Myokine

From Infogalactic: the planetary knowledge core
Jump to: navigation, search

Lua error in package.lua at line 80: module 'strict' not found.

A myokine is one of several hundred cytokines or other small proteins (~5–20 kDa) and proteoglycan peptides that are produced and released by muscle cells (myocytes) in response to muscular contractions.[1] They have autocrine, paracrine and/or endocrine effects; their systemic effects occur at picomolar concentrations.[2][3]

Receptors for myokines are found on muscle, fat, liver, pancreas, bone, heart, immune, and brain cells. The location of these receptors explain the fact that myokines have multiple functions. Foremost, they are involved in exercise-associated metabolic changes, as well as in the metabolic changes following training adaptation.[1] They also participate in tissue regeneration and repair, maintenance of healthy bodily functioning, immunomodulation; and cell signaling, expression and differentiation.[1]

History

The present definition of the term myokine is attributed to Dr. Bente Klarlund Pedersen et al., who suggested its use in 2003.[4] In 2008, the first myokine, myostatin, was identified.[3][5] The gp130 receptor cytokine IL-6 (Interleukin 6) was the first myokine found to be secreted into the blood stream in response to muscle contractions.[6]

Repetitive skeletal muscle contractions

There is an emerging understanding of skeletal muscle as a secretory organ, and of myokines as mediators of physical fitness through the practice of regular physical exercise (aerobic exercise and strength training), as well as new awareness of the anti-inflammatory and thus disease prevention aspects of exercise. Different muscle fiber types -slow twitch muscle fibers, oxidative muscle fibers, intermediate twitch muscle fibers, and fast twitch muscle fibers - release different clusters of myokines during contraction. This implies that variation of exercise types, particularly aerobic training/endurance training and muscle contraction against resistance (strength training) may offer differing myokine-induced benefits. This topic has been discussed by fitness training specialists.[7]

"Some myokines exert their effects within the muscle itself. Thus, myostatin, LIF, IL-6 and IL-7 are involved in muscle hypertrophy and myogenesis, whereas BDNF and IL-6 are involved in AMPK-mediated fat oxidation. IL-6 also appears to have systemic effects on the liver, adipose tissue and the immune system, and mediates crosstalk between intestinal L cells and pancreatic islets. Other myokines include the osteogenic factors IGF-1 and FGF-2; FSTL-1, which improves the endothelial function of the vascular system; and the PGC-1alpha-dependent myokine irisin, which drives brown fat-like development. Studies in the past few years suggest the existence of yet unidentified factors, secreted from muscle cells, which may influence cancer cell growth and pancreas function. Many proteins produced by skeletal muscle are dependent upon contraction; therefore, physical inactivity probably leads to an altered myokine response, which could provide a potential mechanism for the association between sedentary behaviour and many chronic diseases."[2]

"In summary, physical inactivity and muscle disuse lead to loss of muscle mass and accumulation of visceral adipose tissue and consequently to the activation of a network of inflammatory pathways, which promote development of insulin resistance, atherosclerosis, neurodegeneration and tumour growth and, thereby, promote the development of a cluster of chronic diseases. By contrast, the finding that muscles produce and release myokines provides a molecular basis for understanding how physical activity could protect against premature mortality.... Given that muscle is the largest organ in the body, the identification of the muscle secretome could set a new agenda for the scientific community. To view skeletal muscle as a secretory organ provides a conceptual basis for understanding how muscles communicate with other organs such as adipose tissue, liver, pancreas, bone and brain. Physical inactivity or muscle disuse potentially leads to an altered or impaired myokine response and/or resistance to the effects of myokines, which explains why lack of physical activity increases the risk of a whole network of diseases, including cardiovascular diseases, T2DM (Type 2 Diabetes Mellitus), cancer and osteoporosis."[2]

