437
views
0
recommends
+1 Recommend
1 collections
    0
    shares

      CVIA now indexed by SCOPUS from February 2024. CVIA received its first Journal Impact Factor (0.5) in the 2023 Journal Citation Reports Release. 

      Interested in becoming a CVIA published author?

      • Platinum Open Access with no APCs. 
      • Fast peer review/Fast publication online after article acceptance.

      Submissions should be made electronically at: https://mc04.manuscriptcentral.com/cvia-journal.

      Please refer to the Author Guidelines at https://cvia-journal.org/instructions-to-authors/ before submission.

       

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Serum Irisin: Pathogenesis and Clinical Research in Cardiovascular Diseases

      Published
      review-article
      Bookmark

            Abstract

            Recently, muscular function/dysfunction has gained importance in the maintenance of metabolic homeostasis in cardiovascular diseases. Skeletal muscle plays a vital role in coordinating the activity and metabolism of endocrine organs by secreting many myokines, especially irisin. Irisin is a polypeptide hormone consisting of 112 amino acids secreted into the blood from muscle and adipose tissues. Serum irisin levels are associated with cardiometabolic risk factors such as obesity and insulin resistance as defined by homeostatic model assessment. Irisin reduces endothelial damage by inhibiting inflammation and oxidative stress, thus playing a key role in maintaining endothelial cell function. Unsurprisingly, low irisin levels cause endothelial dysfunction and increase the incidence of atherosclerosis. We aimed to summarize the studies on this issue since we have not found any review in the literature on the role of serum irisin levels in the process of atherosclerosis and other cardiovascular events in cardiovascular diseases.

            Main article text

            Introduction

            Cardiovascular diseases (CVDs) are one of the leading causes of death and morbidity worldwide [1]. Diabetes mellitus (DM) increases the risk of CVDs by 1.5–2 times in men and 2–3 times in women. The risk of atherosclerotic disease in individuals with diabetes is 2–3 times higher than in individuals without DM [2]. In the last decade, muscle and adipose tissues have been described as regulators of endocrine metabolism in the body [3]. Skeletal muscle is considered to be an endocrine organ that secretes many myokines, including follistatin, myostatin, activin A, and irisin, which appear to play a vital role in coordinating the activity and metabolism of other endocrine organs [4].

            Many biological processes related to energy metabolism are regulated by peroxisome proliferator– activated receptor γ (PPARγ) coactivator 1α, which also stimulates certain muscle gene products such as fibronectin type III domain–containing protein 5 (FNDC5) [5]. FNDC5 is a membrane protein released into the circulation after exercise and produces a derivative called irisin [5]. Irisin is a polypeptide hormone containing 112 amino acids secreted into the bloodstream from muscle and adipose tissues. Participates in the transformation of white adipocytes into brown adipocytes, both in vitro and in vivo, and stimulates the expression of thermogenic genes, including the gene encoding the mitochondrial brown fat separation protein 1 [5, 6]. Increased thermogenesis is a recommended mechanism by which irisin can increase insulin sensitivity and reduce blood glucose levels and body weight [5, 7]. Irisin, sirtuin, and vaspin have been proposed as regulators of PPARγ, and it appears all of these adipokines might be associated with PPARγ level [8].

            In sedentary life, serum irisin levels are associated with cardiometabolic risk factors such as obesity and insulin resistance as defined by homeostatic model assessment [9]. Insulin resistance may result in microangiopathy and macroangiopathy, peripheral arterial disease, vascular dysfunction, hypertension, and endothelial cell and cardiomyocyte dysfunction. Directly related to insulin resistance and coronary artery disease (CAD), irisin increases the risk of coronary artery disease, myocardial infarction (MI), and heart failure [10].

            Plasma irisin levels have been suggested to be positively correlated with body mass index. Irisin is a physiological protective factor against obesity, and its increased levels compensate for the increase in body weight. In obese individuals, physiological irisin cannot maintain the balance between stored and consumed energy, and thus the secretion of irisin is increased for the transfer of stored fat in skeletal muscle and adipose tissues [11].

            Given the characteristics of irisin mentioned above, it may be considered a potential target for monitoring and intervention in people with type 2 (DM) and cardiovascular complications [12]. It can also be studied as a new target for the treatment of concomitant diseases such as obesity and diabetes and nonalcoholic fatty liver disease. However, despite this potential, many questions remain unanswered about the benefits of irisin as a therapeutic target. An important issue in this context is the role of irisin in metabolic syndrome and the associated complications, especially CVDs [10]. In this context, since there is no specific review of serum irisin levels and CVDs in the literature to our knowledge, we aimed to evaluate and synthesize research related to serum irisin levels in CVDs.

            Khorasani et al. [12] reported irisin level was significantly higher in diabetic patients without CVDs than in diabetic patients with CVDs. To date, relations between irisin and DM [13, 14], obesity [15], and CVDs [16] have been reported. In some studies, irisin level was significantly lower in DM and MI patients than in control individuals [14, 1618].

            Studies comparing circulating irisin levels with levels in a control group have shown that irisin has a protective effect against the development of CVDs [1619]. Several potential mechanisms have been proposed in this regard. Irisin plays a key role in maintaining endothelial cell function, and low levels of irisin lead to endothelial dysfunction and increase the incidence of atherosclerosis [20, 21]. In addition, irisin reduces endothelial damage by inhibiting inflammation and oxidative stress [22]. Hyperhomocysteinemia (which increases the risk of heart disease) has an inverse correlation with irisin levels [23]. Circulating irisin levels may be an indicator of increased heart muscle damage in patients with myocardial lesions [24]. In addition, low circulating irisin levels may increase the accumulation of advanced glycation end products, which lead to vascular complications in DM patients [21]. Irisin also plays an important role in lipid metabolism, and decreased irisin levels may lead to hyperlipidemia in CAD [25]. Despite these findings, the explanation of the function and mechanism of irisin in CVDs is not yet satisfactory.

            Calan and Demirpence [26] found that circulating irisin levels were higher in patients with acromegaly. High levels of circulating irisin in patients with acromegaly were associated with increased epicardial adipose tissue and carotid intima-media thickness. Increased irisin levels may be a useful marker for cardiovascular risk in patients with acromegaly, but further research is needed to clarify the findings.

            The relationship between metabolic syndrome and irisin in the pediatric population has been described [27]. The results suggest that overweight and obese children with metabolic syndrome have significantly lower irisin levels than those without metabolic syndrome.

            Shim et al. [28] found an inverse relationship between glucose and triglyceride levels and irisin levels in overweight/obese children. Irisin is negatively associated with metabolic syndrome parameters (central obesity, high blood pressure, high blood sugar, high serum triglycerides, and low serum high-density lipoprotein (HDL)) also in adult populations [29]. Interaction of irisin with lipids and glucose may result in cell dysfunction and metabolic syndrome [30].

            Fan et al. [31] demonstrated the protective effects of irisin in the presence of hyperglycemia stress in reperfusion-treated cardiomyocytes. Irisin treatment supported survival of cardiomyocytes by modulating mitochondrial function via adenosine 5′-monophosphate–activated protein kinase (AMPK). The findings suggest that irisin is a promising therapeutic agent in the treatment of cardiac muscle damage in DM patients.

            Elizondo-Montemayor et al. [32] conducted the first study showing reduced irisin levels in children and adolescents with type 2 DM compared with healthy controls. On the basis of experimental evidence, the reduction of circulating irisin levels in the pediatric population with type 2 DM can be attributed to the reduction of muscular secretion of FDNC5. The mechanism that may demonstrate that irisin has a potential protective mechanism against endothelial damage and vascular disease is the relationship between irisin and high-sensitivity C-reactive protein. They observed that the level of high-sensitivity C-reactive protein was higher in the type 2 DM group and positively correlated with anthropometric and metabolic markers, indicating that type 2 DM was a systemic inflammatory condition in the pediatric population. Low concentrations of irisin in this population may potentially increase the metabolic and inflammatory components of type 2 DM [32].

            It is well known that physical exercise benefits human health at any age. However, the exact mechanism by which physical exercise improves health is unknown. Recent studies of exercise-related FNDC5/irisin have begun to illuminate this mystery. Exercise-induced FNDC5/irisin has been shown to be protective against cardiovascular damage after an ischemic event, to improve neuronal function in patients with Alzheimer’s disease, and to play a role in macrophage and adipocyte regulation. Experiments have shown that FNDC5/irisin reduces cardiac regeneration, neovascularization, and cardiac fibrosis due to the interaction of Nkx-2.5-positive cardiac progenitor cells. It has also improved macrophage functions that can provide protection against defects in the cardiac conduction system [33].

            Similarly, in a mouse model of Alzheimer’s disease, FNDC5/irisin has been shown to alleviate memory impairment in mice with reduced memory performance [33]. In the same study, FNDC5/irisin was shown to be associated with regulation of osteocytes and adipocytes by signaling through the cytoplasmic membrane–integrated protein αVβ5 integrin receptor. Although these recent discoveries have strengthened the importance of FNDC5/irisin, many details about how FNDC5/irisin contributes to exercise physiology are unknown and deserve further research.

            Irisin level is reduced in patients with heart failure following MI. It is negatively correlated with high troponin I, creatine kinase MB (CK-MB), TNF, total cholesterol, low-density lipoprotein cholesterol, and triglyceride levels. In contrast, it is positively correlated with left ventricular ejection fraction (LVEF) and high-density lipoprotein cholesterol level. Troponin I and CK-MB are useful biomarkers in the diagnosis of MI and are useful in predicting the incidence of post-MI complications [34]. Higher irisin concentration results in greater ATP loss due to its uncoupling property. With a gradual decrease in serum and tissue irisin levels, it is aimed to protect myocardial cells by saving the energy required for ischemic myocardial cells. This causes more uncoupling with higher levels of irisin, leading to loss of ATP and more heat generation. The increased irisin levels cause more damage to myocardium and cause a decrease in ventricular function in MI [18].

            Yu et al. [35] demonstrated increased FNDC5/irisin expression in hypertrophic hearts, cardiomyocytes, and angiotensin II-treated skeletal muscle cells. Administration of recombinant irisin alleviated pathological cardiac hypertrophy and improved cardiac function both in vivo and in vitro. Irisin application activated AMPK–mammalian target of rapamycin signaling, which further reduces cardiac hypertrophy. Irisin was initially described as a skeletal muscle–derived protein [5]. However, an increasing number of studies have found expression of irisin in a variety of other organs, including heart muscle, brain, liver, and the gastrointestinal tract [36, 37]. Irisin expression appears to be relatively high in muscle-rich organs. Studies also show that heart muscle may have higher irisin expression than skeletal muscle [38].

            Chen et al. [39] reported that irisin reduces angiotensin II-induced cardiac fibrosis due to inhibition of redox-sensitive transforming growth factor β1–SMAD2/3 signaling via NRF2. As an inference, irisin therapeutics are thought to be useful to reduce cardiac fibrosis related to angiotensin II. Bi et al. [40] demonstrated that exogenous irisin attenuates hepatic ischemia-reperfusion damage by limiting mitochondrial division, supporting mitochondrial biogenesis, and reducing oxidative stress. Irisin treatment seems to be a new and promising treatment approach for ischemia-reperfusion injury in liver.

            Bashar et al. [41] recommended recombinant irisin treatment and regular exercise to protect people at risk of acute MI. They made this suggestion on the basis of a negative correlation between serum irisin and lipid peroxidation, CK-MB, troponin I, collagen deposition, and the percentage of caspase 3 expression in a trial with rats. They also showed a positive correlation between serum irisin and total antioxidant status.

            On the other hand, Ho et al. [42] showed that overexpression of irisin resulted in reactive oxygen species production and hypoxia-induced apoptosis in mice and cardiomyocytes. Irisin may have a double-edged sword effect on energy metabolism and tissue repair. Excessive irisin expression may increase energy wastage during the heart healing process. Prevention of overexpression of irisin after MI may be important. Further studies are needed to determine the exact mechanisms of the irisin/mitochondrial pathway in the myocardium and whether modulation of irisin levels may have therapeutic benefits in MI.

            Asadi et al. [43] showed that irisin causes dose-dependent neuroprotective effects in stroke-induced brain injury. This effect is achieved by suppressing apoptosis and enhancing brain-induced neurotrophic factor expression in the ischemic area. Tu et al. [44] claimed that irisin is a new and independent prognostic marker that improves the risk classification currently used in stroke patients.

            Kalkan et al. [45] reported that serum adropin and irisin levels were significantly increased in cachectic heart failure patients with reduced ejection fraction, and were significantly correlated with low-LVEF-related markers such as brain-type natriuretic peptide level and New York Heart Association class. The results show that adropin and irisin levels are correlated with the severity of heart failure and are reasonable candidates to be new markers for cardiac cachexia in patients with low-LVEF heart failure.

            Moreno-Perez et al. [46] demonstrated higher irisin concentrations were associated with higher insulin resistance, nonalcoholic fatty liver, and subclinical atherosclerosis in HIV-positive men without DM. Waist-hip ratio is the strongest predictor of insulin homeostasis, and plasminogen activator inhibitor 1 levels and lipodystrophy are the main determinants of subclinical atherosclerosis, but the study asserts that irisin is also positively associated with other cardiovascular risk factors and may be considered as a marker of subclinical atherosclerosis. Among the factors associated with HIV infection, only the duration of exposure to nucleoside reverse transcriptase inhibitors was associated with higher concentrations of irisin. At present, primary and secondary prevention of CVDs are by the management of traditional cardiovascular risk factors, and only extensive studies with clinical outcomes to clarify the role of irisin in cardiovascular and metabolic risk in the HIV-positive population will provide a definitive answer [46].

            He et al. [47] first proved a relationship between serum irisin levels and vascular calcification in hemodialysis patients. Lower irisin levels, long-term hemodialysis, and advanced age were independent risk factors for vascular calcification in hemodialysis patients.

            Deng [25] showed that a decrease in serum irisin concentration was associated with the presence and severity of CAD. Likewise, Hisamatsu et al. [48] demonstrated that higher serum irisin levels were associated with lower coronary atherosclerosis burden in the Japanese male population. This relationship was different from the traditional cardiometabolic pathways. Hsieh et al. [49] demonstrated that higher serum irisin levels were associated with increased major adverse cardiovascular events (MACEs) and outcome rates in patients after ST-elevation MI. As confidence in new myokines continues to increase for the treatment of obesity, identification of the myocardial effects of myokines will become more important. Whether pharmaceutical agents that increase or decrease irisin levels may be applicable to the treatment or prevention of MI has yet to be determined. Further studies are needed to determine the exact mechanisms of the irisin/mitochondrial pathway in the myocardium and whether modulation of irisin levels may have therapeutic benefits in MI.

            Conclusions

            Irisin is a recently discovered molecule produced by muscles. It has been shown to be associated with different metabolic markers. Current studies suggest that irisin is a promising therapeutic agent in diseases such as DM, metabolic syndrome, and CVDs. Further studies are needed for the use of irisin in clinical practice.

            Conflict of Interest

            The authors declare that they have no conflicts of interest.

            References

            1. American DiabetesAssociation. Standards of medical care in diabetes – 2018 abridged for primary care providers. Clin Diabetes 2018;36:14–37.

            2. MelmedS, PolonskyKS, LarsenPR, KronenbergHM. Williams textbook of endocrinology. Elsevier Health Sciences 2015.

            3. RosenED, SpiegelmanBM. Adipocytes as regulators of energy balance and glucose homeostasis. Nature 2006;444:847–53.

            4. VamviniMT, AronisKN, PanagiotouG, HuhJY, ChamberlandJP, BrinkoetterMT, et al. Irisin mRNA and circulating levels in relation to other myokines in healthy and morbidly obese humans. Eur J Endocrinol 2013;169:829–34.

            5. BostromP, WuJ, JedrychowskiMP, KordeA, YeL, LoJC, et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012;481:463–8.

