruhland, C.; Fallon, M.; Newman, G.R.; Mackenzie, R.K.; Williams, G.T. Morphologic Changes in the Peritoneal Membrane of Patients with Renal Disease. J. Am. Soc. Nephrol. 2002, 13, 470–479. [Google Scholar] [CrossRef] [PubMed]
  27. Mateijsen, M.A.; van der Wal, A.C.; Hendriks, P.M.; Zweers, M.M.; Mulder, J.; Struijk, D.G.; Krediet, R.T. Vascular and Interstitial Changes in the Peritoneum of CAPD Patients with Peritoneal Sclerosis. Perit. Dial. Int. 1999, 19, 517–525. [Google Scholar] [CrossRef]
  28. Dobbie, J.W.; Zaki, M.; Wilson, L. Ultrastructural Studies on the Peritoneum with Special Reference to Chronic Ambulatory Peritoneal Dialysis. Scott. Med. J. 1981, 26, 213–223. [Google Scholar] [CrossRef]
  29. Lambie, M.L.; John, B.; Mushahar, L.; Huckvale, C.; Davies, S.J. The Peritoneal Osmotic Conductance Is Low Well before the Diagnosis of Encapsulating Peritoneal Sclerosis Is Made. Kidney Int. 2010, 78, 611–618. [Google Scholar] [CrossRef] [Green Version]
  30. Honda, K.; Hamada, C.; Nakayama, M.; Miyazaki, M.; Sherif, A.M.; Harada, T.; Hirano, H. Impact of Uremia, Diabetes, and Peritoneal Dialysis Itself on the Pathogenesis of Peritoneal Sclerosis: A Quantitative Study of Peritoneal Membrane Morphology. Clin. J. Am. Soc. Nephrol. 2008, 3, 720–728. [Google Scholar] [CrossRef] [Green Version]
  31. Dobbie, J.W. Peritoneal Ultrastructure and Changes with Continuous Ambulatory Peritoneal Dialysis. Perit. Dial. Int. 1993, 13, S585–S587. [Google Scholar] [CrossRef] [PubMed]
  32. Dobbie, J.W.; Lloyd, J.K.; Gall, C.A. Categorization of Ultrastructural Changes in Peritoneal Mesothelium, Stroma and Blood Vessels in Uremia and CAPD Patients. Adv. Perit. Dial. 1990, 6, 3–12. [Google Scholar] [PubMed]
  33. Zhou, Q.; Bajo, M.-A.; Del Peso, G.; Yu, X.; Selgas, R. Preventing Peritoneal Membrane Fibrosis in Peritoneal Dialysis Patients. Kidney Int. 2016, 90, 515–524. [Google Scholar] [CrossRef] [PubMed]
  34. Nieto, M.A.; Huang, R.Y.-J.; Jackson, R.A.; Thiery, J.P. EMT: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Yáñez-Mó, M.; Lara-Pezzi, E.; Selgas, R.; Ramírez-Huesca, M.; Domínguez-Jiménez, C.; Jiménez-Heffernan, J.A.; Aguilera, A.; Sánchez-Tomero, J.A.; Bajo, M.A.; Alvarez, V.; et al. Peritoneal Dialysis and Epithelial-to-Mesenchymal Transition of Mesothelial Cells. N. Engl. J. Med. 2003, 348, 403–413. [Google Scholar] [CrossRef] [PubMed]
  36. Jang, Y.-H.; Shin, H.-S.; Sun Choi, H.; Ryu, E.-S.; Jin Kim, M.; Ki Min, S.; Lee, J.-H.; Kook Lee, H.; Kim, K.-H.; Kang, D.-H. Effects of Dexamethasone on the TGF-Β1-Induced Epithelial-to-Mesenchymal Transition in Human Peritoneal Mesothelial Cells. Lab. Investig. 2013, 93, 194–206. [Google Scholar] [CrossRef] [Green Version]
  37. Masola, V.; Bonomini, M.; Onisto, M.; Ferraro, P.M.; Arduini, A.; Gambaro, G. Biological Effects of XyloCore, a Glucose Sparing PD Solution, on Mesothelial Cells: Focus on Mesothelial-Mesenchymal Transition, Inflammation and Angiogenesis. Nutrients 2021, 13, 2282. [Google Scholar] [CrossRef]
  38. Piera-Velazquez, S.; Mendoza, F.A.; Jimenez, S.A. Endothelial to Mesenchymal Transition (EndoMT) in the Pathogenesis of Human Fibrotic Diseases. J. Clin. Med. 2016, 5, 45. [Google Scholar] [CrossRef]
  39. Wang, L.; Balzer, M.S.; Rong, S.; Menne, J.; von Vietinghoff, S.; Dong, L.; Gueler, F.; Jang, M.-S.; Xu, G.; Timrott, K.; et al. Protein Kinase C α Inhibition Prevents Peritoneal Damage in a Mouse Model of Chronic Peritoneal Exposure to High-Glucose Dialysate. Kidney Int. 2016, 89, 1253–1267. [Google Scholar] [CrossRef]
  40. Aroeira, L.S.; Aguilera, A.; Sánchez-Tomero, J.A.; Bajo, M.A.; del Peso, G.; Jiménez-Heffernan, J.A.; Selgas, R.; López-Cabrera, M. Epithelial to Mesenchymal Transition and Peritoneal Membrane Failure in Peritoneal Dialysis Patients: Pathologic Significance and Potential Therapeutic Interventions. J. Am. Soc. Nephrol. 2007, 18, 2004–2013. [Google Scholar] [CrossRef] [Green Version]
  41. Ito, T.; Yorioka, N.; Yamamoto, M.; Kataoka, K.; Yamakido, M. Effect of Glucose on Intercellular Junctions of Cultured Human Peritoneal Mesothelial Cells. J. Am. Soc. Nephrol. 2000, 11, 1969–1979. [Google Scholar] [CrossRef] [PubMed]
  42. Kang, D.-H. Loosening of the Mesothelial Barrier as an Early Therapeutic Target to Preserve Peritoneal Function in Peritoneal Dialysis. Kidney Res. Clin. Pract. 2020, 39, 136–144. [Google Scholar] [CrossRef] [PubMed]
  43. Strippoli, R.; Loureiro, J.; Moreno, V.; Benedicto, I.; Pérez Lozano, M.L.; Barreiro, O.; Pellinen, T.; Minguet, S.; Foronda, M.; Osteso, M.T.; et al. Caveolin-1 Deficiency Induces a MEK-ERK1/2-Snail-1-Dependent Epithelial-Mesenchymal Transition and Fibrosis during Peritoneal Dialysis. EMBO Mol. Med. 2015, 7, 102–123. [Google Scholar] [CrossRef] [PubMed]
  44. Mutsaers, S.E. The Mesothelial Cell. Int. J. Biochem. Cell Biol. 2004, 36, 9–16. [Google Scholar] [CrossRef]
  45. Morelle, J.; Sow, A.; Fustin, C.-A.; Fillée, C.; Garcia-Lopez, E.; Lindholm, B.; Goffin, E.; Vandemaele, F.; Rippe, B.; Öberg, C.M.; et al. Mechanisms of Crystalloid versus Colloid Osmosis across the Peritoneal Membrane. J. Am. Soc. Nephrol. 2018, 29, 1875–1886. [Google Scholar] [CrossRef]
  46. Zimmeck, T.; Tauer, A.; Fuenfrocken, M.; Pischetsrieder, M. How to Reduce 3-Deoxyglucosone and Acetaldehyde in Peritoneal Dialysis Fluids. Perit. Dial. Int. 2002, 22, 350–356. [Google Scholar] [CrossRef]
  47. Erixon, M.; Lindén, T.; Kjellstrand, P.; Carlsson, O.; Ernebrant, M.; Forsbäck, G.; Wieslander, A.; Jönsson, J.A. PD Fluids Contain High Concentrations of Cytotoxic GDPs Directly after Sterilization. Perit. Dial. Int. 2004, 24, 392–398. [Google Scholar] [CrossRef]
  48. Lui, S.L.; Yung, S.; Yim, A.; Wong, K.M.; Tong, K.L.; Wong, K.S.; Li, C.S.; Au, T.C.; Lo, W.K.; Ho, Y.W.; 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, 966–975. [Google Scholar] [CrossRef]
  49. Shaw, S.; Akyol, M.; Bell, J.; Briggs, J.D.; Dominiczak, M.H. Effects of Continuous Ambulatory Peritoneal Dialysis and Kidney Transplantation on Advanced Glycation Endproducts in the Skin and Peritoneum. Cell. Mol. Biol. 1998, 44, 1061–1068. [Google Scholar]
  50. Selgas, R.; del Peso, G.; Bajo, M.A.; Castro, M.A.; Molina, S.; Cirugeda, A.; Sánchez-Tomero, J.A.; Castro, M.J.; Alvarez, V.; Corbí, A.; et al. Spontaneous VEGF Production by Cultured Peritoneal Mesothelial Cells from Patients on Peritoneal Dialysis. Perit. Dial. Int. 2000, 20, 798–801. [Google Scholar] [CrossRef] [Green Version]
  51. Vriese, A.S.D.; Tilton, R.G.; Stephan, C.C.; Lameire, N.H. Vascular Endothelial Growth Factor Is Essential for Hyperglycemia-Induced Structural and Functional Alterations of the Peritoneal Membrane. J. Am. Soc. Nephrol. 2001, 12, 1734–1741. [Google Scholar] [CrossRef] [PubMed]
  52. Michels, W.M.; Zweers, M.M.; Smit, W.; Korevaar, J.; Struijk, D.G.; van Westrhenen, R.; Krediet, R.T. Does Lymphatic Absorption Change with the Duration of Peritoneal Dialysis? Perit. Dial. Int. 2004, 24, 347–352. [Google Scholar] [CrossRef] [PubMed]
  53. Mortier, S.; Faict, D.; Lameire, N.H.; De Vriese, A.S. Benefits of Switching from a Conventional to a Low-GDP Bicarbonate/Lactate-Buffered Dialysis Solution in a Rat Model. Kidney Int. 2005, 67, 1559–1565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Zweers, M.M.; Struijk, D.G.; Smit, W.; Krediet, R.T. Vascular Endothelial Growth Factor in Peritoneal Dialysis: A Longitudinal Follow-Up. J. Lab. Clin. Med. 2001, 137, 125–132. [Google Scholar] [CrossRef]
  55. Aroeira, L.S.; Aguilera, A.; Selgas, R.; Ramírez-Huesca, M.; Pérez-Lozano, M.L.; Cirugeda, A.; Bajo, M.A.; del Peso, G.; Sánchez-Tomero, J.A.; Jiménez-Heffernan, J.A.; et al. Mesenchymal Conversion of Mesothelial Cells as a Mechanism Responsible for High Solute Transport Rate in Peritoneal Dialysis: Role of Vascular Endothelial Growth Factor. Am. J. Kidney Dis. 2005, 46, 938–948. [Google Scholar] [CrossRef]
  56. Boulanger, E.; Grossin, N.; Wautier, M.-P.; Taamma, R.; Wautier, J.-L. Mesothelial RAGE Activation by AGEs Enhances VEGF Release and Potentiates Capillary Tube Formation. Kidney Int. 2007, 71, 126–133. [Google Scholar] [CrossRef] [Green Version]
  57. Gerber, S.A.; Rybalko, V.Y.; Bigelow, C.E.; Lugade, A.A.; Foster, T.H.; Frelinger, J.G.; Lord, E.M. Preferential Attachment of Peritoneal Tumor Metastases to Omental Immune Aggregates and Possible Role of a Unique Vascular Microenvironment in Metastatic Survival and Growth. Am. J. Pathol. 2006, 169, 1739–1752. [Google Scholar] [CrossRef] [Green Version]
  58. Apte, R.S.; Chen, D.S.; Ferrara, N. VEGF in Signaling and Disease: Beyond Discovery and Development. Cell 2019, 176, 1248–1264. [Google Scholar] [CrossRef] [Green Version]
  59. Vempati, P.; Popel, A.S.; Mac Gabhann, F. Extracellular Regulation of VEGF: Isoforms, Proteolysis, and Vascular Patterning. Cytokine Growth Factor Rev. 2014, 25, 1–19. [Google Scholar] [CrossRef] [Green Version]
  60. Kariya, T.; Nishimura, H.; Mizuno, M.; Suzuki, Y.; Matsukawa, Y.; Sakata, F.; Maruyama, S.; Takei, Y.; Ito, Y. TGF-Β1-VEGF-A Pathway Induces Neoangiogenesis with Peritoneal Fibrosis in Patients Undergoing Peritoneal Dialysis. Am. J. Physiol. Renal. Physiol. 2018, 314, F167–F180. [Google Scholar] [CrossRef]
  61. Pérez-Lozano, M.L.; Sandoval, P.; Rynne-Vidal, A.; Aguilera, A.; Jiménez-Heffernan, J.A.; Albar-Vizcaíno, P.; Majano, P.L.; Sánchez-Tomero, J.A.; Selgas, R.; López-Cabrera, M. Functional Relevance of the Switch of VEGF Receptors/Co-Receptors during Peritoneal Dialysis-Induced Mesothelial to Mesenchymal Transition. PLoS ONE 2013, 8, e60776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Massagué, J. How Cells Read TGF-Beta Signals. Nat. Rev. Mol. Cell Biol. 2000, 1, 169–178. [Google Scholar] [CrossRef] [PubMed]
  63. Shi, Y.; Massagué, J. Mechanisms of TGF-Beta Signaling from Cell Membrane to the Nucleus. Cell 2003, 113, 685–700. [Google Scholar] [CrossRef] [Green Version]
  64. Tomino, Y. Mechanisms and Interventions in Peritoneal Fibrosis. Clin. Exp. Nephrol. 2012, 16, 109–114. [Google Scholar] [CrossRef] [PubMed]
  65. Kang, D.H.; Hong, Y.S.; Lim, H.J.; Choi, J.H.; Han, D.S.; Yoon, K.I. High Glucose Solution and Spent Dialysate Stimulate the Synthesis of Transforming Growth Factor-Beta1 of Human Peritoneal Mesothelial Cells: Effect of Cytokine Costimulation. Perit Dial. Int. 1999, 19, 221–230. [Google Scholar] [CrossRef]
  66. Naiki, Y.; Maeda, Y.; Matsuo, K.; Yonekawa, S.; Sakaguchi, M.; Iwamoto, I.; Hasegawa, H.; Kanamaru, A. Involvement of TGF-Beta Signal for Peritoneal Sclerosing in Continuous Ambulatory Peritoneal Dialysis. J. Nephrol. 2003, 16, 95–102. [Google Scholar] [PubMed]
  67. Balzer, M.S.; Helmke, A.; Ackermann, M.; Casper, J.; Dong, L.; Hiss, M.; Kiyan, Y.; Rong, S.; Timrott, K.; von Vietinghoff, S.; et al. Protein Kinase C Beta Deficiency Increases Glucose-Mediated Peritoneal Damage via M1 Macrophage Polarization and up-Regulation of Mesothelial Protein Kinase C Alpha. Nephrol. Dial. Transplant. 2019, 34, 947–960. [Google Scholar] [CrossRef]
  68. Witowski, J.; Wisniewska, J.; Korybalska, K.; Bender, T.O.; Breborowicz, A.; Gahl, G.M.; Frei, U.; Passlick-Deetjen, J.; Jörres, A. Prolonged Exposure to Glucose Degradation Products Impairs Viability and Function of Human Peritoneal Mesothelial Cells. J. Am. Soc. Nephrol. 2001, 12, 2434–2441. [Google Scholar] [CrossRef]
  69. Kim, Y.S.; Kim, B.C.; Song, C.Y.; Hong, H.K.; Moon, K.C.; Lee, H.S. Advanced Glycosylation End Products Stimulate Collagen MRNA Synthesis in Mesangial Cells Mediated by Protein Kinase C and Transforming Growth Factor-Beta. J. Lab. Clin. Med. 2001, 138, 59–68. [Google Scholar] [CrossRef]
