Vol 27, No 3 (2020)
Original articles — Basic science and experimental cardiology
Published online: 2018-09-13

open access

Page views 1456
Article views/downloads 702
Get Citation

Connect on Social Media

Connect on Social Media

Characteristics of circulating endothelial cells obtained from non-ST-segment elevation myocardial infarction patients with additional diastolic dysfunction of left ventricle observed in echocardiography

Pawel Burchardt123, Maura Farinacci4, Magdalena Mayer5, Klaus Luecke6, Thomas Krahn7, Jaroslaw Manczak2, Marek Slomczynski2, Jaroslaw Hiczkiewicz3, Janusz Rzezniczak2
Pubmed: 30234891
Cardiol J 2020;27(3):295-302.

Abstract

Background: Circulating endothelial cells (CEC) may be used to find new strategies for the early di­agnosis of cardiovascular diseases. The major objective of the project is to broaden knowledge of CEC biology by determining their phenotypic characteristics. The additional aim is to clarify whether on the basis of these information it is possible to identify the origin of CEC release (from various cardiovascular compartments).

Methods: Circulating endothelial cells were collected from arterial blood prior to angiography, as well as from arterial and venous blood obtained after angiography/coronary angioplasty, from 18 patients with non-ST-segment elevation myocardial infarction (NSTEMI). CECs were quantified by flow cytometry and defined as Syto16 (dye)+, CD45dim/neg, CD31+ and CD146+. The additional CD36+ was establish as a marker of endothelial cells released from small vessels of the microcirculation.

Results: The total number of CECs increased significantly after the percutaneous transluminal coronary angioplasty (PTCA) in the arterial system. Number of CECs isolated at similar time points (after invasive procedure) did not differ significantly between arteries and veins, but the number of CD36+ CECs after coronary angioplasty was significantly higher in the venous system, than in the arterial system.

Conclusions: The number of CD36+ in artery samples obtained after coronary angioplasty (PTCA) had tendency to be decreased (in comparison to the sample obtained before angiography). It was major difference between those who had PTCA performed vs. those who had not.

Article available in PDF format

View PDF Download PDF file

References

  1. Montalescot G, Sechtem U, Achenbach S, et al. 2013 ESC guidelines on the management of stable coronary artery disease: the Task Force on the management of stable coronary artery disease of the European Society of Cardiology. Eur Heart J. 2013; 34(38): 2949–3003.
  2. Burchardt P, Goździcka-Józefiak A, Siminiak T. [IGF-1--a new risk factor for coronary atherosclerosis]. Kardiol Pol. 2006; 64(11): 1297–1302.
  3. Kassab GS, Rider CA, Tang NJ, et al. Morphometry of pig coronary arterial trees. Am J Physiol. 1993; 265(1 Pt 2): H350–H365.
  4. Bugiardini R, Pozzati A, Ottani F, et al. Vasotonic angina: a spectrum of ischemic syndromes involving functional abnormalities of the epicardial and microvascular coronary circulation. J Am Coll Cardiol. 1993; 22(2): 417–425.
  5. Crea F. Angina pectoris and normal coronary arteries: cardiac syndrome X. Heart. 2004; 90(4): 457–463.
  6. Maseri A, Crea F, Kaski J, et al. Mechanisms of angina pectoris in syndrome X. J Am Coll Cardiol. 1991; 17(2): 499–506.
  7. Bugiardini R, Bairey Merz CN. Angina with "normal" coronary arteries: a changing philosophy. JAMA. 2005; 293(4): 477–484.
  8. Erdbruegger U, Haubitz M, Woywodt A. Circulating endothelial cells: a novel marker of endothelial damage. Clin Chim Acta. 2006; 373(1-2): 17–26.
  9. Bouvier CA, Gaynor E, Cintron JR, et al. Circulating endothelium as an indication of vascular injury. Thromb Diath Haemorrh. 1970; 40: 163.
  10. Woywodt A, Blann AD, Kirsch T, et al. Isolation and enumeration of circulating endothelial cells by immunomagnetic isolation: proposal of a definition and a consensus protocol. J Thromb Haemost. 2006; 4(3): 671–677.
  11. Hladovec J, Prerovský I, Stanĕk V, et al. Circulating endothelial cells in acute myocardial infarction and angina pectoris. Klin Wochenschr. 1978; 56(20): 1033–1036.
  12. Goon PKY, Lip GYH, Boos CJ, et al. Circulating endothelial cells, endothelial progenitor cells, and endothelial microparticles in cancer. Neoplasia. 2006; 8(2): 79–88.
  13. Mutin M, Canavy I, Blann A, et al. Direct evidence of endothelial injury in acute myocardial infarction and unstable angina by demonstration of circulating endothelial cells. Blood. 1999; 93(9): 2951–2958.
  14. Schmidt DE, Manca M, Hoefer IE. Circulating endothelial cells in coronary artery disease and acute coronary syndrome. Trends Cardiovasc Med. 2015; 25(7): 578–587.
  15. Burger D, Touyz RM. Cellular biomarkers of endothelial health: microparticles, endothelial progenitor cells, and circulating endothelial cells. J Am Soc Hypertens. 2012; 6(2): 85–99.
  16. Lanuti P, Santilli F, Marchisio M, et al. A novel flow cytometric approach to distinguish circulating endothelial cells from endothelial microparticles: relevance for the evaluation of endothelial dysfunction. J Immunol Methods. 2012; 380(1-2): 16–22.
  17. Sinning JM, Losch J, Walenta K, et al. Circulating CD31+/Annexin V+ microparticles correlate with cardiovascular outcomes. Eur Heart J. 2011; 32(16): 2034–2041.
  18. Blann AD, Woywodt A, Bertolini F, et al. Circulating endothelial cells. Biomarker of vascular disease. Thromb Haemost. 2005; 93(2): 228–235.
  19. Sabatier F, Camoin-Jau L, Anfosso F, et al. Circulating endothelial cells, microparticles and progenitors: key players towards the definition of vascular competence. J Cell Mol Med. 2009; 13(3): 454–471.
  20. Bertolini F, Shaked Y, Mancuso P, et al. The multifaceted circulating endothelial cell in cancer: towards marker and target identification. Nat Rev Cancer. 2006; 6(11): 835–845.
  21. Solovey A, Lin Y, Browne P, et al. Circulating activated endothelial cells in sickle cell anemia. N Engl J Med. 1997; 337(22): 1584–1590.