Role of myokines in the maintenance of whole body metabolic homeostasis pdf

50 380 0
Role of myokines in the maintenance of whole body metabolic homeostasis pdf

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

MINERVA ENDOCRINOLOGICA EDIZIONI MINERVA MEDICA This provisional PDF corresponds to the article as it appeared upon acceptance A copyedited and fully formatted version will be made available soon The final version may contain major or minor changes Role of myokines in the maintenance of whole-body metabolic homeostasis Tatiana Y KOSTROMINOVA Minerva Endocrinol 2016 Mar 22 [Epub ahead of print] MINERVA ENDOCRINOLOGICA Rivista sulle Malattie del Sistema Endocrino pISSN 0391-1977 - eISSN 1827-1634 Article type: Review Article The online version of this article is located at http://www.minervamedica.it Subscription: Information about subscribing to Minerva Medica journals is online at: http://www.minervamedica.it/en/how-to-order-journals.php Reprints and permissions: For information about reprints and permissions send an email to: journals.dept@minervamedica.it - journals2.dept@minervamedica.it - journals6.dept@minervamedica.it COPYRIGHT© 2016 EDIZIONI MINERVA MEDICA Role of myokines in the maintenance of whole-body metabolic homeostasis Tatiana Y Kostrominova Department of Anatomy and Cell Biology, Indiana University School of Medicine-Northwest, Gary, IN, USA Corresponding author: Tatiana Y Kostrominova Department of Anatomy and Cell Biology, Indiana University School of MedicineNorthwest, Gary, IN, USA E-mail: tkostrom@iun.edu This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no ABSTRACT Obesity is reaching epidemic proportions in developed countries and is on the rise in developing countries Obesity-related changes in lipid and glucose metabolism predispose to the development of metabolic syndrome and type diabetes Skeletal muscle constitutes about 40 percent of total body weight and is unique compared to other muscle types since it is one of the most important organs for insulin-dependent glucose metabolism in humans Abnormalities in skeletal muscle lipid and glucose metabolism as well as abnormal accumulation of intramyocellular lipids could predispose for the development of type diabetes Skeletal muscle synthesizes and secretes factors with autocrine/paracrine/endocrine functions that can regulate skeletal muscle metabolism as well as affect other organs These factors secreted by skeletal muscle are called myokines Secretion and action of myokines is regulated by physiological conditions Some myokines have positive effect on metabolism, improving functions of multiple organs Yet, other myokines are released under pathological conditions and might exacerbate abnormal metabolic functions Expression and/or secretion of a number of myokines are regulated by exercise and therefore might mediate positive effects of physical activity on whole-body metabolism In the current review we summarized current knowledge on some of the myokines with important physiological functions in lipid and glucose metabolism A better understanding of the effects of myokines on whole-body metabolism can aid in development of the future pharmacologic therapies for counteracting the current worldwide obesity epidemic and obesity-mediated abnormalities This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no Key words: skeletal muscle, myokines, obesity, metabolism This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no TEXT Introduction Obesity has reached epidemic proportions worldwide and the rate of obesity continues to increase According to the World Health Organization, in 2014 about 39% (1.9 billion) of adults worldwide were overweight and 13% (over 600 million) were obese The World Obesity Federation estimates that if current trends continue, around billion adults worldwide will be overweight by the 2025 In order to combat the increasing rate of obesity it is essential to understand molecular mechanisms and factors that mediate the development of obesity-induced abnormalities Due to the extensive studies performed in the last two decades the critical role of factors secreted by adipose tissue (adipokines) in regulation of the whole-body lipid and glucose metabolism is now well established There are more than 600 factors secreted by adipocytes and circulating in the blood that could have autocrine/paracrine/endocrine functions [1] Some adipokines have beneficial effects on metabolism and improve insulin sensitivity, as well as lipid and glucose metabolism (adiponectin, vaspin, FGF21) Their secretion usually is decreased by obesity Other adpokines have negative effect on metabolism, promoting insulin resistance and dyslipidemia (resistin, visfatin, RBP4) and their secretion is increased by obesity Current medical textbooks describe adipose tissue as an adipokine-secreting endocrine organ, with a well-established role in whole-body metabolism Adipokines circulating in the blood can affect the function of many organs, including skeletal muscle A number of previous and current This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no studies are focused on using adipokines for the development of the treatment for obesity-induced abnormalities in humans Currently it is much less appreciated that skeletal muscle also can secrete many of the same factors with autocrine/paracrine/endocrine functions that are secreted by adipocytes, as well as some muscle-specific factors These factors secreted by skeletal muscle are called myokines This term, derived from Greek words for “muscle” and “motion” was proposed to describe factors with endocrine function that are produced and secreted by skeletal muscle cells [2] Skeletal muscle is the second largest organ of the human body It is the largest target organ for insulinstimulated glucose utilization Therefore, abnormalities in skeletal muscle glucose and lipid metabolism can lead to pronounced whole-body metabolic abnormalities Skeletal muscle-secreted myokines can regulate metabolism by endocrine mechanism acting on distant organs such as liver and adipose tissue (Figure 1) Myokines are also capable of regulating muscle functions by an autocrine mechanism as well as function of the located nearby tendon and bone tissues by a paracrine mechanism (Figure 1) A number of myokines have already been identified For many of myokines we have only limited knowledge We know that they potentially can be myokines but their functions are not yet studied Analysis of the secretory profile of human skeletal muscle cells has identified 305 proteins as potential myokines [3] Expression of some of these myokines is regulated by exercise Fifteen out of the 236 proteins first identified in cultured skeletal muscle cells were later shown to be significantly up-regulated in human skeletal muscle in vivo in response to strength training [4] This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no A number of myokines have been previously described in detail (FGF21, adiponectin, LIF, IL-6, CTRPs, irisin, apelin, lipocalins, etc.) although the mechanism regulating their secretion by skeletal muscle and their effect on other organs and whole-body metabolism are not yet completely elucidated Multiple studies suggest that myokine secretion is regulated by diverse physiological changes including obesity, cancer cachexia, insulin resistance and exercise Unlike adipokines, myokines are not yet mentioned in medical textbooks and are not discussed in medical school curricula As a result, the role of myokines as regulators of the whole-body lipid and glucose metabolism is currently underappreciated in the medical community A better understanding of the mechanisms by which myokines can affect obesity-induced abnormalities will help to develop future therapies In this review we will focus on selected myokines that, in our opinion, play crucial roles in whole-body lipid and glucose metabolism and have a potential to be used in the future as diagnostic or therapeutic tools to diminish obesity-induced abnormalities and prevent type diabetes Myostatin, also known as GDF-8, is a member of TGF beta family and is one of the first described myokines [5] Binding of myostatin to the activin receptor type IIB promotes skeletal muscle atrophy in many physiological conditions, including denervation and fasting Myostatin also inhibits activation of skeletal muscle-specific stem cells, satellite cells, in response to injury and slows down muscle differentiation [6] Myostatin mediates skeletal muscle atrophy through activation of the forkhead box O transcription factors (FoxO) leading to increased expression of muscle- This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no specific E3 ligases, atrogin-1 and MuRF1 (muscle RING-finger protein 1) E3 ligases target muscle myofibrillar and intracellular proteins for degradation through the ubiquitin–proteasome system Myostatin also increases skeletal muscle oxidative stress via NF-κB- and NADPH-mediated signaling cascades [7] Myostatin-null mice have significantly increased lean skeletal muscle mass when compared with wild type littermates [5] Increased muscle mass results from both hypertrophy (increase in muscle fiber size) and hyperplasia (proliferation of muscle stem cells to increase the number of muscle fibers) [5] Myostatin-null mice also are protected from dietinduced obesity, they have enhanced peripheral tissue fatty acid oxidation and increased thermogenesis [8] Myostatin-induced catabolic processes mediate skeletal muscle atrophy in cancer cachexia [9] Myostatin expression can be regulated by endocrine hormones In rats hypothyroidism is associated with increased myostatin mRNA expression [10] It is currently unclear whether hypothyroidism in humans is also associated with increased myostatin levels in skeletal muscle The endocrine function of myostatin could be mediated by its effects on adipose tissue Myostatin treatment has been shown to increase proliferation of adipocytes and inhibit their differentiation [11] Myostatin treatment also influenced the expression and secretion of adiponectin, resistin, and visfatin by adipocytes [11] Brain-derived neurotrophic factor (BDNF) is a myokine that affects myogenesis and skeletal muscle regeneration [12] The expression of BDNF is high in myoblasts but it is decreased in differentiated myotubes [13] BDNF effects are This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no mediated by binding to specific cell surface receptors Human myocytes express p75NTR and not TrkB BDNF receptors [14] BDNF is expressed at different levels in fast and slow muscle fibers Mostly slow rat soleus muscle has 2-fold higher levels of BDNF expression than mostly fast gastrocnemius muscle [15] BDNF has beneficial effects on