51
|
Das DK, Graham ZA, Cardozo CP. Myokines in skeletal muscle physiology and metabolism: Recent advances and future perspectives. Acta Physiol (Oxf) 2020; 228:e13367. [PMID: 31442362 DOI: 10.1111/apha.13367] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/11/2019] [Accepted: 08/03/2019] [Indexed: 12/13/2022]
Abstract
Myokines are molecules produced and secreted by skeletal muscle to act in an auto-, para- and endocrine manner to alter physiological function of target tissues. The growing number of effects of myokines on metabolism of distant tissues provides a compelling case for crosstalk between skeletal muscle and other tissues and organs to regulate metabolic homoeostasis. In this review, we summarize and discuss the current knowledge regarding the impact on metabolism of several canonical and recently identified myokines. We focus specifically on myostatin, β-aminoisobutyric acid, interleukin-15, meteorin-like and myonectin, and discuss how these myokines are induced and regulated as well as their overall function. We also review how these myokines may serve as potential prognostic biomarkers that reflect whole-body metabolism and how they may be attractive therapeutic targets for treating muscle and metabolic diseases.
Collapse
Affiliation(s)
- Dibash K. Das
- National Center for the Medical Consequences of Spinal Cord Injury James J. Peters VA Medical Center Bronx NY USA
- Department of Medicine Icahn School of Medicine at Mount Sinai New York NY USA
| | - Zachary A. Graham
- Birmingham VA Medical Center University of Alabama‐Birmingham Birmingham AL USA
- Department of Cell, Developmental, and Integrative Biology University of Alabama‐Birmingham Birmingham AL USA
| | - Christopher P. Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury James J. Peters VA Medical Center Bronx NY USA
- Department of Medicine Icahn School of Medicine at Mount Sinai New York NY USA
- Department of Rehabilitation Medicine Icahn School of Medicine at Mount Sinai New York NY USA
| |
Collapse
|
52
|
Dent JR, Hetrick B, Tahvilian S, Sathe A, Greyslak K, LaBarge SA, Svensson K, McCurdy CE, Schenk S. Skeletal muscle mitochondrial function and exercise capacity are not impaired in mice with knockout of STAT3. J Appl Physiol (1985) 2019; 127:1117-1127. [PMID: 31513449 DOI: 10.1152/japplphysiol.00003.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) was recently found to be localized to mitochondria in a number of tissues and cell types, where it modulates oxidative phosphorylation via interactions with the electron transport proteins, complex I and complex II. Skeletal muscle is densely populated with mitochondria although whether STAT3 contributes to skeletal muscle oxidative capacity is unknown. In the present study, we sought to elucidate the contribution of STAT3 to mitochondrial and skeletal muscle function by studying mice with muscle-specific knockout of STAT3 (mKO). First, we developed a novel flow cytometry-based approach to confirm that STAT3 is present in skeletal muscle mitochondria. However, contrary to findings in other tissue types, complex I and complex II activity and maximal mitochondrial respiratory capacity in skeletal muscle were comparable between mKO mice and floxed/wild-type littermates. Moreover, there were no genotype differences in endurance exercise performance, skeletal muscle force-generating capacity, or the adaptive response of skeletal muscle to voluntary wheel running. Collectively, although we confirm the presence of STAT3 in skeletal muscle mitochondria, our data establish that STAT3 is dispensable for mitochondrial and physiological function in skeletal muscle.NEW & NOTEWORTHY Whether signal transducer and activator of transcription 3 (STAT3) can regulate the activity of complex I and II of the electron transport chain and mitochondrial oxidative capacity in skeletal muscle, as it can in other tissues, is unknown. By using a mouse model lacking STAT3 in muscle, we demonstrate that skeletal muscle mitochondrial and physiological function, both in vivo and ex vivo, is not impacted by the loss of STAT3.
Collapse
Affiliation(s)
- Jessica R Dent
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California
| | - Byron Hetrick
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Shahriar Tahvilian
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California
| | - Abha Sathe
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California
| | - Keenan Greyslak
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Samuel A LaBarge
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California
| | - Kristoffer Svensson
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Simon Schenk
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California.,Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California
| |
Collapse
|
53
|
Song P, Kwon Y, Joo JY, Kim DG, Yoon JH. Secretomics to Discover Regulators in Diseases. Int J Mol Sci 2019; 20:ijms20163893. [PMID: 31405033 PMCID: PMC6720857 DOI: 10.3390/ijms20163893] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/01/2019] [Accepted: 08/08/2019] [Indexed: 01/03/2023] Open
Abstract
Secretory proteins play important roles in the cross-talk of individual functional units, including cells. Since secretory proteins are essential for signal transduction, they are closely related with disease development, including metabolic and neural diseases. In metabolic diseases, adipokines, myokines, and hepatokines are secreted from respective organs under specific environmental conditions, and play roles in glucose homeostasis, angiogenesis, and inflammation. In neural diseases, astrocytes and microglia cells secrete cytokines and chemokines that play roles in neurotoxic and neuroprotective responses. Mass spectrometry-based secretome profiling is a powerful strategy to identify and characterize secretory proteins. This strategy involves stepwise processes such as the collection of conditioned medium (CM) containing secretome proteins and concentration of the CM, peptide preparation, mass analysis, database search, and filtering of secretory proteins; each step requires certain conditions to obtain reliable results. Proteomic analysis of extracellular vesicles has become a new research focus for understanding the additional extracellular functions of intracellular proteins. Here, we provide a review of the insights obtained from secretome analyses with regard to disease mechanisms, and highlight the future prospects of this technology. Continued research in this field is expected to provide valuable information on cell-to-cell communication and uncover new pathological mechanisms.
