Irisin relaxes rat thoracic aorta through inhibiting signaling pathways implicating protein kinase C

Irisin relaxes rat thoracic aorta through inhibiting signaling pathways implicating protein kinase C

Background/aim: Irisin, a newly identified exercise-derived myokine, has been found involved in a peripheral vasodilator effect. However, little is known regarding the potential vascular activity of irisin, and the mechanisms underlying its effects on vascular smooth muscle have not been fully elucidated. This study was aimed to investigate the effects of irisin on vascular smooth muscle contractility in rat thoracic aorta, and the hypothesis that protein kinase C (PKC) may have a role in these effects. Materials and methods: Isometric contraction-relaxation responses of thoracic aorta rings were measured with an isolated organ bath model. The steady contraction was induced with 10 µM phenylephrine (PHE), and then the concentration-dependent responses of irisin (0.001-1 µM) were examined. The time-matched vehicle control (double distilled water) group was also formed. To evaluate the role of PKC, endothelium-intact thoracic aorta rings were incubated with 150 nM bisindolylmaleimide I (BIM I) for 20 min before the addition of 10 µM PHE and irisin. Also, a vehicle control group was formed for dimethyl sulfoxide (DMSO). Results: Irisin exerted the vasorelaxant effects at concentrations of 0.01, 0.1, and 1 µM compared to the control group (p < 0.001). Besides, PKC inhibitor BIM I incubation significantly inhibited the relaxation responses induced by varying concentrations of irisin (p: 0.000 for 0.01 µM; p: 0.000 for 0.1 µM; p: 0.000 for 1 µM). However, DMSO, a solvent of BIM I, did not modulate the relaxant effects of irisin (p > 0.05). Conclusion: In conclusion, physiological findings were obtained regarding the functional relaxing effects of irisin in rat thoracic aorta. The findings demonstrated that irisin induces relaxation responses in endothelium-intact thoracic aorta rings in a concentration-dependent manner. Furthermore, this study is the first to report that irisin-induced relaxation responses are regulated probably via activating signaling pathways implicating PKC.