nerve growth and regeneration via paracrine mechanism [16] Following denervation skeletal muscle increases BDNF expression and retrograde transport of BDNF to spinal cord [16] BDNF also affects whole-body metabolism via autocrine and endocrine mechanisms Systemic treatment of mice with BDNF reduces total food intake and inhibits weight gain [17] These effects are mediated by increased GLUT4 expression in skeletal muscle [17] In obese diabetic mice BDNF treatment enhances glucose utilization in skeletal muscle and brown adipose tissue [18] Insulin-like growth factor (IGF-1) is released by skeletal muscle and affects skeletal muscle growth Mutant mice with reduced IGF-1 content in muscle are ~30% smaller, have reduced levels of circulating IGF-1, have smaller muscles and decreased bone mineral density [19] Growth defects are diminished by administration of recombinant IGF-1 [19] IGF-1 regulates whole-body metabolism Skeletal muscle-specific inactivation of IFG-1 receptors leads to the development of insulin resistance [20] High fat diet-induced obesity in mice reduces expression of IGF-1 in intact muscles and diminishes up-regulation of IGF-1 in response to muscle injury [21] This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no Fibroblast Growth Factor (FGF-2), also known as basic FGF, is a myokine that has pleiotropic effects in a range of cell types During myogenic differentiation expression of FGFs and FGF receptors are downregulated [22] FGF-2 secreted by muscle cells may act as a paracrine and autocrine regulator of skeletal muscle development in vivo [23] During ischemia, increased expression of FGF-2 in skeletal muscle promotes angiogenesis via its paracrine effects on blood vessels [24] Treatment with FGF-2 stimulates proliferation of tendon cells [25] suggesting that skeletal muscle released FGF-2 may have paracrine effects on tendon FGF-2 also plays an important role in bone formation and remodeling Mechanical wounding of muscle cells increases release of FGF-2 into media [26] and may have paracrine effect on tendon and bone Fibroblast growth factor 21 (FGF21) is a well described adipocytokine/ hepatokine/ myokine with glucose and lipid-lowering properties The liver is considered the major source of the plasma FGF21 Under normal physiological conditions the level of hepatic FGF21 expression is low but it increases during prolonged fasting, liver injury, exposure to toxic chemicals and viral infections [27] In mice chronic infusion of FGF21 improves insulin responsiveness due to the reduced diacylglycerol content and reduced protein kinase C activation in skeletal muscle [28] In human clinical trials FGF21 treatment of obese type diabetic patients improved total body weight, dyslipidemia, insulin sensitivity and adiponectin levels [29] [27] FGF21 works through the FGF receptors (FGFR1 and FGFR2) and requires transmembrane protein beta-Klotho (KLB) as a co-receptor for activation of This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 13 Miura P, Amirouche A, Clow C, Belanger G, Jasmin BJ (2012) Brain-derived neurotrophic factor expression is repressed during myogenic differentiation by miR-206 J Neurochem 120: 230-238 14 Colombo E, Bedogni F, Lorenzetti I, Landsberger N, Previtali SC, et al (2013) Autocrine and immune cell-derived BDNF in human skeletal muscle: implications for myogenesis and tissue regeneration J Pathol 231: 190-198 15 Ogborn DI, Gardiner PF (2010) Effects of exercise and muscle type on BDNF, NT-4/5, and TrKB expression in skeletal muscle Muscle Nerve 41: 385-391 16 Gao L, Li LH, Xing RX, Ou S, Liu GD, et al (2012) Gastrocnemius-derived BDNF promotes motor function recovery in spinal cord transected rats Growth Factors 30: 167-175 17 Suwa M, Yamamoto KI, Nakano H, Sasaki H, Radak Z, et al (2010) Brainderived neurotrophic factor treatment increases the skeletal muscle glucose transporter protein expression in mice Physiol Res 59: 619-623 18 Yamanaka M, Tsuchida A, Nakagawa T, Nonomura T, Ono-Kishino M, et al (2007) Brain-derived neurotrophic factor enhances glucose utilization in peripheral tissues of diabetic mice Diabetes Obes Metab 9: 59-64 19 Barton ER, Park S, James JK, Makarewich CA, Philippou A, et al (2012) Deletion of muscle GRP94 impairs both muscle and body growth by inhibiting local IGF production FASEB J 26: 3691-3702 20 Fernandez AM, Kim JK, Yakar S, Dupont J, Hernandez-Sanchez C, et al (2001) Functional inactivation of the IGF-I and insulin receptors in skeletal muscle causes type diabetes Genes Dev 15: 1926-1934 21 Brown LA, Lee DE, Patton JF, Perry RA, Jr., Brown JL, et al (2015) Dietinduced obesity alters anabolic signalling in mice at the onset of skeletal muscle regeneration Acta Physiol (Oxf) 215: 46-57 22 Moore JW, Dionne C, Jaye M, Swain JL (1991) The mRNAs encoding acidic FGF, basic FGF and FGF receptor are coordinately downregulated during myogenic differentiation Development 111: 741-748 23 Hannon K, Kudla AJ, McAvoy MJ, Clase KL, Olwin BB (1996) Differentially expressed fibroblast growth factors regulate skeletal muscle development through autocrine and paracrine mechanisms J Cell Biol 132: 1151-1159 24 Walgenbach KJ, Gratas C, Shestak KC, Becker D (1995) Ischaemia-induced expression of bFGF in normal skeletal muscle: a potential paracrine mechanism for mediating angiogenesis in ischaemic skeletal muscle Nat Med 1: 453-459 25 Takahashi S, Nakajima