60
1 SGK1/FOXO3 signaling in hypothalamic POMC neurons mediates glucocorticoid-increased adiposity Yalan Deng 1 , Yuzhong Xiao 1 , Feixiang Yuan 1 , Yaping Liu 2 , Xiaoxue Jiang 1 , Jiali Deng 1 , Geza Fejes-Toth 3 , Aniko Naray-Fejes-Toth 3 , Shanghai Chen 1 , Yan Chen 1 , Hao Ying 1 , Qiwei Zhai 1 , Yousheng Shu #2 and Feifan Guo #1 1 Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, China 200031 2 State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, Beijing Normal University, Beijing, China 100875 3 Department of Physiology, Dartmouth Medical School, Lebanon, New Hampshire 03756-0001, USA Contact Information # Correspondence should be addressed to Feifan Guo; E-mail: [email protected]; Address: 320 Yueyang Road, Shanghai, China 200031 and Yousheng Shu; E-mail: [email protected]; Address: Beijing Normal University, Beijing, China 100875. E-mail: [email protected] Tel: 86 21 54920945; Fax: 86 21 54920291. [email protected] Tel: 86 10 58804976; Fax: 86 10 58804976. Page 1 of 60 Diabetes Diabetes Publish Ahead of Print, published online January 10, 2018

Page 1 of 60 Diabetes · 2018. 1. 8. · SGK1 allele (SGK1loxp/loxp) mice (18) were kindly provided by Dr Geza Fejes-Toth and Dr Aniko Naray-Fejes-Toth (Dartmouth Medical School,

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

  • 1

    SGK1/FOXO3 signaling in hypothalamic POMC neurons

    mediates glucocorticoid-increased adiposity

    Yalan Deng1, Yuzhong Xiao

    1, Feixiang Yuan

    1, Yaping Liu

    2, Xiaoxue Jiang

    1,

    Jiali Deng1, Geza Fejes-Toth

    3, Aniko Naray-Fejes-Toth

    3, Shanghai Chen

    1, Yan Chen

    1,

    Hao Ying1, Qiwei Zhai

    1, Yousheng Shu

    #2 and Feifan Guo

    #1

    1 Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences,

    Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, University

    of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, China 200031

    2 State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern

    Institute for Brain Research, School of Brain and Cognitive Sciences, Beijing Normal

    University, Beijing, China 100875

    3Department of Physiology, Dartmouth Medical School, Lebanon, New Hampshire

    03756-0001, USA

    Contact Information

    # Correspondence should be addressed to Feifan Guo; E-mail: [email protected];

    Address: 320 Yueyang Road, Shanghai, China 200031 and Yousheng Shu; E-mail:

    [email protected]; Address: Beijing Normal University, Beijing, China 100875.

    E-mail: [email protected] Tel: 86 21 54920945; Fax: 86 21 54920291.

    [email protected] Tel: 86 10 58804976; Fax: 86 10 58804976.

    Page 1 of 60 Diabetes

    Diabetes Publish Ahead of Print, published online January 10, 2018

  • 2

    Running title: POMC SGK1/FOXO3 signaling regulates adiposity.

    The word count: 4161 The number of tables and figures: 0 and 6

    ABSTRACT

    Although central nervous system has been implicated in glucocorticoid-induced

    fat mass gain, the underlying mechanisms are poorly understood. The aim of our

    current study was to investigate the possible involvement of hypothalamic serum- and

    glucocorticoid-regulated kinase 1 (SGK1) in glucocorticoid-increased adiposity. It is

    well-known that SGK1 expression is induced by acute glucocorticoid treatment,

    interestingly, we found its expression was decreased in the arcuate nucleus of the

    hypothalamus, including POMC neurons, following chronic dexamethasone (Dex)

    treatment. To study a role of SGK1 in POMC neurons, mice with development or

    adult-onset SGK1 deletion in POMC neurons (PSKO) were then produced. As

    observed in Dex-treated mice, PSKO mice exhibited increased adiposity and

    decreased energy expenditure. Consistently, mice overexpressing constitutively active

    SGK1 in POMC neurons (PSOE) had the opposite phenotype and prevented from

    Dex-increased adiposity. Finally, Dex decreased hypothalamic α-melanocyte

    stimulating hormone (α-MSH) content and its precursor Pomc expression via

    SGK1/Forkhead box O3 (FOXO3) signaling and intracerebroventricular injection of

    α-MSH or adenovirus-mediated FOXO3 knockdown in ARC largely reversed the

    metabolic alterations in PSKO mice. These results demonstrate that POMC

    SGK1/FOXO3 signaling mediates glucocorticoid-increased adiposity, providing new

    Page 2 of 60Diabetes

  • 3

    insights into mechanistic link between glucocorticoid and fat accumulation and

    important hints for possible treatment targets for obesity.

    INTRODUCTION

    In addition to the overwhelming beneficial effects of glucocorticoid for

    anti-inflammatory purposes, chronic glucocorticoid treatment is shown to cause

    numerous adverse metabolic outcomes, including fat mass gain (1). Recent studies

    have elucidated several peripheral mechanisms underlying glucocorticoid-induced fat

    mass increase. For example, glucocorticoid induces adipocyte differentiation (1-3),

    alters lipid metabolism in adipose tissue (1-3) and inhibits browning of white adipose

    tissue (WAT) or thermogenesis of brown adipose tissue (BAT) (4; 5). In fact, body fat

    mass is also largely controlled by the central nervous system (CNS) (6-8). Specific

    populations of neurons in the arcuate nucleus (ARC) of hypothalamus also play

    fundamental roles in the regulation of energy balance and lipid metabolism (6-8). In

    particular, neurons coexpressing orexigenic neuropeptides agouti-related protein

    (AgRP) and neuropeptide Y (NPY) along with neurons coexpressing anorexigenic

    pro-opiomelanocortin (POMC) precursor and cocaine and amphetamine-related

    transcript (CART) are extensively involved in the regulation of appetite, body weight

    and metabolism (6-8). POMC is a protein expressed and secreted from POMC

    neurons and cleaved by prohormone convertases to produce α-melanocyte stimulating

    hormone (α-MSH) (8). α-MSH binds to the melanocortin 4 receptor (MC4R) and

    functions as a key hub linking the CNS to peripheral organs through the sympathetic

    Page 3 of 60 Diabetes

  • 4

    nervous system (SNS), whereas dysfunction of this signaling axis leads to obesity in

    mice and humans (9; 10). Activation of SNS promotes the release of norepinephrine

    (NE) that binds to β-adrenergic receptor 3 (ADRB3) and stimulates WAT lipolysis

    and BAT thermogenesis (11-14). Although previous studies have shown

    glucocorticoid regulates food intake and energy expenditure (15; 16), the central

    signals mediating glucocorticoid’s effect are poorly understood.