Role in regulating heart architecture

Heart muscle is subject to two kinds of stress: physiologic stress, i.e. exercise; and pathologic stress, i.e. disease related. Likewise, the heart has two potential responses to either stress: cardiac hypertrophy, which is a normal, physiologic, adaptive growth; or cardiac remodeling, which is an abnormal, pathologic, maladaptive growth. Upon being subjected to either stress, the heart "chooses" to turn on one of the responses and turn off the other. If it has chosen the abnormal path,i.e. remodeling, exercise can reverse this choice by turning off remodeling and turning on hypertrophy. The mechanism for reversing this choice is the microRNA miR-222 in cardiac muscle cells, which exercise up-regulates via unknown myokines. miR-222 represses genes involved in fibrosis and cell-cycle control.[8]

Immunomodulation

Immunomodulation and immunoregulation were a particular focus of early myokine research, as, according to Dr. Bente Klarlund Pedersen and her colleagues, "the interactions between exercise and the immune system provided a unique opportunity to evaluate the role of underlying endocrine and cytokine mechanisms."[1]

Specific myokines

Myostatin

Both aerobic exercise and strength training (resistance exercise) attenuate myostatin expression, and myostatin inactivation potentiates the beneficial effects of endurance exercise on metabolism.[9]

Interleukins

Aerobic exercise provokes a systemic cytokine response, including, for example, IL-6, IL-1 receptor antagonist (IL-1ra), and IL-10 (Interleukin 10). IL-6 was serendipitously discovered as a myokine because of the observation that it increased in an exponential fashion proportional to the length of exercise and the amount of muscle mass engaged in the exercise. This increase is followed by the appearance of IL-1ra and the anti-inflammatory cytokine IL-10. In general, the cytokine response to exercise and sepsis differs with regard to TNF-α. Thus, the cytokine response to exercise is not preceded by an increase in plasma-TNF-α. Following exercise, the basal plasma IL-6 concentration may increase up to 100-fold, but less dramatic increases are more frequent. The exercise-induced increase of plasma IL-6 occurs in an exponential manner and the peak IL-6 level is reached at the end of the exercise or shortly thereafter. It is the combination of mode, intensity, and duration of the exercise that determines the magnitude of the exercise-induced increase of plasma IL-6.[6]

IL-6 had previously been classified as a proinflammatory cytokine. Therefore, it was first thought that the exercise-induced IL-6 response was related to muscle damage.[10] However, it has become evident that eccentric exercise is not associated with a larger increase in plasma IL-6 than exercise involving concentric “nondamaging” muscle contractions. This finding clearly demonstrates that muscle damage is not required to provoke an increase in plasma IL-6 during exercise. As a matter of fact, eccentric exercise may result in a delayed peak and a much slower decrease of plasma IL-6 during recovery.[3]

IL-6, among an increasing number of other recently identified myokines, thus remains an important topic of myokine research. It appears in muscle tissue and in the circulation during exercise at levels up to one hundred times basal rates, as noted, and is seen as having a beneficial impact on health and bodily functioning in most circumstances. P. Munoz-Canoves et al. write: "It appears consistently in the literature that IL-6, produced locally by different cell types, has a positive impact on the proliferative capacity of muscle stem cells. This physiological mechanism functions to provide enough muscle progenitors in situations that require a high number of these cells, such as during the processes of muscle regeneration and hypertrophic growth after an acute stimulus. IL-6 is also the founding member of the myokine family of muscle-produced cytokines. Indeed, muscle-produced IL-6 after repeated contractions also has important autocrine and paracrine benefits, acting as a myokine, in regulating energy metabolism, controlling, for example, metabolic functions and stimulating glucose production. It is important to note that these positive effects of IL-6 and other myokines are normally associated with its transient production and short-term action."[11]

Interleukin 15

Interleukin-15 may play a significant role in the reduction of the volume of visceral (intra-abdominal) fat. IL-15 may accumulate within the muscle as a consequence of regular training. There is a negative association between plasma IL-15 concentration and trunk fat mass, but not limb fat mass.[12]

Brain-derived neurotrophic factor

<templatestyles src="Module:Hatnote/styles.css"></templatestyles>

Brain-derived neurotrophic factor (BDNF) is also a myokine, though BDNF produced by contracting muscle is not released into circulation. Rather, BDNF produced in skeletal muscle appears to enhance the oxidation of fat. Skeletal muscle activation through exercise also contributes to an increase in BDNF secretion in the brain. A beneficial effect of BDNF on neuronal function has been noted in multiple studies.[12][13] Dr. Pedersen writes, "Neurotrophins are a family of structurally related growth factors, including brain-derived neurotrophic factor (BDNF), which exert many of their effects on neurons primarily through Trk receptor tyrosine kinases. Of these, BDNF and its receptor TrkB are most widely and abundantly expressed in the brain. However, recent studies show that BDNF is also expressed in non-neurogenic tissues, including skeletal muscle. BDNF has been shown to regulate neuronal development and to modulate synaptic plasticity. BDNF plays a key role in regulating survival, growth and maintenance of neurons, and BDNF has a bearing on learning and memory. However, BDNF has also been identified as a key component of the hypothalamic pathway that controls body mass and energy homeostasis.

"Most recently, we have shown that BDNF appears to be a major player not only in central metabolic pathways but also as a regulator of metabolism in skeletal muscle. Hippocampal samples from Alzheimer’s disease donors show decreased BDNF expression and individuals with Alzheimer’s disease have low plasma levels of BDNF. Also, patients with major depression have lower levels of serum BDNF than normal control subjects. Other studies suggest that plasma BDNF is a biomarker of impaired memory and general cognitive function in ageing women and a low circulating BDNF level was recently shown to be an independent and robust biomarker of mortality risk in old women. Interestingly, low levels of circulating BDNF are also found in obese individuals and those with type 2 diabetes. In addition, we have demonstrated that there is a cerebral output of BDNF and that this is inhibited during hyperglycaemic clamp conditions in humans. This last finding may explain the concomitant finding of low circulating levels of BDNF in individuals with type 2 diabetes, and the association between low plasma BDNF and the severity of insulin resistance.

"BDNF appears to play a role in both neurobiology and metabolism. Studies have demonstrated that physical exercise may increase circulating BDNF levels in humans. To identify whether the brain is a source of BDNF during exercise, eight volunteers rowed for 4 h while simultaneous blood samples were obtained from the radial artery and the internal jugular vein. To further identify the putative cerebral region(s) responsible for BDNF release, mouse brains were dissected and analysed for BDNF mRNA expression following treadmill exercise. In humans, a BDNF release from the brain was observed at rest and increased 2- to 3-fold during exercise. Both at rest and during exercise, the brain contributed 70–80% of the circulating BDNF, while this contribution decreased following 1 h of recovery. In mice, exercise induced a 3- to 5-fold increase in BDNF mRNA expression in the hippocampus and cortex, peaking 2 h after the termination of exercise. These results suggest that the brain is a major but not the sole contributor to circulating BDNF. Moreover, the importance of the cortex and hippocampus as sources of plasma BDNF becomes even more prominent in the response to exercise.”[12]

With respect to studies of exercise and brain function, a 2010 report is of particular interest. Erickson et al. have shown that the volume of the anterior hippocampus increased by 2% in response to aerobic training in a randomized controlled trial with 120 older adults. The authors also summarize several previously-established research findings relating to exercise and brain function: (1) Aerobic exercise training increases grey and white matter volume in the prefrontal cortex of older adults and increases the functioning of key nodes in the executive control network. (2) Greater amounts of physical activity have been associated with sparing of prefrontal and temporal brain regions over a 9-y period, which reduces the risk for cognitive impairment. (3) Hippocampal and medial temporal lobe volumes are larger in higher-fit older adults (larger hippocampal volumes have been demonstrated to mediate improvements in spatial memory). (4) Exercise training increases cerebral blood volume and perfusion of the hippocampus.[13]

Regarding the 2010 study, the authors conclude: "We also demonstrate that increased hippocampal volume is associated with greater serum levels of BDNF, a mediator of neurogenesis in the dentate gyrus. Hippocampal volume declined in the control group, but higher preintervention fitness partially attenuated the decline, suggesting that fitness protects against volume loss. Caudate nucleus and thalamus volumes were unaffected by the intervention. These theoretically important findings indicate that aerobic exercise training is effective at reversing hippocampal volume loss in late adulthood, which is accompanied by improved memory function."[13]

Myonectin (CTRP15)

Seldin, Peterson, Byerly, Wei and Wong clarify that most myokines are also secreted by non-muscle cells. In 2012, they reported: "The expression of all myokines described to date is not restricted to skeletal muscle; they are generally expressed by a variety of cell types, and most are, in fact, expressed at much higher levels by nonmuscle tissues. Prior to (our) study, no myokine has been discovered to be preferentially expressed by skeletal muscle. While characterizing the metabolic function of the C1q/TNF-related protein (CTRP) family of proteins we recently uncovered, we identified myonectin (CTRP15) as a novel member of the family on the basis of sequence homology in the shared C1q domain, the signature that defines this protein family.... (Myonectin) is a novel nutrient-responsive myokine secreted by skeletal muscle to regulate whole-body fatty acid metabolism.... Circulating levels of myonectin were tightly regulated by the metabolic state; fasting suppressed, but refeeding dramatically increased, its mRNA and serum levels. Although mRNA and circulating levels of myonectin were reduced in a diet-induced obese state, voluntary exercise increased its expression and circulating levels. Accordingly, myonectin transcript was up-regulated by compounds (forskolin, epinephrine, ionomycin) that raise cellular cAMP or calcium levels.... In vitro results of myonectin expression in myotubes suggest that exercise-induced rises in intracellular calcium levels may also up-regulate myonectin expression in intact skeletal muscle.... Consistent with enhanced mRNA expression in skeletal muscle of mice subjected to voluntary exercise, circulating levels of myonectin also increased, suggesting a potential role of myonectin in exercise-induced physiology.

"Given that exercise induces myonectin expression in skeletal muscle, we next addressed whether short- and long-term changes in nutritional/metabolic state also regulate myonectin expression and circulating levels. Surprisingly, an overnight fast greatly suppressed myonectin expression, but a 2-h refeeding period (following an overnight fast) dramatically up-regulated its mRNA expression in skeletal muscle. Intriguingly, refeeding induced myonectin mRNA expression to a much greater extent in soleus than in plantaris muscle fiber of both male and female mice (data not shown), suggesting that myonectin expression may be regulated differentially depending on muscle fiber type. Consistent with the mRNA data, fasting reduced, but refeeding substantially increased, circulating levels of myonectin.... As compared with mice fed an isocaloric matched low-fat diet, mice fed a high-fat diet had lower myonectin mRNA and serum levels, suggesting that obesity-induced alteration in energy balance may be linked to dysregulation of myonectin-mediated processes in the obese state.... A relatively modest rise in serum myonectin levels was sufficient to lower (by >30%) nonesterified free fatty acid (NEFA) levels over time relative to vehicle-injected controls. However, no significant difference was observed in serum triacylglycerol levels between the two groups of mice. These data suggest a potential role of myonectin in regulating systemic fatty acid metabolism.... Treatment of adipocytes with recombinant myonectin (5 micrograms/ml) also enhanced fatty acid uptake to the same extent as insulin.... To determine whether myonectin-mediated enhancement of lipid uptake is specific to adipocytes, we also tested the effect of myonectin on lipid uptake in rat H4IIE hepatocytes. We observed a modest (>25%) but consistent increase in fatty acid uptake into hepatocytes stimulated with myonectin (5 micrograms/ml), an effect similar to cells treated with a saturating dose of insulin (50 nM).... Together, these results indicate that myonectin promotes lipid uptake into adipocytes and hepatocytes via transcriptional up-regulation of genes involved in fatty acid uptake....

"We provide the first characterization of myonectin, with in vitro and in vivo evidence that it is a novel myokine with important metabolic function. Unlike the other CTRPs characterized to date, myonectin (CTRP15) is expressed and secreted predominantly by skeletal muscle.... (Our) results suggest that myonectin is a nutrient-responsive metabolic regulator secreted by skeletal muscle in response to changes in cellular energy state resulting from glucose or fatty acid fluxes. Many metabolically relevant secreted proteins (e.g. adiponectin, leptin, resistin, and RBP) and the signaling pathways they regulate in tissues are known to be dysregulated in the condition of obesity. The reduction in expression and circulating levels of myonectin in the obese state may represent yet another component of the complex metabolic circuitry dysregulated by excess caloric intake. Although exercise has long been known to have profound positive impacts on systemic insulin sensitivity and energy balance, the underlying mechanisms remain incompletely understood. That voluntary exercise dramatically increases the expression and circulating levels of myonectin to promote fatty acid uptake into cells may underlie one of the beneficial effects of physical exercise.... A modest rise in the circulating levels of myonectin resulting from recombinant protein administration is sufficient to lower serum NEFA without altering serum triglyceride levels. Unlike CTRP1, CTRP3 and CTRP12, injection of recombinant myonectin into mice appears to have no glucose-lowering effect. Reduction in circulating NEFA is not due to suppression of adipose tissue lipolysis; rather, it results from increased fatty acid uptake by adipocytes and hepatocytes. Although the myonectin-mediated enhancement of lipid uptake in vitro appears modest (25–50%), in fact, the magnitude of this effect is comparable with cells stimulated with 50 nM insulin, a saturating dose that leads to maximum increase in fatty acid uptake.... In accordance with myonectin mediating its metabolic effect through a transcriptional mechanism, a reduction in circulating NEFA in mice occurred only 2 h after recombinant protein injection, a lag period presumably required for mRNA and protein synthesis."[14]

Decorin

Decorin is an example of a proteoglycan which functions as a myokine. Kanzleiter et al have established that this myokine is secreted during muscular contraction against resistance, and plays a role in muscle growth. They reported on July 1, 2014: "The small leucine-rich proteoglycan decorin has been described as a myokine for some time. However, its regulation and impact on skeletal muscle (had) not been investigated in detail. In (our recent) study, we report decorin to be differentially expressed and released in response to muscle contraction using different approaches. Decorin is released from contracting human myotubes, and circulating decorin levels are increased in response to acute resistance exercise in humans. Moreover, decorin expression in skeletal muscle is increased in humans and mice after chronic training. Because decorin directly binds myostatin, a potent inhibitor of muscle growth, we investigated a potential function of decorin in the regulation of skeletal muscle growth. In vivo overexpression of decorin in murine skeletal muscle promoted expression of the pro-myogenic factor Mighty, which is negatively regulated by myostatin. We also found Myod1 and follistatin to be increased in response to decorin overexpression. Moreover, muscle-specific ubiquitin ligases atrogin1 and MuRF1, which are involved in atrophic pathways, were reduced by decorin overexpression. In summary, our findings suggest that decorin secreted from myotubes in response to exercise is involved in the regulation of muscle hypertrophy and hence could play a role in exercise-related restructuring processes of skeletal muscle."[15]

Irisin

Discovery of Irisin

Irisin is a cleaved version of FNDC5. Boström and coworkers named the cleaved product irisin, after the Greek messenger goddess Iris.[16] FNDC5 was initially discovered in 2002 by two independent groups of researchers.[17][18][19]

Function of Irisin

Rana et al. reported in January 2014 that irisin (fibronectin type III domain-containing protein 5 or FNDC5), a recently described myokine hormone produced and secreted by acutely exercising skeletal muscles, is thought to bind white adipose tissue cells via undetermined receptors. Irisin has been reported to promote a brown adipose tissue-like phenotype upon white adipose tissue by increasing cellular mitochondrial density and expression of uncoupling protein-1, thereby increasing adipose tissue energy expenditure via thermogenesis. This is considered important, because excess visceral adipose tissue in particular distorts the whole body energy homeostasis, increases the risk of cardiovascular disease and raises exposure to a milieu of adipose tissue-secreted hormones (adipokines) that promote inflammation and cellular aging. The authors enquired whether the favorable impact of irisin on white adipose tissue might be associated with maintenance of telomere length, a well-established genetic marker in the aging process. They conclude that these data support the view that irisin may have a role in the modulation not only of energy balance but also the aging process.[20]

However, exogenous irisin may aid in heightening energy expenditure, and thus in reducing obesity. Boström et al. reported on December 14, 2012: "Since the conservation of calories would likely provide an overall survival advantage for mammals, it appears paradoxical that exercise would stimulate the secretion of a polypeptide hormone that increases thermogenesis and energy expenditure. One explanation for the increased irisin expression with exercise in mouse and man may have evolved as a consequence of muscle contraction during shivering. Muscle secretion of a hormone that activates adipose thermogenesis during this process might provide a broader, more robust defense against hypothermia. The therapeutic potential of irisin is obvious. Exogenously administered irisin induces the browning of subcutaneous fat and thermogenesis, and it presumably could be prepared and delivered as an injectable polypeptide. Increased formation of brown or beige/brite fat has been shown to have anti-obesity, anti-diabetic effects in multiple murine models, and adult humans have significant deposits of UCP1-positive brown fat. (Our data show) that even relatively short treatments of obese mice with irisin improves glucose homeostasis and causes a small weight loss. Whether longer treatments with irisin and/or higher doses would cause more weight loss remains to be determined. The worldwide, explosive increase in obesity and diabetes strongly suggests exploring the clinical utility of irisin in these and related disorders. Another potentially important aspect of this work relates to other beneficial effects of exercise, especially in some diseases for which no effective treatments exist. The clinical data linking exercise with health benefits in many other diseases suggests that irisin could also have significant effects in these disorders."[21]

While the murine findings reported by Boström et al. appear encouraging, other researchers have questioned whether irisin operates in a similar manner in humans. For example, Timmons et al. noted that over 1,000 genes are upregulated by exercise and examined how expression of FNDC5 was affected by exercise in ~200 humans. They found that it was upregulated only in highly active elderly humans, casting doubt on the conclusions of Boström et al.[22] Further discussion of this issue can be found in the Wikipedia entry for irisin under the "function" heading.

Osteonectin or Secreted protein acidic and rich in cysteine (SPARC)

A novel myokine Osteonectin, or SPARC, plays a vital role in bone mineralization, cell-matrix interactions, and collagen binding. Osteonectin inhibits tumorigenesis in mice. Osteonectin can be classed as a myokine, as it was found that even a single bout of exercise increased its expression and secretion in skeletal muscle in both mice and humans.[23]

References

  1. 1.0 1.1 1.2 1.3 Bente Klarlund Pedersen , Thorbjörn C. A. Åkerström , Anders R. Nielsen , Christian P. Fischer. "Role of myokines in exercise and metabolism." Journal of Applied Physiology | Published 1 September 2007 Vol. 103no. 1093-1098DOI: 10.1152/japplphysiol.00080.2007
  2. 2.0 2.1 2.2 Pedersen BK; Febbraio MA. "Muscles, exercise and obesity: skeletal muscle as a secretory organ." Nat Rev Endocrinol 2012; 8(8): 457-465.
  3. 3.0 3.1 3.2 Pedersen BK. "Muscle as a secretory organ." American Physiological Society. Compr Physiol 3:1337-1362, 2013. http://www.inflammation-metabolism.dk/index.php?pageid=21&pmid=23897689
  4. Pedersen, B. K. et al. Searching for the exercise factor: is IL‑6 a candidate? J. Muscle Res. Cell Motil. 24, 113–119 (2003).
  5. Allen DL, Cleary AS, Speaker KJ, Lindsay SF, Uyenishi J, Reed JM, MaddenMC, MehanRS. "Myostatin, activin receptor IIb, and follistatinlike-3 gene expression are altered in adipose tissue and skeletal muscle of obese mice." Am J Physiol Endocrinol Metab 294: E918-E927, 2008.
  6. 6.0 6.1 Pedersen BK, Febbraio MA. "Muscle as an endocrine organ: Focus on muscle-derived interleukin-6." Physiol Rev 88: 1379-1406, 2008.
  7. McGuff, Doug, MD. "The Amazing Power of Myokines." http://www.bodybyscience.net/home.html/?p=1340 12 January 2014.
  8. Hill, Joseph A, Braking Bad Hypertrophy, NEJM, May 28, 2015,2160-2162, DOI: 10.1056/NEJMcibr1504187
  9. Allen DL, Hittel DS, McPherron AC. "Expression and function of myostatin in obesity, diabetes, and exercise adaptation." Med Sci Sports Exerc 43: 1828-1835, 2011.
  10. Bruunsgaard H, Galbo H, Halkjaer-Kristensen J, Johansen TL, MacLean DA, Pedersen BK. "Exercise-induced increase in interleukin-6 is related to muscle damage." J Physiol Lond 499: 833-841, 1997.
  11. P. Munoz-Canoves et al. "IL-6 myokine signaling in skeletal muscle: a double-edged sword?" The FEBS Journal 280 (2013) 4131–4148, 2013, 7 May 2013.
  12. 12.0 12.1 12.2 Pedersen, Bente Klarlund. "Muscles and their myokines." The Journal of Experimental Biology 214, 337-346. © 2011. Published by The Company of Biologists Ltd. doi:10.1242/jeb.048074
  13. 13.0 13.1 13.2 Erickson, KI, et al. "Exercise training increases size of hippocampus and improves memory." The Proceedings of the National Academy of Sciences vol. 108 no. 7 > Kirk I. Erickson, 3017–3022, doi: 10.1073/pnas.1015950108
  14. Marcus M. Seldin, Jonathan M. Peterson, Mardi S. Byerly, Zhikui Wei, and G. William Wong. "Myonectin (CTRP15), a Novel Myokine That Links Skeletal Muscle to Systemic Lipid Homeostasis." THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 287, NO. 15, pp. 11968–11980, April 6, 2012, doi: 10.1074/jbc.M111.336834 originally published online February 17, 2012
  15. Kanzleiter T, Rath M, Görgens SW, Jensen J, Tangen DS, Kolnes AJ, Kolnes KJ, Lee S, Eckel J, Schürmann A, and Eckardt K. "The myokine decorin is regulated by contraction and involved in muscle hypertrophy." Biochem Biophys Res Commun. 2014 Jul 1. pii: S0006-291X(14)01197-8. doi: 10.1016/j.bbrc.2014.06.123. [Epub ahead of print]
  16. Lua error in package.lua at line 80: module 'strict' not found.
  17. Lua error in package.lua at line 80: module 'strict' not found.
  18. Lua error in package.lua at line 80: module 'strict' not found.
  19. Lua error in package.lua at line 80: module 'strict' not found.
  20. Karan S. Rana, Muhammad Arif, Eric J. Hill, Sarah Aldred, David A. Nagel, Alan Neville, Harpal S. Randeva, Clifford J. Bailey, Srikanth Bellary & James E. Brown. "Plasma irisin levels predict telomere length in healthy adults." AGE (2014) 36:995–1001. DOI 10.1007/s11357-014-9620-9. Published online: 29 January 2014.
  21. Pontus Boström, Jun Wu, Mark P. Jedrychowski, Anisha Korde, Li Ye, James C. Lo, Kyle A. Rasbach, Elisabeth Almer Boström, Jang Hyun Choi, Jonathan Z. Long, Shingo Kajimura, Maria Cristina Zingaretti, Birgitte F. Vind, Hua Tu, Saverio Cinti, Kurt Højlund, Steven P. Gygi, and Bruce M. Spiegelman. "A PGC1α-dependent myokine that drives browning of white fat and thermogenesis." Nature 481(7382): 463–468. doi:10.1038/nature10777. 2012 December 14
  22. Timmons JA, Baar K, Davidsen PK, Atherton PJ (2012). "Is irisin a human exercise gene?" Nature 488 (7413): E9–10; discussion E10–1. doi:10.1038/nature11364. PMID 22932392
  23. Wataru Aoi, Yuji Naito, Tomohisa Takagi, Yuko Tanimura, Yoshikazu Takanami, Yukari Kawai, Kunihiro Sakuma, Liu Po Hang, Katsura Mizushima, Yasuko Hirai, Ryota Koyama, Sayori Wada, Akane Higashi, Satoshi Kokura, Hiroshi Ichikawa and Toshikazu Yoshikawa. "A novel myokine, secreted protein acidic and rich in cysteine (SPARC), suppresses colon tumorigenesis via regular exercise." Gut (2012). doi:10.1136/gutjnl-2011-300776. 9 August 2012.

External links