Skip to main content
Top
Published in: Diabetologia 6/2013

Open Access 01-06-2013 | Article

Hypothalamic ATF3 is involved in regulating glucose and energy metabolism in mice

Authors: Y.-S. Lee, T. Sasaki, M. Kobayashi, O. Kikuchi, H.-J. Kim, H. Yokota-Hashimoto, M. Shimpuku, V.-Y. Susanti, Y. Ido-Kitamura, K. Kimura, H. Inoue, M. Tanaka-Okamoto, H. Ishizaki, J. Miyoshi, S. Ohya, Y. Tanaka, S. Kitajima, T. Kitamura

Published in: Diabetologia | Issue 6/2013

Login to get access

Abstract

Aims/hypothesis

The pancreas and hypothalamus are critical for maintaining nutrient and energy homeostasis, and combined disorders in these organs account for the onset of the metabolic syndrome. Activating transcription factor 3 (ATF3) is an adaptive response transcription factor. The physiological role of ATF3 in the pancreas has been controversial, and its role in the hypothalamus remains unknown. To elucidate the roles of ATF3 in these organs, we generated pancreas- and hypothalamus-specific Atf3 knockout (PHT-Atf3-KO) mice in this study.

Methods

We crossed mice bearing floxed Atf3 alleles with Pdx1-cre mice, in which cre is specifically expressed in the pancreas and hypothalamus, and analysed metabolic variables, pancreatic morphology, food intake, energy expenditure and sympathetic activity in adipose tissue. We also used a hypothalamic cell line to investigate the molecular mechanism by which ATF3 regulates transcription of the gene encoding agouti-related protein (Agrp).

Results

Although PHT-Atf3-KO mice displayed better glucose tolerance, neither plasma glucagon nor insulin level was altered in these mice. However, these mice exhibited higher insulin sensitivity, which was accompanied by a leaner phenotype due to decreased food intake and increased energy expenditure. We also observed decreased hypothalamic Agrp expression in PHT-Atf3-KO mice. Importantly, an increase in ATF3 levels is induced by fasting or low glucose in the hypothalamus. We also showed that ATF3 interacts with forkhead box-containing protein, O subfamily 1 (FoxO1) on the Agrp promoter and activates Agrp transcription.