___

  • 1. Boström P, Wu J, Jedrychowski MP, Korde A, Ye L et al. A PGC1- α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012; 481: 463-468. doi: 10.1038/nature10777
  • 2. Sanchis-Gomar F, Lippi G, Mayero S, Perez-Quilis C, GarcíaGiménez JL. Irisin: a new potential hormonal target for the treatment of obesity and type 2 diabetes. Journal of Diabetes, 2012; 4: 196. doi: 10.1111/j.1753-0407.2012.00194.x
  • 3. Polyzos SA, Anastasilakis AD, Efstathiadou ZA, Makras P, Perakakis N et al. Irisin in metabolic diseases. Endocrine 2018; 59: 260-274. doi: 10.1007/s12020-017-1476-1
  • 4. Korta P, Pocheć E, Mazur-Biały A. Irisin as a multifunctional protein: implications for health and certain diseases. Medicina (Kaunas) 2019; 55: 485. doi: 10.3390/medicina55080485
  • 5. Ma C, Ding H, Deng Y, Liu H, Xiong X et al. Irisin: a new code uncover the relationship of skeletal muscle and cardiovascular health during exercise. Frontiers in Physiology 2021; 12: 620608. doi: 10.3389/fphys.2021.620608
  • 6. Brunner H, Cockcroft JR, Deanfield J, Donald A, Ferrannini E et al. Endothelial function and dysfunction. Part II: association with cardiovascular risk factors and diseases. A statement by the Working Group on Endothelins and Endothelial Factors of the European Society of Hypertension. Journal of Hypertension 2005; 23: 233-246. doi: 10.1097/00004872-200502000-00001
  • 7. Jespersen B, Tykocki NR, Watts SW, Cobbett PJ. Measurement of smooth muscle function in the isolated tissue bath-applications to pharmacology research. Journal of Visualized Experiments 2015; 19: 52324. doi: 10.3791/52324
  • 8. Maciorkowska M, Musiałowska D, Małyszko J. Adropin and irisin in arterial hypertension, diabetes mellitus and chronic kidney disease. Advances in Clinical and Experimental Medicine 2019; 28: 1571-1575. doi: 10.17219/acem/104551
  • 9. Byun K, Lee S. The potential role of irisin in vascular function and atherosclerosis: a review. International Journal of Molecular Sciences 2020; 21: 7184. doi: 10.3390/ijms21197184
  • 10. Voutyritsa E, Kyriakos G, Patsouras A, Damaskos C, Garmpi A et al. Experimental agents for the treatment of atherosclerosis: new directions. Journal of Experimental Pharmacology 2021; 13: 161-179. doi: 10.2147/JEP.S265642
  • 11. Pedersen BK, Akerström TC, Nielsen AR, Fischer CP. Role of myokines in exercise and metabolism. Journal of Applied Physiology 2007; 103: 1093-1098. doi: 10.1152/ japplphysiol.00080.2007
  • 12. Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nature Reviews Endocrinology 2012; 8: 457-465. doi: 10.1038/nrendo.2012.49
  • 13. Strasser B. Physical activity in obesity and metabolic syndrome. Annals of the New York Academy of Sciences 2013; 1281: 141- 159. doi: 10.1111/j.1749-6632.2012.06785.x
  • 14. Demirel S, Şahintürk S, Özyener F. Irisin and its effects on vascular contractility. Medical Journal of Suleyman Demirel University 2021; 28: 163-170. doi: 10.17343/sdutfd.718412
  • 15. Zhang W, Chang L, Zhang C, Zhang R, Li Z et al. Central and peripheral irisin differentially regulate blood pressure. Cardiovascular Drugs and Therapy 2015; 29: 121-127. doi: 10.1007/s10557-015-6580-y
  • 16. Han F, Zhang S, Hou N, Wang D, Sun X. Irisin improves endothelial function in obese mice through the AMPK-eNOS pathway. American Journal of Physiology-Heart and Circulatory 2015; 309: H1501-H1508. doi: 10.1152/ajpheart.00443.2015
  • 17. Jiang M, Wan F, Wang F, Wu Q. Irisin relaxes mouse mesenteric arteries through endothelium-dependent and endotheliumindependent mechanisms. Biochemical and Biophysical Research Communications 2015; 468: 832-836. doi: 10.1016/j. bbrc.2015.11.040
  • 18. Fu J, Han Y, Wang J, Liu Y, Zheng S et al. Irisin lowers blood pressure by improvement of endothelial dysfunction via AMPK-Akt-eNOS-NO pathway in the spontaneously hypertensive rat. Journal of the American Heart Association 2016; 5: e003433. doi: 10.1161/JAHA.116.003433
  • 19. Hou N, Liu Y, Han F, Wang D, Hou X et al. Irisin improves perivascular adipose tissue dysfunction via regulation of the heme oxygenase-1/adiponectin axis in diet-induced obese mice. Journal of Molecular and Cellular Cardiology 2016; 99: 188-196. doi: 10.1016/j.yjmcc.2016.09.005
  • 20. Ye L, Xu M, Hu M, Zhang H, Tan X et al. TRPV4 is involved in irisin-induced endothelium-dependent vasodilation. Biochemical and Biophysical Research Communications 2017; 495: 41-45. doi: 10.1016/j.bbrc.2017.10.160
  • 21. Cosentino-Gomes D, Rocco-Machado N, Meyer-Fernandes JR. Cell signaling through protein kinase C oxidation and activation. International Journal of Molecular Sciences 2012; 13: 10697-10721. doi: 10.3390/ijms130910697
  • 22. Somlyo AP, Somlyo AV. Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiological Reviews 2003; 83: 1325- 1258. doi: 10.1152/physrev.00023.2003
  • 23. Massett MP, Ungvari Z, Csiszar A, Kaley G, Koller A. Different roles of PKC and MAP kinases in arteriolar constrictions to pressure and agonists. American Journal of Physiology-Heart and Circulatory 2002; 283: H2282-H2287. doi: 10.1152/ ajpheart.00544.2002
  • 24. Korzick DH, Laughlin MH, Bowles DK. Alterations in PKC signalling underlie enhanced myogenic tone in exercisetrained porcine coronary resistance arteries. Journal of Applied Physiology 2004; 96: 1425-1432. doi: 10.1152/ japplphysiol.01077.2003
  • 25. Soloviev AI, Kizub IV. Mechanisms of vascular dysfunction evoked by ionizing radiation and possible targets for its pharmacological correction. Biochemical Pharmacology 2018; 159: 121-139. doi: 10.1016/j.bcp.2018.11.019
  • 26. Zhu D, Wang H, Zhang J, Zhang X, Xin C et al. Irisin improves endothelial function in type 2 diabetes through reducing oxidative/nitrative stresses. Journal of Molecular and Cellular Cardiology 2015; 87: 138-147. doi: 10.1016/j.yjmcc.2015.07.015
  • 27. Partovian C, Zhuang Z, Moodie K, Lin M, Ouchi N et al. PKCalpha activates eNOS and increases arterial blood flow in vivo. Circulation Research 2005; 97: 482-487. doi: 10.1161/01. RES.0000179775.04114.45
  • 28. Wang Y, Zhou H, Wu B, Zhou Q, Cui D et al. Protein kinase C isoforms distinctly regulate propofol-induced endotheliumdependent and endothelium-independent vasodilation. Journal of Cardiovascular Pharmacology 2015; 66: 276-284. doi: 10.1097/FJC.0000000000000275
  • 29. Clark SG, Fuchs LC. Role of nitric oxide and Ca++-dependent K+ channels in mediating heterogeneous microvascular responses to acetylcholine in different vascular beds. Journal of Pharmacology and Experimental Therapeutics 1997; 282: 1473-1479. PMID: 9316861
  • 30. Mulvany MJ, Aalkjaer C. Structure and function of small arteries. Physiological Reviews 1990; 70: 921-961. doi: 10.1152/ physrev.1990.70.4.921
  • 31. de Wit C, Wölfle SE. EDHF and gap junctions: important regulators of vascular tone within the microcirculation. Current Pharmaceutical Biotechnology 2007; 8: 11-25. doi: 10.2174/138920107779941462
Turkish Journal of Medical Sciences-Cover
  • ISSN: 1300-0144
  • Yayın Aralığı: Yılda 6 Sayı
  • Yayıncı: TÜBİTAK
Sayıdaki Diğer Makaleler

