Insulin regulates lipolysis and fat mass by upregulating growth/differentiation factor ... · 2019....

49
1 Insulin regulates lipolysis and fat mass by upregulating growth/differentiation factor 3 in adipose tissue macrophages Yun Bu 1 , Katsuhide Okunishi 1 , Satomi Yogosawa 1 , Kouichi Mizuno 1 , Chester W. Brown 2 , and Tetsuro Izumi 1,3 1 Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan; 2 Division of Genetics, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163; and 3 Research Program for Signal Transduction, Division of Endocrinology, Metabolism and Signal Research, Gunma University Initiative for Advanced Research, Gunma University, Maebashi 371- 8512, Japan. A short running title: Roles of the insulin-GDF3-ALK7 axis in obesity Address correspondence to: Tetsuro Izumi, Phone: +81-27-220-8856; E-mail: [email protected]. Or to Katsuhide Okunishi, Phone: +81-27-220-8877; E-mail: [email protected]. The word count: 4077-Endnote92=3985>4000 words (Introduction to Discussion) The number of figures: 6

Transcript of Insulin regulates lipolysis and fat mass by upregulating growth/differentiation factor ... · 2019....

  • 1

    Insulin regulates lipolysis and fat mass by upregulating growth/differentiation

    factor 3 in adipose tissue macrophages

    Yun Bu1, Katsuhide Okunishi1, Satomi Yogosawa1, Kouichi Mizuno1, Chester W.

    Brown2, and Tetsuro Izumi1,3

    1Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular

    Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi

    371-8512, Japan; 2Division of Genetics, Department of Pediatrics, University of

    Tennessee Health Science Center, Memphis, TN 38163; and 3Research Program for

    Signal Transduction, Division of Endocrinology, Metabolism and Signal Research,

    Gunma University Initiative for Advanced Research, Gunma University, Maebashi 371-

    8512, Japan.

    A short running title: Roles of the insulin-GDF3-ALK7 axis in obesity

    Address correspondence to: Tetsuro Izumi, Phone: +81-27-220-8856; E-mail:

    [email protected]. Or to Katsuhide Okunishi, Phone: +81-27-220-8877; E-mail:

    [email protected].

    The word count: 4077-Endnote92=3985>4000 words (Introduction to Discussion)

    The number of figures: 6

  • 2

    Abstract

    Previous genetic studies in mice have shown that functional loss of activin receptor-like

    kinase 7 (ALK7), a type I transforming growth factor (TGF)-β receptor, increases

    lipolysis to resist fat accumulation in adipocytes. Although growth/differentiation factor

    3 (GDF3) has been suggested to function as a ligand of ALK7 under nutrient-excess

    conditions, it is unknown how GDF3 production is regulated. Here, we show that a

    physiologically low level of insulin converts CD11c- adipose tissue macrophages

    (ATMs) into GDF3-producing, CD11c+ macrophages ex vivo, and directs ALK7-

    dependent accumulation of fat in vivo. Depletion of ATMs by clodronate upregulates

    adipose lipases and reduces fat mass in ALK7-intact obese mice, but not in their ALK7-

    deficient counterparts. Furthermore, depletion of ATMs or transplantation of GDF3-

    deficient bone marrow negates the in vivo effects of insulin on both lipolysis and fat

    accumulation in ALK7-intact mice. The GDF3-ALK7 axis between ATMs and

    adipocytes represents a previously unrecognized mechanism by which insulin regulates

    both fat metabolism and mass.

  • 3

    Introduction

    The worldwide prevalence of obesity increases morbidity and mortality and imposes a

    growing public health burden. Most excess food intake is converted into fat, and

    specifically into triglycerides (TGs), which is stored in adipocytes of white adipose

    tissue (WAT). As adipocytes accumulate fat and increase in size, they start to secrete

    proinflammatory adipocytokines, recruit or polarize macrophages and other

    hematopoietic cells inside WAT, and cause chronic inflammation and obesity-related

    disorders {Gregor, 2011 #37}. The TG content in adipocytes is determined by the

    balance between the synthesis and breakdown of TG. While TG synthesis depends on

    the uptake of nutrients, the rate of lipid removal through lipolysis is proportional to the

    total fat mass as well as the activities of lipases, and is regulated by external factors,

    such as catecholamine and insulin. It is important to understand the mechanisms of fat

    accumulation to dissect the pathophysiology of obesity. Our previous genetic analyses

    using F2 progeny between the Tsumura, Suzuki, obese diabetes (TSOD) and control

    BALB/c mice revealed a naturally occurring mutation in Acvr1c encoding the type I

    TGF-β receptor, ALK7, in BALB/c mice {Hirayama, 1999 #6}{Mizutani, 2006

    #7}{Yogosawa, 2013a #3}{Yogosawa, 2013b #4}. The mutation gives rise to a stop

  • 4

    codon in the kinase domain of ALK7. The congenic strain, T.B-Nidd5/3, is isogenic

    with TSOD mice except for the BALB/c-derived ALK7 mutation and exhibits decreased

    adiposity due to enhanced lipolysis. Activation of ALK7 downregulates the master

    regulators of adipogenesis, C/EBPα and PPARγ, in differentiated adipocytes, which

    leads to suppression of lipolysis and to increases in adipocyte size and TG content.

    To understand the regulatory mechanisms associated with ALK7, it is essential

    to determine its physiological ligand. TGF-β family members such as Nodal, inhibin-βB

    (activin B or activin AB), GDF3, and GDF11 bind ALK7 and mediate its signals under

    specific conditions {Reissmann, 2001 #19}{Tsuchida, 2004 #20}{Andersson, 2006

    #21}{Andersson, 2008 #5}. Among them, GDF3 seems to function under nutrient-

    excess conditions, because both GDF3- and ALK7-knockout mice attenuate fat

    accumulation in the face of high-fat diet (HFD)-induced obesity {Andersson, 2008

    #5}{Shen, 2009 #22}. However, it has not been shown that GDF3 directly activates

    ALK7 in adipocytes. Besides, neither the producer nor the upstream regulator of GDF3

    under nutrient-excess conditions is known. In the present study, we establish GDF3 as

    the physiological ligand that activates ALK7 in adipocytes, and CD11c+ ATMs as the

    main cell source of GDF3. We further demonstrate that insulin upregulates GDF3 in

    ATMs ex vivo, and stimulates fat accumulation in vivo through the GDF3-ALK7

  • 5

    signaling pathway. Our findings reveal a novel mechanism by which insulin regulates

    adiposity through ATMs in addition to its classically defined direct effect on adipocytes.

  • 6

    Research Design and Methods

    Animal procedures. Animal experiments were performed in accordance with the rules

    and regulations of the Animal Care and Experimentation Committee, Gunma University.

    The TSOD mouse was originally established from an outbred ddY strain as an inbred

    strain with obesity and urinary glucose {Suzuki, 1999 #32}. The congenic mouse strain,

    T.B-Nidd5/3, was developed and characterized elsewhere {Mizutani, 2006

    #7}{Yogosawa, 2013 #3}. The GDF3-knockout mouse with a genetic background of

    C57BL/6J was previously described {Shen, 2009 #22}. C57BL/6N and BALB/cA mice

    were purchased from CLEA Japan. Only male mice were phenotypically characterized

    in the present study. Mice had ad libitum access to water and standard laboratory chow

    (CE-2; CLEA Japan) in an air-conditioned room with 12-h light-dark cycles. A HFD

    (55% fat, 28% carbohydrate, and 17% protein in calorie percentage; Oriental Yeast) was

    given to mice from 4 weeks of age for the indicated duration. For macrophage

    depletion, mice liposomes containing 110 mg/kg body weight of clodronate

    (ClodronateLiposomes.org) was injected intraperitoneally twice per week. For the in

    vivo insulin administration, saline or 0.75 U/kg body weight of insulin (Humulin R;

    Lilly), the amount generally used for insulin tolerance tests, was injected

  • 7

    intraperitoneally twice daily. For BM transplantation, recipient C57BL/6N mice at the

    ages of 8-10 weeks were irradiated twice with an individual dose of 5.4 Gy with a 3-h

    interval, and subsequently received an intravenous injection of 2 × 106 BM cells from

    donor wild-type or GDF3-knockout mice. Mice were sacrificed after anesthetization by

    isoflurane inhalation. Blood was collected from the inferior vena cava using 23-gauge

    needles and syringes. Serum non-esterified fatty acid (NEFA) levels were measured as a

    marker of lipolysis by NEFA C-test (Wako).

    Cell fractionation of epiWAT. EpiWAT was minced and digested with 1 mg/ml

    collagenase type I (Invitrogen) for 1 h at 37°C during shaking. The digested cells were

    filtered through a 250-µm nylon mesh (Kyoshin Rikoh) and centrifuged at 50 × g for 10

    min. The floating adipocytes were washed with PBS twice. After dispersing the pellet

    containing the SVF, the medium was filtered through a 40-µm nylon mesh and

    centrifuged at 300 × g for 10 min. The pellet was then incubated with erythrocyte-lysing

    buffer consisting of 155 mM NH4Cl, 5.7 mM K2HPO4, and 0.1 mM EDTA at room

    temperature for 1 min and washed with PBS twice.

