Open access peer-reviewed chapter

Preservation of Peritoneal Membrane Structure and Function in Peritoneal Dialysis

Written By

Mathew George Kunthara

Submitted: 22 March 2023 Reviewed: 12 April 2023 Published: 29 July 2023

DOI: 10.5772/intechopen.111586

From the Edited Volume

Updates on Renal Replacement Therapy

Edited by Henry H.L. Wu

Chapter metrics overview

66 Chapter Downloads

View Full Metrics

Abstract

Peritoneal dialysis (PD) is a type of renal replacement therapy which is based on the use of peritoneum, which acts as a semipermeable membrane with diffusion and convection. Long term use can produce structural and functional changes of the membrane by the activation of the resident fibroblasts and infiltrating inflammatory cells, mesothelial to mesenchymal transition, further leading to fibrosis, angiogenesis and ultrafiltration failure. This is due to use of bioincompatible fluids, frequent peritoneal inflammation, uremic milieu and other multiple factors. The peritoneal fibrosis has two parts: fibrosis and inflammation, which induces each other via TGF/SMAD pathway and IL-6 signaling, respectively. The advent of newer biocompatible fluids along with additives has significantly reduced the production of glucose degradation products (GDPs). In addition, the identification of the biomarkers in peritoneal effluent is necessary, which, after being correlated with peritoneal biopsy, may help us to guide future studies and assessment of the efficacy of therapeutic interventions. Various interventions are being tried based on experimental studies from animal models, pharmacology and gene therapy with promising results, with new insights in near future. This article reviews the main aspects associated with the functional and structural alterations related to PD and discusses interventions whereby we may prevent them to preserve the peritoneal membrane.

Keywords

  • peritoneal dialysis
  • ultrafiltration
  • encapsulating peritoneal sclerosis
  • mesothelial to mesenchymal transition (MMT)
  • VEGF (vascular endothelial growth factors)
  • GDP (glucose degradation products)

1. Introduction

Peritoneal dialysis (PD) is a life-sustaining therapy used by >100,000 patients with ESRD worldwide, accounting for approximately 10 to 15% of the dialysis population [1]. Despite these benefits, only a small number of dialysis patients receive PD, in Europe about 13% and in the USA about 10% and 6% in India [2, 3]. The major obstacles for a successful long-term PD are infections and the deleterious functional alterations in the peritoneal membrane following prolonged exposure to dialysis fluids; which is responsible for increased morbidity and mortality. These alterations, such as progressive fibrosis and vasculogenesis, leading to increased solute transport and ultrafiltration (UF) failure, are seen in more than 50% of patients on PD.

Rippe proposed the existence of three pores of different sizes in peritoneal membrane: a large pore of 100–200 Å corresponding to interendothelial cell clefts allowing transport of large molecular weight solutes; a small pore of 40–60 Å, which allows for transport of water and low molecular weight (LMW)solutes and an ultrasmall pore of 4–6 Å that allows for the passage of only water.

Advertisement

2. The normal peritoneal transport barrier

2.1 Distributed concept of ultrafiltration barrier

The three potential barriers to both solute and water are (1) anatomic peritoneum (2) cellular-interstitial matrix surrounding the blood vessels (3) capillary endothelium. Blood vessels are the main source of UF and the water flow from the capillaries to the interstitium depends on the difference between the capillary luminal pressures and the effective pressure on the interstitial side. The concentration profile occurs due to diffusion of the small solutes via the tissue interstitium along with simultaneous uptake into the capillaries. The largest gradients of osmotic pressure will therefore be across blood vessels closest to the peritoneum.

Figure 1A describes the distributed concept of UF barrier and Figure 1B shows changes in membrane [4].

Figure 1.

(A and B) Distributed concept of normal ultrafiltration barrier. Dextrose diffuses from dialysate into tissue and sets up an osmotic pressure profile (thick curved line).

The distributed model proposes that “the influence of a specific capillary on PD transport is the function of that capillary’s proximity to mesothelial to the dialysate interface”. The proliferation of vessels near the interface increases the “effective” peritoneal surface area; especially during peritonitis and following exposure to high glucose containing fluids.

