NLRP3 inflammasome expression in peripheral blood monocytes of coronary heart disease patients and its modulation by rosuvastatin

Jian Zhu, Shili Wu, Sigan Hu, Hui Li, Miaonan Li, Xu Geng, Hongju Wang, Jian Zhu, Shili Wu, Sigan Hu, Hui Li, Miaonan Li, Xu Geng, Hongju Wang

Abstract

Nucleotide‑binding oligomerization domain, leucine rich repeat, and pyrin domain‑containing protein 3 (NLRP3) inflammasome has been implicated in a series of physiological and pathological processes. However, its correlation in coronary heart disease (CHD) still remains to be elucidated. The present study aimed to determine the expression of NLRP3 inflammasome in peripheral blood monocytes (PBMCs) of stable angina pectoris (SAP) and acute myocardial infarction (AMI) patients. In addition, the effect of rosuvastatin on their activities was analyzed in vitro. A total of 60 participants with SAP (n=20), AMI (n=20) and non‑CHD controls (n=20) were enrolled. Fluorescence‑activated cell sorting, real‑time PCR, western blotting and enzyme‑linked immunosorbent assay were performed to reveal the role of NLRP3 inflammasome. NLRP3 inflammasome was expressed in the PBMCs, and revealed an increased expression along the downstream interleukin (IL)‑1β and IL‑18 in both SAP and AMI groups, compared to the control group. Moreover, there was a more marked increase in the expression of these indicators in AMI patients when compared to SAP patients. Interference with rosuvastatin in vitro revealed that the expression of NLRP3 inflammasome and its downstream cytokines were significantly downregulated in both SAP and AMI groups in a time‑dependent manner. The activation of NLRP3 inflammasome may be involved in the development of CHD, and rosuvastatin could attenuate the inflammatory process of atherosclerosis by downregulating NLRP3 expression and its downstream mediators. These findings indicated a potential role of NLRP3 in the pathogenesis and management of CHD, and also provided new insights into the mechanistic framework of rosuvastatin activity.

Figures

Figure 1.
Figure 1.
Upregulation of NLRP3 inflammasome mRNA levels in PBMCs in patients with SAP and AMI, compared with non-CHD controls. (A) PBMCs were isolated from peripheral blood and the positive rate of CD14 was calculated to be ≥95% of PBMCs by flow cytometry. RT-PCR assays were performed to quantify the mRNA levels of (B) NLRP3, (C) ASC and (D) caspase-1 in PBMCs of each group. *P

Figure 2.

Increase in protein expression levels…

Figure 2.

Increase in protein expression levels of NLRP3 inflammasome and secretion of its downstream…

Figure 2.
Increase in protein expression levels of NLRP3 inflammasome and secretion of its downstream interleukins in PBMCs of patients with SAP and AMI, compared with non-CHD controls. (A) Western blotting was used to detect the protein expression of NLRP3, ASC and caspase-1 in PBMCs of each group. (B) Quantitative results are illustrated for the three indicators using western blot analysis. (C) Plasma levels of IL-1β and IL-18 from each group were measured by ELISA. *P

Figure 3.

Effect of rosuvastatin on NLRP3…

Figure 3.

Effect of rosuvastatin on NLRP3 inflammasome mRNA levels in PBMCs in vitro .…

Figure 3.
Effect of rosuvastatin on NLRP3 inflammasome mRNA levels in PBMCs in vitro. PBMCs were isolated from the peripheral blood of patients with non-CHD, SAP and AMI, respectively, followed by proliferation and treatment with rosuvastatin at a concentration of 20 µM for the indicated time-points. RT-PCR assay was performed to investigate the influence of rosuvastatin on the mRNA levels of (A) NLRP3, (B) ASC and (C) caspase-1 in PBMCs of each group. *P<0.05; **P<0.01. NLRP3, nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3; PBMCs, peripheral blood monocytes; CHD, coronary heart disease; SAP, stable angina pectoris; AMI, acute myocardial infarction; ASC, apoptosis-associated speck-like protein containing a CARD; NS, not significant.

Figure 4.

Effect of rosuvastatin on protein…

Figure 4.

Effect of rosuvastatin on protein expression levels of NLRP3 inflammasome in PBMCs and…

Figure 4.
Effect of rosuvastatin on protein expression levels of NLRP3 inflammasome in PBMCs and secretion of its downstream cytokines in supernatants in vitro. PBMCs were isolated from the peripheral blood of patients with non-CHD, SAP and AMI, respectively, followed by proliferation and treatment with rosuvastatin at a concentration of 20 µM for the indicated time-points. (A) Western blotting was conducted to analyze the influence of rosuvastatin on the protein levels of NLRP3, ASC and caspase-1 in PBMCs of each group. (B) Quantitative results for modulating the protein expression of the aforementioned three indicators by rosuvastatin are illustrated. (C) Modulation of IL-1β and IL-18 secretion in the supernatants by rosuvastatin of each group was measured by ELISA. *P<0.05, **P<0.01 and #P<0.05 (compared to the 0-h group); ΔP<0.05 (compared to the 12-h group). NLRP3, nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3; PBMCs, peripheral blood monocytes; CHD, coronary heart disease; SAP, stable angina pectoris; AMI, acute myocardial infarction; ASC, apoptosis-associated speck-like protein containing a CARD; NS, not significant.

Figure 5.

Proposed model for NLRP3 inflammasome…

Figure 5.

Proposed model for NLRP3 inflammasome signaling pathway involved in inflammatory pathogenesis of coronary…

Figure 5.
Proposed model for NLRP3 inflammasome signaling pathway involved in inflammatory pathogenesis of coronary atherosclerosis. Stimulated PBMCs orchestrate activation of NLRP3 inflammasome due to the upregulation of the expression of NLRP3, ASC, caspase-1, and their downstream mediators, leading to atherosclerotic progression. NLRP3, nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3; PBMCs, peripheral blood monocytes; ASC, apoptosis-associated speck-like protein containing a CARD.
Similar articles
Cited by
References
    1. Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, et al. Heart disease and stroke statistics-2017 update: A report from the American heart association. Circulation. 2017;135:e146–e603. doi: 10.1161/CIR.0000000000000485. - DOI - PMC - PubMed
    1. Welsh P, Grassia G, Botha S, Sattar N, Maffia P. Targeting inflammation to reduce cardiovascular disease risk: A realistic clinical prospect? Br J Pharmacol. 2017;174:3898–3913. doi: 10.1111/bph.13818. - DOI - PMC - PubMed
    1. Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–1131. doi: 10.1056/NEJMoa1707914. - DOI - PubMed
    1. Ridker PM, Hennekens CH, Buring JE, Rifai N. C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N Engl J Med. 2000;342:836–843. doi: 10.1056/NEJM200003233421202. - DOI - PubMed
    1. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nuñez G, Schnurr M, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464:1357–1361. doi: 10.1038/nature08938. - DOI - PMC - PubMed
Show all 43 references
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 2.
Figure 2.
Increase in protein expression levels of NLRP3 inflammasome and secretion of its downstream interleukins in PBMCs of patients with SAP and AMI, compared with non-CHD controls. (A) Western blotting was used to detect the protein expression of NLRP3, ASC and caspase-1 in PBMCs of each group. (B) Quantitative results are illustrated for the three indicators using western blot analysis. (C) Plasma levels of IL-1β and IL-18 from each group were measured by ELISA. *P

Figure 3.

Effect of rosuvastatin on NLRP3…

Figure 3.

Effect of rosuvastatin on NLRP3 inflammasome mRNA levels in PBMCs in vitro .…

Figure 3.
Effect of rosuvastatin on NLRP3 inflammasome mRNA levels in PBMCs in vitro. PBMCs were isolated from the peripheral blood of patients with non-CHD, SAP and AMI, respectively, followed by proliferation and treatment with rosuvastatin at a concentration of 20 µM for the indicated time-points. RT-PCR assay was performed to investigate the influence of rosuvastatin on the mRNA levels of (A) NLRP3, (B) ASC and (C) caspase-1 in PBMCs of each group. *P<0.05; **P<0.01. NLRP3, nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3; PBMCs, peripheral blood monocytes; CHD, coronary heart disease; SAP, stable angina pectoris; AMI, acute myocardial infarction; ASC, apoptosis-associated speck-like protein containing a CARD; NS, not significant.

Figure 4.

Effect of rosuvastatin on protein…

Figure 4.

Effect of rosuvastatin on protein expression levels of NLRP3 inflammasome in PBMCs and…

Figure 4.
Effect of rosuvastatin on protein expression levels of NLRP3 inflammasome in PBMCs and secretion of its downstream cytokines in supernatants in vitro. PBMCs were isolated from the peripheral blood of patients with non-CHD, SAP and AMI, respectively, followed by proliferation and treatment with rosuvastatin at a concentration of 20 µM for the indicated time-points. (A) Western blotting was conducted to analyze the influence of rosuvastatin on the protein levels of NLRP3, ASC and caspase-1 in PBMCs of each group. (B) Quantitative results for modulating the protein expression of the aforementioned three indicators by rosuvastatin are illustrated. (C) Modulation of IL-1β and IL-18 secretion in the supernatants by rosuvastatin of each group was measured by ELISA. *P<0.05, **P<0.01 and #P<0.05 (compared to the 0-h group); ΔP<0.05 (compared to the 12-h group). NLRP3, nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3; PBMCs, peripheral blood monocytes; CHD, coronary heart disease; SAP, stable angina pectoris; AMI, acute myocardial infarction; ASC, apoptosis-associated speck-like protein containing a CARD; NS, not significant.

Figure 5.

Proposed model for NLRP3 inflammasome…

Figure 5.

Proposed model for NLRP3 inflammasome signaling pathway involved in inflammatory pathogenesis of coronary…

Figure 5.
Proposed model for NLRP3 inflammasome signaling pathway involved in inflammatory pathogenesis of coronary atherosclerosis. Stimulated PBMCs orchestrate activation of NLRP3 inflammasome due to the upregulation of the expression of NLRP3, ASC, caspase-1, and their downstream mediators, leading to atherosclerotic progression. NLRP3, nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3; PBMCs, peripheral blood monocytes; ASC, apoptosis-associated speck-like protein containing a CARD.
Figure 3.
Figure 3.
Effect of rosuvastatin on NLRP3 inflammasome mRNA levels in PBMCs in vitro. PBMCs were isolated from the peripheral blood of patients with non-CHD, SAP and AMI, respectively, followed by proliferation and treatment with rosuvastatin at a concentration of 20 µM for the indicated time-points. RT-PCR assay was performed to investigate the influence of rosuvastatin on the mRNA levels of (A) NLRP3, (B) ASC and (C) caspase-1 in PBMCs of each group. *P<0.05; **P<0.01. NLRP3, nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3; PBMCs, peripheral blood monocytes; CHD, coronary heart disease; SAP, stable angina pectoris; AMI, acute myocardial infarction; ASC, apoptosis-associated speck-like protein containing a CARD; NS, not significant.
Figure 4.
Figure 4.
Effect of rosuvastatin on protein expression levels of NLRP3 inflammasome in PBMCs and secretion of its downstream cytokines in supernatants in vitro. PBMCs were isolated from the peripheral blood of patients with non-CHD, SAP and AMI, respectively, followed by proliferation and treatment with rosuvastatin at a concentration of 20 µM for the indicated time-points. (A) Western blotting was conducted to analyze the influence of rosuvastatin on the protein levels of NLRP3, ASC and caspase-1 in PBMCs of each group. (B) Quantitative results for modulating the protein expression of the aforementioned three indicators by rosuvastatin are illustrated. (C) Modulation of IL-1β and IL-18 secretion in the supernatants by rosuvastatin of each group was measured by ELISA. *P<0.05, **P<0.01 and #P<0.05 (compared to the 0-h group); ΔP<0.05 (compared to the 12-h group). NLRP3, nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3; PBMCs, peripheral blood monocytes; CHD, coronary heart disease; SAP, stable angina pectoris; AMI, acute myocardial infarction; ASC, apoptosis-associated speck-like protein containing a CARD; NS, not significant.
Figure 5.
Figure 5.
Proposed model for NLRP3 inflammasome signaling pathway involved in inflammatory pathogenesis of coronary atherosclerosis. Stimulated PBMCs orchestrate activation of NLRP3 inflammasome due to the upregulation of the expression of NLRP3, ASC, caspase-1, and their downstream mediators, leading to atherosclerotic progression. NLRP3, nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3; PBMCs, peripheral blood monocytes; ASC, apoptosis-associated speck-like protein containing a CARD.

References

    1. Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, et al. Heart disease and stroke statistics-2017 update: A report from the American heart association. Circulation. 2017;135:e146–e603. doi: 10.1161/CIR.0000000000000485.
    1. Welsh P, Grassia G, Botha S, Sattar N, Maffia P. Targeting inflammation to reduce cardiovascular disease risk: A realistic clinical prospect? Br J Pharmacol. 2017;174:3898–3913. doi: 10.1111/bph.13818.
    1. Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–1131. doi: 10.1056/NEJMoa1707914.
    1. Ridker PM, Hennekens CH, Buring JE, Rifai N. C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N Engl J Med. 2000;342:836–843. doi: 10.1056/NEJM200003233421202.
    1. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nuñez G, Schnurr M, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464:1357–1361. doi: 10.1038/nature08938.
    1. Kingsbury SR, Conaghan PG, McDermott MF. The role of the NLRP3 inflammasome in gout. J Inflamm Res. 2011;4:39–49.
    1. Toldo S, Mezzaroma E, Mauro AG, Salloum F, Van Tassell BW, Abbate A. The inflammasome in myocardial injury and cardiac remodeling. Antioxid Redox Signal. 2015;22:1146–1161. doi: 10.1089/ars.2014.5989.
    1. Satoh M, Tabuchi T, Itoh T, Nakamura M. NLRP3 inflammasome activation in coronary artery disease: Results from prospective and randomized study of treatment with atorvastatin or rosuvastatin. Clin Sci (Lond) 2014;126:233–241. doi: 10.1042/CS20130043.
    1. Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome: A sensor for metabolic danger? Science. 2010;327:296–300. doi: 10.1126/science.1184003.
    1. Rajamäki K, Lappalainen J, Oörni K, Välimäki E, Matikainen S, Kovanen PT, Eklund KK. Cholesterol crystals activate the NLRP3 inflammasome in human macrophages: A novel link between cholesterol metabolism and inflammation. PLoS One. 2010;5:e11765. doi: 10.1371/journal.pone.0011765.
    1. No authors listed, corp-author. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: The scandinavian simvastatin survival study (4S) Lancet. 1994;344:1383–1389.
    1. Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Jr, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG, et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359:2195–2207. doi: 10.1056/NEJMoa0807646.
    1. Schwartz GG, Olsson AG, Ezekowitz MD, Ganz P, Oliver MF, Waters D, Zeiher A, Chaitman BR, Leslie S, Stern T, Myocardial Ischemia Reduction with Aggressive Cholesterol Lowering (MIRACL) Study Investigators Effects of atorvastatin on early recurrent ischemic events in acute coronary syndromes: The MIRACL study: A randomized controlled trial. JAMA. 2001;285:1711–1718. doi: 10.1001/jama.285.13.1711.
    1. Zacà V, Rastogi S, Imai M, Wang M, Sharov VG, Jiang A, Goldstein S, Sabbah HN. Chronic monotherapy with rosuvastatin prevents progressive left ventricular dysfunction and remodeling in dogs with heart failure. J Am Coll Cardiol. 2007;50:551–557. doi: 10.1016/j.jacc.2007.04.050.
    1. Ren Y, Zhu H, Fan Z, Gao Y, Tian N. Comparison of the effect of rosuvastatin versus rosuvastatin/ezetimibe on markers of inflammation in patients with acute myocardial infarction. Exp Ther Med. 2017;14:4942–4950.
    1. Karlson BW, Nicholls SJ, Lundman P, Barter PJ, Palmer MK. Modeling statin-induced reductions of cardiovascular events in primary prevention: A VOYAGER meta-Analysis. Cardiology. 2018;140:30–34. doi: 10.1159/000488311.
    1. Fihn SD, Gardin JM, Abrams J, Berra K, Blankenship JC, Dallas AP, Douglas PS, Foody JM, Gerber TC, Hinderliter AL, et al. 2012 ACCF/AHA/ACP/AATS/PCNA/SCAI/STS guideline for the diagnosis and management of patients with stable ischemic heart disease: Executive summary: A report of the American college of cardiology foundation/American heart association task force on practice guidelines, and the American College of physicians, American association for thoracic surgery, preventive cardiovascular nurses association, society for cardiovascular angiography and interventions, and society of thoracic surgeons. Circulation. 2012;126:3097–3137. doi: 10.1161/CIR.0b013e318277d6a0.
    1. Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, White HD, Joint ESC/ACCF/AHA/WHF Task Force for the Universal Definition of Myocardial Infarction. Katus HA, Lindahl B, Morrow DA, et al. Third universal definition of myocardial infarction. Circulation. 2012;126:2020–2035. doi: 10.1161/CIR.0b013e31826e1058.
    1. Giampietro C, Lionetti MC, Costantini G, Mutti F, Zapperi S, La Porta CA. Cholesterol impairment contributes to neuroserpin aggregation. Sci Rep. 2017;7:43669. doi: 10.1038/srep43669.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262.
    1. Marchant DJ, Boyd JH, Lin DC, Granville DJ, Garmaroudi FS, McManus BM. Inflammation in myocardial diseases. Circ Res. 2012;110:126–144. doi: 10.1161/CIRCRESAHA.111.243170.
    1. Weber C, Soehnlein O. ApoE controls the interface linking lipids and inflammation in atherosclerosis. J Clin Invest. 2011;121:3825–3827. doi: 10.1172/JCI60457.
    1. Husain K, Hernandez W, Ansari RA, Ferder L. Inflammation, oxidative stress and renin angiotensin system in atherosclerosis. World J Biol Chem. 2015;6:209–217. doi: 10.4331/wjbc.v6.i3.209.
    1. Guo H, Callaway JB, Ting JP. Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21:677–687. doi: 10.1038/nm.3893.
    1. Martinon F, Burns K, Tschopp J. The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10:417–426. doi: 10.1016/S1097-2765(02)00599-3.
    1. Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014;157:1013–1022. doi: 10.1016/j.cell.2014.04.007.
    1. Muñoz-Planillo R, Kuffa P, Martinez-Colón G, Smith BL, Rajendiran TM, Núñez G. K+ efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity. 2013;38:1142–1153. doi: 10.1016/j.immuni.2013.05.016.
    1. Alfonso-Loeches S, Ureña-Peralta JR, Morillo-Bargues MJ, Oliver-De La Cruz J, Guerri C. Role of mitochondria ROS generation in ethanol-induced NLRP3 inflammasome activation and cell death in astroglial cells. Front Cell Neurosci. 2014;8:216. doi: 10.3389/fncel.2014.00216.
    1. Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA, Latz E. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008;9:847–856. doi: 10.1038/ni.1631.
    1. Toldo S, Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol. 2018;15:203–214. doi: 10.1038/nrcardio.2017.161.
    1. Galea J, Armstrong J, Gadsdon P, Holden H, Francis SE, Holt CM. Interleukin-1 beta in coronary arteries of patients with ischemic heart disease. Arterioscler Thromb Vasc Biol. 1996;16:1000–1006. doi: 10.1161/01.ATV.16.8.1000.
    1. Blankenberg S, Tiret L, Bickel C, Peetz D, Cambien F, Meyer J, Rupprecht HJ, AtheroGene Investigators Interleukin-18 is a strong predictor of cardiovascular death in stable and unstable angina. Circulation. 2002;106:24–30. doi: 10.1161/01.CIR.0000020546.30940.92.
    1. Takahashi M. NLRP3 inflammasome as a novel player in myocardial infarction. Int Heart J. 2014;55:101–105. doi: 10.1536/ihj.13-388.
    1. Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, Izawa A, Takahashi Y, Masumoto J, Koyama J, et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 2011;123:594–604. doi: 10.1161/CIRCULATIONAHA.110.982777.
    1. Toldo S, Marchetti C, Mauro AG, Chojnacki J, Mezzaroma E, Carbone S, Zhang S, Van Tassell B, Salloum FN, Abbate A. Inhibition of the NLRP3 inflammasome limits the inflammatory injury following myocardial ischemia-reperfusion in the mouse. Int J Cardiol. 2016;209:215–220. doi: 10.1016/j.ijcard.2016.02.043.
    1. Bullón P, Cano-García FJ, Alcocer-Gómez E, Varela-López A, Roman-Malo L, Ruiz-Salmerón RJ, Quiles JL, Navarro-Pando JM, Battino M, Ruiz-Cabello J, et al. Could NLRP3-inflammasome be a cardiovascular risk biomarker in acute myocardial infarction patients? Antioxid Redox Signal. 2017;27:269–275. doi: 10.1089/ars.2016.6970.
    1. Verma S, Buchanan MR, Anderson TJ. Endothelial function testing as a biomarker of vascular disease. Circulation. 2003;108:2054–2059. doi: 10.1161/01.CIR.0000089191.72957.ED.
    1. Szmitko PE, Wang CH, Weisel RD, de Almeida JR, Anderson TJ, Verma S. New markers of inflammation and endothelial cell activation: Part I. Circulation. 2003;108:1917–1923. doi: 10.1161/01.CIR.0000089190.95415.9F.
    1. Yip HK, Wu CJ, Hang CL, Chang HW, Yang CH, Hsieh YK, Fang CY, Fu M, Yeh KH, Chen MC. Levels and values of inflammatory markers in patients with angina pectoris. Int Heart J. 2005;46:571–581. doi: 10.1536/ihj.46.571.
    1. Fiechter M, Ghadri JR, Jaguszewski M, Siddique A, Vogt S, Haller RB, Halioua R, Handzic A, Kaufmann PA, Corti R, et al. Impact of inflammation on adverse cardiovascular events in patients with acute coronary syndromes. J Cardiovasc Med (Hagerstown) 2013;14:807–814. doi: 10.2459/JCM.0b013e3283609350.
    1. Ference BA, Ginsberg HN, Graham I, Ray KK, Packard CJ, Bruckert E, Hegele RA, Krauss RM, Raal FJ, Schunkert H, et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European atherosclerosis society consensus panel. Eur Heart J. 2017;38:2459–2472. doi: 10.1093/eurheartj/ehx144.
    1. Tousoulis D, Andreou I, Tsiatas M, Miliou A, Tentolouris C, Siasos G, Papageorgiou N, Papadimitriou CA, Dimopoulos MA, Stefanadis C. Effects of rosuvastatin and allopurinol on circulating endothelial progenitor cells in patients with congestive heart failure: The impact of inflammatory process and oxidative stress. Atherosclerosis. 2011;214:151–157. doi: 10.1016/j.atherosclerosis.2010.11.002.
    1. Altaf A, Qu P, Zhao Y, Wang H, Lou D, Niu N. NLRP3 inflammasome in peripheral blood monocytes of acute coronary syndrome patients and its relationship with statins. Coron Artery Dis. 2015;26:409–421. doi: 10.1097/MCA.0000000000000255.

Source: PubMed

3
購読する