Signaling specificity of interleukin-6 action on glucose and lipid metabolism in skeletal muscle.

We identified signaling pathways by which IL-6 regulates skeletal muscle differentiation and metabolism. Primary human skeletal muscle cells were exposed to IL-6 (25 ng/ml either acutely or for several days), and small interfering RNA gene silencing was applied to measure glucose and fat metabolism. Chronic IL-6 exposure increased myotube fusion and formation and the mRNA expression of glucose transporter 4, peroxisome proliferator activated receptor (PPAR)alpha, PPARdelta, PPARgamma, PPARgamma coactivator 1, glycogen synthase, myocyte enhancer factor 2D, uncoupling protein 2, fatty acid transporter 4, and IL-6 (P < 0.05), whereas glucose transporter 1, CCAAT/enhancer-binding protein-alpha, and uncoupling protein 3 were decreased. IL-6 increased glucose incorporation into glycogen, glucose uptake, lactate production, and fatty acid uptake and oxidation, concomitant with increased phosphorylation of AMP-activated protein kinase (AMPK), signal transducer and activator of transcription 3, and ERK1/2. IL-6 also increased phosphatidylinositol (PI) 3-kinase activity (450%; P < 0.05), which was blunted by subsequent insulin-stimulation (P < 0.05). IL-6-mediated glucose metabolism was suppressed, but lipid metabolism was unaltered, by inhibition of PI3-kinase with LY294002. The small interfering RNA-directed depletion of AMPK reduced IL-6-mediated fatty acid oxidation and palmitate uptake but did not reduce glycogen synthesis. In summary, IL-6 increases glycogen synthesis via a PI3-kinase-dependent mechanism and enhances lipid oxidation via an AMPK-dependent mechanism in skeletal muscle. Thus, IL-6 directly promotes skeletal muscle differentiation and regulates muscle substrate utilization, promoting glycogen storage and lipid oxidation.

[1]  E. Schleicher,et al.  Direct Cross-talk of Interleukin-6 and Insulin Signal Transduction via Insulin Receptor Substrate-1 in Skeletal Muscle Cells* , 2006, Journal of Biological Chemistry.

[2]  D. Neculoiu,et al.  Evaluation of viral replication in children with chronic hepatitis B with and without interferon treatment. , 2005, Romanian journal of gastroenterology.

[3]  M. T. Nunes,et al.  NF-κB, MEF2A, MEF2D and HIF1-a involvement on insulin- and contraction-induced regulation of GLUT4 gene expression in soleus muscle , 2005, Molecular and Cellular Endocrinology.

[4]  P. Neufer,et al.  Substrate availability and transcriptional regulation of metabolic genes in human skeletal muscle during recovery from exercise. , 2005, Metabolism: clinical and experimental.

[5]  R. Mooney,et al.  Interleukin-6 depletion selectively improves hepatic insulin action in obesity. , 2005, Endocrinology.

[6]  H. El-Gabalawy,et al.  Emerging biologic therapies in rheumatoid arthritis: cell targets and cytokines , 2005, Current opinion in rheumatology.

[7]  F. Lönnqvist,et al.  Enhanced insulin-stimulated glycogen synthesis in response to insulin, metformin or rosiglitazone is associated with increased mRNA expression of GLUT4 and peroxisomal proliferator activator receptor gamma co-activator 1 , 2005, Diabetologia.

[8]  A. Sharma,et al.  Obesity, hypertension and insulin resistance , 2005, Acta Diabetologica.

[9]  Bente Klarlund Pedersen,et al.  The anti-inflammatory effect of exercise. , 2005, Journal of applied physiology.

[10]  S. Shoelson,et al.  Local and systemic insulin resistance resulting from hepatic activation of IKK-β and NF-κB , 2005, Nature Medicine.

[11]  M. Febbraio,et al.  Acute IL-6 treatment increases fatty acid turnover in elderly humans in vivo and in tissue culture in vitro. , 2005, American journal of physiology. Endocrinology and metabolism.

[12]  S. Jee,et al.  Interleukin-6 induced basic fibroblast growth factor-dependent angiogenesis in basal cell carcinoma cell line via JAK/STAT3 and PI3-kinase/Akt pathways. , 2004, The Journal of investigative dermatology.

[13]  U. Smith,et al.  Short-term infusion of interleukin-6 does not induce insulin resistance in vivo or impair insulin signalling in rats , 2004, Diabetologia.

[14]  M. Febbraio,et al.  Suppression of plasma free fatty acids upregulates peroxisome proliferator-activated receptor (PPAR) alpha and delta and PPAR coactivator 1alpha in human skeletal muscle, but not lipid regulatory genes. , 2004, Journal of molecular endocrinology.

[15]  C. Bruce,et al.  Cytokine regulation of skeletal muscle fatty acid metabolism: effect of interleukin-6 and tumor necrosis factor-alpha. , 2004, American journal of physiology. Endocrinology and metabolism.

[16]  G. Hjälm,et al.  The 5′-AMP-activated Protein Kinase γ3 Isoform Has a Key Role in Carbohydrate and Lipid Metabolism in Glycolytic Skeletal Muscle* , 2004, Journal of Biological Chemistry.

[17]  B. Pedersen,et al.  AMPK activity is diminished in tissues of IL-6 knockout mice: the effect of exercise. , 2004, Biochemical and biophysical research communications.

[18]  M. Febbraio,et al.  Interleukin-6 and insulin sensitivity: friend or foe? , 2004, Diabetologia.

[19]  M. Febbraio,et al.  Skeletal myocytes are a source of interleukin‐6 mRNA expression and protein release during contraction: evidence of fiber type specificity , 2004, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[20]  L. Kuller,et al.  The Chronic Inflammatory Hypothesis for the Morbidity Associated with Morbid Obesity: Implications and Effects of Weight Loss , 2004, Obesity surgery.

[21]  G. Cline,et al.  Differential effects of interleukin-6 and -10 on skeletal muscle and liver insulin action in vivo. , 2004, Diabetes.

[22]  P. Muñoz-Cánoves,et al.  p38 MAPK-induced nuclear factor-kappaB activity is required for skeletal muscle differentiation: role of interleukin-6. , 2004, Molecular biology of the cell.

[23]  M. Ishii,et al.  Cytoplasmic c‐Fos induced by the YXXQ‐derived STAT3 signal requires the co‐operative MEK/ERK signal for its nuclear translocation , 2004, Genes to cells : devoted to molecular & cellular mechanisms.

[24]  U. Smith,et al.  High local concentrations and effects on differentiation implicate interleukin-6 as a paracrine regulator. , 2004, Obesity research.

[25]  T. Kanda,et al.  Interleukin-6 and cardiovascular diseases. , 2004, Japanese heart journal.

[26]  L. Goodyear,et al.  5’ Adenosine Monophosphate-Activated Protein Kinase, Metabolism and Exercise , 2004, Sports medicine.

[27]  D. Constantin-Teodosiu,et al.  The effect of repeated muscle biopsy sampling on ATP and glycogen resynthesis following exercise in man , 2004, European Journal of Applied Physiology and Occupational Physiology.

[28]  U. Smith,et al.  Interleukin-6 (IL-6) Induces Insulin Resistance in 3T3-L1 Adipocytes and Is, Like IL-8 and Tumor Necrosis Factor-α, Overexpressed in Human Fat Cells from Insulin-resistant Subjects* , 2003, Journal of Biological Chemistry.

[29]  A. Steensberg,et al.  Interleukin-6 production by contracting human skeletal muscle: autocrine regulation by IL-6. , 2003, Biochemical and biophysical research communications.

[30]  A. Steensberg,et al.  IL-6 enhances plasma IL-1ra, IL-10, and cortisol in humans. , 2003, American journal of physiology. Endocrinology and metabolism.

[31]  A. Krook,et al.  RNA interference-mediated reduction in GLUT1 inhibits serum-induced glucose transport in primary human skeletal muscle cells. , 2003, Biochemical and biophysical research communications.

[32]  B. Saltin,et al.  Interleukin-6 stimulates lipolysis and fat oxidation in humans. , 2003, The Journal of clinical endocrinology and metabolism.

[33]  M. Febbraio,et al.  Skeletal muscle interleukin-6 and tumor necrosis factor-alpha release in healthy subjects and patients with type 2 diabetes at rest and during exercise. , 2003, Metabolism: clinical and experimental.

[34]  S. Keller The insulin-regulated aminopeptidase: a companion and regulator of GLUT4. , 2003, Frontiers in bioscience : a journal and virtual library.

[35]  T. Zimmers,et al.  Suppressor of Cytokine Signaling-3 (SOCS-3), a Potential Mediator of Interleukin-6-dependent Insulin Resistance in Hepatocytes* , 2003, The Journal of Biological Chemistry.

[36]  C. Stewart,et al.  Role of insulin‐like growth factor binding protein‐3 (IGFBP‐3) in the differentiation of primary human adult skeletal myoblasts , 2003, Journal of cellular physiology.

[37]  N. Jessen,et al.  Effects of AICAR and exercise on insulin-stimulated glucose uptake, signaling, and GLUT-4 content in rat muscles. , 2003, Journal of applied physiology.

[38]  Stefan Rose-John,et al.  The human interleukin‐6 (IL‐6) receptor exists as a preformed dimer in the plasma membrane , 2003, FEBS letters.

[39]  E. Kraegen,et al.  Malonyl-CoA and AMP-activated protein kinase (AMPK): possible links between insulin resistance in muscle and early endothelial cell damage in diabetes. , 2001, Biochemical Society transactions.

[40]  M. Febbraio Signaling pathways for IL-6 within skeletal muscle. , 2003, Exercise immunology review.

[41]  M. Febbraio,et al.  IL-6 and TNF-α expression in, and release from, contracting human skeletal muscle , 2002 .

[42]  R. Mooney,et al.  Interleukin-6 induces cellular insulin resistance in hepatocytes. , 2002, Diabetes.

