全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Peritoneal Membrane Injury and Peritoneal Dialysis

DOI: 10.1155/2014/573685

Full-Text   Cite this paper   Add to My Lib

Abstract:

For patients with chronic renal failure, peritoneal dialysis (PD) is a common, life sustaining form of renal replacement therapy that is used worldwide. Exposure to nonbiocompatible dialysate, inflammation, and uremia induces longitudinal changes in the peritoneal membrane. Application of molecular biology techniques has led to advances in our understanding of the mechanism of injury of the peritoneal membrane. This understanding will allow for the development of strategies to preserve the peritoneal membrane structure and function. This may decrease the occurrence of PD technique failure and improve patient outcomes of morbidity and mortality. 1. Introduction PD involves both diffusive and convective clearance driven mainly by glucose-based hyperosmolar PD fluid. The peritoneal membrane overlies the surface of all intra-abdominal organs, the diaphragm, and the parietal peritoneal wall. The peritoneal membrane is a fairly simple structure, with a superficial epithelial-like cell layer—the mesothelium—which is attached to a basement membrane (Figure 1). Beneath the basement membrane is a submesothelial layer consisting of connective tissue, fibroblasts, and blood vessels. Under optimal conditions, the peritoneum acts as an efficient, semipermeable dialysis membrane, enabling removal of metabolites, uremic toxins, salt, and water from the patient. Figure 1: Changes in the peritoneal membrane with dialysis treatment. (a) Normal peritoneal membrane consists of an intact mesothelium overlying a thin submesothelial compact zone containing extracellular matrix, blood vessels, and a few scattered cells—fibroblasts and peritoneal macrophages. (b) After time on dialysis, activated fibroblasts or myofibroblasts appear along with increased submesothelial extracellular matrix and angiogenesis. Mesothelial cells are injured and sometimes denuded from the peritoneal surface. The rate of removal of these products from the blood correlates with the vascular surface area in contact with PD fluids in the peritoneal cavity [1]. Peritoneal membrane solute transport is commonly quantified as a dialysate to plasma ratio of solute (i.e., d/p creatinine). Increased peritoneal membrane solute transport should confer benefit for the patient as blood clearance would be more efficient. However, many studies have demonstrated the opposite [2]. A meta-analysis of observational studies demonstrated that every 0.1 increase in d/p creatinine carries a 15% increased risk of mortality [3]. This risk may be modified by the use of nocturnal cycling PD and use of alternate fluids such as

References

[1]  M. Numata, M. Nakayama, S. Nimura, M. Kawakami, B. Lindholm, and Y. Kawaguchi, “Association between an increased surface area of peritoneal microvessels and a high peritoneal solute transport rate,” Peritoneal Dialysis International, vol. 23, no. 2, pp. 116–122, 2003.
[2]  M. Rumpsfeld, S. P. McDonald, and D. W. Johnson, “Higher peritoneal transport status is associated with higher mortality and technique failure in the Australian and New Zealand peritoneal dialysis patient populations,” Journal of the American Society of Nephrology, vol. 17, no. 1, pp. 271–278, 2006.
[3]  K. S. Brimble, M. Walker, P. J. Margetts, K. K. Kundhal, and C. G. Rabbat, “Meta-analysis: peritoneal membrane transport, mortality, and technique failure in peritoneal dialysis,” Journal of the American Society of Nephrology, vol. 17, no. 9, pp. 2591–2598, 2006.
[4]  S. H. Chung, O. Heimbürger, and B. Lindholm, “Poor outcomes for fast transporters on PD: The rise and fall of a clinical concern,” Seminars in Dialysis, vol. 21, no. 1, pp. 7–10, 2008.
[5]  D. Sobiecka, J. Waniewski, A. Weryński, and B. Lindholm, “Peritoneal fluid transport in CAPD patients with different transport rates of small solutes,” Peritoneal Dialysis International, vol. 24, no. 3, pp. 240–251, 2004.
[6]  A. H. Tzamaloukas, M. C. Saddler, G. H. Murata et al., “Symptomatic fluid retention in patients on continuous peritoneal dialysis,” Journal of the American Society of Nephrology, vol. 6, no. 2, pp. 198–206, 1995.
[7]  Z. Tonbul, L. Altintepe, ?. S?zlü, M. Yeksan, A. Yildiz, and S. Türk, “The association of peritoneal transport properties with 24-hour blood pressure levels in CARP patients,” Peritoneal Dialysis International, vol. 23, no. 1, pp. 46–52, 2003.
[8]  R. N. Foley, P. S. Parfrey, J. D. Harnett, G. M. Kent, D. C. Murray, and P. E. Barre, “Impact of hypertension on cardiomyopathy, morbidity and mortality in end-stage renal disease,” Kidney International, vol. 49, no. 5, pp. 1379–1385, 1996.
[9]  S. Sezer, E. Tutal, Z. Arat, et al., “Peritoneal transport status influence on atherosclerosis/inflammation in CAPD patients,” Journal of Renal Nutrition, vol. 15, no. 4, pp. 427–434, 2005.
[10]  B. Rippe, G. Stelin, and B. Haraldsson, “Computer simulations of peritoneal fluid transport in CAPD,” Kidney International, vol. 40, no. 2, pp. 315–325, 1991.
[11]  B. Rippe and D. Venturoli, “Simulations of osmotic ultrafiltration failure in CAPD using a serial three-pore membrane/fiber matrix model,” The American Journal of Physiology—Renal Physiology, vol. 292, no. 3, pp. F1035–F1043, 2007.
[12]  P. J. Margetts, S. Gyorffy, M. Kolb et al., “Antiangiogenic and antifibrotic gene therapy in a chronic infusion model of peritoneal dialysis in rats,” Journal of the American Society of Nephrology, vol. 13, no. 3, pp. 721–728, 2002.
[13]  P. J. Margetts, M. Kolb, T. Galt, C. M. Hoff, T. R. Shockley, and J. Gauldie, “Gene transfer of transforming growth factor-β1 to the rat peritoneum: effects on membrane function,” Journal of the American Society of Nephrology, vol. 12, no. 10, pp. 2029–2039, 2001.
[14]  P. J. Margetts and D. N. Churchill, “Acquired ultrafiltration dysfunction in peritoneal dialysis patients,” Journal of the American Society of Nephrology, vol. 13, no. 11, pp. 2787–2794, 2002.
[15]  J. D. Williams, K. J. Craig, N. Topley et al., “Morphologic changes in the peritoneal membrane of patients with renal disease,” Journal of the American Society of Nephrology, vol. 13, no. 2, pp. 470–479, 2002.
[16]  J. D. Williams, K. J. Craig, N. Topley, et al., “Morphologic changes in the peritoneal membrane of patients with renal disease,” Journal of the American Society of Nephrology, vol. 13, no. 2, pp. 470–479, 2002.
[17]  S. J. Davies, “Longitudinal relationship between solute transport and ultrafiltration capacity in peritoneal dialysis patients,” Kidney International, vol. 66, no. 6, pp. 2437–2445, 2004.
[18]  G. Garosi and N. di Paolo, “Peritoneal sclerosis: one or two nosological entities,” Seminars in Dialysis, vol. 13, no. 5, pp. 297–308, 2000.
[19]  J. Rubin, G. A. Herrera, and D. Collins, “An autopsy study of the peritoneal cavity from patients on continuous ambulatory peritoneal dialysis,” The American Journal of Kidney Diseases, vol. 18, no. 1, pp. 97–102, 1991.
[20]  K. Honda and H. Oda, “Pathology of encapsulating peritoneal sclerosis,” Peritoneal Dialysis International, vol. 25, no. 4, pp. S19–S29, 2005.
[21]  H. Acloque, M. S. Adams, K. Fishwick, M. Bronner-Fraser, and M. A. Nieto, “Epithelial-mesenchymal transitions: the importance of changing cell state in development and disease,” Journal of Clinical Investigation, vol. 119, no. 6, pp. 1438–1449, 2009.
[22]  A. Moustakas and C.-H. Heldin, “Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression,” Cancer Science, vol. 98, no. 10, pp. 1512–1520, 2007.
[23]  R. Kalluri and R. A. Weinberg, “The basics of epithelial-mesenchymal transition,” Journal of Clinical Investigation, vol. 119, no. 6, pp. 1420–1428, 2009.
[24]  B. Hinz, S. H. Phan, V. J. Thannickal, A. Galli, M.-L. Bochaton-Piallat, and G. Gabbiani, “The myofibroblast: one function, multiple origins,” The American Journal of Pathology, vol. 170, no. 6, pp. 1807–1816, 2007.
[25]  F. R. Bob, G. Gluhovschi, D. Herman et al., “Histological, immunohistochemical and biological data in assessing interstitial fibrosis in patients with chronic glomerulonephritis,” Acta Histochemica, vol. 110, no. 3, pp. 196–203, 2008.
[26]  L. S. Aroeira, J. Loureiro, G. T. González-Mateo, et al., “Characterization of epithelial-to-mesenchymal transition of mesothelial cells in a mouse model of chronic peritoneal exposure to high glucose dialysate,” Peritoneal Dialysis International, vol. 28, supplement 5, pp. S29–S33, 2008.
[27]  E. J. Oh, H. M. Ryu, S. Y. Choi et al., “Impact of low glucose degradation product bicarbonate/lactate-buffered dialysis solution on the epithelial-mesenchymal transition of peritoneum,” The American Journal of Nephrology, vol. 31, no. 1, pp. 58–67, 2009.
[28]  L. S. Aroeira, A. Aguilera, J. A. Sánchez-Tomero 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, vol. 18, no. 7, pp. 2004–2013, 2007.
[29]  P. J. Margetts, P. Bonniaud, L. Liu et al., “Transient overexpression of TGF-β1 induces epithelial mesenchymal transition in the rodent peritoneum,” Journal of the American Society of Nephrology, vol. 16, no. 2, pp. 425–436, 2005.
[30]  M.-A. Yu, K.-S. Shin, J. H. Kim et al., “HGF and BMP-7 ameliorate high glucose-induced epithelial-to-mesenchymal transition of peritoneal mesothelium,” Journal of the American Society of Nephrology, vol. 20, no. 3, pp. 567–581, 2009.
[31]  P. Patel, Y. Sekiguchi, K.-H. Oh, S. E. Patterson, M. R. J. Kolb, and P. J. Margetts, “Smad3-dependent and-independent pathways are involved in peritoneal membrane injury,” Kidney International, vol. 77, no. 4, pp. 319–328, 2010.
[32]  P. J. Margetts, “Twist: a new player in the epithelial-mesenchymal transition of the peritoneal mesothelial cells,” Nephrology Dialysis Transplantation, vol. 27, no. 11, pp. 3978–3981, 2012.
[33]  C. Li, Y. Ren, X. Jia et al., “Twist overexpression promoted epithelial-to-mesenchymal transition of human peritoneal mesothelial cells under high glucose,” Nephrology Dialysis Transplantation, vol. 27, no. 11, pp. 4119–4124, 2012.
[34]  J. H. Cho, J. Y. Do, E. J. Oh et al., “Are ex vivo mesothelial cells representative of the in vivo transition from epithelial-to-mesenchymal cells in peritoneal membrane?” Nephrology Dialysis Transplantation, vol. 27, no. 5, pp. 1768–1779, 2012.
[35]  I. Hirahara, Y. Ishibashi, S. Kaname, E. Kusano, and T. Fujita, “Methylglyoxal induces peritoneal thickening by mesenchymal-like mesothelial cells in rats,” Nephrology Dialysis Transplantation, vol. 24, no. 2, pp. 437–447, 2009.
[36]  Y. Liu, “New insights into epithelial-mesenchymal transition in kidney fibrosis,” Journal of the American Society of Nephrology, vol. 21, no. 2, pp. 212–222, 2010.
[37]  B. D. Humphreys, S.-L. Lin, A. Kobayashi et al., “Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis,” American Journal of Pathology, vol. 176, no. 1, pp. 85–97, 2010.
[38]  Y. T. Chen, Y. T. Chang, S. Y. Pan, et al., “Lineage tracing reveals distinctive fates for mesothelial cells and submesothelial fibroblasts during peritoneal injury,” Journal of the American Society of Nephrology. In press.
[39]  P. Patel, J. West-Mays, M. Kolb, J.-C. Rodrigues, C. M. Hoff, and P. J. Margetts, “Platelet derived growth factor B and epithelial mesenchymal transition of peritoneal mesothelial cells,” Matrix Biology, vol. 29, no. 2, pp. 97–106, 2010.
[40]  Y. Sekiguchi, J. Zhang, S. Patterson et al., “Rapamycin inhibits transforming growth factor β-induced peritoneal angiogenesis by blocking the secondary hypoxic response,” Journal of Cellular and Molecular Medicine, vol. 16, no. 8, pp. 1934–1945, 2012.
[41]  J. Zhang, K.-H. Oh, H. Xu, and P. J. Margetts, “Vascular endothelial growth factor expression in peritoneal mesothelial cells undergoing transdifferentiation,” Peritoneal Dialysis International, vol. 28, no. 5, pp. 497–504, 2008.
[42]  L. S. Aroeira, A. Aguilera, R. Selgas et al., “Mesenchymal conversion of mesothelial cells as a mechanism responsible for high solute transport rate in peritoneal dialysis: role of vascular endothelial growth factor,” American Journal of Kidney Diseases, vol. 46, no. 5, pp. 938–948, 2005.
[43]  A. B. Roberts, M. B. Sporn, and R. K. Assoian, “Transforming growth factor type β: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro,” Proceedings of the National Academy of Sciences of the United States of America, vol. 83, no. 12, pp. 4167–4171, 1986.
[44]  P. J. Margetts, P. Bonniaud, L. Liu et al., “Transient overexpression of TGF-1 induces epithelial mesenchymal transition in the rodent peritoneum,” Journal of the American Society of Nephrology, vol. 16, no. 2, pp. 425–436, 2005.
[45]  Y. Kyuden, T. Ito, T. Masaki, N. Yorioka, and N. Kohno, “TGF-β1 induced by high glucose is controlled by angiotensin-converting enzyme inhibitor and angiotensin II receptor blocker on cultured human peritoneal mesothelial cells,” Peritoneal Dialysis International, vol. 25, no. 5, pp. 483–491, 2005.
[46]  G. Conti, A. Amore, P. Cirina, L. Peruzzi, S. Balegno, and R. Coppo, “Glycated adducts induce mesothelial cell transdifferentiation: role of glucose and icodextrin dialysis solutions,” Journal of Nephrology, vol. 21, no. 3, pp. 426–437, 2008.
[47]  P. J. Margetts, M. Kolb, L. Yu et al., “Inflammatory cytokines, angiogenesis, and fibrosis in the rat peritoneum,” The American Journal of Pathology, vol. 160, no. 6, pp. 2285–2294, 2002.
[48]  Y. Sun, F. Zhu, X. Yu et al., “Treatment of established peritoneal fibrosis by gene transfer of Smad7 in a rat model of peritoneal dialysis,” American Journal of Nephrology, vol. 30, no. 1, pp. 84–94, 2009.
[49]  H. Guo, J. C. K. Leung, M. F. Lam et al., “Smad7 transgene attenuates peritoneal fibrosis in uremic rats treated with peritoneal dialysis,” Journal of the American Society of Nephrology, vol. 18, no. 10, pp. 2689–2703, 2007.
[50]  J. Loureiro, M. Schilte, A. Aguilera et al., “BMP-7 blocks mesenchymal conversion of mesothelial cells and prevents peritoneal damage induced by dialysis fluid exposure,” Nephrology Dialysis Transplantation, vol. 25, no. 4, pp. 1098–1108, 2010.
[51]  A. S. Gangji, K. S. Brimble, and P. J. Margetts, “Association between markers of inflammation, fibrosis and hypervolemia in peritoneal dialysis patients,” Blood Purification, vol. 28, no. 4, pp. 354–358, 2009.
[52]  J.-H. Cho, I.-K. Hur, C.-D. Kim et al., “Impact of systemic and local peritoneal inflammation on peritoneal solute transport rate in new peritoneal dialysis patients: a 1-year prospective study,” Nephrology Dialysis Transplantation, vol. 25, no. 6, pp. 1964–1973, 2010.
[53]  L. Liu, C.-X. Shi, A. Ghayur et al., “Prolonged peritoneal gene expression using a helper-dependent adenovirus,” Peritoneal Dialysis International, vol. 29, no. 5, pp. 508–516, 2009.
[54]  W. Peng, Q. Zhou, X. Ao, R. Tang, and Z. Xiao, “Inhibition of Rho-kinase alleviates peritoneal fibrosis and angiogenesis in a rat model of peritoneal dialysis,” Renal Failure, vol. 35, no. 7, pp. 958–966, 2013.
[55]  R. Strippoli, I. Benedicto, M. L. Perez Lozano et al., “Inhibition of transforming growth factor-activated kinase 1 (TAK1) blocks and reverses epithelial to mesenchymal transition of mesothelial cells,” PLoS ONE, vol. 7, no. 2, Article ID e31492, 2012.
[56]  S. Kokubo, N. Sakai, K. Furuichi et al., “Activation of p38 mitogen-activated protein kinase promotes peritoneal fibrosis by regulating fibrocytes,” Peritoneal Dialysis International, vol. 32, no. 1, pp. 10–19, 2012.
[57]  P. J. Margetts, M. Kolb, L. Yu, C. M. Hoff, and J. Gauldie, “A chronic inflammatory infusion model of peritoneal dialysis in rats,” Peritoneal Dialysis International, vol. 21, no. 3, pp. S368–S372, 2001.
[58]  J. Plum, S. Hermann, A. Fussh?ller et al., “Peritoneal sclerosis in peritoneal dialysis patients related to dialysis settings and peritoneal transport properties,” Kidney International, Supplement, vol. 59, no. 78, pp. S42–S47, 2001.
[59]  M. A. M. Mateijsen, A. C. van der Wal, P. M. E. M. Hendriks et al., “Vascular and interstitial changes in the peritoneum of CAPD patients with peritoneal sclerosis,” Peritoneal Dialysis International, vol. 19, no. 6, pp. 517–525, 1999.
[60]  M. C. Dickson, J. S. Martin, F. M. Cousins, A. B. Kulkarni, S. Karlsson, and R. J. Akhurst, “Defective haematopoiesis and vasculogenesis in transforming growth factor-β1 knock out mice,” Development, vol. 121, no. 6, pp. 1845–1854, 1995.
[61]  T. O. Daniel and D. Abrahamson, “Endothelial signal integration in vascular assembly,” Annual Review of Physiology, vol. 62, pp. 649–671, 2000.
[62]  S. P. Oh, T. Seki, K. A. Goss et al., “Activin receptor-like kinase 1 modulates transforming growth factor-β1 signaling in the regulation of angiogenesis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 97, no. 6, pp. 2626–2631, 2000.
[63]  J. Folkman and P. A. D'Amore, “Blood vessel formation: what is its molecular basis?” Cell, vol. 87, no. 7, pp. 1153–1155, 1996.
[64]  T. Sánchez-Elsner, L. M. Botella, B. Velasco, A. Corbí, L. Attisano, and C. Bernabéu, “Synergistic cooperation between hypoxia and transforming growth factor-beta pathways on human vascular endothelial growth factor gene expression,” The Journal of Biological Chemistry, vol. 276, no. 42, pp. 38527–38535, 2001.
[65]  G. Breier, S. Blum, J. Peli et al., “Transforming growth factor- and RAS regulate the VEGF/VEGF-receptor system during tumor angiogenesis,” International Journal of Cancer, vol. 97, no. 2, pp. 142–148, 2002.
[66]  M. C. Iglesias-de la Cruz, F. N. Ziyadeh, M. Isono et al., “Effects of high glucose and TGF-β1 on the expression of collagen IV and vascular endothelial growth factor in mouse podocytes,” Kidney International, vol. 62, no. 3, pp. 901–913, 2002.
[67]  B. B. Scott, P. F. Zaratin, A. Colombo, M. J. Hansbury, J. D. Winkler, and J. R. Jackson, “Constitutive expression of angiopoietin-1 and -2 and modulation of their expression by inflammatory cytokines in rheumatoid arthritis synovial fibroblasts,” The Journal of Rheumatology, vol. 29, no. 2, pp. 230–239, 2002.
[68]  P. J. Margetts, M. Kolb, C. M. Hoff, and J. Gauldie, “The role of angiopoietins in resolution of angiogenesis resulting from adenovirual mediated gene transfer of TGF1 or VEGF to the rat peritoneum,” Journal of the American Society of Nephrology, vol. 12, Abstract 433A, no. 10, 2001.
[69]  V. E. Belozerov and E. G. van Meir, “Hypoxia inducible factor-1: a novel target for cancer therapy,” Anti-Cancer Drugs, vol. 16, no. 9, pp. 901–909, 2005.
[70]  P. K. Majumder, P. G. Febbo, R. Bikoff et al., “mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1-dependent pathways,” Nature Medicine, vol. 10, no. 6, pp. 594–601, 2004.
[71]  H. O. Akman, H. Zhang, M. A. Q. Siddiqui, W. Solomon, E. L. P. Smith, and O. A. Batuman, “Response to hypoxia involves transforming growth factor-β2 and Smad proteins in human endothelial cells,” Blood, vol. 98, no. 12, pp. 3324–3331, 2001.
[72]  H. Zhang, H. O. Akman, E. L. P. Smith et al., “Cellular response to hypoxia involves signaling via Smad proteins,” Blood, vol. 101, no. 6, pp. 2253–2260, 2003.
[73]  E. Papakonstantinou, A. J. Aletras, M. Roth, M. Tamm, and G. Karakiulakis, “Hypoxia modulates the effects of transforming growth factor-β isoforms on matrix-formation by primary human lung fibroblasts,” Cytokine, vol. 24, no. 1-2, pp. 25–35, 2003.
[74]  M. Kolb, P. Bonniaud, T. Galt, et al., “Differences in the fibrogenic response after transfer of active transforming growth factor-β1 gene to lungs of “fibrosis-prone” and “fibrosis-resistant” mouse strains,” American Journal of Respiratory Cell and Molecular Biology, vol. 27, no. 2, pp. 141–150, 2002.
[75]  P. Bonniaud, P. J. Margetts, M. Kolb et al., “Adenoviral gene transfer of connective tissue growth factor in the lung induces transient fibrosis,” The American Journal of Respiratory and Critical Care Medicine, vol. 168, no. 7, pp. 770–778, 2003.
[76]  D. F. Higgins, M. P. Biju, Y. Akai, A. Wutz, R. S. Johnson, and V. H. Haase, “Hypoxic induction of Ctgf is directly mediated by Hif-1,” The American Journal of Physiology—Renal Physiology, vol. 287, no. 6, pp. F1223–F1232, 2004.
[77]  D. F. Higgins, K. Kimura, W. M. Bernhardt et al., “Hypoxia promotes fibrogenesis in vivo via HIF-1 stimulation of epithelial-to-mesenchymal transition,” Journal of Clinical Investigation, vol. 117, no. 12, pp. 3810–3820, 2007.
[78]  K. Chaudhary and R. Khanna, “Biocompatible peritoneal dialysis solutions: do we have one?” Clinical Journal of the American Society of Nephrology, vol. 5, no. 4, pp. 723–732, 2010.
[79]  P. G. Blake, A. K. Jain, and S. Yohanna, “Biocompatible peritoneal dialysis solutions: many questions but few answers,” Kidney International, vol. 84, no. 5, pp. 864–866, 2013.
[80]  D.-H. Kang, Y.-S. Hong, H. J. Lim, J.-H. Choi, D.-S. Han, and K.-L. Yoon, “High glucose solution and spent dialysate stimulate the synthesis of transforming growth factor-β1 of human peritoneal mesothelial cells: effect of cytokine costimulation,” Peritoneal Dialysis International, vol. 19, no. 3, pp. 221–230, 1999.
[81]  S. J. Davies, L. Phillips, P. F. Naish, and G. I. Russell, “Peritoneal glucose exposure and changes in membrane solute transport with time on peritoneal dialysis,” Journal of the American Society of Nephrology, vol. 12, no. 5, pp. 1046–1051, 2001.
[82]  Y. Wen, Q. Guo, X. Yang, et al., “High glucose concentrations in peritoneal dialysis are associated with all-cause and cardiovascular disease mortality in continuous ambulatory peritoneal dialysis patients,” Peritoneal Dialysis International, 2013.
[83]  A. S. de Vriese, R. G. Tilton, C. C. Stephan, and N. H. Lameire, “Vascular endothelial growth factor is essential for hyperglycemia-induced structural and functional alterations of the peritoneal membrane,” Journal of the American Society of Nephrology, vol. 12, no. 8, pp. 1734–1741, 2001.
[84]  G. Clerbaux, J. Francart, P. Wallemacq, A. Robert, and E. Goffin, “Evaluation of peritoneal transport properties at onset of peritoneal dialysis and longitudinal follow-up,” Nephrology Dialysis Transplantation, vol. 21, no. 4, pp. 1032–1039, 2006.
[85]  A. S. Rodrigues, M. Martins, J. C. Korevaar et al., “Evaluation of peritoneal transport and membrane status in peritoneal dialysis: focus on incident fast transporters,” The American Journal of Nephrology, vol. 27, no. 1, pp. 84–91, 2007.
[86]  A. S. de Vriese, A. Flyvbjerg, S. Mortier, R. G. Tilton, and N. H. Lameire, “Inhibition of the interaction of age-rage prevents hyperglycemia-induced fibrosis of the peritoneal membrane,” Journal of the American Society of Nephrology, vol. 14, no. 8, pp. 2109–2118, 2003.
[87]  Y. Cho, D. W. Johnson, J. C. Craig, et al., “Biocompatible dialysis fluids for peritoneal dialysis,” Cochrane Database of Systematic Reviews, vol. 3, Article ID CD007554, 2014.
[88]  D. W. Johnson, F. G. Brown, M. Clarke et al., “The effect of low glucose degradation product, neutral pH versus standard peritoneal dialysis solutions on peritoneal membrane function: the balANZ trial,” Nephrology Dialysis Transplantation, vol. 27, no. 12, pp. 4445–4453, 2012.
[89]  C.-Y. Lin, W.-P. Chen, L.-Y. Yang, A. Chen, and T.-P. Huang, “Persistent transforming growth factor-beta 1 expression may predict peritoneal fibrosis in CAPD patients with frequent peritonitis occurrence,” American Journal of Nephrology, vol. 18, no. 6, pp. 513–519, 1998.
[90]  P. J. Margetts, M. Kolb, L. Yu et al., “Inflammatory cytokines, angiogenesis, and fibrosis in the rat peritoneum,” American Journal of Pathology, vol. 160, no. 6, pp. 2285–2294, 2002.
[91]  G. del Peso, M. J. Fernández-Reyes, C. Hevia et al., “Factors influencing peritoneal transport parameters during the first year on peritoneal dialysis: peritonitis is the main factor,” Nephrology Dialysis Transplantation, vol. 20, pp. 1201–1206, 2005.
[92]  A. T. van Diepen, E. S. Van, D. G. Struijk, and R. T. Krediet, “The first peritonitis episode alters the natural course of peritoneal membrane characteristics in peritoneal dialysis patients,” Peritoneal Dialysis International, 2014.
[93]  T. Hirano, K. Yasukawa, H. Harada et al., “Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin,” Nature, vol. 324, no. 6092, pp. 73–76, 1986.
[94]  K.-H. Oh, J. Y. Jung, M. O. Yoon et al., “Intra-peritoneal interleukin-6 system is a potent determinant of the baseline peritoneal solute transport in incident peritoneal dialysis patients,” Nephrology Dialysis Transplantation, vol. 25, no. 5, pp. 1639–1646, 2010.
[95]  M. Lambie, J. Chess, K. L. Donovan et al., “Independent effects of systemic and peritoneal inflammation on peritoneal dialysis survival,” Journal of the American Society of Nephrology, vol. 24, no. 12, pp. 2071–2080, 2013.
[96]  S. Combet, M.-L. Ferrier, M. van Landschoot, et al., “Chronic uremia induces permeability changes, increased nitric oxide synthase expression, and structural modifications in the peritoneum,” Journal of the American Society of Nephrology, vol. 12, no. 10, pp. 2146–2157, 2001.
[97]  K. Honda, C. Hamada, M. Nakayama et al., “Impact of uremia, diabetes, and peritoneal dialysis itself on the pathogenesis of peritoneal sclerosis: a quantitative study of peritoneal membrane morphology,” Clinical Journal of the American Society of Nephrology, vol. 3, no. 3, pp. 720–728, 2008.
[98]  G. Gillerot, E. Goffin, C. Michel et al., “Genetic and clinical factors influence the baseline permeability of the peritoneal membrane,” Kidney International, vol. 67, no. 6, pp. 2477–2487, 2005.
[99]  Y.-H. Hwang, M.-J. Son, J. Yang et al., “Effects of interleukin-6 T15A single nucleotide polymorphism on baseline peritoneal solute transport rate in incident peritoneal dialysis patients,” Peritoneal Dialysis International, vol. 29, no. 1, pp. 81–88, 2009.
[100]  T. Y.-H. Wong, C.-C. Szeto, C. Y.-K. Szeto, K.-B. Lai, K.-M. Chow, and P. K.-T. Li, “Association of ENOS polymorphism with basal peritoneal membrane function in uremic patients,” The American Journal of Kidney Diseases, vol. 42, no. 4, pp. 781–786, 2003.
[101]  A. Akcay, H. Micozkadioglu, F. B. Atac, E. Agca, and F. N. Ozdemir, “Relationship of ENOS and RAS gene polymorphisms to initial peritoneal transport status in peritoneal dialysis patients,” Nephron-Clinical Practice, vol. 104, no. 1, pp. c41–c46, 2006.
[102]  Y. Maruyama, M. Numata, M. Nakayama et al., “Relationship between the -374T/A receptor of advanced glycation end products gene polymorphism and peritoneal solute transport status at the initiation of peritoneal dialysis,” Therapeutic Apheresis and Dialysis, vol. 11, no. 4, pp. 301–305, 2007.
[103]  C.-C. Szeto, K.-M. Chow, P. Poon, C. Y.-K. Szeto, T. Y.-H. Wong, and P. K.-T. Li, “Genetic polymorphism of VEGF: impact on longitudinal change of peritoneal transport and survival of peritoneal dialysis patients,” Kidney International, vol. 65, no. 5, pp. 1947–1955, 2004.
[104]  Y.-T. Lee, Y.-C. Tsai, Y.-K. Yang et al., “Association between interleukin-10 gene polymorphism ?592 (A/C) and peritoneal transport in patients undergoing peritoneal dialysis,” Nephrology, vol. 16, no. 7, pp. 663–671, 2011.
[105]  M. Numata, M. Nakayama, T. Hosoya 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, vol. 62, no. 6, pp. 455–460, 2004.
[106]  P. J. Margetts, C. Hoff, L. Liu et al., “Transforming growth factor β-induced peritoneal fibrosis is mouse strain dependent,” Nephrology Dialysis Transplantation, vol. 28, no. 8, pp. 2015–2027, 2013.
[107]  A. Leelahavanichkul, Q. Yan, X. Hu et al., “Angiotensin II overcomes strain-dependent resistance of rapid CKD progression in a new remnant kidney mouse model,” Kidney International, vol. 78, no. 11, pp. 1136–1153, 2010.
[108]  S. Hillebrandt, C. Goos, S. Matern, and F. Lammert, “Genome-wide analysis of hepatic fibrosis in inbred mice identifies the susceptibility locus Hfib1 on chromosome 15,” Gastroenterology, vol. 123, no. 6, pp. 2041–2051, 2002.
[109]  S. W. M. Van Den Borne, V. A. M. Van De Schans, A. E. Strzelecka et al., “Mouse strain determines the outcome of wound healing after myocardial infarction,” Cardiovascular Research, vol. 84, no. 2, pp. 273–282, 2009.
[110]  P. W. Noble, C. E. Barkauskas, and D. Jiang, “Pulmonary fibrosis: patterns and perpetrators,” Journal of Clinical Investigation, vol. 122, no. 8, pp. 2756–2762, 2012.
[111]  E. M. Zeisberg and M. Zeisberg, “The role of promoter hypermethylation in fibroblast activation and fibrogenesis,” The Journal of Pathology, vol. 229, no. 2, pp. 264–273, 2013.
[112]  G. Balasubramaniam, E. A. Brown, A. Davenport, et al., “The Pan-Thames EPS study: treatment and outcomes of encapsulating peritoneal sclerosis,” Nephrology Dialysis Transplantation, vol. 24, no. 10, pp. 3209–3215, 2009.
[113]  W. Bechtel, S. McGoohan, E. M. Zeisberg et al., “Methylation determines fibroblast activation and fibrogenesis in the kidney,” Nature Medicine, vol. 16, no. 5, pp. 544–550, 2010.
[114]  J. Mann and D. A. Mann, “Epigenetic regulation of wound healing and fibrosis,” Current Opinion in Rheumatology, vol. 25, no. 1, pp. 101–107, 2013.
[115]  M. Pang and S. Zhuang, “Histone deacetylase: a potential therapeutic target for fibrotic disorders,” Journal of Pharmacology and Experimental Therapeutics, vol. 335, no. 2, pp. 266–272, 2010.
[116]  K. van Beneden, I. Mannaerts, M. Pauwels, C. van den Branden, and L. A. van Grunsven, “HDAC inhibitors in experimental liver and kidney fibrosis,” Fibrogenesis and Tissue Repair, vol. 6, article 1, 2013.
[117]  C. Huang, M. Xu, and B. Zhu, “Epigenetic inheritance mediated by histone lysine methylation: maintaining transcriptional states without the precise restoration of marks?” Philosophical Transactions of the Royal Society B: Biological Sciences, vol. 368, no. 1609, Article ID 20110332, 2013.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133