A new device for bronchoscopy for better protection

Aslıhan GÜRÜN KAYA, Aydın ÇİLEDAĞ, Akın KAYA, Miraç ÖZ, Serhat EROL, İncifer KARNAK ORHUN, Demet KARNAK

Evaluation of the frequency and intensity of COVID-19 in patients with ankylosing spondylitis under anti-TNF therapy

Sümeyye Merve TÜRK, Zeynep ÖZTÜRK, Damla KARATAŞ, Ünal ERKORKMAZ, Emel GÖNÜLLÜ

Irisin relaxes rat thoracic aorta through inhibiting signaling pathways implicating protein kinase C

Fadıl ÖZYENER, Serdar ŞAHİNTÜRK, Naciye İŞBİL, Sadettin DEMİREL

The impact of bone marrow-derived mesenchymal stem cells on experimental testicular torsion in rats

Emrah ŞENEL, Sabri DEMİR, Mehmet ZENGİN, Ahmet ERTÜRK, Yasemin Dere GÜNAL, Miyase ÇINAR, Siyami KARAHAN, Dinçer YILDIZ

Clinically mild encephalitis/encephalopathy with a reversible splenial lesion of the corpus callosum in childhood: a single-center experience

Betül Emine DERİNKUYU, Melek Melehat OĞUZ, Ebru AZAPAĞASI, Ülkühan ÖZTOPRAK, Erhan AKSOY, Halil ÇELİK, Deniz YÜKSEL, Nesrin CEYLAN, Suna ÖZDEM

Linguistic validation of a widely used recovery score: quality of recovery-15 (QoR-15)

Sami EKSERT, Mehmet Burak EŞKİN, Serkan ŞENKAL, Umut KARA, Fatih ŞİMŞEK, Mehmet Özgür ÖZHAN, Mehmet Emin İNCE, Gökhan ÖZKAN, Ümit ALAKUŞ, Hasan KAMBUROĞLU

The role of leptin in primary Sjögren syndrome: a clinical and histopathological assessment study

Kayhan BAŞAK, Mehmet Engin TEZCAN, Mustafa ERDOĞAN

Validity and reliability of Fried frailty phenotype in Turkish population

Hacer DOĞAN VARAN, Muhammet Cemal KIZILARSLANOĞLU, Rana TUNA DOĞRUL, Olgun DENİZ, Süheyla ÇÖTELİ, Berna GÖKER

Pasireotide treatment in Cushing’s disease: A single tertiary center’s experience

Pınar KADIOĞLU, Serdar ŞAHİN, Emre DURCAN, Hande Mefkure ÖZKAYA, Gular KARİMOV, Şeyda Gül ÖZCAN

Tp-e interval, Tp-e/QT, and Tp-e/QTc ratios in patients with primary hyperparathyroidism and their relationship with cardiac arrhythmic events

Ferhat GÖKAY, Yücel YILMAZ, Yasin ŞİMŞEK, Şaban KELEŞOĞLU