[43]  M. Febbraio,et al.  Muscle‐derived interleukin‐6: mechanisms for activation and possible biological roles , 2002, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[44]  J. Zierath,et al.  Isoform-specific regulation of 5' AMP-activated protein kinase in skeletal muscle from obese Zucker (fa/fa) rats in response to contraction. , 2002, Diabetes.

[45]  Olle Ljunqvist,et al.  Metformin increases AMP-activated protein kinase activity in skeletal muscle of subjects with type 2 diabetes. , 2002, Diabetes.

[46]  E. Ravussin,et al.  Increased Fat Intake, Impaired Fat Oxidation, and Failure of Fat Cell Proliferation Result in Ectopic Fat Storage, Insulin Resistance, and Type 2 Diabetes Mellitus , 2002, Annals of the New York Academy of Sciences.

[47]  C. Marboe,et al.  The role of IL6 cytokines in acute cardiac allograft rejection. , 2002, Transplant immunology.

[48]  W. Winder Energy-sensing and signaling by AMP-activated protein kinase in skeletal muscle. , 2001, Journal of applied physiology.

[49]  L Wood,et al.  Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. , 2001, American journal of physiology. Endocrinology and metabolism.

[50]  L. Goodyear,et al.  5' AMP-activated protein kinase activation causes GLUT4 translocation in skeletal muscle. , 1999, Diabetes.

[51]  D. Hardie,et al.  AMP-activated protein kinase, a metabolic master switch: possible roles in Type 2 diabetes. , 1999, American journal of physiology. Endocrinology and metabolism.

[52]  B. Kemp,et al.  Dealing with energy demand: the AMP-activated protein kinase. , 1999, Trends in biochemical sciences.

[53]  G. Hill Cardiopulmonary bypass-induced inflammation: is it important? , 1998, Journal of cardiothoracic and vascular anesthesia.

[54]  R. Somwar,et al.  Stimulation of glucose and amino acid transport and activation of the insulin signaling pathways by insulin lispro in L6 skeletal muscle cells. , 1998, Clinical therapeutics.

[55]  J S Yudkin,et al.  Journal of Clinical Endocrinology and Metabolism Printed in U.S.A. Copyright © 1997 by The Endocrine Society Subcutaneous Adipose Tissue Releases Interleukin-6, But Not Tumor Necrosis Factor-�, in Vivo* , 2022 .

[56]  H. Tilg,et al.  IL-6 and APPs: anti-inflammatory and immunosuppressive mediators. , 1997, Immunology today.

[57]  Michel Georges,et al.  A deletion in the bovine myostatin gene causes the double–muscled phenotype in cattle , 1997, Nature Genetics.

[58]  Hurel Sj Insulin action in cultured human skeletal muscle. , 1997 .

[59]  G. Kannourakis,et al.  Leukemia inhibitory factor and interleukin‐6 are produced by diseased and regenerating skeletal muscle , 1996, Muscle & nerve.

[60]  S. Okazaki,et al.  Effects of calcitonin gene‐related peptide and interleukin 6 on myoblast differentiation , 1996, Cell proliferation.

[61]  S. O’Rahilly,et al.  Two Naturally Occurring Mutant Insulin Receptors Phosphorylate Insulin Receptor Substrate-1 (IRS-1) but Fail to Mediate the Biological Effects of Insulin , 1996, The Journal of Biological Chemistry.

[62]  H. Goebel,et al.  Cytokine Expression Profile in Idiopathic Inflammatory Myopathies , 1996, Journal of neuropathology and experimental neurology.

[63]  R. Henry,et al.  The expression of TNF alpha by human muscle. Relationship to insulin resistance. , 1996, The Journal of clinical investigation.

[64]  A. Billiau,et al.  Anti-gamma interferon and anti-interleukin-6 antibodies affect staphylococcal enterotoxin B-induced weight loss, hypoglycemia, and cytokine release in D-galactosamine-sensitized and unsensitized mice , 1995, Infection and immunity.

[65]  M. Kluger,et al.  Cytokines and the acute phase response to influenza virus in mice. , 1995, The American journal of physiology.

[66]  R. Hohlfeld,et al.  Constitutive and cytokine-induced production of interleukin-6 by human myoblasts. , 1994, Immunology letters.

[67]  P. Heinrich,et al.  Protein kinase C activity is rate limiting for shedding of the interleukin-6 receptor. , 1992, Biochemical and biophysical research communications.

[68]  C. Dinarello,et al.  Correlations and interactions in the production of interleukin-6 (IL-6), IL-1, and tumor necrosis factor (TNF) in human blood mononuclear cells: IL-6 suppresses IL-1 and TNF. , 1990, Blood.

[69]  K. Kristensson,et al.  Interferon-gamma-like immunoreactivity and T-cell marker expression in rat skeletal muscle fibres , 1989, Brain Research.

[70]  T. Hirano,et al.  Interleukin-6 triggers the association of its receptor with a possible signal transducer, gp130 , 1989, Cell.

[71]  Blom Pc,et al.  Exhaustive running: inappropriate as a stimulus of muscle glycogen super-compensation. , 1987 .

[72]  N. Vøllestad,et al.  Exhaustive running: inappropriate as a stimulus of muscle glycogen super-compensation. , 1987, Medicine and science in sports and exercise.