Collapse
Affiliation(s)
- Parkyong Song
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Yonghoon Kwon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Jae-Yeol Joo
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, Korea
| | - Do-Geun Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Korea
| | - Jong Hyuk Yoon
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, Korea.
| |
Collapse
|
54
|
Jiang H, Yoshioka Y, Yuan S, Horiuchi Y, Yamashita Y, Croft KD, Ashida H. Enzymatically modified isoquercitrin promotes energy metabolism through activating AMPKα in male C57BL/6 mice. Food Funct 2019; 10:5188-5202. [PMID: 31380532 DOI: 10.1039/c9fo01008d] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Quercetin possesses various health beneficial functions, but its poor bioavailability limits these functions. Enzymatically modified isoquercitrin (EMIQ) is a quercetin glycoside with a greater bioavailability than quercetin. In this study, we investigated whether EMIQ regulates energy metabolism in mice and its underlying molecular mechanism. Male C57BL/6 mice were fed a normal diet with different doses of EMIQ or quercetin (0.02%, 0.1% and 0.5%) for two weeks. Supplementation with 0.1% EMIQ significantly decreased white adipose tissue (WAT) weight. Supplementation with 0.02% and 0.1% EMIQ promoted phosphorylation of adenosine monophosphate activated protein kinase (AMPK) in the WAT, liver, and muscle. In the WAT, 0.1% EMIQ downregulated peroxisome proliferator-activated receptor (PPAR)γ, CCAAT-enhancer-binding protein (C/EBP)α, C/EBPβ, and sterol regulatory element-binding protein 1 expression, as well as upregulated mitochondrial uncoupling protein (UCP) 2 and carnitine palmitoyltransferase-1 expression. Supplementation with 0.1% EMIQ also promoted the expression of thermogenesis-associated factors including PPARγ coactivator α (PGC-1α), UCP1, PR-domain containing protein 16, and sirtuin 1 in the WAT. In the liver, EMIQ promoted the phosphorylation of acetyl-CoA carboxylase, and increased the expression of PPARα, constitutive androstane-receptor, and farnesoid X receptor. Furthermore, supplementation with 0.02% or 0.1% EMIQ suppressed the plasma glucose level accompanied by the translocation of glucose transporter 4 to the plasma membrane of the muscle. Our results suggest that EMIQ is a potential food additive for the regulation of energy metabolism through AMPK phosphorylation.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
55
|
Nadeau L, Aguer C. Interleukin-15 as a myokine: mechanistic insight into its effect on skeletal muscle metabolism. Appl Physiol Nutr Metab 2019; 44:229-238. [DOI: 10.1139/apnm-2018-0022] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Interleukin (IL)-15 is a cytokine with important immunological functions. It is highly expressed in skeletal muscle and is believed to be a myokine, a hypothesis supported by the rapid increase in circulating levels of IL-15 in response to exercise. Treatment with high doses of IL-15 results in metabolic adaptations such as improved insulin sensitivity and whole-body fatty acid oxidation and protection from high-fat-diet-induced obesity and insulin resistance. IL-15 secreted by contracting muscle may therefore act as an endocrine factor to improve adiposity and energy metabolism in different tissues. Most studies have used supraphysiological doses of IL-15 that do not represent circulating IL-15 in response to exercise. However, evidence shows that IL-15 levels are higher in muscle interstitium and that IL-15 might improve muscle glucose homeostasis and oxidative metabolism in an autocrine/paracrine manner. Nevertheless, how IL-15 signals in skeletal muscle to improve muscle energy metabolism is not understood completely, especially because the absence of the α subunit of the IL-15 receptor (IL-15Rα) results in a phenotype similar to that of overexpressing/oversecreting IL-15 in mice. In this article, we review the literature to propose a model for the regulation of IL-15 by the soluble form of IL-15Rα to explain why some findings in the literature seem, at first glance, to be contradictory.
