Investigation of the Effects of Cilostazol on the Myocardial Ischemia-Reperfusion Injury of Rats

Anar Amrah 1 *
More Detail
1 Republican Diagnostical Center, Baku, Azerbaijan
* Corresponding Author
J CLIN MED KAZ, Volume 21, Issue 2, pp. 59-65. https://doi.org/10.23950/jcmk/14494
OPEN ACCESS 439 Views 238 Downloads
Download Full Text (PDF)
Author Contributions: Conceptualization, formal analysis, investigation, methodology, project administration, supervision; validation, visualization, roles/writing – original draft, writing – review and editing, A.A. The author has read and agreed to the published version of the manuscript.

ABSTRACT

Background. Myocardial ischemia, occurring as a consequence of imbalance between oxygen supply and demand, causes a rapid metabolic and structural
impairment within the tissue. After a period of ischemia, sudden onset of reperfusion causes a transition to aerobic metabolism within living cells. Afterwards, emerging substrates initiate a chain of reactions leading to tissue injury. This situation is called “ischemia reperfusion injury”. Despite all technical advancements in anesthesia, myocardial protection and cardiac surgical techniques, we still face the clinical reflections of ischemia reperfusion (IR) injury.
Materials and methods. The protective effect of cilostasole on IR injury in an animal model of experimental myocardial ischemia and reperfusion was investigated. In this regional myocardial ischemia model, male Wistar-Albino rats were used as subjects and they were allocated into three groups; ischemia (n=8), sham (n=8), and cilostazole (n=8). LAD was occluded for 45 minutes, and then reperfused for three hours. Rats received Cilostazole 20 mg/kg/d by gastric gavage once daily. During IR hemodynamic parameters were recorded. Serum analysis for CK-MB and Troponin T were analysed at 180th minute of ischemia. Ischemic zone was measured by dying with Evans Blue and infarct area was measured by dying with triphenyltetrazolium chloride.
Results. Before the onset of LAD occlusion, as well as at 25th, 60th and 120th minutes of occlusion, all groups were similar in terms of blood pressure and pulse rate.
The total area, affected area and necrotic area were calculated by using formulas; affected area ratio= affected area/total area X 100, necrotic area ratio = necrotic area/total affected area X 100, necrotic area and affected area ratio = necrotic area /affected area X 100.  Affected area and total area ratio was significantly higher in IR group, compared with cilostazole group (t=8.965; p<0.001). Similarly, necrotic area and total area ratio was higher in IR group, compared with cilostazole group (t=8.965; p<0.001). The necrotic area and affected area ratios were similar in IR and cilostazole groups (t=0.245; p=0.810). CK-MB level differences were not statistically significant between two groups (Z=0.382; p=0.721).
Troponin levels were similar between IR and cilostazole groups and the difference was not statistically significant (Z=0.630; p=0.574). Pathological specimens of the heart were scanned for myocytolysis, PMNL and hemorrhage.  The difference between mean value of MDA enzyme levels were statistically significant (p<0.001) between all groups.  MDA enzyme levels, from higher to lower was IR, cilostazole and Sham group.  SOD levels (F=5.910; p=0.009) were significantly lower in Sham group when compared with IR group (p=0.008). The differences between Sham and cilostazole groups and IR and cilostazole gropus were not statistically significant (p=0.008). According to planimetric values and enzyme levels, cilostazole was found to be effective in reducing the ischemic zone, without effecting the necrotic zone in cardiac ischemia reperfusion damage. Therefore cilostazole has protective effects agains ischemia reperfusion damage.

Conclusion. This study explored how cilostazol affects myocardial ischemia-reperfusion injury in rats, finding that cilostazol administration during reperfusion may protect against such injury. Through various analyses, we observed positive outcomes associated with cilostazol treatment, suggesting its potential in reducing myocardial damage. Further research is needed to understand the underlying mechanisms and optimize therapeutic strategies, but our findings highlight cilostazol's promise in improving clinical outcomes in cardiac interventions.

CITATION

Amrah A. Investigation of the Effects of Cilostazol on the Myocardial Ischemia-Reperfusion Injury of Rats. J CLIN MED KAZ. 2024;21(2):59-65. https://doi.org/10.23950/jcmk/14494

REFERENCES

  • Steenberger C, Hill ML, Jennings RB. Volume regulation and plasma membrane injury in aerobic, anearobic, and ischemic myocardium in vitro. Circ Res 1987;57:864-75
  • Writing Committee Members. Gerhard-Herman MD, Gornik HL, Barrett C, Barshes NR, Corriere MA, Drachman DE, Fleisher LA, Fowkes FGR, Hamburg NM, Kinlay S, Lookstein R, Misra S, Mureebe L, Olin JW, Patel RAG, Regensteiner JG, Schanzer A, Shishehbor MH, Stewart KJ, Treat-Jacobson D, Walsh ME, ACC/AHA Task Force Members. Halperin JL, Levine GN, Al-Khatib SM, Birtcher KK, Bozkurt B, Brindis RG, Cigarroa JE, Curtis LH, Fleisher LA, Gentile F, Gidding S, Hlatky MA, Ikonomidis J, Joglar J, Pressler SJ, Wijeysundera DN. 2016 AHA/ACC Guideline on the Management of Patients with Lower Extremity Peripheral Artery Disease: Executive Summary. Vasc Med. 2017 Jun;22(3):NP1-NP43. [PubMed]
  • Niu PP, Guo ZN, Jin H, Xing YQ, Yang Y. Antiplatelet regimens in the long-term secondary prevention of transient ischaemic attack and ischaemic stroke: an updated network meta-analysis. BMJ Open. 2016 Mar 17;6(3):e009013. [PMC free article] [PubMed]
  • Powers WJ, Rabinstein AA, Ackerson T, et al.. Guidelines for the early management of patients with acute ischemic stroke: 2019 update to the 2018 guidelines for the early management of acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2019;50:e344–418. 10.1161/STR.0000000000000211 [PubMed] [CrossRef] [Google Scholar]
  • Li, J., X. Xiang, X. Gong, Y. Shi, J. Yang, and Z. Xu. Cilostazol protects mice against myocardium ischemic/ reperfusion injury by activating a PPARgamma/JAK2/ STAT3 pathway. Biomed. Pharmacother. 94:995–1001, 2017. https://doi.org/10.1016/j.biopha.2017.07.143; (PMID: 28810537).
  • Shimizu, T., T. Osumi, K. Niimi, and K. Nakagawa. Physico-chemical properties and stability of cilostazol. Arzneimittelforschung 35(7a):1117–1123, 1985.Shaw LM, Kaplan B, Kaufman D. Toxic effects of immunosuppressivedrugs: Mechanism and strategies for controlling them. Clin Chem 1996;42:1316-1321
  • Plosker GL and Foster RH. cilostazol: A further update of its pharmacology and therapeutic use in the management of organ transplantation. Drugs 2000;59:323-389
  • Teagtmeyer H, King LM, Jones BE. Energy substrate metabolism, myocardial ischemia, and targets of pharmacotherapy. Am J Cardiol 1998;82:54-60
  • Kloner RA, Bolli R, Marban E, Reinlib L, Braunwald E. Medical and cellular implication of stunning, hibernation, and precondition: an NHLBI workshop. Circulation 1998;97(18):1848-1867
  • Kloner RA, Jenning RB. Consequences of brief ischemia: stunning, preconditioning, and their clinical implications: part 1. Circulation 2001;104(24):2981-89
  • Kloner RA, Jennings RB. Consequences of brief ischemia: stunning, preconditioning and their clinical implications: part 2. Circulation. 2001;104(25):3158-67
  • Murry CE, Richard VJ, Reimer KA, Jennings RB. İschemic preconditioning slows energy metabolism and delays ultrastructural damage during a sustained ischemic episode. Circ Res. 1990;66(4):913-931
  • Diethard P, UteB, Markus k, Manfred D. Cardioprotective effects of the serine protease inhibitor aprotinin after regional ischemia and reperfusion injury. Jour. Thorasic and Card.Surg. 2002;124:5:942949
  • Downey JM, Cohen MV, Mitochondrial K channel opening during index ischemia and following myocardial reperfusion in ischemic rat hearts. J.Mol.Cell Cardiol. 2001;33:651-3
  • Chiara M, Daniela G, Maria M, Shiela L. Further evidence that melanokortins prevent myocardial reperfusion injury by activating melanocortin MC3 receptors. Eur. Jour.Pharm. 2003;447:227-234
  • Thomsan JA, Hess ML. The oxygen free radical system: A fundemantal mechanism in the production of myocardial necrosis. Prog Cardiovasc Dis 1986;28:449-462
  • Southorn PA, Powis G. Free radicals in medicine. 1. Chemical nature and biological reactions. Mayo Clin Proc 1998;63:381-89
  • Wakabayashi H, Karasawa Y, Tanaka S, Kokudo Y. The effect of FK506 on warm ischemia and reperfusion injury in the rat liver. Surg Today. 1994;24:994-1002
  • Sark MF, Zetti GM, Farghali H, Nalessic FH. FK506 amelorites the hepatic injury associated with ischemia and reperfusion in rats.
  • Zini R, Simon N, Morin C, Tillement JR. cilostazol decreases in vitro oxidative phosphorilation of mitochondria from rat forebrain. Life Sci. 1998;63:257368
  • Dhar DK, Takemoto Y, Uchidae M, Ono T. FK506 maintains cellular calcium homeostasis in ischemia-reperfusion injury of the canine liver. J Surg. Res 1996;60:142-146
  • Klein HH, Pushmann S, Shaper J, Schaper W. The mechanism of the tetrazolium reaction in identifying experimental myocardial infarction. Arch Pathol. Anat.1981;393:287-297