HVEM-deficient mice fed a high-fat diet are protected from adipose tissue inflammation and glucose intolerance

Similar documents
Supplementary Figures

Effect of High-fat or High-glucose Diet on Obesity and Visceral Adipose Tissue in Mice

Supplementary Figure 1. DNA methylation of the adiponectin promoter R1, Pparg2, and Tnfa promoter in adipocytes is not affected by obesity.

General Laboratory methods Plasma analysis: Gene Expression Analysis: Immunoblot analysis: Immunohistochemistry:

Supplemental Table 1: Demographics and characteristics of study participants. Male, n (%) 3 (20%) 6 (50%) Age, years [mean ± SD] 33.3 ± ± 9.

Metabolic ER stress and inflammation in white adipose tissue (WAT) of mice with dietary obesity.

Yiying Zhang, PhD Research Scientist. Research Summary:

A crucial role for HVEM and BTLA in preventing intestinal inflammation

Supplementary Figure 1

Males- Western Diet WT KO Age (wks) Females- Western Diet WT KO Age (wks)

Supplementary Information

Inflammasome-mediated caspase-1 activity Gatekeeper of inflammation in the adipose tissue. Rinke Stienstra

New therapeutic targets for T2DM

Supplemental Information Supplementary Table 1. Tph1+/+ Tph1 / Analyte Supplementary Table 2. Tissue Vehicle LP value

Naohito AOKI, Erina ARAKAWA and Miyuki ITO. Department of Life Science, Graduate School of Bioresources, Mie University Tsu ABSTRACT

MICRORNA-223 REGULATES MACROPHAGE POLARIZATION AND DIET- INDUCED INSULIN RESISTANCE. A Thesis CONG MENG

Tissue factor-par2 signaling promotes diet-induced obesity and adipose

Primer sequences Target Sequence F Sequence R TNF-α (Tnfa) TCAGCCGATTTGCTATCTCAT A

Crosstalk between Adiponectin and IGF-IR in breast cancer. Prof. Young Jin Suh Department of Surgery The Catholic University of Korea

Supplementary Figure 1

Supplementary Information

Supporting Information

GPR120 *** * * Liver BAT iwat ewat mwat Ileum Colon. UCP1 mrna ***

IL-17 in health and disease. March 2014 PSO13-C051n

Possible roles of adiponectin in inflammatory process of rheumatoid arthritis

University of California, San Diego La Jolla CA 92093

Pathologic Stage. Lymph node Stage

Supplementary Figure 1 Chemokine and chemokine receptor expression during muscle regeneration (a) Analysis of CR3CR1 mrna expression by real time-pcr

Role of Th17 cells in the immunopathogenesis of dry eye disease

FAPESP Week /21/2017

The Immune System: The Mind Body Connection. Presented by Margaret Kemeny, Ph.D. Department of Psychiatry, University of California, San Francisco

Effect of BI-1 on insulin resistance through regulation of CYP2E1

Psoriasis and the metabolic syndrome

Supplemental Information. Increased 4E-BP1 Expression Protects. against Diet-Induced Obesity and Insulin. Resistance in Male Mice

Osteopontin mediates obesity-induced adipose tissue macrophage infiltration and insulin resistance in mice

We obtained Male C57BL/6J, ob/ob, and Cd8a-deficient mice from Charles River Japan

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

Supplementary Information:

Supplemental Table 1. Plasma NEFA and liver triglyceride levels in ap2-hif1ako and ap2-hif2ako mice under control and high fat diets.

Leptin deficiency suppresses progression of atherosclerosis in apoe-deficient mice

HIV AND INFLAMMATION: A NEW THREAT

Role of JAKs in myeloid cells and autoimmune diseases. Satoshi Kubo, Kunihiro Yamaoka and Yoshiya Tanaka

Supplementary Information

Epigenetic regulation of macrophage polarization and inflammation by DNA methylation in obesity

STAT4 Deficiency Reduces Obesity-Induced Insulin Resistance and Adipose Tissue Inflammation

Protein extraction and western blot analysis Protein extraction was performed as

SUPPLEMENTARY INFORMATION

ΦΛΕΓΜΟΝΗ ΚΑΙ ΔΙΑΒΗΤΗΣ

IMMUNOLOGICAL MEMORY. CD4 T Follicular Helper Cells. Memory CD8 T Cell Differentiation

HSN301 Diet and Disease Entire Note Summary

IMMUNOTOOLS: EFFECT OF NOTCH-DEFICIENT MACROPHAGES TO AUTOIMMUNE DISEASE WIPAWEE WONGCHANA

NK cells link obesity-induced adipose stress to inflammation and insulin resistance

Cytokines modulate the functional activities of individual cells and tissues both under normal and pathologic conditions Interleukins,

Cigarette Smoke Exposure and HIV-Related Neurologic Disease Progression Basic Mechanisms and Clinical Consequences

A microrna-34a/fgf21 Regulatory Axis and Browning of White Fat

SUPPLEMENTARY FIGURE 1

HHS Public Access Author manuscript Obesity (Silver Spring). Author manuscript; available in PMC 2014 November 01.

Defective Hepatic Autophagy in Obesity Promotes ER Stress and Causes Insulin Resistance

Supplementary Figure 1. Deletion of Smad3 prevents B16F10 melanoma invasion and metastasis in a mouse s.c. tumor model.

PANDER KO mice on high-fat diet are glucose intolerant yet resistant to fasting hyperglycemia and hyperinsulinemia

A Central Role of MG53 in Metabolic Syndrome. and Type-2 Diabetes

Supplementary Materials for

SUPPLEMENTARY INFORMATION. Supplemental Figure 1. Body weight and blood glucose parameters of chow-diet (CD)

Supplementary Figure 1. mtor LysM and Rictor LysM mice have normal cellularity and percentages of hematopoe>c cells. a. Cell numbers of lung, liver,

Molecular pathways linking metabolic inflammation and thermogenesis G. Solinas Summary

Supplementary Table 2. Plasma lipid profiles in wild type and mutant female mice submitted to a HFD for 12 weeks wt ERα -/- AF-1 0 AF-2 0

T Cell Activation, Costimulation and Regulation

B cells: a fundamental role in the pathogenesis of rheumatoid arthritis?

Nature Medicine: doi: /nm.3922

renoprotection therapy goals 208, 209

Supplementary Materials for

12/10/2009. Department of Pathology, Case Western Reserve University. Mucosal Cytokine Network in IBD

AdPLA ablation increases lipolysis and prevents obesity induced by high fat feeding or leptin deficiency

Attribution: University of Michigan Medical School, Department of Microbiology and Immunology

Interleukin-6; pathogenesis and treatment of autoimmune inflammatory diseases

Roger J. Davis. Metabolic Stress Signaling by the JNK Pathway

SUPPLEMENTARY INFORMATION

Microbiology 204: Cellular and Molecular Immunology

Metabolic Syndrome. DOPE amines COGS 163

pplementary Figur Supplementary Figure 1. a.

Inflammation & Type 2 Diabetes Prof. Marc Y. Donath

Analysis of AVP functions via V1a and V1b receptors with knockout mice. Akito Tanoue

Dietary regulation of nuclear receptors in obesity-related metabolic syndrome

Supplementary Figure 1: TSLP receptor skin expression in dcssc. A: Healthy control (HC) skin with TSLP receptor expression in brown (10x

ENHANCEMENT OF BROWN ADIPOSE TISSUE DEVELOPMENT IN VIVO BY A NOVEL INSULIN SENSITIZER

Combined Rho-kinase inhibition and immunogenic cell death triggers and propagates immunity against cancer

Oncolytic Immunotherapy: A Local and Systemic Antitumor Approach

Adipose Tissue as an Endocrine Organ. Abdel Moniem Ibrahim, MD Professor of Physiology Cairo University

Basic Immunology. Cytokines, cytokine receptors. Lecture 8th. Timea Berki MD, PhD

LIPOCALIN 2 DEFICIENCY INFLUENCES TRANSFORMING GROWTH FACTOR-β EFFECT ON INFLAMMATION AND EXTRACELLULAR MATRIX REMODELING IN INGUINAL ADIPOCYTES

Tomasz J Guzik. Diabetes, perivascular adipose tissue and inflammation.

control kda ATGL ATGLi HSL 82 GAPDH * ** *** WT/cTg WT/cTg ATGLi AKO/cTg AKO/cTg ATGLi WT/cTg WT/cTg ATGLi AKO/cTg AKO/cTg ATGLi iwat gwat ibat

Autoimmune Diseases. Betsy Kirchner CNP The Cleveland Clinic

Potential Role of Sphingosine 1-Phosphate in the. Pathogenesis of Rheumatoid Arthritis

Cytokines, adhesion molecules and apoptosis markers. A comprehensive product line for human and veterinary ELISAs

IL METABOLISMO EPATICO DEI CARBOIDRATI IN FISIOLOGIA E PATOLOGIA


Stewart et al. CD36 ligands promote sterile inflammation through assembly of a TLR 4 and 6 heterodimer

Programmed necrosis, not apoptosis, is a key mediator of cell loss and DAMP-mediated inflammation in dsrna-induced retinal degeneration

Comparison of Cryopreserved Amniotic Membrane and Umbilical Cord Tissues for use in Foot and Ankle Reconstructive Procedures

Transcription:

FEBS Letters 585 (2011) 2285 2290 journal homepage: www.febsletters.org HVEM-deficient mice fed a high-fat diet are protected from adipose tissue inflammation and glucose intolerance Ha-Jung Kim a, Hong-Min Kim b, Chu-Sook Kim a, Choon-Soo Jeong b, Hye-Sun Choi b, Teruo Kawada c, Byung-Sam Kim b, Rina Yu a, a Department of Food Science and Nutrition, University of Ulsan, Ulsan 680-749, South Korea b Department of Biological Science, University of Ulsan, Ulsan 680-749, South Korea c Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan article info abstract Article history: Received 21 March 2011 Revised 12 May 2011 Accepted 26 May 2011 Available online 12 June 2011 Edited by Laszlo Nagy Keywords: Obesity Inflammation Adipose tissue Insulin resistance HVEM is a member of the TNF receptor superfamily that plays a role in the development of various inflammatory diseases. In this study, we show that HVEM deficiency attenuates adipose tissue inflammatory responses and glucose intolerance in diet-induced obesity. Feeding a high-fat diet (HFD) to HVEM-deficient mice elicited a reduction in the number of macrophages and T cells infiltrated into adipose tissue. Proinflammatory cytokine levels in the adipose tissue decreased in HFDfed HVEM-deficient mice, while levels of the anti-inflammatory cytokine IL-10 increased. Moreover, glucose intolerance and insulin sensitivity were markedly improved in the HFD-fed HVEM-deficient mice. These findings indicate that HVEM may be a useful target for combating obesity-induced inflammatory responses and insulin resistance. Ó 2011 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved. 1. Introduction Inflammation, a hallmark of obesity, is closely associated with the development of obesity-related complications such as type II diabetes and atherosclerosis. Adipose tissue inflammatory components such as immune cells (e.g., macrophages and T cells) and cytokines/chemokines play a role in obesity-induced inflammatory processes [1,2], and modulation of the inflammatory components ameliorates not only the obesity-induced inflammatory responses but also metabolic complications [3 5]. However, the precise mechanisms and molecules underlying adipose immune cell crosstalk in obesity-related inflammation remain to be elucidated. HVEM/TNFRSF14 (herpes simplex virus glycoprotein D for herpes virus entry mediator) is a member of the TNF receptor superfamily (TNFRSF), and the membrane receptor for LIGHT/TNFSF14 (lymphotoxin-related inducible ligand that competes for glycoprotein D binding to herpes virus entry mediator on T cells) [6,7]. HVEM is widely and constitutively expressed in various human and rodent tissues, and strongly expressed on resting/activated immune cells (e.g., T cells, B cells, and monocytes) and non-immune cells (e.g., endothelial cells and adipocytes) [6,8,9]. The interaction Corresponding author. Address: Department of Food Science and Nutrition, University of Ulsan, Mugeo-dong, Nam-ku, Ulsan 680-749, South Korea. Fax: +82 52 259 1698. E-mail address: rinayu@ulsan.ac.kr (R. Yu). between HVEM and its ligand is involved in various inflammatory processes, and thus is implicated in the development of a number of inflammatory diseases (e.g., inflammatory bowel disease, rheumatoid arthritis, and nephritis) [8,10]. Blockade of HVEM/LIGHT activation attenuates the severity of autoimmune diseases such as type I diabetes mellitus and inflammatory bowel disease [11,12], and therefore HVEM and its ligand are considered to be therapeutic targets in those inflammatory diseases. Recently a link between LIGHT/HVEM and obesity has been reported by Bassols et al. [13,14]. Moreover our previous study has demonstrated that soluble LIGHT promotes T cells/macrophages infiltration/activation in vitro, and the LIGHT-induced inflammatory responses are blunted by neutralizing antibody or disruption of HVEM [9], indicating that LIGHT/HVEM interaction plays a role in adipose tissue inflammation through HVEM interaction with LIGHT. However, it remains unclear whether blockade of HVEM could protect obesity-induced inflammation and pathologies in vivo. In this study, we fed HVEM-deficient mice a high-fat diet and demonstrated that HVEM deficiency ameliorates not only adipose tissue inflammatory responses but also glucose intolerance. HVEM may be a novel target for treating obesity-induced inflammatory responses and complications. 2. Materials and methods See Supplementary material. 0014-5793/$36.00 Ó 2011 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved. doi:10.1016/j.febslet.2011.05.057

2286 H.-J. Kim et al. / FEBS Letters 585 (2011) 2285 2290 2.1. Statistical analyses Data are presented as means ± S.E. Statistical analysis was performed using ANOVA and Duncan s multiple-range test. Differences were considered to be significant at P < 0.05. 3. Results 3.1. Body weight change, food intake, and adiposity in HFD-fed HVEMdeficient mice To see whether HVEM deficiency alters obesity-related phenotypes, we fed HVEM-deficient mice a HFD, and measured diet intake, body weight gain and adiposity. RT-PCR analysis displayed that HVEM mrna was absent in adipose tissue, liver, and spleen of HVEM-deficient mice (Fig. 1A). We observed a significant reduction in body weight after 12 weeks in 45% HFD-fed HVEM-deficient mice and after 5 weeks in 60% HFD-fed HVEM-deficient mice compared with the HFD-fed wild type control mice (Fig. 1B), and no difference was observed in the lean mice fed a LFD (Fig. 1B). Gross inspection of abdominal dissections revealed smaller masses of adipose tissue in the HVEM-deficient mice on a high-fat diet than in the control obese mice (Fig. 1C). Adipose tissue weights in the HFD-fed HVEM-deficient mice were significantly lower than those in the HFD-fed wild type mice (Fig. 1D). Total diet intake of the HFD-fed HVEM-deficient mice was not significantly different from the HFD-fed wild type control (Fig. 1E). To see whether the reduced body weight gain and adiposity observed in HFD-fed HVEM deficient mice was due to increased energy expenditure, we measured UCP-1 expression in brown adipose tissue, which plays a crucial role in the control of energy expenditure. The levels of UCP-1 protein in brown adipose tissue were significantly higher in the HVEM deficient obese mice than in the WT control mice (Fig. 1F), indicating that HVEM deficiency increases thermogenesis, thus increasing energy expenditure. There was no phenotypic difference in fat mass and diet intake between LFD-fed HVEM-deficient lean mice and LFD-fed wild type lean mice (Fig. 1D and E). 3.2. Adipose macrophages and T cells in HFD-fed HVEM-deficient mice To determine whether HVEM deficiency alters adipose tissue inflammatory responses, we examined the increment of adipose immune cell infiltration in obesity. Histochemical examination showed that the adipocytes of the HFD-fed HVEM-deficient mice were smaller then those of the HFD-fed WT controls (6463 ± 347 lm 2 in the HVEM-deficient mice vs. 7937 ± 670 lm 2 in the WT mice), and the percentage of hypertrophic adipocytes (P12 000 lm 2 ) was significantly lower in the HFD-fed HVEM-deficient mice than the HFD-fed WT mice (Fig. 2A). Moreover, the numbers of cells infiltrated into adipose tissue were markedly reduced in the HFD-fed HVEM-deficient mice compared with the wild type obese controls (Fig. 2A). Flow cytometry revealed that total cell numbers in the SVF from adipose tissue also decreased markedly (Fig. 2B), as did the numbers of macrophages (CD11b+/ F4/80+) and T cells (CD4+ or CD8+) (Fig. 2C and D). Moreover, the HFD-fed HVEM-deficient mice contained fewer activated T cells (CD44+/CD62 ) (Fig. 2E), indicating that HVEM deficiency reduces adipose tissue leukocytes recruitment and their activation Fig. 1. Body weight change, food intake, and adiposity in HVEM-deficient obese mice. HVEM-deficient mice and wild type mice were fed 60% high-fat diet or 45% high-fat diet for 10 and 19 weeks, respectively. (A) HVEM mrna in tissues was detected by RT-PCR. (B) Total body weight gain. (C) Gross body shape. (D) Adipose tissue weights (Ep, epididymal; Re, renal; Me, mesenteric; Sc, subcutaneous) of wild type mice and HVEM-deficient mice fed a low-fat diet (LFD) or a high-fat diet (HFD). (E) Amount of total diet intake. (F) UCP-1 protein levels. UCP-1 protein in the brown adipose tissue of each mice was examined by Western blot analysis using the indicated antibodies. Data are means ± S.E. P < 0.05, # P < 0.005, ## P < 0.001 compared to wild type obese mice.

H.-J. Kim et al. / FEBS Letters 585 (2011) 2285 2290 2287 Fig. 2. Adipose macrophages and T cells in HFD-fed HVEM-deficient mice. (A) Histochemistry of adipose tissue and adipocyte size. Arrows show infiltrated cells. Sections obtained from the animals were stained with hematoxylin and eosin. Original magnification, 200 and 400. Data are means ± S.E. P < 0.05, # P < 0.005 compared to wild type obese mice. Immune cells in the adipose stromal vascular fraction were analyzed. Stromal vascular cells were isolated from the epididymal adipose tissue of HFD-fed HVEM-deficient mice and HFD-fed wild type mice. (B) Stromal vascular fraction cells (C) Macrophages (D) T cells (E) Activated T cells. The isolated cells were labeled with conjugated antibodies to CD11b, F4/80, CD4, CD8, CD44, CD62L and analyzed by flow cytometry. The values are numbers of cells per gram epididymal adipose tissue shown in fold decrease. Data are representative of three independent experiments. in the HFD-fed HVEM-deficient mice. There was no difference in the leukocyte infiltration between LFD-fed HVEM-deficient lean mice and LFD-fed wild type lean mice (data not shown). 3.3. Cytokine levels in epididymal adipose tissue of HFD-fed HVEMdeficient mice To examine whether HVEM deficiency affects inflammatory cytokine profiles in adipose tissue, we measured TNFa, MCP-1 and IL-6 mrna levels in epididymal adipose tissue. The levels of cytokine gene expression were significantly lower in the HFD-fed HVEM-deficient mice than in the HFD-fed wild type mice (Fig. 3A), and this was confirmed at the protein level (Fig. 3B). IL- 10 is an anti-inflammatory cytokine which is associated with insulin sensitivity [15]. It has been shown that low IL-10 levels dispose to the development of type 2 diabetes and metabolic syndrome [16]. We tested whether HVEM deficiency affects the IL-10 mrna and protein levels in epididymal adipose tissue. As shown in Fig. 3C and D, IL-10 was increased in the HFD-fed HVEM-deficient mice compared with HFD-fed wild type mice. No differences were observed in the cytokine levels between LFD-fed HVEM-deficient lean mice and LFD-fed wild type lean mice (Fig. 3A and B). 3.4. HVEM deficiency reduces obesity-induced glucose intolerance and insulin resistance To see whether the attenuation of adipose tissue inflammation in the HFD-fed HVEM-deficient mice is associated with glucose tolerance, we measured fasting glucose levels and performed oral glucose tolerance test. Fasting glucose and insulin levels in the 45% HFD-fed HVEM-deficient mice were slightly lower than in the HFD-fed wild type mice (Fig. 4A), and the oral glucose tolerance test demonstrated that the impairment of glucose tolerance was less severe in the HFD-fed HVEM-deficient mice than in the HFDfed wild type mice (Fig. 4B). The 45% HFD-fed HVEM-deficient mice had significantly lower levels of plasma glucose than the wild type

2288 H.-J. Kim et al. / FEBS Letters 585 (2011) 2285 2290 Fig. 3. Levels of cytokines in the adipose tissue of HFD-fed HVEM-deficient mice. Epididymal adipose tissue was isolated from HVEM-deficient mice and wild type mice fed a LFD or HFD, and total RNA was extracted. (A) Cytokine mrna levels. (B) Cytokine protein levels (TNFa, MCP-1, and IL-6). Anti-inflammatory IL-10 (C) mrna and (D) protein levels. The expression of inflammatory genes (TNFa, MCP-1, and IL-6) and anti-inflammatory (IL-10) was measured by real time-pcr. The levels of proteins in the adipose tissue homogenates were measured by ELISA and normalized with total protein content, which was determined using the BCA protein assay. Values are means ± S.E. of animals (n =4orn = 5) for each group. P < 0.05, # P < 0.005, ## P < 0.001 compared to the HFD-fed wild-type mice. obese controls at 90, 120 and 150 min (Fig. 4B). The improvement of glucose intolerance was also confirmed in 60% HFD-fed HVEMdeficient mice at 90 min (Fig. 4B). Additionally, Western blot analysis revealed that insulin responsiveness assessed by phosphorylations of IRS-1 and/or Akt was greater in HFD-fed HVEM-deficient mice than HFD-fed wild type control (Fig. 4C), and moreover expression levels of GLUT4 protein were also increased in the HFD-fed HVEM-deficient mice (Fig. 4D). These results indicate that HVEM-deficiency attenuates HFD-induced glucose intolerance and improved insulin sensitivity. 4. Discussion Obesity-induced inflammation mimics chronic inflammatory conditions accompanied by increased inflammatory cells such as activated macrophages/t cells and mediators such as cytokines/ chemokines. In this study, we demonstrate that HVEM deficiency reduces obesity-induced adipose tissue inflammatory responses and glucose intolerance. HVEM plays important roles in various inflammatory processes. Studies have shown that dysregulation of HVEM and its ligand system, which exaggerates T cell responses, can lead to chronic inflammation [17,18]. Although we and others have suggested an association between HVEM or LIGHT and obesity parameters in humans and mice [13,14], the pathogenic effects and underlying mechanism of HVEM have not been fully clarified. Generally, key events in obesity-induced inflammatory processes are characterized by the localized recruitment of activated macrophage/t cell subsets into adipose tissue and increased inflammatory cytokine/ chemokine release from these cells. We previously showed that soluble LIGHT directly induced macrophage and T cells migration in vitro [9], and the LIGHT-induced inflammatory actions were blunted by blockade of HVEM [9]. In view of the in vitro observation, we hypothesized that disruption of HVEM protects obesityinduced adipose tissue inflammatory responses in vivo. Using HVEM-deficient mice fed a high-fat diet, we for the first time found that HVEM deficiency markedly reduced the accumulation of macrophages and activated CD4+ and/or CD8+ T cells in the adipose tissue of HFD-fed mice. The findings not only verify our in vitro findings, but also suggest that HVEM plays a crucial role in obesity-induced infiltration of T cells/macrophages into adipose tissue. It has been shown that the interaction between HVEM and LIGHT provides a pro-inflammatory signal to T cells or macrophages to produce inflammatory cytokines [17 19]. Previously, our in vitro study has demonstrated that blocking of HVEM inhibits the activation of macrophages stimulated by soluble LIGHT to produce inflammatory cytokines [14]. Thus we thought that HVEM deficiency may reduce the release of inflammatory cytokines by adipose tissue-derived soluble LIGHT, which increases in obese condition. Indeed, we found that levels of inflammatory cytokine/ chemokine such as TNFa, IL-6, and MCP-1 are lower in the adipose tissue of HFD-fed HVEM-deficient mice. This notion is also supported by the evidence that HVEM-deficient mice have reduced numbers of activated CD4+ and CD8+ T cells in their adipose tissue. In addition, it should be noted that adipocytes constitutively express HVEM [13] and activation of HVEM by LIGHT enhances the

H.-J. Kim et al. / FEBS Letters 585 (2011) 2285 2290 2289 Fig. 4. HVEM deficiency ameliorates obesity-induced glucose intolerance in obese mice fed a high-fat diet. (A) Fasting glucose and insulin levels (B) Oral glucose tolerance test. Animals were fed a high fat diet 45% HFD (8 weeks, n = 12, male) for 14 weeks. Or, animals were fed 60% HFD (8 weeks, n = 4, male) for 7 weeks. Mice fed a high-fat diet were fasted 12 h before receiving oral administration of a 20% glucose solution at a dose of 2 g/kg, and blood samples were taken at the indicated times. Levels of glucose were measured with an enzymatic assay kit. (C and D) Insulin responses. After fasting 5 h, mice were stimulated with or without insulin for 4 min. Expression of p-akt, total Akt, p- IRS-1, total IRS-1 and GLUT4 proteins in the skeletal muscle of each mice was examined by Western blot analysis using the indicated antibodies. Values are means ± S.E. P < 0.05, # P < 0.005 compared to the HFD-fed wild type mice. release MCP-1 and IL-6 [14]. These findings together indicate that HVEM deficiency blunts the HVEM-mediated inflammatory crosstalk between adipose cells and T cells/macrophages in obesity, which may be favorable for protecting adipose tissue inflammation. Given the role of T cells/macrophages in vicious cycle of adipose tissue inflammation, the findings indicate that HVEM could be a valid target to disrupt a pathogenic loop in the recruitment of T cells and macrophages and their activation, which are crucial for adipose tissue inflammation in obesity condition. Obese adipose tissue T cells and macrophages participate in not only for promoting adipose tissue inflammation but also insulin resistance [4]. Obesity-induced chronic activation of pro-inflammatory signaling pathways in the adipose immune cells results in elevated levels of TNFa, IL-6, and MCP-1, which inhibit insulin signaling [3 5]. It has been shown that depletion of CD8+ T cells lower macrophage infiltration and adipose tissue inflammation and ameliorates systemic insulin resistance [20], while reduction of CD4+ Th1 cells suppresses insulin resistance [5]. We found that HFD-induced glucose intolerance and/or insulin sensitivity estimated by phosphorylations of IRS-1 and Akt were significantly attenuated in HFD-fed HVEM-deficient mice. This indicates that HVEM deficiency reduced adipose tissue inflammation through decreased inflammatory CD4+ and/or CD8+ T cells and cytokines, which in turn ameliorates the HFD-induced pathological processes. Additionally, given that IL-10 elicits insulin-sensitizing activity and protects IL-6 and TNFa-induced cellular insulin resistance [21,22], the attenuation of inflammation observed in the HFD-fed HVEMdeficient mice may at least in part be attributed to the increased IL-10 in the adipose tissue. HVEM not only transmits inflammatory signals which activate T cells through the interaction with LIGHT, but also induces antiinflammatory signals by engaging coinhibitory receptor BTLA (B and T lymphocyte attenuator) and/or CD160 which inhibits T cell proliferation [23]. We previously observed that levels of HVEM and LIGHT mrna were significantly increased in obese mice compared with lean control [9], whereas BTLA expression was markedly reduced [14]. With respect to this, it is seemingly that LIGHT-mediated inflammatory signals may be in favor of obesity

2290 H.-J. Kim et al. / FEBS Letters 585 (2011) 2285 2290 condition, and this may be disrupted in HFD-fed HVEM-deficient mice, leading to the improvement of obesity-related inflammation and complications. In conclusion, we have for the first time demonstrated that HVEM deficiency reduces adipose tissue inflammatory responses as assessed by the decrease in infiltration of T cells/macrophages and inflammatory cytokine levels, and thus counteracts the glucose intolerance in mice fed a high-fat diet. HVEM may be a novel target for reducing obesity-induced inflammatory responses and complications. Conflict of interest None. Acknowledgements This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (KRF-2007-313-F00148), and supported by the SRC program (Center for Food and Nutritional Genomics: grant number 2010-0001886) of the NRF of Korea, funded by the Ministry of Education, Science and Technology. T.K. was supported by the Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sport, Science and Technology of Japan (22228001). BSK was supported by the Korea Research Foundation grant funded by the Korean Government (MOEHRD) (BRL-2010-0087350). Appendix A. Supplementary data Supplementary data associated with this article can be found, in the online version, at doi:10.1016/j.febslet.2011.05.057. References [1] Weisberg, S.P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R.L. and Ferrante Jr., A.W. (2003) Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest. 112, 1796 1808. [2] Xu, H. et al. (2003) Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Invest. 112, 1821 1830. [3] Hotamisligil, G.S., Shargill, N.S. and Spiegelman, B.M. (1993) Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 259, 87 91. [4] Shoelson, S.E., Lee, J. and Goldfine, A.B. (2006) Inflammation and insulin resistance. J. Clin. Invest. 116, 1793 1801. [5] Winer, S. et al. (2009) Normalization of obesity-associated insulin resistance through immunotherapy. Nat. Med. 15, 921 929. [6] Kwon, B.S. et al. (1997) A newly identified member of the tumor necrosis factor receptor superfamily with a wide tissue distribution and involvement in lymphocyte activation. J. Biol. Chem. 272, 14272 14276. [7] Schneider, K., Potter, K.G. and Ware, C.F. (2004) Lymphotoxin and LIGHT signaling pathways and target genes. Immunol. Rev. 202, 49 66. [8] Otterdal, K. et al. (2006) Platelet-derived LIGHT induces inflammatory responses in endothelial cells and monocytes. Blood 108, 928 935. [9] Kim, H.M., Jeong, C.S., Choi, H.S., Kawada, T. and Yu, R. (2011) LIGHT/TNFSF14 enhances adipose tissue inflammatory responses through its interaction with HVEM. FEBS Lett. 585, 579 584. [10] Kim, W.J., Kang, Y.J., Koh, E.M., Ahn, K.S., Cha, H.S. and Lee, W.H. (2005) LIGHT is involved in the pathogenesis of rheumatoid arthritis by inducing the expression of pro-inflammatory cytokines and MMP-9 in macrophages. Immunology 114, 272 279. [11] Ware, C.F. (2009) Targeting the LIGHT HVEM pathway. Adv. Exp. Med. Biol. 647, 146 155. [12] Wang, J., Anders, R.A., Wang, Y., Turner, J.R., Abraham, C., Pfeffer, K. and Fu, Y.X. (2005) The critical role of LIGHT in promoting intestinal inflammation and Crohn s disease. J. Immunol. 174, 8173 8182. [13] Bassols, J., Moreno, J.M., Ortega, F., Ricart, W. and Fernandez-Real, J.M. (2010) Characterization of herpes virus entry mediator as a factor linked to obesity. Obesity (Silver Spring) 18, 239 246. [14] Bassols, J., Moreno-Navarrete, J.M., Ortega, F., Ricart, W. and Fernandez-Real, J.M. (2010) LIGHT is associated with hypertriglyceridemia in obese subjects and increased cytokine secretion from cultured human adipocytes. Int. J. Obes. (Lond.) 34, 146 156. [15] de Luca, C. and Olefsky, J.M. (2008) Inflammation and insulin resistance. FEBS Lett. 582, 97 105. [16] van Exel, E., Gussekloo, J., de Craen, A.J., Frolich, M., Bootsma-Van Der Wiel, A. and Westendorp, R.G. (2002) Low production capacity of interleukin-10 associates with the metabolic syndrome and type 2 diabetes: the Leiden 85- Plus Study. Diabetes 51, 1088 1092. [17] Croft, M. (2003) Co-stimulatory members of the TNFR family: keys to effective T-cell immunity? Nat. Rev. Immunol. 3, 609 620. [18] Kwon, B., Kim, B.S., Cho, H.R., Park, J.E. and Kwon, B.S. (2003) Involvement of tumor necrosis factor receptor superfamily (TNFRSF) members in the pathogenesis of inflammatory diseases. Exp. Mol. Med. 35, 8 16. [19] Granger, S.W. and Rickert, S. (2003) LIGHT HVEM signaling and the regulation of T cell-mediated immunity. Cytokine Growth Factor Rev. 14, 289 296. [20] Nishimura, S. et al. (2009) CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat. Med. 15, 914 920. [21] Cintra, D.E. et al. (2008) Interleukin-10 is a protective factor against dietinduced insulin resistance in liver. J. Hepatol. 48, 628 637. [22] Hong, E.G. et al. (2009) Interleukin-10 prevents diet-induced insulin resistance by attenuating macrophage and cytokine response in skeletal muscle. Diabetes 58, 2525 2535. [23] Shui, J.W., Steinberg, M.W. and Kronenberg, M. (2011) Regulation of inflammation, autoimmunity, and infection immunity by HVEM BTLA signaling. J. Leukoc. Biol. 89, 517 523.