Page 1 of 56

1 Orally active osteoanabolic agent 6CβDglucopyranosyl(2S, 3S)(+) 5,7, 3',4'

2 tetrahydroxydihydroflavonol binds to receptors, with a preference for AdipoR1,

3 induces adiponectinassociated signaling and improves metabolic health in a rodent model of

4 diabetes.

5 Running title: GTDF is an orally active adiponectin mimetic

6 Abhishek Kumar Singh1,#, Amit Arvind Joharapurkar2,#, Mohd. Parvez Khan3, Jay Sharan Mishra1,

7 Nidhi Singh1, Manisha Yadav1, Zakir Hossain4, Kainat Khan3, Sudhir Kumar5, Nirav Anilkumar

8 Dhanesha2, Devendra Pratap Mishra5, Rakesh Maurya5, Sharad Sharma6, Mukul Rameshchandra

9 Jain2, Arun Kumar Trivedi1, Madan Madhav Godbole7, Jiaur Rahaman Gayen4 , Naibedya

10 Chattopadhyay2 and Sabyasachi Sanyal1,*

11

12 Authors’ affiliations:

13 1Biochemistry Division, CSIRCentral Drug Research Institute, 10 Janakipuram Extn, Sitapur Road, Lucknow

14 226031, UP, India. 2Zydus Research Center, SarkhejBavla NH#8A, Moraiya, Ahmedabad 382210, Gujarat,

15 India. 3Division of Endocrinology, CSIRCentral Drug Research Institute, 10 Janakipuram Extn, Sitapur Road,

16 Lucknow 226031, UP, India. 4Division of Phramacokinetics, CSIRCentral Drug Research Institute, 10

17 Janakipuram Extn, Sitapur Road, Lucknow 226031, UP, India. 5Division of Medicinal and Process Chemistry,

18 CSIRCentral Drug Research Institute, 10 Janakipuram Extn, Sitapur Road, Lucknow 226031, UP, India.

19 6Division of Toxicology, CSIRCentral Drug Research Institute, 10 Janakipuram Extn, Sitapur Road, Lucknow

20 226031, UP, India. 7Department of Molecular Medicine, Sanjay Gandhi Postgraduate Institute of Medical

21 Sciences, Lucknow 226014, UP, India.

22 # These authors contributed equally

23 *Address correspondence to Sabyasachi sanyal, e mail: [email protected], Biochemistry Division,

24 CSIRCentral Drug Research Institute, 10 Janakipuram Extn, Sitapur Road, Lucknow 226031, UP,

25 India ; Tel: (+91)9792366322; Fax: (+91)(522)2771941

26 Word Count 5412

27 Number of Figures 7

1

Diabetes Publish Ahead of Print, published online May 21, 2014 Diabetes Page 2 of 56

1 ABSTRACT

2 Adiponectin is an adipocytokine that signals through plasma membranebound adiponectin

3 receptors (AdipoR) 1 and 2. Plasma adiponectin depletion is associated with type 2

4 diabetes, and cardiovascular diseases. Adiponectin therapy however, is yet

5 unavailable owing to its large size, complex multimerization and functional differences of the

6 multimers. We report discovery and characterization of 6CβDglucopyranosyl(2S, 3S)

7 (+) 5,7, 3',4' tetrahydroxydihydroflavonol (GTDF) as an orally active adiponectin mimetic.

8 GTDF interacted with both AdipoRs, with a preference for AdipoR1. It induced adiponectin

9 associated signaling and enhanced glucose uptake and fatty acid oxidation in vitro, which

10 were augmented or abolished by AdipoR1 overexpression or silencing respectively. GTDF

11 improved metabolic health, characterized by elevated glucoseclearance, βcellsurvival,

12 reduced steatohepatitis, browning of white adipose tissue and improved lipid profile in an

13 AdipoR1expressing but not an AdipoR1depleted strain of diabetic mice. The discovery of

14 GTDF as an adiponectin mimetic provides a promising therapeutic tool for the treatment of

15 metabolic diseases.

16

17

18

19

20

21

22

23

24

25 2

Page 3 of 56 Diabetes

1 The antiinflammatory adipocytokine adiponectin (1; 2) signals through adiponectin

2 receptors (AdipoR) 1 and 2 (3). Tcadherin, a cadherin family member that lacks

3 transmembrane and cytoplasmic domains also binds adiponectin, and is proposed to affect its

4 bioavailability (4). Plasma adiponectin depletion is associated with type 2 diabetes, obesity

5 and cardiovascular diseases (57). Adiponectin administration or overexpression ameliorates

6 resistance, metabolic syndrome and atherosclerosis in animals (3; 812) and enhances

7 pancreatic β cell survival (13). These evidences make AdipoRs important therapeutic targets

8 for metabolic diseases.

9 Structurally, adiponectin belongs to the complement 1q family (1; 14; 15).

10 Adipoenctin monomer is a 30 kDa consisting of an Nterminal collagenous and a C

11 terminal globular domains (1). Mammalian plasma adiponectin is present in several

12 multimeric forms; lowmolecularweight dimer or trimers, mediummolecularweight

13 hexamers or highmolecularweight (HMW) dodecamers and 18 mers (10; 1618). The

14 globular domain of adiponectin (gAd) can form trimers and was initially shown to exist as a

15 proteolytic cleavage product in human plasma (10), although subsequent studies failed to

16 detect it in circulation, its ability to modulate AdipoRs is undisputed. All these forms display

17 different levels of physiological activity and the HMW complex is considered the most

18 clinically relevant form (10; 1618). The HMW fulllength adiponectin and gAd

19 preferentially signal through AdipoR2 and AdipoR1 respectively (3). Given the

20 multimerizationrelated complexities of adiponectin structure and function, it appears that

21 small molecule AdipoR ligands may provide the only viable therapeutic option against

22 diseases associated with defects in adiponectin expression or action.

23 We have previously identified GTDF, a novel natural analog of the dietary flavonoid

24 , as a potent orally bioavailable osteoanabolic compound that induced proliferation,

25 differentiation and mineralization of cultured primary osteoblasts at a nanomolar 3

Diabetes Page 4 of 56

1 concentration that was 1000 fold less than effective concentration of quercetin or queretinO

2 glucoside, and restored trabecular bones of osteopenic rats on a par with parathyroid

3 (19). While studying its mechanism of action, we found that GTDF induced rapid AMP–

4 dependent protein kinase (AMPK), AKT and p38 phosphorylation and elevated PGC1α

5 expression in osteoblasts. GTDF also deacetylated tumor suppressor P53 via indirect

6 activation of NADdependent deacetylase Sirtuin1 (sirt1) [(20), and manuscript in

7 preparation, SS and NC)]. Literature search revealed that adiponectin elicits similar cellular

8 signaling (1; 3; 11). Interestingly, the quercetin group of compounds display functional

9 properties similar to adiponectin, such as AMPK activation, glucose uptake enhancement,

10 induction of fatty acid oxidationrelated and amelioration of diabetes and insulin

11 resistance in vivo (2123). We thus asked if GTDF, quercetin or other naturally occurring

12 quercetin analogs could be adiponectin mimetics. Here we report detailed characterization of

13 GTDF as an adiponectin mimetic that improves metabolic health in a rodent model of

14 diabetes.

15

16 RESEARCH DESIGN AND METHODS

17 Materials and kits: All cell culture reagents were from Invitrogen, Life Technologies

18 (Carlsbad, CA). Fine chemicals were from SigmaAldrich (St. Louis, MO) unless otherwise

19 indicated. Epoxy agarose beads conjugated with GTDF were constructed at Shantani Biotech

20 (Pune, India). Globular adiponectin (gAd) was purchased from Enzo Life Sciences

21 (Farmingdale, NY) and ATGen Global (Gyeonggido, South Korea) and compared; gAd

22 from both sources showed identical activity; gAd from ATGen Global was used in this study.

23 GW7647 was from SigmaAldrich, full length human AdipoR1 and AdipoR2 mammalian

24 expression constructs were from Open Biosystems (Huntsville, AL). 125IgAd was from

25 Phoenix Pharmaceuticals (Burlingame, CA), 125I (20 MBq) used for radiolabeling GTDF was 4

Page 5 of 56 Diabetes

1 from BARC (Mumbai, India). Glycogen assay kit was from SigmaAldrich (St. Louis,

2 Missouri). Serum concentrations of triglycerides, nonesterified free fatty acids (NEFA),

3 HDL, LDL,VLDL, total cholesterol, betahydroxybutyrate, and creatinine were determined

4 using kits purchased from Pointe Scientific (Canton, MI, USA).Circulating , insulin,

5 C, , adiponectin, ghrelin, corticosterone and markers of inflammation (MCP1,

6 TNFα, and IL6) were estimated using linked immunosorbent assay (ELISA) assay

7 kits (R&D Systems, Minneapolis, MN). Serum concentrations of sodium and potassium were

8 measured using a Cobas c system (Roche Diagnostics; Indianapolis, IN). TUNEL assay kit

9 was from Roche Applied Science (Indianapolis, IN). Plasma membrane extraction kit was

10 from Biovision (Milpitas, CA).

11 Cell culture and induction of differentiation

12 HEK293, CHO, C2C12 and 3T3L1 celllines were cultured as reported (24). Mouse stromal

13 vascular fraction (SVF) was prepared from epididymal fat pad by collagenase digestion.

14 Human SVFs were prepared from human lipoaspirates (subcutaneous), collected following

15 approval of Institutional Ethical Committee. Cells were differentiated in presence of GTDF

16 or vehicle for 10d (3T3L1 and mouse SVF) or 21d (human SVF), using standard procedure,

17 and then analyzed by QPCR, immunoblotting or microscopy.

18 Iodination and purification of 125I-GTDF

19 GTDF was radiolabeled as previously described for quercetin1 with minor modifications.

20 Briefly, 10l of 125I (20 MBq; BARC, Mumbai, India) was added to 100g GTDF in 5%

21 acetic acid/methanol, then chloramineT (4g in MilliQ H2O) was added, and the mixture

22 was allowed to react at room temperature (240C) for 5 min. The reaction was terminated by

23 adding 60l sodium metabisulphite (4mg/ml in MilliQ H2O).The reaction mixture was dried

24 by passing nitrogen and was dissolved into methanol (100 l). Reverse phase TLC (RP18

5

Diabetes Page 6 of 56

1 F254s, Merck, 8cm in length) was used to purify 125IGTDF from free iodine and unlabeled

2 compound using methanolwater (40%60%) as mobile phase. Following run, the TLC plate

3 was cut into pieces of 0.5mm each and the distribution of radioactivity along the plate was

4 measured in a gamma Counter. TLC of the blank reaction suggested the location of free 125I

5 in the TLC plate. The RF value of the labeled compound was determined by gamma

6 counting. The area showing maximum activity at distance of 40 to 60 mm was eluted from

7 the TLC plate, and was washed with methanol, centrifuged, decanted and dried under N2.

8 Overexpression and silencing experiments

9 Plasmid transfections were performed with Lipofectamine LTX (Life Technologies

10 Carlsbad, CA) reagent according to manufacturer’s protocols. For C2C12 transfections,

11 C2C12 myoblasts were trypsinized and transfected. The cells were then grown till 6070%

12 confluence. They were then differentiated and assessed as required. For RNA interference,

13 siRNAs (siAdipoR1; L063377010010and siC; siRNA against luciferase GL3 duplex D

14 0014000150) were purchased from Thermo Scientific (Pittsburgh, PA). Cells were

15 transfected with 0.1µM of each siRNA using DharmaFECT 1 transfection reagent (Thermo

16 Scientific). 72 h after transfection cells were treated and were analyzed as required.

17 QPCR, Western blotting, co-immunoprecipitation

18 These studies were performed as previously described (24). List of primer sequences for

19 qPCR has been provided in supplementary table 5. AntiPGC1α (ST1202; Millipore,

20 Billerica, MA) was used at 1:2000 dilution and antiCD36 (188361AP; Proteintech

21 (Chicago, IL) was used at 1:1000 dilution. AntiUCP1 (ab10983) UCP3 (ab3477) and

22 PPARα (ab8934); Abcam (Cambridge, MA) were used at 1:1000 dilutions. Phospho AMPK

23 (Thr172), AMPK, phospho p38(Thr 180/ Tyr 182), p38, phospho ACC, ACC, Glut4, P53,

24 AcetylP53 (Lys 382), C/EBPβ, and βactin antibodies were from Cell Signalling

6

Page 7 of 56 Diabetes

1 Technology (Beverly, MA) and were used at 1:1000 except βactin which was used at

2 1:3000 dilution. Antibodies against AdipoR1 (sc46748), AdipoR2 (sc46755) and N

3 cadherin (sc1502) were from Santacruz Biotechnology (Dallas, TX) and used at 1:1000

4 dilutions. For immunohistochemistry, insulin antibody (Cell Signalling Technology; Beverly,

5 MA) was used at a dilution of 1:200, phycoerythrin tagged Ki67 antibody (BD

6 Biosciences;San Diego, CA) was used at a dilution of 1:100 and anti UCP1 (ab10983) was

7 used at a dilution of 1:250.