M, Kobayashi M, Wakabayashi I, Miyakoshi N, et al (2002) Effect of recombinant basic fibroblast growth factor (bFGF) on This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 35 fibroblast-like cells from human rotator cuff tendon Tohoku J Exp Med 198: 207-214 26 Hamrick MW, McNeil PL, Patterson SL (2010) Role of muscle-derived growth factors in bone formation J Musculoskelet Neuronal Interact 10: 64-70 27 Kharitonenkov A, Adams AC (2014) Inventing new medicines: The FGF21 story Mol Metab 3: 221-229 28 Camporez JP, Jornayvaz FR, Petersen MC, Pesta D, Guigni BA, et al (2013) Cellular mechanisms by which FGF21 improves insulin sensitivity in male mice Endocrinology 154: 3099-3109 29 Gaich G, Chien JY, Fu H, Glass LC, Deeg MA, et al (2013) The effects of LY2405319, an FGF21 analog, in obese human subjects with type diabetes Cell Metab 18: 333-340 30 Smith R, Duguay A, Bakker A, Li P, Weiszmann J, et al (2013) FGF21 can be mimicked in vitro and in vivo by a novel anti-FGFR1c/beta-Klotho bispecific protein PLoS One 8: e61432 31 Izumiya Y, Bina HA, Ouchi N, Akasaki Y, Kharitonenkov A, et al (2008) FGF21 is an Akt-regulated myokine FEBS Lett 582: 3805-3810 32 Hojman P, Pedersen M, Nielsen AR, Krogh-Madsen R, Yfanti C, et al (2009) Fibroblast growth factor-21 is induced in human skeletal muscles by hyperinsulinemia Diabetes 58: 2797-2801 33 Keipert S, Ost M, Johann K, Imber F, Jastroch M, et al (2014) Skeletal muscle mitochondrial uncoupling drives endocrine cross-talk through the induction of FGF21 as a myokine Am J Physiol Endocrinol Metab 306: E469-482 34 Mashili FL, Austin RL, Deshmukh AS, Fritz T, Caidahl K, et al (2011) Direct effects of FGF21 on glucose uptake in human skeletal muscle: implications for type diabetes and obesity Diabetes Metab Res Rev 27: 286-297 35 Voigt A, Katterle Y, Kahle M, Kluge R, Schurmann A, et al (2015) Skeletal muscle mitochondrial uncoupling prevents diabetes but not obesity in NZO mice, a model for polygenic diabesity Genes Nutr 10: 57 36 Ribas F, Villarroya J, Hondares E, Giralt M, Villarroya F (2014) FGF21 expression and release in muscle cells: involvement of MyoD and regulation by mitochondria-driven signalling Biochem J 463: 191-199 37 Guridi M, Tintignac LA, Lin S, Kupr B, Castets P, et al (2015) Activation of mTORC1 in skeletal muscle regulates whole-body metabolism through FGF21 Sci Signal 8: ra113 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 36 38 Tsai S, Sitzmann JM, Dastidar SG, Rodriguez AA, Vu SL, et al (2015) Musclespecific 4E-BP1 signaling activation improves metabolic parameters during aging and obesity J Clin Invest 125: 2952-2964 39 Crooks DR, Natarajan TG, Jeong SY, Chen C, Park SY, et al (2014) Elevated FGF21 secretion, PGC-1alpha and ketogenic enzyme expression are hallmarks of iron-sulfur cluster depletion in human skeletal muscle Hum Mol Genet 23: 24-39 40 Lee MS, Choi SE, Ha ES, An SY, Kim TH, et al (2012) Fibroblast growth factor21 protects human skeletal muscle myotubes from palmitate-induced insulin resistance by inhibiting stress kinase and NF-kappaB Metabolism 61: 11421151 41 Holland WL, Adams AC, Brozinick JT, Bui HH, Miyauchi Y, et al (2013) An FGF21-adiponectin-ceramide axis controls energy expenditure and insulin action in mice Cell Metab 17: 790-797 42 Joki Y, Ohashi K, Yuasa D, Shibata R, Ito M, et al (2015) FGF21 attenuates pathological myocardial remodeling following myocardial infarction through the adiponectin-dependent mechanism Biochem Biophys Res Commun 459: 124-130 43 Berti L, Irmler M, Zdichavsky M, Meile T, Bohm A, et al (2015) Fibroblast growth factor 21 is elevated in metabolically unhealthy obesity and affects lipid deposition, adipogenesis, and adipokine secretion of human abdominal subcutaneous adipocytes Mol Metab 4: 519-527 44 Wang W, Xing W, Zhang H, Ding M, Shang L, et al (2013) Reduced highmolecular-weight adiponectin is an independent risk factor for cardiovascular lesions in hypercholesterolaemic patients Clin Endocrinol (Oxf) 78: 539-544 45 Goto A, Ohno Y, Ikuta A, Suzuki M, Ohira T, et al (2013) Up-regulation of adiponectin expression in antigravitational soleus muscle in response to unloading followed by reloading, and functional overloading in mice PLoS One 8: e81929 46 Delaigle AM, Jonas JC, Bauche IB, Cornu O, Brichard SM (2004) Induction of adiponectin in skeletal muscle by inflammatory cytokines: in vivo and in vitro studies Endocrinology 145: 5589-5597 47 Jortay J, Senou M, Abou-Samra M, Noel L, Robert A, et al (2012) Adiponectin and skeletal muscle: pathophysiological implications in metabolic stress Am J Pathol 181: 245-256 48 Mao X, Kikani CK, Riojas RA, Langlais P, Wang L, et al (2006) APPL1 binds to adiponectin receptors and mediates adiponectin signalling and function Nat Cell Biol 8: 516-523 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 37 49 Di Chiara T, Licata A, Argano C, Duro G, Corrao S, et al (2013) Plasma adiponectin: A contributing factor for cardiac changes in visceral obesityassociated hypertension Blood Press 50 Ji ZY, Li HF, Lei Y, Rao YW, Tan ZX, et al (2015) Association of adiponectin gene polymorphisms with an elevated risk of diabetic peripheral neuropathy in type diabetes patients J Diabetes Complications 29: 887-892 51 Hou M, Chu Z, Liu T, Lv H, Sun L, et al (2015) A high-fat maternal diet decreases adiponectin receptor-1 expression in offspring J Matern Fetal