Collapse
Affiliation(s)
- Lucien Nadeau
- Institut du Savoir Montfort – Recherche, 713 Montreal Road, Ottawa, ON K1K 0T2, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Céline Aguer
- Institut du Savoir Montfort – Recherche, 713 Montreal Road, Ottawa, ON K1K 0T2, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
56
|
Jiang H, Yamashita Y, Nakamura A, Croft K, Ashida H. Quercetin and its metabolite isorhamnetin promote glucose uptake through different signalling pathways in myotubes. Sci Rep 2019; 9:2690. [PMID: 30804434 PMCID: PMC6389993 DOI: 10.1038/s41598-019-38711-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/23/2018] [Indexed: 12/22/2022] Open
Abstract
Quercetin and its metabolite isorhamnetin elicit various beneficial effects on human health. However, their bioavailability is low. In this study, we investigated whether low concentrations in the physiological range could promote glucose uptake in L6 myotubes, as well as the underlying molecular mechanisms. We found that 0.1 nM and 1 nM quercetin or 1 nM isorhamnetin significantly increased glucose uptake via translocation of glucose transporter type 4 (GLUT4) to the plasma membrane of L6 myotubes. Quercetin principally activated the CaMKKβ/AMPK signalling pathway at these concentrations, but also activated IRS1/PI3K/Akt signalling at 10 nM. In contrast, 1 nM and 10 nM isorhamnetin principally activated the JAK/STAT pathway. Treatment with siAMPKα and siJAK2 abolished quercetin- and isorhamnetin-induced GLUT4 translocation, respectively. However, treatment with siJAK3 did not affect isorhamnetin-induced GLUT4 translocation, indicating that isorhamnetin induced GLUT4 translocation mainly through JAK2, but not JAK3, signalling. Thus, quercetin preferably activated the AMPK pathway and, accordingly, stimulated IRS1/PI3K/Akt signalling, while isorhamnetin activated the JAK2/STAT pathway. Furthermore, after oral administration of quercetin glycoside at 10 and 100 mg/kg body weight significantly induced GLUT4 translocation to the plasma membrane of skeletal muscles in mice. In the same animals, plasma concentrations of quercetin aglycone form were 4.95 and 6.80 nM, respectively. In conclusion, at low-concentration ranges, quercetin and isorhamnetin promote glucose uptake by increasing GLUT4 translocation via different signalling pathways in skeletal muscle cells; thus, these compounds may possess beneficial functions for maintaining glucose homeostasis by preventing hyperglycaemia at physiological concentrations.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan
| | - Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan
| | - Asuka Nakamura
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan
| | - Kevin Croft
- School of Biomedical Science, The University of Western Australia, Perth, WA, 6009, Australia
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan.
| |
Collapse
|
57
|
Overexpression of Interleukin-15 exhibits improved glucose tolerance and promotes GLUT4 translocation via AMP-Activated protein kinase pathway in skeletal muscle. Biochem Biophys Res Commun 2019; 509:994-1000. [DOI: 10.1016/j.bbrc.2019.01.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 01/05/2019] [Indexed: 01/12/2023]
|
58
|
Lee JH, Jun HS. Role of Myokines in Regulating Skeletal Muscle Mass and Function. Front Physiol 2019; 10:42. [PMID: 30761018 PMCID: PMC6363662 DOI: 10.3389/fphys.2019.00042] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/14/2019] [Indexed: 12/27/2022] Open
Abstract
Loss of skeletal muscle mass and strength has recently become a hot research topic with the extension of life span and an increasingly sedentary lifestyle in modern society. Maintenance of skeletal muscle mass is considered an essential determinant of muscle strength and function. Myokines are cytokines synthesized and released by myocytes during muscular contractions. They are implicated in autocrine regulation of metabolism in the muscle as well as in the paracrine/endocrine regulation of other tissues and organs including adipose tissue, the liver, and the brain through their receptors. Till date, secretome analysis of human myocyte culture medium has revealed over 600 myokines. In this review article, we summarize our current knowledge of major identified and characterized myokines focusing on their biological activity and function, particularly in muscle mass and function.
Collapse
Affiliation(s)
- Jong Han Lee
- College of Pharmacy, Gachon University, Incheon, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea
| | - Hee-Sook Jun
- College of Pharmacy, Gachon University, Incheon, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea.,Gachon University Gil Medical Center, Gachon Medical and Convergence Institute, Incheon, South Korea
| |
Collapse
|
59
|
Kim S, Choi JY, Moon S, Park DH, Kwak HB, Kang JH. Roles of myokines in exercise-induced improvement of neuropsychiatric function. Pflugers Arch 2019; 471:491-505. [PMID: 30627775 DOI: 10.1007/s00424-019-02253-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/18/2018] [Accepted: 01/03/2019] [Indexed: 01/28/2023]
Abstract
Exercise is a well-known non-pharmacological intervention to improve brain functions, including cognition, memory, and motor coordination. Contraction of skeletal muscles during exercise releases humoral factors that regulate the whole-body metabolism via interaction with other non-muscle organs. Myokines are muscle-derived effectors that regulate body metabolism by autocrine, paracrine, or endocrine action and were reportedly suggested as "exercise factors" that can improve the brain function. However, several aspects remain to be elucidated, namely the specific activities of myokines related to the whole-body metabolism or brain function, the mechanisms of regulation of other organs or cells, the sources of "exercise factors" that regulate brain function, and their mechanisms of interaction with non-muscle organs. In this paper, we present the physiological functions of myokines secreted by exercise, including regulation of the whole-body metabolism by interaction with other organs and adaptation of skeletal muscles to exercise. In addition, we discuss the functions of myokines that possibly contribute to exercise-induced improvement of brain function. Among several myokines, brain-derived neurotrophic factor (BDNF) is the most studied myokine that regulates adult neurogenesis and