Involvement of proinflammatory S100A9/A8 in the atherocalcinosis of aortic valves

Authors

  • R. А. Moskalenko Sumy State University, Ukraine,
  • А. M. Romaniuk Sumy State University, Ukraine Umeå University, Sweden,
  • I. O. Iashchishyn Sumy State University, Ukraine Umeå University, Sweden,
  • I.-M. S. Zakorko Sumy State University, Ukraine,
  • А. M. Piddubnyi Sumy State University, Ukraine,
  • Ye. O. Chernov Umeå University, Sweden,
  • L. O. Morozova-Roche Umeå University, Sweden,

DOI:

https://doi.org/10.14739/2310-1237.2017.1.98183

Keywords:

aortic valves, calgranulin A, calgranulin B

Abstract

According to the results of the Euro-Heart Survey on Vascular Heart Disease the most common pathology is nonrheumatic aortic stenosis, it is also called as calcific aortic valve stenosis (CAVS), as in its pathogenesis the process of biomineralization of valve cusps and ring plays the main role.

The aim of the work is the immunohistochemical study of mineralized tissue of aortic heart valves, which are affected by atherocalcinosis.

Materials and methods. 30 samples of mineralized aortic valves (I group) and 10 samples of aortic valve without evidence of biomineralization (II group  -– control) were studied. Immunohistochemical study of expression of collagen (Collagen I), CD68, myeloperoxidase (MPO), calgranulin A (S100A8), calgranulin B (S100A9), caspase 3 (Casp 3) and osteopontin (OPN) was conducted in AV tissue of both groups.

Results. In CAV tissues the fibrillar component (collagen I) growths was found, but the quantitative and qualitative compositions of CD68+ circulating inflammatory cells are not significantly different from the control group. CAVs contain much more MPO+ -cells (p <0.001) in comparison to the group of AVs without biomineralization.

Our data show a significant increase of the S100A9 and OPN expression in the mineralized tissue of AVs (p < 0.01). Also, a higher expression level of Casp3 and MPO was found in CAVs (p < 0.05). Comparing the first and the second groups of AVs connection between the expression of S100A8 was not determined.

Conclusion. High Casp 3 expression confirms the increased level of cell elimination in the CAVs tissue, which is obviously connected with the impact of high local concentrations of S100A9. These facts can contribute to the development of pathological biomineralization of AV. Since osteopontin inhibits the hydroxyapatite formation by binding to the surface of the crystals, its hyperproduction is a counteracting factor against biomineralization in AV tissue.

References

Iung, B., Baron, G., Butchart, E. G., Delahaye, F., Gohlke-Bärwolf, C., Levang, O. W. et al. (2003) A prospective survey of patients with valvular heart disease in Europe: The Euro Heart Survey on Valvular Heart Disease. Eur Heart J, 24, 1231–43. doi: https://doi.org/10.1016/S0195-668X(03)00201-X.

The task force on the management of valvular hearth disease of European Society of Cardiology (2007). Guidelines on the management of valvular heath disease.

Tornos, P. (2006) Valvular hearth disease in women. Rev Esp Cardiol, 59, 832–6.

Palorari, A., Loardi, C., Mussoni, L., Cavallotti, L., Camera, M., Biglioli, P., et al. (2009) Nonrheumatic calcific aortic stenosis: an overview from basic science to pharmacological prevention. European Journal of Cardio-thoracic Surgery, 35, 493–504. doi: 10.1016/j.ejcts.2008.11.033.

Dzemeshkevich, S. L., Stivenson, L. W., & Alexi-Meschischvili, V. V. (2004) Bolezni aortalʹnogo klapana: funccia, diagnostika, lecheniye [Diseases of the aortic valve: function, diagnosis, treatment]. Moscow: Geotar-Med. [in Russian].

Yang, X., Fullerton, D. A., Su, X., , Ao, L., Cleveland, J. C. Jr., & Meng, X. (2009) Pro-osteogenic phenotype of human aortic valve interstitial cells is associated with higher levels of Toll-like receptors 2 and 4 and enhanced expression of bone morphogenetic protein 2. J. Am. Coll. Cardiol, 53(6), 491–500. doi: 10.1016/j.jacc.2008.09.052.

Vogl, T., Gharibyan, A., & Morozova-Roche, L. A. (2012) Pro-inflammatory S100A8 and S100A9 proteins: self-assembly into multifunctional native and amyloid complexes. Int. J. Mol. Sci, 13, 2893–2917. doi: 10.3390/ijms13032893.

Sainger, R., Grau, J. B., Poggio, P., Branchetti, E., Bavaria, J. E., Gorman, J. H. et al. (2012) Dephosphorilation of circulating human osteopontin correlates with severe vascular calcification in patients with calcific aortic valve disease. Biomarkers, 17(2), 111–8. doi: 10.3109/1354750X.2011.642407.

Moskalenko, R., Romaniuk, A., Zakorko, I-M., Levchenko, , D. O., Piddubnyi, A. M., & Hetmanska, V. M. et al. (2016) Pathological biomineralization in heart valves affected by atherosclerosis. J. Clin. Exp. Med. Res, 4(4), 564–571.

Hamlin, N. J., & Price, P. A. (2004) Mineralization of decalcified bone occurs under cell culture conditions and requires bovine serum but not cells. Calcif Tissue Int, 75(3), 231–42. doi: 10.1007/s00223-004-0190-1.

Butcher, J. T., & Mahler, G. J. (2006) Inflammatory regulation of valvular remodeling: the good (?), the bad, and the ugly. J. Tissue. Eng, 12(7), 905–915.

Miller, J. D., Weiss, R. M., & Heistad, D. D. (2006) Calcific aortic valve stenosis: methods, models, and mechanisms. Circ Res, 108, 1392–1412, doi: 10.1161/CIRCRESAHA.110.234138.

Damo, S. M., Kehl-Fie, T. E., Sugitani, N., Holt, M. E., Rathi, S., Murphy, W. J.,et al. (2013) Molecular basis for manganese sequestration by calprotectin and roles in the innate immune response to invading bacterial pathogens. PNAS, 110(10), 3841–3846. doi: 10.1073/pnas.1220341110.

Franke, S., Rüster, C., Pester, J., Hofmann, G., Oelzner, P., & Wolf, G. (2011) Advanced glycation end products affect growth and function of osteoblasts. Clin Exp Rheumatol, 29(4), 650–60.

Grau, J. B., Poggio, P., Sainger, R., Vernick, W. J., Seefried, W. F., Branchetti, E., et al. (2012) Analysis of osteopontin levels for the identification of asymptomatic patients with calcific aortic valve disease. Ann Thorac Surg, 93, 79–86. doi: 10.1016/j.athoracsur.2011.08.036.

Passmore, M., Nataatmadja, M., Fung, Y. L., Pearse, B., Gabriel, S., Tesar, P., & Fraser, J. F. (2015) Osteopontin alters endothelial and valvular interstitial cell behavior in calcific aortic valve stenosis through HMGB1 regulation. Eur J Cardiothorac Surg, 48, e20–e29. doi: 10.1093/ejcts/ezv244.

Steitz, S. A., Speer, M. Y., McKee, M. D. et al. (2002) Osteopontin inhibits mineral deposition and promotes regression of ectopic calcification. Am. J. Pathol, 161(6), 2035–46. doi: 10.1016/S0002-9440(10)64482-3.

Edgeworth, J., Gorman, M., Bennett, R., Freemont, P., & Hogg, N. (1991) Identification of p 8, 14 as a highly abundant heterodimeric calcium binding protein complex of myeloid cells. Journal of Biological Chemistry, 266(12), 7706–7713.

Chen, J. H., Chen, W. L., Sider, K. L., Yip, C. Y., & Simmons, C. A. (2011) β-catenin mediates mechanically regulated, transforming growth factor-β1-induced myofibroblast differentiation of aortic valve interstitial cells. Arterioscler Thromb Vasc Biol, 31(3), 590–7. doi: 10.1161/ATVBAHA.110.220061.

Downloads

How to Cite

1.
Moskalenko RА, Romaniuk АM, Iashchishyn IO, Zakorko I-MS, Piddubnyi АM, Chernov YO, Morozova-Roche LO. Involvement of proinflammatory S100A9/A8 in the atherocalcinosis of aortic valves. Pathologia [Internet]. 2017Apr.7 [cited 2024Apr.25];(1). Available from: http://pat.zsmu.edu.ua/article/view/98183

Issue

Section

Original research