    The cells in the stromal-vascular fraction (SVF) were resuspended in PBS, 2

    mM EDTA, and 2% FBS, and were incubated with excess Fc block (anti-CD16/CD32

  • 8

    antibodies, BD Bioscience) to block Fc receptor-mediated, nonspecific antibody

    binding. Cell surface markers were stained on ice in the dark for 20 min using CD11b-

    phycoerythrin-Cy7, F4/80-allophycocyanin (TONBO Biosciences), and CD11c-

    phycoerythrin (BD Biosciences) monoclonal antibodies. Some cells were stained as

    negative controls with fluorochrome-matched isotype control antibodies. After

    excluding dead cells by staining with 7-aminoactinomycin D, live cells were subjected

    to characterization of cell populations or to sorting of specific cell populations by

    FACSVerse or FACSAriaII flow cytometers (BD Biosciences).

    RNA preparation and gene expression analyses. RNA was extracted using Sepasol-

    RNA I Super (Nacalai Tesque). Total RNA (1 µg) was reverse-transcribed using oligo-

    (dT)12-18 primer and Superscript III (Invitrogen). Quantitative PCR was performed with

    SYBR premix Ex Taq (Takara Bio) using a LightCyler 480 (Roche). The results were

    normalized against 36B4 mRNA expression. The primer sequences are listed in

    Supplementary Table 1.

    Antibodies, immunoblotting, and immunostaining. Rabbit polyclonal anti-ALK7

    antibody was described previously {Yogosawa, 2013 #3}. Rabbit monoclonal

  • 9

    antibodies toward Smad3, phospho-Smad3 (Ser 423/425), Akt, and phospho-Akt (Ser

    473) were purchased from Cell Signaling Technology. Rat monoclonal anti-Cripto and

    goat polyclonal anti-GDF3 antibodies were purchased from R&D Systems. Mouse

    monoclonal antibodies toward β-actin and α-tubulin were purchased from Sigma-

    Aldrich. For immunoblotting, isolated adipocytes and the SVF were lysed with buffer

    (20 mM HEPES pH 7.4, 150 mM NaCl, 1% Triton X-100, 0.2 mM EDTA, and 1 mM

    dithiothreitol) containing protease and phosphatase inhibitors. The protein extracts (8-10

    µg for macrophages and 20 µg for other cells) were loaded onto polyacrylamide gels for

    electrophoresis. For imaging of whole-mount epididymal WAT (epiWAT), euthanized

    mice were perfused with 40 ml of fresh 1% paraformaldehyde (PFA) in PBS via

    intracardiac injection over a few minutes. EpiWAT was subdivided into small pieces

    (~0.1 cm3) by scissors, and was then fixed in 1% PFA in PBS and blocked in 5% BSA

    in PBS at room temperature for 30 min. For immunostaining of CD11c+ ATMs, cells

    attached on slide glasses by Cytospin (Thermo Fisher Scientific) were fixed with 3.7%

    PFA in PBS for 30 min at room temperature. With permeabilization by 0.1% Triton X-

    100, the tissues or the cells were incubated with 10 µg/ml of anti-GDF3 antibody or

    control IgG overnight at 4°C followed by the Alexa Fluor 488-conjugated secondary

    antibody (diluted at 1:500; Invitrogen) for 1 h at room temperature, and were onserved

  • 10

    under a laser scanning confocal microscope. The concentration of GDF3 in a medium

    was measured by mouse GDF3 ELISA kit (Elabscience Biotechnology).

    Vector construction and luciferase reporter assay. The binding site of Smad3 and

    Smad4 (CAGA)14 {Dennler, 1998 #9} was inserted into the pGL4.10[luc2] vector

    (Promega). HEK293T cells cultured in Dulbecco’s modified Eagle’s medium containing

    10% FBS and 1 mM L-glutamine were transfected with 20 ng of the reporter plasmid,

    10 ng of the control plasmid pGLA474[hRluc/TK], 12.5 ng of plasmid containing

    ALK7 cDNA {Yogosawa, 2013 #3}, and 6.25 ng of that containing Cripto cDNA

    derived from mouse embryo, using Lipofectamine 2000 reagent (Invitrogen). After 48

    h, the recombinant proteins of human GDF3, bone morphogenetic protein (BMP) 3,

    activin B, and TGF-β1 (R&D Systems) were added to the medium. After a further 24 h,

    the luciferase activities were measured by the Dual-Luciferase Reporter Assay System

    (Promega). The light units were normalized to Renilla luciferase activity.

    Lipolysis assay. Isolated mouse adipocytes (600 µl) were incubated at 37°C for 3 h in

    Krebs-Ringer Hepes buffer (20 mM HEPES pH 7.4, 120 mM NaCl, 5 mM KCl, 2 mM

    CaCl2, 1 mM MgCl2 and 1 mM KH2PO4) containing 2 mM glucose and 1% fatty acid-

  • 11

    free BSA. Lipolysis was assessed by measuring the concentration of glycerol in the

    buffer using a Free Glycerol Determination Kit (Sigma-Aldrich).

    Statistical analysis. All quantitative data were expressed as mean ± SD. Data analysis

    employed GraphPad Prism software. The p values were calculated using Student’s t-test

    or one-way ANOVA with Tukey’s multiple comparison test, as appropriate, to determine

    significant differences between group means.

  • 12

    Results

    GDF3 produced from CD11c+ ATMs functions as a ligand of ALK7 in adipocytes.

    Because ALK7-knockout mice show reduced fat accumulation when fed a HFD, but

    exhibit normal weight when fed regular chow {Andersson, 2008 #5}, the ALK7 signal

    could be activated under nutrient-excess conditions. We thus screened TGF-β

    superfamily members that exhibit differential expressions depending on nutritional

    states and also between the absence or presence of functional ALK7. For this purpose,

    we isolated tissues potentially involved in nutritional metabolism from ALK7-intact

    C57BL/6 and ALK7-deficient BALB/c lean mouse strains fed either regular chow or a

    HFD. We also isolated these tissues from ALK7-intact TSOD and ALK7-deficient T.B-

    Nidd5/3 obese strains, both of which have the same genetic background {Mizutani,

    2006 #7}{Yogosawa, 2013a #3}. Among the 33 members of the mammalian TGF-β

    superfamily {Shi, 2011 #8}, GDF3 and BMP3, inhibin-βB, and TGF-β1 showed

    differential expression in WAT (Fig. 1A and Supplementary Fig. 1). Their expression in

    WAT was strongly upregulated in C57BL/6 mice fed a HFD compared with those fed

    regular chow. Some of them were also upregulated in obese TSOD mice fed regular

    chow and even in ALK7-deficient BALB/c mice fed a HFD. In contrast to the other

  • 13

    three ligands, GDF3 showed a remarkably high and specific expression in epiWAT of

    TSOD and HFD-fed C57BL/6 mice, consistent with previous findings {Shen, 2009

    #22}{Yogosawa, 2013a #3}. We then examined the ligand activity through ALK7 in

    HEK293T cells expressing a luciferase reporter containing a Smad3/4 responsive

    element {Dennler, 1998 #9}, which acts downstream of ALK7 in adipocytes

    {Yogosawa, 2013a #3}. Consistent with the previous finding {Andersson, 2008 #5},

    GDF3 activated the reporter in a dose-dependent fashion only in the presence of

    exogenously expressed ALK7 and Cripto, a co-receptor that enhances signaling via the

    type I and type II receptor kinase complex {Chen, 2006 #42} (Fig. 1B). In contrast,

    BMP3 did not show such enhancement. Activin B, a dimer of inhibin-βB, and TGF-β1

    activated the reporter even in the absence of ALK7 and Cripto, although activing B

    induced slight activation with the receptor expression. These findings make GDF3 the

    most likely candidate ligand for ALK7.

    ALK7-deficient T.B-Nidd5/3 mice at 7 weeks of age showed a significant

    reduction in epiWAT weight (~ 0.6 g), which was almost equal to the reduction of body

    weight, compared with control TSOD mice (Supplementary Fig. 2A). The mice

    exhibited increased levels of mRNA encoding the transcription factors PPARγ and

    C/EBPα, and their downstream genes encoding adipose triglyceride lipase (ATGL) and

  • 14

    hormone-sensitive lipase (HSL), as previously reported in older mice {Yogosawa,

    2013a #3}. Serum NEFA reflecting enhanced lipolysis were also elevated relative to

    control TSOD mice. Therefore, the ALK7-deficient phenotypes become overt at 7

    weeks of age. GDF3 inhibited lipolysis in adipocytes from TSOD mice at this age,

    whereas neither BMP3, activin B, nor TGF-β1 did so (Fig. 2A), consistent with the

    findings from the luciferase assays (Fig. 1B). Importantly, GDF3 inhibited lipolysis and

    activated the downstream Smad3 by phosphorylation only in ALK7-intact adipocytes

    from TSOD mice, but not in ALK7-deficient adipocytes from T.B-Nidd5/3 mice (Fig.

    2B and C). These findings establish that GDF3 can signal through ALK7 in adipocytes.

    Because GDF3 is expressed in thymus, spleen, and bone marrow (BM) as well

    as in WAT (Fig. 1A), as originally reported {McPherron, 1993 #11}, it might be

    expressed in hematopoietic cells rather than adipocytes in WAT. To identify the cell

    source of GDF3, we first dissociated the epiWAT into the SVF and mature adipocytes,

    then further fractionated SVF cells by fluorescence activated cell sorting using

    fluorochrome-conjugated antibodies targeting macrophage surface markers {Gordon,

    2005 #14}. GDF3 transcripts were enriched in the SVF, particularly in CD11b+ F4/80+

    macrophages (defined as ATMs), with the greatest elevation seen in those expressing

    CD11c (Fig. 2D and Supplementary Fig. 2B). Immunostaining with anti-GDF3 antibody

  • 15

    revealed that most of the CD11c+ ATMs express GDF3 (94.8 ± 2.2%; n = 3:

    approximately a hundred cells were examined in total). GDF-positive cells located

    around individual adipocytes in WAT, consistent with its localization in ATMs. In

    contrast, BMP3 and inhibin-βB were expressed mainly in mature adipocytes, whereas

    TGF-β1 was ubiquitously expressed in every cell fraction (Supplementary Fig. 2C).

    Concomitant increases in the CD11c and GDF3 transcripts were also found in the SVF

    of HFD-fed C57BL/6 mice (Supplementary Fig. 1D). Although inflammasome

    activation has recently been shown to induce GDF3 in ATMs from aged mice {Camell,

    2017 #40}, the GDF3 induction in TSOD or HFD-treated C57BL/6 mice was not

    accompanied by upregulation of inflammasome activation-related genes, such as tumor

    necrosis factor-α (TNFα), monocyte chemotactic protein-1 (MCP-1), NLRP3, and

    Caspase 1 (Supplementary Fig. 1B and D).

    Macrophage depletion reverses the effects of ALK7 on adiposity. To evaluate the

    role of GDF3-producing ATMs in vivo, we intraperitoneally injected clodronate to

    deplete macrophages {Van Rooijen, 1994 #33}. Clodronate treatment partially but

    significantly decreased the percentage of ATMs, including that of CD11c+ ATMs, as

    well as the expression of F4/80, CD11c, and GDF3, in both TSOD and T.B-Nidd5/3

  • 16

    mice (Fig. 3A and Supplementary Fig. 3A and B). However, clodronate decreased total

    body weight, particularly epiWAT weight, only in TSOD mice, indicating that the drug’s

    effects depend on intact ALK7. Furthermore, it increased the PPARγ, C/EBPα, ATGL,

    and HSL transcripts, and the serum NEFA concentration normalized to the epiWAT

    weight, in TSOD mice (Fig. 3B). Therefore, the effects of macrophage depletion from

    ALK7-intact TSOD mice are remarkably similar to the phenotypic changes in ALK7-

    deficient T.B-Nidd5/3 mice when compared to control TSOD mice {Mizutani, 2006

    #7}{Yogosawa, 2013a #3}, indicating that the GDF3-ALK7 axis represents a major link

    between macrophages and adipocytes in the regulation of whole body lipid metabolism

    and fat accumulation.

    Insulin upregulates GDF3 in ATMs. We next explored the external factors that

    increase GDF3 production under nutrient-excess conditions. CD11c- ATMs isolated

    from epiWAT of TSOD mice showed elevated levels of the GDF3 transcript during

    culture in FBS-containing medium (Supplementary Fig. 4A), suggesting that some FBS

    component converts CD11c- to CD11c+ ATMs and concomitantly induces GDF3

    expression. Because obesity is frequently coincident with hyperinsulinemia, we

    suspected that insulin might upregulate GDF3. Plasma insulin concentrations are

  • 17

    approximately ~170 pM in lean BALB/c mice and ~1.7 nM in obese TSOD mice

    {Hirayama, 1999 #6}. Ex vivo administration of 10 µU/ml of insulin (61 pM) increased

    both CD11c and GDF3 expressions after a 24-h culture in CD11c- macrophages derived

    from epiWAT of TSOD mice, and wortmannin, an inhibitor of phosphatidylinositol-3-

    kinase, inhibited insulin-induced GDF3 upregulation (Fig. 4A). Insulin also increased

    the expression of the typical M2 markers, arginase (Arg1) and chitinase-like 3 (Ym1),

    but not that of the M1 markers, TNFα and MCP-1. Although 61 pM insulin induced

    GDF3 in ATMs, it increased GDF3 only weakly in macrophages derived from lung,

    peritoneum, or BM of TSOD mice (Supplementary Fig. 4B). This was evident in the

    low level of expression of the insulin receptor in these macrophages in contrast to that

    in ATMs. These findings indicate the tissue selectivity of insulin sensitivity in

    macrophages.

    Although the above findings raise the possibility that insulin inhibits lipolysis

    and accumulates fat in adipocytes through upregulation of GDF3 in ATMs, insulin is

    generally believed to do so by directly acting on adipocytes. We next investigated the

    effects of insulin on isolated adipocytes. We confirmed that insulin phosphorylates the

    downstream Akt kinase, but does not activate Smad3 by a non-canonical pathway, in

    adipocytes (Fig. 4B). However, the concentration of insulin (61 pM) we administered ex

  • 18

    vivo to ATMs (Fig. 4A) did not inhibit basal or catecholamine-induced lipolysis in

    adipocytes, although a higher concentration of insulin (25 nM) did so (Fig. 4C). These

    findings indicate that a much higher dose of insulin is required to directly inhibit

    lipolysis in adipocytes than is required to upregulate GDF3 in ATMs. Although ALK7

    deficiency has been reported to enhance catecholamine-induced lipolysis in adipocytes

    {Guo, 2014 #26}, we found that unstimulated lipolysis is already elevated, and that the

    extent of stimulation by catecholamine is not changed, in ALK7-deficient adipocytes

    (Fig. 4C). These findings confirm the previous finding that ALK7 deficiency elevates

    basal lipolysis by affecting the expression levels of adipose lipases {Yogosawa, 2013a

    #3}.

    To reinforce the functional significance of insulin’s activity through GDF3

    production from ATMs, we performed reconstitution assays by incubating adipocytes

    with the supernatant of CD11c- ATMs that had been treated with or without 61 pM insulin.

    Note that this concentration of insulin does not directly inhibit lipolysis in isolated

    adipocytes (Fig. 4C). Insulin induced secretion of GDF3 into their supernatants (1-2

    ng/ml), which dose-dependently increased the phosphorylation of Smad3 and inhibited

    lipolysis in adipocytes of ALK7-intact TSOD mice, but not in those of ALK7-deficient

    T.B-Nidd5/3 mice (Fig. 4D and E). We confirmed that 61 pM insulin did not change the

  • 19

    expression levels of inhibin-βB and TGF-β1 in the ATMs (Supplementary Fig. 4C), both

    of which can induce Smad3 phosphorylation in adipocytes. These findings indicate that

    the insulin-stimulated release of GDF3 from ATMs successfully inhibits lipolysis in

    adipocytes ex vivo.

    Insulin inhibits lipolysis and accumulates fat in an ALK7-dependent manner in

    vivo. To clarify whether insulin functions through the GDF3-ALK7 signaling pathway

    in vivo, we intraperitoneally administered insulin twice a day for 2 weeks to TSOD and

    T.B-Nidd5/3 mice. This in vivo insulin treatment elevated the WAT weight, and

    decreased the levels of the ATGL and HSL transcripts and serum NEFA, in an ALK7-

    dependent manner (Fig. 5A), suggesting that insulin inhibits lipolysis and accumulates

    fat through the upregulation of GDF3 in ATMs.

    In order to exclude the possibility that insulin’s effects via the GDF3-ALK7

    axis are applicable only to the TSOD strain, for which the molecular pathogenesis of

    obesity and diabetes is unknown {Hirayama, 1999 #6}, we administered insulin to a

    commonly used C57BL/6 strain fed a HFD that indeed expressed ALK7 in WAT

    (Supplementary Fig. 5A). Insulin increased adiposity in parallel with reductions in

    expression of adipose lipases in epiWAT and serum NEFA concentration in C57BL/6

  • 20

    mice (Fig. 5B). However, no such effects were found in ALK7-deficient BALB/c mice

    fed a HFD. These findings indicate that insulin’s effects via ALK7 under nutrient-excess

    conditions continue irrespective of the mouse strain.

    GDF3 mediates the activity of insulin to promote adiposity in vivo. To further

    substantiate the role of the GDF3-ALK7 axis in insulin activity in vivo, we injected

    clodronate to deplete macrophages and then administered insulin to C57BL/6 mice fed a

    HFD. We confirmed that neither clodronate nor insulin treatment alters the food intake

    of mice (Supplementary Fig. 5B). Clodronate treatment markedly decreased ATMs,

    including CD11c+ ATMs, and concomitantly reduced GDF3 levels in the SVF (Fig. 6A

    and Supplementary Fig. 5B). Remarkably, it eliminated the in vivo effects of insulin to

    increase CD11c and GDF3 in the SVF and adiposity in the whole body, and to decrease

    adipose lipases and the serum concentration of NEFA. These findings demonstrate that

    insulin can regulate fat metabolism and mass through its effects on macrophages in

    vivo.

    Finally, we performed BM transplantation experiments to directly prove the

    involvement of GDF3 in the insulin activity. We transplanted the BM of GDF3-deficient

    C57BL/6 mice {Shen, 2009 #22} to wild-type C57BL/6 mice to evade the cell

  • 21

    elimination by the immune system due to major histocompatibility complex mismatch.

    The recipient mice were then fed a HFD and treated with insulin. We confirmed that

    GDF3 deficiency in BM cells does not affect the numbers of ATM (Supplementary Fig.

    5C). In contrast to the mice harboring the wild-type BM, those harboring the GDF3-

    deficient BM and thus losing GDF3 in the SVF failed to mediate the in vivo effects of

    insulin to inhibit lipolysis in the WAT (Fig. 6B). These findings demonstrate that GDF3

    production is necessary for insulin to regulate fat metabolism and mass under nutrient-

    excess conditions.

  • 22

    Discussion

    We showed that GDF3 produced from CD11c+ ATMs acts as a ligand of ALK7 in

    adipocytes to inhibit lipolysis and accumulate fat under nutrient-excess conditions. The

    GDF3-ALK7 axis within WAT should represent the major interactive mechanism

    between macrophages and adipocytes in the regulation of adiposity, because

    nonselective macrophage depletion by clodronate highlights the ALK7-specific effects,

    such as decreases in body and epiWAT weights, and increases in the expressions of

    C/EBPα, PPARγ, ATGL, and HSL, as well as NEFA production in WAT, in ALK7-intact

    TSOD mice, but not in their ALK7-deficient counterparts. While many studies have

    focused on the effects of macrophages in the formation of chronic inflammation

    associated with obesity, the present study demonstrates the role of ATMs in fat

    accumulation per se. Although CD11c+ macrophages are conventionally understood to

    be M1 macrophages that are recruited to and/or polarized in obese WAT to induce a

    chronic inflammatory state {Lumeng, 2007 #15}, the GDF3-producing cells express a

    substantial level of M2 markers. Similar to our findings, it has recently been shown that

    a prototypical M2 marker, CD301b, as well as Arg1, is selectively expressed in CD11c+

    mononuclear phagocytes including ATMs, and that depleting these cells leads to weight

  • 23

    loss and increased insulin sensitivity in mice {Kumamoto, 2016 #28}.

    We found that a physiologically low concentration of insulin alters the

    properties of CD11c- ATMs ex vivo by increasing the expressions of CD11c and GDF3.

    Moreover, in vivo insulin administration inhibits lipolysis and expands WAT in an

    ALK7-dependent manner, which indicates that insulin regulates fat metabolism and

    mass via the GDF3-ALK7 axis. Consistently, the in vivo effects of insulin on WAT are

    absent after depletion of macrophages or transplantation of GDF3-deficient BM. It is

    intriguing that ATMs appear to specifically express a high level of insulin receptor

    compared with macrophages in other tissues. Although insulin is generally thought to

    inhibit lipolysis directly in adipocytes by regulating the cAMP-mediated signaling

    pathway {Burns, 1979 #29}{Choi, 2006 #30}{Lafontan, 2009 #24}{Degerman, 2011

    #31}, and/or by suppressing transcription of adipose lipases {Kralisch, 2005

    #16}{Kershaw, 2006 #17}{Kim, 2006 #18}, these actions in adipocytes have been

    detected only at higher concentrations of insulin (1-100 nM) than those applied to ATMs

    in the present study (61 pM). In fact, we observed that 25 nM insulin can directly inhibit

    both basal and catecholamine-stimulated lipolysis in adipocytes, whereas 61 pM insulin

    cannot. Therefore, insulin can differentially regulate fat metabolism and mass

    depending on its local concentration in WAT.

  • 24

    Given that GDF3 induction in ATMs requires a minimal concentration of

    insulin, the GDF3-ALK7 pathway should be active at the beginning of

    hyperinsulinemia under nutrient-excess conditions. This has a clinical implication for

    the importance of “early intervention” in adiposity before the manifestation of insulin

    resistance. Insulin resistance-related chronic hyperinsulinemia may accelerate fat

    accumulation even under the same energy balance via activation of the GDF3-ALK7

    axis, which makes it much harder for obese individuals to reduce adiposity. Future

    research should focus on novel targeting strategies for this pathway, such as inhibitors

    of GDF3 and ALK7, specific depletion of ATMs, and macrophage-specific inhibition of

    insulin receptor expression.

    In summary, we present a novel mechanism of obesity (Fig. 6C). Under

    nutrient-excess conditions, insulin efficiently activates insulin receptor expressed on

    CD11c- ATMs and converts them to CD11c+ ATMs to produce GDF3. GDF3 locally

    stimulates ALK7 on adipocytes and activates Smads 2-4 to downregulate PPARγ,

    C/EBPα, and also adipose lipases to store excess nutrient as fat {Yogosawa, 2013a #3}.

    However, persistent activation of this physiological pathway enlarges adipocytes and

    may change adipocytokine repertoires to cause chronic inflammation and insulin

    resistance. In fact, ALK7-intact, aged obese mice exhibit elevated levels of

  • 25

    proinflammatory MCP-1 and TNFα, a reduced level of insulin-sensitizing adiponectin,

    and greater glucose intolerance, compared with their ALK7-deficient counterparts

    {Yogosawa, 2013a #3}{Yogosawa, 2013b #4}. As such, the insulin-GDF3-ALK7 axis

    plays a pivotal role in both physiological and pathological fat accumulation in WAT.

  • 26

    Acknowledgments. We are grateful to T. Nara, E. Kobayashi and T. Ushigome for

    colony maintenance of mice, S. Shigoka for assistance in preparing the manuscript, and

    the staffs at Bioresource Center, Gunma University for their help in breeding of mice.

    Funding. This work was supported by JSPS KAKENHI Grant Numbers JP24659442

    and JP25126702 to T.I., and JP25860739 to S.Y. It was also supported by grants from

    Japan Diabetes Foundation and from Novo Nordisk Insulin Study Award (to T.I.).

    Duality of Interest. The authors have declared that no conflict of interest exists.

    Author contributions. Y.B., K.O., S.Y., and K.M. performed experiments, C.W.B

    provided experimental reagents, and K.O. and T.I. designed experiments and wrote the

    paper.

    The guarantor of the study. T.I. is the guarantor of this work and, as such, had full

    access to all the data in the study and takes responsibility for the integrity of the data

    and the accuracy of the data analysis.

  • 27

    References

    1. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol 2011;29:415-445

    2. Hirayama I, Yi Z, Izumi S, Arai I, Suzuki W, Nagamachi Y, Kuwano H, Takeuchi T, Izumi T. Genetic analysis of obese diabetes in the TSOD mouse. Diabetes 1999;48:1183-1191

    3. Mizutani S, Gomi H, Hirayama I, Izumi T. Chromosome 2 locus Nidd5 has a potent effect on adiposity in the TSOD mouse. Mamm Genome 2006;17:375-384

    4. Yogosawa S, Mizutani S, Ogawa Y, Izumi T. Activin receptor-like kinase 7 suppresses lipolysis to accumulate fat in obesity through downregulation of peroxisome proliferator-activated receptor γ and C/EBPα. Diabetes 2013;62:115-123

    5. Yogosawa S, Izumi T. Roles of activin receptor-like kinase 7 signaling and its target, peroxisome proliferator-activated receptor γ, in lean and obese adipocytes. Adipocyte 2013;2:246-250

    6. Reissmann E, Jornvall H, Blokzijl A, Andersson O, Chang C, Minchiotti G, Persico MG, Ibanez CF, Brivanlou AH. The orphan receptor ALK7 and the Activin receptor ALK4 mediate signaling by Nodal proteins during vertebrate development. Genes Dev 2001;15:2010-2022

    7. Tsuchida K, Nakatani M, Yamakawa N, Hashimoto O, Hasegawa Y, Sugino H. Activin isoforms signal through type I receptor serine/threonine kinase ALK7. Mol Cell Endocrinol 2004;220:59-65

    8. Andersson O, Reissmann E, Ibanez CF. Growth differentiation factor 11 signals through the transforming growth factor-β receptor ALK5 to regionalize the anterior-posterior axis. EMBO Rep 2006;7:831-837

    9. Andersson O, Korach-Andre M, Reissmann E, Ibáñez CF, Bertolino P. Growth/differentiation factor 3 signals through ALK7 and regulates accumulation of adipose tissue and diet-induced obesity. Proc Natl Acad Sci U S A 2008;105:7252-7256

    10. Shen JJ, Huang L, Li L, Jorgez C, Matzuk MM, Brown CW. Deficiency of growth differentiation factor 3 protects against diet-induced obesity by selectively acting on white adipose. Mol Endocrino. 2009;23:113-123

    11. Suzuki W, Iizuka S, Tabuchi M, Funo S, Yanagisawa T, Kimura M, Sato T, Endo

  • 28

    T, Kawamura H. A new mouse model of spontaneous diabetes derived from ddY strain. Exp Anim 1999;48:181-189

    12. Dennler S, Itoh S, Vivien D, ten Dijke P, Huet S, Gauthier JM. Direct binding of Smad3 and Smad4 to critical TGFβ-inducible elements in the promoter of human plasminogen activator inhibitor-type 1 gene. EMBO J 1998;17:3091-3100

    13. Shi M, Zhu J, Wang R, Chen X, Mi L, Walz T, Springer TA. Latent TGF-beta structure and activation. Nature 2011;474:343-349

    14. Chen C, Ware SM, Sato A, Houston-Hawkins DE, Habas R, Matzuk MM, Shen MM, and Brown CW. The Vg1-related protein Gdf3 acts in a Nodal signaling pathway in the pre-gastrulation mouse embryo. Development 2006;133:319-329

    15. McPherron AC, Lee SJ. GDF-3 and GDF-9: two new members of the transforming growth factor-β superfamily containing a novel pattern of cysteines. J Biol Chem 1993;268:3444-3449

    16. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol 2005;5:953-964

    17. Camell CD, Sander J, Spadaro O, Lee A, Nguyen KY, Wing A, Goldberg EL, Youm YH, Brown CW, Elsworth J, et al. Inflammasome-driven catecholamine catabolism in macrophages blunts lipolysis during ageing. Nature 2017;550:119-23

    18. Van Rooijen N, Sanders A. Liposome mediated depletion of macrophages: mechanism of action, preparation of liposomes and applications. J Immunol Methods 1994;174:83-9319. Guo T, Marmol P, Moliner A, Björnholm M, Zhang C, Shokat KM, Ibanez CF. Adipocyte ALK7 links nutrient overload to catecholamine resistance in obesity. Elife 2014;3:e03245

    20. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 2007;117:175-184

    21. Kumamoto Y, Camporez JP, Jurczak MJ, Shanabrough M, Horvath T, Shulman GI, Iwasaki A. CD301b+ mononuclear phagocytes maintain positive energy balance through secretion of resistin-like molecule alpha. Immunity 2016;45:583-596

    22. Burns TW, Terry BE, Langley PE, Robison GA. Insulin inhibition of lipolysis of human adipocytes: the role of cyclic adenosine monophosphate. Diabetes 1979;28:957-961

    23. Choi YH, Park S, Hockman S, Zmuda-Trzebiatowska E, Svennelid F, Haluzik M, Gavrilova O, Ahmad F, Pepin L, Napolitano M, et al. Alterations in

  • 29

    regulation of energy homeostasis in cyclic nucleotide phosphodiesterase 3B-null mice. J Clin Invest 2006;116:3240-3251

    24. Lafontan M, Langin D. Lipolysis and lipid mobilization in human adipose tissue. Prog Lipid Res 2009;48:275-297

    25. Degerman E, Ahmad F, Chung YW, Guirguis E, Omar B, Stenson L, Manganiello V. From PDE3B to the regulation of energy homeostasis. Curr Opin Pharmacol 2011;11:676-82

    26. Kralisch S, Klein J, Lossner U, Bluher M, Paschke R, Stumvoll M, Fasshauer M. Isoproterenol, TNFα, and insulin downregulate adipose triglyceride lipase in 3T3-L1 adipocytes. Mol Cell Endocrinol 2005;240:43-49

    27. Kershaw EE, Hamm JK, Verhagen LA, Peroni O, Katic M, Flier JS. Adipose triglyceride lipase: function, regulation by insulin, and comparison with adiponutrin. Diabetes 2006;55:148-57

    28. Kim JY, Tillison K, Lee JH, Rearick DA, Smas CM. The adipose tissue triglyceride lipase ATGL/PNPLA2 is downregulated by insulin and TNF-α in 3T3-L1 adipocytes and is a target for transactivation by PPARγ. Am J Physiol Endocrinol Metab 2006;291:E115-127

  • 30

    Figure Legends

    Figure 1. Screening TGF-β superfamily members to identify ALK7 ligands

    (A) TSOD mice and their ALK7-deficient counterparts, T.B-Nidd5/3 mice, fed regular

    chow (RC) were sacrificed at 10 weeks of age. C57BL/6 (B6) and BALB/c (BALB)

    mice fed either RC or a HFD from 4 weeks of age were sacrificed at 14 weeks of age.

    Total RNA was isolated from the indicated tissues, including epididymal WAT

    (epiWAT), inguinal WAT (ingWAT), and brown adipose tissue (BAT), and mRNA levels

    of GDF3, BMP3, inhibin-βB, and TGF-β1 were quantified and normalized to the

    average values in epiWAT of C57BL/6 mice fed RC (n = 3). (B) HEK239T cells were

    transfected with plasmids encoding ALK7 and/or Cripto. The protein levels of ALK7

    and Cripto were examined at 48 h post-transfection by immunoblotting (left panel).

    HEK293T cells were transfected with plasmids encoding ALK7 and Cripto and

    simultaneously with a luciferase reporter fused with the Smad-binding promoter

    element. At 48 h post-transfection, different concentrations (0, 50, 150, and 400 ng/ml)

    of the indicated recombinant protein were added to the cells. The luciferase activities

    were measured after further 24 h (middle panel: GDF3, n = 4; BMP3, n = 3; right panel,

    n = 3). *p < 0.05, **p < 0.01, ***p < 0.001; Student’s t-test. #p < 0.05, ##p < 0.01, ###p <

  • 31

    0.001; one-way ANOVA.

    Figure 2. GDF3 acts on ALK7 within WAT

    (A-C) Primary adipocytes derived from epiWAT of 7-week-old TSOD or T.B-Nidd5/3

    mice were incubated with the indicated recombinant protein (400 ng/ml) for 3 h (A, B)

    or 30 min (C). Glycerol release was measured and normalized to that of control TSOD

    adipocytes (A, B, n = 3). Phosphorylation of Smad3 in adipocytes was examined by

    immunoblotting with the indicated antibodies (C). The band with a black arrowhead in

    the p-Smad3 panel is a nonspecific protein. (D) EpiWAT of 7-week-old TSOD mice

    were biochemically separated into adipocytes and the SVF. The SVF was then

    fractionated by FACS as shown in Supplementary Fig. 1B. GDF3 mRNA levels were

    quantified in each of the cell fractions (left panel: epiWAT, n = 4; adipocytes, n = 3;

    SVF, n = 4; CD11b- cells in the SVF, n = 5; CD11b+ cells in the SVF, n = 4; CD11b+

    F4/80- non-macrophage cells, n = 3; CD11b+ F4/80+ macrophages, n = 3; CD11c-

    macrophages, n = 4; and CD11c+ macrophages, n = 6). #p < 0.05, ##p < 0.01, ###p <

    0.001; one-way ANOVA.

    Figure 3. Effects of macrophage depletion by clodronate

  • 32

    PBS or clodronate encapsulated in liposomes (CLO) was injected intraperitoneally into

    TSOD and T.B-Nidd5/3 mice twice a week for 3 weeks from 4 weeks of age. Three

    days after the final injection at 7 weeks of age, the SVF was isolated from epiWAT. (A)

    The mRNA level of GDF3 in SVF (TSOD, n = 8; T.B-Nidd5/3, n = 4), body weights at

    4 weeks of age, and body and epiWAT weights and their ratio at 7 weeks of age (n = 5)

    in mice with or without CLO treatment. (B) The mRNA levels of adipose transcription

    factors and lipases in epiWAT and serum NEFA concentration normalized to the

    epiWAT weight (TSOD, n = 8; T.B-Nidd5/3, n = 4). #p < 0.05, ##p < 0.01, ###p < 0.001;

    one-way ANOVA.

    Figure 4. Effects of insulin administered to CD11c- ATMs, adipocytes.

    (A) CD11c- macrophages from epiWAT of 7-week-old TSOD mice (1.5 × 106 cells/24-

    well dish) were incubated with or without 61 pM insulin in Krebs-Ringer Hepes buffer

    for 24 h (left panel; n = 7). Some were pretreated with the indicated concentration of

    wortmannin 10 min before the 24-h incubation (right panel; n = 3). The mRNA levels of

    the indicated genes were quantified and normalized to those without insulin incubation

    in each experiment (middle panel). Insulin-induced phosphorylation of Akt in CD11c-

    macrophages pretreated with or without 100 nM wortmannin was examined by

  • 33

    immunoblotting with the indicated antibodies (right panel). (B) Primary adipocytes

    isolated from epiWAT of TSOD or T.B-Nidd5/3 mice were incubated for 30 min with 0

    M, 61 pM, or 25 nM of insulin or with 400 ng/ml of GDF3. The cell extracts were

    immunoblotted with the indicated antibodies. The band with a black arrowhead in the p-

    Smad3 panel is a nonspecific protein. (C) Adipocytes were incubated with or without 10

    µM isoproterenol plus 0 M, 61 pM, or 25 nM insulin for 3 h. Glycerol levels in the

    medium were measured and normalized to those of TSOD adipocytes without insulin or

    isoproterenol incubation in each experiment (n = 3). (D, E) CD11c- ATMs from TSOD

    mice were cultured with or without 61 pM insulin for 24 h as in (A). The conditioned

    medium of the macrophages was harvested after centrifugation of the culture plate at

    300 × g for 10 min at 4°C, and the concentration of GDF3 was measured (D, upper

    panel; n = 4). The conditioned medium warmed to 37°C was incubated with adipocytes

    of TSOD mice for 30 min to examine its effect on Smad3 phosphorylation (D, lower

    panel), or with adipocytes of TSOD (n = 5) or T.B-Nidd5/3 (n = 3) mice for 3 h to

    examine its effect on glycerol release (E). *p < 0.05, **p < 0.01, ***p < 0.001; Student’s t-

    test. #p < 0.05, ##p < 0.01, ###p < 0.001; one-way ANOVA.

    Figure 5. Effects of insulin administered to a whole body.

  • 34

    (A) Saline or insulin (0.75 U/kg body weight) was injected intraperitoneally into TSOD

    (upper panels; n = 10) and T.B-Nidd5/3 (lower panels; n = 8) mice twice daily for 2

    weeks from 5 weeks of age. Shown are the weight ratio of epiWAT to total body, mRNA

    levels of ATGL and HSL in epiWAT, and serum NEFA concentration normalized to the

    epiWAT weight. (B) Saline or insulin was injected to C57BL/6 (upper panels; n = 8) and

    BALB/c mice (lower panels; n = 8) that had been fed a HFD for 3 weeks from 4 weeks

    of age, and the effects of insulin were examined as in (A). *p < 0.05, **p < 0.01;

    Student’s t-test.

    Figure 6. Insulin regulates fat metabolism and mass through upregulation of GDF3

    in ATMs

    (A) C57BL/6 mice fed a HFD (n = 7 per group) were treated with PBS or clodronate

    from 4 weeks of age for 3 weeks as described in Fig. 3, and were also treated with saline

    or insulin from 5 weeks of age for 2 weeks as described in Fig. 5B. Shown are GDF3

    mRNA levels in the SVF the weight ratio of epiWAT to total body, serum NEFA

    concentration normalized to the epiWAT weight, and ATGL and HSL mRNA levels in

    epiWAT. (B) The BM of wild-type (WT) or GDF3-knockout (KO) C57BL/6 mice at 8-

    10 weeks of age were transplanted into wild-type C57BL/6 mice. The recipient mice

  • 35

    were fed a HFD for 3 weeks and treated with insulin for 2 weeks from 6 and 7 weeks

    after the BM transfer, respectively (n = 6-9 per group). #p < 0.05, ##p < 0.01, ###p <

    0.001; one-way ANOVA. (C) Scheme of the insulin-GDF3-ALK7 axis. See text in

    Discussion.

  • 0

    5

    10

    15

    20

    25

    30

    35

    40

    Brain Thymus Liver Spleen BAT epiWATingWAT BM Muscle

    Rel

    ativ

    eex

    pres

    sion

    GDF3

    *

    **

    B6-HFDB6-RC

    T.B-Nidd5/3-RC

    BALB-HFDBALB-RC

    TSOD-RCALK7(+)

    ALK7(-)

    ***

    Figure 1

    B

    ALK7

    Cripto

    -actin

    ALK7

    Cripto

    ++

    ++

    A

    ***0

    1

    2

    3

    4

    5

    6

    7

    8

    Brain Thymus Liver Spleen BAT epiWATingWAT BM Muscle

    BMP3

    **

    0

    2

    4

    6

    8

    10

    12

    14

    Brain Thymus Liver Spleen BAT epiWATingWAT BM Muscle

    Rel

    ativ

    eex

    pres

    sion

    Inhibin-B

    *

    *

    *

    0

    5

    10

    15

    20

    25

    30

    Brain Thymus Liver Spleen BAT epiWATingWAT BM Muscle

    TGF-β1

    ** ** * *

    0

    20

    40

    60

    80

    100

    120

    140

    ALK7+

    Cripto

    GDF3 BMP3

    ##

    Rel

    ativ

    e lu

    cife

    rase

    act

    ivity 0 50 150 400

    (ng/mL)

    + +

    #

    0

    5

    10

    15

    20

    25

    30

    350 50 150 400

    (ng/mL)

    ###

    ##

    #####

    GDF3 Activin B TGF-1

    Rel

    ativ

    e lu

    cife

    rase

    act

    ivity

    ### ######

    ###

    + + +

    #

    ###

  • 0

    5

    10

    15

    20

    25

    epiW

    AT

    adip

    ocyte

    SV

    F

    CD

    11b

    -

    CD

    11b

    +

    F4

    /80

    -

    F4

    /80

    +

    CD

    11c-

    CD

    11

    c+

    epiWAT SVF CD11b+ CD11b+F4/80+

    GDF3 mRNA expression

    A C

    Rela

    tive e

    xpre

    ssio

    n

    TSOD CD11c+ ATM

    ATM

    control IgG anti-GDF3

    ###

    ###

    ###

    ##

    a-tubulin

    TSOD ATM

    GDF3

    50kD

    37kD

    CD11c

    + -

    Figure 2

    TSOD epiWAT

    control IgG

    anti-GDF3

    0

    0.5

    1

    1.5

    2

    - + - +

    TSOD T.B-Nidd5/3R

    ela

    tive g

    lycero

    l re

    lease

    #

    Primary adipocyte

    GDF3

    ######

    ###

    B

    p-Smad3

    total-Smad3

    b-actin

    GDF3 ++

    TSOD T.B-Nidd5/3

    Primary adipocyte

    : non-specific bandRela

    tive g

    lycero

    l re

    lease

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    TSOD adipocyte

    D

    Control GDF3 BMP3 Activin B TGF-b1

    ## ##

    ###

  • 0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    0

    1

    2

    3

    4

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    0

    0.5

    1

    1.5

    2

    2.5

    3

    3.5

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    05

    1015202530354045

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    BW (g) (after 3-wk Tx)

    Figure 3

    05

    1015202530354045

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    BW (g)(at the start)

    00.2

    0.40.60.8

    1

    1.21.41.6

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    epiWAT (g)

    ALK7 +

    0

    0.005

    0.01

    0.015

    0.02

    0.025

    0.03

    0.035

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    Ratio epiWAT/BW

    ####

    ####

    #######

    ALK7 + ALK7 + ALK7 +

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    0

    1

    2

    3

    4

    5

    6

    7

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    0

    1

    2

    3

    4

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    mRNA expression in epiWATB

    Rel

    ativ

    e ex

    pres

    sion

    #

    #

    #####

    CEBP

    ###

    ###

    ###

    ###

    ######

    ATGL HSL

    ######

    NEFA/epiWAT(mM/g)

    #

    +ALK7 +

    A

    ##

    ###

    PPAR

    + + +ALK7 ALK7 ALK7 ALK7

    Rel

    ativ

    e ex

    pres

    sion

    ###

    #####

    GDF3 mRNA in SVF

    #

    ALK7 +

  • 0

    5

    10GDF3 mRNA in TSOD-

    CD11c- ATM

    00.5

    11.5

    22.5

    33.5

    4

    - 61pM

    25nM

    - 61pM

    25nM

    - 61pM

    25nM

    - 61pM

    25nM

    TSOD T.B-Nidd5/3

    TSOD T.B-Nidd5/3

    Basal Isoproterenol 10 μM

    # #

    0

    1

    2

    3

    4

    Gene induction by ex vivo Insulin

    ***

    0

    0.2

    0.4

    0.6

    0.8

    1

    InsulinWortmannin

    (nM) 100 1000

    A

    Rel

    ativ

    e ex

    pres

    sion

    + + +

    CD11c GDF3 TNF MCP1 Arg1 Ym1

    Rel

    ativ

    e ex

    pres

    sion

    Rel

    ativ

    e gl

    ycer

    ol re

    leas

    eCB

    p-AKT

    β-actin

    Insulin

    TSOD T.B-Nidd5/3

    Primary adipocyte

    AKT

    61pM

    p-Smad3total-Smad3

    25nM

    ALK7 +

    TSOD

    +

    GDF3

    400ng/ml

    61pM

    25nM

    * **

    **

    ## #####

    Insulin

    # #

    #

    ###

    D

    p-Smad3

    -actin

    total-Smad3

    CD11c-ATM

    Insulin

    ++

    Sups

    E

    Sups

    Rel

    ativ

    e gl

    ycer

    ol re

    leas

    e

    CD11c-ATM

    Insulin

    +

    +

    +

    TSOD T.B-Nidd5/3###

    ## #

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    GDF3 in the sups

    0

    500

    1000

    1500

    2000

    pg/m

    L

    TSOD adipocyte

    #

    ###

    ###

    Figure 4

    ###

    +

    Insulin + +

    TSOD CD11c- ATM

    Wortmannin (nM)

    100

    p-AKT

    AKT

    β-actin

    CD11c-ATM

    Insulin

    ++

    Sups

    +

    ++

    +

  • 00.20.40.60.8

    11.21.41.61.8

    0

    0.5

    1

    1.5

    0

    0.01

    0.02

    0.03

    0

    0.5

    1

    1.5

    00.20.40.60.8

    11.21.41.6

    00.20.40.60.8

    11.2

    ARatio

    epiWAT/BWATGL mRNA

    in epiWATHSL mRNA in epiWAT

    NEFA/epiWAT(mM/g)

    B

    00.0050.01

    0.0150.02

    0.0250.03

    0.035

    0

    0.5

    1

    1.5

    0

    0.5

    1

    1.5

    Rel

    ativ

    e ex

    pres

    sion

    0

    1

    2

    3

    C57BL/6: ALK7 (+)

    BALB/c: ALK7 (-)

    0

    0.01

    0.02

    0.03

    0

    0.5

    1

    1.5

    0

    0.5

    1

    1.5

    2

    0

    1

    2

    3

    4

    Rel

    ativ

    e ex

    pres

    sion

    *

    ** *

    TSOD: ALK7 (+)

    T.B-Nidd5/3: ALK7 (-)

    0

    0.01

    0.02

    0.03

    0.04

    Insulin

    ***

    Rel

    ativ

    e ex

    pres

    sion

    *

    0

    0.5

    1

    1.5

    *

    Insulin

    Rel

    ativ

    e ex

    pres

    sion

    Figure 5

    ++++

    Ratio epiWAT/BW

    ATGL mRNA in epiWAT

    HSL mRNA in epiWAT

    NEFA/epiWAT(mM/g)

    ++++

    Ratio epiWAT/BW

    ATGL mRNA in epiWAT

    HSL mRNA in epiWAT

    NEFA/epiWAT(mM/g)

    Insulin ++++

    Ratio epiWAT/BW

    ATGL mRNA in epiWAT

    HSL mRNA in epiWAT

    NEFA/epiWAT(mM/g)

    Insulin ++++

  • 0

    0.5

    1

    1.5

    2

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6

    0

    0.005

    0.01

    0.015

    0.02

    0.025

    0.03

    0.035

    0

    1

    2

    3

    4

    5

    6

    B

    BM

    Insulin

    WT WT KO KO

    0

    0.5

    1

    1.5

    2

    2.5

    3

    3.5

    0

    0.5

    1

    1.5

    2

    2.5

    A

    CLO

    Insulin ++

    +

    +

    Rela

    tive e

    xpre

    ssio

    nGDF3 mRNA

    in SVF

    epiWAT/BW

    ratio

    NEFA/epiWAT

    (mM/g)

    ATGL mRNA

    in epiWAT

    HSL mRNA

    in epiWAT

    #

    #

    ###

    ## ###

    0

    0.5

    1

    1.5

    2

    2.5

    ##

    ##

    ######

    ###

    #

    ###

    ###

    Rela

    tive e

    xpre

    ssio

    n

    ###

    Figure 6

    Insulin

    Lipolysis

    Obesity

    Insulin

    Resistance

    CD11c+ATMIR

    CD11c-ATM

    GDF3

    Inflammation

    ALK7Adipocyte

    Fat Accumulation

    C

    +

    +

    +

    +

    ++

    +

    +

    +

    +

    +

    ++

    +

    +

    +

    GDF3 mRNA

    in SVF

    epiWAT/BW

    ratio

    NEFA/epiWAT

    (mM/g)

    ATGL mRNA

    in epiWAT

    HSL mRNA

    in epiWAT

    ### ###

    ###

    Rela

    tive e

    xpre

    ssio

    n

    + +WT WT KO KO

    + +WT WT KO KO

    + +

    ##

    Rela

    tive e

    xpre

    ssio

    nWT WT KO KO

    + +

    #

    ##

    ###

    WT WT KO KO

    + +

    ##

    ###

    ###

    0

    0.5

    1

    1.5

    2

    2.5

    0

    0.005

    0.01

    0.015

    0.02

    0.025

    0.03

    0.035

    0.04

    0.045

    0

    0.5

    1

    1.5

    2

    2.5

    3

  • 0

    10

    20

    30

    40

    50

    60

    70

    80

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF5

    0

    50

    100

    150

    200

    250

    300

    350

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF2 (BMP9)

    0

    500

    1000

    1500

    2000

    2500

    3000

    3500

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF6 (BMP13)

    0

    200

    400

    600

    800

    1000

    1200

    1400

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF1

    *

    B6-HFD

    B6-NC

    T.B-Nidd5/3-NC

    BALB-HFD

    BALB-NC

    TSOD-NC

    ALK7(+)

    ALK7(-)

    Supplemental Figure 1-1 (to be continued)

    Rela

    tive e

    xpre

    ssio

    n

    0

    200

    400

    600

    800

    1000

    1200

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF7 (BMP12)

    0

    100

    200

    300

    400

    500

    600

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF8

    0

    1

    2

    3

    4

    5

    6

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF9

    0

    10

    20

    30

    40

    50

    60

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF9b (BMP15)

    * *

    **

    * *

    * **

    *

    *

  • 0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6

    1.8

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF10 (BMP3b)

    *

    0

    5

    10

    15

    20

    25

    30

    35

    40

    45

    50

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF11 (BMP11)

    B6-HFD

    B6-NC

    T.B-Nidd5/3-NC

    BALB-HFD

    BALB-NC

    TSOD-NC

    ALK7(+)

    ALK7(-)

    0

    20

    40

    60

    80

    100

    120

    140

    160

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    GDF15

    0

    2

    4

    6

    8

    10

    12

    14

    16

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    BMP2

    0

    1

    2

    3

    4

    5

    6

    7

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    BMP4

    0

    20

    40

    60

    80

    100

    120

    140

    160

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    BMP5

    0

    5

    10

    15

    20

    25

    30

    35

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    BMP6

    0

    50

    100

    150

    200

    250

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    BMP7

    Rela

    tive e

    xpre

    ssio

    n

    Supplemental Figure 1-2 (to be continued)

    *

    *

    *

    *

  • 0

    10

    20

    30

    40

    50

    60

    70

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    BMP8A

    0

    1000

    2000

    3000

    4000

    5000

    6000

    7000

    8000

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    Inhibin-βE

    0

    500

    1000

    1500

    2000

    2500

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    BMP8BR

    ela

    tive e

    xpre

    ssio

    n

    B6-HFD

    B6-NC

    T.B-Nidd5/3-NC

    BALB-HFD

    BALB-NC

    TSOD-NC

    ALK7(+)

    ALK7(-)

    0

    100

    200

    300

    400

    500

    600

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    BMP10

    0

    1000

    2000

    3000

    4000

    5000

    6000

    7000

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    Inhibin-α

    0

    10

    20

    30

    40

    50

    60

    70

    80

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    Inhibin-βA

    0

    20000

    40000

    60000

    80000

    100000

    120000

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    Inhibin-βC

    0

    2

    4

    6

    8

    10

    12

    14

    16

    18

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    TGF-β2

    Supplemental Figure 1-3 (to be continued)

    *

    * * **

    **

    *

  • 0

    50

    100

    150

    200

    250

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    Nodal

    0

    0.5

    1

    1.5

    2

    2.5

    3

    3.5

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    TGF-β3R

    ela

    tive e

    xpre

    ssio

    n

    B6-HFD

    B6-NC

    T.B-Nidd5/3-NC

    BALB-HFD

    BALB-NC

    TSOD-NC

    ALK7(+)

    ALK7(-)

    0

    200

    400

    600

    800

    1000

    1200

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    AMH

    0

    500

    1000

    1500

    2000

    2500

    3000

    3500

    4000

    4500

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    Lefty1

    0

    1000

    2000

    3000

    4000

    5000

    6000

    7000

    8000

    9000

    10000

    Brain Thymus Liver Spleen BAT epiWAT ingWAT BM Muscle

    Lefty2

    Supplemental Figure 1-4

    *

    * *

    *

  • Figure S2

    B

    A

    Rela

    tive e

    xpre

    ssio

    n

    0

    0.5

    1

    1.5

    2

    2.5

    3

    3.5

    4

    TSOD T.B-Nidd5/3

    HSL

    **

    Rela

    tive e

    xpre

    ssio

    n

    Rela

    tive e

    xpre

    ssio

    n

    Rela

    tive e

    xpre

    ssio

    n

    0

    10

    20

    30

    40

    50

    TSOD T.B-Nidd5/3

    BW(g)

    0

    0.5

    1

    1.5

    2

    TSOD T.B-Nidd5/3

    epiWAT weight (g)

    ***

    0

    0.005

    0.01

    0.015

    0.02

    0.025

    0.03

    0.035

    0.04

    0.045

    TSOD T.B-Nidd5/3

    Ratio epiWAT/BW

    ***

    0

    0.5

    1

    1.5

    2

    TSOD T.B-Nidd5/3

    NEFA (mM)

    **

    0

    0.5

    1

    1.5

    2

    2.5

    TSOD T.B-Nidd5/3

    NEFA/epiWAT(mM/g)

    **

    0

    1

    2

    3

    4

    5

    6

    7

    8

    TSOD T.B-Nidd5/3

    PPARg

    *

    0

    1

    2

    3

    4

    5

    6

    7

    8

    TSOD T.B-Nidd5/3

    CEBPa

    *

    0

    0.5

    1

    1.5

    2

    2.5

    3

    3.5

    4

    4.5

    TSOD T.B-Nidd5/3

    ATGL

    **

    mRNA in epiWAT

    0

    1

    2

    3

    4

    5

    6

    7

    8

    TSOD T.B-Nidd5/3

    Food Intake(g/day)

    CD11c

    TSOD ATM

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6

    CD11c- CD11c+

    *

    D

    0

    0.1

    0.2

    0.3

    0.4

    0.5

    0.6

    0.7

    0.8

    epiWAT

    weight (g)

    **

    0

    0.005

    0.01

    0.015

    0.02

    0.025

    0.03

    0.035

    Ratio

    epiWAT/BW

    ***

    mRNA in SVF

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    C57BL/6 (3 wks NC vs HFD)

    TNFa

    *

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6MCP1

    0

    1

    2

    3

    4

    5

    Arg1

    0

    1

    2

    3

    4

    5

    6

    *

    Ym1

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    CD11c- CD11c+

    TNFa

    Rela

    tive e

    xpre

    ssio

    n

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    CD11c- CD11c+

    MCP1

    mRNA

    0

    0.5

    1

    1.5

    2

    2.5

    CD11c- CD11c+

    Arg1

    0

    0.5

    1

    1.5

    2

    2.5

    CD11c- CD11c+

    Ym1

    0

    0.4

    0.8

    1.2

    1.6

    NLRP3

    0

    0.5

    1

    1.5

    2

    CAS1

    RC HFD RC HFD RC HFD RC HFD RC HFD RC HFD RC HFD RC HFD

    0

    5

    10

    15

    20

    25

    0

    1

    2

    3

    4

    5

    6

    GDF3 CD11c

    Rela

    tive e

    xpre

    ssio

    n

    ***

    RC HFDRC HFD

    CD11b-PE-Cy7

    F4

    /80

    -AP

    C

    CD

    11

    c-P

    E

    CD11b-PE-Cy7

    CD11b+

    F4/80+ ATM

    73.7%

    CD11c+

    ATM

    17.8%

    C

    0

    0.5

    1

    1.5

    2

    2.5

    ep

    iWA

    T

    ad

    ipocyte

    SV

    F

    CD

    11

    b-

    CD

    11

    b+

    F4/8

    0-

    F4/8

    0+

    CD

    11

    c-

    CD

    11

    c+

    SVF CD11b+ CD11b+F4/80+

    BMP3 mRNA

    Rela

    tive e

    xpre

    ssio

    n

    ATM

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6

    1.8

    2

    epiWAT

    adipocyte

    SVF

    CD11b-

    CD11b+

    F4/80-

    F4/80+

    CD11c-

    CD11c+

    SVF CD11b+ CD11b+

    F4/80+

    Inhibin-bB mRNA

    ATM

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6

    1.8

    2

    epiWAT

    adipocyte

    SVF

    CD11b-

    CD11b+

    F4/80-

    F4/80+

    CD11c-

    CD11c+

    SVF CD11b+ CD11b+

    F4/80+

    TGF-b1 mRNA

    ATM

    ##

    ###

    ###

    ###

    ###

    ###

    ###

    ###

  • 0

    10

    20

    30

    40

    50

    60

    70

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    % of ATM in SVF

    ###

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    0

    0.5

    1

    1.5

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    Figure S3

    0

    1

    2

    3

    4

    5

    6

    7

    8

    9

    10

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    % of CD11c+ ATM in SVF

    0

    2

    4

    6

    8

    10

    12

    14

    16

    PBS CLO PBS CLO

    TSOD T.B-Nidd5/3

    % of CD11c+ in ATM

    ##

    ##

    ##

    A

    B

    ALK7 +

    ALK7 + +

    Rela

    tive

    exp

    ressio

    n

    Rela

    tive

    exp

    ressio

    n

    ATM

    56.3%

    CD11c+

    11.49%

    F4

    /80

    -AP

    C

    CD11b-

    PE-Cy7

    PBS

    CD

    11

    c-P

    E

    CD11b-

    PE-Cy7

    CLO

    ATM

    35.26%

    CD11c+

    4.35%

    T.B-Nidd5/3

    ATM

    58.49%

    ATM

    40.84%

    CD11c+

    6.57%

    CD11c+

    2.8%

    ###

    ++

    TSOD

    PBS CLO

    ####

    ##

    ###

    F4/80 mRNA

    in epiWAT

    # #

    #

    #

    CD11c mRNA

    in epiWAT

    #

    ##

  • 0

    1

    2

    3

    4

    0

    0.5

    1

    1.5

    2

    Lung.MΦ P.MΦ BMDM

    Figure S4

    A

    Rela

    tive e

    xpre

    ssio

    n

    B

    TSODR

    ela

    tive e

    xpre

    ssio

    n

    CD11c- ATM

    C

    Rela

    tive e

    xpre

    ssio

    n

    Gene induction

    by 61 pM insulin

    0

    0.5

    1

    1.5

    Inhibin-bB TGF-b1

    TSOD

    CD11c- ATM

    TSOD

    GDF3 mRNA induced

    by 61 pM insulin

    TSODTSOD

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    CD11c-ATM

    Lung.MΦ P.MΦ BMDM

    IR mRNA

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6

    CD11c-ATM

    Lung.MΦ P.MΦ BMDM

    GDF3 mRNA (after 24 h culture without insulin)

    no

    culture

    24 h culture

    FBS(-) FBS(+)

    GDF3 mRNA

    ###

    ##

    #

    ####

    ###

    ###

  • 0

    1

    2

    3

    Figure S5

    C

    0

    10

    20

    30

    40

    50

    60

    70

    % of ATM in SVF

    BM

    Insulin + +WT WT KO KO

    + +WT WT KO KO

    0

    1

    2

    3

    4

    numbers of SVF cells

    (x106 cells/g)

    B

    ++

    +

    ++

    ++

    +0

    10

    20

    30

    40

    50

    60

    70

    % of ATM in SVF

    ##

    ###

    0

    1

    2

    3

    4

    5

    % of CD11c+ ATM in SVF

    ##

    ###

    Insulin

    CLO

    ######

    ++

    +

    +0

    1

    2

    3

    4

    5

    Food intake (g/day)

    ATSOD T.B-Nidd5/3 C57BL/6 BALB/c

    ALK7

    b-actin

    Rela

    tive e

    xpre

    ssio

    n

    ++

    +

    +

    CD11c mRNA

    in SVF

    ##

    ### ###

    Diabetes Figure 1 New (2 column) 20180406Diabetes Figure 2 New (2 column) 20180406Diabetes Figure 3 New (2 column) 20180406Diabetes Figure 4 New (2 column) 20180406Diabetes Figure 5 New (2 column) 20180406Diabetes Figure 6 New (2 column) 20180406-newDiabetes Supplemental Figures N 20180406-new