1
|
Seghers F, Tintillier M, Morelle J. Recent advances in the understanding of the peritoneal membrane. Curr Opin Nephrol Hypertens 2024:00041552-990000000-00188. [PMID: 39291741 DOI: 10.1097/mnh.0000000000001028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
PURPOSE OF REVIEW The efficiency of peritoneal dialysis (PD) as a life-sustaining replacement therapy for patients with kidney failure relies on the integrity and function of the peritoneal membrane. Here, we review the most recent advances in the understanding of the peritoneal membrane and its role in PD. RECENT FINDINGS A recent update of the ISPD guidelines proposed a revised definition of membrane dysfunction, emphasizing the importance of fluid balance in patients treated with PD and identified three main mechanisms leading to insufficient peritoneal ultrafiltration (UF). The Bio-PD study, the first genomewide association study in PD, demonstrated that 20% of the interindividual variability in the peritoneal solute transfer rate is genetically determined, and identified several loci of potential relevance for peritoneal transport. A candidate-gene approach identified and characterized a common and functional variant in the promoter of the AQP1 gene associated with water transport and clinical outcomes in PD. Innovative strategies to preserve the integrity of the peritoneal membrane and to enhance UF are also discussed, including the use of gliflozins; steady glucose concentration PD; modulation of GLUT proteins; and cytoprotective additives. SUMMARY A comprehensive understanding of the peritoneal membrane and of the mechanisms driving UF may help individualizing PD prescription and improving outcomes in patients treated with PD.
Collapse
Affiliation(s)
- François Seghers
- Division of Nephrology, University Hospitals Namur (CHU UCL Namur), Namur
| | - Michel Tintillier
- Division of Nephrology, University Hospitals Namur (CHU UCL Namur), Namur
| | - Johann Morelle
- Division of Nephrology, University Hospitals Namur (CHU UCL Namur), Namur
- de Duve Institute, UCLouvain, Brussels, Belgium
| |
Collapse
|
2
|
Witowski J, Sikorska D, Catar R. Reporting Practices for Animal Studies on Peritoneal Dialysis Conducted in 2021-2023 after the Introduction of the ARRIVE 2.0 Guidelines. Nephron Clin Pract 2024:1-11. [PMID: 38934165 DOI: 10.1159/000539892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION The first version of Animal Research: Reporting of in vivo Experiments (ARRIVE 1.0) guidelines was introduced to improve reporting of animal research but did not lead to major improvements in this respect. This applied also to animal studies on peritoneal dialysis (PD). Here, we examined the performance of the revised version of these guidelines (ARRIVE 2.0). METHODS Eighty-nine relevant articles published in 2018-2020 (ARRIVE 1.0 period) and 97 published in 2021-2023 (ARRIVE 2.0 period) were identified in PubMed® and analyzed for completeness and transparency of reporting. RESULTS In both periods, most studies were carried out in Asia, on rodents, and concerned the peritoneal pathophysiology. During ARRIVE 2.0, more studies were published in higher impact factor journals with the focus on pharmacology and immunology. Compared to ARRIVE 1.0, general aspects of study design and reporting improved during ARRIVE 2.0 period in studies generated in Europe and USA but did not change significantly in Asia. Detailed analysis showed no global improvement in completeness of reporting key information included in the ARRIVE 2.0 Essential 10 checklist. Articles from both periods were deficient in sample size calculations, use of blinding, recording adverse events and drop-outs, and specification of appropriate statistical methods. The level of reporting during ARRIVE 2.0 did not correspond to the journal impact factor and the presence of recommendations for the use of ARRIVE 2.0 in their instructions to authors. CONCLUSION So far, ARRIVE 2.0 has not produced significant improvements in the reporting of animal studies in PD.
Collapse
Affiliation(s)
- Janusz Witowski
- Department Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Dorota Sikorska
- Department Rheumatology, Rehabilitation and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Rusan Catar
- Department Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
3
|
Jagirdar RM, Pitaraki E, Rouka E, Papazoglou ED, Bartosova M, Zebekakis P, Schmitt CP, Zarogiannis SG, Liakopoulos V. Differential effects of biocompatible peritoneal dialysis fluids on human mesothelial and endothelial cells in 2D and 3D phenotypes. Artif Organs 2024; 48:484-494. [PMID: 38151979 DOI: 10.1111/aor.14703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
INTRODUCTION Peritoneal dialysis (PD) is a life maintaining treatment in patients with end-stage renal disease. Its chronic application leads to peritoneal mesothelial layer denudation and fibrotic transformation along with vascular activation of inflammatory pathways. The impact of different PD fluids (PDF) on mesothelial and endothelial cell function and repair mechanisms are not comprehensively described. MATERIALS AND METHODS Mesothelial (MeT-5A) and endothelial cells (EA.hy926) were cultured in 1:1 ratio with cell medium and different PDF (icodextrin-based, amino acid-based, and glucose-based). Cell adhesion, cell migration, and cell proliferation in 2D and spheroid formation and collagen gel contraction assays in 3D cell cultures were performed. RESULTS Cell proliferation and cell-mediated gel contraction were both significantly decreased in all conditions. 3D spheroid formation was significantly reduced with icodextrin and amino acid PDF, but unchanged with glucose PDF. Adhesion was significantly increased by amino acid PDF in mesothelial cells and decreased by icodextrin and amino acid PDF in endothelial cells. Migration capacity was significantly decreased in mesothelial cells by all three PDF, while endothelial cells remained unaffected. CONCLUSIONS In 3D phenotypes the effects of PDF are more uniform in both mesothelial and endothelial cells, mitigating spheroid formation and gel contraction. On the contrary, effects on 2D phenotypes are more uniform in the icodextrin and amino acid PDF as opposed to glucose ones and affect mesothelial cells more variably. 2D and 3D comparative assessments of PDF effects on the main peritoneal membrane cell barriers, the mesothelial and endothelial, could provide useful translational information for PD studies.
Collapse
Affiliation(s)
- Rajesh M Jagirdar
- 2nd Department of Nephrology, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Eleanna Pitaraki
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Erasmia Rouka
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Eleftherios D Papazoglou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Pantelis Zebekakis
- First Department of Internal Medicine, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sotirios G Zarogiannis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Vassilios Liakopoulos
- 2nd Department of Nephrology, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
4
|
Morelle J, Lambie M, Öberg CM, Davies S. The Peritoneal Membrane and Its Role in Peritoneal Dialysis. Clin J Am Soc Nephrol 2024; 19:244-253. [PMID: 37616463 PMCID: PMC10861113 DOI: 10.2215/cjn.0000000000000282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023]
Abstract
A healthy and functional peritoneal membrane is key to achieving sufficient ultrafiltration and restoring fluid balance, a major component of high-quality prescription in patients treated with peritoneal dialysis (PD). Variability in membrane function at the start of PD or changes over time on treatment influence dialysis prescription and outcomes, and dysfunction of the peritoneal membrane contributes to fluid overload and associated complications. In this review, we summarize the current knowledge about the structure, function, and pathophysiology of the peritoneal membrane with a focus on clinical implications for patient-centered care. We also discuss the molecular and genetic mechanisms of solute and water transport across the peritoneal membrane, including the role of aquaporin water channels in crystalloid versus colloid osmosis; why and how to assess membrane function using peritoneal equilibration tests; the etiologies of membrane dysfunction and their specific management; and the effect of genetic variation on membrane function and outcomes in patients treated with PD. This review also identifies the gaps in current knowledge and perspectives for future research to improve our understanding of the peritoneal membrane and, ultimately, the care of patients treated with PD.
Collapse
Affiliation(s)
- Johann Morelle
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- UCLouvain School of Medicine, UCLouvain, Brussels, Belgium
| | - Mark Lambie
- Faculty of Medicine and Health Sciences, Keele University, Keele, United Kingdom
| | - Carl M. Öberg
- Division of Nephrology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Simon Davies
- Faculty of Medicine and Health Sciences, Keele University, Keele, United Kingdom
| |
Collapse
|
5
|
Dai H, Shan Y, Yu M, Wang F, Zhou Z, Sun J, Sheng L, Huang L, Sheng M. Network pharmacology, molecular docking and experimental verification of the mechanism of huangqi-jixuecao herb pair in treatment of peritoneal fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116874. [PMID: 37437794 DOI: 10.1016/j.jep.2023.116874] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/30/2023] [Accepted: 07/01/2023] [Indexed: 07/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Huangqi-Jixuecao herb pair (HQJXCHP) is a traditional herbal formula composed of two widely applied TCM prescriptions, Huangqi (Astragalus membranaceus (Fisch.) Bunge) and Jixuecao (Centella asiatica (L.) Urb.), used for hundreds of years to replenish qi and clear away heat. However, the therapeutic effects of HQJXCHP against peritoneal fibrosis (PF) and potential targets are currently unclear. AIMS OF THE STUDY The main objective of this study was preliminary prediction and validation of the effects and molecular mechanisms of action of HQJXCHP against PF based on network pharmacology analysis and experimental verification. MATERIALS AND METHODS The ingredients of HQJXCHP were analyzed via HPLC-Q-TOF/MS. Bioactive compounds of HQJXCHP used for network pharmacology analysis were obtained from the TCMSP database. HQJXCHP-related therapeutic targets in PF were obtained from the GeneCards, OMIM, Therapeutic Targets and PharmGkb databases. Therapeutic target-related signaling pathways were predicted via GO and KEGG pathway enrichment analyses. The targets of HQJXCHO were further validated in a PDS-induced PF mouse model in vivo and PMCs MMT model in vitro. RESULTS A total of 23 bioactive compounds of HQJXCHP related 188 target genes were retrieved. The HQJXCHP compound-target and PF-related target networks identified 131 common target genes. Subsequent protein-protein interaction (PPI) network analysis results disclosed Akt1, TP53, TNF, VEGFA and CASP3 as the top five key targets of HQJXCHP. Further molecular docking data revealed strong affinity of the two key compounds of HQJXCHP, quercetin and kaempferol, for these key targets. GO and KEGG pathway enrichment analyses further showed that PI3K/Akt, IL-17, TNF and TLR pathways contribute to the therapeutic effects of HQJXCHP on PF. An in vivo PDS-induced PF mouse model and in vitro PMCs mesothelial-to-mesenchymal transition (MMT) model with or without HQJXCHP intervention were used to confirm the effects and mechanisms of action of HQJXCHP. Western blot and qRT-PCR results showed that HQ, JXC and HQJXCHP reduced PDS-induced inflammatory cell aggregation and peritoneal thickening through suppressing the MMT process, among which HQJXCHP exerted the greatest therapeutic effect. Moreover, HQJXCHP inhibited activation of the PI3K/Akt, IL-17, TNF and TLR signaling pathways induced by PDS. CONCLUSIONS This is the first study to employ network pharmacology and molecular docking analyses to predict the targets of HQJXCHP with therapeutic effects on PDS-related PF. Data from in vivo and in vitro validation experiments collectively showed that HQJXCHP delays the PF process through inhibiting PI3K/Akt, IL-17, TNF and TLR signaling pathways. Overall, our findings highlight the successful application of network pharmacology theory to provide a scientific basis for clinical utility of HQJXCHP against PF.
Collapse
Affiliation(s)
- Huibo Dai
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Shan
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Manshu Yu
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Funing Wang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziren Zhou
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyi Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyan Huang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meixiao Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
6
|
Levai E, Marinovic I, Bartosova M, Zhang C, Schaefer B, Jenei H, Du Z, Drozdz D, Klaus G, Arbeiter K, Romero P, Schwenger V, Schwab C, Szabo AJ, Zarogiannis SG, Schmitt CP. Human peritoneal tight junction, transporter and channel expression in health and kidney failure, and associated solute transport. Sci Rep 2023; 13:17429. [PMID: 37833387 PMCID: PMC10575882 DOI: 10.1038/s41598-023-44466-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023] Open
Abstract
Next to the skin, the peritoneum is the largest human organ, essentially involved in abdominal health and disease states, but information on peritoneal paracellular tight junctions and transcellular channels and transporters relative to peritoneal transmembrane transport is scant. We studied their peritoneal localization and quantity by immunohistochemistry and confocal microscopy in health, in chronic kidney disease (CKD) and on peritoneal dialysis (PD), with the latter allowing for functional characterizations, in a total of 93 individuals (0-75 years). Claudin-1 to -5, and -15, zonula occludens-1, occludin and tricellulin, SGLT1, PiT1/SLC20A1 and ENaC were consistently detected in mesothelial and arteriolar endothelial cells, with age dependent differences for mesothelial claudin-1 and arteriolar claudin-2/3. In CKD mesothelial claudin-1 and arteriolar claudin-2 and -3 were more abundant. Peritonea from PD patients exhibited increased mesothelial and arteriolar claudin-1 and mesothelial claudin-2 abundance and reduced mesothelial and arteriolar claudin-3 and arteriolar ENaC. Transperitoneal creatinine and glucose transport correlated with pore forming arteriolar claudin-2 and mesothelial claudin-4/-15, and creatinine transport with mesothelial sodium/phosphate cotransporter PiT1/SLC20A1. In multivariable analysis, claudin-2 independently predicted the peritoneal transport rates. In conclusion, tight junction, transcellular transporter and channel proteins are consistently expressed in peritoneal mesothelial and endothelial cells with minor variations across age groups, specific modifications by CKD and PD and distinct associations with transperitoneal creatinine and glucose transport rates. The latter deserve experimental studies to demonstrate mechanistic links.Clinical Trial registration: The study was performed according to the Declaration of Helsinki and is registered at www.clinicaltrials.gov (NCT01893710).
Collapse
Affiliation(s)
- Eszter Levai
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
- HUNREN SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Iva Marinovic
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Conghui Zhang
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Betti Schaefer
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Hanna Jenei
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Zhiwei Du
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Dorota Drozdz
- Jagiellonian University Medical College, Krakow, Poland
| | | | - Klaus Arbeiter
- Department of Pediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | - Philipp Romero
- Division of Pediatric Surgery, Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Vedat Schwenger
- Department of Nephrology, Klinikum der Landeshauptstadt Stuttgart, Stuttgart, Germany
| | | | - Attila J Szabo
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
- HUNREN SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Sotirios G Zarogiannis
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Department of Physiology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Claus Peter Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
| |
Collapse
|
7
|
Zhang C, Bartosova M, Marinovic I, Schwab C, Schaefer B, Vondrak K, Ariceta G, Zaloszyc A, Ranchin B, Taylan C, Büscher R, Oh J, Mehrabi A, Schmitt CP. Peritoneal transformation shortly after kidney transplantation in pediatric patients with preceding chronic peritoneal dialysis. Nephrol Dial Transplant 2023; 38:2170-2181. [PMID: 36754369 DOI: 10.1093/ndt/gfad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND The unphysiological composition of peritoneal dialysis (PD) fluids induces progressive peritoneal fibrosis, hypervascularization and vasculopathy. Information on these alterations after kidney transplantation (KTx) is scant. METHODS Parietal peritoneal tissues were obtained from 81 pediatric patients with chronic kidney disease stage 5 (CKD5), 72 children on PD with low glucose degradation product (GDP) PD fluids, and from 20 children 4-8 weeks after KTx and preceding low-GDP PD. Tissues were analyzed by digital histomorphometry and quantitative immunohistochemistry. RESULTS While chronic PD was associated with peritoneal hypervascularization, after KTx vascularization was comparable to CKD5 level. Submesothelial CD45 counts were 40% lower compared with PD, and in multivariable analyses independently associated with microvessel density. In contrast, peritoneal mesothelial denudation, submesothelial thickness and fibrin abundance, number of activated, submesothelial fibroblasts and of mesothelial-mesenchymal transitioned cells were similar after KTx. Diffuse peritoneal podoplanin positivity was present in 40% of the transplanted patients. In subgroups matched for age, PD vintage, dialytic glucose exposure and peritonitis incidence, submesothelial hypoxia-inducible factor 1-alpha abundance and angiopoietin 1/2 ratio were lower after KTx, reflecting vessel maturation, while arteriolar and microvessel p16 and cleaved Casp3 were higher. Submesothelial mast cell count and interleukin-6 were lower, whereas transforming growth factor-beta induced pSMAD2/3 was similar as compared with children on PD. CONCLUSIONS Peritoneal membrane damage induced with chronic administration of low-GDP PD fluids was less severe after KTx. While peritoneal microvessel density, primarily defining PD transport and ultrafiltration capacity, was normal after KTx and peritoneal inflammation less pronounced, diffuse podoplanin positivity and profibrotic activity were prevalent.
Collapse
Affiliation(s)
- Conghui Zhang
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Iva Marinovic
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Constantin Schwab
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Betti Schaefer
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Karel Vondrak
- Department of Pediatrics, University Hospital Motol, Prague, Czech Republic
| | - Gema Ariceta
- Hospital Universitario Materno-Infantil Vall d'Hebron, Barcelona, Spain
| | - Ariane Zaloszyc
- Department of Pediatrics 1, University Hospital of Strasbourg, Strasbourg, France
| | - Bruno Ranchin
- Service de Néphrologie Pédiatrique, Hôpital Femme Mere Enfant, Lyon, France
| | - Christina Taylan
- Pediatric Nephrology, Children's and Adolescent's Hospital, University Hospital of Cologne, Cologne, Germany
| | - Rainer Büscher
- Pediatric Nephrology, University Children's Hospital, Essen, Germany
| | - Jun Oh
- Department of Pediatric Nephrology, University Children's Medical Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Arianeb Mehrabi
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
8
|
MUW researcher of the month. Wien Klin Wochenschr 2022; 134:892-893. [PMID: 36512129 DOI: 10.1007/s00508-022-02143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
9
|
Proteome-Wide Differential Effects of Peritoneal Dialysis Fluid Properties in an In Vitro Human Endothelial Cell Model. Int J Mol Sci 2022; 23:ijms23148010. [PMID: 35887356 PMCID: PMC9317527 DOI: 10.3390/ijms23148010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/27/2023] Open
Abstract
To replace kidney function, peritoneal dialysis (PD) utilizes hyperosmotic PD fluids with specific physico-chemical properties. Their composition induces progressive damage of the peritoneum, leading to vasculopathies, decline of membrane function, and PD technique failure. Clinically used PD fluids differ in their composition but still remain bioincompatible. We mapped the molecular pathomechanisms in human endothelial cells induced by the different characteristics of widely used PD fluids by proteomics. Of 7894 identified proteins, 3871 were regulated at least by 1 and 49 by all tested PD fluids. The latter subset was enriched for cell junction-associated proteins. The different PD fluids individually perturbed proteins commonly related to cell stress, survival, and immune function pathways. Modeling two major bioincompatibility factors of PD fluids, acidosis, and glucose degradation products (GDPs) revealed distinct effects on endothelial cell function and regulation of cellular stress responses. Proteins and pathways most strongly affected were members of the oxidative stress response. Addition of the antioxidant and cytoprotective additive, alanyl-glutamine (AlaGln), to PD fluids led to upregulation of thioredoxin reductase-1, an antioxidant protein, potentially explaining the cytoprotective effect of AlaGln. In conclusion, we mapped out the molecular response of endothelial cells to PD fluids, and provided new evidence for their specific pathomechanisms, crucial for improvement of PD therapies.
Collapse
|
10
|
Zarogiannis SG, Schmitt CP. Molecular Mechanisms of Peritoneal Membrane Pathophysiology. Biomolecules 2022; 12:biom12060757. [PMID: 35740882 PMCID: PMC9220859 DOI: 10.3390/biom12060757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 02/01/2023] Open
Abstract
The peritoneal membrane is the largest internal membrane of the human body, having a surface area that approximates the surface area of the skin [...]
Collapse
Affiliation(s)
- Sotirios G. Zarogiannis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece
- Correspondence: ; Tel.: +30 2410 685558
| | - Claus Peter Schmitt
- Pediatric Nephology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, 69210 Heidelberg, Germany;
| |
Collapse
|
11
|
Bartosova M, Zarogiannis SG, Schmitt CP. How peritoneal dialysis transforms the peritoneum and vasculature in children with chronic kidney disease-what can we learn for future treatment? Mol Cell Pediatr 2022; 9:9. [PMID: 35513740 PMCID: PMC9072612 DOI: 10.1186/s40348-022-00141-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/09/2022] [Indexed: 01/04/2023] Open
Abstract
Children with chronic kidney disease (CKD) suffer from inflammation and reactive metabolite-induced stress, which massively accelerates tissue and vascular aging. Peritoneal dialysis (PD) is the preferred dialysis mode in children, but currently used PD fluids contain far supraphysiological glucose concentrations for fluid and toxin removal and glucose degradation products (GDP). While the peritoneal membrane of children with CKD G5 exhibits only minor alterations, PD fluids trigger numerous molecular cascades resulting in major peritoneal membrane inflammation, hypervascularization, and fibrosis, with distinct molecular and morphological patterns depending on the GDP content of the PD fluid used. PD further aggravates systemic vascular disease. The systemic vascular aging process is particularly pronounced when PD fluids with high GDP concentrations are used. GDP induce endothelial junction disintegration, apoptosis, fibrosis, and intima thickening. This review gives an overview on the molecular mechanisms of peritoneal and vascular transformation and strategies to improve peritoneal and vascular health in patients on PD.
Collapse
Affiliation(s)
- Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Sotirios G Zarogiannis
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany.,Department of Physiology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
12
|
Krediet RT. Aging of the Peritoneal Dialysis Membrane. Front Physiol 2022; 13:885802. [PMID: 35574465 PMCID: PMC9096116 DOI: 10.3389/fphys.2022.885802] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Long-term peritoneal dialysis as currently performed, causes structural and functional alterations of the peritoneal dialysis membrane. This decay is brought about by the continuous exposure to commercially available glucose-based dialysis solutions. This review summarizes our knowledge on the peritoneum in the initial phase of PD, during the first 2 years and the alterations in function and morphology in long-term PD patients. The pseudohypoxia hypothesis is discussed and how this glucose-induced condition can be used to explain all peritoneal alterations in long-term PD patients. Special attention is paid to the upregulation of hypoxia inducing factor-1 and the subsequent stimulation of the genes coding for glucose transporter-1 (GLUT-1) and the growth factors transforming growth factor-β (TGFβ), vascular endothelial growth factor (VEGF), plasminogen growth factor activator inhibitor-1 (PAI-1) and connective tissue growth factor (CTGF). It is argued that increased pseudohypoxia-induced expression of GLUT-1 in interstitial fibroblasts is the key factor in a vicious circle that augments ultrafiltration failure. The practical use of the protein transcripts of the upregulated growth factors in peritoneal dialysis effluent is considered. The available and developing options for prevention and treatment are examined. It is concluded that low glucose degradation products/neutral pH, bicarbonate buffered solutions with a combination of various osmotic agents all in low concentration, are currently the best achievable options, while other accompanying measures like the use of RAAS inhibitors and tamoxifen may be valuable. Emerging developments include the addition of alanyl glutamine to the dialysis solution and perhaps the use of nicotinamide mononucleotide, available as nutritional supplement.
Collapse
|
13
|
Krediet RT, Parikova A. Relative Contributions of Pseudohypoxia and Inflammation to Peritoneal Alterations with Long-Term Peritoneal Dialysis Patients. Clin J Am Soc Nephrol 2022; 17:1259-1266. [PMID: 35168992 PMCID: PMC9435980 DOI: 10.2215/cjn.15371121] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Long-term peritoneal dialysis (PD) is associated with alterations in peritoneal function, like the development of high small solute transfer rates and impaired ultrafiltration. Also morphologic changes can develop, the most prominent being loss of mesothelium, vasculopathy and interstitial fibrosis. Current research suggests peritoneal inflammation as the driving force for these alterations. In this review the available evidence for inflammation is examined and a new hypothesis is put forward consisting of high glucose-induced pseudohypoxia. Hypoxia of cells is characterized by a high NADH/NAD+ ratio in their cytosol. Pseudohypoxia is similar, but occurs when excessive amounts of glucose are metabolized, as is the case for peritoneal interstitial cells in PD. The glucose- induced high NADH/NAD+ ratio upregulates the hypoxia-inducible factor-1 gene, which stimulates not only the glucose transporter-1 gene, but also many profibrotic genes like TGFβ, VEGF, PAI-1 and CTGF, all known to be involved in the development of peritoneal fibrosis. This review discusses the causes and consequences of pseudohypoxia in PD and the available options for treatment and prevention. Reducing peritoneal exposure to the excessively high dialysate glucose load is the cornerstone to avoid the pseudohypoxia-induced alterations. This can partly be done by the use of icodextrin, or by combinations of low molecular weight osmotic agents, all in a low dose. The addition of alanyl-glutamine to the dialysis solution needs further clinical investigation.
Collapse
Affiliation(s)
- Raymond T. Krediet
- Division of Nephrology, Department of Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Alena Parikova
- Department of Nephrology, Transplant Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
14
|
Impact of Metabolomics Technologies on the Assessment of Peritoneal Membrane Profiles in Peritoneal Dialysis Patients: A Systematic Review. Metabolites 2022; 12:metabo12020145. [PMID: 35208219 PMCID: PMC8879920 DOI: 10.3390/metabo12020145] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 02/07/2023] Open
Abstract
Peritoneal dialysis (PD) is an effective and frequent dialysis modality in adults, particularly preferred in infants and young children with end-stage renal disease (ESRD). Long-term exposure of the peritoneal membrane to dialysis solutions results in severe morphologic and functional alterations. Peritoneal dialysis effluent biomarkers are based on omics technologies, which could predict the onset or confirm the diagnosis of peritoneal membrane dysfunction, would allow the development of accurate early prognostic tools and, potentially, the identification of future therapeutic targets. The purpose of our study was to critically review the literature on the impact and the effectiveness of metabolomics technologies in peritoneal health. The main search was performed in electronic databases (PubMed/MEDLINE, Embase and Cochrane Central Register of Controlled Trials) from inception to December 2020, using various combinations of Medical Subject Headings (MeSH). The main search highlighted nine studies, of which seven were evaluated in detail. Metabolomics technologies may provide significant input in the recognition of peritoneal membrane dysfunction in PD patients and provide evidence of early intervention strategies that could protect peritoneum health and function.
Collapse
|
15
|
Hausinger R, Schmaderer C, Heemann U, Bachmann Q. Innovationen in der Peritonealdialyse. DER NEPHROLOGE 2022; 17:85-91. [PMID: 34786026 PMCID: PMC8588934 DOI: 10.1007/s11560-021-00542-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 12/03/2022]
Abstract
Die Peritonealdialyse (PD) war früher eine geläufige Behandlung beim akuten dialysepflichtigen Nierenversagen. Zugunsten kontinuierlicher, extrakorporaler Nierenersatzverfahren verschwand sie von der Bildfläche der westlichen Welt, wohingegen sie in strukturarmen Ländern aufgrund ihrer Simplizität und geringen Ressourcenintensität weiter eingesetzt wird. Die Engpässe in der medizinischen Versorgung im Rahmen der COVID-19(„coronavirus disease 2019“)-Pandemie führten kürzlich zu erneuter weltweiter Beachtung der PD als sichere Option beim akuten dialysepflichtigen Nierenversagen. Von der Einführung biokompatibler Lösungen vor 20 Jahren war eine Reduktion von Mortalität oder technischem Versagen erwartet worden. Leider konnten Studien dieses bisher allenfalls andeuten, nicht aber beweisen. Eine innovative Option stellen immunmodulatorische Adjuvanzien dar, die die lokale Immunkompetenz verbessern und den Verlust der Funktion des Peritoneums verhindern sollen. Derzeit rückt die Vision einer tragbaren künstlichen Niere immer näher. Auch eine Intensivierung der Dialysedosis erscheint mit minimaler Dialysatmenge erreichbar. In Zeiten der globalen Erderwärmung könnten durch die Regeneration von Dialysat nicht nur relevante Mengen an Wasser eingespart, sondern auch die CO2-Bilanz günstig beeinflusst werden. Zusammenfassend erlebt die PD derzeit einen zweiten Frühling. Dieser Artikel beschreibt die derzeitigen und zukünftigen Entwicklungen dieses Verfahrens.
Collapse
|
16
|
Herzog R, Sacnun JM, González-Mateo G, Bartosova M, Bialas K, Wagner A, Unterwurzacher M, Sobieszek IJ, Daniel-Fischer L, Rusai K, Pascual-Antón L, Kaczirek K, Vychytil A, Schmitt CP, López-Cabrera M, Alper SL, Aufricht C, Kratochwill K. Lithium preserves peritoneal membrane integrity by suppressing mesothelial cell αB-crystallin. Sci Transl Med 2021; 13:13/608/eaaz9705. [PMID: 34433641 DOI: 10.1126/scitranslmed.aaz9705] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/30/2021] [Accepted: 08/04/2021] [Indexed: 01/18/2023]
Abstract
Life-saving renal replacement therapy by peritoneal dialysis (PD) is limited in use and duration by progressive impairment of peritoneal membrane integrity and homeostasis. Preservation of peritoneal membrane integrity during chronic PD remains an urgent but long unmet medical need. PD therapy failure results from peritoneal fibrosis and angiogenesis caused by hypertonic PD fluid (PDF)-induced mesothelial cytotoxicity. However, the pathophysiological mechanisms involved are incompletely understood, limiting identification of therapeutic targets. We report that addition of lithium chloride (LiCl) to PDF is a translatable intervention to counteract PDF-induced mesothelial cell death, peritoneal membrane fibrosis, and angiogenesis. LiCl improved mesothelial cell survival in a dose-dependent manner. Combined transcriptomic and proteomic characterization of icodextrin-based PDF-induced mesothelial cell injury identified αB-crystallin as the mesothelial cell protein most consistently counter-regulated by LiCl. In vitro and in vivo overexpression of αB-crystallin triggered a fibrotic phenotype and PDF-like up-regulation of vascular endothelial growth factor (VEGF), CD31-positive cells, and TGF-β-independent activation of TGF-β-regulated targets. In contrast, αB-crystallin knockdown decreased VEGF expression and early mesothelial-to-mesenchymal transition. LiCl reduced VEGF release and counteracted fibrosis- and angiogenesis-associated processes. αB-crystallin in patient-derived mesothelial cells was specifically up-regulated in response to PDF and increased in peritoneal mesothelial cells from biopsies from pediatric patients undergoing PD, correlating with markers of angiogenesis and fibrosis. LiCl-supplemented PDF promoted morphological preservation of mesothelial cells and the submesothelial zone in a mouse model of chronic PD. Thus, repurposing LiCl as a cytoprotective PDF additive may offer a translatable therapeutic strategy to combat peritoneal membrane deterioration during PD therapy.
Collapse
Affiliation(s)
- Rebecca Herzog
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Juan Manuel Sacnun
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria.,Zytoprotec GmbH, 1090 Vienna, Austria
| | - Guadalupe González-Mateo
- Tissue and Organ Homeostasis, Molecular Biology Centre Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany
| | - Katarzyna Bialas
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Zytoprotec GmbH, 1090 Vienna, Austria
| | - Anja Wagner
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Markus Unterwurzacher
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Isabel J Sobieszek
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Lisa Daniel-Fischer
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Krisztina Rusai
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Lucía Pascual-Antón
- Tissue and Organ Homeostasis, Molecular Biology Centre Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Klaus Kaczirek
- Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Andreas Vychytil
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria
| | - Claus Peter Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany
| | - Manuel López-Cabrera
- Tissue and Organ Homeostasis, Molecular Biology Centre Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Seth L Alper
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Christoph Aufricht
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria. .,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
17
|
How to Improve the Biocompatibility of Peritoneal Dialysis Solutions (without Jeopardizing the Patient's Health). Int J Mol Sci 2021; 22:ijms22157955. [PMID: 34360717 PMCID: PMC8347640 DOI: 10.3390/ijms22157955] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/23/2022] Open
Abstract
Peritoneal dialysis (PD) is an important, if underprescribed, modality for the treatment of patients with end-stage kidney disease. Among the barriers to its wider use are the deleterious effects of currently commercially available glucose-based PD solutions on the morphological integrity and function of the peritoneal membrane due to fibrosis. This is primarily driven by hyperglycaemia due to its effects, through multiple cytokine and transcription factor signalling-and their metabolic sequelae-on the synthesis of collagen and other extracellular membrane components. In this review, we outline these interactions and explore how novel PD solution formulations are aimed at utilizing this knowledge to minimise the complications associated with fibrosis, while maintaining adequate rates of ultrafiltration across the peritoneal membrane and preservation of patient urinary volumes. We discuss the development of a new generation of reduced-glucose PD solutions that employ a variety of osmotically active constituents and highlight the biochemical rationale underlying optimization of oxidative metabolism within the peritoneal membrane. They are aimed at achieving optimal clinical outcomes and improving the whole-body metabolic profile of patients, particularly those who are glucose-intolerant, insulin-resistant, or diabetic, and for whom daily exposure to high doses of glucose is contraindicated.
Collapse
|
18
|
Bartosova M, Zhang C, Schaefer B, Herzog R, Ridinger D, Damgov I, Levai E, Marinovic I, Eckert C, Romero P, Sallay P, Ujszaszi A, Unterwurzacher M, Wagner A, Hildenbrand G, Warady BA, Schaefer F, Zarogiannis SG, Kratochwill K, Schmitt CP. Glucose Derivative Induced Vasculopathy in Children on Chronic Peritoneal Dialysis. Circ Res 2021; 129:e102-e118. [PMID: 34233458 DOI: 10.1161/circresaha.121.319310] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Maria Bartosova
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Conghui Zhang
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Betti Schaefer
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Rebecca Herzog
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria (R.H., M.U., A.W., K.K.)
| | - David Ridinger
- Kirchhoff Institute for Physics (D.R., G.H.), University of Heidelberg, Heidelberg, Germany
| | - Ivan Damgov
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Eszter Levai
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
- ELKH-SE, Pediatrics and Nephrology Research Group, Budapest, Hungary (E.L.)
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary (E.L., P.S.)
| | - Iva Marinovic
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Christoph Eckert
- Institute of Pathology (C.E.), University of Heidelberg, Heidelberg, Germany
| | - Philipp Romero
- Division of Pediatric Surgery, Department of General, Visceral and Transplantation Surgery (P.R.), University of Heidelberg, Heidelberg, Germany
| | - Peter Sallay
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary (E.L., P.S.)
| | - Akos Ujszaszi
- Division of Nephrology, Heidelberg University Hospital, Heidelberg, Germany (A.U.)
| | - Markus Unterwurzacher
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria (R.H., M.U., A.W., K.K.)
| | - Anja Wagner
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria (R.H., M.U., A.W., K.K.)
| | - Georg Hildenbrand
- Kirchhoff Institute for Physics (D.R., G.H.), University of Heidelberg, Heidelberg, Germany
| | | | - Franz Schaefer
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Sotirios G Zarogiannis
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
- Department of Physiology, Faculty of Medicine, University of Thessaly, Larissa, Greece (S.G.Z.)
| | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria (R.H., M.U., A.W., K.K.)
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
19
|
Masola V, Bonomini M, Onisto M, Ferraro PM, 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. [PMID: 34209455 PMCID: PMC8308380 DOI: 10.3390/nu13072282] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Glucose-based solutions remain the most used osmotic agents in peritoneal dialysis (PD), but unavoidably they contribute to the loss of peritoneal filtration capacity. Here, we evaluated at a molecular level the effects of XyloCore, a new PD solution with a low glucose content, in mesothelial and endothelial cells. Cell viability, integrity of mesothelial and endothelial cell membrane, activation of mesothelial and endothelial to mesenchymal transition programs, inflammation, and angiogenesis were evaluated by several techniques. Results showed that XyloCore preserves mesothelial and endothelial cell viability and membrane integrity. Moreover XyloCore, unlike glucose-based solutions, does not exert pro-fibrotic, -inflammatory, and -angiogenic effects. Overall, the in vitro evidence suggests that XyloCore could represent a potential biocompatible solution promising better outcomes in clinical practice.
Collapse
Affiliation(s)
- Valentina Masola
- Division of Nephrology and Dialysis, Department of Medicine, Piazzale A. Stefani 1, 37126 Verona, Italy;
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy;
| | - Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS.Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy;
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy;
| | - Pietro Manuel Ferraro
- U.O.S. Terapia Conservativa della Malattia Renale Cronica, U.O.C. Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00178 Rome, Italy;
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00178 Rome, Italy
| | - Arduino Arduini
- R&D Department, Iperboreal Pharma Srl, 65122 Pescara, Italy;
| | - Giovanni Gambaro
- Division of Nephrology and Dialysis, Department of Medicine, Piazzale A. Stefani 1, 37126 Verona, Italy;
| |
Collapse
|
20
|
Terri M, Trionfetti F, Montaldo C, Cordani M, Tripodi M, Lopez-Cabrera M, Strippoli R. Mechanisms of Peritoneal Fibrosis: Focus on Immune Cells-Peritoneal Stroma Interactions. Front Immunol 2021; 12:607204. [PMID: 33854496 PMCID: PMC8039516 DOI: 10.3389/fimmu.2021.607204] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Peritoneal fibrosis is characterized by abnormal production of extracellular matrix proteins leading to progressive thickening of the submesothelial compact zone of the peritoneal membrane. This process may be caused by a number of insults including pathological conditions linked to clinical practice, such as peritoneal dialysis, abdominal surgery, hemoperitoneum, and infectious peritonitis. All these events may cause acute/chronic inflammation and injury to the peritoneal membrane, which undergoes progressive fibrosis, angiogenesis, and vasculopathy. Among the cellular processes implicated in these peritoneal alterations is the generation of myofibroblasts from mesothelial cells and other cellular sources that are central in the induction of fibrosis and in the subsequent functional deterioration of the peritoneal membrane. Myofibroblast generation and activity is actually integrated in a complex network of extracellular signals generated by the various cellular types, including leukocytes, stably residing or recirculating along the peritoneal membrane. Here, the main extracellular factors and the cellular players are described with emphasis on the cross-talk between immune system and cells of the peritoneal stroma. The understanding of cellular and molecular mechanisms underlying fibrosis of the peritoneal membrane has both a basic and a translational relevance, since it may be useful for setup of therapies aimed at counteracting the deterioration as well as restoring the homeostasis of the peritoneal membrane.
Collapse
Affiliation(s)
- Michela Terri
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Claudia Montaldo
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Marco Cordani
- instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA) Nanociencia, Madrid, Spain
| | - Marco Tripodi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Manuel Lopez-Cabrera
- Programa de Homeostasis de Tejidos y Organos, Centro de Biología Molecular “Severo Ochoa”-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
21
|
Herzog R, Bartosova M, Tarantino S, Wagner A, Unterwurzacher M, Sacnun JM, Lichtenauer AM, Kuster L, Schaefer B, Alper SL, Aufricht C, Schmitt CP, Kratochwill K. Peritoneal Dialysis Fluid Supplementation with Alanyl-Glutamine Attenuates Conventional Dialysis Fluid-Mediated Endothelial Cell Injury by Restoring Perturbed Cytoprotective Responses. Biomolecules 2020; 10:biom10121678. [PMID: 33334074 PMCID: PMC7765520 DOI: 10.3390/biom10121678] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 12/18/2022] Open
Abstract
Long-term clinical outcome of peritoneal dialysis (PD) depends on adequate removal of small solutes and water. The peritoneal endothelium represents the key barrier and peritoneal transport dysfunction is associated with vascular changes. Alanyl-glutamine (AlaGln) has been shown to counteract PD-induced deteriorations but the effect on vascular changes has not yet been elucidated. Using multiplexed proteomic and bioinformatic analyses we investigated the molecular mechanisms of vascular pathology in-vitro (primary human umbilical vein endothelial cells, HUVEC) and ex-vivo (arterioles of patients undergoing PD) following exposure to PD-fluid. An overlap of 1813 proteins (40%) of over 3100 proteins was identified in both sample types. PD-fluid treatment significantly altered 378 in endothelial cells and 192 in arterioles. The HUVEC proteome resembles the arteriolar proteome with expected sample specific differences of mainly immune system processes only present in arterioles and extracellular region proteins primarily found in HUVEC. AlaGln-addition to PD-fluid revealed 359 differentially abundant proteins and restored the molecular process landscape altered by PD fluid. This study provides evidence on validity and inherent limitations of studying endothelial pathomechanisms in-vitro compared to vascular ex-vivo findings. AlaGln could reduce PD-associated vasculopathy by reducing endothelial cellular damage, restoring perturbed abundances of pathologically important proteins and enriching protective processes.
Collapse
Affiliation(s)
- Rebecca Herzog
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (M.U.)
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (B.S.); (C.P.S.)
| | - Silvia Tarantino
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Zytoprotec GmbH, 1090 Vienna, Austria
| | - Anja Wagner
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (M.U.)
| | - Markus Unterwurzacher
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (M.U.)
| | - Juan Manuel Sacnun
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Zytoprotec GmbH, 1090 Vienna, Austria
| | - Anton M. Lichtenauer
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
| | - Lilian Kuster
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
| | - Betti Schaefer
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (B.S.); (C.P.S.)
| | - Seth L. Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Christoph Aufricht
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (B.S.); (C.P.S.)
| | - Klaus Kratochwill
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (M.U.)
- Correspondence: ; Tel.: +43-140400-80
| |
Collapse
|
22
|
Alanyl-Glutamine Restores Tight Junction Organization after Disruption by a Conventional Peritoneal Dialysis Fluid. Biomolecules 2020; 10:biom10081178. [PMID: 32823646 PMCID: PMC7464725 DOI: 10.3390/biom10081178] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
Understanding and targeting the molecular basis of peritoneal solute and protein transport is essential to improve peritoneal dialysis (PD) efficacy and patient outcome. Supplementation of PD fluids (PDF) with alanyl-glutamine (AlaGln) increased small solute transport and reduced peritoneal protein loss in a recent clinical trial. Transepithelial resistance and 10 kDa and 70 kDa dextran transport were measured in primary human endothelial cells (HUVEC) exposed to conventional acidic, glucose degradation products (GDP) containing PDF (CPDF) and to low GDP containing PDF (LPDF) with and without AlaGln. Zonula occludens-1 (ZO-1) and claudin-5 were quantified by Western blot and immunofluorescence and in mice exposed to saline and CPDF for 7 weeks by digital imaging analyses. Spatial clustering of ZO-1 molecules was assessed by single molecule localization microscopy. AlaGln increased transepithelial resistance, and in CPDF exposed HUVEC decreased dextran transport rates and preserved claudin-5 and ZO-1 abundance. Endothelial clustering of membrane bound ZO-1 was higher in CPDF supplemented with AlaGln. In mice, arteriolar endothelial claudin-5 was reduced in CPDF, but restored with AlaGln, while mesothelial claudin-5 abundance was unchanged. AlaGln supplementation seals the peritoneal endothelial barrier, and when supplemented to conventional PD fluid increases claudin-5 and ZO-1 abundance and clustering of ZO-1 in the endothelial cell membrane.
Collapse
|
23
|
Bonomini M, Borras FE, Troya-Saborido M, Carreras-Planella L, Di Liberato L, Arduini A. Proteomic Research in Peritoneal Dialysis. Int J Mol Sci 2020; 21:ijms21155489. [PMID: 32752018 PMCID: PMC7432538 DOI: 10.3390/ijms21155489] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023] Open
Abstract
Peritoneal dialysis (PD) is an established home care, cost-effective renal replacement therapy (RRT), which offers several advantages over the most used dialysis modality, hemodialysis. Despite its potential benefits, however, PD is an under-prescribed method of treating uremic patients. Infectious complications (primarily peritonitis) and bio-incompatibility of PD solutions are the main contributors to PD drop-out, due to their potential for altering the functional and anatomical integrity of the peritoneal membrane. To improve the clinical outcome of PD, there is a need for biomarkers to identify patients at risk of PD-related complications and to guide personalized interventions. Several recent studies have shown that proteomic investigation may be a powerful tool in the prediction, early diagnosis, prognostic assessment, and therapeutic monitoring of patients on PD. Indeed, analysis of the proteome present in PD effluent has uncovered several proteins involved in inflammation and pro-fibrotic insult, in encapsulating peritoneal sclerosis, or even in detecting early changes before any measurable modifications occur in the traditional clinical parameters used to evaluate PD efficacy. We here review the proteomic studies conducted thus far, addressing the potential use of such omics methodology in identifying potential new biomarkers of the peritoneal membrane welfare in relation to dialytic prescription and adequacy.
Collapse
Affiliation(s)
- Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS. Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy;
- Correspondence:
| | - Francesc E. Borras
- Nephrology Department, Campus Can Ruti, Germans Trias i Pujol Research Institute (IGTP), REMAR-IGTP Group, Germans Trias i Pujol University Hospital, Carretera de Can Ruti, Camí de les Escoles s/n, 08916 Barcelona, Spain; (F.E.B.); (M.T.-S.); (L.C.-P.)
| | - Maribel Troya-Saborido
- Nephrology Department, Campus Can Ruti, Germans Trias i Pujol Research Institute (IGTP), REMAR-IGTP Group, Germans Trias i Pujol University Hospital, Carretera de Can Ruti, Camí de les Escoles s/n, 08916 Barcelona, Spain; (F.E.B.); (M.T.-S.); (L.C.-P.)
| | - Laura Carreras-Planella
- Nephrology Department, Campus Can Ruti, Germans Trias i Pujol Research Institute (IGTP), REMAR-IGTP Group, Germans Trias i Pujol University Hospital, Carretera de Can Ruti, Camí de les Escoles s/n, 08916 Barcelona, Spain; (F.E.B.); (M.T.-S.); (L.C.-P.)
| | - Lorenzo Di Liberato
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS. Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy;
| | - Arduino Arduini
- Department of Research and Development, CoreQuest Sagl, Tecnopolo, 6934 Bioggio, Switzerland;
| |
Collapse
|
24
|
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:biom10050768. [PMID: 32423139 PMCID: PMC7277773 DOI: 10.3390/biom10050768] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023] Open
Abstract
One of the main limitations to successful long-term use of peritoneal dialysis (PD) as a renal replacement therapy is the harmful effects of PD solutions to the structure and function of the peritoneal membrane (PM). In PD, the PM serves as a semipermeable membrane that, due to exposure to PD solutions, undergoes structural alterations, including peritoneal fibrosis, vasculopathy, and neoangiogenesis. In recent decades, oxidative stress (OS) has emerged as a novel risk factor for mortality and cardiovascular disease in PD patients. Moreover, it has become evident that OS plays a pivotal role in the pathogenesis and development of the chronic, progressive injury of the PM. In this review, we aimed to present several aspects of OS in PD patients, including the pathophysiologic effects on the PM, clinical implications, and possible therapeutic antioxidant strategies that might protect the integrity of PM during PD therapy.
Collapse
Affiliation(s)
- Stefanos Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Evangelia Dounousi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Marios Salmas
- Department of Anatomy, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | | | - Vassilios Liakopoulos
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
- Correspondence: ; Tel.: +30-2310-994-694
| |
Collapse
|
25
|
Borceux P, Morelle J, Goffin E. Complement system activation and peritoneal membrane alterations: Culprit or innocent bystander? Perit Dial Int 2020; 40:115-123. [PMID: 32063185 DOI: 10.1177/0896860819896242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Peritoneal dialysis (PD) accounts for approximately 10% of the dialysis population worldwide. Major concern limiting long-term PD success is the loss of the peritoneal membrane function after prolonged exposure to dialysis solutions. The complement system is a major component of the innate immune system, which provides a first-line defense against pathogens. Uncontrolled activation of the complement system directly contributes to the pathophysiology of rare and common kidney diseases and to a growing number of nonrenal diseases. Here, we review currently available evidence of complement activation in patients treated with PD and its association with structural and functional alterations of the peritoneal membrane. Mainly, evidence point toward a local, intraperitoneal, production of complement molecules in response to PD exposure. Dialysis fluids, particularly glucose, play a role in complement activation and dysregulation leading to untoward PD-related pathophysiological processes such as peritoneal fibrosis, angiogenesis, and vasculopathy and, perhaps, encapsulating peritoneal fibrosis development. These findings could lead to further development and use of anticomplement therapeutics in PD patients to prevent membrane damage.
Collapse
Affiliation(s)
- Pauline Borceux
- Division of Nephrology, Cliniques universitaires St Luc, Brussels, Belgium.,Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Johann Morelle
- Division of Nephrology, Cliniques universitaires St Luc, Brussels, Belgium.,Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Eric Goffin
- Division of Nephrology, Cliniques universitaires St Luc, Brussels, Belgium.,Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| |
Collapse
|
26
|
Boehm M, Herzog R, Klinglmüller F, Lichtenauer AM, Wagner A, Unterwurzacher M, Beelen RHJ, Alper SL, Aufricht C, Kratochwill K. The Peritoneal Surface Proteome in a Model of Chronic Peritoneal Dialysis Reveals Mechanisms of Membrane Damage and Preservation. Front Physiol 2019; 10:472. [PMID: 31156443 PMCID: PMC6530346 DOI: 10.3389/fphys.2019.00472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 04/04/2019] [Indexed: 12/31/2022] Open
Abstract
Peritoneal dialysis (PD) fluids are cytotoxic to the peritoneum. Recent studies have shown that alanyl-glutamine (AlaGln) modulates the cellular stress response, improves mesothelial cell survival, reduces submesothelial thickening in experimental models of PD, and in clinical studies improves PD effluent cell stress and immune responses. However, the mechanisms of AlaGln-mediated membrane protection are not yet fully understood. Here, we explore those mechanisms through application of a novel proteomics approach in a clinically relevant in vivo model in rats. Experimental PD was performed for 5 weeks using conventional single-chamber bag (SCB) or neutral dual-chamber bag (DCB), PD fluid (PDF), with or without AlaGln supplementation, via a surgically implanted catheter. Rats subjected to a single dwell without catheter implantation served as controls. The peritoneal surface proteome was directly harvested by detergent extraction and subjected to proteomic analysis by two-dimensional difference gel electrophoresis (2D-DiGE) with protein identification by mass spectrometry. An integrated bioinformatic approach was applied to identify proteins significantly affected by the treatments despite biological variation and interfering high abundance proteins. From 505 of 744 common spots on 59 gels, 222 unique proteins were identified. Using UniProt database information, proteins were assigned either as high abundance plasma proteins, or as cellular proteins. Statistical analysis employed an adapted workflow from RNA-sequencing, the trimmed mean of M-values (TMM) for normalization, and a mixed model for computational identification of significantly differentially abundant proteins. The most prominently enriched pathways after 5 weeks chronic treatment with SCB or DCB, PDFs belonged to clusters reflecting tissue damage and cell differentiation by cytoskeletal reorganization, immune responses, altered metabolism, and oxidative stress and redox homeostasis. Although the AlaGln effect was not as prominent, associated enriched pathways showed mostly regression to control or patterns opposite that of the PDF effect. Our study describes the novel peritoneal surface proteome through combined proteomic and bioinformatic analyses, and assesses changes elicited by chronic experimental PD. The biological processes so identified promise to link molecular mechanisms of membrane damage and protection in the in vivo rat model to pathomechanisms and cytoprotective effects observed in vitro and in clinical PD.
Collapse
Affiliation(s)
- Michael Boehm
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Rebecca Herzog
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Florian Klinglmüller
- Center for Medical Statistics, Informatics, and Intelligent Systems-CeMSIIS, Medical University of Vienna, Vienna, Austria
| | - Anton M Lichtenauer
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Anja Wagner
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Unterwurzacher
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Robert H J Beelen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Seth L Alper
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Christoph Aufricht
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Klaus Kratochwill
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
27
|
Wiesenhofer FM, Herzog R, Boehm M, Wagner A, Unterwurzacher M, Kasper DC, Alper SL, Vychytil A, Aufricht C, Kratochwill K. Targeted Metabolomic Profiling of Peritoneal Dialysis Effluents Shows Anti-oxidative Capacity of Alanyl-Glutamine. Front Physiol 2019; 9:1961. [PMID: 30719009 PMCID: PMC6348277 DOI: 10.3389/fphys.2018.01961] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/28/2018] [Indexed: 01/25/2023] Open
Abstract
Readily available peritoneal dialysis (PD) effluents from PD patients in the course of renal replacement therapy are a potentially rich source for molecular markers for predicting clinical outcome, monitoring the therapy, and therapeutic interventions. The complex clinical phenotype of PD patients might be reflected in the PD effluent metabolome. Metabolomic analysis of PD effluent might allow quantitative detection and assessment of candidate PD biomarkers for prognostication and therapeutic monitoring. We therefore subjected peritoneal equilibration test effluents from 20 stable PD patients, obtained in a randomized controlled trial (RCT) to evaluate cytoprotective effects of standard PD solution (3.86% glucose) supplemented with 8 mM alanyl-glutamine (AlaGln) to targeted metabolomics analysis. One hundred eighty eight pre-defined metabolites, including free amino acids, acylcarnitines, and glycerophospholipids, as well as custom metabolic indicators calculated from these metabolites were surveyed in a high-throughput assay requiring only 10 μl of PD effluent. Metabolite profiles of effluents from the cross-over trial were analyzed with respect to AlaGln status and clinical parameters such as duration of PD therapy and history of previous episodes of peritonitis. This targeted approach detected and quantified 184 small molecules in PD effluent, a larger number of detected metabolites than in all previous metabolomic studies in PD effluent combined. Metabolites were clustered within substance classes regarding concentrations after a 4-h dwell. PD effluent metabolic profiles were differentiated according to PD patient sub-populations, revealing novel changes in small molecule abundance during PD therapy. AlaGln supplementation of PD fluid altered levels of specific metabolites, including increases in alanine and glutamine but not glutamate, and reduced levels of small molecule indicators of oxidative stress, such as methionine sulfoxide. Our study represents the first application of targeted metabolomics to PD effluents. The observed metabolomic changes in PD effluent associated with AlaGln-supplementation during therapy suggested an anti-oxidant effect, and were consistent with the restoration of important stress and immune processes previously noted in the RCT. High-throughput detection of PD effluent metabolomic signatures and their alterations by therapeutic interventions offers new opportunities for metabolome-clinical correlation in PD and for prescription of personalized PD therapy.
Collapse
Affiliation(s)
- Florian M Wiesenhofer
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Rebecca Herzog
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Boehm
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Anja Wagner
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Unterwurzacher
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Andreas Vychytil
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christoph Aufricht
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Bartosova M, Schmitt CP. Biocompatible Peritoneal Dialysis: The Target Is Still Way Off. Front Physiol 2019; 9:1853. [PMID: 30700974 PMCID: PMC6343681 DOI: 10.3389/fphys.2018.01853] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/07/2018] [Indexed: 01/01/2023] Open
Abstract
Peritoneal dialysis (PD) is a cost-effective, home-based therapy for patients with end-stage renal disease achieving similar outcome as compared to hemodialysis. Still, a minority of patients only receive PD. To a significant extend, this discrepancy is explained by major limitations regarding PD efficiency and sustainability. Due to highly unphysiological composition of PD fluids, the peritoneal membrane undergoes rapid morphological and long-term functional alterations, which limit the treatment and contribute to adverse patient outcome. This review is focused on the peritoneal membrane ultrastructure and its transformation in patients with kidney disease and chronic PD, underlying molecular mechanisms, and potential systemic sequelae. Current knowledge on the impact of conventional and second-generation PD fluids is described; novel strategies and innovative PD fluid types are discussed.
Collapse
Affiliation(s)
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine Heidelberg, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
29
|
Wilkie M, Davies S. Does alanyl-glutamine supplementation offer potential to improve peritoneal dialysate biocompatibility? Kidney Int 2018; 94:1050-1052. [PMID: 30466563 DOI: 10.1016/j.kint.2018.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/13/2018] [Indexed: 11/27/2022]
Abstract
Peritoneal dialysis technique survival remains challenging due to peritonitis and long-term alterations to peritoneal membrane function and integrity. Up to now, the development of less bio-incompatible dialysis solutions have not had a major impact on these aspects of the therapy. A novel approach, supplementing dialysis solutions with a cytoprotective additive, alanyl-glutamine, has shown benefits to surrogate biomarkers of cell function in a randomized controlled study.
Collapse
Affiliation(s)
- Martin Wilkie
- Sheffield Kidney Institute, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK.
| | - Simon Davies
- Institute for Science and Technology in Medicine (ISTM) at Keele, Staffordshire, UK
| |
Collapse
|