  70. Mlambo, N.C.; Hylander, B.; Brauner, A. Increased Levels of Transforming Growth Factor Beta 1 and Basic Fibroblast Growth Factor in Patients on CAPD: A Study during Non-Infected Steady State and Peritonitis. Inflammation 1999, 23, 131–139. [Google Scholar] [CrossRef]
  71. Wang, T.; Ghen, Y.G.; Ye, R.G.; Mai, W.Y.; Zhen, Z.H.; Li, H.Q. Enhanced Expression of TGF-Beta 1 by Peritoneal Macrophages in CAPD Patients. Adv. Perit. Dial. 1995, 11, 11–14. [Google Scholar] [PubMed]
  72. Margetts, P.J.; Kolb, M.; Galt, T.; Hoff, C.M.; Shockley, T.R.; Gauldie, J. Gene Transfer of Transforming Growth Factor-Beta1 to the Rat Peritoneum: Effects on Membrane Function. J. Am. Soc. Nephrol. 2001, 12, 2029–2039. [Google Scholar] [CrossRef] [PubMed]
  73. Margetts, P.J.; Hoff, C.; Liu, L.; Korstanje, R.; Walkin, L.; Summers, A.; Herrick, S.; Brenchley, P. Transforming Growth Factor β-Induced Peritoneal Fibrosis Is Mouse Strain Dependent. Nephrol. Dial. Transplant. 2013, 28, 2015–2027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Yakymovych, I.; Ten Dijke, P.; Heldin, C.H.; Souchelnytskyi, S. Regulation of Smad Signaling by Protein Kinase C. FASEB J. 2001, 15, 553–555. [Google Scholar] [CrossRef] [PubMed]
  75. Moustakas, A.; Souchelnytskyi, S.; Heldin, C.H. Smad Regulation in TGF-Beta Signal Transduction. J. Cell Sci. 2001, 114 Pt 24, 4359–4369. [Google Scholar] [CrossRef]
  76. Nie, J.; Dou, X.; Hao, W.; Wang, X.; Peng, W.; Jia, Z.; Chen, W.; Li, X.; Luo, N.; Lan, H.Y.; et al. Smad7 Gene Transfer Inhibits Peritoneal Fibrosis. Kidney Int. 2007, 72, 1336–1344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Peng, W.; Dou, X.; Hao, W.; Zhou, Q.; Tang, R.; Nie, J.; Lan, H.Y.; Yu, X. Smad7 Gene Transfer Attenuates Angiogenesis in Peritoneal Dialysis Rats. Nephrology 2013, 18, 138–147. [Google Scholar] [CrossRef]
  78. Sun, Y.; Zhu, F.; Yu, X.; Nie, J.; Huang, F.; Li, X.; Luo, N.; Lan, H.Y.; Wang, Y. Treatment of Established Peritoneal Fibrosis by Gene Transfer of Smad7 in a Rat Model of Peritoneal Dialysis. Am. J. Nephrol. 2009, 30, 84–94. [Google Scholar] [CrossRef] [Green Version]
  79. Loureiro, J.; Schilte, M.; Aguilera, A.; Albar-Vizcaíno, P.; Ramírez-Huesca, M.; Pérez-Lozano, M.L.; González-Mateo, G.; Aroeira, L.S.; Selgas, R.; Mendoza, L.; et al. BMP-7 Blocks Mesenchymal Conversion of Mesothelial Cells and Prevents Peritoneal Damage Induced by Dialysis Fluid Exposure. Nephrol. Dial. Transplant. 2010, 25, 1098–1108. [Google Scholar] [CrossRef] [Green Version]
  80. Silva, F.M.O.; Costalonga, E.C.; Silva, C.; Carreira, A.C.O.; Gomes, S.A.; Sogayar, M.C.; Fanelli, C.; Noronha, I.L. Tamoxifen and Bone Morphogenic Protein-7 Modulate Fibrosis and Inflammation in the Peritoneal Fibrosis Model Developed in Uremic Rats. Mol. Med. 2019, 25, 41. [Google Scholar] [CrossRef] [Green Version]
  81. Yu, M.-A.; Shin, K.-S.; Kim, J.H.; Kim, Y.-I.; Chung, S.S.; Park, S.-H.; Kim, Y.-L.; Kang, D.-H. HGF and BMP-7 Ameliorate High Glucose-Induced Epithelial-to-Mesenchymal Transition of Peritoneal Mesothelium. J. Am. Soc. Nephrol. 2009, 20, 567–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Matsuo, H.; Tamura, M.; Kabashima, N.; Serino, R.; Tokunaga, M.; Shibata, T.; Matsumoto, M.; Aijima, M.; Oikawa, S.; Anai, H.; et al. Prednisolone Inhibits Hyperosmolarity-Induced Expression of MCP-1 via NF-KappaB in Peritoneal Mesothelial Cells. Kidney Int. 2006, 69, 736–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Patel, P.; Sekiguchi, Y.; Oh, K.-H.; Patterson, S.E.; Kolb, M.R.J.; Margetts, P.J. Smad3-Dependent and -Independent Pathways Are Involved in Peritoneal Membrane Injury. Kidney Int. 2010, 77, 319–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Liu, Q.; Mao, H.; Nie, J.; Chen, W.; Yang, Q.; Dong, X.; Yu, X. Transforming Growth Factor {beta}1 Induces Epithelial-Mesenchymal Transition by Activating the JNK-Smad3 Pathway in Rat Peritoneal Mesothelial Cells. Perit. Dial. Int. 2008, 28 (Suppl. 3), S88–S95. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, Q.; Zhang, Y.; Mao, H.; Chen, W.; Luo, N.; Zhou, Q.; Chen, W.; Yu, X. A Crosstalk between the Smad and JNK Signaling in the TGF-β-Induced Epithelial-Mesenchymal Transition in Rat Peritoneal Mesothelial Cells. PLoS ONE 2012, 7, e32009. [Google Scholar] [CrossRef] [Green Version]
  86. Kokubo, S.; Sakai, N.; Furuichi, K.; Toyama, T.; Kitajima, S.; Okumura, T.; Matsushima, K.; Kaneko, S.; Wada, T. Activation of P38 Mitogen-Activated Protein Kinase Promotes Peritoneal Fibrosis by Regulating Fibrocytes. Perit. Dial. Int. 2012, 32, 10–19. [Google Scholar] [CrossRef] [Green Version]
  87. Zhang, Y.; Huang, Q.; Chen, Y.; Peng, X.; Wang, Y.; Li, S.; Wu, J.; Luo, C.; Gong, W.; Yin, B.; et al. Parthenolide, an NF-ΚB Inhibitor, Alleviates Peritoneal Fibrosis by Suppressing the TGF-β/Smad Pathway. Int. Immunopharmacol. 2020, 78, 106064. [Google Scholar] [CrossRef]
  88. Krediet, R.T. Acquired Decline in Ultrafiltration in Peritoneal Dialysis: The Role of Glucose. J. Am. Soc. Nephrol. 2021, 32, 2408–2415. [Google Scholar] [CrossRef]
  89. Frazier, K.; Williams, S.; Kothapalli, D.; Klapper, H.; Grotendorst, G.R. Stimulation of Fibroblast Cell Growth, Matrix Production, and Granulation Tissue Formation by Connective Tissue Growth Factor. J. Investig. Dermatol. 1996, 107, 404–411. [Google Scholar] [CrossRef] [Green Version]
  90. Perbal, B. CCN Proteins: Multifunctional Signalling Regulators. Lancet 2004, 363, 62–64. [Google Scholar] [CrossRef]
  91. Grotendorst, G.R.; Okochi, H.; Hayashi, N. A Novel Transforming Growth Factor Beta Response Element Controls the Expression of the Connective Tissue Growth Factor Gene. Cell Growth Differ. 1996, 7, 469–480. [Google Scholar] [PubMed]
  92. Holmes, A.; Abraham, D.J.; Sa, S.; Shiwen, X.; Black, C.M.; Leask, A. CTGF and SMADs, Maintenance of Scleroderma Phenotype Is Independent of SMAD Signaling. J. Biol. Chem. 2001, 276, 10594–10601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Wang, S.; Denichilo, M.; Brubaker, C.; Hirschberg, R. Connective Tissue Growth Factor in Tubulointerstitial Injury of Diabetic Nephropathy. Kidney Int. 2001, 60, 96–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zarrinkalam, K.H.; Stanley, J.M.; Gray, J.; Oliver, N.; Faull, R.J. Connective Tissue Growth Factor and Its Regulation in the Peritoneal Cavity of Peritoneal Dialysis Patients. Kidney Int. 2003, 64, 331–338. [Google Scholar] [CrossRef]
  95. Mizutani, M.; Ito, Y.; Mizuno, M.; Nishimura, H.; Suzuki, Y.; Hattori, R.; Matsukawa, Y.; Imai, M.; Oliver, N.; Goldschmeding, R.; et al. Connective Tissue Growth Factor (CTGF/CCN2) Is Increased in Peritoneal Dialysis Patients with High Peritoneal Solute Transport Rate. Am. J. Physiol. Renal. Physiol. 2010, 298, F721–F733. [Google Scholar] [CrossRef] [Green Version]
  96. Sakamoto, N.; Sugimura, K.; Kawashima, H.; Tsuchida, K.; Takemoto, Y.; Naganuma, T.; Tatsumi, S.; Nakatani, T. Influence of Glucose and Inflammatory Cytokines on TGF-Beta1 and CTGF MRNA Expressions in Human Peritoneal Mesothelial Cells. Int. J. Mol. Med. 2005, 15, 907–911. [Google Scholar]
  97. Abrahams, A.C.; Habib, S.M.; Dendooven, A.; Riser, B.L.; van der Veer, J.W.; Toorop, R.J.; Betjes, M.G.H.; Verhaar, M.C.; Watson, C.J.E.; Nguyen, T.Q.; et al. Patients with Encapsulating Peritoneal Sclerosis Have Increased Peritoneal Expression of Connective Tissue Growth Factor (CCN2), Transforming Growth Factor-Β1, and Vascular Endothelial Growth Factor. PLoS ONE 2014, 9, e112050. [Google Scholar] [CrossRef] [Green Version]
  98. Leung, J.C.K.; Chan, L.Y.Y.; Tam, K.Y.; Tang, S.C.W.; Lam, M.F.; Cheng, A.S.; Chu, K.M.; Lai, K.N. Regulation of CCN2/CTGF and Related Cytokines in Cultured Peritoneal Cells under Conditions Simulating Peritoneal Dialysis. Nephrol. Dial. Transplant. 2009, 24, 458–469. [Google Scholar] [CrossRef] [Green Version]
  99. Toda, N.; Mori, K.; Kasahara, M.; Koga, K.; Ishii, A.; Mori, K.P.; Osaki, K.; Mukoyama, M.; Yanagita, M.; Yokoi, H. Deletion of Connective Tissue Growth Factor Ameliorates Peritoneal Fibrosis by Inhibiting Angiogenesis and Inflammation. Nephrol. Dial. Transplant. 2018, 33, 943–953. [Google Scholar] [CrossRef]
  100. Toda, N.; Mukoyama, M.; Yanagita, M.; Yokoi, H. CTGF in Kidney Fibrosis and Glomerulonephritis. Inflamm. Regen. 2018, 38, 14. [Google Scholar] [CrossRef]
  101. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Li, X.Y.; Wu, J.; Luo, D.; Chen, W.X.; Zhu, G.L.; Zhang, Y.X.; Bi, Z.M.; Feng, B.H. Effect of high glucose-based peritoneal dialysis fluids on NLRP3-IL-1β in human peritoneal mesothelial cells. Beijing Da Xue Xue Bao Yi Xue Ban 2017, 49, 954–960. [Google Scholar] [PubMed]
  103. Wu, J.; Li, X.; Zhu, G.; Zhang, Y.; He, M.; Zhang, J. The Role of Resveratrol-Induced Mitophagy/Autophagy in Peritoneal Mesothelial Cells Inflammatory Injury via NLRP3 Inflammasome Activation Triggered by Mitochondrial ROS. Exp. Cell Res. 2016, 341, 42–53. [Google Scholar] [CrossRef] [PubMed]
  104. Hishida, E.; Ito, H.; Komada, T.; Karasawa, T.; Kimura, H.; Watanabe, S.; Kamata, R.; Aizawa, E.; Kasahara, T.; Morishita, Y.; et al. Crucial Role of NLRP3 Inflammasome in the Development of Peritoneal Dialysis-Related Peritoneal Fibrosis. Sci. Rep. 2019, 9, 10363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Bertoli, S.V.; Barone, M.T.; Vago, L.; Bonetto, S.; De Vecchi, A.; Scalamogna, A.; Barbiano di Belgiojoso, G. Changes in Peritoneal Membrane after Continuous Ambulatory Peritoneal Dialysis--a Histopathological Study. Adv. Perit. Dial. 1999, 15, 28–31. [Google Scholar]
  106. Devuyst, O.; Margetts, P.J.; Topley, N. The Pathophysiology of the Peritoneal Membrane. J. Am. Soc. Nephrol. 2010, 21, 1077–1085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Yung, S.; Chan, T.M. Intrinsic Cells: Mesothelial Cells—Central Players in Regulating Inflammation and Resolution. Perit. Dial. Int. 2009, 29 (Suppl. 2), S21–S27. [Google Scholar] [CrossRef]
  108. Topley, N.; Jörres, A.; Luttmann, W.; Petersen, M.M.; Lang, M.J.; Thierauch, K.H.; Müller, C.; Coles, G.A.; Davies, M.; Williams, J.D. Human Peritoneal Mesothelial Cells Synthesize Interleukin-6: Induction by IL-1 Beta and TNF Alpha. Kidney Int. 1993, 43, 226–233. [Google Scholar] [CrossRef] [Green Version]
  109. Kato, S.; Yuzawa, Y.; Tsuboi, N.; Maruyama, S.; Morita, Y.; Matsuguchi, T.; Matsuo, S. Endotoxin-Induced Chemokine Expression in Murine Peritoneal Mesothelial Cells: The Role of Toll-like Receptor 4. J. Am. Soc. Nephrol. 2004, 15, 1289–1299. [Google Scholar]
  110. Yang, X.; Zhang, H.; Hang, Y.; Yan, H.; Lin, A.; Huang, J.; Ni, Z.; Qian, J.; Fang, W. Intraperitoneal Interleukin-6 Levels Predict Peritoneal Solute Transport Rate: A Prospective Cohort Study. Am. J. Nephrol. 2014, 39, 459–465. [Google Scholar] [CrossRef]
  111. Jiang, N.; Zhang, Z.; Fang, W.; Qian, J.; Mou, S.; Ni, Z. High Peritoneal Glucose Exposure Is Associated with Increased Incidence of Relapsing and Recurrent Bacterial Peritonitis in Patients Undergoing Peritoneal Dialysis. Blood Purif. 2015, 40, 72–78. [Google Scholar] [CrossRef] [PubMed]
  112. Feurino, L.W.; Zhang, Y.; Bharadwaj, U.; Zhang, R.; Li, F.; Fisher, W.E.; Brunicardi, F.C.; Chen, C.; Yao, Q.; Min, L. IL-6 Stimulates Th2 Type Cytokine Secretion and Upregulates VEGF and NRP-1 Expression in Pancreatic Cancer Cells. Cancer Biol. Ther. 2007, 6, 1096–1100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Yang, X.; Yan, H.; Jiang, N.; Yu, Z.; Yuan, J.; Ni, Z.; Fang, W. IL-6 Trans-Signaling Drives a STAT3-Dependent Pathway That Leads to Structural Alterations of the Peritoneal Membrane. Am. J. Physiol. Renal. Physiol. 2020, 318, F338–F353. [Google Scholar] [CrossRef] [PubMed]
  114. Witowski, J.; Pawlaczyk, K.; Breborowicz, A.; Scheuren, A.; Kuzlan-Pawlaczyk, M.; Wisniewska, J.; Polubinska, A.; Friess, H.; Gahl, G.M.; Frei, U.; et al. IL-17 Stimulates Intraperitoneal Neutrophil Infiltration through the Release of GRO Alpha Chemokine from Mesothelial Cells. J. Immunol. 2000, 165, 5814–5821. [Google Scholar] [CrossRef] [Green Version]
  115. Rodrigues, A.; Cabrita, A.; Maia, P.; Guimarães, S. Peritoneal Rest May Successfully Recover Ultrafiltration in Patients Who Develop Peritoneal Hyperpermeability with Time on Continuous Ambulatory Peritoneal Dialysis. Adv. Perit. Dial. 2002, 18, 78–80. [Google Scholar]
  116. Ferrantelli, E.; Liappas, G.; Vila Cuenca, M.; Keuning, E.D.; Foster, T.L.; Vervloet, M.G.; Lopéz-Cabrera, M.; Beelen, R.H.J. The Dipeptide Alanyl-Glutamine Ameliorates Peritoneal Fibrosis and Attenuates IL-17 Dependent Pathways during Peritoneal Dialysis. Kidney Int. 2016, 89, 625–635. [Google Scholar] [CrossRef]
  117. Henderson, J.; O’Reilly, S. The Emerging Role of Metabolism in Fibrosis. Trends Endocrinol. Metab. 2021, 32, 639–653. [Google Scholar] [CrossRef]
  118. Jiang, L.; Xiao, L.; Sugiura, H.; Huang, X.; Ali, A.; Kuro-o, M.; Deberardinis, R.J.; Boothman, D.A. Metabolic Reprogramming during TGFβ1-Induced Epithelial-to-Mesenchymal Transition. Oncogene 2015, 34, 3908–3916. [Google Scholar] [CrossRef] [Green Version]
  119. Kierans, S.J.; Taylor, C.T. Regulation of glycolysis by the hypoxia-inducible factor (HIF): Implications for cellular physiology. J. Physiol. 2021, 599, 23–37. [Google Scholar] [CrossRef]
  120. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [Green Version]
  121. Zhang, D.; Tang, Z.; Huang, H.; Zhou, G.; Cui, C.; Weng, Y.; Liu, W.; Kim, S.; Lee, S.; Perez-Neut, M.; et al. Metabolic Regulation of Gene Expression by Histone Lactylation. Nature 2019, 574, 575–580. [Google Scholar] [CrossRef] [PubMed]
  122. Yang, L.; Venneti, S.; Nagrath, D. Glutaminolysis: A Hallmark of Cancer Metabolism. Annu. Rev. Biomed. Eng. 2017, 19, 163–194. [Google Scholar] [CrossRef] [PubMed]
  123. Cheng, S.-C.; Quintin, J.; Cramer, R.A.; Shepardson, K.M.; Saeed, S.; Kumar, V.; Giamarellos-Bourboulis, E.J.; Martens, J.H.A.; Rao, N.A.; Aghajanirefah, A.; et al. MTOR- and HIF-1α-Mediated Aerobic Glycolysis as Metabolic Basis for Trained Immunity. Science 2014, 345, 1250684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Hewitson, T.D.; Smith, E.R. A Metabolic Reprogramming of Glycolysis and Glutamine Metabolism Is a Requisite for Renal Fibrogenesis-Why and How? Front. Physiol. 2021, 12, 645857. [Google Scholar] [CrossRef]
  125. Aufricht, C.; Beelen, R.; Eberl, M.; Fischbach, M.; Fraser, D.; Jörres, A.; Kratochwill, K.; LópezCabrera, M.; Rutherford, P.; Schmitt, C.-P.; et al. Biomarker Research to Improve Clinical Outcomes of Peritoneal Dialysis: Consensus of the European Training and Research in Peritoneal Dialysis (EuTRiPD) Network. Kidney Int. 2017, 92, 824–835. [Google Scholar] [CrossRef] [Green Version]
  126. Jones, S.A.; Fraser, D.J.; Fielding, C.A.; Jones, G.W. Interleukin-6 in Renal Disease and Therapy. Nephrol. Dial. Transplant. 2015, 30, 564–574. [Google Scholar] [CrossRef] [Green Version]
  127. Lopes Barreto, D.; Krediet, R.T. Current Status and Practical Use of Effluent Biomarkers in Peritoneal Dialysis Patients. Am. J. Kidney Dis. 2013, 62, 823–833. [Google Scholar] [CrossRef]
  128. Lopez-Anton, M.; Bowen, T.; Jenkins, R.H. MicroRNA Regulation of Peritoneal Cavity Homeostasis in Peritoneal Dialysis. Biomed. Res. Int. 2015, 2015, 929806. [Google Scholar] [CrossRef] [Green Version]
  129. Kondou, A.; Begou, O.; Dotis, J.; Karava, V.; Panteris, E.; Taparkou, A.; Gika, H.; Printza, N. Impact of Metabolomics Technologies on the Assessment of Peritoneal Membrane Profiles in Peritoneal Dialysis Patients: A Systematic Review. Metabolites 2022, 12, 145. [Google Scholar] [CrossRef]
  130. Corciulo, S.; Nicoletti, M.C.; Mastrofrancesco, L.; Milano, S.; Mastrodonato, M.; Carmosino, M.; Gerbino, A.; Corciulo, R.; Russo, R.; Svelto, M.; et al. AQP1-Containing Exosomes in Peritoneal Dialysis Effluent As Biomarker of Dialysis Efficiency. Cells 2019, 8, 330. [Google Scholar] [CrossRef] [Green Version]
  131. Szeto, C.C.; Johnson, D.W. Low GDP Solution and Glucose-Sparing Strategies for Peritoneal Dialysis. Semin. Nephrol. 2017, 37, 30–42. [Google Scholar] [CrossRef] [PubMed]
  132. Bartosova, M.; Schmitt, C.P. Biocompatible Peritoneal Dialysis: The Target Is Still Way Off. Front. Physiol. 2018, 9, 1853. [Google Scholar] [CrossRef] [PubMed]
  133. Blake, P.G. Is the Peritoneal Dialysis Biocompatibility Hypothesis Dead? Kidney Int. 2018, 94, 246–248. [Google Scholar] [CrossRef] [PubMed]
  134. Sugiyama, N.; Tawada, M.; Sun, T.; Suzuki, Y.; Kinashi, H.; Yamaguchi, M.; Katsuno, T.; Aten, J.; Vlahu, C.A.; van Kuppevelt, T.H.; et al. Low-GDP, pH-Neutral Solutions Preserve Peritoneal Endothelial Glycocalyx during Long-Term Peritoneal Dialysis. Clin. Exp. Nephrol. 2021, 25, 1035–1046. [Google Scholar] [CrossRef]
  135. Schaefer, B.; Bartosova, M.; Macher-Goeppinger, S.; Sallay, P.; Vörös, P.; Ranchin, B.; Vondrak, K.; Ariceta, G.; Zaloszyc, A.; Bayazit, A.K.; et al. Neutral PH and Low-Glucose Degradation Product Dialysis Fluids Induce Major Early Alterations of the Peritoneal Membrane in Children on Peritoneal Dialysis. Kidney Int. 2018, 94, 419–429. [Google Scholar] [CrossRef]
  136. Burkart, J. Metabolic Consequences of Peritoneal Dialysis. Semin. Dial. 2004, 17, 498–504. [Google Scholar] [CrossRef]
  137. Wang, I.-K.; Lin, C.-L.; Chen, H.-C.; Lin, S.-Y.; Chang, C.-T.; Yen, T.-H.; Sung, F.-C. Risk of New-Onset Diabetes in End-Stage Renal Disease Patients Undergoing Dialysis: Analysis from Registry Data of Taiwan. Nephrol. Dial. Transplant. 2018, 33, 670–675. [Google Scholar] [CrossRef]
  138. Jones, M.; Hagen, T.; Boyle, C.A.; Vonesh, E.; Hamburger, R.; Charytan, C.; Sandroni, S.; Bernard, D.; Piraino, B.; Schreiber, M.; et al. Treatment of Malnutrition with 1.1% Amino Acid Peritoneal Dialysis Solution: Results of a Multicenter Outpatient Study. Am. J. Kidney Dis. 1998, 32, 761–769. [Google Scholar] [CrossRef]
  139. Johnson, D.W.; Agar, J.; Collins, J.; Disney, A.; Harris, D.C.H.; Ibels, L.; Irish, A.; Saltissi, D.; Suranyi, M. Recommendations for the Use of Icodextrin in Peritoneal Dialysis Patients. Nephrology 2003, 8, 1–7. [Google Scholar] [CrossRef]
  140. Holmes, C.J. Glucotoxicity in Peritoneal Dialysis--Solutions for the Solution! Adv. Chronic Kidney Dis. 2007, 14, 269–278. [Google Scholar] [CrossRef]
  141. Goossen, K.; Becker, M.; Marshall, M.R.; Bühn, S.; Breuing, J.; Firanek, C.A.; Hess, S.; Nariai, H.; Sloand, J.A.; Yao, Q.; et al. Icodextrin Versus Glucose Solutions for the Once-Daily Long Dwell in Peritoneal Dialysis: An Enriched Systematic Review and Meta-Analysis of Randomized Controlled Trials. Am. J. Kidney Dis. 2020, 75, 830–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Higuchi, C.; Kuriyma, J.; Sakura, H. Effect of Neutral PH Icodextrin Peritoneal Dialysis Fluid on Mesothelial Cells. Ther. Apher. Dial. 2018, 22, 656–661. [Google Scholar] [CrossRef] [PubMed]
  143. Asola, M.; Virtanen, K.; Någren, K.; Helin, S.; Taittonen, M.; Kastarinen, H.; Anderstam, B.; Knuuti, J.; Metsärinne, K.; Nuutila, P. Amino-Acid-Based Peritoneal Dialysis Solution Improves Amino-Acid Transport into Skeletal Muscle. Kidney Int. Suppl. 2008, 73, S131–S136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Plum, J.; Erren, C.; Fieseler, C.; Kirchgessner, J.; Passlick-Deetjen, J.; Grabensee, B. An Amino Acid-Based Peritoneal Dialysis Fluid Buffered with Bicarbonate versus Glucose/Bicarbonate and Glucose/Lactate Solutions: An Intraindividual Randomized Study. Perit. Dial. Int. 1999, 19, 418–428. [Google Scholar] [CrossRef] [PubMed]
  145. Park, M.S.; Heimbürger, O.; Bergström, J.; Waniewski, J.; Werynski, A.; Lindholm, B. Peritoneal Transport during Dialysis with Amino Acid-Based Solutions. Perit. Dial. Int. 1993, 13, 280–288. [Google Scholar] [CrossRef] [PubMed]
  146. Bonomini, M.; Zammit, V.; Divino-Filho, J.C.; Davies, S.J.; Di Liberato, L.; Arduini, A.; Lambie, M. The Osmo-Metabolic Approach: A Novel and Tantalizing Glucose-Sparing Strategy in Peritoneal Dialysis. J. Nephrol. 2021, 34, 503–519. [Google Scholar] [CrossRef]
  147. Longo, N.; Frigeni, M.; Pasquali, M. Carnitine Transport and Fatty Acid Oxidation. Biochim. Biophys. Acta 2016, 1863, 2422–2435. [Google Scholar] [CrossRef]
  148. Arduini, A.; Bonomini, M.; Savica, V.; Amato, A.; Zammit, V. Carnitine in Metabolic Disease: Potential for Pharmacological Intervention. Pharmacol. Ther. 2008, 120, 149–156. [Google Scholar] [CrossRef]
  149. Wang, Y.M.; van Eys, J. Nutritional Significance of Fructose and Sugar Alcohols. Annu. Rev. Nutr. 1981, 1, 437–475. [Google Scholar] [CrossRef]
  150. Mäkinen, K.K. Can the Pentitol-Hexitol Theory Explain the Clinical Observations Made with Xylitol? Med. Hypotheses 2000, 54, 603–613. [Google Scholar] [CrossRef]
  151. Wölnerhanssen, B.K.; Cajacob, L.; Keller, N.; Doody, A.; Rehfeld, J.F.; Drewe, J.; Peterli, R.; Beglinger, C.; Meyer-Gerspach, A.C. Gut Hormone Secretion, Gastric Emptying, and Glycemic Responses to Erythritol and Xylitol in Lean and Obese Subjects. Am. J. Physiol. Endocrinol. Metab. 2016, 310, E1053–E1061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Gill, V.; Kumar, V.; Singh, K.; Kumar, A.; Kim, J.-J. Advanced Glycation End Products (AGEs) May Be a Striking Link Between Modern Diet and Health. Biomolecules 2019, 9, 888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Gaggiotti, E.; Arduini, A.; Bonomini, M.; Valentini, G.; Sacchi, G.; Sansoni, E.; Salvo, D.; Di Paolo, N. Prevention of Peritoneal Sclerosis: A New Proposal to Substitute Glucose with Carnitine Dialysis Solution (Biocompatibility Testing in Vitro and in Rabbits). Int. J. Artif. Organs 2005, 28, 177–187. [Google Scholar] [CrossRef] [PubMed]
  154. Bonomini, M.; Pandolfi, A.; Di Liberato, L.; Di Silvestre, S.; Cnops, Y.; Di Tomo, P.; D’Arezzo, M.; Monaco, M.P.; Giardinelli, A.; Di Pietro, N.; et al. L-Carnitine Is an Osmotic Agent Suitable for Peritoneal Dialysis. Kidney Int. 2011, 80, 645–654. [Google Scholar] [CrossRef] [Green Version]
  155. Bazzato, G.; Coli, U.; Landini, S.; Fracasso, A.; Morachiello, P.; Righetto, F.; Scanferla, F.; Onesti, G. Xylitol as Osmotic Agent in CAPD: An Alternative to Glucose for Uremic Diabetic Patients? Trans. Am. Soc. Artif. Intern. Organs 1982, 28, 280–286. [Google Scholar]
  156. Bonomini, M.; Di Liberato, L.; Zammit, V.; Arduini, A. Current Opinion on Usage of L-Carnitine in End-Stage Renal Disease Patients on Peritoneal Dialysis. Molecules 2019, 24, 3449. [Google Scholar] [CrossRef] [Green Version]
  157. Bonomini, M.; Di Liberato, L.; Del Rosso, G.; Stingone, A.; Marinangeli, G.; Consoli, A.; Bertoli, S.; De Vecchi, A.; Bosi, E.; Russo, R.; et al. Effect of an L-Carnitine-Containing Peritoneal Dialysate on Insulin Sensitivity in Patients Treated with CAPD: A 4-Month, Prospective, Multicenter Randomized Trial. Am. J. Kidney Dis. 2013, 62, 929–938. [Google Scholar] [CrossRef]
  158. Bonomini, M.; Di Silvestre, S.; Di Tomo, P.; Di Pietro, N.; Mandatori, D.; Di Liberato, L.; Sirolli, V.; Chiarelli, F.; Indiveri, C.; Pandolfi, A.; et al. Effect of Peritoneal Dialysis Fluid Containing Osmo-Metabolic Agents on Human Endothelial Cells. Drug Des. Devel. Ther. 2016, 10, 3925–3932. [Google Scholar] [CrossRef] [Green Version]