2.2 Pore matrix concept of endothelial barrier

Flessner [4] has postulated a new concept which says that the large and small pores will be represented as a single entity with the difference in transport characteristics; being a function of the density of intercellular glycoprotein matrix; as in Figure 2. This additional layer of glycocalyx alters the microenvironment near the true, size-selective boundary.

Figure 2.

Pore-matrix concept of endothelial barrier incorporating the luminal glycocalyx [4].

Moreover, albumin concentration below the glycocalyx but above the tight junction is likely much lower than that in the interstitium. This is because the albumin is unable to diffuse against the ultrafiltrate flow through the gap in the glycocalyx. The glycocalyx density is decreased by perfusion of oxidized LDL, adenosine, ischemia reperfusion injury and TNF-α.

Advertisement

3. Natural history of peritoneal membrane in CAPD

The peritoneal fibrosis has two parts; fibrosis process itself and the inflammation which is promoted by the non-physiologic content of solutions and infections. In the fibrotic process, there is loss of mesothelial cells with fibroblastoid changes leading to mesothelial-to-mesenchymal transition mediated by TGF-ß (Transforming growth factor) and VEGF (Vascular endothelial growth factor) signaling pathways. The inflammation pathway is mediated by the IL-6 (Interleukin-6) and other chemokines. Both the pathways are interlinked to each other and will be potentiating each other.

Advertisement

4. Regulation of peritoneal inflammation and leukocyte trafficking

During acute episodes of peritonitis, there is early activation of proinflammatory cytokines (TNF-, IL-1, and IFN-) and rapid recruitment of neutrophils with subsequent replacement by monocytes. This initial influx of neutrophils is due to the expression of CXC chemokine, MIP-1/KC, and the release of sIL-6R which facilitates the formation of sIL-6R/IL-6 complexes. These trans-signaling complexes suppress the release of other CXC chemokines, ensuring clearance of neutrophils, and simultaneously promoting the secretion of the CC chemokines, such as monocyte chemoattractant protein 1 (MCP-1) and RANTES, triggering the recruitment of mononuclear leukocytes and regulate the process of apoptosis. The IL-6/sIL-6R signaling also selectively promotes T cell recruitment into the peritoneal membrane through a gp130-dependent, STAT1/3-dependent activation pathway (as shown in Figure 3).

Figure 3.

Molecular network that regulate EMT [5]. Courtesy: Gonzalez 2016. Abbreviations: TGF ß—Transforming growth factor ß; GF—growth factor; TKr—Tyrosine kinase receptor; IL-6—interleukin 6; and EMT/MMT—Epithelial/Mesothelial to mesenchymal transition.

The most consistent change observed in peritoneal tissues of a patient on PD is an increase in the sub-mesothelial thickness associated with peritoneal fibrosis. The use of non-physiologic PD solutions along with uremic milieu, has led to the production of advanced glycation end products (AGEs) in peritoneal tissues which induces vasculogenesis and fibrosis. The interaction between fibrosis and angiogenesis may occur at the level of inducing cytokines; TGF-ß leading to SMAD pathway and inflammatory cytokines induce VEGF and angiogenesis; this is how EMT (epithelial to mesenchymal transition)/MMT (mesothelial to mesenchymal transition) occurs (shown in Figure 4).

Figure 4.

Key events during EMT (Courtesy: [6]).

There are two pathologic types of PD related fibrosis. Most common type is simple peritoneal sclerosis which is seen in almost all patients. The other one is Encapsulating peritoneal fibrosis (EPS) that evolves rapidly with intense fibrosis and inflammation leading to life threatening visceral encapsulation (as shown in Figure 5).

Figure 5.

Natural history of peritoneal membrane changes (Courtesy: [5]).

Advertisement

5. Consequences of peritoneal fibrosis

The peritoneum is an acellular, avascular layer of tissue. Significant scarring of the peritoneum is often present after 6 or more years of CAPD. Solute transport is rapid across this avascular, acellular layer and uptake into abnormal blood capillaries is rapid. However, with the loss of the interstitial cell matrix and the increase in the distance of the blood capillaries from the peritoneum, the water transport to peritoneal cavity will be nearly zero [4]. Immunolocalization of collagen 1α-1 revealed that this protein was predominantly expressed in the sub-mesothelial compact zone of EPS peritoneal samples, whereas non-EPS patients exhibited diffuse and homogeneous Col1a-1 staining.