Neonatal Med 28: 216-221 52 Sattar AA, Sattar R (2012) Globular adiponectin activates Akt in cultured myocytes Biochem Biophys Res Commun 424: 753-757 53 Qiao L, Kinney B, Yoo HS, Lee B, Schaack J, et al (2012) Adiponectin increases skeletal muscle mitochondrial biogenesis by suppressing mitogenactivated protein kinase phosphatase-1 Diabetes 61: 1463-1470 54 Choudhary S, Sinha S, Zhao Y, Banerjee S, Sathyanarayana P, et al (2011) NF-kappaB-inducing kinase (NIK) mediates skeletal muscle insulin resistance: blockade by adiponectin Endocrinology 152: 3622-3627 55 Abou-Samra M, Lecompte S, Schakman O, Noel L, Many MC, et al (2015) Involvement of adiponectin in the pathogenesis of dystrophinopathy Skelet Muscle 5: 25 56 Delaigle AM, Senou M, Guiot Y, Many MC, Brichard SM (2006) Induction of adiponectin in skeletal muscle of type diabetic mice: In vivo and in vitro studies Diabetologia 49: 1311-1323 57 Xu A, Sweeney G (2015) Emerging role of autophagy in mediating widespread actions of ADIPOQ/adiponectin Autophagy 11: 723-724 58 Okada-Iwabu M, Iwabu M, Ueki K, Yamauchi T, Kadowaki T (2015) Perspective of Small-Molecule AdipoR Agonist for Type Diabetes and Short Life in Obesity Diabetes Metab J 39: 363-372 59 Schaffler A, Buechler C (2012) CTRP family: linking immunity to metabolism Trends Endocrinol Metab 23: 194-204 60 Seldin MM, Peterson JM, Byerly MS, Wei Z, Wong GW (2012) Myonectin (CTRP15), a novel myokine that links skeletal muscle to systemic lipid homeostasis J Biol Chem 287: 11968-11980 61 Wei Z, Lei X, Seldin MM, Wong GW (2012) Endopeptidase cleavage generates a functionally distinct isoform of C1q/tumor necrosis factor-related protein-12 (CTRP12) with an altered oligomeric state and signaling specificity J Biol Chem 287: 35804-35814 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 38 62 Kim KY, Kim HY, Kim JH, Lee CH, Kim DH, et al (2006) Tumor necrosis factoralpha and interleukin-1beta increases CTRP1 expression in adipose tissue FEBS Lett 580: 3953-3960 63 Han S, Park JS, Lee S, Jeong AL, Oh KS, et al (2015) CTRP1 protects against diet-induced hyperglycemia by enhancing glycolysis and fatty acid oxidation J Nutr Biochem 64 Maeda T, Abe M, Kurisu K, Jikko A, Furukawa S (2001) Molecular cloning and characterization of a novel gene, CORS26, encoding a putative secretory protein and its possible involvement in skeletal development J Biol Chem 276: 3628-3634 65 Otani M, Furukawa S, Wakisaka S, Maeda T (2015) A novel adipokine C1q/TNF-related protein is expressed in developing skeletal muscle and controls myoblast proliferation and differentiation Mol Cell Biochem 409: 271-282 66 Akiyama H, Otani M, Sato S, Toyosawa S, Furukawa S, et al (2013) A novel adipokine C1q/TNF-related protein (CTRP1) regulates chondrocyte proliferation and maturation through the ERK1/2 signaling pathway Mol Cell Endocrinol 369: 63-71 67 Akiyama H, Furukawa S, Wakisaka S, Maeda T (2007) CTRP3/cartducin promotes proliferation and migration of endothelial cells Mol Cell Biochem 304: 243-248 68 Peterson JM, Wei Z, Wong GW (2010) C1q/TNF-related protein-3 (CTRP3), a novel adipokine that regulates hepatic glucose output J Biol Chem 285: 39691-39701 69 Kopp A, Bala M, Weigert J, Buchler C, Neumeier M, et al (2010) Effects of the new adiponectin paralogous protein CTRP-3 and of LPS on cytokine release from monocytes of patients with type diabetes mellitus Cytokine 49: 51-57 70 Park SY, Choi JH, Ryu HS, Pak YK, Park KS, et al (2009) C1q tumor necrosis factor alpha-related protein isoform is increased in mitochondrial DNAdepleted myocytes and activates AMP-activated protein kinase J Biol Chem 284: 27780-27789 71 Lim S, Choi SH, Koo BK, Kang SM, Yoon JW, et al (2012) Effects of aerobic exercise training on C1q tumor necrosis factor alpha-related protein isoform (myonectin): association with insulin resistance and mitochondrial DNA density in women J Clin Endocrinol Metab 97: E88-93 72 Wei Z, Peterson JM, Lei X, Cebotaru L, Wolfgang MJ, et al (2012) C1q/TNFrelated protein-12 (CTRP12), a novel adipokine that improves insulin sensitivity and glycemic control in mouse models of obesity and diabetes J Biol Chem 287: 10301-10315 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 39 73 Enomoto T, Ohashi K, Shibata R, Kambara T, Uemura Y, et al (2013) Transcriptional regulation of an insulin-sensitizing adipokine adipolin/CTRP12 in adipocytes by Kruppel-like factor 15 PLoS One 8: e83183 74 Fathi R, Ghanbari-Niaki A, Kraemer RR, Talebi-Garakani E, Saghebjoo M (2010) The effect of exercise intensity on plasma and tissue acyl ghrelin concentrations in fasted rats Regul Pept 165: 133-137 75 Filigheddu N, Gnocchi VF, Coscia M, Cappelli M, Porporato PE, et al (2007) Ghrelin and des-acyl ghrelin promote differentiation and fusion of C2C12 skeletal muscle cells Mol Biol Cell 18: 986-994 76 Yu AP, Pei XM, Sin TK, Yip SP, Yung BY, et al (2014) Acylated and unacylated ghrelin inhibit doxorubicin-induced apoptosis in skeletal muscle Acta Physiol (Oxf) 211: 201-213 77 von Haehling S, Anker SD (2015) Treatment of cachexia: An overview of recent developments Int J Cardiol 184: 736-742 78 Pietra C, Takeda Y, Tazawa-Ogata N, Minami M, Yuanfeng X, et al (2014) Anamorelin HCl (ONO-7643), a novel ghrelin receptor agonist, for the treatment of cancer anorexia-cachexia syndrome: preclinical profile J Cachexia Sarcopenia Muscle 5: 329-337 79 Tam BT, Pei XM, Yung BY, Yip SP, Chan LW, et al (2015) Unacylated ghrelin restores insulin and autophagic signaling in skeletal muscle of diabetic mice Pflugers Arch 467: 2555-2569 80 Barazzoni R, Zanetti M, Semolic A, Cattin MR, Pirulli A, et al (2011) High-fat diet with acyl-ghrelin treatment leads to weight gain with low inflammation, high oxidative capacity and normal triglycerides in rat muscle PLoS One 6: e26224 81 Barazzoni R, Bosutti A, Stebel M, Cattin MR, Roder E, et al (2005) Ghrelin regulates mitochondrial-lipid metabolism gene expression and tissue fat distribution in liver and skeletal muscle Am J Physiol Endocrinol Metab 288: E228-235 82 Nishizawa H, Matsuda M, Yamada Y, Kawai K, Suzuki E, et al (2004) Musclin, a novel skeletal muscle-derived secretory factor J Biol Chem 279: 1939119395 83 Kita S, Nishizawa H, Okuno Y, Tanaka M, Yasui A, et al (2009) Competitive binding of musclin to natriuretic peptide receptor with atrial natriuretic peptide J Endocrinol 201: 287-295 84 Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, et al (2012) A PGC1-alphadependent myokine that drives brown-fat-like development of white fat and thermogenesis Nature 481: 463-468 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 40 85 Moreno-Navarrete JM, Ortega F, Serrano M, Guerra E, Pardo G, et al (2013) Irisin is expressed and produced by human muscle and adipose tissue in association with obesity and insulin resistance J Clin Endocrinol Metab 98: E769-778 86 Zhang Y, Li R, Meng Y, Li S, Donelan W, et al (2014) Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling Diabetes 63: 514-525 87 Vaughan RA, Gannon NP, Mermier CM, Conn CA (2015) Irisin, a unique noninflammatory myokine in stimulating skeletal muscle metabolism J Physiol Biochem 71: 679-689 88 Zhang C, Ding Z, Lv G, Li J, Zhou P, et al (2014) Lower irisin level in patients with type diabetes mellitus: A case-control study and meta-analysis J Diabetes 89 Gannon NP, Vaughan RA, Garcia-Smith R, Bisoffi M, Trujillo KA (2015) Effects of the exercise-inducible myokine irisin on malignant and non-malignant breast epithelial cell behavior in vitro Int J Cancer 136: E197-202 90 Krzysik-Walker SM, Hadley JA, Pesall JE, McFarland DC, Vasilatos-Younken R, et al (2011) Nampt/visfatin/PBEF affects expression of myogenic regulatory factors and is regulated by interleukin-6 in chicken skeletal muscle cells Comp Biochem Physiol A Mol Integr Physiol 159: 413-421 91 Lee JO, Kim N, Lee HJ, Lee YW, Kim JK, et al (2015) Visfatin, a novel adipokine, stimulates glucose uptake through the Ca2 +-dependent AMPKp38 MAPK pathway in C2C12 skeletal muscle cells J Mol Endocrinol 54: 251-262 92 Harasim E, Chabowski A, Gorski J (2011) Lack of downstream insulin-mimetic effects of visfatin/eNAMPT on glucose and fatty acid metabolism in skeletal muscles Acta Physiol (Oxf) 202: 21-28 93 Yue P, Jin H, Aillaud M, Deng AC, Azuma J, et al (2010) Apelin is necessary for the maintenance of insulin sensitivity Am J Physiol Endocrinol Metab 298: E59-67 94 Dray C, Knauf C, Daviaud D, Waget A, Boucher J, et al (2008) Apelin stimulates glucose utilization in normal and obese insulin-resistant mice Cell Metab 8: 437-445 95 Higuchi K, Masaki T, Gotoh K, Chiba S, Katsuragi I, et al (2007) Apelin, an APJ receptor ligand, regulates body adiposity and favors the messenger ribonucleic acid expression of uncoupling proteins in mice Endocrinology 148: 2690-2697 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 41 96 Yang H, Zhao L, Zhang J, Tang CS, Qi YF, et al (2015) Effect of Treadmill Running on Apelin and APJ Expression in Adipose Tissue and Skeletal Muscle in Rats Fed a High-fat Diet Int J Sports Med 36: 535-541 97 Besse-Patin A, Montastier E, Vinel C, Castan-Laurell I, Louche K, et al (2013) Effect of endurance training on skeletal muscle myokine expression in obese men: identification of apelin as a novel myokine Int J Obes (Lond) 98 Bertrand C, Pignalosa A, Wanecq E, Rancoule C, Batut A, et al (2013) Effects of dietary eicosapentaenoic acid (EPA) supplementation in high-fat fed mice on lipid metabolism and apelin/APJ system in skeletal muscle PLoS One 8: e78874 99 Cavallo MG, Sentinelli F, Barchetta I, Costantino C, Incani M, et al (2012) Altered glucose homeostasis is associated with increased serum apelin levels in type diabetes mellitus PLoS One 7: e51236 100 Hui X, Zhu W, Wang Y, Lam KS, Zhang J, et al (2009) Major urinary protein-1 increases energy expenditure and improves glucose intolerance through enhancing mitochondrial function in skeletal muscle of diabetic mice J Biol Chem 284: 14050-14057 101 Cho KW, Zhou Y, Sheng L, Rui L (2011) Lipocalin-13 regulates glucose metabolism by both insulin-dependent and insulin-independent mechanisms Mol Cell Biol 31: 450-457 102 Mauer J, Denson JL, Bruning JC (2015) Versatile functions for IL-6 in metabolism and cancer Trends Immunol 36: 92-101 103 Al-Khalili L, Bouzakri K, Glund S, Lonnqvist F, Koistinen HA, et al (2006) Signaling specificity of interleukin-6 action on glucose and lipid metabolism in skeletal muscle Mol Endocrinol 20: 3364-3375 104 Carey AL, Steinberg GR, Macaulay SL, Thomas WG, Holmes AG, et al (2006) Interleukin-6 increases insulin-stimulated glucose disposal in humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated protein kinase Diabetes 55: 2688-2697 105 Guerra J, Ferrer B, Giralt M, Comes G, Carrasco J, et al (2015) Muscular interleukin-6 differentially regulates skeletal muscle adaptation to high-fat diet in a sex-dependent manner Cytokine 74: 145-151 106 Nicola NA, Babon JJ (2015) Leukemia inhibitory factor (LIF) Cytokine Growth Factor Rev 26: 533-544 107 Broholm C, Laye MJ, Brandt C, Vadalasetty R, Pilegaard H, et al (2011) LIF is a contraction-induced myokine stimulating human myocyte proliferation J Appl Physiol (1985) 111: 251-259 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 42 108 Williams RL, Hilton DJ, Pease S, Willson TA, Stewart CL, et al (1988) Myeloid leukaemia inhibitory factor maintains the developmental potential of embryonic stem cells Nature 336: 684-687 109 Reardon KA, Kapsa RM, Davis J, Kornberg AJ, Austin L, et al (2000) Increased levels of leukemia inhibitory factor mRNA in muscular dystrophy and human muscle trauma Muscle Nerve 23: 962-966 110 Austin L, Bower JJ, Bennett TM, Lynch GS, Kapsa R, et al (2000) Leukemia inhibitory factor ameliorates muscle fiber degeneration in the mdx mouse Muscle Nerve 23: 1700-1705 111 Curtis R, Scherer SS, Somogyi R, Adryan KM, Ip NY, et al (1994) Retrograde axonal transport of LIF is increased by peripheral nerve injury: correlation with increased LIF expression in distal nerve Neuron 12: 191-204 112 Poulton IJ, McGregor NE, Pompolo S, Walker EC, Sims NA (2012) Contrasting roles of leukemia inhibitory factor in murine bone development and remodeling involve region-specific changes in vascularization J Bone Miner Res 27: 586-595 113 Brandt N, O'Neill HM, Kleinert M, Schjerling P, Vernet E, et al (2015) Leukemia inhibitory factor increases glucose uptake in mouse skeletal muscle Am J Physiol Endocrinol Metab 309: E142-153 114 Broholm C, Brandt C, Schultz NS, Nielsen AR, Pedersen BK, et al (2012) Deficient leukemia inhibitory factor signaling in muscle precursor cells from patients with type diabetes Am J Physiol Endocrinol Metab 303: E283-292 115 Skaper SD, Selak I, Manthorpe M, Varon S (1984) Chemically defined requirements for the survival of cultured 8-day chick embryo ciliary ganglion neurons Brain Res 302: 281-290 116 Johnson RW, White JD, Walker EC, Martin TJ, Sims NA (2014) Myokines (muscle-derived cytokines and chemokines) including ciliary neurotrophic factor (CNTF) inhibit osteoblast differentiation Bone 64: 47-56 117 Ip NY, McClain J, Barrezueta NX, Aldrich TH, Pan L, et al (1993) The alpha component of the CNTF receptor is required for signaling and defines potential CNTF targets in the adult and during development Neuron 10: 89102 118 Larsen JV, Hansen M, Moller B, Madsen P, Scheller J, et al (2010) Sortilin facilitates signaling of ciliary neurotrophic factor and related helical type cytokines targeting the gp130/leukemia inhibitory factor receptor beta heterodimer Mol Cell Biol 30: 4175-4187 119 Hiatt K, Lewis D, Shew M, Bijangi-Vishehsaraei K, Halum S (2014) Ciliary neurotrophic factor (CNTF) promotes skeletal muscle progenitor cell (MPC) This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 43 viability via the phosphatidylinositol 3-kinase-Akt pathway J Tissue Eng Regen Med 8: 963-968 120 Huang S, Wang F, Hong G, Wan S, Kang H (2002) Protective effects of ciliary neurotrophic factor on denervated skeletal muscle J Huazhong Univ Sci Technolog Med Sci 22: 148-151 121 Steinberg GR, Watt MJ, Ernst M, Birnbaum MJ, Kemp BE, et al (2009) Ciliary neurotrophic factor stimulates muscle glucose uptake by a PI3-kinasedependent pathway that is impaired with obesity Diabetes 58: 829-839 122 Huang G, Buckler-Pena D, Nauta T, Singh M, Asmar A, et al (2013) Insulin responsiveness of glucose transporter in 3T3-L1 cells depends on the presence of sortilin Mol Biol Cell 24: 3115-3122 123 Tsuchiya Y, Hatakeyama H, Emoto N, Wagatsuma F, Matsushita S, et al (2010) Palmitate-induced down-regulation of sortilin and impaired GLUT4 trafficking in C2C12 myotubes J Biol Chem 285: 34371-34381 124 Kaddai V, Jager J, Gonzalez T, Najem-Lendom R, Bonnafous S, et al (2009) Involvement of TNF-alpha in abnormal adipocyte and muscle sortilin expression in obese mice and humans Diabetologia 52: 932-940 125 Watt MJ, Dzamko N, Thomas WG, Rose-John S, Ernst M, et al (2006) CNTF reverses obesity-induced insulin resistance by activating skeletal muscle AMPK Nat Med 12: 541-548 126 Pennica D, Shaw KJ, Swanson TA, Moore MW, Shelton DL, et al (1995) Cardiotrophin-1 Biological activities and binding to the leukemia inhibitory factor receptor/gp130 signaling complex J Biol Chem 270: 10915-10922 127 Pennica D, Swanson TA, Shaw KJ, Kuang WJ, Gray CL, et al (1996) Human cardiotrophin-1: protein and gene structure, biological and binding activities, and chromosomal localization Cytokine 8: 183-189 128 Oppenheim RW, Wiese S, Prevette D, Armanini M, Wang S, et al (2001) Cardiotrophin-1, a muscle-derived cytokine, is required for the survival of subpopulations of developing motoneurons J Neurosci 21: 1283-1291 129 Rind HB, Butowt R, von Bartheld CS (2005) Synaptic targeting of retrogradely transported trophic factors in motoneurons: comparison of glial cell linederived neurotrophic factor, brain-derived neurotrophic factor, and cardiotrophin-1 with tetanus toxin J Neurosci 25: 539-549 130 Holtmann B, Wiese S, Samsam M, Grohmann K, Pennica D, et al (2005) Triple knock-out of CNTF, LIF, and CT-1 defines cooperative and distinct roles of these neurotrophic factors for motoneuron maintenance and function J Neurosci 25: 1778-1787 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 44 131 Moreno-Aliaga MJ, Perez-Echarri N, Marcos-Gomez B, Larequi E, Gil-Bea FJ, et al (2011) Cardiotrophin-1 is a key regulator of glucose and lipid metabolism Cell Metab 14: 242-253 132 Castano D, Larequi E, Belza I, Astudillo AM, Martinez-Anso E, et al (2014) Cardiotrophin-1 eliminates hepatic steatosis in obese mice by mechanisms involving AMPK activation J Hepatol 60: 1017-1025 133 Malik N, Kallestad JC, Gunderson NL, Austin SD, Neubauer MG, et al (1989) Molecular cloning, sequence analysis, and functional expression of a novel growth regulator, oncostatin M Mol Cell Biol 9: 2847-2853 134 Sun D, Li S, Wu H, Zhang M, Zhang X, et al (2015) Oncostatin M (OSM) protects against cardiac ischaemia/reperfusion injury in diabetic mice by regulating apoptosis, mitochondrial biogenesis and insulin sensitivity J Cell Mol Med 19: 1296-1307 135 Xiao F, Wang H, Fu X, Li Y, Ma K, et al (2011) Oncostatin M inhibits myoblast differentiation and regulates muscle regeneration Cell Res 21: 350-364 136 Hojman P, Dethlefsen C, Brandt C, Hansen J, Pedersen L, et al (2011) Exercise-induced muscle-derived cytokines inhibit mammary cancer cell growth Am J Physiol Endocrinol Metab 301: E504-510 137 Komori T, Tanaka M, Furuta H, Akamizu T, Miyajima A, et al (2015) Oncostatin M is a potential agent for the treatment of obesity and related metabolic disorders: a study in mice Diabetologia 58: 1868-1876 138 Scheler M, Irmler M, Lehr S, Hartwig S, Staiger H, et al (2013) Cytokine response of primary human myotubes in an in vitro exercise model Am J Physiol Cell Physiol 305: C877-886 139 Shin KO, Bae JY, Woo J, Jang KS, Kim KS, et al (2015) The effect of exercise on expression of myokine and angiogenesis mRNA in skeletal muscle of high fat diet induced obese rat J Exerc Nutrition Biochem 19: 91-98 140 Benrick A, Wallenius V, Asterholm IW (2012) Interleukin-6 mediates exerciseinduced increase in insulin sensitivity in mice Exp Physiol 97: 1224-1235 141 Matthews VB, Allen TL, Risis S, Chan MH, Henstridge DC, et al (2010) Interleukin-6-deficient mice develop hepatic inflammation and systemic insulin resistance Diabetologia 53: 2431-2441 142 Cuppini R, Sartini S, Agostini D, Guescini M, Ambrogini P, et al (2007) Bdnf expression in rat skeletal muscle after acute or repeated exercise Arch Ital Biol 145: 99-110 143 Breen EC, Johnson EC, Wagner H, Tseng HM, Sung LA, et al (1996) Angiogenic growth factor mRNA responses in muscle to a single bout of exercise J Appl Physiol (1985) 81: 355-361 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 45 144 Cho JK, Kim S, Hong HR, Yoon JH, Kang H (2015) Exercise Training Improves Whole Body Insulin Resistance via Adiponectin Receptor Int J Sports Med 145 Covington JD, Tam CS, Bajpeyi S, Galgani JE, Noland RC, et al (2015) Myokine Expression in Muscle and Myotubes in Response to Exercise Stimulation Med Sci Sports Exerc 146 Broholm C, Mortensen OH, Nielsen S, Akerstrom T, Zankari A, et al (2008) Exercise induces expression of leukaemia inhibitory factor in human skeletal muscle J Physiol 586: 2195-2201 147 Alfieri A, Martone D, Randers MB, Labruna G, Mancini A, et al (2015) Effects of long-term football training on the expression profile of genes involved in muscle oxidative metabolism Mol Cell Probes 29: 43-47 148 Loffler D, Muller U, Scheuermann K, Friebe D, Gesing J, et al (2015) Serum irisin levels are regulated by acute strenuous exercise J Clin Endocrinol Metab 100: 1289-1299 149 Tsuchiya Y, Ando D, Takamatsu K, Goto K (2015) Resistance exercise induces a greater irisin response than endurance exercise Metabolism 64: 10421050 Acknowledgements: The author is grateful to Dr Stephen Echtenkamp for the helpful discussions and advice during preparation of this manuscript This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 46 Funding: This work was supported by the funds from the IUSM-NW Conflicts of interest: None This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 47 TITLES OF FIGURES Figure Skeletal muscle-secreted myokines can work via autocrine, paracrine, and endocrine mechanisms Abbreviations: BDNF, brain-derived neurotrophic factor; IGF-1, insulin-like growth factor 1; FGF-2, fibroblast growth factor 2; FGF21, fibroblast growth factor 21; CTRP, C1q/TNF-related proteins; LCN13, lipocalin 13; IL-6, interleukin 6; LIF, leukemia inhibitory factor; CNTF, ciliary neurotrophic factor; CT-1, cardiotrophin-1 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no 48 This document is protected by international copyright laws No additional reproduction is authorized It is permitted for personal use to download and save only one file and print only one copy of this Article It is not permitted to make additional copies (either sporadically or systematically, either printed or electronic) of the Article for any purpose It is not permitted to distribute the electronic copy of the article through online internet and/or intranet file sharing systems, electronic mailing or any other means which may allow access to the Article The use of all or an part of the Article for any Commercial Use is not permitted The creation of derivative works from the Article is not permitted The production of reprints for personal or commercial use is no

Ngày đăng: 22/07/2016, 08:48

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan