Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with...

12
Yuan Zhang, 1,2 Rui Li, 1,2 Yan Meng, 1 Shiwu Li, 3 William Donelan, 3 Yan Zhao, 2 Lei Qi, 2 Mingxiang Zhang, 2 Xingli Wang, 2 Taixing Cui, 2,4 Li-Jun Yang, 3 and Dongqi Tang 1,2,4 Irisin Stimulates Browning of White Adipocytes Through Mitogen-Activated Protein Kinase p38 MAP Kinase and ERK MAP Kinase Signaling The number and activity of brown adipocytes are linked to the ability of mammals to resist body fat accumulation. In some conditions, certain white adipose tissue (WAT) depots are readily convertible to a ‘‘brown-like’’ state, which is associated with weight loss. Irisin, a newly identied hormone, is secreted by skeletal muscles into circulation and promotes WAT browningwith unknown mechanisms. In the current study, we demonstrated in mice that recombinant irisin decreased the body weight and improved glucose homeostasis. We further showed that irisin upregulated uncoupling protein-1 (UCP-1; a regulator of thermogenic capability of brown fat) expression. This effect was possibly mediated by irisin-induced phosphorylation of the p38 mitogen-activated protein kinase (p38 MAPK) and extracellular signalrelated kinase (ERK) signaling pathways. Inhibition of the p38 MAPK by SB203580 and ERK by U0126 abolished the upregulatory effect of irisin on UCP-1. In addition, irisin also promoted the expression of betatrophin, another newly identied hormone that promotes pancreatic b-cell proliferation and improves glucose tolerance. In summary, our data suggest that irisin can potentially prevent obesity and associated type 2 diabetes by stimulating expression of WAT browning-specic genes via the p38 MAPK and ERK pathways. Diabetes 2014;63:514525 | DOI: 10.2337/db13-1106 Obesity is the most common metabolic disorder in the world. Obesity develops when energy intake exceeds energy expenditure (1) and is principally characterized by excess adipose tissue (2,3). Obese subjects appear to have a high risk of developing type 2 diabetes (2). Increasing energy expenditure has emerged as a potential and at- tractive strategy to prevent obesity (4). Although regu- lators of the energy expenditure are not entirely clear, adipocytes appear to play a central role in modulating energy balance and nutrient ux in vertebrates. Two different types of adipose tissue including white adipose tissue (WAT) and brown adipose tissue (BAT) have been widely studied. White adipocytes store energy (e.g., triglycerides), whereas brown adipocytes consume energy (5,6). Many studies have shown that the changes 1 Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, Peoples Republic of China 2 Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Peoples Republic of China 3 Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 4 Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC Corresponding authors: Dongqi Tang, [email protected], Taixing Cui, taixing [email protected], and Li-Jun Yang, [email protected].edu. Received 16 July 2013 and accepted 9 October 2013. This article contains Supplementary Data online at http://diabetes .diabetesjournals.org/lookup/suppl/doi:10.2337/db13-1106/-/DC1. © 2014 by the American Diabetes Association. See http://creativecommons .org/licenses/by-nc-nd/3.0/ for details. See accompanying commentary, p. 381. 514 Diabetes Volume 63, February 2014 SIGNAL TRANSDUCTION

Transcript of Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with...

Page 1: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

Yuan Zhang,1,2 Rui Li,1,2 Yan Meng,1 Shiwu Li,3 William Donelan,3 Yan Zhao,2 Lei Qi,2 Mingxiang Zhang,2 Xingli Wang,2

Taixing Cui,2,4 Li-Jun Yang,3 and Dongqi Tang1,2,4

Irisin Stimulates Browning ofWhite Adipocytes ThroughMitogen-Activated ProteinKinase p38 MAP Kinase andERK MAP Kinase Signaling

The number and activity of brown adipocytes arelinked to the ability of mammals to resist body fataccumulation. In some conditions, certain whiteadipose tissue (WAT) depots are readily convertibleto a ‘‘brown-like’’ state, which is associated withweight loss. Irisin, a newly identified hormone, issecreted by skeletal muscles into circulation andpromotes WAT “browning” with unknownmechanisms. In the current study, we demonstratedin mice that recombinant irisin decreased the bodyweight and improved glucose homeostasis. Wefurther showed that irisin upregulated uncouplingprotein-1 (UCP-1; a regulator of thermogeniccapability of brown fat) expression. This effect waspossibly mediated by irisin-induced phosphorylationof the p38 mitogen-activated protein kinase (p38MAPK) and extracellular signal–related kinase (ERK)signaling pathways. Inhibition of the p38 MAPK bySB203580 and ERK by U0126 abolished theupregulatory effect of irisin on UCP-1. In addition,irisin also promoted the expression of betatrophin,another newly identified hormone that promotespancreatic b-cell proliferation and improves glucose

tolerance. In summary, our data suggest that irisincan potentially prevent obesity and associated type2 diabetes by stimulating expression of WATbrowning-specific genes via the p38 MAPK and ERKpathways.Diabetes 2014;63:514–525 | DOI: 10.2337/db13-1106

Obesity is the most common metabolic disorder in theworld. Obesity develops when energy intake exceedsenergy expenditure (1) and is principally characterized byexcess adipose tissue (2,3). Obese subjects appear to havea high risk of developing type 2 diabetes (2). Increasingenergy expenditure has emerged as a potential and at-tractive strategy to prevent obesity (4). Although regu-lators of the energy expenditure are not entirely clear,adipocytes appear to play a central role in modulatingenergy balance and nutrient flux in vertebrates.

Two different types of adipose tissue including whiteadipose tissue (WAT) and brown adipose tissue (BAT)have been widely studied. White adipocytes store energy(e.g., triglycerides), whereas brown adipocytes consumeenergy (5,6). Many studies have shown that the changes

1Center for Stem Cell and Regenerative Medicine, The Second Hospital ofShandong University, Jinan, People’s Republic of China2Shandong University Qilu Hospital Research Center for Cell Therapy, KeyLaboratory of Cardiovascular Remodeling and Function Research, Qilu Hospitalof Shandong University, Jinan, People’s Republic of China3Department of Pathology, Immunology, and Laboratory Medicine, University ofFlorida College of Medicine, Gainesville, FL4Department of Cell Biology and Anatomy, University of South Carolina School ofMedicine, Columbia, SC

Corresponding authors: Dongqi Tang, [email protected], Taixing Cui, [email protected], and Li-Jun Yang, [email protected].

Received 16 July 2013 and accepted 9 October 2013.

This article contains Supplementary Data online at http://diabetes.diabetesjournals.org/lookup/suppl/doi:10.2337/db13-1106/-/DC1.

© 2014 by the American Diabetes Association. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.

See accompanying commentary, p. 381.

514 Diabetes Volume 63, February 2014

SIG

NALTRANSDUCTIO

N

Page 2: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

in BAT activity can profoundly affect adaptive thermo-genesis and glucose homeostasis (7,8). The thermogeniccapability of brown fat is mainly mediated by the pres-ence of a mitochondria uncoupling protein-1 (UCP-1),which uncouples the electron transport chain from en-ergy production and results in the release of potentialenergy obtained from food as heat. The expression ofUCP-1 is regulated by several transcriptional factors,including peroxisome proliferator–activated receptorg (PPARg) coactivator-1a (PGC-1a), which can be in-duced by cold exposure and/or b-adrenergic signaling (9).In several rodent models, browning of WAT depotsappears to be protective against diet-induced metabolicdisorders, including obesity and diabetes (10,11).

Irisin, secreted by skeletal muscles and increased withexercise, is a small polypeptide hormone containing 111amino acids with an estimated molecular mass of 22 kDa.It has been shown that PGC-1a and exercise upregulatethe expression of fibronectin type III domain containing5 (Fndc5), a type I transmembrane protein of skeletalmuscle. Fndc5 is then proteolysed at amino acid position30 and 140 to give rise to irisin (12). Overexpression ofirisin by adenoviral vector increases total body energyexpenditure, induces modest weight loss, and improvesglucose intolerance in high fat–fed mice (12). Age andskeletal muscle mass are the primary predictors of thecirculating level, with young male athletes having irisinlevels several fold higher than those in middle-aged obesewomen (13). The circulating irisin is significantly lower inindividuals with type 2 diabetes compared with non-diabetic control subjects (14,15). Moreover, the plasmairisin level is negatively associated with the blood ureanitrogen level in chronic kidney disease patients (16) andpositively associated with aerobic performance in heartfailure patients (17). Taking into account the relationshipbetween irisin and energy expenditure, irisin can now beregarded as another secreted protein stimulating brownadipocyte thermogenesis, besides liver fibroblast growthfactor 21 (18) and cardiac natriuretic peptides (19).However, little is known about the molecular mechanismand signaling pathways mediating the induction of thefunctional “brown-like” adipocyte phenotype by irisin.

In the current study, we established an efficient sys-tem for the expression and purification of humanrecombinant irisin (r-irisin) in Pichia pastoris and de-livered the r-irisin in mice through intraperitoneal in-jection. We also treated primary adipocytes and 3T3-L1–derived adipocytes with r-irisin to detect its direct effectson the fat cell browning process in order to understandthe underlying “browning” mechanisms. Our studydemonstrated that administration of r-irisin in vivo de-creased body weight, promoted brown fat–specific geneexpression in subcutaneous white adipose, and improvedglucose tolerance, which is consistent with a previousstudy (12). In addition, from cell culture experiments,we showed that irisin stimulated brown fat–specific geneexpression via the activation of the p38 mitogen-activated

protein kinase (p38 MAPK) and extracellular signal–related kinase (ERK) pathways.

RESEARCH DESIGN AND METHODS

Expression and Purification of Human Irisin FromPichia

Human irisin cDNA (360 bp) was designed and synthe-sized by Life Technologies. The synthesized cDNA wascloned into EcoR1/Xba1 sites of the pPICZaA plasmid.Pichia pastoris X-33 was transformed with a linearizedpPICZaA-irisin plasmid according to the kit manual(Pichia Easycomp Transformation Kit; Invitrogen). Theculture of yeast and induction of protein expression wereperformed as previously described (20).

The r-irisin protein in the supernatant was purified bya two-step method (20). First, r-irisin was precipitatedwith 60% saturated ammonium sulfate. The precipitatedproteins were collected and dissolved in buffer A (25mmol/L HEPES, pH 7.9, 10% glycerol, 0.1 mol/L KCl, 0.2mmol/L EDTA, and 0.5 mmol/L dithiothreitol) and di-alyzed against the same buffer. The resulting sample wasloaded onto a concanavalin A–agarose column. The col-umn was first washed with 0.2 mol/L KCl in buffer A, andproteins were eluted with 1.5 mol/L KCl in buffer A. Theelution fractions were analyzed by Western blotting. Thepurified irisin was dialyzed against 10% glycerol in PBSand stored at 280°C.

Site-Directed Mutagenesis of Irisin cDNA

There are two N-glycosylation sites in human irisinprotein (Asn7 and Asn52). To confirm the glycosylation,we constructed two mutant irisin cDNA clones, N7H andN52H, by using a site-directed mutagenesis kit (Stra-tagene). In brief, the pPICZaA-irisin plasmid was used asa template for site-directed mutagenesis PCR with thepoint-mutation primers. After Dpn1 digestion, the PCRfragments were transformed into Escherichia coli Top10.The cDNA of the clones was purified with a Qiagenplasmid midi-purification kit. Each mutant cDNA wassequenced completely to ensure that no other basechanges were produced. The mutation primers were asfollows: N7H forward, 59-tccatccgctccagtgcacgttactgttagacacttgaaggc, and reverse, 59-tgtctaacagtaacgtgcctggagcggatggagagaattc; N52H forward, 59-gattcatccaagaggtgcacactactactagatcctgtgctttg, and reverse, 59-gatctagtagtagtgtgcacctcttggatgaatctcaacattc.

Cell Culture

Primary Adipocyte CultureMale Sprague-Dawley rats (200 g) were used in the study,and the adipocytes were isolated using a method as de-scribed previously (21). In brief, subcutaneous fat padswere minced with scissors and digested in type I colla-genase (1 mg/mL; Sigma-Aldrich) at 37°C in a water bathshaker. After 90 min, the contents were immediatelyfiltered in mesh and centrifuged at 1,000 rpm for 10 min.The layer of floating cells was then washed three times

diabetes.diabetesjournals.org Zhang and Associates 515

Page 3: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

with PBS. Adipocytes were incubated in Dulbecco’smodified Eagle’s medium (DMEM)/F12 medium supple-mented with 10% FBS and 100 units/mL penicillin-streptomycin (Sigma-Aldrich) in an atmosphere of 5%CO2 at 37°C.

Differentiation of 3T3-L1 Preadipocytes Into MatureAdipocytesMurine preadipocyte (3T3-L1) cells were purchased fromthe Chinese Academy of Sciences Cell Bank (Shanghai,China) and induced to differentiate into mature adipo-cytes as described previously (22). In brief, preadipocyteswere cultured using basic medium (DMEM supplementedwith 10% bovine calf serum and 100 units/mL penicillin-streptomycin) at 37°C in a 5% CO2 incubator. Afterconfluence (day 1), the cells were initiated for the adi-pocyte differentiation by incubating with induction me-dium (basic medium supplemented with 0.5 mmol/Lisobutylmethylxanthine, 0.25 mmol/L dexamethasone,and 10 mmol/L insulin). Two days after induction (day3), the medium was changed to basic medium supple-mented with insulin only for an additional 2 days. Onday 5, the medium was switched back to basic mediumfor another 2 days. R-irisin was added to the 3T3-L1 cellsat day 3 in adipocyte differentiation for a total of 4 daysstimulation. On day 7, 3T3-L1 mature adipocytes with orwithout r-irisin were processed for Oil Red O staining forconfirming adipocyte differentiation, RNA extraction forreverse transcription-quantitative PCR (RT-qPCR), im-munocytochemistry (ICC) with anti–UCP-1 antibodies,and Western blotting.

Binding of Irisin on the Cell Membrane of Adipocytes

On day 7 of differentiation, 3T3-L1–derived adipocyteswere incubated with or without irisin for 30 min. Afterwashing, the cells were incubated with anti-irisin rabbitpolyclonal antibody (1:1,000; Phoenix Pharmaceuticals,

Inc.) for 1 h. Then FITC–anti-rabbit IgG antibody wasadded and incubated for another hour. After washing,the cells were examined using a confocal fluorescencemicroscope. Additionally, a luciferase-irisin fusion proteinwas constructed into pREN2-irisin expression vector andproduced as described previously (23). 3T3-L1–derivedadipocytes were incubated in culture with eitherluciferase-irisin or luciferase. ICC was performed usingantiluciferase antibody (Santa Cruz Biotechnology) asdescribed above.

RNA Isolation and Real-Time PCR

Total RNA was extracted from cells with or without irisintreatment by using Trizol reagent (Invitrogen). Reversetranscription of 2 mg total RNA was performed with theHigh-Capacity cDNA Reverse Transcription Kit (AppliedBiosystems). Real-time PCR in triplicate was performedwith SYBR Green Master Mix (Applied Biosystems), andgene-specific primers are listed in Table 1. The 22Δct

method was used to calculate the relative expression ofgenes using b-actin RNA as an internal control.

ICC

3T3-L1–derived adipocytes were fixed with ice-cold meth-anol for 10 min at 4°C. The cells were then washed threetimes with PBS and blocked in hydrogen peroxide blockingsolution for 10 min at room temperature. The cells wereincubated with primary antibody of UCP-1 (1:500; Sigma-Aldrich, Oakville, ONT, Canada) overnight at 4°C. Afterwashing, the cells were incubated with horseradish perox-idase (HRP)-conjugated goat anti-rabbit IgG antibody for30 min at room temperature. After washing with PBS, thecells were incubated in avidin-biotinylated HRP for another30 min. Diaminobenzidine was used for ICC and hema-toxylin was used for counterstaining nuclei. Positive andnegative expression of protein UCP-1 was represented bydark brown in cytoplasm, respectively.

Table 1—Primer sequences for RT-qPCR

Gene Forward primer Reverse primer

UCP-1 AGGCTTCCAGTACCATTAGGT CTGAGTGAGGCAAAGCTGATTT

PGC-1a CCCTGCCATTGTTAAGACC TGCTGCTGTTCCTGTTTTC

Cox7a CTCTGGTCCGGTCTTTTAGC GTACTGGGAGGTCATTGTCGG

Elovl3 TTCTCACGCGGGTTAAAAATGG GAGCAACAGCTAGACGACCAC

PPARa AGAGCCCCATCTGTCCTCTC ACTGGTAGTCTGCAAAACCAAA

TMEM26 TTCCTGTTGCATTCCCTGGTC GCCGGAGAAAGCCATTTGT

Ebf3 TCACCCTCCCTTCAAACTGTA GTTTCACTGCGGACATGACAT

Cidea TGCTCTTCTGTATCGCCCAGT GCCGTGTTAAGGAATCTGCTG

Prdm16 CCCCACATTCCGCTGTGAT CTCGCAATCCTTGCACTCA

aP2 GGAACACGTCGTGGGATAATG GGCAGACTTTGGATGCTTCTT

Adipoq GCACTGGCAAGTTCTACTGCAA GTAGGTGAAGAGAACGGCCTTGT

Betatrophin CAGGATGCCACACAGGAGCTTC TCAGCTGCACTTGTAGTCTC

Adipoq, adiponectin; Prdm16, PR domain containing 16.

516 Mechanism of Irisin Function Diabetes Volume 63, February 2014

Page 4: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

Western Blotting

An equal amount of proteins from cell lysates was loadedin each well of a 12% SDS-PAGE. After electrophoresis,proteins were transferred to polyvinylidene fluoridemembranes, blocked for 1 h with 5% fat-free milk atroom temperature, and blotted with the indicated primaryantibodies overnight at 4°C at 1:1,000 dilution (rabbitanti-irisin, anti–UCP-1 [Sigma-Aldrich], anti-ERK1/2,anti–phospho-ERK1/2, anti-AKT, anti–phospho-AKT,anti-JNK, anti–phospho-JNK, anti–p38 MAPK, and anti–phospho-p38 MAPK antibodies [Cell Signaling Technology,Inc.]); rabbit anti-GAPDH antibody was used at 1:10,000(Sigma-Aldrich). After washing with Tris-buffered salinewith Tween, the membranes were incubated with HRP-conjugated secondary antibodies for 1 h at room temper-ature. Then, immune complexes were detected using theECL method, and immunoreactive bands were quantified bydensitometric analysis using Alpha Imager 2200.

Animal Studies

C57BL/6 mice fed with high-fat diet (HFD) (60% fat) for10 weeks with an initial body weight in the range of 35–43 g were purchased from the animal center of ShandongUniversity. Obese mice were randomly divided into twogroups: control (n = 8) and r-irisin treated (n = 8). Themice were treated daily with purified r-irisin at a dose of0.5 mg/g body weight/day by intraperitoneal injection for14 days, and the mice in the control group were givendaily saline at the same volume. At the end of r-irisintreatment (day 14), mouse body weights were recordedand some mice were subjected to intraperitoneal glucosetolerance test before being killed. Mouse subcutaneousfat tissues were harvested for immunohistochemistryand total RNA extraction for subsequent RT-qPCR.

Statistical Analysis

Results are presented as mean 6 SE of at least threeindependent experiments, and each experiment wasconducted in triplicate. Statistical significance among

multiple groups was analyzed by one-way ANOVA fol-lowed by Student t test for comparison of the resultsbetween two groups using the SPSS software package(SPSS 17.0). P , 0.05 was considered to be statisticallysignificant.

RESULTS

Expression and Characterization of Purified HumanRecombinant Irisin

In order to achieve high-yield expression of human irisingene in a yeast expression system, we designed andconstructed an optimized irisin cDNA coding sequencefor yeast codon usage. The synthesized gene and proteinsequence of r-irisin are shown in Supplementary Fig. 1.The resulting plasmid pPICZaA-irisin was transformedinto P. pastoris. Eight clones were selected from 50 singlecolonies that grew on yeast extract peptone dextrose(YPD) containing zeocin (100 mg/mL) for testing thecapacity of r-irisin protein expression. To examine theprotein expression, the secreted r-irisin at various timepoints was separated by SDS-PAGE and stained withCoomassie brilliant blue. There were three protein bandspresent on the gel with a molecular weight of 25, 22, and15 kDa (Fig. 1A). By Western blotting analysis, all threebands are human irisin protein (Fig. 1B). The proteinidentity of all three bands was also confirmed by MALDI-TOF mass spectrometry (Supplementary Fig. 2).

Human irisin protein has two putative N-glycosylationsites at asparagine 7 (Asn7) and asparagine 52 (Asn52).The two higher molecular weights (25 and 22 kDa) of thesecreted irisin protein compared with the native form ofirisin were likely glycosylated. Treatment of the purifiedr-irisin with PNGase F to remove N-linked glycans andsubsequent Western blotting analysis revealed (Fig. 1C)that the enzyme-treated r-irisin exhibited a single bandat low molecular weight (15 kDa), confirming that mostof the secreted r-irisin expressed by the P. pastoris wasglycosylated.

Figure 1—Expression of r-irisin and glycosylation of r-irisin. A: Culture supernatants (40 mL) from the S1 clone fermentation were collecteddaily up to day 6. The human r-irisin protein in the supernatants was visualized by Coomassie blue staining and confirmed by Westernblotting with anti-irisin antibodies (B). C: PNGase-F treatment of r-irisin removed glycosylation and confirmed by Western blot. D: SDS-PAGE analysis of wild-type (WT) and mutant forms of irisin (N7H and N52H) expression and deglycosylation by PNGase-F treatment.

diabetes.diabetesjournals.org Zhang and Associates 517

Page 5: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

Figure 2—Effects of r-irisin and mutants on the expression of brown adipocyte–specific genes in 3T3-L1–derived adipocytes. 3T3-L1–derivedadipocytes were treated with r-irisin (20 nmol/L) as described in RESEARCH DESIGN AND METHODS. A: Expression of brown/beige adipocyte marker,betatrophin, and white adipocyte marker (aP2 and adipoq) genes were measured by RT-qPCR. *P< 0.05; **P< 0.01 vs. untreated. B: Westernblot and densitometric analysis for UCP-1 protein in cells with indicated treatments. **P< 0.01.C: Oil Red O staining for lipid droplets confirmedadipocyte differentiation (top) and quantification of UCP-1–positive cells in ICC using Imagepro Plus software (bottom). Data were expressed asmean 6 SE of three independent experiments. **P < 0.01 vs. adipocytes without irisin. D: Biological function of mutant irisin. 3T3-L1–derived

518 Mechanism of Irisin Function Diabetes Volume 63, February 2014

Page 6: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

To confirm the glycosylated sites, we mutated each ofthe two glycosylation sites at Asn7 or Asn52, in-dividually. The mutant r-irisin protein from each mutant(N7H or N52H) showed only two bands with molecularweight 22 and 15 kDa. The 22-kDa protein likely repre-sents the mutant irisin that was glycosylated on one ofthe two glycosylated sites (N7H or N52H) since this bandwas still sensitive to PNGase-F treatment (Fig. 1D). Ourexperiments confirm that human irisin indeed has twoglycosylated sites at Asn7 and Asn52.

R-irisin Stimulates the Browning of Primary Adipocytesand 3T3-L1–Derived Adipocytes

To investigate the browning effect of r-irisin on adipo-cytes, we used both 3T3-L1–derived adipocytes and pri-mary rat adipocytes as cellular models treated with orwithout human r-irisin as described in RESEARCH DESIGN

AND METHODS. Gene expression studies showed thattreating 3T3-L1–derived adipocytes with r-irisin(20 nmol/L) led to a rapid upregulation of brown cellmarkers: UCP-1, PGC-1a, transmembrane protein 26(TMEM26), early B cell factor 3 (Ebf3), elongation of very

long chain fatty acids-like 3 (Elovl3), cell death-inducingDNA fragmentation factor (Cidea), and cytochromec oxidase, subunit VII a (Cox7a) (all P value ,0.01)(Fig. 2A). UCP-1 is a special mitochondrial protein thatdissipates chemical energy to heat. PGC-1a is a powerfultranscriptional coactivator that is a master regulator ofmitochondrial biogenesis, including UCP-1 and Fndc5 inbrown fat cells. The increased expression of beige cellmarker TMEM26 (24) and brown cell markers such asCidea and Cox7a indicated that the r-irisin may exerteffects on converting white adipose cells into beige-likeadipocytes and brown adipose cells. Interestingly, treat-ment of adipocytes with r-irisin also significantly increasedthe expression of the betatrophin (ANGPTL8) gene (3.5360.97–fold, P , 0.01). Betatrophin, a newly identifiedpancreatic b-cell growth hormone, is primarily expressedin liver and fat in mice (25). This result suggests thatincreased betatrophin expression by irisin may, at least inpart, explain improved insulin resistance by irisin.

The increased UCP-1 expression at the protein level byr-irisin in 3T3-L1–derived adipocytes was also demon-strated both by Western blotting (Fig. 2B) and ICC study

adipocytes were treated as indicated, and expression levels of UCP-1 and PGC-1a genes were measured by qPCR. *P < 0.05; **P < 0.01vs. control. E: UCP-1 protein expression was detected by Western blotting and measured by densitometry. N7H refers to mutated irisinafter mutating asparagine 7 to histidine. N52H refers to mutated irisin after mutating asparagine 52 to histidine. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Figure 3—Browning effect of r-irisin on rat primary adipocytes. Primary rat adipocytes were treated with r-irisin at indicated concen-trations for 6 h. A: The expression levels of UCP-1 and PGC-1a genes were measured by RT-qPCR. B: UCP-1 protein expression wasmeasured by Western blotting and quantified by densitometry. GAPDH, glyceraldehyde-3-phosphate dehydrogenase. *P < 0.05;**P < 0.01 vs. untreated.

diabetes.diabetesjournals.org Zhang and Associates 519

Page 7: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

Figure 4—Irisin stimulates adipocyte browning via p38/ERK pathways. Primary rat adipocytes and 3T3-L1–derived adipocytes weretreated with PBS (control [con]) or r-irisin (20 nmol/L) at indicated time points. Phosphorylated and total p38 and ERK1/2 in cell lysateswere analyzed by Western blotting with anti–P-p38 or anti–P-ERK antibodies. Expression levels of P-p38 and total p38 protein in primaryadipocytes (A) and 3T3-L1 adipocytes (B) were measured by corresponding densitometric quantification. Expression levels of P-ERK andtotal ERK in primary (C) and 3T3-L1–derived (D) adipocytes were measured with corresponding densitometric quantification. Densito-metric analysis of the related bands was expressed as relative optical density of the bands, corrected using respective total protein asa loading control and normalized against the untreated control. Data were expressed as mean 6 SE of three independent experiments.*P < 0.05; **P < 0.01 vs. control. E: 3T3-L1–derived adipocytes were pretreated for 30 min with p38 inhibitor SB023580 or ERK inhibitorU0126 at indicated concentrations followed by r-irisin treatment. Western blotting detected phosphorylated and total p38 and ERKproteins.

520 Mechanism of Irisin Function Diabetes Volume 63, February 2014

Page 8: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

(Fig. 2C, bottom) with confirmed adipocyte differentia-tion (Fig. 2C, Oil Red O staining, top). Quantification ofUCP-1 ICC studies in r-irisin–treated 3T3-L1–derived adi-pocytes revealed a significant increase in UCP-1–positiveadipocytes over controls (2.786 0.63–fold, P, 0.01) withmultilocular lipid droplets compared with those treatedwith saline (Fig. 2C). Moreover, treatment of primarycultured rat adipocytes with r-irisin over a range of con-centrations (2, 20, and 100 nmol/L) (Fig. 3) was able tosignificantly increase the expression of the UCP-1 and PGC-1a genes. Considering that UCP-1 mRNA was increasedfourfold compared with those that were untreated using20 nmol/L r-irisin, we chose 20 nmol/L r-irisin for thefollowing experiments.

Effect of Mutated Irisin on Browning

To determine whether glycosylation in irisin has a sig-nificant effect on biological function, we next examinedthe expression of browning markers in 3T3-L1–derivedadipocytes treated with Asn7- or Asn52-mutated irisin

protein. As shown in Fig. 2D, the effects ofupregulation of UCP-1 and PGC-1a by irisin wassignificantly reduced by the mutated irisin at either site.This loss of function was also confirmed at the proteinlevel, with significant UCP-1 reduction compared withtreatment with the wild-type irisin (Fig. 2E), suggestingthat the posttranslational glycosylation of the secretedirisin from yeast enhances the biological function of thebrowning effect evidenced by increased expression ofUCP-1 and PGC-1a. However, we cannot exclude thepossibility that the decreased biological activity observedin the N7H and N52H mutant irisin was due to mecha-nisms other than the altered glycosylation status.

Identifying Signaling Pathways of Irisin-MediatedBrowning

Although the browning function mediated by irisin hasbeen extensively studied, the molecular signaling path-ways that specifically mediate browning are unclear. Herewe showed that p38 MAPK and ERK play a key role in

Figure 5—Inhibiting p38 MAPK and ERK prevents the expression of UCP-1. Primary rat (A and C) and 3T3-L1 (B and D) adipocytes werepretreated with SB (10 mmol/L) or U0 (10 mmol/L) and both for 30 min, and then r-irisin (20 nmol/L) was supplemented for an additional 4days. UCP-1 expression at mRNA level by qPCR analysis (A and B) and at protein levels by Western blotting (C and D) was examined. ForC and D, the bar graph represents the ratio of UCP-1/glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and is normalized against thecontrol group. Data are expressed as mean 6 SE of three independent experiments. *P < 0.05; **P < 0.01 vs. control group; ++P < 0.01;+P < 0.05 vs. irisin group.

diabetes.diabetesjournals.org Zhang and Associates 521

Page 9: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

mediating the irisin browning function. As shown inFig. 4, the phosphorylated p38 MAPK (P-p38) andphosphorylated ERK (P-ERK) were significantly increasedat as early as 5 min, peaked between 10 and 20 min, anddecreased at 30 min after the r-irisin treatment in bothprimary rat (Fig. 4A and C) and 3T3-L1 adipocytes (Fig.4B and D). The increased levels of the P-p38 and P-ERKproteins after the r-irisin treatment were statisticallysignificant, as quantified by densitometry.

To further examine the possible involvements of p38MAPK and ERK pathways in irisin-induced browningeffects, 3T3-L1 adipocytes were treated with or withoutSB203580 (p-38MAPK inhibitor) or U0126 (ERK inhibitor)individually in the presence of irisin. P38 and ERK phos-phorylation induced by irisin was gradually suppressed withincreasing concentrations of the p38 and ERK inhibitors(Fig. 4E), whereas there was no reduction in the amount oftotal p38 or ERK protein. The irisin-induced UCP-1 upre-gulation was significantly reduced by either SB203580 orU0126. Moreover, the combination of both inhibitors al-most completely abolished the effect of irisin-mediatedincreasing expression of UCP-1 mRNA (Fig. 5A and B) andprotein (Fig. 5C and D) levels. These findings indicate thatthe irisin-induced browning effect marked by enhancedexpression of the browning gene UCP-1 was mediated viaactivation of p38 MAPK and ERK signaling pathways.

A previous study showed that irisin significantlyimproves glucose tolerance and insulin resistance in themice fed with HFD (12); the mechanism of this effect,

however, was unknown. AKT is an important signalingmolecule in the insulin signaling pathway. We thereforeinvestigated the effect of irisin on the activation of AKTusing an antibody to phosphorylated serine 473 of AKT.Unlike insulin (100 nmol/L), which can significantlypromote the phosphorylation of AKT (SupplementaryFig. 3B), irisin stimulation of 3T3-L1 adipocytes showedno significant effects on the phosphorylation of AKT inall testing time points (Supplementary Fig. 3A). In ad-dition, there was no additive or synergistic effect be-tween insulin and irisin on the level of phosphorylationof AKT (Supplementary Fig. 3B), indicating that the AKTsystem was not involved in the browning action of irisin.Furthermore, we showed that treatment of adipocyteswith irisin had no effect on the phosphorylation of c-JunNH2-terminal kinases (JNKs), indicating that the JNKsignaling pathway is not involved in the irisin-mediatedbrowning process (Supplementary Fig. 3C).

Detection of Irisin Binding to Cell Membrane ofAdipocytes

So far, there is no direct evidence showing a specific mem-brane receptor for irisin in adipocytes. Based on the resultsof the activation of P-p38 and P-ERK signaling transductionby irisin, we infer that the adipocyte membrane mighthave receptors for irisin. Ligand binding to its membranereceptor has historically been determined using a radioactiveisotopically labeled ligand. However, the potential hazardsof radioisotope handling and the environmental impact of

Figure 6—Binding of irisin to the plasma membrane of 3T3-L1 adipocytes. Confocal microscopy of adipocytes treated with irisin, anti-irisin antibody, and FITC–anti-rabbit IgG antibody (A). Fluorescence microscopy of adipocytes treated with luciferase as control or lu-ciferase-irisin fusion protein (B). The red arrows indicate the cell membrane of the adipocytes bound with irisin fusion protein.

522 Mechanism of Irisin Function Diabetes Volume 63, February 2014

Page 10: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

radioisotope disposal make this approach less desirable.Therefore, we used live cell binding methods to observe thebinding property of irisin to the cell membrane. The 3T3-L1 adipocytes were briefly incubated with r-irisin. Ther-irisin bound on the cell surface membrane was visualizedby immunofluorescence after sequential incubation withanti-irisin antibody and FITC–anti-rabbit IgG antibody. Asshown in Fig. 6A, the green fluorescence can only be seenon the membrane of irisin-treated cells. To further confirmthe irisin binding to the cell membrane, we addedluciferase-irisin fusion protein or luciferase alone (as con-trol) into the cell culture medium. Irisin binding to the cellmembrane was visualized by antiluciferase antibody. Asshown in Fig. 6B, the green fluorescence was observedclearly with increasing intensity along the membrane onluciferase-irisin–incubated cells, whereas no clear membranesignal was observed on luciferase-treated cells. These datasuggest that 3T3-L1–derived adipocytes may have yet-to-be-identified irisin receptor on the cell membrane.

R-irisin Affects the Body Weight and GlucoseHomeostasis In Vivo

To examine potential therapeutic effects of r-irisin, wetreated both normal and fat mice with daily injections ofr-irisin for 14 days. Figure 7A shows that the mRNA

levels of brown adipocyte marker genes, including UCP-1,PGC-1a, Cox7a, Elovl3, PPARa, and Cidea, were signifi-cantly elevated in the adipose tissues of the normal micereceiving r-irisin (**P , 0.01). Similar gene expressionresults were also observed in adipose tissue isolated fromfat mice fed with HFD (Supplementary Fig. 4). Consis-tent with 3T3-L1–derived adipocytes, the expression ofbetatrophin (ANGPTL8) was also significantly increased(*P , 0.05) in the adipose tissue of the mice treated withr-irisin compared with the control mice (Fig. 7B). Thechanges in browning gene expression in the sub-cutaneous adipose tissues were accompanied by an in-crease in the number of UCP-1–positive, multilocularadipocytes (Fig. 7C). The body weights of the r-irisin–treated fat mice were reduced after 14 days comparedwith the saline-treated control fat mice (Fig. 7D).R-irisin treatment of fat mice significantly improvedglucose tolerance, as demonstrated by a quicker re-covery curve in intraperitoneal glucose tolerance test(Fig. 7E) and significantly reduced levels of fasting in-sulin (Fig. 7F) when compared with the control fatmice. These results suggest that in vivo treatment of fatmice with r-irisin can decrease body weight, promotesthe expression of brown-specific genes, and improvesHFD-induced insulin resistance.

Figure 7—In vivo effects of r-irisin on body weight and brown-specific gene expression. The mice were intraperitoneally given eitherr-irisin (0.5 mg/g/day) or saline daily for 14 days and were terminated at day 14 posttreatment. Subcutaneous fat pads were collected andlevels of transcripts were analyzed by qPCR for expression of the indicated brown cell–specific genes (A) and betatrophin (B). Repre-sentative images from immunohistochemistry against UCP-1 in normal and r-irisin–treated fat mice (C ). C57BL/6 mice fed HFD for 10weeks were intraperitoneally injected with saline or r-irisin, and all analyses were performed at 14 days thereafter (n = 8 for both groups).D: Body weights of mice 14 days after injection with saline or r-irisin (using paired Student t test: control group, P = 0.3196, and irisin-treated group, P = 0.006). E: Intraperitoneal glucose tolerance test. F: Fasting plasma insulin levels measured using radioimmunoassay.Data are expressed as mean 6 SE (**P < 0.01; *P < 0.05 vs. control).

diabetes.diabetesjournals.org Zhang and Associates 523

Page 11: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

DISCUSSION

PGC-1a–dependent irisin, a novel myokine, is derivedfrom cleaving Fndc5 protein. Irisin promotes brown fat–like development and thermogenesis in WAT both invitro and in vivo. The discovery of irisin has created anopportunity to further understand the role of adipocytesin obesity, diabetes, and other associated metabolic dis-orders (12,13,26,27). However, the molecular mecha-nisms and cellular signaling pathways responsible for thebrowning effect of irisin have not been elucidated.

In this study, we successfully constructed the yeastexpression plasmid containing a synthesized optimalcodon usage, human irisin-coding sequence and gener-ated pure human recombinant irisin protein in P. pastoriswith high yield that is fully biologically functional. TheP. pastoris system is widely used for heterogenic proteinexpression, with the capacity to generate post-translational modified proteins (28). The humanrecombinant irisin protein expressed in yeast showeda predominant band of ;22 kDa, which is similar to theglycosylated irisin previously reported (12). UsingPNGase-F digestion and site-directed mutagenesis anal-ysis, we for the first time confirmed two glycosylationsites in human irisin located at the position of asparagine7 and asparagine 52. Given the glycosylation, a commonfeature of secreted protein, of irisin in mice and humans(12), we site directedly mutated the two glycosylatedsites to detect the biological effect of the mutant irisin.As shown in Fig. 2D and E, posttranslational glycosyla-tion of the secreted irisin enhances the expression ofUCP-1 and PGC-1a, compared with mutated irisin, in-dicating that yeast-produced irisin is superior to thatproduced in a bacterial expression system.

It is now clear that there are two distinct types ofbrown adipose cells. One is the classical brown fat thatarises from a myf5, muscle-like cell lineage (29,30). An-other is the brown adipocytes that are found in-terspersed within the WAT, in response to chemical orhormonal stimulation, environmental changes, cold ex-posure, and defined genetic manipulation (31–33). Thesebrown fat cells, UCP-1 positive, are designated as beige orbrown-in-white (brite) cells (11,34). In the current study,we demonstrated that r-irisin indeed increased the ex-pression of thermogenic genes, including UCP-1, PGC-1a,Cox7a, Ebf3, and Elovl3, in both primary and cell line–derived adipocytes. We also found that the expression ofTMEM26, a marker of beige cells, was increased whenr-irisin was added to the cultured 3T3-L1 preadipocytesduring the differentiation. Wu et al. (24) also showedthat beige cell precursors have preferential sensitivity tothe browning effect of irisin. Our results showed thatirisin significantly elevated the expression of TMEM26,which suggests that irisin may influence the number ofbeige-like cells.

Irisin functions as a muscle-derived energy expendi-ture signal that directly communicates with adipose

tissue and induces browning. It is therefore important toidentify the molecular pathways mediating the browningfunction by irisin. Boström et al. (12) identified PPARa asa potential factor for the expression of UCP-1, but otherpathways may also be involved. PGC-1a, a master regu-lator of mitochondrial biogenesis and function, acts asa cold-inducible protein that controls adaptive thermo-genesis in BAT, and its expression is regulated by the ac-tivation of cAMP/PKA/p38 MAPK signaling pathways(35,36). Moreover, other molecules, such as AMPK andPPARd, have been suggested as exercise mimetics (37,38).Our data show that irisin significantly increased the levelsof P-p38 MAPK and P-ERK in primary adipocytes and3T3-L1–derived adipocytes, and this effect was abolishedby inhibiting p38 and ERK phosphorylation (Fig. 5) toprevent irisin-induced UCP-1 expression. We suggest thatthe p38 and ERK signaling pathways play a central role inthe irisin-induced emergence of brown adipocytes.

Our study further suggests a possible existence of anirisin-specific receptor on the membrane of adipocytes.Although more experiments are needed to characterizeand validate the existence and specificity of this receptor,our study at least provides the first evidence for itsexistence.

In conclusion, the recombinant irisin increases theexpression of thermogenic genes in vitro and in vivo. Itstimulates expression of brown cell–specific genes inadipocytes via p38 and ERK signaling pathways. Thiseffect is likely mediated through a specific membranereceptor on adipocytes. Our findings have provided ini-tial experimental evidence in supporting possible thera-peutic usage of recombinant irisin for the treatmentof obesity and diabetes as well as associated metabolicdisorders.

Acknowledgments. The authors thank Dr. Jinghua Yang, ShandongUniversity, for LC-MS/MS analysis. The authors are also indebted toDr. Xiangdong Wang, Shandong University, for much helpful discussion andinformation regarding 3T3-L1 cell differentiation.

Funding. This work was supported by National Qianren Scholar ProgramSpecial Funding of Shandong University, Taishan Scholar Special Funding ofShandong Province, and in part by National Institute of Health CareerDevelopment Award (K01) to D.T.

Duality of Interest. No potential conflicts of interest relevant to thisarticle were reported.

Author Contributions. Y. Zhang designed and performed theexperiments and wrote the manuscript. R.L. and Y.M. participated in thedesign of the research with a special focus on recombinant gene expressionin yeast. S.L. participated in the design and execution of the experiments,and W.D. participated in writing the manuscript. Y. Zhao and L.Q.contributed to animal experiments. M.Z. developed the method forpurification of irisin. X.W., T.C., and L.-J.Y. participated in the discussionduring the project design and revised and edited the manuscript. D.T.participated in the design and overall supervision of the experiments. D.T. isthe guarantor of this work and, as such, had full access to all the data in thestudy and takes responsibility for the integrity of the data and the accuracyof the data analysis.

524 Mechanism of Irisin Function Diabetes Volume 63, February 2014

Page 12: Irisin Stimulates Browning of White Adipocytes Through ... · munocytochemistry (ICC) with anti–UCP-1 antibodies, and Western blotting. Binding of Irisin on the Cell Membrane of

References1. Speakman J.R. FTO effect on energy demand versus food intake. Nature

2010;464:E1; discussion E2

2. Gesta S, Tseng YH, Kahn CR. Developmental origin of fat: tracking obesityto its source. Cell 2007;131:242–256

3. Park KW, Halperin DS, Tontonoz P. Before they were fat: adipocyte pro-genitors. Cell Metab 2008;8:454–457

4. Frühbeck G, Becerril S, Sáinz N, Garrastachu P, García-Velloso MJ. BAT:a new target for human obesity? Trends Pharmacol Sci 2009;30:387–396

5. Cannon B, Nedergaard J. Brown adipose tissue: function and physiologicalsignificance. Physiol Rev 2004;84:277–359

6. Cinti S. Adipocyte differentiation and transdifferentiation: plasticity of theadipose organ. J Endocrinol Invest 2002;25:823–835

7. Cederberg A, Grønning LM, Ahrén B, Taskén K, Carlsson P, Enerbäck S.FOXC2 is a winged helix gene that counteracts obesity, hypertriglyceri-demia, and diet-induced insulin resistance. Cell 2001;106:563–573

8. Feldmann HM, Golozoubova V, Cannon B, Nedergaard J. UCP1 ablationinduces obesity and abolishes diet-induced thermogenesis in mice exemptfrom thermal stress by living at thermoneutrality. Cell Metab 2009;9:203–209

9. Barbera MJ, Schluter A, Pedraza N, Iglesias R, Villarroya F, Giralt M.Peroxisome proliferator-activated receptor alpha activates transcription ofthe brown fat uncoupling protein-1 gene. A link between regulation of thethermogenic and lipid oxidation pathways in the brown fat cell. J BiolChem 2001;276:1486–1493

10. Cao L, Choi EY, Liu X, et al. White to brown fat phenotypic switch inducedby genetic and environmental activation of a hypothalamic-adipocyte axis.Cell Metab 2011;14:324–338

11. Seale P, Conroe HM, Estall J, et al. Prdm16 determines the thermogenicprogram of subcutaneous white adipose tissue in mice. J Clin Invest 2011;121:96–105

12. Boström P, Wu J, Jedrychowski MP, et al. A PGC1-a-dependent myokinethat drives brown-fat-like development of white fat and thermogenesis.Nature 2012;481:463–468

13. Huh JY, Panagiotou G, Mougios V, et al. FNDC5 and irisin in humans: I.Predictors of circulating concentrations in serum and plasma and II. mRNAexpression and circulating concentrations in response to weight loss andexercise. Metabolism 2012;61:1725–1738

14. Liu JJ, Wong MD, Toy WC, et al. Lower circulating irisin is associated withtype 2 diabetes mellitus. J Diabetes Complications 2013;27:365–369

15. Choi YK, Kim MK, Bae KH, et al. Serum irisin levels in new-onset type 2diabetes. Diabetes Res Clin Pract 2013;100:96–101

16. Wen MS, Wang CY, Lin SL, Hung KC. Decrease in irisin in patients withchronic kidney disease. PLoS ONE 2013;8:e64025

17. Lecker SH, Zavin A, Cao P, et al. Expression of the irisin precursor FNDC5in skeletal muscle correlates with aerobic exercise performance in patientswith heart failure. Circ Heart Fail 2012;5:812–818

18. Hondares E, Rosell M, Gonzalez FJ, Giralt M, Iglesias R, Villarroya F.Hepatic FGF21 expression is induced at birth via PPARalpha in response tomilk intake and contributes to thermogenic activation of neonatal brownfat. Cell Metab 2010;11:206–212

19. Bordicchia M, Liu D, Amri EZ, et al. Cardiac natriuretic peptides act via p38MAPK to induce the brown fat thermogenic program in mouse and humanadipocytes. J Clin Invest 2012;122:1022–1036

20. Li SW, Sun Y, Donelan W, et al. Expression, purification, and character-ization of recombinant human pancreatic duodenal homeobox-1 protein inPichia pastoris. Protein Expr Purif 2010;72:157–161

21. Fernyhough ME, Vierck JL, Hausman GJ, Mir PS, Okine EK, Dodson MV.Primary adipocyte culture: adipocyte purification methods may lead toa new understanding of adipose tissue growth and development. Cyto-technology 2004;46:163–172

22. Miegueu P, Cianflone K, Richard D, St-Pierre DH. Motilin stimulates pre-adipocyte proliferation and differentiation and adipocyte lipid storage. Am JPhysiol Endocrinol Metab 2011;301:E758–E766

23. Zeng FY, McLean AJ, Milligan G, Lerner M, Chalmers DT, Behan DP.Ligand specific up-regulation of a Renilla reniformis luciferase-tagged,structurally unstable muscarinic M3 chimeric G protein-coupled receptor.Mol Pharmacol 2003;64:1474–1484

24. Wu J, Boström P, Sparks LM, et al. Beige adipocytes are a distinct type ofthermogenic fat cell in mouse and human. Cell 2012;150:366–376

25. Yi P, Park JS, Melton DA. Betatrophin: a hormone that controls pancreaticb cell proliferation. Cell 2013;153:747–758

26. Spiegelman BM, Korsmeyer SJ. Irisin and the therapeutic benefits ofexercise. BMC Proceedings 2012;6(Suppl. 3):O23

27. Swick A.G., Orena S., O’Connor A. Irisin levels correlate with energyexpenditure in a subgroup of humans with energy expenditure greater thanpredicted by fat free mass. Metabolism 2013;62:1070–1073

28. Li Z, Moy A, Sohal K, et al. Expression and characterization of recombinanthuman secretory leukocyte protease inhibitor (SLPI) protein from Pichiapastoris. Protein Expr Purif 2009;67:175–181

29. Seale P, Bjork B, Yang W, et al. PRDM16 controls a brown fat/skeletalmuscle switch. Nature 2008;454:961–967

30. Lepper C, Fan CM. Inducible lineage tracing of Pax7-descendant cellsreveals embryonic origin of adult satellite cells. Genesis 2010;48:424–436

31. Langin, D. Recruitment of brown fat and conversion of white into brownadipocytes: strategies to fight the metabolic complications of obesity?Biochim Biophys Acta 2010;1801:372–376

32. Lefterova MI, Lazar MA. New developments in adipogenesis. Trends En-docrinol Metab 2009;20:107–114

33. Cinti S. Between brown and white: novel aspects of adipocyte differenti-ation. Ann Med 2011;43:104–115

34. Petrovic N, Walden TB, Shabalina IG, Timmons JA, Cannon B, NedergaardJ. Chronic peroxisome proliferator-activated receptor gamma (PPAR-gamma) activation of epididymally derived white adipocyte cultures revealsa population of thermogenically competent, UCP1-containing adipocytesmolecularly distinct from classic brown adipocytes. J Biol Chem 2010;285:7153–7164

35. Collins S, Yehuda-Shnaidman E, Wang H. Positive and negative control ofUcp1 gene transcription and the role of b-adrenergic signaling networks.Int J Obes (Lond) 2010;34(Suppl. 1):S28–S33

36. Fernandez-Marcos PJ, Auwerx J. Regulation of PGC-1a, a nodal regulatorof mitochondrial biogenesis. Am J Clin Nutr 2011;93:884S–90

37. Shan T, Liang X, Bi P, Kuang S. Myostatin knockout drives browning ofwhite adipose tissue through activating the AMPK-PGC1a-Fndc5 pathwayin muscle. FASEB J 2013;27:1981–1989

38. Narkar VA, Downes M, Yu RT, et al. AMPK and PPARdelta agonists areexercise mimetics. Cell 2008;134:405–415

diabetes.diabetesjournals.org Zhang and Associates 525