Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2016, Vol. 10 Issue (3) : 320-329    https://doi.org/10.1007/s11684-016-0463-x
RESEARCH ARTICLE
p-Cresyl sulfate promotes the formation of atherosclerotic lesions and induces plaque instability by targeting vascular smooth muscle cells
Hui Han1,2,Yanjia Chen1,2,Zhengbin Zhu1,Xiuxiu Su1,Jingwei Ni1,Run Du1,Ruiyan Zhang1,2,*(),Wei Jin1,*()
1. Department of Cardiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
2. Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
 Download: PDF(459 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Coronary atherosclerosis is a major complication of chronic kidney disease. This condition contributes to the increased mortality in dialysis patients. p-Cresyl sulfate (PCS) is a prototype of protein-bound uremic toxins that cannot be efficiently removed through routine dialysis procedures. In the present study, ApoE−/− mice that underwent 5/6 nephrectomy were randomly divided into two groups, namely, vehicle-treated group (n = 20) and PCS-treated group (n = 20). Mice were sacrificed for en face and immunohistological analyses after 8 or 24 weeks of high-fat diet. Rat aortic vascular smooth muscle cells (VSMCs) were treated with phosphate buffer solution or 500 µmol/L PCS for in vitro evaluation. PCS-treated mice were observed to suffer increased atherosclerotic lesions after eight weeks of PCS administration. Moreover, 24 weeks of PCS administration also markedly increased the vulnerability index of aortic plaques. PCS was also observed to facilitate the migration and proliferation of VSMCs during the progression of the disease. Moreover, PCS disturbed the balance between matrix metalloproteinases and tissue inhibitor of metalloproteinases within the plaques. Thus, PCS played a vital role in promoting atherogenesis and disturbing the stability of formed plaques probably by targeting VSMCs.

Keywords p-cresyl sulfate      atherosclerosis      plaque stability      vascular smooth muscle cell     
Corresponding Author(s): Ruiyan Zhang,Wei Jin   
Just Accepted Date: 19 July 2016   Online First Date: 12 August 2016    Issue Date: 30 August 2016
 Cite this article:   
Hui Han,Yanjia Chen,Zhengbin Zhu, et al. p-Cresyl sulfate promotes the formation of atherosclerotic lesions and induces plaque instability by targeting vascular smooth muscle cells[J]. Front. Med., 2016, 10(3): 320-329.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-016-0463-x
https://academic.hep.com.cn/fmd/EN/Y2016/V10/I3/320
Variables 8 weeks 24 weeks
Vehicle mice PCS mice Vehicle mice PCS mice
Body weight (g) 27.50±0.67 27.10±0.67 37.30±0.76 36.75±0.70
TG (mmol/L) 1.43±0.20 2.07±0.35 1.37±0.12 2.27±0.34
TC (mmol/L) 3.35±1.33 3.23±0.15 4.00±0.12 4.17±0.15
HDL-C (mmol/L) 1.70±0.11 1.75±0.97 1.54±0.16 1.62±0.19
LDL-C (mmol/L) 2.39±0.14 2.37±0.06 2.84±0.82 2.93±0.24
SCr (µmol/L) 66.00±8.11 72.33±8.58 75.67±6.44 86.17±7.98
PCS (µg/ml) 0.68 (0.63–0.74) 5.54 (4.29–6.94)# 0.72 (0.66–0.79) 5.83 (4.35–7.40)#
Tab.1  Characteristics of the animal models
Fig.1  Mice treated with p-cresyl sulfate (PCS) showed increased atherosclerotic plaque areas after eight weeks of PCS administration. (A) Representative aortic images of en face staining with oil red O in vehicle-treated (vehicle) and PCS-treated (PCS) mice. (B) Representative sectional images of aortic roots staining with H&E and oil red O in vehicle and PCS mice (bar= 200 µm). (C and D) Quantitative analysis of lesion areas for en face and cross-sectional staining. Data are presented as mean±SEM (n = 6–8). # P<0.05 compared with vehicle.
Fig.2  Mice treated with PCS show altered plaque contents and increased plaque vulnerability index after 24 weeks of treatment. (A) Representative sectional images of aortic roots stained with oil red O, MOMA-2, α-SMA, and Sirius red in vehicle-treated (vehicle) and PCS-treated (PCS) mice (bar= 100 µm). (B) Quantitative analysis of positively stained areas for oil red O, MOMA-2, α-SMA, and Sirius red in vehicle and PCS mice. (C) Quantification of vulnerability index in vehicle and PCS mice. Data are presented as mean±SEM (n = 6–8). # P<0.05 compared with vehicle. MOMA-2, monocyte/macrophage; α-SMA, α-smooth muscle actin.
Fig.3  PCS treatment promotes the migration and proliferation of VSMCs. (A) The number of cells that migrated through the pores was determined in high-power fields for each group. (B) PCNA expression in mice treated with vehicle and PCS (bar= 100 µm; blue, DAPI-labeled cell nuclei; green, PCNA; red, α-actin). (C) Quantitative analysis of migrated cells through the pores. (D) Quantitative analysis of PCNA-positive cells in the sections of aortic roots. (E) Colorimetric assay of the absorbance at 450 nm for each group. Data are presented as mean±SEM (n = 6). # P<0.05 compared with vehicle. PCNA, proliferating cell nuclear antigen; DAPI, 4′,6-diamidino-2-phenylindole.
Fig.4  PCS treatment disturbs the balance between MMPs and TIMPs. (A) Representative blots of aortic samples in vehicle-treated (vehicle) and PCS-treated (PCS) mice. (B) Representative sectional images of aortic roots staining with MMP-2 and MMP-9 in vehicle and PCS mice after 24 weeks of treatment (bar= 200 µm). (C) Representative gelatin zymography of aortic samples in vehicle and PCS mice. (D) Quantitative analysis of blots of aortic samples in vehicle and PCS mice. (E and F) Quantitative analysis of positively stained areas for MMP-2 and MMP-9 in vehicle and PCS mice. (G and H) Quantitative analysis of gelatin zymography of aortic samples in vehicle and PCS mice. Data are presented as mean±SEM (n = 6). # P<0.05 compared with vehicle. MMP, matrix metalloproteinase; TIMP, tissue inhibitor of metalloproteinase; VSMC, vascular smooth muscle cell.
Fig.5  PCS treatment facilitates the apoptosis of VSMCs in vitro. (A and C) Evaluation of apoptotic incidence by flow cytometry analysis via double staining with annexin V-FITC and PI. (B) Representative blots showing the expression of Bax and Bcl-2. (D) Quantitative analysis of blots in VSMCs. Data are presented as mean±SEM (n = 6). # P<0.05 compared with vehicle. Annexin V-FITC, fluorescein isothiocyanate-labeled human recombinant annexin V; PI, propidium iodide; Bcl-2, B cell lymphoma 2; Bax, Bcl-2-associated X protein.
1 Collins AJ, Foley RN, Chavers B, Gilbertson D, Herzog C, Johansen K, Kasiske B, Kutner N, Liu J, St Peter W, Guo H, Gustafson S, Heubner B, Lamb K, Li S, Li S, Peng Y, Qiu Y, Roberts T, Skeans M, Snyder J, Solid C, Thompson B, Wang C, Weinhandl E, Zaun D, Arko C, Chen SC, Daniels F, Ebben J, Frazier E, Hanzlik C, Johnson R, Sheets D, Wang X, Forrest B, Constantini E, Everson S, Eggers P, Agodoa L. US Renal Data System 2011 Annual Data Report. Am J Kidney Dis 2012; 59(1 Suppl 1): A7, e1–420
https://doi.org/10.1053/j.ajkd.2011.11.015
2 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(17): 2154–2169
https://doi.org/10.1161/01.CIR.0000095676.90936.80 pmid: 14581387
3 Formanowicz D, Wanic-Kossowska M, Pawliczak E, Radom M, Formanowicz P. Usefulness of serum interleukin-18 in predicting cardiovascular mortality in patients with chronic kidney disease — systems and clinical approach. Sci Rep 2015; 5: 18332
https://doi.org/10.1038/srep18332 pmid: 26669254
4 Sirich TL, Meyer TW, Gondouin B, Brunet P, Niwa T. Protein-bound molecules: a large family with a bad character. Semin Nephrol 2014; 34(2): 106–117
https://doi.org/10.1016/j.semnephrol.2014.02.004 pmid: 24780467
5 Ito S, Yoshida M. Protein-bound uremic toxins: new culprits of cardiovascular events in chronic kidney disease patients. Toxins (Basel) 2014; 6(2): 665–678
https://doi.org/10.3390/toxins6020665 pmid: 24561478
6 Vanholder R, Schepers E, Pletinck A, Nagler EV, Glorieux G. The uremic toxicity of indoxyl sulfate and p-cresyl sulfate: a systematic review. J Am Soc Nephrol 2014; 25(9): 1897–1907
https://doi.org/10.1681/ASN.2013101062 pmid: 24812165
7 Itoh Y, Ezawa A, Kikuchi K, Tsuruta Y, Niwa T. Protein-bound uremic toxins in hemodialysis patients measured by liquid chromatography/tandem mass spectrometry and their effects on endothelial ROS production. Anal Bioanal Chem 2012; 403(7): 1841–1850
https://doi.org/10.1007/s00216-012-5929-3 pmid: 22447217
8 Miyamoto Y, Watanabe H, Noguchi T, Kotani S, Nakajima M, Kadowaki D, Otagiri M, Maruyama T. Organic anion transporters play an important role in the uptake of p-cresyl sulfate, a uremic toxin, in the kidney. Nephrol Dial Transplant 2011; 26(8): 2498–2502
https://doi.org/10.1093/ndt/gfq785 pmid: 21303967
9 Wu IW, Hsu KH, Hsu HJ, Lee CC, Sun CY, Tsai CJ, Wu MS. Serum free p-cresyl sulfate levels predict cardiovascular and all-cause mortality in elderly hemodialysis patients — a prospective cohort study. Nephrol Dial Transplant 2012; 27(3): 1169–1175
https://doi.org/10.1093/ndt/gfr453 pmid: 21891772
10 Liabeuf S, Barreto DV, Barreto FC, Meert N, Glorieux G, Schepers E, Temmar M, Choukroun G, Vanholder R, Massy ZA; European Uraemic Toxin Work Group (EUTox). Free p-cresylsulphate is a predictor of mortality in patients at different stages of chronic kidney disease. Nephrol Dial Transplant 2010; 25(4): 1183–1191
https://doi.org/10.1093/ndt/gfp592 pmid: 19914995
11 Curcio A, Torella D, Indolfi C. Mechanisms of smooth muscle cell proliferation and endothelial regeneration after vascular injury and stenting: approach to therapy. Circ J 2011; 75(6): 1287–1296
12 Newby AC, Libby P, van der Wal AC. Plaque instability — the real challenge for atherosclerosis research in the next decade? Cardiovasc Res 1999; 41(2): 321–322
pmid: 10341831
13 Muteliefu G, Enomoto A, Jiang P, Takahashi M, Niwa T. Indoxyl sulphate induces oxidative stress and the expression of osteoblast-specific proteins in vascular smooth muscle cells. Nephrol Dial Transplant 2009; 24(7): 2051–2058
https://doi.org/10.1093/ndt/gfn757 pmid: 19164326
14 Ito S, Osaka M, Higuchi Y, Nishijima F, Ishii H, Yoshida M. Indoxyl sulfate induces leukocyte-endothelial interactions through up-regulation of E-selectin. J Biol Chem 2010; 285(50): 38869–38875
https://doi.org/10.1074/jbc.M110.166686 pmid: 20937831
15 Feigenbaum J, Neuberg CA. Simplified method for the preparation of aromatic sulfuric acid esters. J Am Chem Soc 1941; 63: 3529–3530
16 Shimizu RT, Blank RS, Jervis R, Lawrenz-Smith SC, Owens GK. The smooth muscle α-actin gene promoter is differentially regulated in smooth muscle versus non-smooth muscle cells. J Biol Chem 1995; 270(13): 7631–7643
https://doi.org/10.1074/jbc.270.13.7631 pmid: 7706311
17 Ni J, Zhang W, Zhu Z, Zhu J, Du R, Jing Y, Lu L, Zhang R. In vivo kinetics of the uremic toxin p-cresyl sulfate in mice with variable renal function. Ther Apher Dial 2014; 18(6): 637–642
https://doi.org/10.1111/1744-9987.12185 pmid: 25256665
18 Han H, Zhu J, Zhu Z, Ni J, Du R, Dai Y, Chen Y, Wu Z, Lu L, Zhang R. p-Cresyl sulfate aggravates cardiac dysfunction associated with chronic kidney disease by enhancing apoptosis of cardiomyocytes. J Am Heart Assoc 2015; 4(6): e001852
https://doi.org/10.1161/JAHA.115.001852 pmid: 26066032
19 Cho KY, Miyoshi H, Kuroda S, Yasuda H, Kamiyama K, Nakagawara J, Takigami M, Kondo T, Atsumi T. The phenotype of infiltrating macrophages influences arteriosclerotic plaque vulnerability in the carotid artery. J Stroke Cerebrovasc Dis 2013; 22(7): 910–918
20 Ross R. Atherosclerosis — an inflammatory disease. N Engl J Med 1999; 340(2): 115–126
https://doi.org/10.1056/NEJM199901143400207 pmid: 9887164
21 Johnson JL. Emerging regulators of vascular smooth muscle cell function in the development and progression of atherosclerosis. Cardiovasc Res 2014; 103(4): 452–460
https://doi.org/10.1093/cvr/cvu171 pmid: 25053639
22 Gittenberger-de Groot AC, DeRuiter MC, Bergwerff M, Poelmann RE. Smooth muscle cell origin and its relation to heterogeneity in development and disease. Arterioscler Thromb Vasc Biol 1999; 19(7): 1589–1594
https://doi.org/10.1161/01.ATV.19.7.1589 pmid: 10397674
23 Rudijanto A. The role of vascular smooth muscle cells on the pathogenesis of atherosclerosis. Acta Med Indones 2007; 39(2): 86–93
pmid: 17933075
24 Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res 2003; 92(8): 827–839
https://doi.org/10.1161/01.RES.0000070112.80711.3D pmid: 12730128
25 Newby AC. Matrix metalloproteinases regulate migration, proliferation, and death of vascular smooth muscle cells by degrading matrix and non-matrix substrates. Cardiovasc Res 2006; 69(3): 614–624
https://doi.org/10.1016/j.cardiores.2005.08.002 pmid: 16266693
26 Langer HF, Haubner R, Pichler BJ, Gawaz M. Radionuclide imaging: a molecular key to the atherosclerotic plaque. J Am Coll Cardiol 2008; 52(1): 1–12
https://doi.org/10.1016/j.jacc.2008.03.036 pmid: 18582628
27 Shiomi M, Ito T, Hirouchi Y, Enomoto M. Fibromuscular cap composition is important for the stability of established atherosclerotic plaques in mature WHHL rabbits treated with statins. Atherosclerosis 2001; 157(1): 75–84
https://doi.org/10.1016/S0021-9150(00)00708-5 pmid: 11427206
28 Yang JM, Dong M, Meng X, Zhao YX, Yang XY, Liu XL, Hao PP, Li JJ, Wang XP, Zhang K, Gao F, Zhao XQ, Zhang MX, Zhang Y, Zhang C. Angiotensin-(1-7) dose-dependently inhibits atherosclerotic lesion formation and enhances plaque stability by targeting vascular cells. Arterioscler Thromb Vasc Biol 2013; 33(8): 1978–1985
https://doi.org/10.1161/ATVBAHA.113.301320 pmid: 23723368
29 Schwarz U, Buzello M, Ritz E, Stein G, Raabe G, Wiest G, Mall G, Amann K. Morphology of coronary atherosclerotic lesions in patients with end-stage renal failure. Nephrol Dial Transplant 2000; 15(2): 218–223
https://doi.org/10.1093/ndt/15.2.218 pmid: 10648668
30 Rössig L, Dimmeler S, Zeiher AM. Apoptosis in the vascular wall and atherosclerosis. Basic Res Cardiol 2001; 96(1): 11–22
https://doi.org/10.1007/s003950170073 pmid: 11215528
[1] Jichun Yang, Kaiyue Jin, Jiajun Xiao, Jing Ma, Duan Ma. Endogenous tissue factor pathway inhibitor in vascular smooth muscle cells inhibits arterial thrombosis[J]. Front. Med., 2017, 11(3): 403-409.
[2] Wen YAN MD, Min FENG MD, Pei-Hua WANG MD, Dao-Wen WANG MD, . Effect of bradykinin on bradykinin-B2 receptor in rat aortic vascular smooth muscle cells and the involved signal transduction pathways[J]. Front. Med., 2010, 4(2): 225-228.
[3] Qigong LIU, Honglian ZHOU, Yan ZENG, Shan YE, Jiani LIU, Zaiying LU. Mechanism of vascular endothelial growth factor on the prevention of restenosis after angioplasty[J]. Front Med Chin, 2009, 3(2): 177-180.
[4] WANG Fei, DAI Yalei, XU Ting, XU Bo, WANG Kaifeng. Effect of IL-10 on formation of foam cell induced by ox-LDL[J]. Front. Med., 2008, 2(3): 298-302.
[5] SUN Mingxiao, WANG Yao, CHI Jiamin. Modulatory effect on dyslipidemia and anti-atherosclerotic function of in newly diagnosed type 2 diabetes patients[J]. Front. Med., 2008, 2(2): 174-177.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed