Deleterious effects of phosphate on vascular and endothelial function via disruption to the nitric oxide pathway

Kathryn K Stevens, Laura Denby, Rajan K Patel, Patrick B Mark, Sarah Kettlewell, Godfrey L Smith, Marc J Clancy, Christian Delles, Alan G Jardine, Kathryn K Stevens, Laura Denby, Rajan K Patel, Patrick B Mark, Sarah Kettlewell, Godfrey L Smith, Marc J Clancy, Christian Delles, Alan G Jardine

Abstract

Background: Hyperphosphataemia is an independent risk factor for accelerated cardiovascular disease in chronic kidney disease (CKD), although the mechanism for this is poorly understood. We investigated the effects of sustained exposure to a high-phosphate environment on endothelial function in cellular and preclinical models, as well as in human subjects.

Methods: Resistance vessels from rats and humans (± CKD) were incubated in a normal (1.18 mM) or high (2.5 mM) phosphate concentration solution and cells were cultured in normal- (0.5 mM) or high-phosphate (3 mM) concentration media. A single-blind crossover study was performed in healthy volunteers, receiving phosphate supplements or a phosphate binder (lanthanum), and endothelial function measured was by flow-mediated dilatation.

Results: Endothelium-dependent vasodilatation was impaired when resistance vessels were exposed to high phosphate; this could be reversed in the presence of a phosphodiesterase-5-inhibitor. Vessels from patients with CKD relaxed normally when incubated in normal-phosphate conditions, suggesting that the detrimental effects of phosphate may be reversible. Exposure to high-phosphate disrupted the whole nitric oxide pathway with reduced nitric oxide and cyclic guanosine monophosphate production and total and phospho endothelial nitric oxide synthase expression. In humans, endothelial function was reduced by chronic phosphate loading independent of serum phosphate, but was associated with higher urinary phosphate excretion and serum fibroblast growth factor 23.

Conclusions: These directly detrimental effects of phosphate, independent of other factors in the uraemic environment, may explain the increased cardiovascular risk associated with phosphate in CKD.

Keywords: cardiovascular risk; chronic kidney disease; endothelial function; nitric oxide; phosphate.

© The Author 2016. Published by Oxford University Press on behalf of ERA-EDTA. All rights reserved.

Figures

FIGURE 1
FIGURE 1
Rat mesenteric vessels incubated in a high-phosphate concentration (2.5 mM) solution have impaired endothelium-dependent and -independent vasodilatation, an effect that can be reversed when the vessels are co-incubated with a phosphodiesterase-5 inhibitor (PDE5I), zaprinast. Vessels were incubated for 16 h in either a normal (1.18 mM) or high (2.5 mM) phosphate concentration solution and all responses are expressed as mean ± SEM. (A and C) Vasodilatation to increasing concentrations of carbachol or SNP, respectively, expressed as a % of maximal contraction with PEP 1 × 10−5 M. (B) Contraction with PEP in the presence and absence of L-NAME. (D and E) Vasodilatation to increasing concentrations of carbachol or SNP, respectively, in the presence of zaprinast (PDE5I) expressed as a % of maximal contraction with PEP 1 × 10−5 M. n = 10–13 vessels (see Table 1).
FIGURE 2
FIGURE 2
Human resistance vessels from patients with (C) and without (A) CKD incubated in a high-phosphate concentration (2.5 mM) solution have impaired endothelium-dependent vasodilatation. Endothelium-independent vasodilatation is impaired in vessels from humans without CKD (B) but preserved in those with CKD (D). Vessels were incubated for 16 h in either a normal (1.18 mM) or high (2.5 mM) phosphate concentration solution and all responses are expressed as mean ± SEM. (A and C) Vasodilatation to increasing concentrations of carbachol expressed as a % of contraction with PEP 1 × 10−5 M in vessels from living kidney donors without CKD (A) and from patients with CKD stage 5 (C). (B and D) Vasodilatation to increasing concentrations of SNP expressed as a % of contraction with PEP 1 × 10−5 M in vessels from living kidney donors without CKD (B) and from patients with CKD stage 5 (D). n = 9–14 vessels (see Table 3).
FIGURE 3
FIGURE 3
HUVECs express reduced total (A) and phospho (B) eNOS and increased nitrotyrosine (C) when cultured in high-phosphate concentration medium (3 mM). PKG expression is reduced in rat VSMCs cultured in high- compared with normal-phosphate concentration medium. **P < 0.05. HUVECs and VSMCs were cultured in 0.5 mM or 3 mM phosphate concentration medium. Ten µg of protein was fractionated on SDS page gels. Primary antibodies for Total eNOS (1:1000), Phospho eNOS (1:200) (both Cell Signalling Technology), nitrotyrosine (1:500, R & D Systems), PKG (1:200, Enzo Life Sciences) and protein expression normalized to GAPDH. Densitometry was performed. In (A)–(D), top panels show representative gel from cells cultured in 0.5 or 3 mM phosphate concentration medium and bottom panels show GAPDH. The panels were all taken from a single gel and each band represents one well from separate six-well plates.
FIGURE 4
FIGURE 4
cGMP concentration is significantly reduced in rat vessels incubated for 24 h in a high-phosphate concentration solution (A), whereas the intracellular calcium level in HUVECs is unchanged in a high-phosphate environment (B). (A) Rat mesenteric resistance vessels were incubated in a 1.18 mM (n = 11) or 2.5 mM (n = 11) phosphate concentration solution for 24 h. Results are expressed as cGMP pM/µg protein. Analysis was by the Mann–Whitney U test. (B) Cells were plated in glass-bottomed plates and cytosolic loading of FURA-2-AM was achieved by incubating the cells with FURA-2-AM. Values are mean ± SEM and are from at least four experiments with the number of cells included indicated by (n).
FIGURE 5
FIGURE 5
Change in FMD from baseline visit measures, following intervention with lanthanum and phosphate expressed as a % change from the baseline visit.
FIGURE 6
FIGURE 6
Possible mechanism of action of phosphate as a cardiovascular risk factor causing both a pro-inflammatory state and endothelial dysfunction. Using evidence presented in this manuscript and previous studies, we propose this as the possible mechanism of action of phosphate resulting in endothelial dysfunction and a pro-inflammatory state. Additionally, we have some evidence of an effect of phosphate on cell growth (data not yet published).

References

    1. Betriu A, Martinez-Alonso M, Arcidiacono MV. et al. Prevalence of subclinical atheromatosis and associated risk factors in chronic kidney disease: the NEFRONA study. Nephrol Dial Transplant 2014; 29: 1415–1422
    1. Ganesh SK, Stack AG, Levin NW. et al. Association of elevated serum PO(4), Ca x PO(4) product, and parathyroid hormone with cardiac mortality risk in chronic hemodialysis patients. J Am Soc Nephrol 2001; 12: 2131–2138
    1. Kestenbaum B, Sampson JN, Rudser KD. et al. Serum phosphate levels and mortality risk among people with chronic kidney disease. J Am Soc Nephrol 2005; 16: 520–528
    1. Achinger SG, Ayus JC. Left ventricular hypertrophy: is hyperphosphatemia among dialysis patients a risk factor? J Am Soc Nephrol 2006; 17(12 Suppl 3): S255–S261
    1. Tonelli M, Pfeffer MA. Kidney disease and cardiovascular risk. Annu Rev Med 2007; 58: 123–139
    1. Block GA, Hulbert-Shearon TE, Levin NW. et al. Association of serum phosphorus and calcium x phosphate product with mortality risk in chronic hemodialysis patients: a national study. Am J Kidney Dis 1998; 31: 607–617
    1. Block GA, Klassen PS, Lazarus JM. et al. Mineral metabolism, mortality, and morbidity in maintenance hemodialysis. J Am Soc Nephrol 2004; 15: 2208–2218
    1. Fellstrom B, Jardine AG, Holdaas H. et al. The effects of rosuvastatin versus placebo on cardiovascular outcomes in patients with end stage renal disease on haemodialysis—results of the AURORA study. NDT Plus 2009; 2(Suppl 2): ii1514
    1. Jardine AG, Fellstrom B, Logan JO. et al. Cardiovascular risk and renal transplantation: post hoc analyses of the Assessment of Lescol in Renal Transplantation (ALERT) Study. Am J Kidney Dis 2005; 46: 529–536
    1. Foley RN, Parfrey PS, Sarnak MJ. Epidemiology of cardiovascular disease in chronic renal disease. J Am Soc Nephrol 1998; 9(12 Suppl): S16–S23
    1. Sarnak MJ, Levey AS, Schoolwerth AC. et al. 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. Hypertension 2003; 42: 1050–1065
    1. Stevens KK, Morgan IR, Patel RK. et al. Serum phosphate and outcome at one year after deceased donor renal transplantation. Clin Transplant 2011; 25: E199–E204
    1. Cirillo M, Botta G, Chiricone D. et al. Glomerular filtration rate and serum phosphate: an inverse relationship diluted by age. Nephrol Dial Transplant 2009; 24: 2123–2131
    1. Connolly GM, Cunningham R, McNamee PT. et al. Elevated serum phosphate predicts mortality in renal transplant recipients. Transplantation 2009; 87: 1040–1044
    1. Bellasi A, Mandreoli M, Baldrati L. et al. Chronic kidney disease progression and outcome according to serum phosphorus in mild-to-moderate kidney dysfunction. Clin J Am Soc Nephrol 2011; 6: 883–891
    1. Chue CD, Edwards NC, Moody WE. et al. Serum phosphate is associated with left ventricular mass in patients with chronic kidney disease: a cardiac magnetic resonance study. Heart 2012; 98: 219–224
    1. Block GA. Control of serum phosphorus: implications for coronary artery calcification and calcific uremic arteriolopathy (calciphylaxis). Curr Opin Nephrol Hypertens 2001; 10: 741–747
    1. Cozzolino M, Rizzo MA, Stucchi A. et al. Sevelamer for hyperphosphataemia in kidney failure: controversy and perspective. Ther Adv Chronic Dis 2012; 3: 59–68
    1. Yilmaz MI, Sonmez A, Saglam M. et al. Comparison of calcium acetate and sevelamer on vascular function and fibroblast growth factor 23 in CKD patients: a randomized clinical trial. Am J Kidney Dis 2012; 59: 177–185
    1. Voormolen N, Noordzij M, Grootendorst DC. et al., High plasma phosphate as a risk factor for decline in renal function and mortality in pre-dialysis patients. Nephrol Dial Transplant 2007; 22: 2909–2916
    1. Tonelli M, Pannu N, Manns B. Oral phosphate binders in patients with kidney failure. N Engl J Med 2010; 362: 1312–1324
    1. Tonelli M, Sacks F, Pfeffer M. et al. Relation between serum phosphate level and cardiovascular event rate in people with coronary disease. Circulation 2005; 112: 2627–2633
    1. Toussaint ND, Pedagogos E, Tan SJ. et al. Phosphate in early chronic kidney disease: associations with clinical outcomes and a target to reduce cardiovascular risk. Nephrology (Carlton) 2012; 17: 433–444
    1. Foley RN, Collins AJ, Herzog CA. et al. Serum phosphorus levels associate with coronary atherosclerosis in young adults. J Am Soc Nephrol 2009; 20: 397–404
    1. Zoccali C, Mallamaci F. Moderator's view: phosphate binders in chronic kidney disease patients: a clear ‘No’ at the moment, but stay tuned. Nephrol Dial Transplant 2016; 31: 196–199
    1. Giachelli CM. The emerging role of phosphate in vascular calcification. Kidney Int 2009; 75: 890–897
    1. Shanahan CM, Crouthamel MH, Kapustin A. et al. Arterial calcification in chronic kidney disease: key roles for calcium and phosphate. Circ Res 2011; 109: 697–711
    1. Van TV, Watari E, Taketani Y. et al. Dietary phosphate restriction ameliorates endothelial dysfunction in adenine-induced kidney disease rats. J Clin Biochem Nutr 2012; 51: 27–32
    1. Gutierrez OM, Mannstadt M, Isakova T. et al. Fibroblast growth factor 23 and mortality among patients undergoing hemodialysis. N Engl J Med 2008; 359: 584–592
    1. Faul C, Amaral AP, Oskouei B. et al. FGF23 induces left ventricular hypertrophy. J Clin Invest 2011; 121: 4393–4408
    1. Gutierrez OM, Januzzi JL, Isakova T. et al. Fibroblast growth factor 23 and left ventricular hypertrophy in chronic kidney disease. Circulation 2009; 119: 2545–2552
    1. Shuto E, Taketani Y, Tanaka R. et al. Dietary phosphorus acutely impairs endothelial function. J Am Soc Nephrol 2009; 20: 1504–1512
    1. Peng A, Wu T, Zeng C. et al. Adverse effects of simulated hyper- and hypo-phosphatemia on endothelial cell function and viability. PLoS One 2011; 6: e23268.
    1. Thambyrajah J, Landray MJ, McGlynn FJ. et al. Abnormalities of endothelial function in patients with predialysis renal failure. Heart 2000; 83: 205–209
    1. Annuk M, Soveri I, Zilmer M. et al. Endothelial function, CRP and oxidative stress in chronic kidney disease. J Nephrol 2005; 18: 721–726
    1. Thijssen DH, Black MA, Pyke KE. et al. Assessment of flow-mediated dilation in humans: a methodological and physiological guideline. Am J Physiol Heart Circ Physiol 2011; 300: H2–12
    1. Van Bortel LM, Duprez D, Starmans-Kool MJ. et al. Clinical applications of arterial stiffness, Task Force III: recommendations for user procedures. Am J Hypertens 2002; 15: 445–452
    1. Keswani AN, Peyton KJ, Durante W. et al. The cyclic GMP modulators YC-1 and zaprinast reduce vessel remodeling through antiproliferative and proapoptotic effects. J Cardiovasc Pharmacol Ther 2009; 14: 116–124
    1. Sebkhi A, Strange JW, Phillips SC. et al. Phosphodiesterase type 5 as a target for the treatment of hypoxia-induced pulmonary hypertension. Circulation 2003; 107: 3230–3235
    1. Morris ST, McMurray JJ, Spiers A. et al. Impaired endothelial function in isolated human uremic resistance arteries. Kidney Int 2001; 60: 1077–1082
    1. Ando R, Ueda S, Yamagishi S. et al. Involvement of advanced glycation end product-induced asymmetric dimethylarginine generation in endothelial dysfunction. Diab Vasc Dis Res 2013; 10: 436–441
    1. Brunet P, Gondouin B, Duval-Sabatier A. et al. Does uremia cause vascular dysfunction? Kidney Blood Press Res 2011; 34: 284–290
    1. Stevens KK, Patel RK, Methven S. et al. Proteinuria and outcome after renal transplantation: ratios or fractions? Transplantation 2013; 96: 65–69
    1. Burnett SM, Gunawardene SC, Bringhurst FR. et al. Regulation of C-terminal and intact FGF-23 by dietary phosphate in men and women. J Bone Miner Res 2006; 21: 1187–1196
    1. Antoniucci DM, Yamashita T, Portale AA. Dietary phosphorus regulates serum fibroblast growth factor-23 concentrations in healthy men. J Clin Endocrinol Metab 2006; 91: 3144–3149
    1. Stevens KK, McQuarrie EP, Sands W. et al. Fibroblast growth factor 23 predicts left ventricular mass and induces cell adhesion molecule formation. Int J Nephrol 2011; 2011: 297070.

Source: PubMed

3
Subscribe