Skip to main content
Top
Published in: Diabetologia 11/2012

01-11-2012 | Article

Reduced Socs3 expression in adipose tissue protects female mice against obesity-induced insulin resistance

Authors: R. Palanivel, M. D. Fullerton, S. Galic, J. Honeyman, K. A. Hewitt, S. B. Jorgensen, G. R. Steinberg

Published in: Diabetologia | Issue 11/2012

Login to get access

Abstract

Aims/hypothesis

Inflammation in obesity increases the levels of the suppressor of cytokine signalling-3 (SOCS3) protein in adipose tissue, but the physiological importance of this protein in regulating whole-body insulin sensitivity in obesity is not known.

Methods

We generated Socs3 floxed (wild-type, WT) and Socs3 aP2 (also known as Fabp4)-Cre null (Socs3 AKO) mice. Mice were maintained on either a regular chow or a high-fat diet (HFD) for 16 weeks during which time body mass, adiposity, glucose homeostasis and insulin sensitivity were assessed.

Results

The HFD increased SOCS3 levels in adipose tissue of WT but not Socs3 AKO mice. WT and Socs3 AKO mice had similar body mass and adiposity, assessed using computed tomography (CT) imaging, irrespective of diet or sex. On a control chow diet there were no differences in insulin sensitivity or glucose tolerance. When fed a HFD, female but not male Socs3 AKO mice had improved glucose tolerance as well as lower fasting glucose and insulin levels compared with WT littermates. Hyperinsulinaemic–euglycaemic clamps and positron emission tomography (PET) imaging demonstrated that improved insulin sensitivity was due to elevated adipose tissue glucose uptake. Increased insulin-stimulated glucose uptake in adipose tissue was associated with enhanced levels and activating phosphorylation of insulin receptor substrate-1 (IRS1).