            6. ZhuD, WangH, ZhangJ, ZhangX, XinC, ZhangF, et al. Irisin improves endothelial function in type 2 diabetes through reducing oxidative/nitrative stresses. J Mol Cell Cardiol 2015;87:138–47.

            7. ZhangY, LiR, MengY, LiS, DonelanW, ZhaoY, et al. Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling. Diabetes 2014;63:514–25.

            8. MirzaeiK, Hossein-NezhadA, KeshavarzSA, KoohdaniF, Saboor-YaraghiAA, HosseiniS, et al. Crosstalk between circulating peroxisome proliferator-activated receptor gamma, adipokines and metabolic syndrome in obese subjects. Diabetol Metab Syndr 2013;5:79.

            9. MorenoM, Moreno-NavarreteJM, SerranoM, OrtegaF, DelgadoE, Sanchez RagnarssonC, et al. Circulating irisin levels are positively associated with metabolic risk factors in sedentary subjects. PLoS One 2015;10:e0124100.

            10. ChenJQ, HuangYY, GusdonAM, QuS. Irisin: a new molecular marker and target in metabolic disorder. Lipids Health Dis 2015;14:2.

            11. ZhangM, ChenP, ChenS, SunQ, ZengQC, ChenJY, et al. The association of new inflammatory markers with type 2 diabetes mellitus and macrovascular complications: a preliminary study. Eur Rev Med Pharmacol Sci 2014;18:1567–72.

            12. KhorasaniZM, BagheriRK, YaghoubiMA, ChobkarS, AghaeeMA, AbbaszadeganMR, et al. The association between serum irisin levels and cardiovascular disease in diabetic patients. Diabetes Metab Syndr 2019;13:786–90.

            13. AtesI, ArikanMF, ErdoganK, KaplanM, YukselM, TopcuogluC, et al. Factors associated with increased irisin levels in the type 1 diabetes mellitus. Endocr Regul 2017;51:1–7.

            14. KhidrEG, AliSS, ElshafeyMM, FawzyOA. Association of irisin and FNDC5 rs16835198 G>T gene polymorphism with type 2 diabetes mellitus and diabetic nephropathy. An Egyptian pilot study. Gene 2017;626:26–31.

            15. ChenN, LiQ, LiuJ, JiaS. Irisin, an exercise-induced myokine as a metabolic regulator: an updated narrative review. Diabetes Metab Res Rev 2016;32:51–9.

            16. AydinS, KobatMA, KalayciM, ErenMN, YilmazM, KulogluT, et al. Decreased saliva/serum irisin concentrations in the acute myocardial infarction promising for being a new candidate biomarker for diagnosis of this pathology. Peptides 2014;56:141–5.

            17. EmanueleE, MinorettiP, Pareja-GaleanoH, Sanchis-GomarF, GaratacheaN, LuciaA. Serum irisin levels, precocious myocardial infarction, and healthy exceptional longevity. Am J Med 2014;127:888–90.

            18. KulogluT, AydinS, ErenMN, YilmazM, SahinI, KalayciM, et al. Irisin: a potentially candidate marker for myocardial infarction. Peptides 2014;55:85–91.

            19. ShenS, GaoR, BeiY, LiJ, ZhangH, ZhouY, et al. Serum irisin predicts mortality risk in acute heart failure patients. Cell Physiol Biochem 2017;42:615–22.

            20. ZhangY, MuQ, ZhouZ, SongH, WuF, JiangM, et al. Protective effect of irisin on atherosclerosis via suppressing oxidized low density lipoprotein induced vascular inflammation and endothelial dysfunction. PLoS One 2016;11:e0158038.

            21. LiZ, WangG, ZhuYJ, LiCG, TangYZ, JiangZH, et al. The relationship between circulating irisin levels and tissues AGE accumulation in type 2 diabetes patients. Biosci Rep 2017;37. pii: BSR20170213. doi: 10.1042/BSR20170213.