For more advanced peritoneal conditions with potential EPS development, EPS-prone states [7] is defined by (i) PD duration >3 years (ii) history of recurrent ± severe peritonitis (iii) presence of acquired UF failure or high-fast membrane transport (iv) high exposure to high GDP PD fluids, (v) repeated hemoperitoneum.

Advertisement

6. Risk factors for peritoneal membrane damage

  1. Diabetes: In diabetes there will be upregulation of vascular endothelial growth factor (VEGF), driven by local hypoxia induced by vasculopathy of the microvasculature and also due to increased GDPs. They also have lower lumen-to-vessel diameter ratios and higher postcapillary venule diameters.

  2. Uraemia: There will be increased expression of several proteoglycan components (versican, matrix metalloproteinase-2 [MMP-2] and hyaluronan) in patients with uraemia along with upregulation of AGE receptors (RAGE). It has been shown that presence of the C allele of RAGE protects against peritoneal fibrosis.

  3. Dietary salt intake: There will be upregulation of TGF-β1 and IL-6 expression in the peritoneal membrane, resulting in an enhanced EMT; in addition, to an increase in peritoneal small solute transport leading to UF failure.

Advertisement

7. Genetic factors

  1. IL-6 POLYMORPHISM: There can be G and C allele variant IL-6 polymorphism.IL-6 level is linked to peritoneal small solute transport and to albumin leakage. Those with GC or CC genotype had much higher IL-6 levels in their serum and in the drained dialysate than did patients with a GG genotype, along with upregulation of IL-6mRNA in the membrane [8].

  2. eNOS: eNOS genotype aa or ab (versus bb) was an independent predictor of reduced peritoneal membrane transport rate [9].

  3. Receptor for AGE: Numata et al. observed that a polymorphism of RAGE, the presence of the C allele in RAGE –429 T/C, were not present in patients with EPS [10]. Anti-RAGE antibodies also prevented the AGE associated upregulation of TGF-β1.

  4. Il-1β: polymorphism has shown increased infection rate (lower with T/T vs. C/T & T/T) [11].

  5. IL-1RN: polymorphism was an independent predictor of technique survival.

  6. CCL18: Increased expression of CCL18 was associated with functional deficiency, increased fibrosis and atherosclerosis.

The risk factors for the peritoneal membrane damage are summarized in Figure 6.

Figure 6.

Factors affecting peritoneal membrane degradation. Abbreviations: IL-6—Interleukin-6; VEGF—vascular endothelial growth factor; TGF—Transforming growth factor; and RAAS—renin angiotensin aldosterone system. Courtesy: Pletinck et al. [12].

Advertisement

8. Diagnosis of peritoneal fibrosis

  1. Effluent biomarkers: Nowadays, early detection of membrane damage can be done with biomarkers. Some of them are CA-125, IL-6 and PAI-1. A low effluent level of CA 125 has recently been found as a prognosis factor for the membrane damage [13]. Cases of plasminogen activator inhibitor and CCL18, in peritoneal effluent are also associated with the prognosis of the membrane [14]. Other biomarkers are VEGF, MMP-2, TGF-ß, CTGF, TNF-α.

  2. Histopathology: is the gold standard. The biopsy findings if using bioincompatible fluids were mesothelial layer disappearance, thickening of the sub-mesothelial compact zone, hyalinizing vasculopathy, angiogenesis, along with co-expression of α-smooth muscle actin and cytokeratin. The biopsy findings in those who used biocompatible fluids were associated with more well-preserved MC layer (56% vs. 26%), mild thickening with less dense sub-mesothelial compact zone (47% vs. 69%) and an absence of hyalinizing vasculopathy (4% vs. 30%) [15].

Advertisement

9. Conventional PD fluids

First generation Fluids: contain 35–40 mM lactate buffer with an acidic pH of 5.5. The low pH will be aggravating the detrimental effects of the high lactate on peritoneal mesothelium. During heat sterilization and storage, more of (GDPs) (e.g., formaldehyde, acetaldehyde, glyoxal, methylglyoxal, 5-hydroxymethylfurfural (5-HMF), 3-Deoxyglucosone (3-DG) and 3,4-dideoxyglucosone3-ene (3,4-DGE)) are formed, leading to membrane damage (shown in Figure 7).

Figure 7.

Potential beneficial effects of newer peritoneal dialysis solutions [16]. (Courtesy: Garcia nature reviews 2012). Abbreviations: RRF—residual renal function; LV—left ventricle; and AGE—advanced glycosylated end products).

Second generation Fluids: The buffers (such as lactate ± bicarbonate) are in separate chamber and are kept in very low pH to prevent formation of GDPs. Prior to use, they are mixed to a pH of 7–7.5 and are administered. They are detailed below.

Icodextrin: is an osmotic agent (Mol.wt-16,800 Da) derived from the starch, used in long night dwell with very low GDP, since they are absorbed into the circulation, with no sodium sieving. It has an UF capacity comparable to 4.25% dextrose fluid. Despite this, acidic PD fluid has been associated with increased local and systemic inflammation with increased permeability and IL-6. Even though this reaction is reaction, long term exposure may irreversibly change peritoneal morphology. The ISPD guidelines recommends use of icodextrin in high transporters for better volume control.

Amino acid solutions: A bag of 1.1% 21 amino acid PD fluid used in one exchange a day provide 22gm of amino acids (2/3rd essential & 1/3rd nonessential) which is 25% of daily requirement and generate an UF equal to 2 L 1.5% dextrose. It has an acidic pH of 6.6 with very low GDPs and low VEGF. For optimized nutrition of malnourished patients and to prevent increased serum nitrogen levels and metabolic acidosis, they should be applied at a ratio of 1:4 with glucose-containing PD fluids [17]. Nutrineal in one exchange with icodextrin (Extraneal) and Physioneal (Baxter) for other exchange as a regimen (NEPP regimen) has shown to preserve mesothelial integrity but with increased VEGF.

Advertisement

10. Newer PD fluids

  1. Trio gambrosol- tri compartment with two small chambers with 50% glucose and last chamber with calcium, magnesium, chloride & lactate.

  2. Physioneal- two chamber with chamber A containing glucose in 1.5%, 2.5%, 4.5% at a pH of 2.1 along with calcium & magnesium salts and chamber B with buffer lactate & bicarbonate at a pH 9.0. Volume of chambers in 3:1 ratio and solutions in both chamber mixed prior infusion, at least 1.6 L instilled during each infusion (to avoid accidental infusion of buffer only chamber and alkalosis).

  3. Balance- double chamber with glucose & electrolyte and other with buffer in equal volumes.

  4. Bicavera-double chamber which has used bicarbonate as buffer used along with glucose containing calcium& magnesium chloride. This is the only PD fluid which used bicarbonate alone as buffer.

  5. Delflex Neutral pH- Only FDA approved neutral pH PD fluid. GDP levels are 55, 70, 95 μmol/L depending on 1.5%, 2.5%, 4.5% glucose content (Table 1).

FluidpHBufferGDP (μmol/L)Osmotic agent
Icodextrin5.8Lactate457.5% polyglucose
Amino acid6.6Lactate1.1% amino acid
Trio Gambro6.3Lactate65Dextrose
Physioneal7.4Bicarbonate+lactate253Dextrose
Balance7.0Lactate42Dextrose
Bicavera7.4Bicarbonate42Dextrose
Delflex NpH7.0Lactate+bicarbonate70Dextrose

Table 1.

Newer PD fluids and constituents.

The Euro-Balance trial, demonstrated improved residual renal function together with decreased peritoneal UF with the pH neutral, low GDP fluid, as compared to the first generation, acidic high GDP solution [18]. The BalANZ trial has shown a lower risk of anuria and lower ultrafiltration and higher solute clearance rates with the use of low GDP fluid during the first 9 months of PD. Peritonitis incidence and severity were reduced in the BalANZ trial [19]. The TRIO trial comparing biocompatible solution (Gambrosol Trio) to standard PD fluid (Dianeal) showed contrasting results with slower rated of GFR decline but with higher peritonitis rate.

11. Novel PD fluid protoypes

The introduction of novel osmotic agents, is a promising way to improve the biocompatibility (Figure 8).

  1. The addition of 3.5% taurine-based PD fluid achieved equivalent ultrafiltration as glucose-based PD fluid and induced less mesothelial and fibroblast cell proliferation(rat model) [20].

  2. Hyperbranched polyglycerol containing PD fluid achieved similar solute and water transport and induced less peritoneal membrane damage (rat model)but data on metabolism are lacking [21].

  3. By the addition of dipeptide alanyl-glutamine to first- and second-generation PD fluid improved mesothelial cell stress response and cell survival with reduced peritoneal fibrosis [22]. A phase 3 trial is needed.

  4. Addition of L-carnitine to acidic, glucose PD fluids resulted in superior ultrafiltration and improved insulin sensitivity [23].

Figure 8.

Novel PD fluid prototypes.

Other preventive strategies:

Peritoneal resting: especially for high solute transport with Type 1 UF failure because this partially reverses some of the functional alterations of peritoneal transport. De souse et al. [24] found decreased D/P creatinine with increased UF capacity, after 4 weeks of peritoneal resting.

12. Newer agents to ameliorate membrane damage

  1. Inhibitors of the RAAS system: The mesenchymal cells can locally activate RAAS; in autocrine and paracrine fashion. The administration of RAAS inhibitors results in blockage of the TGF-β, fibronectin and VEGF. Koleysnk et al. [21]and Jing et al. [25], found that ACE inhibitors appeared to have a slower rate of decline in ultrafiltration and residual function, effectively protect against peritoneal fibrosis in long-term peritoneal dialysis.

  2. Hyaluronic acid: Preservation of hyaluronan concentration in PD effluent is deemed to be a marker of preservation of peritoneal integrity. It has protective role against abrasion and infection, through the initiation of increased synthesis of growth factors.

  3. I.P. Tinzaparin & Bemiparin: Del peso et al. [26] showed an improvement in UF capacity.

  4. Paricalcitol: VDR activator reduced IL-17 and increased Tregs leading to antifibrotic and anti-inflammatory effects.

  5. Rapamycin: mTOR inhibitors diminish IL-17 and decreases fibrosis with anti- MMT action but delayed healing, limiting its use in specific situations. It also decreases synthesis of VEGF.

  6. Tamoxifen: Estrogen receptor modulator, which inhibit MMT, reduces membrane thickness, invasion of the compact zone by mesenchymal mesothelial cells leading to reduced peritoneal MC migration and improved fibrinolytic capacity. A Dutch study showed a decreased mortality among patients with EPS after treatment with Tamoxifen.

  7. Nebivolol & Heparin (IP): increases fibrinolytic capacity associated with increased tPA levels. Apart from anticoagulation, heparin also has anti-inflammatory, immunomodulatory, antiangiogenic, antiproliferative, antifibrotic properties. Low-molecular-weight heparins can also inhibit VEGF and fibroblast growth factor activity.

  8. Benfotiamine: A derivative of thiamine, has been associated with decreased AGE and decreased oxidation by increasing transketolase [27].

  9. Pyridoxamine: beneficial role against UF failure; by reduction in accumulation of AGEs and the expression of angiogenic cytokines leading to decreased transport rates for small solutes and reduced blood vessel density [28].

  10. NSAIDs: In a rat model, oral administration of celecoxib drastically reduced prostaglandin E2, angiogenesis and lymph angiogenesis [29] and preserved ultrafiltration. Liu et al. observed that selective COX inhibition resulted in blunting of TGF-β production by mesothelial cells when exposed to high glucose concentrations and resulted in reduction of fibrosis and blunted ultrafiltration failure. No data on oral administration.

  11. Sulodexide: consists of 80% LMW heparin and 20% heparan sulphate. Apart from anticoagulation, it has immunomodulating, anti-inflammatory and antiproliferative, and anti-angiogenic properties. Oral sulodexide inhibits either VEGF directly by binding to it or by inhibiting its interaction with receptor.

  12. PPAR Ƴ agonist: Rosiglitazone, decreases AGE and fibrosis but the adverse effects have limited its use. The anti-inflammatory properties were mediated by an increase in peritoneal levels of IL-10 along with recruitment of CD4+ CD25+ FoxP3+ cellsD3+ lymphocytes.

  13. Tranilast: proposed to have some effects on peritoneal MCs, being the therapeutic potential for the treatment of peritoneal fibrosis [30].

The potential MMT modulators untested in PD are depicted in Table 2:

Antifibrotic agentsMechanism of action
1) TetrapeptideTGF ß inhibition
2) DipyridamoleTGF ß inhibition
3) PentoxyfyllineInhibition of ECM production
4) EmodinInhibition of ECM production
5) SimvastatinIncreases fibrinolytic activity
Antiangiogenic
1) Anecortave acetateInhibits VEGF production
2) PegaptanibInhibits VEGF-VEGFR binding
3) Anti-VEGFRIIBlocks receptor VEGFRII
4) TNP 470Decreases VEGF expression
Inhibition of EMT
1) Rho-ROCK inhibitor (Y27632)TGF-β/Smads inhibitors
2) Anti-oxidant agentNF-κB inhibition
3) Notch inhibitorsInhibit the induction of snail and repression of VE-cadherin
4) JNK inhibitors (PS600125)Inhibition of both ZEB and Rho pathway
5) CBR1 antagonistsTGF-β/Smads inhibitors

Table 2.

Potential MMT modulators untested IN PD.

Courtesy: Gonzalez 2016.

A study in a rat model, by using the stem cells demonstrated that the xenografts of human umbilical mesenchymal stem cells prevented the PD-induced membrane alterations [31].

By the advent of the proteomics and functional genomics analysis of the MC and the EMT, these fine biomarkers can be used for the accurate follow-up of the progressive peritoneal membrane deterioration. They also will identify the master molecules which governs the mesenchymal transition of MC. These molecular profiles of the EMT process in future might become an excellent tool to test the biocompatibility of newer PD fluids. Although further studies are needed, it is expected that increasing knowledge will provide a novel approach for therapeutic benefits in the treatment of peritoneal fibrosis, thus maintaining the peritoneal membrane for an extended period in PD patients.

Conflicts of interest

None.

Disclosures

None.

Source of funding

Nil.

Abbreviations

AGE

advanced glycation end products

EMT

epithelial to mesenchymal transition

EPS

encapsulating peritoneal sclerosis

GDP

glucose degradation product

IL-6

interleukin 6

LMW

low molecular weight

MC

mesothelial cells

MMT

mesothelial to mesenchymal transition

PD

peritoneal dialysis

RAGE

receptor for AGE

RANTES

regulated on activation, normal T cell expressed and secreted

RRF

residual renal function

STAT

signal transducer and activator of transcription

TGF

transforming growth factor

UF

ultrafiltration

VEGF

vascular endothelial growth factor

References

  1. 1. ESRD Patients in 2004: Global Overview of Patient Numbers, Treatment Modalities and Associated Trends. Available from: https://pubmed.ncbi.nlm.nih.gov/16204281/ [Accessed: January 14, 2021]
  2. 2. Mehrotra R, Devuyst O, Davies SJ, Johnson DW. The current state of peritoneal dialysis. Journal of American Society Nephrology. 2016;27(11):3238-3252
  3. 3. Kramer A, Pippias M, Noordzij M, Stel VS, Afentakis N, Ambühl PM, et al. The European Renal Association – European Dialysis and Transplant Association (ERA-EDTA) registry annual report 2015: A summary. Clinical Kidney Journal. 2018;11(1):108-122
  4. 4. Flessner MF. Peritoneal ultrafiltration: Physiology and failure. Peritoneal Dialysis – From Basic Concepts to Clinical Excellence. 2009;163:7-14
  5. 5. Gónzalez-Mateo G, Gallardo JM, AntonioSánchez-Tomero J, Majano P, Flores-Maldonado E, Paniagua R, et al. Pharmacological Preservation of Peritoneal Membrane in Peritoneal Dialysis. Some Special Problems in Peritoneal Dialysis. 2016. Available from: https://www.intechopen.com/books/some-special-problems-in-peritoneal-dialysis/pharmacological-preservation-of-peritoneal-membrane-in-peritoneal-dialysis [Accessed: January 14, 2021]
  6. 6. Aroeira LS, Aguilera A, Sánchez-Tomero JA, Bajo MA, del Peso G, Jiménez-Heffernan JA, et al. Epithelial to mesenchymal transition and peritoneal membrane failure in peritoneal dialysis patients: Pathologic significance and potential therapeutic interventions. Journal of the American Society of Nephrology. 2007;18(7):2004-2013
  7. 7. de Sousa-Amorim E, del Peso G, Bajo MA, Alvarez L, Ossorio M, Gil F, et al. Can EPS development be avoided with early interventions? The potential role of tamoxifen—A Single-Center Study. Peritoneal Dialysis International. 2014;34(6):582-593
  8. 8. Pecoits-Filho R, Carvalho MJ, Stenvinkel P, Lindholm B, Heimbürger O. Systemic and intraperitoneal interleukin-6 system during the first year of peritoneal dialysis. Peritoneal Dialysis International. 2006;26(1):53-63
  9. 9. Wong TY-H, Szeto C-C, Szeto CY-K, Lai K-B, Chow K-M, Li PK-T. Association of ENOS polymorphism with basal peritoneal membrane function in uremic patients. American Journal of Kidney Diseases. 2003;42(4):781-786
  10. 10. Numata M, Nakayama M, Hosoya T, Hoff CM, Holmes CJ, Schalling M, et al. Possible pathologic involvement of receptor for advanced glycation end products (RAGE) for development of encapsulating peritoneal sclerosis in Japanese CAPD patients. Clinical Nephrology. 2004;62(6):455-460
  11. 11. Shu K-H, Chuang Y-W, Huang S-T, Cheng C-H, Wu M-J, Chen C-H, et al. Association of interleukin-1β gene polymorphism and peritonitis in uremic patients undergoing peritoneal dialysis. Blood Purification. 2011;32(3):156-160
  12. 12. Pletinck A, Vanholder R, Veys N, Van Biesen W. Protecting the peritoneal membrane: Factors beyond peritoneal dialysis solutions. Nature Reviews Nephrology. 2012;8(9):542-550
  13. 13. Barreto DL, Hoekstra T, Halbesma N, Leegte M, Boeschoten EW, Dekker FW, et al. The association of effluent Ca125 with peritoneal dialysis technique failure. Peritoneal Dialysis International. 2015;35(7):683-690
  14. 14. Ahmad S, North BV, Qureshi A, Malik A, Bhangal G, Tarzi RM, et al. CCL18 in peritoneal dialysis patients and encapsulating peritoneal sclerosis. European Journal of Clinical Investigation. 2010;40(12):1067-1073
  15. 15. del Peso G, Jiménez-Heffernan JA, Selgas R, Remón C, Ossorio M, Fernández-Perpén A, et al. Biocompatible dialysis solutions preserve peritoneal mesothelial cell and Vessel Wall integrity. A case-control study on human biopsies. Peritoneal Dialysis International. 2016;36(2):129-134
  16. 16. García-López E, Lindholm B, Davies S. An update on peritoneal dialysis solutions. Nature Reviews Nephrology. 2012;8(4):224-233
  17. 17. Dombros NV, Prutis K, Tong M, Anderson GH, Harrison J, Sombolos K, et al. Six-month overnight intraperitoneal amino-acid infusion in continuous ambulatory peritoneal dialysis (CAPD) patients--no effect on nutritional status. Peritoneal Dialysis International. 1990;10(1):79-84
  18. 18. Williams JD, Topley N, Craig KJ, Mackenzie RK, Pischetsrieder M, Lage C, et al. The Euro-Balance Trial: The effect of a new biocompatible peritoneal dialysis fluid (balance) on the peritoneal membrane. Kidney International. 2004;66(1):408-418
  19. 19. Johnson DW, Brown FG, Clarke M, Boudville N, Elias TJ, Foo MWY, et al. Effects of biocompatible versus standard fluid on peritoneal dialysis outcomes. JASN. 2012;23(6):1097-1107
  20. 20. Nishimura H, Ikehara O, Naito T, Higuchi C, Sanaka T. Evaluation of taurine as an osmotic agent for Peritoneal Dialysis Solution. 2009. DOI: 10.1177/089686080902900216
  21. 21. Du C, Mendelson AA, Guan Q, Dairi G, Chafeeva I, da Roza G, et al. Hyperbranched polyglycerol is superior to glucose for long-term preservation of peritoneal membrane in a rat model of chronic peritoneal dialysis. Journal of Translational Medicine. 2016;14(1):338
  22. 22. Kratochwill K, Boehm M, Herzog R, Lichtenauer AM, Salzer E, Lechner M, et al. Alanyl–glutamine dipeptide restores the cytoprotective stress proteome of mesothelial cells exposed to peritoneal dialysis fluids. Nephrology Dialysis Transplantation. 2012;27(3):937-946
  23. 23. Bonomini M, Liberato LD, Rosso GD, Stingone A, Marinangeli G, Consoli A, 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. American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation. 2013;62(5):929-938
  24. 24. Sousa ED, Peso GD, Alvarez L, Ros S, Mateus A, Aguilar A, et al. Peritoneal resting with heparinized lavage reverses peritoneal type I membrane failure. A Comparative Study of the Resting Effects on Normal Membranes: Peritoneal Dialysis International. 2014. DOI: 10.3747/pdi.2013.00286
  25. 25. Effect of renin–angiotensin system inhibitors on prevention of peritoneal fibrosis in peritoneal dialysis patients - JING - 2010 - Nephrology - Wiley Online Library. DOI: 10.1111/j.1440-1797.2009.01162.x
  26. 26. Effect of Self-administered Intraperitoneal Bemiparin on Peritoneal Transport and Ultrafiltration Capacity in Peritoneal Dialysis Patients with Membrane Dysfunction. A Randomized, Multi-centre Open Clinical Trial | Nephrology Dialysis Transplantation | Oxford Academic. Available from: https://academic.oup.com/ndt/article/27/5/2051/1838368 [Accessed: January 28, 2021]
  27. 27. Kihm LP, Müller-Krebs S, Klein J, Ehrlich G, Mertes L, Gross M-L, et al. Benfotiamine protects against peritoneal and kidney damage in peritoneal dialysis. Journal of American Society Nephrology. 2011;22(5):914-926
  28. 28. Kakuta T, Tanaka R, Satoh Y, Izuhara Y, Inagi R, Nangaku M, et al. Pyridoxamine improves functional, structural, and biochemical alterations of peritoneal membranes in uremic peritoneal dialysis rats. Kidney International. 2005;68(3):1326-1336
  29. 29. Cyclooxygenase-2 Mediates Dialysate-Induced Alterations of the Peritoneal Membrane | American Society of Nephrology. Available from: https://jasn.asnjournals.org/content/20/3/582 [Accessed: January 27, 2021]
  30. 30. Fukuyama J, Ichikawa K, Hamano S, Shibata N. Tranilast suppresses the vascular intimal hyperplasia after balloon injury in rabbits fed on a high-cholesterol diet. European Journal of Pharmacology. 1996;318(2):327-332
  31. 31. The Therapeutic Potential of Human Umbilical Mesenchymal Stem Cells from Wharton’s Jelly in the Treatment of Rat Peritoneal Dialysis-Induced Fibrosis - Fan - 2016 - STEM CELLS Translational Medicine - Wiley Online Library. DOI: 10.5966/sctm.2015-0001

Written By

Mathew George Kunthara

Submitted: 22 March 2023 Reviewed: 12 April 2023 Published: 29 July 2023