Conclusions/interpretation

These data demonstrate that inhibiting SOCS3 production in adipose tissue of female mice is effective for improving whole-body insulin sensitivity in obesity.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Mori H, Hanada R, Hanada T et al (2004) Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity. Nat Med 10:739–743PubMedCrossRef Mori H, Hanada R, Hanada T et al (2004) Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity. Nat Med 10:739–743PubMedCrossRef
3.
go back to reference Myers MG Jr (2004) Leptin receptor signaling and the regulation of mammalian physiology. Recent Prog Horm Res 59:287–304PubMedCrossRef Myers MG Jr (2004) Leptin receptor signaling and the regulation of mammalian physiology. Recent Prog Horm Res 59:287–304PubMedCrossRef
4.
go back to reference Ueki K, Kondo T, Kahn CR (2004) Suppressor of cytokine signaling 1 (SOCS-1) and SOCS-3 cause insulin resistance through inhibition of tyrosine phosphorylation of insulin receptor substrate proteins by discrete mechanisms. Mol Cell Biol 24:5434–5446PubMedCrossRef Ueki K, Kondo T, Kahn CR (2004) Suppressor of cytokine signaling 1 (SOCS-1) and SOCS-3 cause insulin resistance through inhibition of tyrosine phosphorylation of insulin receptor substrate proteins by discrete mechanisms. Mol Cell Biol 24:5434–5446PubMedCrossRef
5.
go back to reference Steinberg GR, Smith AC, Wormald S, Malenfant P, Collier C, Dyck DJ (2004) Endurance training partially reverses dietary-induced leptin resistance in rodent skeletal muscle. Am J Physiol Endocrinol Metab 286:E57–E63PubMedCrossRef Steinberg GR, Smith AC, Wormald S, Malenfant P, Collier C, Dyck DJ (2004) Endurance training partially reverses dietary-induced leptin resistance in rodent skeletal muscle. Am J Physiol Endocrinol Metab 286:E57–E63PubMedCrossRef
6.
go back to reference Steinberg GR, McAinch AJ, Chen MB et al (2006) The suppressor of cytokine signaling 3 inhibits leptin activation of AMP-kinase in cultured skeletal muscle of obese humans. J Clin Endocrinol Metab 91:3592–3597PubMedCrossRef Steinberg GR, McAinch AJ, Chen MB et al (2006) The suppressor of cytokine signaling 3 inhibits leptin activation of AMP-kinase in cultured skeletal muscle of obese humans. J Clin Endocrinol Metab 91:3592–3597PubMedCrossRef
7.
go back to reference Emanuelli B, Peraldi P, Filloux C et al (2001) SOCS-3 inhibits insulin signaling and is up-regulated in response to tumor necrosis factor-alpha in the adipose tissue of obese mice. J Biol Chem 276:47944–47949PubMed Emanuelli B, Peraldi P, Filloux C et al (2001) SOCS-3 inhibits insulin signaling and is up-regulated in response to tumor necrosis factor-alpha in the adipose tissue of obese mice. J Biol Chem 276:47944–47949PubMed
8.
go back to reference Wang Z, Zhou YT, Kakuma T et al (2000) Leptin resistance of adipocytes in obesity: role of suppressors of cytokine signaling. Biochem Biophys Res Commun 277:20–26PubMedCrossRef Wang Z, Zhou YT, Kakuma T et al (2000) Leptin resistance of adipocytes in obesity: role of suppressors of cytokine signaling. Biochem Biophys Res Commun 277:20–26PubMedCrossRef
9.
go back to reference Ghanim H, Aljada A, Daoud N, Deopurkar R, Chaudhuri A, Dandona P (2007) Role of inflammatory mediators in the suppression of insulin receptor phosphorylation in circulating mononuclear cells of obese subjects. Diabetologia 50:278–285PubMedCrossRef Ghanim H, Aljada A, Daoud N, Deopurkar R, Chaudhuri A, Dandona P (2007) Role of inflammatory mediators in the suppression of insulin receptor phosphorylation in circulating mononuclear cells of obese subjects. Diabetologia 50:278–285PubMedCrossRef
10.
go back to reference Ghanim H, Abuaysheh S, Sia CL et al (2009) Increase in plasma endotoxin concentrations and the expression of Toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: implications for insulin resistance. Diabetes Care 32:2281–2287PubMedCrossRef Ghanim H, Abuaysheh S, Sia CL et al (2009) Increase in plasma endotoxin concentrations and the expression of Toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: implications for insulin resistance. Diabetes Care 32:2281–2287PubMedCrossRef
11.
go back to reference Emanuelli B, Peraldi P, Filloux C, Sawka-Verhelle D, Hilton D, van Obberghen E (2000) SOCS-3 is an insulin-induced negative regulator of insulin signaling. J Biol Chem 275:15985–15991PubMedCrossRef Emanuelli B, Peraldi P, Filloux C, Sawka-Verhelle D, Hilton D, van Obberghen E (2000) SOCS-3 is an insulin-induced negative regulator of insulin signaling. J Biol Chem 275:15985–15991PubMedCrossRef
12.
go back to reference Senn JJ, Klover PJ, Nowak IA et al (2003) Suppressor of cytokine signaling-3 (SOCS-3), a potential mediator of interleukin-6-dependent insulin resistance in hepatocytes. J Biol Chem 278:13740–13746PubMedCrossRef Senn JJ, Klover PJ, Nowak IA et al (2003) Suppressor of cytokine signaling-3 (SOCS-3), a potential mediator of interleukin-6-dependent insulin resistance in hepatocytes. J Biol Chem 278:13740–13746PubMedCrossRef
13.
go back to reference Rui L, Yuan M, Frantz D, Shoelson S, White MF (2002) SOCS-1 and SOCS-3 block insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2. J Biol Chem 277:42394–42398PubMedCrossRef Rui L, Yuan M, Frantz D, Shoelson S, White MF (2002) SOCS-1 and SOCS-3 block insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2. J Biol Chem 277:42394–42398PubMedCrossRef
14.
go back to reference Shi H, Tzameli I, Bjorbaek C, Flier JS (2004) Suppressor of cytokine signaling 3 is a physiological regulator of adipocyte insulin signaling. J Biol Chem 279:34733–34740PubMedCrossRef Shi H, Tzameli I, Bjorbaek C, Flier JS (2004) Suppressor of cytokine signaling 3 is a physiological regulator of adipocyte insulin signaling. J Biol Chem 279:34733–34740PubMedCrossRef
15.
go back to reference Bjorbaek C, Lavery HJ, Bates SH et al (2000) SOCS3 mediates feedback inhibition of the leptin receptor via Tyr985. J Biol Chem 275:40649–40657CrossRef Bjorbaek C, Lavery HJ, Bates SH et al (2000) SOCS3 mediates feedback inhibition of the leptin receptor via Tyr985. J Biol Chem 275:40649–40657CrossRef
16.
go back to reference Bjorbaek C, El-Haschimi K, Frantz JD, Flier JS (1999) The role of SOCS-3 in leptin signaling and leptin resistance. J Biol Chem 274:30059–30065PubMedCrossRef Bjorbaek C, El-Haschimi K, Frantz JD, Flier JS (1999) The role of SOCS-3 in leptin signaling and leptin resistance. J Biol Chem 274:30059–30065PubMedCrossRef
17.
go back to reference Howard JK, Cave BJ, Oksanen LJ, Tzameli I, Bjorbaek C, Flier JS (2004) Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of Socs3. Nat Med 10:734–738PubMedCrossRef Howard JK, Cave BJ, Oksanen LJ, Tzameli I, Bjorbaek C, Flier JS (2004) Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of Socs3. Nat Med 10:734–738PubMedCrossRef
18.
go back to reference Reed AS, Unger EK, Olofsson LE, Piper ML, Myers MG Jr, Xu AW (2010) Functional role of suppressor of cytokine signaling 3 upregulation in hypothalamic leptin resistance and long-term energy homeostasis. Diabetes 59:894–906PubMedCrossRef Reed AS, Unger EK, Olofsson LE, Piper ML, Myers MG Jr, Xu AW (2010) Functional role of suppressor of cytokine signaling 3 upregulation in hypothalamic leptin resistance and long-term energy homeostasis. Diabetes 59:894–906PubMedCrossRef
19.
go back to reference Bjornholm M, Munzberg H, Leshan RL et al (2007) Mice lacking inhibitory leptin receptor signals are lean with normal endocrine function. J Clin Invest 117:1354–1360PubMedCrossRef Bjornholm M, Munzberg H, Leshan RL et al (2007) Mice lacking inhibitory leptin receptor signals are lean with normal endocrine function. J Clin Invest 117:1354–1360PubMedCrossRef
20.
go back to reference Wang MY, Orci L, Ravazzola M, Unger RH (2005) Fat storage in adipocytes requires inactivation of leptin's paracrine activity: implications for treatment of human obesity. Proc Natl Acad Sci U S A 102:18011–18016PubMedCrossRef Wang MY, Orci L, Ravazzola M, Unger RH (2005) Fat storage in adipocytes requires inactivation of leptin's paracrine activity: implications for treatment of human obesity. Proc Natl Acad Sci U S A 102:18011–18016PubMedCrossRef
21.
go back to reference Wang MY, Lee Y, Unger RH (1999) Novel form of lipolysis induced by leptin. J Biol Chem 274:17541–17544PubMedCrossRef Wang MY, Lee Y, Unger RH (1999) Novel form of lipolysis induced by leptin. J Biol Chem 274:17541–17544PubMedCrossRef
22.
go back to reference Orci L, Cook WS, Ravazzola M et al (2004) Rapid transformation of white adipocytes into fat-oxidizing machines. Proc Natl Acad Sci U S A 101:2058–2063PubMedCrossRef Orci L, Cook WS, Ravazzola M et al (2004) Rapid transformation of white adipocytes into fat-oxidizing machines. Proc Natl Acad Sci U S A 101:2058–2063PubMedCrossRef
23.
go back to reference Gaidhu MP, Anthony NM, Patel P, Hawke TJ, Ceddia RB (2010) Dysregulation of lipolysis and lipid metabolism in visceral and subcutaneous adipocytes by high-fat diet: role of ATGL, HSL, and AMPK. Am J Physiol Cell Physiol 298:C961–C971PubMedCrossRef Gaidhu MP, Anthony NM, Patel P, Hawke TJ, Ceddia RB (2010) Dysregulation of lipolysis and lipid metabolism in visceral and subcutaneous adipocytes by high-fat diet: role of ATGL, HSL, and AMPK. Am J Physiol Cell Physiol 298:C961–C971PubMedCrossRef
24.
go back to reference Gauthier MS, O’Brien EL, Bigornia S et al (2010) Decreased AMP-activated protein kinase activity is associated with increased inflammation in visceral adipose tissue and with whole-body insulin resistance in morbidly obese humans. Biochem Biophys Res Commun 404:382–387PubMedCrossRef Gauthier MS, O’Brien EL, Bigornia S et al (2010) Decreased AMP-activated protein kinase activity is associated with increased inflammation in visceral adipose tissue and with whole-body insulin resistance in morbidly obese humans. Biochem Biophys Res Commun 404:382–387PubMedCrossRef
25.
go back to reference Krebs DL, Hilton DJ (2001) SOCS proteins: negative regulators of cytokine signaling. Stem Cells 19:378–387PubMedCrossRef Krebs DL, Hilton DJ (2001) SOCS proteins: negative regulators of cytokine signaling. Stem Cells 19:378–387PubMedCrossRef
26.
go back to reference Shi H, Cave B, Inouye K, Bjorbaek C, Flier JS (2006) Overexpression of suppressor of cytokine signaling 3 in adipose tissue causes local but not systemic insulin resistance. Diabetes 55:699–707PubMedCrossRef Shi H, Cave B, Inouye K, Bjorbaek C, Flier JS (2006) Overexpression of suppressor of cytokine signaling 3 in adipose tissue causes local but not systemic insulin resistance. Diabetes 55:699–707PubMedCrossRef
27.
go back to reference Jamieson E, Chong MM, Steinberg GR et al (2005) Socs1 deficiency enhances hepatic insulin signaling. J Biol Chem 280:31516–31521PubMedCrossRef Jamieson E, Chong MM, Steinberg GR et al (2005) Socs1 deficiency enhances hepatic insulin signaling. J Biol Chem 280:31516–31521PubMedCrossRef
28.
go back to reference Torisu T, Sato N, Yoshiga D et al (2007) The dual function of hepatic SOCS3 in insulin resistance in vivo. Genes Cells 12:143–154PubMedCrossRef Torisu T, Sato N, Yoshiga D et al (2007) The dual function of hepatic SOCS3 in insulin resistance in vivo. Genes Cells 12:143–154PubMedCrossRef
29.
go back to reference Sachithanandan N, Fam BC, Fynch S et al (2010) Liver specific SOCS3 deletion in mice enhances hepatic insulin sensitivity and lipogenesis resulting in fatty liver and obesity. Hepatology 52:1632–1642PubMedCrossRef Sachithanandan N, Fam BC, Fynch S et al (2010) Liver specific SOCS3 deletion in mice enhances hepatic insulin sensitivity and lipogenesis resulting in fatty liver and obesity. Hepatology 52:1632–1642PubMedCrossRef
30.
go back to reference Kiu H, Greenhalgh CJ, Thaus A et al (2009) Regulation of multiple cytokine signalling pathways by SOCS3 is independent of SOCS2. Growth Factors 27:384–393PubMedCrossRef Kiu H, Greenhalgh CJ, Thaus A et al (2009) Regulation of multiple cytokine signalling pathways by SOCS3 is independent of SOCS2. Growth Factors 27:384–393PubMedCrossRef
31.
go back to reference Abel ED, Peroni O, Kim JK et al (2001) Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 409:729–733PubMedCrossRef Abel ED, Peroni O, Kim JK et al (2001) Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 409:729–733PubMedCrossRef
32.
go back to reference Galic S, Fullerton MD, Schertzer JD et al (2011) Hematopoietic AMPK beta1 reduces mouse adipose tissue macrophage inflammation and insulin resistance in obesity. J Clin Invest 121:4903–4915PubMedCrossRef Galic S, Fullerton MD, Schertzer JD et al (2011) Hematopoietic AMPK beta1 reduces mouse adipose tissue macrophage inflammation and insulin resistance in obesity. J Clin Invest 121:4903–4915PubMedCrossRef
33.
go back to reference O’Neill HM, Maarbjerg SJ, Crane JD et al (2011) AMP-activated protein kinase (AMPK) beta1beta2 muscle null mice reveal an essential role for AMPK in maintaining mitochondrial content and glucose uptake during exercise. Proc Natl Acad Sci U S A 108:16092–16097PubMedCrossRef O’Neill HM, Maarbjerg SJ, Crane JD et al (2011) AMP-activated protein kinase (AMPK) beta1beta2 muscle null mice reveal an essential role for AMPK in maintaining mitochondrial content and glucose uptake during exercise. Proc Natl Acad Sci U S A 108:16092–16097PubMedCrossRef
34.
go back to reference Dzamko N, Vandenderen BJ, Hevener AL et al (2010) AMPK β1 deletion reduces appetite preventing obesity and hepatic insulin resistance. J Biol Chem 285:115–122PubMedCrossRef Dzamko N, Vandenderen BJ, Hevener AL et al (2010) AMPK β1 deletion reduces appetite preventing obesity and hepatic insulin resistance. J Biol Chem 285:115–122PubMedCrossRef
35.
go back to reference Saab C, Labiris N, Chirakal R, Farncombe T (2007) A database of normal PET and SPECT radiotracer distributions in rats and mice. J Nucl Med Meeting Abstracts 48:292P-b- Saab C, Labiris N, Chirakal R, Farncombe T (2007) A database of normal PET and SPECT radiotracer distributions in rats and mice. J Nucl Med Meeting Abstracts 48:292P-b-
36.
go back to reference Watt MJ, Dzamko N, Thomas WG et al (2006) CNTF reverses obesity-induced insulin resistance by activating skeletal muscle AMPK. Nat Med 12:541–548PubMedCrossRef Watt MJ, Dzamko N, Thomas WG et al (2006) CNTF reverses obesity-induced insulin resistance by activating skeletal muscle AMPK. Nat Med 12:541–548PubMedCrossRef
37.
go back to reference Chen Z-P, McConell GK, Michell BJ, Snow RJ, Canny BJ, Kemp BE (2000) AMPK signaling in contracting human skeletal muscle: acetyl-CoA carboxylase and NO synthase phosphorylation. Am J Physiol 279:E1202–E1206 Chen Z-P, McConell GK, Michell BJ, Snow RJ, Canny BJ, Kemp BE (2000) AMPK signaling in contracting human skeletal muscle: acetyl-CoA carboxylase and NO synthase phosphorylation. Am J Physiol 279:E1202–E1206
38.
go back to reference Maeda K, Cao H, Kono K et al (2005) Adipocyte/macrophage fatty acid binding proteins control integrated metabolic responses in obesity and diabetes. Cell Metab 1:107–119PubMedCrossRef Maeda K, Cao H, Kono K et al (2005) Adipocyte/macrophage fatty acid binding proteins control integrated metabolic responses in obesity and diabetes. Cell Metab 1:107–119PubMedCrossRef
39.
go back to reference Jiang C, Qu A, Matsubara T et al (2011) Disruption of hypoxia-inducible factor 1 in adipocytes improves insulin sensitivity and decreases adiposity in high-fat diet-fed mice. Diabetes 60:2484–2495PubMedCrossRef Jiang C, Qu A, Matsubara T et al (2011) Disruption of hypoxia-inducible factor 1 in adipocytes improves insulin sensitivity and decreases adiposity in high-fat diet-fed mice. Diabetes 60:2484–2495PubMedCrossRef
40.
go back to reference Wueest S, Rapold RA, Schumann DM et al (2010) Deletion of Fas in adipocytes relieves adipose tissue inflammation and hepatic manifestations of obesity in mice. J Clin Invest 120:191–202PubMedCrossRef Wueest S, Rapold RA, Schumann DM et al (2010) Deletion of Fas in adipocytes relieves adipose tissue inflammation and hepatic manifestations of obesity in mice. J Clin Invest 120:191–202PubMedCrossRef
41.
go back to reference Kumar A, Lawrence JC Jr, Jung DY et al (2010) Fat cell-specific ablation of rictor in mice impairs insulin-regulated fat cell and whole-body glucose and lipid metabolism. Diabetes 59:1397–1406PubMedCrossRef Kumar A, Lawrence JC Jr, Jung DY et al (2010) Fat cell-specific ablation of rictor in mice impairs insulin-regulated fat cell and whole-body glucose and lipid metabolism. Diabetes 59:1397–1406PubMedCrossRef
42.
go back to reference Hirosumi J, Tuncman G, Chang L et al (2002) A central role for JNK in obesity and insulin resistance. Nature 420:333–336PubMedCrossRef Hirosumi J, Tuncman G, Chang L et al (2002) A central role for JNK in obesity and insulin resistance. Nature 420:333–336PubMedCrossRef
43.
go back to reference Gao Z, Hwang D, Bataille F et al (2002) Serine phosphorylation of insulin receptor substrate 1 by inhibitor kappa B kinase complex. J Biol Chem 277:48115–48121PubMedCrossRef Gao Z, Hwang D, Bataille F et al (2002) Serine phosphorylation of insulin receptor substrate 1 by inhibitor kappa B kinase complex. J Biol Chem 277:48115–48121PubMedCrossRef
44.
go back to reference Croker BA, Krebs DL, Zhang JG et al (2003) SOCS3 negatively regulates IL-6 signaling in vivo. Nat Immunol 4:540–545PubMedCrossRef Croker BA, Krebs DL, Zhang JG et al (2003) SOCS3 negatively regulates IL-6 signaling in vivo. Nat Immunol 4:540–545PubMedCrossRef
45.
go back to reference Galic S, Oakhill JS, Steinberg GR (2010) Adipose tissue as an endocrine organ. Mol Cell Endocrinol 316:129–139PubMedCrossRef Galic S, Oakhill JS, Steinberg GR (2010) Adipose tissue as an endocrine organ. Mol Cell Endocrinol 316:129–139PubMedCrossRef
46.
go back to reference Bence KK, Delibegovic M, Xue B et al (2006) Neuronal PTP1B regulates body weight, adiposity and leptin action. Nat Med 12:917–924PubMedCrossRef Bence KK, Delibegovic M, Xue B et al (2006) Neuronal PTP1B regulates body weight, adiposity and leptin action. Nat Med 12:917–924PubMedCrossRef
47.
go back to reference Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS (2006) TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116:3015–3025PubMedCrossRef Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS (2006) TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116:3015–3025PubMedCrossRef
48.
go back to reference Arnold SF, Obourn JD, Jaffe H, Notides AC (1995) Phosphorylation of the human estrogen receptor on tyrosine 537 in vivo and by src family tyrosine kinases in vitro. Mol Endocrinol 9:24–33PubMedCrossRef Arnold SF, Obourn JD, Jaffe H, Notides AC (1995) Phosphorylation of the human estrogen receptor on tyrosine 537 in vivo and by src family tyrosine kinases in vitro. Mol Endocrinol 9:24–33PubMedCrossRef
49.
go back to reference Leong GM, Moverare S, Brce J et al (2004) Estrogen up-regulates hepatic expression of suppressors of cytokine signaling-2 and -3 in vivo and in vitro. Endocrinology 145:5525–5531PubMedCrossRef Leong GM, Moverare S, Brce J et al (2004) Estrogen up-regulates hepatic expression of suppressors of cytokine signaling-2 and -3 in vivo and in vitro. Endocrinology 145:5525–5531PubMedCrossRef
50.
go back to reference Ribas V, Nguyen MT, Henstridge DC et al (2010) Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERalpha-deficient mice. Am J Physiol Endocrinol Metab 298:E304–E319PubMedCrossRef Ribas V, Nguyen MT, Henstridge DC et al (2010) Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERalpha-deficient mice. Am J Physiol Endocrinol Metab 298:E304–E319PubMedCrossRef
Metadata
Title
Reduced Socs3 expression in adipose tissue protects female mice against obesity-induced insulin resistance
Authors
R. Palanivel
M. D. Fullerton
S. Galic
J. Honeyman
K. A. Hewitt
S. B. Jorgensen
G. R. Steinberg
Publication date
01-11-2012
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 11/2012
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-012-2665-3

Other articles of this Issue 11/2012

Diabetologia 11/2012 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.