8 Glucose-uptake

9 Fully differentiated C2C12 myotubes on 24 well plates were treated with vehicle (0.1%

10 DMSO) or 0.01 µM GTDF for 24h, following which the cells were serum starved for 3h. The

11 cells were then washed three times in warm (370 C) HEPES buffer solution ( HBS; 140 mM

12 sodium chloride, 20 mM HEPES, 5 mM potassium chloride, 2.5 mM sulfate, 1

13 mM calcium chloride, pH 7.4) and then were treated with warm HBS or 0.1µM insulin (in

14 HBS) for 20 min. Subsequently cells were washed 3X in warm HBS and were then

15 incubated in 250µl transport solution (HBS containing with 1µCi 3Hdeoxyglucose (Perkin

16 Elmer) and 10µM unlabeled 2deoxyglucose (SigmaAldrich) per well for 5 min. Then the

17 transport solution was aspirated and the cells were washed 3X with icecold stop solution

18 (0.9% NaCl and 25mM dextrose). Subsequently the cells were lysed in 100µl 0.5N NaOH

19 and 5µl lysate was used for determination of protein concentration (by Bichinconic acid

20 assay: SigmaAldrich), and rest of the lysate were used to measure cellular radioactivity in a

21 betacounter (Beckman Coulter, New Delhi, India).

22 Fatty acid oxidation

23 C2C12 myotubes plated in 12 well plates were treated with vehicle (0.1% DMSO) or 0.01µM

24 GTDF for 2h, 24h or 120h. Following treatment, the cells were washed 3X in warm HBS and

25 were then incubated with medium containing 0.75 mM palmitate (conjugated to 2% fatty acid 7

Diabetes Page 8 of 56

1 free BSA)/14C palmitate at 2 µCi/ml) for 2h. Following this incubation period, 1 ml of the

2 culture medium was removed and transferred to a sealable tube, the cap of which housed a

3 Whatman (GF/B) filter paper disc that had been presoaked with 1M potassium hydroxide.

14 4 CO2 trapped in the media were then released by acidification of media using 60% (vol/vol)

5 perchloric acid and gently agitating the tubes at 37°C for 2 h. Radioactivity that had become

6 adsorbed onto the filter discs were then be quantified by liquid scintillation counting in a

7 betacounter (Beckman Coulter).

8

9 Animal Experiments

10 Animal studies were approved by Institutional Animal Ethics Committee (IAEC) of Zydus

11 Research Centre, Ahmedabad. This facility is accredited to AALAC. Mice were individually

12 housed at 21◦C, in 12h light:12h dark cycles. All animals had access to normal chow diet

13 and water ad libitum. 12week old male BL6 (2530g), db/db or BKSdb/db (4550g) were

14 divided into different treatment groups randomly (db/db; n=8/group and BKSdb/db;

15 n=6/group). Vehicle groups received 1% Carboxymethylcellulose and the other groups

16 received GTDF or pioglitazone (10mg/kg), once a day, by oral gavage for 30 days. Feed

17 intake and body weight were measured every day. Blood was obtained from tail snips and

18 glucose levels were measured using a glucometer. Glucose tolerance tests were performed on

19 day 29 after overnight fasting. Pyruvate tolerance test was performed on Day 31, after an

20 overnight fast. On day 32, the animals were fasted for six hours and sacrificed. Plasma and

21 tissues were collected and stored at 80oC until further analysis. Hepatic glycogen was

22 estimated using a glycogen assay kit. IPGTT, IPPTT, estimation of circulating biomarkers,

23 histology and immunohistochemistry were performed using standard procedures.

24 EDL muscle culture and treatment

8

Page 9 of 56 Diabetes

1 EDL muscle strips along with tendons were dissected out from anesthetized mice and were

2 tied to stainless steel clutch pins by the tendons, without stretching. Muscles were

3 preincubated for 30 min at 37°C in oxygenated (95% O2/5% CO2) Krebs–Henseleit solution

4 (pH 7.4), then again for 30 min in the same medium with or without 2.5 g/ml gAd or 0.1µM

5 GTDF. The incubation media were gassed continuously with 95% O2/5% CO2 through

6 hypodermic needle. At the end of this incubation, tendons were removed, the muscles were

7 blotted on gauze pads and snap frozen in liquid N2 and were stored at −80°C until further

8 analysis.

9

10 Data analysis and Statistics

11 Results are expressed as mean ± SEM. All data were analyzed using GraphPad Prism 5.0

12 (GraphPad, San Diego, CA). Animal data involving more than two groups were analyzed

13 using twoway ANOVA followed by Bonferroni post test or one way ANOVA followed by

14 Tukey’s multiple comparison test as appropriate. In vitro or in vivo data involving two

15 experimental groups were analyzed using twotailed unpaired Student’s ttest or Mann

16 Whitney U test as appropriate.

17

18 RESULTS

19 GTDF binds to AdipoRs and mimics adiponectin-associated signaling events in vitro.

20 We assessed GTDF, quercetin and its natural analogs (chemical structures in Fig. 1)

21 in a “peroxisome proliferatoractivated (PPAR) α activitation assay”, a

22 previously reported functional assay for AdipoRs (3), in HEK293 cells that express

23 endogenous AdipoR1 (3). All the quercetin analogs tested enhanced PPARα activity in

24 presence of PPARα agonist GW7647 (Fig. 2A). GTDF maximally activated the reporter at

25 0.01µM, while dihydroquercetin (DHQ) an enantiomer of the aglycone form of GTDF and 9

Diabetes Page 10 of 56

1 quercetin6CβDglucopyranoside (QCG) did so at 0.1µM, and quercetin, quercetin3Oβ

2 Dglucopyranoside (QOG) or quercetin3OαLrhamnopyranoside (QR) caused maximal

3 activation at 1µM (Fig. 2A). Amplitudes of activation of PPARα ligand activity by GTDF

4 and gAd were comparable (Fig. 2B). GTDF did not activate PPARα in absence of GW7647,

5 indicating that it was not a PPARα agonist per se (Fig. ).

6 To assess if GTDF and AdipoRs could physically interact, we immobilized GTDF on

7 agarose beads and performed a pulldown assay with purified plasma membrane from C2C12

8 myotubes that expressed both AdipoR1 and R2 (3). GTDF beads but not control beads

9 successfully pulled down AdipoR1 and R2, and free GTDF or QCG competed with this

10 binding (Fig. 2D). Cold GTDF competed with 125Iadiponectin for binding to C2C12 cells in

11 a radioligand binding assay (Fig. 2E). Radioligand saturation assays with 125I labeled GTDF

12 (Fig. 2F) or 125IgAd (Supplementary Figure 2) revealed that both these ligands bound to

13 AdipoRdeficient chinese hamster ovary (CHO) cells (25) transfected with AdipoR1 or R2

14 but not empty expression plasmids (Fig. 2G) (GTDF: Kd and BMAX; 4.90nM and

15 1410fmol/mg of protein for AdipoR1 and 326 nM and 3950 fmol/mg of protein for AdipoR2;

16 gAd: Kd and BMAX 0.25 g/ml and 130ng/mg for AdipoR1 and 0.526 g/ml and 200ng/mg for

17 AdipoR2). Thus GTDF displayed about 70 fold more affinity towards AdipoR1 over

18 AdipoR2.

19 Consistent with adiponectinassociated rapid signaling events (1; 26) GTDF induced

20 AMPK, acetyl coA carboxylase (ACC), and p38 phosphorylation in C2C12 myotubes (Fig.

21 2H). While p38 phosphorylation was rapid and sustained, AMPK and ACC phosphorylation

22 peaked at 1 and 10 min respectively, and returned to the basal level at 60 min (Fig. 2H). A

23 similar pattern of AMPK and ACC phosphorylation by adiponectin has been reported earlier

24 (11).

10

Page 11 of 56 Diabetes

1 Apart from adiponectin glutamate receptors also activate AMPK and p38 (27), and we

2 therefore assessed if GTDF could modulate these two receptors, however, GTDF failed to

3 bind to or activate/repress any of the ionotropic or metabotropic glutamate receptors (mGlu),

4 while quercetin strongly activated mGlu2 and inhibited mGlu 4,6 and 8 (Supplementary

5 Tables S1 and S2). To also assess if these signaling events were mediated through one or

6 more Gprotein coupled receptors (GPCR), GPCR modulation by GTDF was assessed and it

7 failed to activate or repress any of the 158 GPCRs tested (Supplementary Tables S3 and S4).

8 Consistent with its activation of adiponectinassociated signaling, GTDF enhanced

9 mRNA and protein levels of factors associated with fatty acid transport [CD36 and fatty acid

10 binding protein 3 (FABP3)], oxidation [(carnityl palmitoyl transferase 1 b; CPT1B), long

11 chain fatty acyl CoA synthetase (FACS), acetyl CoA oxidase (ACOX1), PPARα and

12 PPARδ)], mitochondrial biogenesis (PPARγ coactivator1α; PGC1α), mitochondrial

13 uncoupling protein (UCP) 3, and glucose transporter 4 (Glut4), while lipoprotein lipase

14 (LPL) and PPARγ were unchanged (Fig. 2I and 2J). Adiponectin deacetylates and activates

15 PGC1α via indirect activation of Sirt1 (26), and consistent with its indirect sirt1 activation

16 (20), GTDF too deacetylated PGC1α in C2C12 myotubes (Fig. 2K). This PGC1α induction

17 and activation led to increased mitochondrial DNA copy number (Fig. 2L). gAd enhances

18 basal glucose uptake (3; 11) and increases insulinsensitivity(28) in C2C12 myotubes, and in

19 consistence, GTDF too enhanced basal and insulinstimulated glucose uptake in these cells

20 (Fig. 2M). Further, GTDF also significantly enhanced basal and insulindependent glucose

21 uptake in a C2C12 myotube model of palmitateinduced insulin resistance (Fig. 2N). Similar

22 to rapid enhancement of fatty acid oxidation by adiponectin (3; 11; 29), GTDF significantly

23 enhanced 14C palmitate oxidation in C2C12 myotubes within 2h, which increased further

24 over time (Fig. 2O).

25 11

Diabetes Page 12 of 56

1 Overexpression or silencing of AdipoR1 augments or mitigates GTDF functions in vitro.

2 GTDF significantly enhanced insulinstimulated glucose uptake in CHO/HIRc cells

3 [CHO cells expressing ectopic human (30)] transfected with AdipoR1 but not

4 empty plasmid (Fig. 3A). Consistent with the binding studies AdipoR1 but not AdipoR2

5 overexpression in C2C12 myotubes significantly enhanced GTDFstimulated AMPK, ACC

6 and p38 phosphorylation (Fig. 3B). Small interfering RNA (siRNA) for AdipoR1

7 (siAdipoR1) but not control siRNA against luciferase (siC) abolished GTDFinduced AMPK,

8 ACC and p38 phosphorylation, without affecting AdipoR2 expression. Further, GTDF

9 stimulated glucose uptake and fatty acid oxidation were abolished by siAdipoR1 (Fig. 3C-

10 3E), while insulinstimulated glucose uptake was unaltered (Fig. 3D). Together with Fig 1

11 and supplementary tables 14, these results demonstrate that GTDF action indeed is

12 AdipoR1specific.

13

14 BKS.Cg-Dock7m +/+ Leprdb/db/J (BKS-db/db) mice have severely depleted plasma

15 membrane-associated AdipoR1 compared to B6.BKS(D)-Leprdb/db/J (db/db) mice.

16 A relevant animal model to test AdipoRspecificity of GTDF would have been

17 AdipoR1/R2 knockout mice subjected to dietinduced obesity. However, our inability to

18 obtain the knockout mice led us to search for an alternate model. Since chronic high level of

19 plasma insulin causes an AdipoRdepletion mediated adiponectinresistance (31), we

20 systematically investigated AdipoR expression in major adiponectin target organs, across

21 different age groups in two different strains of deficient obese and diabetic

22 mice; db/db (in C57BL/6J background) and BKSdb/db (in BLKS/J background), and wild

23 type healthy C57BL/6J (BL6).

24 Comparison of 12 week old male BL6, db/db and BKSdb/db mice revealed that both

25 diabetic mice had lower AdipoR mRNAs in skeletal muscle, liver and eWAT compared to 12

Page 13 of 56 Diabetes

1 the healthy BL6 mice, however, between the diabetic strains only modest differences were

2 observed (Fig. 4A).

3 Examination of total and plasma membrane (PM) associated AdipoR protein levels

4 however, revealed striking differences between the two diabetic strains. While BKSdb/db

5 displayed >80% decrease in total and PMassociated and therefore functional AdipoR1

6 expression in skeletal muscle, BL6 and db/db did not show significant difference (Fig. 4B).