    Serum- and Glucocorticoid-regulated Kinase 1 (SGK1) belongs to the family of

    serine/threonine kinases and its coding region was originally isolated from rat

    mammary tumour cells (17). SGK1 is ubiquitously expressed in various tissues,

    including hypothalamus (17) and functions via activation of glucocorticoid receptor

    (GR), retinoid X receptor (RXR), peroxisomeproliferator-activated receptor γ (PPARγ)

    and nuclear factor κB (NF-κB) (17). It has been shown that SGK1 is involved in the

    regulation of many processes, including hypertension, epithelial sodium channel

    activity and insulin sensitivity (17-19). SGK1 also mediates many important functions

    of glucocorticoids, including insulin secretion and hippocampal neurogenesis (20; 21).

    Although extensive studies have been carried out, a role of hypothalamic SGK1 in the

    regulation of energy homeostasis is unknown. Furthermore, it is well-known that

    SGK1 is an early response gene that can be induced by acute glucocorticoid treatment

    in various cells and animal models (20-22), however, the effect of chronic

    glucocorticoid treatment on SGK1 expression remains largely unknown. In fact, the

    expression of SGK1 in the context of glucocorticoid-induced metabolic effects could

    be very important, as studies show that sometimes SGK1 may have opposing effects

    Page 4 of 60Diabetes

  • 5

    of glucocorticoid (23).

    Despite the above unknown facts, as a downstream target of glucocorticoid (17)

    expressed in the hypothalamus (17), it is conceivable to speculate that SGK1 may

    contribute to the central action of glucocorticoid. Therefore, the aim of our current

    study was to test this hypothesis first by determining the expression of SGK1 in the

    hypothalamus and followed by investigating its possible contribution to

    glucocorticoid-increased adiposity.

    By constructing mice with development or adult-onset knockout of SGK1, or

    over-expression of SGK1 in POMC neurons, we demonstrate a crucial role for SGK1

    expressed in POMC neurons in glucocorticoid-increased adiposity and provide a

    novel mechanistic link between glucocorticoid treatment and body fat mass gain.

    RESEARCH DESIGN AND METHODS

    Mice and diets

    The POMC-Cre mice (24) and POMC-cre:ERT2

    (24) were kindly provided by

    Prof. Joel K. Elmquist and Tiemin Liu from Southwestern Medical Center, and floxed

    SGK1 allele (SGK1loxp/loxp

    ) mice (18) were kindly provided by Dr Geza Fejes-Toth

    and Dr Aniko Naray-Fejes-Toth (Dartmouth Medical School, Hanover, NH, USA).

    To generate POMC neuron-specific SGK1 knockout mice, POMC-Cre mice were

    crossed with SGK1loxp/loxp

    mice. To generate an inducible POMC specific SGK1

    knockout mice, POMC-cre:ERT2

    mice were crossed with SGK1loxp/loxp

    mice.

    Tamoxifen (0.15 g/kg; Sigma, MO, USA) suspended in corn oil (Sigma, MO, USA)

    Page 5 of 60 Diabetes

  • 6

    was intraperitoneally (i.p.) injected to 8-week-old male SGK1loxp/loxp

    or SGK1loxp/loxp

    ×

    POMC-cre:ERT2

    littermate mice for 5 consecutive days to generate mice with adult

    onset of SGK1 deletion in POMC neurons (PSKO-ER). Dexamethasone (Dex)

    treatment were conducted with male WT, control or PSKO mice, or male AAV-CA

    SGK1/AAV-null ARC injection POMC-cre mice by i.p. injection with PBS or 5

    mg/kg Dex every other day for 6 weeks or 2 hours (1; 25). All the mice were in

    C57BL/6J background. Mice were maintained on a 12:12 hr light-dark cycle (lights

    on 7:00/off 19:00) at 25 ℃ with free access to water and standard chow diet. In vivo

    studies were conducted in accordance with the guidelines of the Institutional Animal

    Care and Use Committee of Shanghai Institute for Nutritional Sciences, Chinese

    Academy of Sciences.

    Intracerebroventricular (i.c.v.) cannulation and ARC administration

    experiments

    I.c.v. cannulation experiments were conducted as previously described (26). Five

    days after recovery, mice were infused with 2 ul of α-MSH peptide (Abcam,

    Cambridge, UK) at a concentration of 1 nmol/uL or 2ul artificial cerebrospinal fluid

    (ACSF) (Tocris, Bristol, UK) and experiments were conducted 24 h later. ARC

    administration experiments were conducted as previously described (6). Mice were

    anesthetized and received bilateral stereotaxic injections of adenovirus expressing

    Forkhead box O3 (FOXO3)-specific short hairpin RNA against FOXO3

    (Ad-shFOXO3) or scrambled control adenovirus (Ad-scrambled), adeno-associated

    Page 6 of 60Diabetes

  • 7

    virus expressing constitutively active mutant rat SGK1 (S422D) (AAV-CA SGK1) or

    control AAV-null into ARC (1.5 mm posterior to the bregma, ± 0.2 mm lateral to

    midline and 6 mm below the surface of the skull). AAV-CA SGK1 expression

    plasmid was constructed in pAAV-Ef1a-DIO-mCherry-2A plasmid (Addgene, MA,

    USA), and SGK1 started to express only in the presence of CRE recombinase.

    Metabolic parameter measurements

    The mice body composition was measured by a nuclear magnetic resonance

    system (Bruker, Rheinstetten, GER). Indirect calorimetry was performed in a

    comprehensive laboratory animal-monitoring system (Columbus Instruments, OH,

    USA), as previously described (27). Rectal temperature of mice was measured at

    14:00 and 17:00 by a rectal probe attached to a digital thermometer (Physitemp,

    Clifton, NJ). The measurement of food intake was conducted as reported previously

    (6).

    POMC neuron identification, count and area

    AI9 (tdTomato) reporter mice (Jackson Laboratory) were mated with or

    AAV-CA SGK1 and AAV-null expressed mCherry red fluorescent-protein were ARC

    injected to POMC-Cre mice to reflect POMC neurons, demonstrated by the

    colocolization with POMC antibodies. The distribution and number of POMC

    neurons were determined as described previously (6). Average somatic area was

    analyzed in > 500 POMC neurons (n = 4 mice/genotype). The area occupied by

    Page 7 of 60 Diabetes

  • 8

    POMC neurons was manually scored using Image J software.

    Hypothalamic α-MSH protein content

    Hypothalamus was prepared as previously described (6) and α-MSH were

    quantified by ELISA kit (Phoenix Pharmaceuticals, CA, USA), according to

    manufacture’s instructions.

    Hypothalamic nuclear and cytoplasmic fractions

    Hypothalamic nuclear and cytoplasmic fractions were isolated as previously

    described (28).