            22. LuJ, XiangG, LiuM, MeiW, XiangL, DongJ. Irisin protects against endothelial injury and ameliorates atherosclerosis in apolipoprotein E-null diabetic mice. Atherosclerosis 2015;243:438–48.

            23. AlisR, Sanchis-GomarF, Pareja-GaleanoH, Hernandez-MijaresA, RomagnoliM, VictorVM, et al. Association between irisin and homocysteine in euglycemic and diabetic subjects. Clin Biochem 2014;47:333–5.

            24. AronisKN, MorenoM, PolyzosSA, Moreno-NavarreteJM, RicartW, DelgadoE, et al. Circulating irisin levels and coronary heart disease: association with future acute coronary syndrome and major adverse cardiovascular events. Int J Obes (Lond) 2015;39:156–61.

            25. DengW. Association of serum irisin concentrations with presence and severity of coronary artery disease. Med Sci Monit 2016;22:4193–7.

            26. CalanM, DemirpenceM. Increased circulating levels of irisin are associated with cardiovascular risk factors in subjects with acromegaly. Hormones (Athens) 2019. doi:10.1007/s42000-019-00151-3.

            27. Elizondo-MontemayorL, Mendoza-LaraG, Gutierrez-DelBosqueG, Peschard-FrancoM, NieblasB, Garcia-RivasG. Relationship of circulating irisin with body composition, physical activity, and cardiovascular and metabolic disorders in the pediatric population. Int J Mol Sci 2018;19:3727.

            28. ShimYS, KangMJ, YangS, HwangIT. Irisin is a biomarker for metabolic syndrome in prepubertal children. Endocr J. 2018;65:23–31.

            29. YanB, ShiX, ZhangH, PanL, MaZ, LiuS, et al. Association of serum irisin with metabolic syndrome in obese Chinese adults. PLoS One 2014;9:e94235.

            30. De MeneckF, de SouzaLV, OliveiraV, do FrancoMC. High irisin levels in overweight/obese children and its positive correlation with metabolic profile, blood pressure, and endothelial progenitor cells. Nutr Metab Cardiovasc Dis 2018;28:756–64.

            31. FanJ, ZhuQ, WuZ, DingJ, QinS, LiuH, et al. Protective effects of irisin on hypoxia-reoxygenation injury in hyperglycemia-treated cardiomyocytes: role of AMPK pathway and mitochondrial protection. J Cell Physiol 2020;235:1165–74.

            32. Elizondo-MontemayorL, Gonzalez-GilAM, Tamez-RiveraO, Toledo-SalinasC, Peschard-FrancoM, Rodríguez-GutiérrezNA, et al. Association between irisin, hs-CRP, and metabolic status in children and adolescents with type 2 diabetes mellitus. Mediators Inflamm 2019;2019:6737318.

            33. ZhouX, XuM, BryantJL, MaJ, XuX. Exercise-induced myokine FNDC5/irisin functions in cardiovascular protection and intracerebral retrieval of synaptic plasticity. Cell Biosci 2019;9:32.

            34. Abd El-MottalebNA, GalalHM, El MaghrabyKM, GadallahAI. Serum irisin level in myocardial infarction patients with or without heart failure. Can J Physiol Pharmacol 2019;97:932–8.

            35. YuQ, KouW, XuX, ZhouS, LuanP, XuX, et al. FNDC5/irisin inhibits pathological cardiac hypertrophy. Clin Sci (Lond) 2019;133:611–27.

            36. GurFM, TimurkaanS, YalcinMH, GirginA, GencerTB. Immunohistochemical localization of irisin in mole rats (Spalax leucodon). Biotech Histochem 2017;92:245–51.

            37. CaiC, XiaoG, QianL, JiangS, LiB, XieS, et al. Gene location, expression, and function of FNDC5 in Meishan pigs. Sci Rep 2017;7:7886.

            38. AydinS, KulogluT, OzercanMR, AlbayrakS, AydinS, BakalU, et al. Irisin immunohistochemistry in gastrointestinal system cancers. Biotech HIstochem 2016;91:242–50.

            39. ChenRR, FanXH, ChenG, ZengGW, XueYG, LiuXT, et al. Irisin attenuates angiotensin II-induced cardiac fibrosis via Nrf2 mediated inhibition of ROS/TGFβ1/Smad2/3 signaling axis. Chem Biol Interact 2019;302:11–21.

            40. BiJ, ZhangJ, RenY, DuZ, LiQ, WangY, et al. Irisin alleviates liver ischemia-reperfusion injury by inhibiting excessive mitochondrial fission, promoting mitochondrial biogenesis and decreasing oxidative stress. Redox Biol 2019;20:296–306.

            41. BasharSM, Samir El-SherbeinySM, BoraieMZ. Correlation between the blood level of irisin and the severity of acute myocardial infarction in exercise-trained rats. J Basic Clin Physiol Pharmacol 2018;30:59–71.

            42. HoMY, WenMS, YehJK, HsiehIC, ChenCC, HsiehMJ, et al. Excessive irisin increases oxidative stress and apoptosis in murine heart. Biochem Biophys Res Commun 2018;503:2493–8.

            43. AsadiY, GorjipourF, BehrouzifarS, VakiliA. Irisin peptide protects brain against ischemic injury through reducing apoptosis and enhancing BDNF in a rodent model of stroke. Neurochem Res 2018;43:1549–60.

            44. TuWJ, QiuHC, CaoJL, LiuQ, ZengXW, ZhaoJZ. Decreased concentration of irisin is associated with poor functional outcome in ischemic stroke. Neurotherapeutics 2018;15:1158–67.

            45. KalkanAK, CakmakHA, ErturkM, KalkanKE, UzunF, TasbulakO, et al. Adropin and irisin in patients with cardiac cachexia. Arq Bras Cardiol 2018;111:39–47.

            46. Moreno-PerezO, Reyes-GarciaR, Muñoz-TorresM, MerinoE, BoixV, ReusS, et al. High irisin levels in nondiabetic HIV-infected males are associated with insulin resistance, nonalcoholic fatty liver disease, and subclinical atherosclerosis. Clin Endocrinol (Oxf) 2018;89:414–23.

            47. HeL, HeWY, ALT, YangWL, ZhangAH. Lower serum irisin levels are associated with increased vascular calcification in hemodialysis patients. Kidney Blood Press Res 2018;43:287–95.

            48. HisamatsuT, MiuraK, ArimaH, FujiyoshiA, KadotaA, KadowakiS, et al. Relationship of serum irisin levels to prevalence and progression of coronary artery calcification: a prospective, population-based study. Int J Cardiol 2018;267:177–82.

            49. HsiehIC, HoMY, WenMS, ChenCC, HsiehMJ, LinCP, et al. Serum irisin levels are associated with adverse cardiovascular outcomes in patients with acute myocardial infarction. Int J Cardiol 2018;261:12–17.

            Author and article information

            Journal
            CVIA
            Cardiovascular Innovations and Applications
            CVIA
            Compuscript (Ireland )
            2009-8782
            2009-8618
            January 2020
            March 2020
            : 4
            : 3
            : 195-200
            Affiliations
            [1] 1Department of Cardiology, Adiyaman Education and Research Hospital, Adiyaman, Turkey
            Author notes
            Correspondence: Lutfu Askin, MD, Cardiology Department, Adiyaman Education and Research Hospital, 2230 Adiyaman, Turkey, Tel.: +90-531-5203486, Fax: +90-4161015, E-mail: lutfuaskin23@ 123456gmail.com
            Article
            cvia20190569
            10.15212/CVIA.2019.0569
            e0d97c99-7f63-440f-9815-ad9459b6c267
            Copyright © 2020 Cardiovascular Innovations and Applications

            This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 Unported License (CC BY-NC 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. See https://creativecommons.org/licenses/by-nc/4.0/.

            History
            : 21 November 2019
            : 09 December 2019
            : 09 January 2020
            Categories
            Reviews

            General medicine,Medicine,Geriatric medicine,Transplantation,Cardiovascular Medicine,Anesthesiology & Pain management
            irisin,body mass index,cardiovascular diseases,insulin resistance

            Comments

            Comment on this article