7 In liver, PM AdipoR1 but not AdipoR2 was strongly depleted (≥80%) in BKSdb/db but not

8 in db/db, while compared to BL6 total AdipoR1 and R2 were diminished in both. In

9 WAT, both diabetic strains displayed >90% reduction in total AdipoR1 protein compared to

10 BL6, while AdipoR2 protein was not detected (Fig. 4B). AdipoR2 could not be detected in

11 PM fraction of skeletal muscle (Fig. 4B) and we failed to generate enough PM extract from

12 WAT for immunoblotting. Together, compared to db/db and BL6, BKSdb/db displayed

13 severely depleted total and PM AdipoR1 protein in skeletal muscle and liver at 12 weeks of

14 age. Consistent with this observation, gAd and GTDF failed to induce AMPK

15 phosphorylation in extensor digitorium longinus (EDL) muscles of BKSdb/db, while it did

16 so in EDL muscles from both BL6 and db/db mice (Fig. 4C). Thus, based on above evidences

17 we selected 12week old male db/db mice as adiponectinsensitive, and age and sex matched

18 BKSdb/db as adiponectinresistant models for the in vivo pharmacological studies.

19

20 GTDF fails to improve diabetic phenotype in BKS-db/db mice.

21 Over a 30d treatment period, GTDF did not alter feedintake or body weight in BKS

22 db/db (Fig. 5A), while PPARγ agonist pioglitazone (Pio) significantly enhanced body weight

23 6d onwards without altering feedintake (Fig. 5B). GTDF failed to alter nonfasting blood

24 glucose, fasting blood glucose and glucose clearance, while Pio significantly improved these

13

Diabetes Page 14 of 56

1 parameters (Fig. 5Cand 5D). GTDF failed to alter plasma glycated haemoglobin (HbA1c)

2 and insulin (Fig. 5E and 5F). GTDF was ineffective in improving the fasting lipid profile in

3 these mice, while pio modestly but significantly reduced plasma TG and VLDL (Fig. 5G).

4 These results indicate that GTDF is ineffective in BKSdb/db mice that display severely

5 depleted AdipoR1 protein in skeletal muscle and liver. That pio improved glycemic

6 parameters in these mice further indicates that GTDF did not function through the PPARγ

7 pathway.

8

9 GTDF ameliorates diabetic phenotype in AdipoR1-expressing db/db mice.

10 In contrast to its inefficacy in BKSdb/db (Fig. 5), GTDF was remarkably effective in

11 AdipoR1expressing db/db mice. It reduced feed intake in db/db but not BL6 (Fig. 6A),

12 which could be correlated with a robust reduction in serum ghrelin (Fig. 6N). GTDFtreated

13 db/db mice displayed reducing trend in body weight, which however was not statistically

14 significant (Fig. 6B) and showed significantly reduced adipose tissue weight (Supplementary

15 Figure 3). GTDF remarkably reduced nonfasting blood glucose in db/db mice on the 7th day

16 of treatment (Fig. 6C). Reduction in feed intake could not be responsible for the GTDF

17 induced reduction in nonfasting glucose level in db/db, as the feed intake was significantly

18 different only after 16d of treatment, while nonfasting blood glucose was drastically reduced

19 at the 7th day and did not fall any further.

20 GTDF modestly yet significantly reduced fasting blood glucose in db/db but not in

21 BL6 (Fig. 6D), which was associated with modestly reduced hepatic gluconeogenesis as

22 revealed by a pyruvate tolerance test (PTT) (Fig. 6E). GTDF treatment prevented increase in

23 HbA1c in db/db, which increased in the vehicletreated db/db during the treatment period

24 (Fig. 6F). In intraperitoneal glucose tolerance tests (IPGTT), GTDFtreated db/db mice

14

Page 15 of 56 Diabetes

1 showed remarkably improved glucose clearance (Fig. 6G), however, both fasting insulin as

2 well as insulin levels during glucose challenge was higher in these mice over vehicletreated

3 controls, while they were unchanged in BL6 mice (Fig. 6G-6I). The enhanced insulin level

4 was associated with increase in pancreatic Cpeptide (Fig. 6J), indicating that insulin

5 production was higher in GTDFtreated db/db mice. However, hypokalemia, generally

6 associated with higher insulin, was not observed (Fig. 6K).

7 Pancreatic histomorphometry revealed that in vehicletreated db/db mice, islet number

8 was diminished in comparison to BL6, and GTDF significantly increased it in the former but

9 not later (Fig. 6Land 6M). Furthermore, about 30% of the β cell population in the islets of

10 vehicletreated db/db mice were apoptotic as determined by TUNEL staining whilst GTDF

11 markedly attenuated the number of apoptotic β cells (Fig. 6L and 6M). The increased islet

12 number in GTDF treated db/db was not due to cellular proliferation as the islets

13 from these mice did not stain for the proliferation marker Ki67 (Fig. 6l), indicating that

14 GTDF might protect beta cells from gluco and lipotoxicityinduced apoptosis. Serum

15 glucagon level was unchanged (Fig. 6N).

16 Consistent with antiinflammatory properties of adiponectin, GTDF treatment in

17 db/db mice significantly lowered serum tumor necrosis factor (TNFα), and interleukin

18 6 (IL6) showed a reducing trend, whilst monocyte / macrophage chemoattractant protein1

19 (MCP1) was unchanged (Fig. 6N). While serum adiponectin showed an increasing trend

20 upon GTDF treatment (p=0.082), leptin level was greatly diminished (Fig. 6N). We also

21 found a robust fall in serum corticosterone (Fig. 6N), indicating that GTDF might be cardio

22 protective. Analysis of serum lipid profile revealed significant reductions in total cholesterol,

23 triglyceride (TG), very lowdensity lipoprotein (VLDL), nonesterified fatty acids (NEFA)

24 and ketone bodies (βOH butyrate) which supports an enhanced fatty acid oxidation rate,

25 while it significantly enhanced high density lipoprotein (HDL) level (Fig. 6N), reiterating its 15

Diabetes Page 16 of 56

1 cardioprotective promise. Low–density lipoprotein (LDL) and creatinine levels were

2 unchanged (Fig. 6N). In light of the lipid profile data, we examined the hepatic histology, and

3 sections from GTDFtreated db/db mice displayed no vacuolation and lipid accumulation,

4 while the vehicletreated db/db exhibited robust lipid accumulation (Fig. 6O). Hepatic

5 glycogen level was unchanged (Fig. 6P). Together, Fig. 5 and 6 indicated that GTDF stalls

6 diabetes progression in AdipoRexpressing db/db mice and improves their overall metabolic

7 health, while it is ineffective in AdipoR1depleted BKSdb/db.

8 GTDF induces browning of white adipose tissue, evidence of involvements of direct and

9 indirect pathways.

10 GTDF induced and activated PGC1α in myocytes and PGC1α is reported to induce

11 FNDC5, a myokine that drives browning of myf5negative white adipocytes (32). GTDF

12 indeed enhanced FNDC5 expression in C2C12 myotubes and increased its secretion in

13 culture medium (Fig. 7A) indicating that it might induce FNDC5 in circulation and thereby

14 promote browning of WAT. Histological analysis of epididymal WAT (eWAT) from GTDF

15 treated db/db mice revealed robust fat mobilization characterized by reduction in adipocyte

16 size (Fig. 7B) and these eWATs stained strongly for the brown adipose marker UCP1 (Fig.

17 7C), indicating that GTDF indeed induced a browning “like” phenomenon.

18 Adipocytespecific transgenic overexpression of adiponectin is reported to mobilize

19 WAT, enhance UCPs and decrease TNFα and leptin level in this tissue (33), indicating that

20 adiponectin may also promote a FNDC5independent browning of WAT through a direct

21 action on adipocytes. Differentiation of 3T3L1 cells in presence of GTDF from day 0 or day

22 2 of differentiation reduced oildroplet accumulation in these cells and significantly reduced

23 triglyceride content (Fig. 7D and 7E). Differentiation of 3T3L1, mouse (epididymal) or

24 human SVFs in presence of GTDF enhanced expressions of UCP1, UCP2, PGC1α, brown

25 adipose determination factor PRDM16, brownfatenriched protein CIDEA and adiponectin 16

Page 17 of 56 Diabetes

1 (Fig. 7F-7H). PRDM16 and ccaat/enhancer protein beta (C/EBPβ) transcriptional complex

2 has been implicated in conversion of myf5 positive myoblasts into BAT (34), and GTDF

3 also induced the expression of c/EBPβ in 3T3L1, mouse and human SVF (Fig. 7H),

4 indicating that this complex may play an important role in the browning phenomenon

5 observed here. Consistent with elevated browning markers, GTDF increased mitochondrial

6 DNA content in mouse SVFs (Fig. 7I). Notably; GTDF did not activate βadrenergic

7 receptors (Supplementary Tables 3 and 4 and Fig. 7J).

8

9

10 DISCUSSION

11 Following comprehensive molecular characterizations, we have identified GTDF as

12 an orally active, small molecule adiponectin mimic that improves metabolic parameters in a

13 preclinical disease setting and holds therapeutic promise in metabolic diseases caused by

14 adiponectin deficiency.

15 GTDF interacted with AdipoRs and displayed a 70 fold higher affinity for AdipoR1

16 over R2. It induced adiponectinassociated rapid signaling, expressions and functional

17 events in C2C12 myotubes which were respectively augmented or abolished by AdipoR1

18 overexpression or silencing.

19 Our inability to procure AdipoR knockout mice inspired us to explore alternate

20 model systems and led to the identification of 12 week old male BKSdb/db mice as

21 AdipoR1deficient, while age and sex matched db/db mice were found to be AdipoR1intact.

22 Intriguingly, these differences were not apparent at the mRNA level but were pronounced in

23 the protein level, especially at the level of PMassociated and hence functional form of

24 AdipoR1 protein. These findings indicate that apart from transcriptional regulation by insulin

25 and Foxo1 (31), AdipoR1 expression and function may also be regulated by post 17

Diabetes Page 18 of 56

1 transcriptional or translational events. Interestingly, a recent report characterized a

2 developmentally regulated alternate splice variant of AdipoR1 in human subjects that was

3 strongly enhanced during skeletal muscle differentiation and was decreased in diabetic

4 patients, and the protein level of AdipoR1 accurately represented these changes but not total

5 mRNA levels that accounted for all AdipoR1 splice variants(35). Our data gains further

6 support from another recent report, which describes that microRNA221 and the RNA

7 binding protein polypyrimidine tract binding protein regulate AdipoR1 protein expression

8 and are induced in genetic and dietinduced mice models of obesity (36).

9 Consistent with AdipoR1 deficiency, BKSdb/db mice were refractory to GTDF,

10 while metabolism in db/db mice was remarkably improved. Interestingly, GTDF reduced

11 feedintake in db/db mice from 16 day onwards. Effect of adiponectin on feedintake is

12 controversial and all three possible outcomes have been reported (3739). While the reason

13 for these differences is unclear, strain of mice, mode of administration and the administered

14 form of adiponectin appear to be responsible. It however, is important to note that we

15 delivered GTDF by oral route, in contrast to systemic delivery or transgenebased

16 overexpression of adiponectin, and thus GTDF may impact satiety related from

17 gut. In agreement with this notion, plasma ghrelin level was greatly reduced in GTDFtreated

18 db/db mice. Ghrelin is chiefly produced by P/D1 cells lining the fundus of stomach and given

19 the current lack of information regarding expression of AdipoRs in these cells it’s presently

20 unclear if GTDFmediated downregulation of ghrelin level is achieved through AdipoRs in

21 these cells. However, since GTDF did not alter feedintake in adiponectininsensitive BKS

22 db/db, it seems probable that these cells may express one or more AdipoRs which may act as

23 sensors for dietderived putative AdipoR ligands.

24 In addition to alteration in feedintake, GTDF remarkably reduced nonfasting blood

25 glucose that was significant from day 7 onwards and did not fall thereafter. GTDF also stalled 18

Page 19 of 56 Diabetes

1 the increase in HbA1c that was observed in vehicletreated db/db mice. Reduction in feed

2 intake could not account for these changes as earlier reports have demonstrated that caloric

3 restriction fails to not only decrease nonfasting blood glucose or HbA1c in db/db mice with

4 manifested diabetes (4042) but also fails to prevent onset of hyperglycemia in db/db mice

5 pairfed for 5 weeks since weaning (43). That GTDF decreased feedintake in db/db only but

6 not in BL6 and BKSdb/db, clearly indicates that toxicity was not involved. It’s further

7 supported by our earlier study where, GTDF not only normalized feedintake in high dose

8 dexamethasonetreated wistar rats, it also prevented dexamethsoneinduced mortality (20).

9 While the effect of GTDF on nonfasting blood glucose and glucose clearance were

10 robust, it caused significant but modest decrease in fasting blood glucose and hepatic

11 gluconeogenesis. Since GTDF showed a preference for AdipoR1, and R2 is the principal

12 AdipoR in liver, it appears that GTDFmediated reduction of blood glucose might be

13 principally achieved by skeletal muscle’s AdipoR1mediated glucose disposal, which is

14 supported by the fact that fasting hepatic glycogen was unaltered upon GTDF treatment.

15 However, a robust reduction in ketone bodies in GTDFtreated db/db mice indicates that in

16 these animals the liver may efficiently oxidize them.

17 BKSdb/db display much severe hyperglycemia than db/db and are susceptible to

18 terminal diabetes characterized by pancreatic β cell degranulation and death, while db/db

19 mice are protected from terminal diabetes due to the unique proliferation capacity of their β

20 cells (44). Thus, in older db/db mice hyperglycemia is corrected through a higher plasma

21 insulin level, although these mice remain severely dyslipidemic (44). In our experimental set

22 up, consistent with a lower plasma insulin (which although was still much higher than healthy

23 BL6) than db/db, the BKSdb/db mice indeed exhibited higher fasting and nonfasting blood

24 glucose. However, β cell proliferation was yet not apparent in db/db as evidenced by lack of

25 Ki67 staining; indicating that during the experimental period the db/db β cells did not yet go 19

Diabetes Page 20 of 56

1 on a proliferative drive. Further, about 30% of β cells from db/db mice were apoptotic and

2 GTDF strongly mitigated this apoptosis without increasing proliferation, indicating that

3 GTDF may protect these cells from gluco and lipotoxic stresses and a similar finding has

4 been reported for adiponectin (45).

5 Despite higher plasma insulin, GTDFtreated db/db mice were not hypokalemic and

6 displayed a remarkably improved lipid profile evidenced by decreased serum total

7 cholesterol, TG, VLDL and NEFA and increased HDL, which together with a marked decline

8 in plasma corticosterone in these mice, indicates that GTDF may have cardioprotective

9 properties. NEFA are breakdown products of TG that are released from adipocytes following

10 lipolysis and are important in diabetic pathogenesis (46). The marked decline in plasma

11 NEFA suggests that in GTDFtreated db/db mice NEFA were either utilized with robust

12 efficiency in liver and skeletal muscle, or adipose TG were efficiently oxidized in situ, or a

13 combination of both. These postulates could be corroborated by the facts that livers of

14 GTDFtreated db/db were free from vacuolation and oildroplets which were

15 characteristically present in vehicletreated db/db mice, and eWAT depot in GTDFtreated

16 db/db mice showed robust mobilization and increased UCP1 expression. Since both db/db

17 and BKSdb/db displayed depleted AdipoR1 protein in eWAT, it is possible that GTDF

18 caused browning of eWAT through an indirect mechanism involving previously described

19 PGC1αinduced myokine; FNDC5/irisin (32). Consistent with PGC1α induction and

20 activation, GTDF induced FNDC5 expression in C2C12 myotubes and its release in culture

21 medium. In addition, GTDF was also capable of directly inducing brown adipose markers

22 and increasing mitochondrial content in 3T3L1, mouse and human SVFs differentiated in its

23 presence, indicating that GTDF may protect against energy imbalancerelated metabolic

24 diseases. However, further studies including GTDF’s effect on energy expenditure is needed

25 to fully understand and realize its potential. 20

Page 21 of 56 Diabetes

1 While this manuscript was under review, OkadaIwabu et al reported identification and

2 characterization of a small molecule AdipoR agonist; Adiporon (47). While Adiporon is 2(4

3 Benzoylphenoxy)N[1(phenylmethyl)4piperidinyl]acetamide, GTDF is a flavone c

4 glucoside and these compounds do not share any structural homology. Functionally,

5 showed comparable affinity to both AdipoR1 and R2 and acted through both these receptors

6 to bring about physiological improvements in diabetic mice, while GTDF showed a stronger

7 affinity for AdipoR1 and given its modest effects on hepatic gluconeogenesis, appears to act

8 mainly via AdipoR1. Incidentally, another recent report identified DHQ (also known as

9 taxifoliol) as one of 9 small molecule AdipoR agonists from a library of 10,000 compounds

10 by a fluorescent polarizationbased screen, however, the in vivo efficacy of DHQ is yet to be

11 elucidated (48). DHQ is an enantiomer of the aglycone form of GTDF and was active in our

12 PPARα ligand activation as well, albeit at a 10 fold higher concentration than GTDF. In

13 contrast to GTDF, DHQ displays a stronger affinity for AdipoR2 (48) and thus studies with

14 DHQ alone or in combination with GTDF will be needed to further explore their therapeutic

15 potential in metabolic diseases.

16 In conclusion, discovery of GTDF as a small molecule adiponectin mimetic, that

17 remarkably improves metabolic health in diabetic mice provides a promising therapeutic tool

18 for treatment of adiponectindeficiencyassociated metabolic diseases. However, given the

19 lack of AdipoR knockout animal models in this study, it remains to be confirmed if all the

20 beneficial metabolic effects of GTDF were indeed routed through AdipoRs alone.

21

22 ACKNOWLEDGEMENTS

23 This work was supported by CSIR grant BSC0201 to SS and NC. AKS was supported by

24 fellowship from CSIR. MY, MPK were supported by ICMR fellowships, JSM, NS, JH were

25 supported by fellowships from UGC, KK was supported by DBT fellowship. 21

Diabetes Page 22 of 56

1

2 AKS, MPK, JSM, NS, MY, KK, DPM, RM, ShS, AKT, JRG, NC and SS have a pending

3 patent pertaining to some of the works in this manuscript. AAJ, NAD, MRJ are employees of

4 Zydus Research Center the research and development arm of Cadila Health Care limited,

5 Ahmedabad, India. This study was in no way sponsored or funded by an industry.

6

7 Part of the work was presented as an abstract in the “International Symposium on Molecular

8 Signaling” held at VisvaBharati University, Santiniketan, India from February 1821, 2013.

9

10 SS conceived the study. AKS, AAJ, JRG, MMG, NC and SS designed experiments. AKS,

11 AAJ, JSM, MPK, NS, MY, KK, JH, SK, NAD, DPM and MMG performed experiments.

12 MRJ, RM, AKT, MMG, NC, JRG and SS supervised experiments. ShS provided materials

13 and contributed to discussion. AKT, JRG, contributed to discussion. All authors analyzed

14 data. SS and NC contributed to discussion, wrote, edited and reviewed the manuscript. SS is

15 the guarantor of this work and, as such, had full access to all the data in the study and takes

16 responsibility for the integrity of the data and the accuracy of the data analysis.

17

18 Authors acknowledge Sophisticated Analytical Instrument Facility in CSIRCDRI for help

19 with confocal microscopy. The authors acknowledge Prem N Yadav, Ph.D. (Division of

20 Pharmacology, CSIR, CDRI) for help with designing of the radioligand binding assays and

21 Durga Prasad Mishra, Ph.D. (Division of Endocrinology, CSIRCDRI) for sharing antibodies.

22

23 REFERENCES

24 1. Kadowaki T, Yamauchi T: Adiponectin and adiponectin receptors. Endocr Rev 2005;26:439451 25 2. Robinson K, Prins J, Venkatesh B: Clinical review: adiponectin biology and its role in 26 inflammation and critical illness. Crit Care 2011;15:221

22

Page 23 of 56 Diabetes

1 3. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, Sugiyama T, Miyagishi M, Hara 2 K, Tsunoda M, Murakami K, Ohteki T, Uchida S, Takekawa S, Waki H, Tsuno NH, Shibata Y, 3 Terauchi Y, Froguel P, Tobe K, Koyasu S, Taira K, Kitamura T, Shimizu T, Nagai R, Kadowaki T: 4 Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 2003;423:762 5 769 6 4. Hug C, Wang J, Ahmad NS, Bogan JS, Tsao TS, Lodish HF: Tcadherin is a receptor for hexameric 7 and highmolecularweight forms of Acrp30/adiponectin. Proceedings of the National Academy of 8 Sciences of the United States of America 2004;101:1030810313 9 5. Hotta K, Funahashi T, Arita Y, Takahashi M, Matsuda M, Okamoto Y, Iwahashi H, Kuriyama H, 10 Ouchi N, Maeda K, Nishida M, Kihara S, Sakai N, Nakajima T, Hasegawa K, Muraguchi M, Ohmoto 11 Y, Nakamura T, Yamashita S, Hanafusa T, Matsuzawa Y: Plasma concentrations of a novel, adipose 12 specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol 13 2000;20:15951599 14 6. Li S, Shin HJ, Ding EL, van Dam RM: Adiponectin levels and risk of type 2 diabetes: a systematic 15 review and metaanalysis. Jama 2009;302:179188 16 7. Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE, Tataranni PA: 17 Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and 18 hyperinsulinemia. J Clin Endocrinol Metab 2001;86:19301935 19 8. Berg AH, Combs TP, Du X, Brownlee M, Scherer PE: The adipocytesecreted protein Acrp30 20 enhances hepatic insulin action. Nature medicine 2001;7:947953 21 9. Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, 22 TsuboyamaKasaoka N, Ezaki O, Akanuma Y, Gavrilova O, Vinson C, Reitman ML, Kagechika H, 23 Shudo K, Yoda M, Nakano Y, Tobe K, Nagai R, Kimura S, Tomita M, Froguel P, Kadowaki T: The 24 fatderived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and 25 obesity. Nature medicine 2001;7:941946 26 10. Fruebis J, Tsao TS, Javorschi S, EbbetsReed D, Erickson MR, Yen FT, Bihain BE, Lodish HF: 27 Proteolytic cleavage product of 30kDa adipocyte complementrelated protein increases fatty acid 28 oxidation in muscle and causes weight loss in mice. Proceedings of the National Academy of Sciences 29 of the United States of America 2001;98:20052010 30 11. Yamauchi T, Kamon J, Minokoshi Y, Ito Y, Waki H, Uchida S, Yamashita S, Noda M, Kita S, 31 Ueki K, Eto K, Akanuma Y, Froguel P, Foufelle F, Ferre P, Carling D, Kimura S, Nagai R, Kahn BB, 32 Kadowaki T: Adiponectin stimulates glucose utilization and fattyacid oxidation by activating AMP 33 activated protein kinase. Nature medicine 2002;8:12881295 34 12. Yamauchi T, Kamon J, Waki H, Imai Y, Shimozawa N, Hioki K, Uchida S, Ito Y, Takakuwa K, 35 Matsui J, Takata M, Eto K, Terauchi Y, Komeda K, Tsunoda M, Murakami K, Ohnishi Y, Naitoh T, 36 Yamamura K, Ueyama Y, Froguel P, Kimura S, Nagai R, Kadowaki T: Globular adiponectin 37 protected ob/ob mice from diabetes and ApoEdeficient mice from atherosclerosis. The Journal of 38 biological chemistry 2003;278:24612468 39 13. Holland WL, Miller RA, Wang ZV, Sun K, Barth BM, Bui HH, Davis KE, Bikman BT, Halberg 40 N, Rutkowski JM, Wade MR, Tenorio VM, Kuo MS, Brozinick JT, Zhang BB, Birnbaum MJ, 41 Summers SA, Scherer PE: Receptormediated activation of ceramidase activity initiates the 42 pleiotropic actions of adiponectin. Nature medicine 2011;17:5563 43 14. Hu E, Liang P, Spiegelman BM: AdipoQ is a novel adiposespecific gene dysregulated in obesity. 44 The Journal of biological chemistry 1996;271:1069710703 45 15. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF: A novel serum protein similar to 46 C1q, produced exclusively in adipocytes. The Journal of biological chemistry 1995;270:2674626749 47 16. Shapiro L, Scherer PE: The crystal structure of a complement1q family protein suggests an 48 evolutionary link to tumor necrosis factor. Curr Biol 1998;8:335338 49 17. Tsao TS, Murrey HE, Hug C, Lee DH, Lodish HF: Oligomerization statedependent activation of 50 NFkappa B signaling pathway by adipocyte complementrelated protein of 30 kDa (Acrp30). The 51 Journal of biological chemistry 2002;277:2935929362 52 18. Tsao TS, Tomas E, Murrey HE, Hug C, Lee DH, Ruderman NB, Heuser JE, Lodish HF: Role of 53 disulfide bonds in Acrp30/adiponectin structure and signaling specificity. Different oligomers activate 54 different signal transduction pathways. The Journal of biological chemistry 2003;278:5081050817 23

Diabetes Page 24 of 56

1 19. Sharan K, Mishra JS, Swarnkar G, Siddiqui JA, Khan K, Kumari R, Rawat P, Maurya R, Sanyal 2 S, Chattopadhyay N: A novel quercetin analogue from a medicinal plant promotes peak bone mass 3 achievement and bone healing after injury and exerts an anabolic effect on osteoporotic bone: the role 4 of aryl hydrocarbon receptor as a mediator of osteogenic action. J Bone Miner Res 2011;26:2096 5 2111 6 20. Khan MP, Mishra JS, Sharan K, Yadav M, Singh AK, Srivastava A, Kumar S, Bhaduaria S, 7 Maurya R, Sanyal S, Chattopadhyay N: A novel flavonoid Cglucoside from Ulmus wallichiana 8 preserves bone mineral density, microarchitecture and biomechanical properties in the presence of 9 glucocorticoid by promoting osteoblast survival: A comparative study with human parathyroid 10 hormone. Phytomedicine : international journal of phytotherapy and phytopharmacology 2013; 11 21. Nieman DC, Williams AS, Shanely RA, Jin F, McAnulty SR, Triplett NT, Austin MD, Henson 12 DA: Quercetin's influence on exercise performance and muscle mitochondrial biogenesis. Med Sci 13 Sports Exerc 42:338345 14 22. Jeong SM, Kang MJ, Choi HN, Kim JH, Kim JI: Quercetin ameliorates hyperglycemia and 15 dyslipidemia and improves antioxidant status in type 2 diabetic db/db mice. Nutr Res Pract 16 2012;6:201207 17 23. Nieman DC, Williams AS, Shanely RA, Jin F, McAnulty SR, Triplett NT, Austin MD, Henson 18 DA: Quercetin's influence on exercise performance and muscle mitochondrial biogenesis. Medicine 19 and science in sports and exercise 2010;42:338345 20 24. Dwivedi SK, Singh N, Kumari R, Mishra JS, Tripathi S, Banerjee P, Shah P, Kukshal V, Tyagi 21 AM, Gaikwad AN, Chaturvedi RK, Mishra DP, Trivedi AK, Sanyal S, Chattopadhyay N, 22 Ramachandran R, Siddiqi MI, Bandyopadhyay A, Arora A, Lundasen T, Anakk SP, Moore DD, 23 Sanyal S: Bile acid receptor agonist GW4064 regulates PPARgamma coactivator1alpha expression 24 through receptorrelated receptor alpha. Molecular endocrinology 2011;25:922932 25 25. Neumeier M, Weigert J, Schaffler A, Weiss T, Kirchner S, Laberer S, Scholmerich J, Buechler C: 26 Regulation of 1 in human hepatocytes by agonists of nuclear receptors. Biochem 27 Biophys Res Commun 2005;334:924929 28 26. Iwabu M, Yamauchi T, OkadaIwabu M, Sato K, Nakagawa T, Funata M, Yamaguchi M, Namiki 29 S, Nakayama R, Tabata M, Ogata H, Kubota N, Takamoto I, Hayashi YK, Yamauchi N, Waki H, 30 Fukayama M, Nishino I, Tokuyama K, Ueki K, Oike Y, Ishii S, Hirose K, Shimizu T, Touhara K, 31 Kadowaki T: Adiponectin and AdipoR1 regulate PGC1alpha and mitochondria by Ca(2+) and 32 AMPK/SIRT1. Nature 2010;464:13131319 33 27. Kim JH, Lee JO, Lee SK, Moon JW, You GY, Kim SJ, Park SH, Park JM, Lim SY, Suh PG, Uhm 34 KO, Song MS, Kim HS: The glutamate agonist homocysteine sulfinic acid stimulates glucose uptake 35 through the calciumdependent AMPKp38 MAPKprotein kinase C zeta pathway in skeletal muscle 36 cells. The Journal of biological chemistry 2011;286:75677576 37 28. Wang C, Mao X, Wang L, Liu M, Wetzel MD, Guan KL, Dong LQ, Liu F: Adiponectin sensitizes 38 insulin signaling by reducing p70 S6 kinasemediated phosphorylation of IRS1. The Journal of 39 biological chemistry 2007;282:79917996 40 29. Yoon MJ, Lee GY, Chung JJ, Ahn YH, Hong SH, Kim JB: Adiponectin increases fatty acid 41 oxidation in skeletal muscle cells by sequential activation of AMPactivated protein kinase, p38 42 mitogenactivated protein kinase, and peroxisome proliferatoractivated receptor alpha. Diabetes 43 2006;55:25622570 44 30. Wilden PA, Backer JM, Kahn CR, Cahill DA, Schroeder GJ, White MF: The insulin receptor with 45 phenylalanine replacing tyrosine1146 provides evidence for separate signals regulating cellular 46 metabolism and growth. Proceedings of the National Academy of Sciences of the United States of 47 America 1990;87:33583362 48 31. Tsuchida A, Yamauchi T, Ito Y, Hada Y, Maki T, Takekawa S, Kamon J, Kobayashi M, Suzuki 49 R, Hara K, Kubota N, Terauchi Y, Froguel P, Nakae J, Kasuga M, Accili D, Tobe K, Ueki K, Nagai 50 R, Kadowaki T: Insulin/Foxo1 pathway regulates expression levels of adiponectin receptors and 51 adiponectin sensitivity. The Journal of biological chemistry 2004;279:3081730822 52 32. Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, Rasbach KA, Bostrom EA, Choi JH, 53 Long JZ, Kajimura S, Zingaretti MC, Vind BF, Tu H, Cinti S, Hojlund K, Gygi SP, Spiegelman BM:

24

Page 25 of 56 Diabetes

1 A PGC1alphadependent myokine that drives brownfatlike development of white fat and 2 thermogenesis. Nature 2012;481:463468 3 33. Bauche IB, El Mkadem SA, Pottier AM, Senou M, Many MC, Rezsohazy R, Penicaud L, Maeda 4 N, Funahashi T, Brichard SM: Overexpression of adiponectin targeted to adipose tissue in transgenic 5 mice: impaired adipocyte differentiation. Endocrinology 2007;148:15391549 6 34. Kajimura S, Seale P, Kubota K, Lunsford E, Frangioni JV, Gygi SP, Spiegelman BM: Initiation of 7 myoblast to brown fat switch by a PRDM16C/EBPbeta transcriptional complex. Nature 8 2009;460:11541158 9 35. Ashwal R, Hemi R, Tirosh A, Gordin R, Yissachar E, CohenDayag A, Rosenberg A, Karasik A, 10 Bluher M, Kanety H: Differential expression of novel adiponectin receptor1 transcripts in skeletal 11 muscle of subjects with normal glucose tolerance and type 2 diabetes. Diabetes 2011;60:936946 12 36. Lustig Y, Barhod E, AshwalFluss R, Gordin R, Shomron N, BaruchUmansky K, Hemi R, 13 Karasik A, Kanety H: RNA binding protein PTB and microRNA221 coregulate AdipoR1 translation 14 and adiponectin signaling. Diabetes 2013; 15 37. Kubota N, Terauchi Y, Yamauchi T, Kubota T, Moroi M, Matsui J, Eto K, Yamashita T, Kamon 16 J, Satoh H, Yano W, Froguel P, Nagai R, Kimura S, Kadowaki T, Noda T: Disruption of adiponectin 17 causes insulin resistance and neointimal formation. The Journal of biological chemistry 18 2002;277:2586325866 19 38. Qi Y, Takahashi N, Hileman SM, Patel HR, Berg AH, Pajvani UB, Scherer PE, Ahima RS: 20 Adiponectin acts in the brain to decrease body weight. Nature medicine 2004;10:524529 21 39. Shklyaev S, Aslanidi G, Tennant M, Prima V, Kohlbrenner E, Kroutov V, CampbellThompson 22 M, Crawford J, Shek EW, Scarpace PJ, Zolotukhin S: Sustained peripheral expression of transgene 23 adiponectin offsets the development of dietinduced obesity in rats. Proceedings of the National 24 Academy of Sciences of the United States of America 2003;100:1421714222 25 40. Tonra JR, Ono M, Liu X, Garcia K, Jackson C, Yancopoulos GD, Wiegand SJ, Wong V: Brain 26 derived neurotrophic factor improves blood glucose control and alleviates fasting hyperglycemia in 27 C57BLKSLepr(db)/lepr(db) mice. Diabetes 1999;48:588594 28 41. Chen D, Liao J, Li N, Zhou C, Liu Q, Wang G, Zhang R, Zhang S, Lin L, Chen K, Xie X, Nan F, 29 Young AA, Wang MW: A nonpeptidic agonist of glucagonlike peptide 1 receptors with efficacy in 30 diabetic db/db mice. Proceedings of the National Academy of Sciences of the United States of 31 America 2007;104:943948 32 42. Sleeman MW, Garcia K, Liu R, Murray JD, Malinova L, Moncrieffe M, Yancopoulos GD, 33 Wiegand SJ: Ciliary neurotrophic factor improves diabetic parameters and hepatic steatosis and 34 increases basal metabolic rate in db/db mice. Proceedings of the National Academy of Sciences of the 35 United States of America 2003;100:1429714302 36 43. Bates SH, Kulkarni RN, Seifert M, Myers MG, Jr.: Roles for leptin receptor/STAT3dependent 37 and independent signals in the regulation of glucose homeostasis. Cell Metab 2005;1:169178 38 44. Davis RC, Castellani LW, Hosseini M, BenZeev O, Mao HZ, Weinstein MM, Jung DY, Jun JY, 39 Kim JK, Lusis AJ, Peterfy M: Early hepatic insulin resistance precedes the onset of diabetes in obese 40 C57BLKSdb/db mice. Diabetes 2010;59:16161625 41 45. Wijesekara N, Krishnamurthy M, Bhattacharjee A, Suhail A, Sweeney G, Wheeler MB: 42 Adiponectininduced ERK and Akt phosphorylation protects against pancreatic beta cell apoptosis 43 and increases insulin and secretion. The Journal of biological chemistry 44 2010;285:3362333631 45 46. Karpe F, Dickmann JR, Frayn KN: Fatty acids, obesity, and insulin resistance: time for a 46 reevaluation. Diabetes 2011;60:24412449 47 47. OkadaIwabu M, Yamauchi T, Iwabu M, Honma T, Hamagami K, Matsuda K, Yamaguchi M, 48 Tanabe H, KimuraSomeya T, Shirouzu M, Ogata H, Tokuyama K, Ueki K, Nagano T, Tanaka A, 49 Yokoyama S, Kadowaki T: A smallmolecule AdipoR agonist for type 2 diabetes and short life in 50 obesity. Nature 2013;503:493499 51 48. Sun Y, Zang Z, Zhong L, Wu M, Su Q, Gao X, Zan W, Lin D, Zhao Y, Zhang Z: Identification of 52 adiponectin receptor agonist utilizing a fluorescence polarization based high throughput assay. PloS 53 one 2013;8:e63354

25

Diabetes Page 26 of 56

1 49. Sharan K, Swarnkar G, Siddiqui JA, Kumar A, Rawat P, Kumar M, Nagar GK, Manickavasagam 2 L, Singh SP, Mishra G, Wahajuddin, Jain GK, Maurya R, Chattopadhyay N: A novel flavonoid, 6C 3 betadglucopyranosyl(2S,3S)(+)3',4',5,7tetrahydroxyflavanone, isolated from Ulmus wallichiana 4 Planchon mitigates ovariectomyinduced osteoporosis in rats. Menopause 2010;17:577586 5 50. Siddiqui JA, Sharan K, Swarnkar G, Rawat P, Kumar M, Manickavasagam L, Maurya R, Pierroz 6 D, Chattopadhyay N: Quercetin6CbetaDglucopyranoside isolated from Ulmus wallichiana 7 planchon is more potent than quercetin in inhibiting osteoclastogenesis and mitigating ovariectomy 8 induced bone loss in rats. Menopause 2011;18:198207 9 51. Siddiqui JA, Swarnkar G, Sharan K, Chakravarti B, Gautam AK, Rawat P, Kumar M, Gupta V, 10 Manickavasagam L, Dwivedi AK, Maurya R, Chattopadhyay N: A naturally occurring rare analog of 11 quercetin promotes peak bone mass achievement and exerts anabolic effect on osteoporotic bone. 12 Osteoporos Int 2011;22:30133027 13 52. Kumar M, Rawat P, Dixit P, Mishra D, Gautam AK, Pandey R, Singh D, Chattopadhyay N, 14 Maurya R: Antiosteoporotic constituents from Indian medicinal plants. Phytomedicine : international 15 journal of phytotherapy and phytopharmacology 2010;17:993999 16 53. Hosseinimehr SJ, Tolmachev V, Stenerlow B: 125Ilabeled quercetin as a novel DNAtargeted 17 radiotracer. Cancer biotherapy & radiopharmaceuticals 2011;26:469475

18

19 20 FIGURE LEGENDS 21 22 Fig. 1. Chemical structures of quercetin and its natural analogs.

23 (A) Quercetin (B) (2R, 3R)dihydroquercetin (C) 6CβDglucopyranosyl(2S, 3S) (+)

24 5,7, 3',4' tetrahydroxydihydroflavonol (GTDF). (D) Quercetin6CβDglucopyranoside

25 (QCG). (E) Quercetin3OβDglucopyranoside (QOG). (F) Quercetin3OαL

26 rhamnopyranoside (QR). GTDF, QCG and DHQ were isolated from the stembark of Ulmus

27 wallichiana Planchon (19; 4951). QR was prepared from stems of Cissus quadrangularis

28 (52). Quercetin and QOG were procured from Sigma Aldrich (St. Louis, MO). Purity of

29 GTDF, DHQ, QCG and QR were > 98%; quercetin was > 95% and QOG was >90%.

30

31 Fig. 2. GTDF is an AdipoR agonist that elicits adiponectin-associated signaling and

32 functional events

33 (A-C) PPARαligand activation assay in HEK293 cells transfected with Gal4UASLuc and

34 Gal4PPARα. 24h after transfection cells were treated as indicated, luciferase activities were

35 normalized with GFP fluorescence obtained from cotransfected eGFPC1 plasmid; used as a

26

Page 27 of 56 Diabetes

1 transfection control. (A) Data is plotted as activity of compounds in presence of PPARα

2 agonist GW7647 at its effective concentration 10 (EC10: 0.001µM) over luciferase activity

3 obtained in presence of GW7647 alone. (B) Comparison of GTDF with gAd (0.1µg/ml). (C)

4 GTDF activity in absence of GW7647. (D) GTDF was immobilized on epoxy agarose beads

5 and GTDF beads or mocktreated control beads were incubated with 50µg plasma membrane

6 extracts from C2C12 myotubes in presence or absence of 100 fold molar excess of GTDF or QCG,

7 following washes the complexes were resolved by SDSPAGE and were analyzed by immunoblotting

8 (see Supplementary Figure 1 for antibody validation data; antiAdipoR1 also detected AdipoR2 in

9 agreement with the supplier information). (E) Competition radioligand binding assay. C2C12

10 myotubes in 12 well plates were incubated with 2µl of 10µCi/ml mouse 125IgAd with or

11 without cold GTDF in PBS supplemented with 0.1% BSA for 12h, following which cells

12 were washed and lysed. A portion of the lysate was used for protein estimation and rest was

13 quantitated in a gammacounter. Nonspecific binding was determined with a 200fold excess

14 of cold gAd. Specific binding was calculated by subtracting nonspecific binding from the

15 total binding. The count per minute (CPM) was normalized with protein concentration and

16 plotted as % binding compared to wells without cold GTDF. (F) 125IGTDF was prepared by

17 chloramine T method as described elsewhere (53) and research design and methods, and

18 purified by thin layer chromatography (TLC). A representative reverse phase TLC result

19 showing blank reaction (upper panel) and 125I GTDF (lower panel) is shown. 125I did not

20 move in the TLC with mobile phase methanol water (40%60%). (G) CHO cells transfected

21 with AdipoR1, AdipoR2 or empty expression plasmids in 12 well plates were incubated with

22 increasing concentrations of 125IGTDF (specific activity 178.2 Ci/ mmol) for 2h at 40C (at 2h

23 the binding equilibrium was established). The cells were then washed and processed as in 2F.

24 (H) Immunoblot analysis in C2C12 myotubes treated with or without GTDF for indicated

25 times. Densitometric analysis of three independent blots using Image J software is given in

27

Diabetes Page 28 of 56

1 the right panel. (I) Quantitative polymerase chain reaction (QPCR)based determination of

2 gene expression in C2C12 myotubeswas performed on a LightCycler 480 (Roche

3 Diagnostics, Mannheim, Germany) using SYBR green chemistry (oligonucleotide sequences

4 in Supplementary Table 5) data were analyzed by CT method using βactin as

5 normalization control. (J) Immunoblotting in C2C12 myotubes treated with GTDF. (K) Co

6 immunoprecipitation followed by immunodetection of acetylated PGC1α. Whole cell

7 extract from C2C12 myotubes were precleared with proteinA sepharose and IgG isotype and

8 were incubated with PGC1α antibody (2µg/reaction) for 3h. Complexes were resolved on

9 8% SDS PAGE and were probed with AcLys antibody. (L) Mitochondrial DNA content

10 determination by QPCR in C2C12 myotubes treated for 48h with GTDF. (M) Glucose uptake

11 in C2C12 myotubes. C2C12 myotubes in 24 well plates were treated with vehicle or GTDF

12 for 24h. Cells were then washed and treated with 0.1µM insulin for 20 min and glucose

13 uptake was quantified as described in research design and methods. (N) C2C12 myotubes

14 were rendered insulinresistant by incubation in 500µM palmitate for 24h (control groups

15 received 4% BSA). GTDF group received 0.01µM GTDF for 12h. Glucose uptake studies

16 were performed as described in figure 2M. (O) Fatty acid oxidation in C2C12 myotubes.

17 C2C12 myotubes in 12 well plates were treated with vehicle (0h) or GTDF for 2h, 24h or

18 120h. Following incubation, fatty acid oxidation was assessed as described in research design

19 and methods. Data are mean ± SEM of 3 (A-C, E, G-I,, K, M-O), or 6 independent

20 experiments (L). All images are representatives of 3 independent experiments. GTDF

21 concentration in all experiments was 0.01µM or as indicated. V; Vehicle. *p<0.05, **p<0.01,

22 ***p<0.001.

23

24 Fig. 3. Overexpression or knockdown of AdipoR1 enhances or mitigates GTDF action

25 respectively 28

Page 29 of 56 Diabetes

1 (A) Immunoblot analysis of C2C12 myotubes transfected with vector or AdipoRs. (B)

2 Immunoblot analysis of C2C12 myotubes transfected with luciferase siRNA (siC) or siRNAs

3 against AdipoR1. (C) Glucose uptake in C2C12 myotubes transfected with indicated siRNAs.

4 Data are mean ± SEM of 4 independent experiments. (D) Fatty acid oxidation in C2C12

5 myotubes transfected with indicated siRNAs. Data are mean ± SEM of 3 independent

6 experiments. In all graphs Images are representative of 3 independent experiments. GTDF

7 concentration in all experiments was 0.01µM. Note: antiAdipoR1 also detects AdipoR2.

8 *p<0.05, **p<0.01, ***p<0.001.

9

10 Fig. 4. Comparison of mRNA and protein expression of AdipoRs and evaluation of

11 adiponectin sensitivity in BL6, db/db or BKS- db/db mice.

12 12week old male mice (n=3/group) were assessed for (A) AdipoR mRNA expression and

13 (B) total or PMassociated AdipoR protein levels in skeletal muscle (quadriceps), liver or

14 eWAT. Bar graphs represent densitometric analysis. βactin and Ncadherin were used as

15 loading controls. Note: antiAdipoR1 also detects AdipoR2.(C). EDL muscles excised from

16 12 week old BL6, db/db or BKSdb/db mice were treated with 2.5 g/ml gAd or vehicle for

17 30 min as described in research designs and methods. Total protein was then isolated and

18 analyzed for pAMPK and AMPK expression. *p<0.05, **p<0.01, ***p<0.001 in comparison

19 to BL6. ##p<0.01compared to db/db.

20

21 Fig. 5. GTDF but not pioglitazone fails to improve metabolic health in BKS-db/db

22 12 week old male db/db mice (n=6/group) were fed vehicle, 10mg/kg GTDF or Pio for 30

23 days. (A) Feed intake. (B) Body weight. (C) Nonfasting blood glucose was analyzed using a

24 glucometer capable of measuring 20900 mg/dl glucose in whole blood (Gluco Dr.

25 Supersensor (Gyeonggido, South Korea). (D) IPGTT; Glucose was intraperitoneally 29

Diabetes Page 30 of 56

1 administered in mice (1g/kg body weight) and at indicated time points blood was collected

2 from tail snips and blood glucose was measured by a glucometer. Inset represents area under

3 the curve (AUC). (E) Glycated haemoglobin level; vehicle vs GTDF 9.9±0.4% vs 9.3 ± 0.3%

4 (84.33 ± 4.29 vs 78.17±2.96 fmol/mol) (F) Fasting insulin was estimated by ELISA (G)

5 Fasting serum lipid profile.. *p<0.05, **p<0.01, ***p<0.001.

6

7 Fig. 6. GTDF improves metabolic parameters in db/db mice

8 12 week old db/db or BL6 mice were fed vehicle or 10mg/kg GTDF for 30 days; n=8/group.

9 (A) Feed intake. (B) Body weight. (C) Nonfasting blood glucose. (D) Fasting blood glucose.

10 (E) PTT; Mice were intraperitoneally administered sodium pyruvate (1g/kg body weight) and

11 blood collected from tail snips were analyzed by a glucometer. Inset represents AUC. (F)

12 Glycated haemoglobin level; graph represents mean ± SEM. HbA1c was comparable between

13 vehicle and GTDF group on 0d; Mean±SD; 8.7±1.03% vs 8.72 ±1.17% (72.0±11.1 vs

14 72±12.8fmol/mol) but was significantly lowered in GTDFtreated group after 30d V vs

15 GTDF; 10.53±0.54% vs 9±0.93% (91±5.9 vs 75±10.2 fmol/mol) (G) IPGTT, inset represents

16 AUC. (H) Insulin level during IPGTT; Blood collected from tail snips at indicated time

17 points during GTT was used for insulin measurement by ELISA. Inset, represents AUC. (I)

18 Fasting insulin. (J) Serum pancreatic Cpeptide level (K) Serum Na+ and K+ levels. (L)

19 Microscopic images from immunostaining of pancreas sections by antiinsulin antibody; top

20 two panels; diaminobenzidine (DAB) staining (light microscopy; Leica DMI6000 B), third

21 and fourth panels from above; confocal microscopy (Carl Zeiss LSM 510 Meta).

22 Immunofluorescence for insulin (green), DAPI (blue) and TUNEL (red), or insulin, DAPI

23 and Ki67 (red); by confocal microscopy are shown. (M) Histomorphometry of pancreatic

24 islets was performed using Image –Pro plus (6.1) software (MediaCybernetics), where two

25 investigators separately evaluated 10 random sections probed with insulin antibody from 4 30

Page 31 of 56 Diabetes

1 mice per group, blinded to experimental design. (N) Fasting serum levels of circulating

2 glucagon, leptin, adiponectin, ghrelin, corticosterone and markers of inflammation (MCP1,

3 TNFα, and IL6) as determined by ELISA. (O) Microscopic images from hematoxylin and

4 eosin (H&E) stained or oil redO stained liver sections. Top panel magnification 10X, bar

5 200µm. Middle and bottom panel magnification 40X, bar 50 µm. (P) Hepatic glycogen

6 content. n=8 for all experiments unless otherwise indicated. *p<0.05, **p<0.01, ***p<0.001.

7

8 Fig. 7. GTDF induces browning in adipocytes

9 (A) Immunodetection of FNDC5 in C2C12 myotubes or culture medium (CM) of C2C12

10 myotubes treated with GTDF. (B) H&E staining of eWAT of db/db mice used in Fig. 6. (C)

11 Immunofluorescent staining of the same eWAT by antiUCP1 antibody, visualized by

12 confocal microscopy. Bottom panel shows images in bright field. (B and C) Each image is

13 representative of 10 random sections from 3 mice per group. (D) Oil redO staining of 3T3L

14 1 adipocytes differentiated with or without GTDF, where GTDF was added on day 0 or day 2

15 of differentiation (10d or 8d respectively). (E) Extracted oil redO from stained cells

16 quantified by spectrophotometry at 490nm. Data are mean ± SEM of 3 independent

17 experiments. (F and G) QPCR analysis of gene expression in 3T3L1 (F) or mouse SVFs (G)

18 differentiated with or without GTDF. (H) 3T3L1, mouse SVF or human SVF were

19 differentiated with or without GTDF using standard protocol and as indicated in 7D, and

20 analyzed by immunoblotting. (I) Mitochondrial DNA content determination by QPCR in

21 mouse SVF differentiated in presence of GTDF. (J) GTDF does not modulate the activity of

22 βadrenergic receptors. HEK293 cells in 24 well plates were transfected with 200 ng cyclic

23 AMP response elementdriven luciferase reporter (CRELuc), 200 ng mammalian expression

24 plasmids for βADR 1, 2 or 3 and 100 ng eGFPC1. 24 h post transfection, cells were

25 treated for 6 h with indicated ligands, lysed and GFP and luciferase activity were 31

Diabetes Page 32 of 56

1 determined using a microplate fluorimeter (Fluostar Omega; BMG Labtech) and a microplate

2 luminometer (GloMax; Promega). The luciferase values were normalized with GFP and

3 plotted as fold luciferase activity over vehicletreated controls. Data are mean ± SEM of 3

4 independent experiments in all graphs. Images are representative of 3 independent

5 experiments. GM; Growth medium (DMEM with 4.5mg/ml glucose, 4.0mM , 1

6 mM sodium pyruvate, 10% FBS and 1X antibioticantimycotic solution) DM; Differentiation

7 medium (GM supplemented with 1.5µg/ml insulin, 0.5mM IBMX, 1.0µM dexamethasone

8 and 1µM rosiglitazone), INS; Insulin medium GM plus 1.5µg/ml insulin). For human SVFs

9 after DM and INS medium cells were cultured for 21d, at which matured adipocyte

10 phenotypes were observed. Cells were given fresh medium containing vehicle or 0.01µM

11 GTDF every day during the differentiation. *p<0.05, **p<0.01, ***p<0.005.

12 Supplementary information includes supplementary Figures 13 and supplementary tables

13 15.

32

Page 33 of 56 Diabetes

Chemical structures of quercetin and its natural analogs.

Diabetes Page 34 of 56

GTDF is an AdipoR agonist that elicits adiponectin-associated signaling and functional events

Page 35 of 56 Diabetes

Overexpression or knockdown of AdipoR1 enhances or mitigates GTDF action respectively

Diabetes Page 36 of 56

Comparison of mRNA and protein expression of AdipoRs and evaluation of adiponectin sensitivity in BL6, db/db or BKS- db/db mice.

Page 37 of 56 Diabetes

GTDF but not pioglitazone fails to improve metabolic health in BKS-db/db 218x266mm (600 x 600 DPI)

Diabetes Page 38 of 56

GTDF improves metabolic parameters in db/db mice 193x208mm (600 x 600 DPI)

Page 39 of 56 Diabetes

GTDF induces browning in adipocytes

Diabetes Page 40 of 56

SUPPLEMENTARY DATA

Supplementary Figure 1. Validation of AdipoR1 and R2 antibodies. 50 extracts from CHO cells transfected with empty plasmid or AdipoR1 and R2 mammalian expression plasmids or C2C12 cells (that express endogenous AdipoRs) were analyzed by immunoblotting with indicated antibodies. AdipoR1 antibody showed some crossreactivity with overexpressed AdipoR2, which was expected as this antibody was raised against a peptide from the Cterminus of AdipoR1 that shares amino acid similarity with AdipoR2. Image representative of 3 independent experiments.

Page 41 of 56 Diabetes

Supplementary Figure 2. Saturation binding of 125IgAd to CHO cells transfected with AdipoR1, AdipoR2 or vector only. CHO cells transfected with AdipoR1, R2 or empty expression vector were incubated with increasing concentrations of 125IgAd in PBS supplemented with 0.1% BSA for 2h, following which cells were washed and lysed. A portion of the lysate was used for protein estimation and rest was quantitated in a gamma counter. Nonspecific binding for each concentration was determined using 200fold excess of cold gAd. Specific binding was calculated by subtracting nonspecific binding from the total binding. The count per minute (CPM) was normalized with protein concentration and plotted.

Diabetes Page 42 of 56

Supplementary Figure 3. GTDF lowers adipose tissue weight in db/db mice. Indicated fat pads from 6 mice in each group were dissected and weighted using an analytical balance (Mettler, Toledo, Osaka, Japan).

Page 43 of 56 Diabetes

Supplementary Table 1: GTDF does not bind to ionotropic glutamate receptors. Wistar rat cerebral cortex (for AMPA receptor, and NMDA receptor agonist, , and polyamine sites), Wistar rat whole brain (minus cerebellum, for ) or whole Wistar rat brain (for nonselective glutamate binding) were incubated with indicated radioligands in absence or presence of indicated doses of GTDF. % inhibition of radioligand binding was calculated using standard formula. Threshold inhibition was set at 50% for consideration of inhibition.

GTDF Receptor, % Radi Specific Kd of Nonspec Incubat Incubati Concentra assay inhibition oliga binding radiolig ific ion on time, tion (M) of specific nd activity and competit buffer temperat radioligand of or ure binding radiolig and

AMPA 1.0 mM 50 mM L Tris 5.0 Glutami HCl, nM 90% KD1= c acid pH 7.4, 10 3 [3H] 200 AMP 0.018 mM A M KSCN 1 15 KD2 = 0.1 7 0.99 90min, M 4°C Kainate 5.0 1.0 mM 50 mM 60 nM 80% KD = L Tris minutes, 10 7 [3H] Glutami HCl, 4°C AMP 0.012 c acid pH 7.4 A M 1 2 0.1 1 NMDA, 2.0 1.0 mM 50 mM 20 Agonism nM L Tris minutes, [3H] 70% KD = Glutami HCl, 4°C 10 3 CGP c acid pH 7.4 3965 0.019 3 M 1 7 0.1 6 NMDA, 0.33 50 mM 30 Glycine nM HEPES minutes, [3H] 85% KD = 10.0 M , pH 4°C 10 7 7.7 MDL 105,5 0.006 MDL 19 M 105,519 Diabetes Page 44 of 56

1 3 0.1 3

NMDA, 1.0 M 45 Phencycli Dizocilp minutes, dine 4.0 ine (+) 25°C nM MK [3H] 94% KD = 801) 10 2 0.0084 TCP M 10 mM Tris 1 5 HCl, 0.1 2 pH 7.4 NMDA, 2.0 50 mM 2 hours, Polyamin nM Tris 4°C e [3H] 80% KD = HCl, 10 4 pH 7.4 Ifenp 0.026 rodil M 10.0 M 1 8 Ifenprod 0.1 3 il Glutamate 50 mM 30 , Non 3.75 Tris minutes, selective nM HCl, 37°C [3H] 90% KD = pH 7.4, 10 17 L 2.5 mM Gluta CaCl2 mic 0.29 acid M 50.0 M L 1 13 Glutami 0.1 14 c acid

Page 45 of 56 Diabetes

Supplementary Table 2. GTDF does not activate or repress metabotropic glutamate receptors at its active dose but quercetin activates mGlu2, and suppresses mGlu4, 6 and 8 activity. Modulation of metabotropic activities by GTDF or quercetin at their pharmacologically active concentrations was assessed. mGlu1, 3, 5 activities were assessed by an aequorin assay, mGlu2 and 7 activities were assessed by cAMP assay, mGlu4, 6 and 8 were assessed by GTPγS binding. PAM; positive allosteric modulator. Threshold activation or repression was set at 50%. % activation or repression beyond the threshold is in red.

Assay mode Conc Co Compound (M) Agonist Antagonist PAM Addition % % % % activation inhibition activation activation Receptor: mGlu1 GTDF 1 0.15 8.98 0.951 2.09 0.1 0.35 1.6 0.791 0.87 Quercetin 1000 0.049 16.41 2.02 3.38 100 0.37 8.911 6.97 23.58 Receptor: mGlu2 GTDF 1 10.64 19.62 17.95 0.1 7.76 9.56 8.59 Quercetin 1000 2.17 18.32 12.65 100 117.85* 7.6 284.27* Receptor: mGlu3 GTDF 1 2.38 5.06 3.55 6.39 0.1 0.78 12.26 3.52 3.94 Quercetin 1000 0.36 6.58 18.96 0.061 100 4.75 11.32 5.068 24.67 Receptor: mGlu4 GTDF 1 5.04 4.44 4.14 0.1 1.63 6.16 5.87 Quercetin 1000 23.15 86.97 33.79 100 7.75 31.65 14.11 Receptor: mGlu5 GTDF 1 0.4 1.15 3.35 2.61 0.1 0.04 3.35 5.14 3.69 Quercetin 1000 0.11 14.66 0.74 0.64 100 0.07 30.92 8.72 3.55 Diabetes Page 46 of 56

Receptor: mGlu6 GTDF 1 2.24 0.04 15.39 0.1 6.65 4.91 4.82 Quercetin 1000 136.684 201.61 141.74 100 20.7469 58.63 39.7 Receptor: mGlu7 GTDF 1 16.46 11.21 8.68 0.1 19.05 0.22 0.836 Quercetin 1000 18.96 13.68 10.19 100 23.22 4.71 8.94 Receptor: mGlu8 GTDF 1 1.15 15.33 10.26 0.1 4.24 1.28 4.6 Quercetin 1000 72.89 159.53 64.26 100 14.3 58.17 10.91

Page 47 of 56 Diabetes

Supplementary Table 3. GTDF is not a GPCR agonist. GTDF at its pharmacologically active dose (0.1M) or high dose (10M) was assessed for its ability to modulate GPCR signalling using pathhunter βarrestin assay for the indicated GPCRs. Threshold for consideration as an activator was set at 50%.

GTDF GTDF Assay % % GPCR ID Conc Conc Mode Activity Activity (M) (M) ADCYAP1R1 Agonist 0.1 2% 10 2% ADORA3 Agonist 0.1 7% 10 9% ADRA1B Agonist 0.1 1% 10 2% ADRA2A Agonist 0.1 3% 10 1% ADRA2B Agonist 0.1 5% 10 3% ADRA2C Agonist 0.1 2% 10 1% ADRB1 Agonist 0.1 1% 10 1% ADRB2 Agonist 0.1 0% 10 1% AGTR1 Agonist 0.1 3% 10 2% AGTRL1 Agonist 0.1 1% 10 2% AVPR1A Agonist 0.1 5% 10 4% AVPR1B Agonist 0.1 2% 10 1% AVPR2 Agonist 0.1 2% 10 4% BDKRB1 Agonist 0.1 5% 10 0% BDKRB2 Agonist 0.1 3% 10 3% BRS3 Agonist 0.1 6% 10 2% C5AR1 Agonist 0.1 2% 10 2% C5LR2 Agonist 0.1 1% 10 14% CALCR Agonist 0.1 1% 10 11% CALCR + RAMP2 Agonist 0.1 7% 10 4% CALCR + RAMP3 Agonist 0.1 6% 10 25% CALCRL + RAMP1 Agonist 0.1 4% 10 1% CALCRL + RAMP2 Agonist 0.1 4% 10 1% CALCRL + RAMP3 Agonist 0.1 3% 10 1% CCKAR Agonist 0.1 2% 10 1% CCKBR Agonist 0.1 5% 10 4% CCR10 Agonist 0.1 3% 10 3% CCR2 Agonist 0.1 2% 10 2% CCR3 Agonist 0.1 5% 10 4% CCR4 Agonist 0.1 5% 10 1% CCR5 Agonist 0.1 4% 10 1% CCR6 Agonist 0.1 2% 10 3% CCR7 Agonist 0.1 8% 10 5% Diabetes Page 48 of 56

CCR8 Agonist 0.1 0% 10 3% CCR9 Agonist 0.1 5% 10 5% CHRM1 Agonist 0.1 4% 10 5% CHRM2 Agonist 0.1 3% 10 1% CHRM3 Agonist 0.1 5% 10 2% CHRM4 Agonist 0.1 6% 10 4% CHRM5 Agonist 0.1 7% 10 29% CMKLR1 Agonist 0.1 2% 10 4% CMKOR1 Agonist 0.1 8% 10 2% CNR1 Agonist 0.1 0% 10 2% CNR2 Agonist 0.1 0% 10 4% CRHR1 Agonist 0.1 2% 10 1% CRHR2 Agonist 0.1 3% 10 1% CRTH2 Agonist 0.1 3% 10 5% CX3CR1 Agonist 0.1 2% 10 0% CXCR1 Agonist 0.1 10% 10 1% CXCR2 Agonist 0.1 2% 10 2% CXCR3 Agonist 0.1 3% 10 3% CXCR4 Agonist 0.1 7% 10 9% CXCR5 Agonist 0.1 8% 10 6% CXCR6 Agonist 0.1 2% 10 3% DRD1 Agonist 0.1 6% 10 3% DRD2L Agonist 0.1 4% 10 2% DRD2S Agonist 0.1 5% 10 3% DRD3 Agonist 0.1 7% 10 14% DRD4 Agonist 0.1 2% 10 2% DRD5 Agonist 0.1 4% 10 2% EDG1 Agonist 0.1 3% 10 5% EDG2 Agonist 0.1 1% 10 2% EDG3 Agonist 0.1 4% 10 5% EDG4 Agonist 0.1 2% 10 1% EDG5 Agonist 0.1 2% 10 7% EDG6 Agonist 0.1 2% 10 12% EDG7 Agonist 0.1 4% 10 2% EDG8 Agonist 0.1 0% 10 1% EDNRA Agonist 0.1 7% 10 3% EDNRB Agonist 0.1 3% 10 3% F2R Agonist 0.1 9% 10 2% F2RL1 Agonist 0.1 1% 10 1% F2RL3 Agonist 0.1 3% 10 1% FPR1 Agonist 0.1 10% 10 4% FPRL1 Agonist 0.1 2% 10 1% Page 49 of 56 Diabetes

FSHR Agonist 0.1 6% 10 11% GALR1 Agonist 0.1 4% 10 1% GALR2 Agonist 0.1 5% 10 6% GCGR Agonist 0.1 3% 10 1% GHSR1A Agonist 0.1 0% 10 2% GIPR Agonist 0.1 3% 10 13% GLP1R Agonist 0.1 1% 10 1% GLP2R Agonist 0.1 1% 10 2% GPR1 Agonist 0.1 4% 10 3% GPR109A Agonist 0.1 11% 10 4% GPR119 Agonist 0.1 4% 10 4% GPR120 Agonist 0.1 1% 10 7% GPR35 Agonist 0.1 4% 10 3% GPR92 Agonist 0.1 7% 10 6% GRPR Agonist 0.1 1% 10 2% HCRTR1 Agonist 0.1 3% 10 1% HCRTR2 Agonist 0.1 3% 10 1% HRH1 Agonist 0.1 0% 10 1% HRH2 Agonist 0.1 2% 10 1% HRH3 Agonist 0.1 14% 10 6% HTR1A Agonist 0.1 2% 10 2% HTR1B Agonist 0.1 4% 10 4% HTR1E Agonist 0.1 7% 10 1% HTR1F Agonist 0.1 3% 10 2% HTR2A Agonist 0.1 3% 10 11% HTR2C Agonist 0.1 3% 10 1% HTR5A Agonist 0.1 1% 10 13% KISS1R Agonist 0.1 3% 10 4% LHCGR Agonist 0.1 1% 10 8% LTB4R Agonist 0.1 3% 10 3% MC1R Agonist 0.1 1% 10 1% MC3R Agonist 0.1 7% 10 10% MC4R Agonist 0.1 4% 10 12% MC5R Agonist 0.1 8% 10 5% MCHR1 Agonist 0.1 1% 10 5% MCHR2 Agonist 0.1 3% 10 2% MLNR Agonist 0.1 3% 10 4% MRGPRX2 Agonist 0.1 6% 10 2% MTNR1A Agonist 0.1 0% 10 4% MTNR1B Agonist 0.1 9% 10 3% NMU1R Agonist 0.1 5% 10 12% NPBWR1 Agonist 0.1 2% 10 6% Diabetes Page 50 of 56

NPBWR2 Agonist 0.1 4% 10 3% NPFFR1 Agonist 0.1 1% 10 2% NPSR1 Agonist 0.1 2% 10 4% NPY1R Agonist 0.1 4% 10 3% NPY2R Agonist 0.1 2% 10 1% NTSR1 Agonist 0.1 7% 10 9% OPRD1 Agonist 0.1 1% 10 3% OPRK1 Agonist 0.1 0% 10 5% OPRL1 Agonist 0.1 0% 10 14% OPRM1 Agonist 0.1 5% 10 5% OXTR Agonist 0.1 3% 10 3% P2RY11 Agonist 0.1 3% 10 2% P2RY12 Agonist 0.1 4% 10 5% P2RY2 Agonist 0.1 2% 10 3% P2RY4 Agonist 0.1 13% 10 10% P2RY6 Agonist 0.1 1% 10 0% PPYR1 (NPY4) Agonist 0.1 0% 10 2% PRLHR Agonist 0.1 7% 10 3% PROKR1 Agonist 0.1 7% 10 1% PROKR2 Agonist 0.1 3% 10 1% PTAFR Agonist 0.1 16% 10 3% PTGER2 Agonist 0.1 0% 10 4% PTGER3 Agonist 0.1 3% 10 3% PTGER4 Agonist 0.1 15% 10 9% PTGIR Agonist 0.1 6% 10 7% PTHR1 Agonist 0.1 3% 10 1% PTHR2 Agonist 0.1 3% 10 1% RXFP3 Agonist 0.1 2% 10 4% SCTR Agonist 0.1 3% 10 1% SSTR2 Agonist 0.1 2% 10 1% SSTR3 Agonist 0.1 0% 10 4% SSTR5 Agonist 0.1 0% 10 6% TACR1 Agonist 0.1 13% 10 20% TACR2 Agonist 0.1 11% 10 4% TACR3 Agonist 0.1 2% 10 1% TBXA2R Agonist 0.1 0% 10 3% TRHR Agonist 0.1 2% 10 2% TSHR Agonist 0.1 1% 10 2% UTR2 Agonist 0.1 1% 10 4% VIPR1 Agonist 0.1 2% 10 2% VIPR2 Agonist 0.1 1% 10 0%

Page 51 of 56 Diabetes

Supplementary Table 4. GTDF is not a GPCR antagonist or inhibitor. GTDF at its pharmacologically active dose (0.1M) or high dose (10M) was assessed for its ability to antagonize or inhibit GPCR signaling in presence of EC80 concentrations of cognate agonists of the indicated GPCRs, using pathhunter βarrestin assay. Threshold inhibition for consideration as an antagonist or inhibitor was set at 50%.

GTDF % GTDF % Assay GPCR ID Conc Inhibiti Conc Inhibiti Mode (M) on (M) on ADCYAP1R1 Antagonist 0.1 3% 10 11% ADORA3 Antagonist 0.1 3% 10 6% ADRA1B Antagonist 0.1 3% 10 12% ADRA2A Antagonist 0.1 13% 10 12% ADRA2B Antagonist 0.1 1% 10 5% ADRA2C Antagonist 0.1 2% 10 5% ADRB1 Antagonist 0.1 13% 10 4% ADRB2 Antagonist 0.1 7% 10 10% AGTR1 Antagonist 0.1 14% 10 4% AGTRL1 Antagonist 0.1 20% 10 5% AVPR1A Antagonist 0.1 14% 10 13% AVPR1B Antagonist 0.1 15% 10 14% AVPR2 Antagonist 0.1 4% 10 1% BDKRB1 Antagonist 0.1 0% 10 3% BDKRB2 Antagonist 0.1 3% 10 2% BRS3 Antagonist 0.1 11% 10 4% C5AR1 Antagonist 0.1 3% 10 8% C5LR2 Antagonist 0.1 2% 10 12% CALCR Antagonist 0.1 4% 10 21% CALCR + RAMP2 Antagonist 0.1 10% 10 3% CALCR + RAMP3 Antagonist 0.1 5% 10 20% CALCRL + RAMP1 Antagonist 0.1 8% 10 1% CALCRL + RAMP2 Antagonist 0.1 6% 10 1% CALCRL + RAMP3 Antagonist 0.1 16% 10 0% CCKAR Antagonist 0.1 1% 10 11% CCKBR Antagonist 0.1 5% 10 13% CCR10 Antagonist 0.1 27% 10 6% CCR2 Antagonist 0.1 2% 10 4% CCR3 Antagonist 0.1 13% 10 1% Diabetes Page 52 of 56

CCR4 Antagonist 0.1 11% 10 1% CCR5 Antagonist 0.1 14% 10 1% CCR6 Antagonist 0.1 13% 10 6% CCR7 Antagonist 0.1 18% 10 7% CCR8 Antagonist 0.1 2% 10 25% CCR9 Antagonist 0.1 8% 10 17% CHRM1 Antagonist 0.1 18% 10 10% CHRM2 Antagonist 0.1 0% 10 8% CHRM3 Antagonist 0.1 1% 10 15% CHRM4 Antagonist 0.1 8% 10 28% CHRM5 Antagonist 0.1 7% 10 20% CMKLR1 Antagonist 0.1 0% 10 2% CMKOR1 Antagonist 0.1 1% 10 1% CNR1 Antagonist 0.1 1% 10 0% CNR2 Antagonist 0.1 2% 10 14% CRHR1 Antagonist 0.1 9% 10 0% CRHR2 Antagonist 0.1 7% 10 10% CRTH2 Antagonist 0.1 14% 10 13% CX3CR1 Antagonist 0.1 9% 10 4% CXCR1 Antagonist 0.1 9% 10 8% CXCR2 Antagonist 0.1 8% 10 1% CXCR3 Antagonist 0.1 14% 10 8% CXCR4 Antagonist 0.1 3% 10 13% CXCR5 Antagonist 0.1 16% 10 17% CXCR6 Antagonist 0.1 5% 10 6% DRD1 Antagonist 0.1 5% 10 21% DRD2L Antagonist 0.1 6% 10 3% DRD2S Antagonist 0.1 12% 10 12% DRD3 Antagonist 0.1 14% 10 0% DRD4 Antagonist 0.1 1% 10 1% DRD5 Antagonist 0.1 12% 10 11% EDG1 Antagonist 0.1 1% 10 17% EDG2 Antagonist 0.1 6% 10 8% EDG3 Antagonist 0.1 0% 10 6% EDG4 Antagonist 0.1 2% 10 10% EDG5 Antagonist 0.1 13% 10 4% EDG6 Antagonist 0.1 8% 10 11% EDG7 Antagonist 0.1 0% 10 1% EDG8 Antagonist 0.1 11% 10 28% EDNRA Antagonist 0.1 1% 10 16% EDNRB Antagonist 0.1 13% 10 10% F2R Antagonist 0.1 9% 10 3% Page 53 of 56 Diabetes

F2RL1 Antagonist 0.1 12% 10 0% F2RL3 Antagonist 0.1 3% 10 2% FPR1 Antagonist 0.1 5% 10 9% FPRL1 Antagonist 0.1 5% 10 4% FSHR Antagonist 0.1 7% 10 37% GALR1 Antagonist 0.1 11% 10 4% GALR2 Antagonist 0.1 19% 10 10% GCGR Antagonist 0.1 3% 10 14% GHSR1A Antagonist 0.1 3% 10 1% GIPR Antagonist 0.1 9% 10 23% GLP1R Antagonist 0.1 2% 10 1% GLP2R Antagonist 0.1 2% 10 2% GPR1 Antagonist 0.1 14% 10 8% GPR109A Antagonist 0.1 15% 10 9% GPR119 Antagonist 0.1 10% 10 25% GPR120 Antagonist 0.1 27% 10 3% GPR35 Antagonist 0.1 6% 10 9% GPR92 Antagonist 0.1 9% 10 10% GRPR Antagonist 0.1 19% 10 9% HCRTR1 Antagonist 0.1 9% 10 7% HCRTR2 Antagonist 0.1 15% 10 9% HRH1 Antagonist 0.1 7% 10 10% HRH2 Antagonist 0.1 6% 10 0% HRH3 Antagonist 0.1 1% 10 24% HTR1A Antagonist 0.1 3% 10 23% HTR1B Antagonist 0.1 1% 10 11% HTR1E Antagonist 0.1 0% 10 11% HTR1F Antagonist 0.1 1% 10 12% HTR2A Antagonist 0.1 3% 10 15% HTR2C Antagonist 0.1 12% 10 15% HTR5A Antagonist 0.1 19% 10 31% KISS1R Antagonist 0.1 7% 10 14% LHCGR Antagonist 0.1 8% 10 8% LTB4R Antagonist 0.1 6% 10 2% MC1R Antagonist 0.1 16% 10 12% MC3R Antagonist 0.1 3% 10 4% MC4R Antagonist 0.1 17% 10 20% MC5R Antagonist 0.1 9% 10 14% MCHR1 Antagonist 0.1 11% 10 20% MCHR2 Antagonist 0.1 3% 10 21% MLNR Antagonist 0.1 4% 10 6% MRGPRX2 Antagonist 0.1 1% 10 18% Diabetes Page 54 of 56

MTNR1A Antagonist 0.1 3% 10 25% MTNR1B Antagonist 0.1 25% 10 13% NMU1R Antagonist 0.1 21% 10 7% NPBWR1 Antagonist 0.1 9% 10 14% NPBWR2 Antagonist 0.1 8% 10 2% NPFFR1 Antagonist 0.1 5% 10 2% NPSR1 Antagonist 0.1 4% 10 17% NPY1R Antagonist 0.1 7% 10 16% NPY2R Antagonist 0.1 8% 10 4% NTSR1 Antagonist 0.1 5% 10 1% OPRD1 Antagonist 0.1 15% 10 9% OPRK1 Antagonist 0.1 0% 10 26% OPRL1 Antagonist 0.1 4% 10 34% OPRM1 Antagonist 0.1 4% 10 16% OXTR Antagonist 0.1 22% 10 8% P2RY11 Antagonist 0.1 11% 10 10% P2RY12 Antagonist 0.1 5% 10 7% P2RY2 Antagonist 0.1 2% 10 5% P2RY4 Antagonist 0.1 12% 10 10% P2RY6 Antagonist 0.1 0% 10 3% PPYR1 (NPY4) Antagonist 0.1 14% 10 15% PRLHR Antagonist 0.1 4% 10 6% PROKR1 Antagonist 0.1 8% 10 14% PROKR2 Antagonist 0.1 10% 10 4% PTAFR Antagonist 0.1 10% 10 16% PTGER2 Antagonist 0.1 2% 10 12% PTGER3 Antagonist 0.1 19% 10 6% PTGER4 Antagonist 0.1 22% 10 7% PTGIR Antagonist 0.1 3% 10 16% PTHR1 Antagonist 0.1 15% 10 0% PTHR2 Antagonist 0.1 5% 10 7% RXFP3 Antagonist 0.1 15% 10 21% SCTR Antagonist 0.1 13% 10 4% SSTR2 Antagonist 0.1 10% 10 7% SSTR3 Antagonist 0.1 16% 10 5% SSTR5 Antagonist 0.1 0% 10 2% TACR1 Antagonist 0.1 3% 10 10% TACR2 Antagonist 0.1 9% 10 12% TACR3 Antagonist 0.1 7% 10 3% TBXA2R Antagonist 0.1 22% 10 12% TRHR Antagonist 0.1 10% 10 11% TSHR Antagonist 0.1 19% 10 2% Page 55 of 56 Diabetes

UTR2 Antagonist 0.1 5% 10 13% VIPR1 Antagonist 0.1 3% 10 2% VIPR2 Antagonist 0.1 3% 10 1%

Diabetes Page 56 of 56

Supplementary Table 5. Oligonucleotide sequences. m; mouse. All sequences in 5' to 3' orientation.

Gene name Forward Reverse

mUCP1 ACAGAAGGATTGCCGAAAC AGCTGATTTGCCTCTGAATG mUCP2 GTTCCTCTGTCTCGTCTTGC GGCCTTGAAACCAACCA mUCP3 TGACCTGCGCCCAGC CCCAGGCGTATCATGGCT mPPARα TCTTCACGATGCTGTCCTCCT GGAACTCGCCTGTGATAAAGC mPPARβ TCCAGAAGAAGAACCGCAACA GGATAGCGTTGTGCGACATG mPPARγ CAGGCCGAGAAGGAGAAGCT GGCTCGCAGATCAGCAGACT mPGC1α AGCCGTGACCACTGACAACGAG CTGCATGGTTCTGAGTGCTAAG mCPT1B CTCCTTTCCTGGCTGAGGT GATCTGGAACTGGGGGATCT mLPL CCAATGGAGGCACTTTCCA TGGTCCACGTCTCCGAGTC mAdiponectin TGTTGGAATGACAGGAGCTGA CACACTGAACGCTGAGCGATAC mLeptin TCTCCGAGACCTCCTCCATCT TTCCAGGACGCCATCCAG mCD36 GGCCAAGCTATTGCGACAT CAGATCCGAACACAGCGTAGA mFABP3 CCCCTCAGCTCAGCACCAT CAGAAAAATCCCAACCCAAGAAT mFAS CGGAAACTTCAGGAAATGTCC TCAGAGACGTGTCACTCCTGG mFACS TCTAGGAGTGAAGGCCAACG GCAATATCTGAGGGCAGTGG mACOX1 GCCCAACTGTGACTTCCATC GCCAGGACTATCGCATGATT mPRDM16 ACAGGCAGGCTAAGAACCAG CGTGGAGAGGAGTGTCTTCAG mCIDEA AAACCATGACCGAAGTAGCC AGGCCAGTTGTGATGACTAAGAC mβactin CCTCACCCTCCCAAAAGC GTGGACTCAGGGCATGGA

Mitochondrial DNA MTcoxII GCCGACTAAATCAAGCAACA CAATGGGCATAAAGCTATGG CytB CATTTATTATCGCGGCCCTA TGTTGGGTTGTTTGATCCTG βGlobin GAAGCAATTCTAGGGAGCAG GGAGCAGCGATTCTGAGTAGA