Conclusions/interpretation

Our results suggest that ATF3 plays an important role in the control of glucose and energy metabolism by regulating Agrp.
Appendix
Available only for authorised users
Literature
1.
go back to reference Kahn R, Buse J, Ferrannini E, Stern M (2005) The metabolic syndrome: time for a critical appraisal: joint statement from the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 28:2289–2304PubMedCrossRef Kahn R, Buse J, Ferrannini E, Stern M (2005) The metabolic syndrome: time for a critical appraisal: joint statement from the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 28:2289–2304PubMedCrossRef
3.
go back to reference Shoelson SE, Lee J, Goldfine AB (2006) Inflammation and insulin resistance. J Clin Invest 116:1793–1801PubMedCrossRef Shoelson SE, Lee J, Goldfine AB (2006) Inflammation and insulin resistance. J Clin Invest 116:1793–1801PubMedCrossRef
4.
go back to reference Barsh GS, Schwartz MW (2002) Genetic approaches to studying energy balance: perception and integration. Nat Rev Genet 3:589–600PubMedCrossRef Barsh GS, Schwartz MW (2002) Genetic approaches to studying energy balance: perception and integration. Nat Rev Genet 3:589–600PubMedCrossRef
6.
go back to reference Kubota N, Terauchi Y, Tobe K et al (2004) Insulin receptor substrate 2 plays a crucial role in beta cells and the hypothalamus. J Clin Invest 114:917–927PubMed Kubota N, Terauchi Y, Tobe K et al (2004) Insulin receptor substrate 2 plays a crucial role in beta cells and the hypothalamus. J Clin Invest 114:917–927PubMed
7.
go back to reference Lin X, Taguchi A, Park S et al (2004) Dysregulation of insulin receptor substrate 2 in beta cells and brain causes obesity and diabetes. J Clin Invest 114:908–916PubMed Lin X, Taguchi A, Park S et al (2004) Dysregulation of insulin receptor substrate 2 in beta cells and brain causes obesity and diabetes. J Clin Invest 114:908–916PubMed
8.
go back to reference Accili D, Arden KC (2004) FoxOs at the crossroads of cellular metabolism, differentiation, and transformation. Cell 117:421–426PubMedCrossRef Accili D, Arden KC (2004) FoxOs at the crossroads of cellular metabolism, differentiation, and transformation. Cell 117:421–426PubMedCrossRef
9.
go back to reference Kitamura T, Feng Y, Kitamura YI et al (2006) Forkhead protein FoxO1 mediates Agrp-dependent effects of leptin on food intake. Nat Med 12:534–540PubMedCrossRef Kitamura T, Feng Y, Kitamura YI et al (2006) Forkhead protein FoxO1 mediates Agrp-dependent effects of leptin on food intake. Nat Med 12:534–540PubMedCrossRef
10.
go back to reference Kim HJ, Kobayashi M, Sasaki T et al (2012) Overexpression of FoxO1 in the hypothalamus and pancreas causes obesity and glucose intolerance. Endocrinology 153:659–671PubMedCrossRef Kim HJ, Kobayashi M, Sasaki T et al (2012) Overexpression of FoxO1 in the hypothalamus and pancreas causes obesity and glucose intolerance. Endocrinology 153:659–671PubMedCrossRef
11.
12.
go back to reference Hai T, Hartman MG (2001) The molecular biology and nomenclature of the activating transcription factor/cAMP responsive element binding family of transcription factors: activating transcription factor proteins and homeostasis. Gene 273:1–11PubMedCrossRef Hai T, Hartman MG (2001) The molecular biology and nomenclature of the activating transcription factor/cAMP responsive element binding family of transcription factors: activating transcription factor proteins and homeostasis. Gene 273:1–11PubMedCrossRef
13.
go back to reference Green TA, Alibhai IN, Unterberg S et al (2008) Induction of activating transcription factors (ATFs) ATF2, ATF3, and ATF4 in the nucleus accumbens and their regulation of emotional behavior. J Neurosci 28:2025–2032PubMedCrossRef Green TA, Alibhai IN, Unterberg S et al (2008) Induction of activating transcription factors (ATFs) ATF2, ATF3, and ATF4 in the nucleus accumbens and their regulation of emotional behavior. J Neurosci 28:2025–2032PubMedCrossRef
14.
go back to reference Hai T, Wolford CC, Chang YS (2010) ATF3, a hub of the cellular adaptive-response network, in the pathogenesis of diseases: is modulation of inflammation a unifying component? Gene Expr 15:1–11PubMedCrossRef Hai T, Wolford CC, Chang YS (2010) ATF3, a hub of the cellular adaptive-response network, in the pathogenesis of diseases: is modulation of inflammation a unifying component? Gene Expr 15:1–11PubMedCrossRef
15.
go back to reference Nawa T, Nawa MT, Adachi MT et al (2002) Expression of transcriptional repressor ATF3/LRF1 in human atherosclerosis: colocalization and possible involvement in cell death of vascular endothelial cells. Atherosclerosis 161:281–291PubMedCrossRef Nawa T, Nawa MT, Adachi MT et al (2002) Expression of transcriptional repressor ATF3/LRF1 in human atherosclerosis: colocalization and possible involvement in cell death of vascular endothelial cells. Atherosclerosis 161:281–291PubMedCrossRef
16.
go back to reference Cai Y, Zhang C, Nawa T et al (2000) Homocysteine-responsive ATF3 gene expression in human vascular endothelial cells: activation of c-Jun NH(2)-terminal kinase and promoter response element. Blood 96:2140–2148PubMed Cai Y, Zhang C, Nawa T et al (2000) Homocysteine-responsive ATF3 gene expression in human vascular endothelial cells: activation of c-Jun NH(2)-terminal kinase and promoter response element. Blood 96:2140–2148PubMed
17.
go back to reference Wang J, Webb G, Cao Y, Steiner DF (2003) Contrasting patterns of expression of transcription factors in pancreatic alpha and beta cells. Proc Natl Acad Sci U S A 100:12660–12665PubMedCrossRef Wang J, Webb G, Cao Y, Steiner DF (2003) Contrasting patterns of expression of transcription factors in pancreatic alpha and beta cells. Proc Natl Acad Sci U S A 100:12660–12665PubMedCrossRef
18.
go back to reference Hartman MG, Lu D, Kim ML et al (2004) Role for activating transcription factor 3 in stress-induced beta-cell apoptosis. Mol Cell Biol 24:5721–5732PubMedCrossRef Hartman MG, Lu D, Kim ML et al (2004) Role for activating transcription factor 3 in stress-induced beta-cell apoptosis. Mol Cell Biol 24:5721–5732PubMedCrossRef
19.
go back to reference Wang J, Cao Y, Steiner DF (2003) Regulation of proglucagon transcription by activated transcription factor (ATF) 3 and a novel isoform, ATF3b, through the cAMP-response element/ATF site of the proglucagon gene promoter. J Biol Chem 278:32899–32904PubMedCrossRef Wang J, Cao Y, Steiner DF (2003) Regulation of proglucagon transcription by activated transcription factor (ATF) 3 and a novel isoform, ATF3b, through the cAMP-response element/ATF site of the proglucagon gene promoter. J Biol Chem 278:32899–32904PubMedCrossRef
20.
go back to reference Zmuda EJ, Qi L, Zhu MX, Mirmira RG, Montminy MR, Hai T (2010) The roles of ATF3, an adaptive-response gene, in high-fat-diet-induced diabetes and pancreatic beta-cell dysfunction. Mol Endocrinol 24:1423–1433PubMedCrossRef Zmuda EJ, Qi L, Zhu MX, Mirmira RG, Montminy MR, Hai T (2010) The roles of ATF3, an adaptive-response gene, in high-fat-diet-induced diabetes and pancreatic beta-cell dysfunction. Mol Endocrinol 24:1423–1433PubMedCrossRef
21.
go back to reference Li D, Yin X, Zmuda EJ et al (2008) The repression of IRS2 gene by ATF3, a stress-inducible gene, contributes to pancreatic beta-cell apoptosis. Diabetes 57:635–644PubMedCrossRef Li D, Yin X, Zmuda EJ et al (2008) The repression of IRS2 gene by ATF3, a stress-inducible gene, contributes to pancreatic beta-cell apoptosis. Diabetes 57:635–644PubMedCrossRef
22.
go back to reference Zhang SJ, Buchthal B, Lau D et al (2011) A signaling cascade of nuclear calcium-CREB-ATF3 activated by synaptic NMDA receptors defines a gene repression module that protects against extrasynaptic NMDA receptor-induced neuronal cell death and ischemic brain damage. J Neurosci 31:4978–4990PubMedCrossRef Zhang SJ, Buchthal B, Lau D et al (2011) A signaling cascade of nuclear calcium-CREB-ATF3 activated by synaptic NMDA receptors defines a gene repression module that protects against extrasynaptic NMDA receptor-induced neuronal cell death and ischemic brain damage. J Neurosci 31:4978–4990PubMedCrossRef
23.
go back to reference Guerra-Crespo M, Perez-Monter C, Janga SC et al (2011) Transcriptional profiling of fetal hypothalamic TRH neurons. BMC Genom 12:222CrossRef Guerra-Crespo M, Perez-Monter C, Janga SC et al (2011) Transcriptional profiling of fetal hypothalamic TRH neurons. BMC Genom 12:222CrossRef
24.
go back to reference Kawauchi J, Zhang C, Nobori K et al (2002) Transcriptional repressor activating transcription factor 3 protects human umbilical vein endothelial cells from tumor necrosis factor-alpha-induced apoptosis through down-regulation of p53 transcription. J Biol Chem 277:39025–39034PubMedCrossRef Kawauchi J, Zhang C, Nobori K et al (2002) Transcriptional repressor activating transcription factor 3 protects human umbilical vein endothelial cells from tumor necrosis factor-alpha-induced apoptosis through down-regulation of p53 transcription. J Biol Chem 277:39025–39034PubMedCrossRef
25.
go back to reference Gu G, Dubauskaite J, Melton DA (2002) Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development 129:2447–2457PubMed Gu G, Dubauskaite J, Melton DA (2002) Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development 129:2447–2457PubMed
26.
go back to reference Taketani K, Kawauchi J, Tanaka-Okamoto M et al (2012) Key role of ATF3 in p53-dependent DR5 induction upon DNA damage of human colon cancer cells. Oncogene 31:2210–2221PubMedCrossRef Taketani K, Kawauchi J, Tanaka-Okamoto M et al (2012) Key role of ATF3 in p53-dependent DR5 induction upon DNA damage of human colon cancer cells. Oncogene 31:2210–2221PubMedCrossRef
27.
go back to reference Kitamura T, Kido Y, Nef S, Merenmies J, Parada LF, Accili D (2001) Preserved pancreatic beta-cell development and function in mice lacking the insulin receptor-related receptor. Mol Cell Biol 21:5624–5630PubMedCrossRef Kitamura T, Kido Y, Nef S, Merenmies J, Parada LF, Accili D (2001) Preserved pancreatic beta-cell development and function in mice lacking the insulin receptor-related receptor. Mol Cell Biol 21:5624–5630PubMedCrossRef
28.
go back to reference Wicksteed B, Brissova M, Yan W et al (2010) Conditional gene targeting in mouse pancreatic ss-Cells: analysis of ectopic Cre transgene expression in the brain. Diabetes 59:3090–3098PubMedCrossRef Wicksteed B, Brissova M, Yan W et al (2010) Conditional gene targeting in mouse pancreatic ss-Cells: analysis of ectopic Cre transgene expression in the brain. Diabetes 59:3090–3098PubMedCrossRef
29.
go back to reference Seale P, Conroe HM, Estall J et al (2011) Prdm16 determines the thermogenic program of subcutaneous white adipose tissue in mice. J Clin Invest 121:96–105PubMedCrossRef Seale P, Conroe HM, Estall J et al (2011) Prdm16 determines the thermogenic program of subcutaneous white adipose tissue in mice. J Clin Invest 121:96–105PubMedCrossRef
30.
go back to reference Ohno H, Shinoda K, Spiegelman BM, Kajimura S (2012) PPARgamma agonists induce a white-to-brown fat conversion through stabilization of PRDM16 protein. Cell Metab 15:395–404PubMedCrossRef Ohno H, Shinoda K, Spiegelman BM, Kajimura S (2012) PPARgamma agonists induce a white-to-brown fat conversion through stabilization of PRDM16 protein. Cell Metab 15:395–404PubMedCrossRef
31.
go back to reference Ebihara K, Ogawa Y, Katsuura G et al (1999) Involvement of agouti-related protein, an endogenous antagonist of hypothalamic melanocortin receptor, in leptin action. Diabetes 48:2028–2033PubMedCrossRef Ebihara K, Ogawa Y, Katsuura G et al (1999) Involvement of agouti-related protein, an endogenous antagonist of hypothalamic melanocortin receptor, in leptin action. Diabetes 48:2028–2033PubMedCrossRef
32.
go back to reference Fukuda M, Jones JE, Olson D et al (2008) Monitoring FoxO1 localization in chemically identified neurons. J Neurosci 28:13640–13648PubMedCrossRef Fukuda M, Jones JE, Olson D et al (2008) Monitoring FoxO1 localization in chemically identified neurons. J Neurosci 28:13640–13648PubMedCrossRef
33.
go back to reference Qian S, Chen H, Weingarth D et al (2002) Neither agouti-related protein nor neuropeptide Y is critically required for the regulation of energy homeostasis in mice. Mol Cell Biol 22:5027–5035PubMedCrossRef Qian S, Chen H, Weingarth D et al (2002) Neither agouti-related protein nor neuropeptide Y is critically required for the regulation of energy homeostasis in mice. Mol Cell Biol 22:5027–5035PubMedCrossRef
34.
go back to reference Wortley KE, Anderson KD, Yasenchak J et al (2005) Agouti-related protein-deficient mice display an age-related lean phenotype. Cell Metab 2:421–427PubMedCrossRef Wortley KE, Anderson KD, Yasenchak J et al (2005) Agouti-related protein-deficient mice display an age-related lean phenotype. Cell Metab 2:421–427PubMedCrossRef
35.
go back to reference Bewick GA, Gardiner JV, Dhillo WS et al (2005) Post-embryonic ablation of AgRP neurons in mice leads to a lean, hypophagic phenotype. FASEB J 19:1680–1682PubMed Bewick GA, Gardiner JV, Dhillo WS et al (2005) Post-embryonic ablation of AgRP neurons in mice leads to a lean, hypophagic phenotype. FASEB J 19:1680–1682PubMed
36.
go back to reference Chen BP, Liang G, Whelan J, Hai T (1994) ATF3 and ATF3 delta Zip. Transcriptional repression versus activation by alternatively spliced isoforms. J Biol Chem 269:15819–15826PubMed Chen BP, Liang G, Whelan J, Hai T (1994) ATF3 and ATF3 delta Zip. Transcriptional repression versus activation by alternatively spliced isoforms. J Biol Chem 269:15819–15826PubMed
Metadata
Title
Hypothalamic ATF3 is involved in regulating glucose and energy metabolism in mice
Authors
Y.-S. Lee
T. Sasaki
M. Kobayashi
O. Kikuchi
H.-J. Kim
H. Yokota-Hashimoto
M. Shimpuku
V.-Y. Susanti
Y. Ido-Kitamura
K. Kimura
H. Inoue
M. Tanaka-Okamoto
H. Ishizaki
J. Miyoshi
S. Ohya
Y. Tanaka
S. Kitajima
T. Kitamura
Publication date
01-06-2013
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 6/2013
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-013-2879-z

Other articles of this Issue 6/2013

Diabetologia 6/2013 Go to the issue
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.