synaptic plasticity. However, the source of circulating BDNF and its upstream effector, insulin-like growth factor (IGF-1), and irisin and the effect size of peripheral BDNF, irisin, and IGF-1 released after exercise should be further investigated. Recently, cathepsin B has been reported to be secreted from skeletal muscles and upregulate BDNF following exercise, which was associated with improved cognitive function. We reviewed the level of evidence for the effect of myokine on the brain function. Level of evidence for the association of the change in circulating myokine following exercise and improvement of neuropsychiatric function is lower than the level of evidence for the benefit of exercise on the brain. Therefore, more clinical evidences for the association of myokine release after exercise and their effect on the brain function are required. Finally, we discuss the effect size of the action of myokines on cognitive benefits of exercise, in addition to other contributors, such as improvement of the cardiovascular system or the effect of "exercise factors" released from non-muscle organs, particularly in patients with sarcopenia.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Pharmacology and Hypoxia-related Disease Research Center, Inha University School of Medicine, Room 1015, 60th Anniversary Hall, 100, Inha-ro, Nam-gu, Incheon, 22212, Republic of Korea.,Department of Kinesiology, Inha University, Incheon, Republic of Korea
| | - Ji-Young Choi
- Department of Pharmacology and Hypoxia-related Disease Research Center, Inha University School of Medicine, Room 1015, 60th Anniversary Hall, 100, Inha-ro, Nam-gu, Incheon, 22212, Republic of Korea
| | - Sohee Moon
- Department of Pharmacology and Hypoxia-related Disease Research Center, Inha University School of Medicine, Room 1015, 60th Anniversary Hall, 100, Inha-ro, Nam-gu, Incheon, 22212, Republic of Korea
| | - Dong-Ho Park
- Department of Kinesiology, Inha University, Incheon, Republic of Korea
| | - Hyo-Bum Kwak
- Department of Kinesiology, Inha University, Incheon, Republic of Korea
| | - Ju-Hee Kang
- Department of Pharmacology and Hypoxia-related Disease Research Center, Inha University School of Medicine, Room 1015, 60th Anniversary Hall, 100, Inha-ro, Nam-gu, Incheon, 22212, Republic of Korea.
| |
Collapse
|
60
|
Nadeau L, Patten DA, Caron A, Garneau L, Pinault-Masson E, Foretz M, Haddad P, Anderson BG, Quinn LS, Jardine K, McBurney MW, Pistilli EE, Harper ME, Aguer C. IL-15 improves skeletal muscle oxidative metabolism and glucose uptake in association with increased respiratory chain supercomplex formation and AMPK pathway activation. Biochim Biophys Acta Gen Subj 2018; 1863:395-407. [PMID: 30448294 DOI: 10.1016/j.bbagen.2018.10.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND IL-15 is believed to play a role in the beneficial impact of exercise on muscle energy metabolism. However, previous studies have generally used supraphysiological levels of IL-15 that do not represent contraction-induced IL-15 secretion. METHODS L6 myotubes were treated acutely (3 h) and chronically (48 h) with concentrations of IL-15 mimicking circulating (1-10 pg/ml) and muscle interstitial (100 pg/ml -20 ng/ml) IL-15 levels with the aim to better understand its autocrine/paracrine role on muscle glucose uptake and mitochondrial function. RESULTS Acute exposure to IL-15 levels representing muscle interstitial IL-15 increased basal glucose uptake without affecting insulin sensitivity. This was accompanied by increased mitochondrial oxidative functions in association with increased AMPK pathway and formation of complex III-containing supercomplexes. Conversely, chronic IL-15 exposure resulted in a biphasic effect on mitochondrial oxidative functions and ETC supercomplex formation was increased with low IL-15 levels but decreased with higher IL-15 concentrations. The AMPK pathway was activated only by high levels of chronic IL-15 treatment. Similar results were obtained in skeletal muscle from muscle-specific IL-15 overexpressing mice that show very high circulating IL-15 levels. CONCLUSIONS Acute IL-15 treatment that mimics local IL-15 concentrations enhances muscle glucose uptake and mitochondrial oxidative functions. That mitochondria respond differently to different levels of IL-15 during chronic treatments indicates that IL-15 might activate two different pathways in muscle depending on IL-15 concentrations. GENERAL SIGNIFICANCE Our results suggest that IL-15 may act in an autocrine/paracrine fashion and be, at least in part, involved in the positive effect of exercise on muscle energy metabolism.
Collapse
Affiliation(s)
- L Nadeau
- Institut du Savoir Montfort - Recherche, Ottawa, ON, Canada; University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology, and Immunology, Ottawa, ON, Canada
| | - D A Patten
- University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology, and Immunology, Ottawa, ON, Canada
| | - A Caron
- Institut du Savoir Montfort - Recherche, Ottawa, ON, Canada; University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology, and Immunology, Ottawa, ON, Canada
| | - L Garneau
- Institut du Savoir Montfort - Recherche, Ottawa, ON, Canada; University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology, and Immunology, Ottawa, ON, Canada
| | - E Pinault-Masson
- Institut du Savoir Montfort - Recherche, Ottawa, ON, Canada; University of Ottawa, Faculty of Science, Ottawa, ON, Canada
| | - M Foretz
- INSERM, U1016, Institut Cochin, Paris 75014, France; CNRS, UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - P Haddad
- Institut du Savoir Montfort - Recherche, Ottawa, ON, Canada; University of Ottawa, Faculty of Science, Ottawa, ON, Canada
| | - B G Anderson
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
| | - L S Quinn
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
| | - K Jardine
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - M W McBurney
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - E E Pistilli
- West Virginia University School of Medicine, Morgantown, WV, United States
| | - M E Harper
- University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology, and Immunology, Ottawa, ON, Canada
| | - C Aguer
- Institut du Savoir Montfort - Recherche, Ottawa, ON, Canada; University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology, and Immunology, Ottawa, ON, Canada.
| |
Collapse
|
61
|
Hingorjo MR, Zehra S, Saleem S, Qureshi MA. Serum Interleukin-15 and its relationship with adiposity Indices before and after short-term endurance exercise. Pak J Med Sci 2018; 34:1125-1131. [PMID: 30344562 PMCID: PMC6191778 DOI: 10.12669/pjms.345.15516] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Objective The myokine interleukin-15 (IL-15) is capable of modifying the metabolism of both skeletal and adipose tissue. This study compares the change in serum levels of IL-15 in obese and non-obese after a single session of submaximal exercise. Methods A cross-sectional study was carried out at Jinnah Medical and Dental College, Karachi, during Aug-Dec 2015, comprising of 133 medical students (aged 17-24 years). Cardiorespiratory fitness was evaluated by Queen's College Step Test. Blood was obtained both before and just after exercise and serum levels of IL-15 determined by enzyme-linked immunosorbent assay. Results Mean serum level of IL-15 was 3.64±1.59 pg/mL. Higher levels of IL-15 were seen in lean subjects compared to overweight/obese, both before and after three minutes of exercise (all Ptrend<.001). The percent increase in IL-15 upon exercise was 12.7% higher in lean. Significant negative association was seen between interleukin-15 and adiposity, especially visceral fat (r = -.288, p=.001). Conclusion Interleukin-15 correlates negatively with adiposity indices, especially visceral fat. With the proven benefit of IL-15 in terms of adipose tissue stores and skeletal muscle mitochondrial biogenesis, endurance exercises, even of short duration, may possess therapeutic potential towards producing a healthier body.
Collapse
Affiliation(s)
- Mozaffer Rahim Hingorjo
- Prof. Mozaffer Rahim Hingorjo, FCPS. Department of Physiology, Jinnah Medical & Dental College, Karachi, Pakistan
| | - Sitwat Zehra
- Dr. Sitwat Zehra, PhD. Department of Physiology, Dr. A.Q. Khan Institute of Biotechnology & Genetic Engineering (KIBGE), Karachi, Pakistan
| | - Saima Saleem
- Dr. Saima Saleem, PhD. Department of Physiology, Dr. A.Q. Khan Institute of Biotechnology & Genetic Engineering (KIBGE), Karachi, Pakistan
| | - Masood Anwar Qureshi
- Prof. Masood Anwar Qureshi, PhD. Department of Physiology, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
62
|
Malemud CJ. The role of the JAK/STAT signal pathway in rheumatoid arthritis. Ther Adv Musculoskelet Dis 2018; 10:117-127. [PMID: 29942363 PMCID: PMC6009092 DOI: 10.1177/1759720x18776224] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/20/2018] [Indexed: 12/30/2022] Open
Abstract
Proinflammatory cytokine activation of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signal transduction pathway is a critical event in the pathogenesis and progression of rheumatoid arthritis. Under normal conditions, JAK/STAT signaling reflects the influence of negative regulators of JAK/STAT, exemplified by the suppressor of cytokine signaling and protein inhibitor of activated STAT. However, in rheumatoid arthritis (RA) both of these regulators are dysfunctional. Thus, continuous activation of JAK/STAT signaling in RA synovial joints results in the elevated level of matrix metalloproteinase gene expression, increased frequency of apoptotic chondrocytes and most prominently 'apoptosis resistance' in the inflamed synovial tissue. Tofacitinib, a JAK small molecule inhibitor, with selectivity for JAK2/JAK3 was approved by the United States Food and Drug Administration (US FDA) for the therapy of RA. Importantly, tofacitinib has demonstrated significant clinical efficacy for RA in the post-US FDA-approval surveillance period. Of note, the success of tofacitinib has spurred the development of JAK1, JAK2 and other JAK3-selective small molecule inhibitors, some of which have also entered the clinical setting, whereas other JAK inhibitors are currently being evaluated in RA clinical trials.
Collapse
|
63
|
Shi H, Munk A, Nielsen TS, Daughtry MR, Larsson L, Li S, Høyer KF, Geisler HW, Sulek K, Kjøbsted R, Fisher T, Andersen MM, Shen Z, Hansen UK, England EM, Cheng Z, Højlund K, Wojtaszewski JFP, Yang X, Hulver MW, Helm RF, Treebak JT, Gerrard DE. Skeletal muscle O-GlcNAc transferase is important for muscle energy homeostasis and whole-body insulin sensitivity. Mol Metab 2018. [PMID: 29525407 PMCID: PMC6001359 DOI: 10.1016/j.molmet.2018.02.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective Given that cellular O-GlcNAcylation levels are thought to be real-time measures of cellular nutrient status and dysregulated O-GlcNAc signaling is associated with insulin resistance, we evaluated the role of O-GlcNAc transferase (OGT), the enzyme that mediates O-GlcNAcylation, in skeletal muscle. Methods We assessed O-GlcNAcylation levels in skeletal muscle from obese, type 2 diabetic people, and we characterized muscle-specific OGT knockout (mKO) mice in metabolic cages and measured energy expenditure and substrate utilization pattern using indirect calorimetry. Whole body insulin sensitivity was assessed using the hyperinsulinemic euglycemic clamp technique and tissue-specific glucose uptake was subsequently evaluated. Tissues were used for histology, qPCR, Western blot, co-immunoprecipitation, and chromatin immunoprecipitation analyses. Results We found elevated levels of O-GlcNAc-modified proteins in obese, type 2 diabetic people compared with well-matched obese and lean controls. Muscle-specific OGT knockout mice were lean, and whole body energy expenditure and insulin sensitivity were increased in these mice, consistent with enhanced glucose uptake and elevated glycolytic enzyme activities in skeletal muscle. Moreover, enhanced glucose uptake was also observed in white adipose tissue that was browner than that of WT mice. Interestingly, mKO mice had elevated mRNA levels of Il15 in skeletal muscle and increased circulating IL-15 levels. We found that OGT in muscle mediates transcriptional repression of Il15 by O-GlcNAcylating Enhancer of Zeste Homolog 2 (EZH2). Conclusions Elevated muscle O-GlcNAc levels paralleled insulin resistance and type 2 diabetes in humans. Moreover, OGT-mediated signaling is necessary for proper skeletal muscle metabolism and whole-body energy homeostasis, and our data highlight O-GlcNAcylation as a potential target for ameliorating metabolic disorders. Type 2 diabetic humans have elevated O-GlcNAc levels in skeletal muscle. Knockout of OGT in muscle elevates whole body insulin sensitivity. Knockout of OGT in muscle increases resistance to diet-induced obesity. Muscle-specific OGT knockout mice have elevated plasma IL-15 levels. OGT in muscle controls Il15 expression by O-GlcNAcylation and inhibition of EZH2.
Collapse
Affiliation(s)
- Hao Shi
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Alexander Munk
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK2200, Denmark
| | - Thomas S Nielsen
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK2200, Denmark
| | - Morgan R Daughtry
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Louise Larsson
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK2200, Denmark
| | - Shize Li
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Kasper F Høyer
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK2200, Denmark; Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK8000, Denmark
| | - Hannah W Geisler
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Karolina Sulek
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK2200, Denmark
| | - Rasmus Kjøbsted
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, DK2100, Denmark
| | - Taylor Fisher
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Marianne M Andersen
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK2200, Denmark
| | - Zhengxing Shen
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Ulrik K Hansen
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK2200, Denmark
| | - Eric M England
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Zhiyong Cheng
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Kurt Højlund
- Department of Endocrinology, Odense University Hospital, Odense, Denmark; Section of Molecular Diabetes and Metabolism, Institute of Molecular Medicine and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, DK2100, Denmark
| | - Xiaoyong Yang
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Matthew W Hulver
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; The Virginia Tech Metabolic Phenotyping Core, Blacksburg, VA 24061, USA
| | - Richard F Helm
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Jonas T Treebak
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK2200, Denmark.
| | - David E Gerrard
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA.
| |
Collapse
|
64
|
Kjøbsted R, Hingst JR, Fentz J, Foretz M, Sanz MN, Pehmøller C, Shum M, Marette A, Mounier R, Treebak JT, Wojtaszewski JFP, Viollet B, Lantier L. AMPK in skeletal muscle function and metabolism. FASEB J 2018; 32:1741-1777. [PMID: 29242278 PMCID: PMC5945561 DOI: 10.1096/fj.201700442r] [Citation(s) in RCA: 317] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skeletal muscle possesses a remarkable ability to adapt to various physiologic conditions. AMPK is a sensor of intracellular energy status that maintains energy stores by fine-tuning anabolic and catabolic pathways. AMPK’s role as an energy sensor is particularly critical in tissues displaying highly changeable energy turnover. Due to the drastic changes in energy demand that occur between the resting and exercising state, skeletal muscle is one such tissue. Here, we review the complex regulation of AMPK in skeletal muscle and its consequences on metabolism (e.g., substrate uptake, oxidation, and storage as well as mitochondrial function of skeletal muscle fibers). We focus on the role of AMPK in skeletal muscle during exercise and in exercise recovery. We also address adaptations to exercise training, including skeletal muscle plasticity, highlighting novel concepts and future perspectives that need to be investigated. Furthermore, we discuss the possible role of AMPK as a therapeutic target as well as different AMPK activators and their potential for future drug development.—Kjøbsted, R., Hingst, J. R., Fentz, J., Foretz, M., Sanz, M.-N., Pehmøller, C., Shum, M., Marette, A., Mounier, R., Treebak, J. T., Wojtaszewski, J. F. P., Viollet, B., Lantier, L. AMPK in skeletal muscle function and metabolism.
Collapse
Affiliation(s)
- Rasmus Kjøbsted
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Janne R Hingst
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Joachim Fentz
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Marc Foretz
- INSERM, Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria-Nieves Sanz
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland, and.,Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Christian Pehmøller
- Internal Medicine Research Unit, Pfizer Global Research and Development, Cambridge, Massachusetts, USA
| | - Michael Shum
- Axe Cardiologie, Quebec Heart and Lung Research Institute, Laval University, Québec, Canada.,Institute for Nutrition and Functional Foods, Laval University, Québec, Canada
| | - André Marette
- Axe Cardiologie, Quebec Heart and Lung Research Institute, Laval University, Québec, Canada.,Institute for Nutrition and Functional Foods, Laval University, Québec, Canada
| | - Remi Mounier
- Institute NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM Unité 1217, CNRS UMR, Villeurbanne, France
| | - Jonas T Treebak
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Benoit Viollet
- INSERM, Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA.,Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
65
|
Paula FMM, Leite NC, Borck PC, Freitas-Dias R, Cnop M, Chacon-Mikahil MPT, Cavaglieri CR, Marchetti P, Boschero AC, Zoppi CC, Eizirik DL. Exercise training protects human and rodent β cells against endoplasmic reticulum stress and apoptosis. FASEB J 2018; 32:1524-1536. [PMID: 29133342 DOI: 10.1096/fj.201700710r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Prolonged exercise has positive metabolic effects in obese or diabetic individuals. These effects are usually ascribed to improvements in insulin sensitivity. We evaluated whether exercise also generates circulating signals that protect human and rodent β cells against endoplasmic reticulum (ER) stress and apoptosis. For this purpose, we obtained serum from humans or mice before and after an 8 wk training period. Exposure of human islets or mouse or rat β cells to human or rodent sera, respectively, obtained from trained individuals reduced cytokine (IL-1β+IFN-γ)- or chemical ER stressor-induced β-cell ER stress and apoptosis, at least in part via activation of the transcription factor STAT3. These findings indicate that exercise training improves human and rodent β-cell survival under diabetogenic conditions and support lifestyle interventions as a protective approach for both type 1 and 2 diabetes.-Paula, F. M. M., Leite, N. C., Borck, P. C., Freitas-Dias, R., Cnop, M., Chacon-Mikahil, M. P. T., Cavaglieri, C. R., Marchetti, P., Boschero, A. C., Zoppi, C. C., Eizirik, D. L. Exercise training protects human and rodent β cells against endoplasmic reticulum stress and apoptosis.
Collapse
Affiliation(s)
- Flavia M M Paula
- Center for Diabetes Research, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nayara C Leite
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil
| | - Patricia C Borck
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil
| | - Ricardo Freitas-Dias
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil.,Department of Physical Therapy, University of Pernambuco, Petrolina, Brazil
| | - Miriam Cnop
- Center for Diabetes Research, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Mara P T Chacon-Mikahil
- Exercise Physiology Laboratory (FISEX), Faculty of Physical Education, University of Campinas (UNICAMP), Campinas, Brazil; and
| | - Claudia R Cavaglieri
- Exercise Physiology Laboratory (FISEX), Faculty of Physical Education, University of Campinas (UNICAMP), Campinas, Brazil; and
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Antonio C Boschero
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil
| | - Claudio C Zoppi
- Department of Structural and Functional Biology, Institute of Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, Brazil
| | - Decio L Eizirik
- Center for Diabetes Research, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
66
|
Dodington DW, Desai HR, Woo M. JAK/STAT - Emerging Players in Metabolism. Trends Endocrinol Metab 2018; 29:55-65. [PMID: 29191719 DOI: 10.1016/j.tem.2017.11.001] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway is crucial for transducing signals from a variety of metabolically relevant hormones and cytokines including growth hormone, leptin, erythropoietin, IL4, IL6 and IFNγ. A growing body of evidence suggests that this pathway is dysregulated in the context of obesity and metabolic disease. Recent development of animal models has been instrumental in identifying the role of JAK/STAT signaling in the peripheral metabolic organs including adipose, liver, muscle, pancreas, and the immune system. In this review we summarize current knowledge about the function of JAK/STAT proteins in the regulation of metabolism, and highlight new potential therapeutic targets for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- David W Dodington
- Toronto General Hospital Research Institute, University Health Network, Toronto, M5G 1L7, Canada
| | - Harsh R Desai
- Toronto General Hospital Research Institute, University Health Network, Toronto, M5G 1L7, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, M5G 1L7, Canada; Division of Endocrinology and Metabolism, Department of Medicine, University Health Network and University of Toronto, Toronto, M5G 2C4, Canada.
| |
Collapse
|
67
|
The role of exercise-induced myokines in regulating metabolism. Arch Pharm Res 2017; 41:14-29. [DOI: 10.1007/s12272-017-0994-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 11/21/2017] [Indexed: 12/25/2022]
|
68
|
Pérez-López A, Valadés D, Vázquez Martínez C, de Cos Blanco AI, Bujan J, García-Honduvilla N. Serum IL-15 and IL-15Rα levels are decreased in lean and obese physically active humans. Scand J Med Sci Sports 2017; 28:1113-1120. [PMID: 28940555 DOI: 10.1111/sms.12983] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2017] [Indexed: 12/23/2022]
Abstract
Circulating IL-15 presence is required to stimulate anti-adipogenic effects of the IL-15/IL-15Rα axis in adipose tissue. Although exercise increases blood IL-15 expression post-exercise, it remains inconclusive whether physical activity can alter the baseline concentrations of this cytokine. The aim of this study was to determine whether physical activity regulates circulating IL-15 and IL-15Rα in lean and obese individuals. Two hundred and seventy-six participants were divided into five groups according to physical activity (PA), body mass and type 2 diabetes mellitus (T2DM) diagnosis: (a) lean PA (N = 25); (b) lean non-PA (N = 28); (c) obese PA (N = 64); (d) obese non-PA (N = 79); and (e) obese non-PA with T2DM (N = 80). Serum IL-15 and IL-15Rα, blood glucose/lipid profile and body composition were measured. Serum IL-15 and IL-15Rα decreased in PA participants compared to non-PA (P < .05), while IL-15 and IL-15Rα increased in obese with T2DM compared to obese without T2DM (P < .05). No differences were observed between lean non-PA and obese PA. Serum IL-15Rα was associated with fasting glucose (R2 = .063), insulin (R2 = .082), HbA1c (R2 = .108), and HOMA (R2 = .057) in obese participants. Circulating IL-15 and IL-15Rα are reduced in lean and obese participants who perform physical activity regularly (≥180 min/week), suggesting a regulative role of physical activity on the circulating concentrations of IL-15 and IL-15Rα at baseline. Moreover, the relationship observed between IL-15Rα and glucose profile may indicate a role of the alpha receptor in glucose metabolism.
Collapse
Affiliation(s)
- A Pérez-López
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Department of Biomedical Sciences, Area of Sport and Physical Education, Faculty of Medicine and Health Sciences, University of Alcalá, Madrid, Spain
| | - D Valadés
- Department of Biomedical Sciences, Area of Sport and Physical Education, Faculty of Medicine and Health Sciences, University of Alcalá, Madrid, Spain
| | - C Vázquez Martínez
- Department of Endocrinology and Nutrition, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | | | - J Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - N García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
69
|
Duan Y, Li F, Wang W, Guo Q, Wen C, Li Y, Yin Y. Interleukin-15 in obesity and metabolic dysfunction: current understanding and future perspectives. Obes Rev 2017; 18:1147-1158. [PMID: 28752527 DOI: 10.1111/obr.12567] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/16/2017] [Accepted: 04/28/2017] [Indexed: 12/13/2022]
Abstract
Obesity rises rapidly and is a major health concern for modern people. Importantly, it is a major risk factor in the development of numerous chronic diseases such as type 2 diabetes mellitus (T2DM). Recently, interleukin (IL)-15 has attracted considerable attention as a potential regulator for the prevention and/or treatment of obesity and T2DM. The beneficial effects include increased loss of fat mass and body weight, improved lipid and glucose metabolism, reduced white adipose tissue inflammation, enhanced mitochondrial function, alterations in the composition of muscle fibres and gut bacterial and attenuated endoplasmic reticulum stress. Although these beneficial effects are somewhat controversial, IL-15, exogenously delivered or endogenously produced, may be a promising target in the prevention and treatment of obesity and T2DM.
Collapse
Affiliation(s)
- Y Duan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - F Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan, China.,Hunan Co-Innovation Center of Safety Animal Production, CICSAP, Changsha, China
| | - W Wang
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, China
| | - Q Guo
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - C Wen
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, China
| | - Y Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Y Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan, China.,Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, China
| |
Collapse
|
70
|
Carson BP. The Potential Role of Contraction-Induced Myokines in the Regulation of Metabolic Function for the Prevention and Treatment of Type 2 Diabetes. Front Endocrinol (Lausanne) 2017; 8:97. [PMID: 28512448 PMCID: PMC5411437 DOI: 10.3389/fendo.2017.00097] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/18/2017] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle represents the largest organ in the body, comprises 36-42% of body weight, and has recently been recognized as having an endocrine function. Proteins expressed and released by muscle that have autocrine, paracrine, and endocrine bioactivities have been termed myokines. It is likely that muscle contraction represents the primary stimulus for the synthesis and secretion of myokines to enable communication with other organs such as the liver, adipose tissue, brain, and auto-regulation of muscle metabolism. To date, several hundred myokines in the muscle secretome have been identified, a sub-population of which are specifically induced by skeletal muscle contraction. However, the bioactivity of many of these myokines and the mechanism through which they act has either not yet been characterized or remains poorly understood. Physical activity and exercise are recognized as a central tenet in both the prevention and treatment of type 2 diabetes (T2D). Recent data suggest humoral factors such as muscle-derived secretory proteins may mediate the beneficial effects of exercise in the treatment of metabolic diseases. This mini-review aims to summarize our current knowledge on the role of contraction-induced myokines in mediating the beneficial effects of physical activity and exercise in the prevention and treatment of T2D, specifically glucose and lipid metabolism. Future directions as to how we can optimize contraction-induced myokine secretion to inform exercise protocols for the prevention and treatment of T2D will also be discussed.
Collapse
Affiliation(s)
- Brian P. Carson
- Health Research Institute, Physical Education and Sport Sciences, University of Limerick, Limerick, Ireland
- *Correspondence: Brian P. Carson,
| |
Collapse
|