  159. Piccapane, F.; Bonomini, M.; Castellano, G.; Gerbino, A.; Carmosino, M.; Svelto, M.; Arduini, A.; Procino, G. A Novel Formulation of Glucose-Sparing Peritoneal Dialysis Solutions with l-Carnitine Improves Biocompatibility on Human Mesothelial Cells. Int. J. Mol. Sci. 2020, 22, 123. [Google Scholar] [CrossRef]
  160. Rago, C.; Lombardi, T.; Di Fulvio, G.; Di Liberato, L.; Arduini, A.; Divino-Filho, J.C.; Bonomini, M. A New Peritoneal Dialysis Solution Containing L-Carnitine and Xylitol for Patients on Continuous Ambulatory Peritoneal Dialysis: First Clinical Experience. Toxins 2021, 13, 174. [Google Scholar] [CrossRef]
  161. Rodrigues-Díez, R.; Aroeira, L.S.; Orejudo, M.; Bajo, M.-A.; Heffernan, J.J.; Rodrigues-Díez, R.R.; Rayego-Mateos, S.; Ortiz, A.; Gonzalez-Mateo, G.; López-Cabrera, M.; et al. IL-17A Is a Novel Player in Dialysis-Induced Peritoneal Damage. Kidney Int. 2014, 86, 303–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Gozdzikiewicz, J.; Borawski, J.; Mysliwiec, M. Pleiotropic Effects of Heparin and Heparinoids in Peritoneal Dialysis. Clin. Appl. Thromb. Hemost. 2009, 15, 92–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Bazzato, G.; Fracasso, A.; Gambaro, G.; Baggio, B. Use of Glycosaminoglycans to Increase Efficiency of Long-Term Continuous Peritoneal Dialysis. Lancet 1995, 346, 740–741. [Google Scholar] [CrossRef] [PubMed]
  164. Sjøland, J.A.; Smith Pedersen, R.; Jespersen, J.; Gram, J. Intraperitoneal Heparin Reduces Peritoneal Permeability and Increases Ultrafiltration in Peritoneal Dialysis Patients. Nephrol. Dial. Transplant. 2004, 19, 1264–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Braide, M.; Haraldsson, B.; Persson, U. Citrate Supplementation of PD Fluid: Effects on Net Ultrafiltration and Clearance of Small Solutes in Single Dwells. Nephrol. Dial. Transplant. 2009, 24, 286–292. [Google Scholar] [CrossRef] [Green Version]
  166. Kratochwill, K.; Boehm, M.; Herzog, R.; Gruber, K.; Lichtenauer, A.M.; Kuster, L.; Csaicsich, D.; Gleiss, A.; Alper, S.L.; Aufricht, C.; et al. Addition of Alanyl-Glutamine to Dialysis Fluid Restores Peritoneal Cellular Stress Responses—A First-In-Man Trial. PLoS ONE 2016, 11, e0165045. [Google Scholar] [CrossRef] [Green Version]
  167. Herzog, R.; Bartosova, M.; Tarantino, S.; Wagner, A.; Unterwurzacher, M.; Sacnun, J.M.; Lichtenauer, A.M.; Kuster, L.; Schaefer, B.; Alper, S.L.; 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, 1678. [Google Scholar] [CrossRef]
  168. Wiesenhofer, F.M.; Herzog, R.; Boehm, M.; Wagner, A.; Unterwurzacher, M.; Kasper, D.C.; Alper, S.L.; Vychytil, A.; Aufricht, C.; Kratochwill, K. Targeted Metabolomic Profiling of Peritoneal Dialysis Effluents Shows Anti-Oxidative Capacity of Alanyl-Glutamine. Front. Physiol. 2018, 9, 1961. [Google Scholar] [CrossRef] [Green Version]
  169. Ohsawa, I.; Ishikawa, M.; Takahashi, K.; Watanabe, M.; Nishimaki, K.; Yamagata, K.; Katsura, K.-I.; Katayama, Y.; Asoh, S.; Ohta, S. Hydrogen Acts as a Therapeutic Antioxidant by Selectively Reducing Cytotoxic Oxygen Radicals. Nat. Med. 2007, 13, 688–694. [Google Scholar] [CrossRef]
  170. Ichihara, M.; Sobue, S.; Ito, M.; Ito, M.; Hirayama, M.; Ohno, K. Beneficial Biological Effects and the Underlying Mechanisms of Molecular Hydrogen—Comprehensive Review of 321 Original Articles. Med. Gas Res. 2015, 5, 12. [Google Scholar] [CrossRef] [Green Version]
  171. Terawaki, H.; Hayashi, Y.; Zhu, W.-J.; Matsuyama, Y.; Terada, T.; Kabayama, S.; Watanabe, T.; Era, S.; Sato, B.; Nakayama, M. Transperitoneal Administration of Dissolved Hydrogen for Peritoneal Dialysis Patients: A Novel Approach to Suppress Oxidative Stress in the Peritoneal Cavity. Med. Gas Res. 2013, 3, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Nakayama, M.; Zhu, W.-J.; Watanabe, K.; Gibo, A.; Sherif, A.M.; Kabayama, S.; Ito, S. Dissolved Molecular Hydrogen (H(2)) in Peritoneal Dialysis (PD) Solutions Preserves Mesothelial Cells and Peritoneal Membrane Integrity. BMC Nephrol. 2017, 18, 327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Lu, H.; Chen, W.; Liu, W.; Si, Y.; Zhao, T.; Lai, X.; Kang, Z.; Sun, X.; Guo, Z. Molecular Hydrogen Regulates PTEN-AKT-MTOR Signaling via ROS to Alleviate Peritoneal Dialysis-Related Peritoneal Fibrosis. FASEB J. 2020, 34, 4134–4146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Herzog, R.; Sacnun, J.M.; González-Mateo, G.; Bartosova, M.; Bialas, K.; Wagner, A.; Unterwurzacher, M.; Sobieszek, I.J.; Daniel-Fischer, L.; Rusai, K.; et al. Lithium Preserves Peritoneal Membrane Integrity by Suppressing Mesothelial Cell AB-Crystallin. Sci. Transl. Med. 2021, 13, eaaz9705. [Google Scholar] [CrossRef] [PubMed]
  175. Diamond, M.P.; El-Hammady, E.; Wang, R.; Saed, G. Regulation of Transforming Growth Factor-Beta, Type III Collagen, and Fibronectin by Dichloroacetic Acid in Human Fibroblasts from Normal Peritoneum and Adhesions. Fertil. Steril. 2003, 79, 1161–1167. [Google Scholar] [CrossRef]
  176. Tian, L.; Wu, D.; Dasgupta, A.; Chen, K.-H.; Mewburn, J.; Potus, F.; Lima, P.D.A.; Hong, Z.; Zhao, Y.-Y.; Hindmarch, C.C.T.; 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, 1723–1745. [Google Scholar] [CrossRef]
  177. Pala, H.G.; Pala, E.E.; Artunc Ulkumen, B.; Erbas, O. Protective Effects of Dichloroacetic Acid on Endometrial Injury and Ovarian Reserve in an Experimental Rat Model of Diabetes Mellitus. J. Obstet. Gynaecol. Res. 2021, 47, 4319–4328. [Google Scholar] [CrossRef]
  178. 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. Renal. Physiol. 2019, 316, F1162–F1172. [Google Scholar] [CrossRef]
  179. Goodwin, J.; Choi, H.; Hsieh, M.-H.; Neugent, M.L.; Ahn, J.-M.; Hayenga, H.N.; Singh, P.K.; Shackelford, D.B.; Lee, I.-K.; Shulaev, V.; et al. Targeting Hypoxia-Inducible Factor-1α/Pyruvate Dehydrogenase Kinase 1 Axis by Dichloroacetate Suppresses Bleomycin-Induced Pulmonary Fibrosis. Am. J. Respir. Cell Mol. Biol. 2018, 58, 216–231. [Google Scholar] [CrossRef]
  180. Lambie, M.; Bonomini, M.; Davies, S.J.; Accili, D.; Arduini, A.; Zammit, V. Insulin Resistance in Cardiovascular Disease, Uremia, and Peritoneal Dialysis. Trends Endocrinol. Metab. 2021, 32, 721–730. [Google Scholar] [CrossRef]
  181. Pajak, B.; Siwiak, E.; Sołtyka, M.; Priebe, A.; Zieliński, R.; Fokt, I.; Ziemniak, M.; Jaśkiewicz, A.; Borowski, R.; Domoradzki, T.; et al. 2-Deoxy-d-Glucose and Its Analogs: From Diagnostic to Therapeutic Agents. Int. J. Mol. Sci. 2019, 21, 234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Wilson, R.B. Hypoxia, Cytokines and Stromal Recruitment: Parallels between Pathophysiology of Encapsulating Peritoneal Sclerosis, Endometriosis and Peritoneal Metastasis. Pleura Peritoneum 2018, 3, 20180103. [Google Scholar] [CrossRef] [PubMed]
  183. Si, M.; Wang, Q.; Li, Y.; Lin, H.; Luo, D.; Zhao, W.; Dou, X.; Liu, J.; Zhang, H.; Huang, Y.; et al. Inhibition of Hyperglycolysis in Mesothelial Cells Prevents Peritoneal Fibrosis. Sci. Transl. Med. 2019, 11, eaav5341. [Google Scholar] [CrossRef]
  184. Horne, A.W.; Ahmad, S.F.; Carter, R.; Simitsidellis, I.; Greaves, E.; Hogg, C.; Morton, N.M.; Saunders, P.T.K. Repurposing Dichloroacetate for the Treatment of Women with Endometriosis. Proc. Natl. Acad. Sci. USA 2019, 116, 25389–25391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Laussel, C.; Léon, S. Cellular Toxicity of the Metabolic Inhibitor 2-Deoxyglucose and Associated Resistance Mechanisms. Biochem. Pharmacol. 2020, 182, 114213. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Graphical representation of metabolic control of fibrosis. Several metabolic processes such as glycolysis, glutaminolysis, and fatty acid oxidation contribute to the deposition and other ECM components. High glucose levels activate glycolysis directly by increasing HIF-1a expression, which in turn amplifies the production of TGF-b1. The latter would not only further sustain high glycolytic rates but also fibrogenesis. The increased production of lactate sustains macrophages polarization toward an inflammatory phenotype, which worsens the fibrosis. In addition, the increased glycolytic rate makes glycolytic intermediates available in larger quantities, contributing to the synthesis of amino acid substrates for collagen synthesis. Moreover, glutamine has a role in collagen synthesis, but it can also contribute to ATP production through oxidative phosphorylation. A key metabolic switch in maintaining a chronically activated fibrogenic state is the pyruvate dehydrogenase complex (PDH).
Figure 1. Graphical representation of metabolic control of fibrosis. Several metabolic processes such as glycolysis, glutaminolysis, and fatty acid oxidation contribute to the deposition and other ECM components. High glucose levels activate glycolysis directly by increasing HIF-1a expression, which in turn amplifies the production of TGF-b1. The latter would not only further sustain high glycolytic rates but also fibrogenesis. The increased production of lactate sustains macrophages polarization toward an inflammatory phenotype, which worsens the fibrosis. In addition, the increased glycolytic rate makes glycolytic intermediates available in larger quantities, contributing to the synthesis of amino acid substrates for collagen synthesis. Moreover, glutamine has a role in collagen synthesis, but it can also contribute to ATP production through oxidative phosphorylation. A key metabolic switch in maintaining a chronically activated fibrogenic state is the pyruvate dehydrogenase complex (PDH).
Ijms 23 04831 g001
Figure 2. Metabolic strategies to control fibrosis could be based on inhibiting glycolysis, fatty acid, and pyruvate oxidation. Glycolysis can be modulated by inhibiting hexokinase 2 by 2-deoxyglucose, though the safety of this approach must be proved. The alternative strategy is coupling glycolysis with the Krebs cycle by inhibiting PDH Kinase using DCA or increasing PDH activity by reducing the intramitochondrial acetyl-CoA pool using L-carnitine.
Figure 2. Metabolic strategies to control fibrosis could be based on inhibiting glycolysis, fatty acid, and pyruvate oxidation. Glycolysis can be modulated by inhibiting hexokinase 2 by 2-deoxyglucose, though the safety of this approach must be proved. The alternative strategy is coupling glycolysis with the Krebs cycle by inhibiting PDH Kinase using DCA or increasing PDH activity by reducing the intramitochondrial acetyl-CoA pool using L-carnitine.
Ijms 23 04831 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Masola, V.; Bonomini, M.; Borrelli, S.; Di Liberato, L.; Vecchi, L.; Onisto, M.; Gambaro, G.; Palumbo, R.; Arduini, A. Fibrosis of Peritoneal Membrane as Target of New Therapies in Peritoneal Dialysis. Int. J. Mol. Sci. 2022, 23, 4831. https://doi.org/10.3390/ijms23094831

AMA Style

Masola V, Bonomini M, Borrelli S, Di Liberato L, Vecchi L, Onisto M, Gambaro G, Palumbo R, Arduini A. Fibrosis of Peritoneal Membrane as Target of New Therapies in Peritoneal Dialysis. International Journal of Molecular Sciences. 2022; 23(9):4831. https://doi.org/10.3390/ijms23094831

Chicago/Turabian Style

Masola, Valentina, Mario Bonomini, Silvio Borrelli, Lorenzo Di Liberato, Luigi Vecchi, Maurizio Onisto, Giovanni Gambaro, Roberto Palumbo, and Arduino Arduini. 2022. "Fibrosis of Peritoneal Membrane as Target of New Therapies in Peritoneal Dialysis" International Journal of Molecular Sciences 23, no. 9: 4831. https://doi.org/10.3390/ijms23094831

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop