Endothelial Microparticles and Systemic Complement Activation in Patients With Chronic Kidney Disease

Diana Jalal, Brandon Renner, Jennifer Laskowski, Erik Stites, James Cooper, Karissa Valente, Zhiying You, Loni Perrenoud, Moglie Le Quintrec, Ismaeel Muhamed, Uwe Christians, Jelena Klawitter, Margaret A Lindorfer, Ronald P Taylor, V Michael Holers, Joshua M Thurman, Diana Jalal, Brandon Renner, Jennifer Laskowski, Erik Stites, James Cooper, Karissa Valente, Zhiying You, Loni Perrenoud, Moglie Le Quintrec, Ismaeel Muhamed, Uwe Christians, Jelena Klawitter, Margaret A Lindorfer, Ronald P Taylor, V Michael Holers, Joshua M Thurman

Abstract

Background: Endothelial microparticles are associated with chronic kidney disease (CKD) and complement activation. We hypothesized that the complement pathway is activated in patients with CKD via endothelial microparticles and that complement activation correlates with endothelial dysfunction in CKD.

Methods and results: We analyzed complement data of 30 healthy subjects, 30 patients with stage III/IV CKD, and 30 renal transplant recipients with stage III/IV CKD, evaluating the potential correlation of complement fragments with brachial artery flow-mediated dilation, Chronic Kidney Disease Epidemiology Collaboration glomerular filtration rate, and urinary albumin/creatinine ratio. Endothelial microparticles were characterized via proteomic analysis and compared between study groups. Complement fragment Ba was significantly increased in CKD and post-kidney transplant CKD. Plasma Ba levels correlated significantly with lower brachial artery flow-mediated dilation, lower Chronic Kidney Disease Epidemiology Collaboration glomerular filtration rate, and higher urinary albumin/creatinine ratio. Factor D levels were significantly higher in the plasma microparticles of patients with CKD versus healthy controls. Plasma microparticles isolated from patients with CKD and containing factor D activated the alternative pathway in vitro.

Conclusion: The alternative complement pathway is activated in CKD and correlates with endothelial dysfunction and markers of CKD. Future studies are needed to evaluate whether endothelial microparticles with increased factor D play a pathologic role in CKD-associated vascular disease.

Clinical trial registration: URL: http://www.clinicaltrials.gov. Unique identifier: NCT02230202.

Keywords: Microparticles complement activation; chronic kidney disease.

© 2018 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley.

Figures

Figure 1
Figure 1
Levels of complement activation fragments are higher in patients with stage III/IV CKD compared with healthy controls. A, Plasma Ba and C5b‐9 levels were significantly higher in stage III/IV CKD and transplant patients compared with healthy controls. There was also a trend toward higher levels of the other complement fragments in CKD patients, but these levels were not significantly higher than in healthy controls. B, Urine Ba levels were higher in patients with stage III/IV CKD compared with healthy controls. ***P<0.001, *P<0.05.
Figure 2
Figure 2
Plasma levels of complement fragment Ba are significantly and inversely correlated with BA‐FMD in all of the recruited participants (correlation coefficient −0.26, P=0.016) suggesting that complement activation is related to endothelial dysfunction.
Figure 3
Figure 3
Complement proteins in microparticles from patients with CKD are altered compared with microparticles in healthy controls. We used an aptamer‐based assay (SOMAscan) to measure microparticle proteins with high sensitivity. A, Factor D levels were significantly higher in the microparticles of patients with CKD and patients with transplants with CKD compared with healthy controls. Factor B levels were lower in these groups compared with healthy controls. B, Levels of C3, iC3b, and C3d were not significantly different among the patient groups. C, Levels of the complement inhibitory protein CD59 was higher in patients with CKD than in healthy controls. Levels of factor H and decay accelerating factor were not statistically different among the 3 groups. ***P<0.001, *P<0.05.
Figure 4
Figure 4
Endothelial microparticle number and bound C3 fragments are not significantly increased in patients with CKD. A, We used sizing beads to identify 0.2 to 1.0 μm by forward scatter and size scatter. Counting beads were added to the plasma before microparticle purification, and were used to compare the number of microparticles isolated from each sample. Endothelial microparticles were analyzed by gating on the CD105+CD41a‐ microparticle population. We also analyzed C3 fragment deposition on the endothelial microparticles. B, The size of the microparticles purified from plasma was slightly larger than those in the original plasma sample when compared by forward scatter (FSC). *P<0.05. Data points from healthy control samples are white, and data points from CKD samples are solid black. C, The size of the purified microparticles was the same in the 3 different patient groups studied. D, The number of microparticles in patients with CKD and patients with transplants was not significantly higher than in healthy controls. E, The amounts of C3 fragments deposited on the endothelial microparticles in patients with CKD and patients with transplants was not significantly different than in healthy controls.
Figure 5
Figure 5
Microparticle‐associated factor D activates plasma complement proteins. To test whether FD in plasma microparticles is catalytically active, we examined whether it could restore complement activity to (FDS. A, Using an alternative pathway assay, we confirmed that purified FD restored activity to FDS. We also confirmed that an anti‐FD reduced complement activity in the FDS reconstituted with FD. B, Similarly, (CKD MPs restored activity to the FDS. The addition of an inhibitory antibody to factor D prevented this response, however, confirming that the effect was due to factor D protein contained within the isolated microparticles. C, Staining of the microparticles for factor D confirmed that it was present on the surface. Anti‐FD indicates anti‐factor D antibody; CKD MPs, microparticles purified from the plasma of a patient with CKD; FD, factor D; and FDS, factor D–depleted serum. ***P<0.001, **P<0.01, *P<0.05.

References

    1. Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro AF III, Feldman HI, Kusek JW, Eggers P, Van Lente F, Greene T, Coresh J; CKD EPI . A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150:604–612.
    1. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med. 2004;351:1296–1305.
    1. Keith DS, Nichols GA, Gullion CM, Brown JB, Smith DH. Longitudinal follow‐up and outcomes among a population with chronic kidney disease in a large managed care organization. Arch Intern Med. 2004;164:659–663.
    1. Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, Culleton B, Hamm LL, McCullough PA, Kasiske BL, Kelepouris E, Klag MJ, Parfrey P, Pfeffer M, Raij L, Spinosa DJ, Wilson PW; American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention . Kidney disease as a risk factor for development of cardiovascular disease: a statement from the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation. 2003;108:2154–2169.
    1. Whaley‐Connell AT, Sowers JR, Stevens LA, McFarlane SI, Shlipak MG, Norris KC, Chen SC, Qiu Y, Wang C, Li S, Vassalotti JA, Collins AJ; Kidney Early Evaluation Program Investigators . CKD in the United States: Kidney Early Evaluation Program (KEEP) and National Health and Nutrition Examination Survey (NHANES) 1999–2004. Am J Kidney Dis. 2008;51:S13–S20.
    1. Rao MV, Qiu Y, Wang C, Bakris G. Hypertension and CKD: Kidney Early Evaluation Program (KEEP) and National Health and Nutrition Examination Survey (NHANES), 1999–2004. Am J Kidney Dis. 2008;51:S30–S37.
    1. Sarnak MJ, Coronado BE, Greene T, Wang SR, Kusek JW, Beck GJ, Levey AS. Cardiovascular disease risk factors in chronic renal insufficiency. Clin Nephrol. 2002;57:327–335.
    1. Silverstein DM. Inflammation in chronic kidney disease: role in the progression of renal and cardiovascular disease. Pediatr Nephrol. 2009;24:1445–1452.
    1. Shlipak MG, Fried LF, Crump C, Bleyer AJ, Manolio TA, Tracy RP, Furberg CD, Psaty BM. Elevations of inflammatory and procoagulant biomarkers in elderly persons with renal insufficiency. Circulation. 2003;107:87–92.
    1. Landray MJ, Wheeler DC, Lip GY, Newman DJ, Blann AD, McGlynn FJ, Ball S, Townend JN, Baigent C. Inflammation, endothelial dysfunction, and platelet activation in patients with chronic kidney disease: the chronic renal impairment in Birmingham (CRIB) study. Am J Kidney Dis. 2004;43:244–253.
    1. Oberg BP, McMenamin E, Lucas FL, McMonagle E, Morrow J, Ikizler TA, Himmelfarb J. Increased prevalence of oxidant stress and inflammation in patients with moderate to severe chronic kidney disease. Kidney Int. 2004;65:1009–1016.
    1. Jalal D, Chonchol M, Etgen T, Sander D. C‐reactive protein as a predictor of cardiovascular events in elderly patients with chronic kidney disease. J Nephrol. 2012;25:719–725.
    1. Nowak KL, Chonchol M, Ikizler TA, Farmer‐Bailey H, Salas N, Chaudhry R, Wang W, Smits G, Tengesdal I, Dinarello CA, Hung AM. IL‐1 inhibition and vascular function in CKD. J Am Soc Nephrol. 2017;28:971–980.
    1. de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, Christ‐Schmidt H, Goldsberry A, Houser M, Krauth M, Lambers Heerspink HJ, McMurray JJ, Meyer CJ, Parving HH, Remuzzi G, Toto RD, Vaziri ND, Wanner C, Wittes J, Wrolstad D, Chertow GM; Investigators BT . Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med. 2013;369:2492–2503.
    1. Anderson HC, Mulhall D, Garimella R. Role of extracellular membrane vesicles in the pathogenesis of various diseases, including cancer, renal diseases, atherosclerosis, and arthritis. Lab Invest. 2010;90:1549–1557.
    1. Beyer C, Pisetsky DS. The role of microparticles in the pathogenesis of rheumatic diseases. Nat Rev Rheumatol. 2010;6:21–29.
    1. Hsu CY, Huang PH, Chiang CH, Leu HB, Huang CC, Chen JW, Lin SJ. Increased circulating endothelial apoptotic microparticle to endothelial progenitor cell ratio is associated with subsequent decline in glomerular filtration rate in hypertensive patients. PLoS One. 2013;8:e68644.
    1. Huang PH, Huang SS, Chen YH, Lin CP, Chiang KH, Chen JS, Tsai HY, Lin FY, Chen JW, Lin SJ. Increased circulating CD31+/annexin V+ apoptotic microparticles and decreased circulating endothelial progenitor cell levels in hypertensive patients with microalbuminuria. J Hypertens. 2010;28:1655–1665.
    1. Jourde‐Chiche N, Dou L, Sabatier F, Calaf R, Cerini C, Robert S, Camoin‐Jau L, Charpiot P, Argiles A, Dignat‐George F, Brunet P. Levels of circulating endothelial progenitor cells are related to uremic toxins and vascular injury in hemodialysis patients. J Thromb Haemost. 2009;7:1576–1584.
    1. Trappenburg MC, van Schilfgaarde M, Frerichs FC, Spronk HM, ten Cate H, de Fijter CW, Terpstra WE, Leyte A. Chronic renal failure is accompanied by endothelial activation and a large increase in microparticle numbers with reduced procoagulant capacity. Nephrol Dial Transplant. 2012;27:1446–1453.
    1. Chironi GN, Boulanger CM, Simon A, Dignat‐George F, Freyssinet JM, Tedgui A. Endothelial microparticles in diseases. Cell Tissue Res. 2009;335:143–151.
    1. Boulanger CM, Amabile N, Guerin AP, Pannier B, Leroyer AS, Mallat CN, Tedgui A, London GM. In vivo shear stress determines circulating levels of endothelial microparticles in end‐stage renal disease. Hypertension. 2007;49:902–908.
    1. Renner B, Klawitter J, Goldberg R, McCullough JW, Ferreira VP, Cooper JE, Christians U, Thurman JM. Cyclosporine induces endothelial cell release of complement‐activating microparticles. J Am Soc Nephrol. 2013;24:1849–1862.
    1. Brown KM, Sacks SH, Sheerin NS. Mechanisms of disease: the complement system in renal injury—new ways of looking at an old foe. Nat Clin Pract Nephrol. 2007;3:277–286.
    1. McCullough JW, Renner B, Thurman JM. The role of the complement system in acute kidney injury. Semin Nephrol. 2013;33:543–556.
    1. Noris M, Remuzzi G. Atypical hemolytic‐uremic syndrome. N Engl J Med. 2009;361:1676–1687.
    1. Pickering MC, Cook HT. Translational mini‐review series on complement factor H: renal diseases associated with complement factor H: novel insights from humans and animals. Clin Exp Immunol. 2008;151:210–230.
    1. Pavlov VI, Skjoedt MO, Siow Tan Y, Rosbjerg A, Garred P, Stahl GL. Endogenous and natural complement inhibitor attenuates myocardial injury and arterial thrombogenesis. Circulation. 2012;126:2227–2235.
    1. Leung VW, Yun S, Botto M, Mason JC, Malik TH, Song W, Paixao‐Cavalcante D, Pickering MC, Boyle JJ, Haskard DO. Decay‐accelerating factor suppresses complement C3 activation and retards atherosclerosis in low‐density lipoprotein receptor‐deficient mice. Am J Pathol. 2009;175:1757–1767.
    1. Fearn A, Sheerin NS. Complement activation in progressive renal disease. World J Nephrol. 2015;4:31–40.
    1. Thurman JM, Wong M, Renner B, Frazer‐Abel A, Giclas PC, Joy MS, Jalal D, Radeva MK, Gassman J, Gipson DS, Kaskel F, Friedman A, Trachtman H. Complement activation in patients with focal segmental glomerulosclerosis. PLoS One. 2015;10:e0136558.
    1. Shechter M, Shechter A, Koren‐Morag N, Feinberg MS, Hiersch L. Usefulness of brachial artery flow‐mediated dilation to predict long‐term cardiovascular events in subjects without heart disease. Am J Cardiol. 2014;113:162–167.
    1. Shechter M, Issachar A, Marai I, Koren‐Morag N, Freinark D, Shahar Y, Shechter A, Feinberg MS. Long‐term association of brachial artery flow‐mediated vasodilation and cardiovascular events in middle‐aged subjects with no apparent heart disease. Int J Cardiol. 2009;134:52–58.
    1. Brevetti G, Silvestro A, Schiano V, Chiariello M. Endothelial dysfunction and cardiovascular risk prediction in peripheral arterial disease: additive value of flow‐mediated dilation to ankle‐brachial pressure index. Circulation. 2003;108:2093–2098.
    1. Yilmaz MI, Stenvinkel P, Sonmez A, Saglam M, Yaman H, Kilic S, Eyileten T, Caglar K, Oguz Y, Vural A, Cakar M, Altun B, Yenicesu M, Carrero JJ. Vascular health, systemic inflammation and progressive reduction in kidney function; clinical determinants and impact on cardiovascular outcomes. Nephrol Dial Transplant. 2011;26:3537–3543.
    1. Celermajer DS, Sorensen K, Ryalls M, Robinson J, Thomas O, Leonard JV, Deanfield JE. Impaired endothelial function occurs in the systemic arteries of children with homozygous homocystinuria but not in their heterozygous parents. J Am Coll Cardiol. 1993;22:854–858.
    1. Jalal DI, Decker E, Perrenoud L, Nowak KL, Bispham N, Mehta T, Smits G, You Z, Seals D, Chonchol M, Johnson RJ. Vascular function and uric acid‐lowering in stage 3 CKD. J Am Soc Nephrol. 2017;28:943–952.
    1. Jalal DI, Jablonski KL, McFann K, Chonchol MB, Seals DR. Vascular endothelial function is not related to serum uric acid in healthy adults. Am J Hypertens. 2012;25:407–413.
    1. DiLillo DJ, Pawluczkowycz AW, Peng W, Kennedy AD, Beum PV, Lindorfer MA, Taylor RP. Selective and efficient inhibition of the alternative pathway of complement by a mAb that recognizes C3b/iC3b. Mol Immunol. 2006;43:1010–1019.
    1. Ogrodowski JL, Hebert LA, Sedmak D, Cosio FG, Tamerius J, Kolb W. Measurement of SC5b‐9 in urine in patients with the nephrotic syndrome. Kidney Int. 1991;40:1141–1147.
    1. Morita Y, Ikeguchi H, Nakamura J, Hotta N, Yuzawa Y, Matsuo S. Complement activation products in the urine from proteinuric patients. J Am Soc Nephrol. 2000;11:700–707.
    1. Thurman JM, Kraus DM, Girardi G, Hourcade D, Kang HJ, Royer PA, Mitchell LM, Giclas PC, Salmon J, Gilkeson G, Holers VM. A novel inhibitor of the alternative complement pathway prevents antiphospholipid antibody‐induced pregnancy loss in mice. Mol Immunol. 2005;42:87–97.
    1. Einfinger K, Badrnya S, Furtmuller M, Handschuh D, Lindner H, Geiger M. Phospholipid binding protein C inhibitor (PCI) is present on microparticles generated in vitro and in vivo. PLoS One. 2015;10:e0143137.
    1. White RT, Damm D, Hancock N, Rosen BS, Lowell BB, Usher P, Flier JS, Spiegelman BM. Human adipsin is identical to complement factor‐D and is expressed at high‐levels in adipose‐tissue. J Biol Chem. 1992;267:9210–9213.
    1. Volanakis JE, Barnum SR, Giddens M, Galla JH. Renal filtration and catabolism of complement protein D. N Engl J Med. 1985;312:395–399.
    1. Hertle E, Arts IC, van der Kallen CJ, Feskens EJ, Schalkwijk CG, Stehouwer CD, van Greevenbroek MM. The alternative complement pathway is longitudinally associated with adverse cardiovascular outcomes. The CODAM study. Thromb Haemost. 2016;115:446–457.
    1. Thurman JM, Le Quintrec M. Targeting the complement cascade: novel treatments coming down the pike. Kidney Int. 2016;90:746–752.
    1. Thurman JM, Holers VM. The central role of the alternative complement pathway in human disease. J Immunol. 2006;176:1305–1310.
    1. Yuan X, Gavriilaki E, Thanassi JA, Yang G, Baines AC, Podos SD, Huang Y, Huang M, Brodsky RA. Small‐molecule factor D inhibitors selectively block the alternative pathway of complement in paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome. Haematologica. 2017;102:466–475.
    1. Kavanagh D, Goodship TH, Richards A. Atypical hemolytic uremic syndrome. Semin Nephrol. 2013;33:508–530.
    1. Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Invest. 2016;126:1152–1162.

Source: PubMed

3
購読する