    Immunofluorescence staining

    Immunofluorescence stainings were performed as previously described (29) with

    anti-SGK1 and anti-p-N-myc downstream-regulated gene 1 (p-NDRG1) (Abcam,

    Cambridge, UK), anti-POMC (Phoenix pharmaceuticals, inc, CA, USA), anti-FOXO3

    (Cell Signaling Technology, MA, USA), anti-p-SGK1 and anti-GR (Santa Cruz

    Biotechnology, CA, USA) and anti-α-MSH (Merck Millipore, Frankfurter, GER).

    Immunofluorescence staining of p-FOXO3 was performed using the Tyramide Signal

    Amplifcation (TSA) Cyanine 3 system (Perkin-Elmer, Boston, MA), and

    anti-p-FOXO3 primary antibody (Cell Signaling Technology, MA, USA) was

    co-incubated with anti-mCherry (Abbkine, California, USA).

    Page 8 of 60Diabetes

  • 9

    RNA isolation and relative quantitative RT-PCR

    RNA isolation and RT-PCR were performed as previously described (27). The

    sequences of primers used in this study are available upon request.

    Western blot analysis

    Western blot analysis was performed as previously described (27) with the

    following primary antibodies, anti-p-FOXO3, anti-FOXO3, anti-lamin B1 and

    anti-p-GR (Cell Signaling Technology, MA, USA), anti-SGK1 and anti-GR (Abcam,

    Cambridge, UK), anti-uncoupling protein 1 (UCP1) and anti-p-SGK1 (Santa Cruz

    Biotechnology, CA, USA), anti-α-tublin and anti-β-actin (Sigma, MO, USA).

    Primary hypothalamic neurons isolation and treatments

    Primary cultures of hypothalamic neurons were prepared as previously

    described (27). On day 7, primary cultured hypothalamic neurons were infected with

    adenovirus expressing SGK1-specific short hairpin RNA (Ad-shSGK1; 108 pfu/60

    cm2 cells) or scrambled control adenovirus, constructed as described previously (19).

    Primary hypothalamic neurons were transfected with siRNA for FOXO3 by

    X-tremeGene siRNA Transfection Reagent (Roche Diagnostics, Mannheim,

    Germany). Constitutively active mutant rat SGK1 (S422D) was subcloned into

    PCMV-MYC plasmid and transfected into primary cultured hypothalamic neurons

    using Lipofectamine 2000 (Life Technologies).

    Page 9 of 60 Diabetes

  • 10

    Statistical analysis

    All values are presented as means ± SEM. Differences between groups were

    analyzed by either Student t test or one-way ANOVA followed by the

    Student-Newman-Keuls (SNK) test. Differences in which P was < 0.05 were

    considered statistically significant.

    RESULTS

    Chronic Dex treatment decreases SGK1 expression in hypothalamic POMC

    neurons

    To investigate the metabolic effects of Dex, C57B6J wild-type (WT) mice were

    i.p. injected with Dex for 6 weeks, the way of which has been commonly used to

    study the role of Dex (1; 30). Dex treatment did not change body weight, though the

    total body fat and abdominal fat mass was increased compared with control treatment,

    possibly due to the decreased lean mass (Fig. S1A-D). Body fat mass is maintained by

    a balance between energy intake and energy expenditure (7). Dex treatment did not

    change food intake, but decreased energy expenditure as measured by 24-h-indirect

    calorimetry (Fig. S1E and S1F). No difference was observed in locomotor activity,

    but the body temperature, levels of BAT thermogenic marker UCP1 (11) and serum

    NE levels were significantly lower in Dex-treated mice (Fig. S1G-J).

    To investigate the possible involvement of hypothalamic SGK1 in Dex-increased

    adiposity, we examined hypothalamic SGK1 expression under this condition and

    interestingly found that hypothalamic SGK1 and p-SGK1 were decreased in

    Page 10 of 60Diabetes

  • 11

    Dex-treated mice (Fig. 1A and 1B). Furthermore, immunofluorescence staining

    showed that SGK1 and p-SGK1 was decreased in ARC of the hypothalamus of

    Dex-treated mice (Fig. 1C-F). Immunofluorescence staining of tdTomato and SGK1

    showed that SGK1 was colocalized with POMC neurons in PBS-treated mice, but

    decreased significantly in POMC neurons of Dex-treated mice (Fig. 1G and 1H). In

    contrast, SGK1 expression was increased in ARC of the hypothalamus by acute

    treatment (Fig. 1I-L).

    Deletion of SGK1 in POMC neurons causes obesity and decreases energy

    expenditure

    Based on the above results, we speculated that knockout of SGK1 expression in

    POMC neurons might mimic Dex-induced metabolic alterations. To test this

    hypothesis, we generated POMC neuron-specific SGK1 knockout (PSKO) mice.

    Immunofluorescence staining of tdTomato and SGK1 showed that SGK1 was

    colocalized with POMC neurons (more than 90 % overlapping with tdTomato) in

    control mice, but almost absent in POMC neurons of PSKO mice (Fig. 2A and

    S2A-C), with no difference in SGK1 staining in PVN and VMH between PSKO and

    control mice (Fig. S2D-G). Consistently, Sgk1 mRNA levels were decreased about

    50 % in ARC, as there are other neurons or neurogliocytes (31; 32), but not other

    brain areas and tissues, of PSKO mice (Fig. 2B). POMC neuron differentiation and

    survival, however, as anatomical assessment of POMC neurons throughout the ARC

    area revealed no significant alterations in neuronal population size, distribution and

    Page 11 of 60 Diabetes

  • 12

    somatic area in PSKO mice (Fig. S3A and S3B). Because POMC promoter also

    drives CRE recombinase expression in the pituitary (33), we examined serum contents

    of hormones secreted from pituitary, including corticosterone and growth hormone

    (34), and found that the levels of these two hormones were not altered in PSKO mice

    (Fig. S3C and S3D).

    Male PSKO mice exhibited a significant increased body weight from the age of

    9-weeks old compared with control mice (Fig. 2C), accompanied by a significant

    increase in total body fat and abdominal fat mass (Fig. 2D and 2E), with unchanged

    lean mass (Fig. S2H). Food intake was not altered, but the energy expenditure was

    markedly decreased and respiratory exchange ratio (RER; VCO2/VO2) was higher in

    PSKO mice (Fig. 2F-H). Again, locomotor activity was not changed, but body

    temperature, BAT UCP1 and serum NE levels were significantly lower in PSKO mice

    (Fig. 2I-L). As observed for male mice, female PSKO mice also displayed similar

    phenotypes (Fig. S4), so we undertook all of the subsequent studies in male mice.

    Inducible loss of SGK1 in POMC neurons in adult mice recapitulates aberrant

    energy homeostasis

    We next asked whether adult-onset loss of SGK1 in POMC neurons had similar

    effects to those of ablation during development. We employed a tamoxifen-inducible

    POMC-cre mouse model (POMC-cre:ERT2

    ) (24) that allows temporal control of CRE

    recombinase activity and can be combined with SGK1flox/flox

    mice producing mice with

    adult-onset deletion of SGK1 (PSKO-ER). Similar phenotypes were observed as

    Page 12 of 60Diabetes

  • 13

    using constitutive POMC-cre mice (Fig. S5).

    Mice with over-expression of SGK1 in POMC neurons are lean and resistant to

    Dex-induced fat accumulation

    We then asked whether over-expression of SGK1 in POMC neurons in mice

    would have the opposite phenotype as observed in PSKO mice and prevented from

    Dex-increased adiposity. For this purpose, we generated POMC neuron-specific

    SGK1 over-expression (PSOE) mice by ARC bilateral stereotaxic injection of

    adeno-associated virus expressing constitutively active mutant rat SGK1 (S422D)

    (AAV-CA SGK1) or control AAV-null to male POMC-Cre mice. The effect of SGK1

    over-expression was validated by immunofluorescence staining of the phosphorylated

    levels of NDRG1 that reflects the activation status of SGK1 (35) (Fig. 3A) and

    increased signals of SGK1 in POMC neurons (more than 90 % overlapping with

    mCherry), but not in PVN and VMH, of PSOE mice (Fig. S6A-H). As predicted, the

    body weight was decreased (starting from 6 weeks after AAV injection), accompanied

    by a decrease in total body fat and abdominal fat mass in PSOE mice (Fig. 3B-D).

    Food intake was not affected, but the energy expenditure was increased and RER was

    decreased in PSOE mice (Fig. 3E-G). No difference was observed in locomotor

    activity, but body temperature, BAT UCP1 and serum NE levels were increased in

    PSOE mice (Fig. 3H-K). Furthermore, PSOE mice were resistant to Dex-induced fat

    accumulation and other metabolic alterations (Fig. 4), with Dex injected 5 weeks after

    AAV injection under no difference in lean mass and fat mass between control and

    Page 13 of 60 Diabetes

  • 14

    PSOE mice (Fig. S6I and S6J). In contrast, Dex had very mild effect on PSKO mice,

    as demonstrated by the slightly decreased body weight and lean mass, increased fat

    mass, with no significant effect on food intake and energy expenditure (Fig. S7).

    Dex decreases hypothalamic αααα-MSH content via SGK1 and administration of

    α-MSH reverse obese phenotype in PSKO mice

    Because previous studies have shown that α-MSH plays a critical role in the

    regulation of energy homeostasis (33), we asked whether it might be involved in

    Dex-induced metabolic alterations. As predicted, a dramatic reduction of α-MSH

    staining was observed in PVN of Dex-treated mice (Fig. 5A and 5B). Similar results

    were obtained in PSKO mice (Fig. 5C and 5D). Consistently, the content of α-MSH

    was significantly decreased in the hypothalamus of PSKO mice when analyzed by

    ELISA (Fig. 5E). Notably, Dex-reduced α-MSH staining was reversed in PSOE mice

    (Fig. 5F and 5G).

    To investigate whether α-MSH could mediate SGK1 regulation of energy

    homeostasis, we i.c.v. administered α-MSH peptide to PSKO or control mice. I.c.v.

    injection of α-MSH to PSKO mice markedly reduced body weight and abdominal fat

    mass and increased rectal temperature compared with mice injected with control

    vehicle (Fig. 5H-J). I.c.v. injection of α-MSH in PSKO mice also blocked UCP1

    protein decrease (Fig. 5K). Similar effects were observed in control mice following

    i.c.v. injection of α-MSH (Fig. 5H-K).

    Page 14 of 60Diabetes

  • 15

    Dex reduces α-MSH precursor POMC expression via SGK1/FOXO3 dependent

    pathway and down-regulation of FOXO3 largely reversed the obesity phenotype

    in PSKO mice

    α-MSH levels are determined by the levels of its precursor POMC, and the

    expression of prohormone convertases that are responsible for the cleavage of POMC

    to α-MSH (8). The reduced α-MSH concentration in Dex-treated mice did not seem to

    be the consequence of decreased expression of processing enzymes, including

    prohormone convertase 1 (Pc1/3), prohormone convertase 2 (Pc2), carboxypeptidase

    E (Cpe), α-amidating monooxygenase (Pam) and prolylcarboxypeptidase (Prcp) (8),

    as gene expression of these enzymes was unchanged (Fig. 6A). On the other hand,

    POMC expression was decreased in Dex-treated mice (Fig. 6A-C). Similar results

    were obtained in PSKO mice (Fig. S8A-C). The effect of Dex on reducing POMC

    expression, however, was reversed by over-expression of SGK1 (Fig 6D and 6E).

    Similarly, SGK1 knockdown decreased Pomc expression and SGK1 over-expression

    increased Pomc expression in primary cultured hypothalamic neurons (Fig. S8D and

    S8E).

    We then investigated the downstream signaling of SGK1 in mediating

    Dex-decreased POMC expression. Previous study shows that SGK1 phosphorylates

    FOXO3 (36), and another member from the same FOXO family FOXO1 inhibits

    Pomc expression (37), suggesting that FOXO3 might have similar function to FOXO1

    as downstream of SGK1 in Dex-induced metabolic alterations. Consistent with this

    possibility, hypothalamic FOXO3 phosphorylation was decreased in Dex-treated mice

    Page 15 of 60 Diabetes

  • 16

    (Fig. 6F). Similar reduction was observed in hypothalamic ARC of PSKO mice (Fig.

    S9A). Furthermore, Dex-decreased hypothalamic FOXO3 phosphorylation was

    reversed in PSOE mice (Fig. 6G and 6H). Similar regulatory effects of SGK1 on

    p-FOXO3 were observed in primary cultures of hypothalamic neurons (Fig. S9B and

    S9C).

    Because the inhibitory effect of SGK1 knockdown on Pomc expression was

    reversed by siRNA-mediated FOXO3 inhibition (Fig. S9D), promoting us to

    investigate the in vivo function of FOXO3 as downstream of SGK1. For this purpose,

    we knocked down FOXO3 expression in ARC of PSKO and control mice by ARC

    administration (6) of adenovirus expressing shRNA directed against the coding region

    of FOXO3 (Ad-shFOXO3) (38) or adenoviruses expressing scrambled sequences

    (Ad-scrambled). The effect of Ad-shFOXO3 was demonstrated by the decreased

    expression of Foxo3 and the corresponding change in Pomc expression in ARC of

    PSKO mice (Fig. 6I). Consistently, immunofluorescence showed that FOXO3 was

    decreased in ARC, but not PVN and VMH, in these mice (Fig. S10A and S10B).

    Ad-shFOXO3 decreased the body weight, total body fat and abdominal fat mass in

    PSKO mice (Fig. 6J-L). Although food intake was not affected (Fig. S10C), the

    decreased energy expenditure and increased RER in PSKO mice were largely

    reversed by Ad-shFOXO3 (Fig. 6M and 6N). No significant difference in locomotor

    activity was detected (Fig. S10D), however, the decreased body temperature, BAT

    UCP1 and serum NE in PSKO mice were upregulated by Ad-shFOXO3 (Fig. 6O-Q).

    Moreover, the reduced α-MSH staining in PSKO mice was also blocked by

    Page 16 of 60Diabetes

  • 17

    Ad-shFOXO3 (Fig. S10E and S10F). Except for the unaltered body weight, similar

    effects were observed in control mice following administration of Ad-shFOXO3 (Fig.

    6I-Q and Fig. S10).

    As glucocortitoid functions via GR (28), we investigated the spatial regulation of

    GR and SGK1/FOXO3, with GR antibodies validated previously (39). Though

    hypothalamic Gr mRNA was unchanged, total GR and phosphorylated GR expression

    were significantly decreased, in Dex-treated mice (Fig. S11A and S11B). Furthermore,

    these three proteins were all expressed in POMC neurons, and hypothalamic nuclear

    p-GR was decreased and FOXO3 was increased, whereas cytoplasmic total and

    phorphorylated proteins examined were all decreased, in Dex-treated mice (Fig. S11C

    and S11D).

    DISCUSSION

    Fat mass accumulation is a serious side effect of glucocorticoid therapy (1).

    Recent studies have elucidated several peripheral mechanisms underlying

    glucocorticoid-induced fat mass gain (1-5). In this study, we demonstrated a novel

    central mechanism mediated by SGK1 underlying glucocorticoid-increased adiposity.

    SGK1 is a well-known downstream target of Dex (20-22). It has been widely

    demonstrated that acute Dex treatment induces SGK1 (20-22). Interestingly, we found

    that SGK1 expression was decreased in ARC POMC neurons of Dex-treated mice.

    The importance of POMC SGK1 in mediating Dex-induced adiposity was

    demonstrated by the observation that knockout of SGK1 expression in POMC

    Page 17 of 60 Diabetes

  • 18

    neurons increased adiposity, while overexpression of SGK1 in POMC neurons

    resulted in lean phenotype and prevented Dex-induced fat mass gain in mice.

    Furthermore, the Dex-induced fat mass gain was much less in PSKO compared with

    control mice. The fat mass, however, could still be increased by Dex treatment in

    PSKO mice, suggesting the existence of other central or peripheral signals involved in

    Dex-increased adiposity (40). Our study provides a novel mechanistic link between

    glucocorticoid treatment and fat mass gain. This is important for understanding the

    mechanisms of glucocorticoid-induced metabolic phenotypes, and also providing

    important hint for the possible treatment target for glucocorticoid-induced side effects.

    In addition, our study reports an unrecognized novel function of SGK1 in POMC

    neurons of the hypothalamus in the regulation of energy homeostasis. These results

    are important for understanding the signals in specific neurons that are critical for

    metabolic control.

    Body fat mass is maintained by a balance between energy intake and energy

    expenditure (7). BAT oxidizes fat to produce heat via increased expression of UCPs,

    which is stimulated by activation of SNS. Deletion of UCP1 induces obesity and

    upregulation of UCP1 increases thermogenesis and energy expenditure in mice (11).

    Consistently, other studies also showed that disruption of SNS activity has significant

    negative impact on energy expenditure (6; 41; 42). Our study showed that Dex

    increased adiposity mainly by decreasing energy expenditure, as food intake was not

    changed in Dex-treated mice. Furthermore, the decreased energy expenditure by Dex

    treatment was most likely due to decreased thermogenesis in BAT as demonstrated by

    Page 18 of 60Diabetes

  • 19

    the decreased body temperature, BAT UCP1 expression and serum NE in these mice.

    Lipolysis in WAT is also regulated by SNS activity (41; 42), which might also affect

    Dex-induced adiposity, and should be studied in the future.

    Extensive evidence indicates that the melanocortin signaling in hypothalamus

    plays an important role in regulating energy homeostasis and lipid metabolism

    through affecting SNS activity in BAT (11-14). In this study, we demonstrated a

    possible role of α-MSH in mediating Dex regulation of adiposity, as α-MSH levels

    were decreased in Dex-treated mice via SGK1 and restoration of hypothalamic

    α-MSH levels by i.c.v. administration of this peptide normalized the inadequate

    energy homeostasis in PSKO mice. Although the beneficial effects of the

    pharmacological treatment are most likely mediated through direct actions on POMC

    neurons, we can not exclude its potential effects on other hypothalamic areas due to

    the delivery route used.

    Our results suggest that the reduced α-MSH content in Dex-treated mice was not

    caused by an altered proteolysis process, but the decreased Pomc expression possibly

    due to glucocorticoid resistance, as Dex is shown to induce Pomc expression (43).

    Furthermore, we found Dex-decreased Pomc expression via SGK1/FOXO3 dependent

    pathway, as the inhibitory effect of Dex on Pomc expression was blocked in mice

    with SGK1 over-expression or FOXO3 inhibition. Many studies, including those

    conducted on FOXO3 knockout mice, have demonstrated that FOXO3 is vital for

    many functions in CNS, including neural stem cell homeostasis, stress and

    Huntington's Disease (44; 45). We showed that it functions as downstream signal of

    Page 19 of 60 Diabetes

  • 20

    SGK1 in the regulation of energy homeostasis. We also demonstrated the spatial

    relationships among GR, SGK1 and FOXO3, providing the basis for the interaction

    and regulation among these proteins.

    In this study, we also demonstrated that adult-onset loss of SGK1 in POMC

    neurons results in a phenotype similar to that of ablation during development. This is

    a key issue, because some works report that multiple hypothalamic neurons express

    POMC in adult mice (24) and pre- and post-natal ablation of certain neurons results in

    disparate feeding behavior, suggesting that phenotypes caused by prenatal ablation

    may be influenced by developmental compensation (24). POMC promoter also drives

    CRE recombinase expression in corticotrophs and melanotrophs (46). The

    contribution from pituitary might not be that significant in the current study, as no

    changes were observed in serum corticosterone and growth hormone content, which

    reflect the function of pituitary (34), between PSKO and control mice.

    Previous studies have shown that POMC neurons are involved in the regulation

    of food intake (29; 47). For reasons unknown, however, we found food intake was not

    significantly affected by Dex treatment, or in PSKO or PSOE mice. Consistent with

    our study, however, previous works also indicate that genetic blockade of

    CNS-MC3R promotes fat accumulation in the absence of hyperphagia (48).

    In contrast to the stimulatory effect of glucocorticoid on SGK1 expression

    (20-22), we observed decreased hypothalamic SGK1 expression following chronic

    Dex treatment, which is to our knowledge a novel observation. We speculate that this

    inhibition is not a direct effect of Dex on SGK1 expression, but rather a consequence

    Page 20 of 60Diabetes

  • 21

    of attenuated Dex-mediated signaling, as it has been previously shown that prolonged

    Dex treatment causes glucocorticoid resistance (49). Because glucocorticoid normally

    functions via GR (28), the difference in SGK1 expression under acute or chronic Dex

    treatment may be caused by differences in GR activity under different conditions, as

    shown by our work and those of others (28; 39). In addition, because chronic Dex

    treatment affects the activity of several regulatory molecules that influence SGK1

    transcription and/or mRNA decay (28; 50) and hypothalamic signals might also be

    affected by peripheral events (6; 16; 26), the possible contribution from these factors

    to hypothalamic SGK1 expression in Dex-treated mice cannot be excluded. These

    possibilities will be explored in future studies.

    In summary, our results demonstrate that SGK1/FOXO3 signaling in POMC

    neurons is crucial for Dex-induced adiposity, which provide novel insights into the

    central mechanisms underlying Dex-induced obesity. In this study, we also

    established that SGK1 in POMC neurons as an essential regulator of systemic energy

    balance. This previously unrecognized role for hypothalamic SGK1 also indicates a

    potential novel drug target in treating obesity and its related metabolic disorders.

    ACKNOWLEDGMENTS

    This work was supported by grants from the National Natural Science

    Foundation (81325005, 81390350, 81471076, 81570777, 81130076, 31271269,

    81400792, 81500622 and 81600623), Basic Research Project of Shanghai Science and

    Technology Commission (16JC1404900 and 17XD1404200) and CAS/SAFEA

    Page 21 of 60 Diabetes

  • 22

    international partnership program for creative research teams. Feifan Guo was also

    supported by the One Hundred Talents Program of CAS.

    AUTHOR CONTRIBUTIONS

    Y.D. researched data, wrote, reviewed and edited the manuscript. Y.X., F.Y.,

    Y.L., X.J., J.D. researched data. S.C. provided research material. A.N-F-T and G.F-T

    generated and provided the floxed SGK1 mice. Q.Z, H.Y., Y.C. directed the project

    and contributed to discussion. F.G. and Y.S. directed the project, contributed to

    discussion and wrote, reviewed, and edited the manuscript. F.G. and Y.S. 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.

    COMPETING FINANCIAL INTERESTS

    The authors declare that they have no competing interests.

    REFERENCES

    1. Ma X, Xu L, Mueller E: Forkhead box A3 mediates glucocorticoid receptor function in adipose

    tissue. Proceedings of the National Academy of Sciences of the United States of America

    2016;113:3377-3382

    2. Cha JY, Kim HJ, Yu JH, Xu J, Kim D, Paul BD, Choi H, Kim S, Lee YJ, Ho GP, Rao F, Snyder SH,

    Kim JW: Dexras1 mediates glucocorticoid-associated adipogenesis and diet-induced obesity.

    Proceedings of the National Academy of Sciences of the United States of America

    2013;110:20575-20580

    3. Chapman AB, Knight DM, Ringold GM: Glucocorticoid regulation of adipocyte differentiation:

    hormonal triggering of the developmental program and induction of a differentiation-dependent gene.

    The Journal of cell biology 1985;101:1227-1235

    4. Kong X, Yu J, Bi J, Qi H, Di W, Wu L, Wang L, Zha J, Lv S, Zhang F, Li Y, Hu F, Liu F, Zhou H,

    Liu J, Ding G: Glucocorticoids transcriptionally regulate miR-27b expression promoting body fat

    Page 22 of 60Diabetes

  • 23

    accumulation via suppressing the browning of white adipose tissue. Diabetes 2015;64:393-404

    5. Soumano K, Desbiens S, Rabelo R, Bakopanos E, Camirand A, Silva JE: Glucocorticoids inhibit the

    transcriptional response of the uncoupling protein-1 gene to adrenergic stimulation in a brown adipose

    cell line. Molecular and cellular endocrinology 2000;165:7-15

    6. Schneeberger M, Dietrich MO, Sebastian D, Imbernon M, Castano C, Garcia A, Esteban Y,

    Gonzalez-Franquesa A, Rodriguez IC, Bortolozzi A, Garcia-Roves PM, Gomis R, Nogueiras R,

    Horvath TL, Zorzano A, Claret M: Mitofusin 2 in POMC neurons connects ER stress with leptin

    resistance and energy imbalance. Cell 2013;155:172-187

    7. Schwartz MW, Porte D, Jr.: Diabetes, obesity, and the brain. Science 2005;307:375-379

    8. Wardlaw SL: Hypothalamic proopiomelanocortin processing and the regulation of energy balance.

    European journal of pharmacology 2011;660:213-219

    9. Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, Gu W, Kesterson

    RA, Boston BA, Cone RD, Smith FJ, Campfield LA, Burn P, Lee F: Targeted disruption of the

    melanocortin-4 receptor results in obesity in mice. Cell 1997;88:131-141

    10. Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T, O'Rahilly S: Clinical spectrum of obesity

    and mutations in the melanocortin 4 receptor gene. The New England journal of medicine

    2003;348:1085-1095

    11. Lowell BB, Spiegelman BM: Towards a molecular understanding of adaptive thermogenesis.

    Nature 2000;404:652-660

    12. Holm C: Molecular mechanisms regulating hormone-sensitive lipase and lipolysis. Biochemical

    Society transactions 2003;31:1120-1124

    13. Hucking K, Hamilton-Wessler M, Ellmerer M, Bergman RN: Burst-like control of lipolysis by the

    sympathetic nervous system in vivo. The Journal of clinical investigation 2003;111:257-264

    14. Morrison SF, Madden CJ, Tupone D: Central neural regulation of brown adipose tissue

    thermogenesis and energy expenditure. Cell metabolism 2014;19:741-756

    15. Veyrat-Durebex C, Deblon N, Caillon A, Andrew R, Altirriba J, Odermatt A, Rohner-Jeanrenaud F:

    Central glucocorticoid administration promotes weight gain and increased 11beta-hydroxysteroid

    dehydrogenase type 1 expression in white adipose tissue. PloS one 2012;7:e34002

    16. Zakrzewska KE, Cusin I, Stricker-Krongrad A, Boss O, Ricquier D, Jeanrenaud B,

    Rohner-Jeanrenaud F: Induction of obesity and hyperleptinemia by central glucocorticoid infusion in

    the rat. Diabetes 1999;48:365-370

    17. Lang F, Artunc F, Vallon V: The physiological impact of the serum and glucocorticoid-inducible

    kinase SGK1. Current opinion in nephrology and hypertension 2009;18:439-448

    18. Fejes-Toth G, Frindt G, Naray-Fejes-Toth A, Palmer LG: Epithelial Na+ channel activation and

    processing in mice lacking SGK1. American journal of physiology Renal physiology

    2008;294:F1298-1305

    19. Liu H, Yu J, Xia T, Xiao Y, Zhang Q, Liu B, Guo Y, Deng J, Deng Y, Chen S, Naray-Fejes-Toth A,

    Fejes-Toth G, Guo F: Hepatic serum- and glucocorticoid-regulated protein kinase 1 (SGK1) regulates

    insulin sensitivity in mice via extracellular-signal-regulated kinase 1/2 (ERK1/2). The Biochemical

    journal 2014;464:281-289

    20. Anacker C, Cattaneo A, Musaelyan K, Zunszain PA, Horowitz M, Molteni R, Luoni A, Calabrese F,

    Tansey K, Gennarelli M, Thuret S, Price J, Uher R, Riva MA, Pariante CM: Role for the kinase SGK1

    in stress, depression, and glucocorticoid effects on hippocampal neurogenesis. Proceedings of the

    National Academy of Sciences of the United States of America 2013;110:8708-8713

    Page 23 of 60 Diabetes

  • 24

    21. Ullrich S, Berchtold S, Ranta F, Seebohm G, Henke G, Lupescu A, Mack AF, Chao CM, Su J,

    Nitschke R, Alexander D, Friedrich B, Wulff P, Kuhl D, Lang F: Serum- and glucocorticoid-inducible

    kinase 1 (SGK1) mediates glucocorticoid-induced inhibition of insulin secretion. Diabetes

    2005;54:1090-1099

    22. van Gemert NG, Meijer OC, Morsink MC, Joels M: Effect of brief corticosterone administration on

    SGK1 and RGS4 mRNA expression in rat hippocampus. Stress 2006;9:165-170

    23. Reiter MH, Vila G, Knosp E, Baumgartner-Parzer SM, Wagner L, Stalla GK, Luger A: Opposite

    effects of serum- and glucocorticoid-regulated kinase-1 and glucocorticoids on POMC transcription

    and ACTH release. American journal of physiology Endocrinology and metabolism

    2011;301:E336-341

    24. Berglund ED, Liu C, Sohn JW, Liu T, Kim MH, Lee CE, Vianna CR, Williams KW, Xu Y, Elmquist

    JK: Serotonin 2C receptors in pro-opiomelanocortin neurons regulate energy and glucose homeostasis.

    The Journal of clinical investigation 2013;123:5061-5070

    25. Hoekstra M, Meurs I, Koenders M, Out R, Hildebrand RB, Kruijt JK, Van Eck M, Van Berkel TJ:

    Absence of HDL cholesteryl ester uptake in mice via SR-BI impairs an adequate adrenal

    glucocorticoid-mediated stress response to fasting. Journal of lipid research 2008;49:738-745

    26. Kleinridders A, Schenten D, Konner AC, Belgardt BF, Mauer J, Okamura T, Wunderlich FT,

    Medzhitov R, Bruning JC: MyD88 signaling in the CNS is required for development of fatty

    acid-induced leptin resistance and diet-induced obesity. Cell metabolism 2009;10:249-259

    27. Xia T, Cheng Y, Zhang Q, Xiao F, Liu B, Chen S, Guo F: S6K1 in the central nervous system

    regulates energy expenditure via MC4R/CRH pathways in response to deprivation of an essential

    amino acid. Diabetes 2012;61:2461-2471

    28. Arango-Lievano M, Lambert WM, Bath KG, Garabedian MJ, Chao MV, Jeanneteau F:

    Neurotrophic-priming of glucocorticoid receptor signaling is essential for neuronal plasticity to stress

    and antidepressant treatment. Proceedings of the National Academy of Sciences of the United States of

    America 2015;112:15737-15742

    29. Koch M, Varela L, Kim JG, Kim JD, Hernandez-Nuno F, Simonds SE, Castorena CM, Vianna CR,

    Elmquist JK, Morozov YM, Rakic P, Bechmann I, Cowley MA, Szigeti-Buck K, Dietrich MO, Gao XB,

    Diano S, Horvath TL: Hypothalamic POMC neurons promote cannabinoid-induced feeding. Nature

    2015;519:45-50

    30. Poggioli R, Ueta CB, Drigo RA, Castillo M, Fonseca TL, Bianco AC: Dexamethasone reduces

    energy expenditure and increases susceptibility to diet-induced obesity in mice. Obesity

    2013;21:E415-420

    31. Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW: Central nervous system control

    of food intake and body weight. Nature 2006;443:289-295

    32. Zhang G, Li J, Purkayastha S, Tang Y, Zhang H, Yin Y, Li B, Liu G, Cai D: Hypothalamic

    programming of systemic ageing involving IKK-beta, NF-kappaB and GnRH. Nature

    2013;497:211-216

    33. Plum L, Lin HV, Dutia R, Tanaka J, Aizawa KS, Matsumoto M, Kim AJ, Cawley NX, Paik JH, Loh

    YP, DePinho RA, Wardlaw SL, Accili D: The obesity susceptibility gene Cpe links FoxO1 signaling in

    hypothalamic pro-opiomelanocortin neurons with regulation of food intake. Nature medicine

    2009;15:1195-1201

    34. Wilson CA, Buckingham JC, Morris ID: Influence of growth hormone, corticosterone,

    corticotrophin and changes in the environmental temperature on pituitary-ovarian function in the

    Page 24 of 60Diabetes

  • 25

    immature rat. The Journal of endocrinology 1985;104:179-183

    35. Murray JT, Campbell DG, Morrice N, Auld GC, Shpiro N, Marquez R, Peggie M, Bain J,

    Bloomberg GB, Grahammer F, Lang F, Wulff P, Kuhl D, Cohen P: Exploitation of KESTREL to

    identify NDRG family members as physiological substrates for SGK1 and GSK3. The Biochemical

    journal 2004;384:477-488

    36. Brunet A, Park J, Tran H, Hu LS, Hemmings BA, Greenberg ME: Protein kinase SGK mediates

    survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a). Molecular

    and cellular biology 2001;21:952-965

    37. Ma W, Fuentes G, Shi X, Verma C, Radda GK, Han W: FoxO1 negatively regulates leptin-induced

    POMC transcription through its direct interaction with STAT3. The Biochemical journal

    2015;466:291-298

    38. Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del Piccolo P, Burden SJ, Di Lisi R,

    Sandri C, Zhao J, Goldberg AL, Schiaffino S, Sandri M: FoxO3 controls autophagy in skeletal muscle

    in vivo. Cell metabolism 2007;6:458-471

    39. Sarabdjitsingh RA, Meijer OC, de Kloet ER: Specificity of glucocorticoid receptor primary

    antibodies for analysis of receptor localization patterns in cultured cells and rat hippocampus. Brain

    research 2010;1331:1-11

    40. Smart JL, Tolle V, Low MJ: Glucocorticoids exacerbate obesity and insulin resistance in

    neuron-specific proopiomelanocortin-deficient mice. The Journal of clinical investigation

    2006;116:495-505

    41. Kaushik S, Arias E, Kwon H, Lopez NM, Athonvarangkul D, Sahu S, Schwartz GJ, Pessin JE,

    Singh R: Loss of autophagy in hypothalamic POMC neurons impairs lipolysis. EMBO reports

    2012;13:258-265

    42. Nogueiras R, Wiedmer P, Perez-Tilve D, Veyrat-Durebex C, Keogh JM, Sutton GM, Pfluger PT,

    Castaneda TR, Neschen S, Hofmann SM, Howles PN, Morgan DA, Benoit SC, Szanto I, Schrott B,

    Schurmann A, Joost HG, Hammond C, Hui DY, Woods SC, Rahmouni K, Butler AA, Farooqi IS,

    O'Rahilly S, Rohner-Jeanrenaud F, Tschop MH: The central melanocortin system directly controls

    peripheral lipid metabolism. The Journal of clinical investigation 2007;117:3475-3488

    43. Wardlaw SL, McCarthy KC, Conwell IM: Glucocorticoid regulation of hypothalamic

    proopiomelanocortin. Neuroendocrinology 1998;67:51-57

    44. Scarpa JR, Jiang P, Losic B, Readhead B, Gao VD, Dudley JT, Vitaterna MH, Turek FW, Kasarskis

    A: Systems Genetic Analyses Highlight a TGFbeta-FOXO3 Dependent Striatal Astrocyte Network

    Conserved across Species and Associated with Stress, Sleep, and Huntington's Disease. PLoS genetics

    2016;12:e1006137

    45. Renault VM, Rafalski VA, Morgan AA, Salih DA, Brett JO, Webb AE, Villeda SA, Thekkat PU,

    Guillerey C, Denko NC, Palmer TD, Butte AJ, Brunet A: FoxO3 regulates neural stem cell homeostasis.

    Cell Stem Cell 2009;5:527-539

    46. Stefaneanu L, Kovacs K, Horvath E, Lloyd RV: In situ hybridization study of pro-opiomelanocortin

    (POMC) gene expression in human pituitary corticotrophs and their adenomas. Virchows Arch A Pathol

    Anat Histopathol 1991;419:107-113

    47. Yang Y, Atasoy D, Su HH, Sternson SM: Hunger states switch a flip-flop memory circuit via a

    synaptic AMPK-dependent positive feedback loop. Cell 2011;146:992-1003

    48. Chen AS, Marsh DJ, Trumbauer ME, Frazier EG, Guan XM, Yu H, Rosenblum CI, Vongs A, Feng Y,

    Cao L, Metzger JM, Strack AM, Camacho RE, Mellin TN, Nunes CN, Min W, Fisher J, Gopal-Truter S,

    Page 25 of 60 Diabetes

  • 26

    MacIntyre DE, Chen HY, Van der Ploeg LH: Inactivation of the mouse melanocortin-3 receptor results

    in increased fat mass and reduced lean body mass. Nature genetics 2000;26:97-102

    49. Sousa e Silva T, Longui CA, Rocha MN, Faria CD, Melo MR, Faria TG, de Souza JA, Rizzo LV:

    Prolonged physical training decreases mRNA levels of glucocorticoid receptor and inflammatory genes.

    Hormone research in paediatrics 2010;74:6-14

    50. Cho H, Park OH, Park J, Ryu I, Kim J, Ko J, Kim YK: Glucocorticoid receptor interacts with

    PNRC2 in a ligand-dependent manner to recruit UPF1 for rapid mRNA degradation. Proceedings of the

    National Academy of Sciences of the United States of America 2015;112:E1540-1549

    FIGURE LEGENDS

    Figure 1. SGK1 expression in hypothalamic POMC neurons under chronic or

    acute Dex treatment.

    (A) Sgk1 expression in hypothalamus;

    (B) SGK1 and p-SGK1 western blot and densitometric quantification in

    hypothalamus;

    (C and D) Immunofluorescence for SGK1 in ARC (arcuate nucleus) sections (C) and

    integrated density quantification (D);

    (E and F) Immunofluorescence for p-SGK1 in ARC sections (E) and integrated

    density quantification (F);

    (G and H) Immunofluorescence for POMC neurons (red), SGK1 (green) and merge

    (yellow) in ARC sections (G) and integrated density quantification in POMC neurons

    and colocalization (H);

    (I) Sgk1 expression in hypothalamus;

    (J) SGK1 western blot and densitometric quantification in hypothalamus;

    (K and L) Immunofluorescence for POMC neurons (red), SGK1 (green) and merge

    (yellow) in ARC sections (K) and integrated density quantification in POMC neurons

    Page 26 of 60Diabetes

  • 27

    and colocalization (L).

    Studies were conducted in 14 to 15-weeks old male wild-type mice (for A-F) or

    POMC-tdTomato indicator mice (for G and H) treated without (- Dex) or with Dex (+

    Dex) for 6 weeks. Studies were conducted in 9-weeks old male wild-type mice (for I

    and J) or POMC-tdTomato indicator mice (for K and L) treated without (- Dex) or

    with Dex (+ Dex) for 2 hours. Data are expressed as mean ± SEM (n = 6-11/group). *:

    p < 0.05 for the effect of with versus without Dex treatment group.

    Figure 2. PSKO mice exhibit obese phenotype and decreased energy expenditure

    as Dex-treated mice.

    (A) Immunofluorescence for POMC neurons (red), SGK1 (green) and merge (yellow)

    in ARC (arcuate nucleus) sections from male POMC-tdTomato indicator mice;

    (B) Sgk1 expression in different tissues (ARC; COR: cortex; WAT: white adipose

    tissue; BAT: brown adipose tissue; LV: liver);

    (C) Body weight curve;

    (D) Total body fat mass;

    (E) Abdominal fat mass;

    (F) Daily food intake;

    (G) Daily energy expenditure;

    (H) Daily RER (respiratory exchange ratio, VCO2/VO2);

    (I) Daily locomotor activity;

    (J) Basal rectal temperature;

    Page 27 of 60 Diabetes

  • 28

    (K) UCP1 western blot and densitometric quantification in BAT;

    (L) Serum NE (norepinephrine).

    All studies were conducted in 12 to 14-weeks old male control (- PSKO) and PSKO

    (+ PSKO) mice. Data are expressed as mean ± SEM (n = 6-16/group). *: p < 0.05 for

    the effect of PSKO group versus control group.

    Figure 3. PSOE mice show lean phenotype and increased energy expenditure.

    (A) Immunofluorescence for POMC neurons (red), p-NDRG1 (green) and merge

    (yellow) in ARC (arcuate nucleus) sections;

    (B) Body weight curve;

    (C) Total body fat mass;

    (D) Abdominal fat mass;

    (E) Daily food intake;

    (F) Daily energy expenditure;

    (G) Daily RER (respiratory exchange ratio, VCO2/VO2);

    (H) Daily locomotor activity;

    (I) Basal rectal temperature;

    (J) UCP1 western blot and densitometric quantification in BAT (brown adipose

    tissue);

    (K) Serum NE (norepinephrine).

    All studies were conducted in 19 to 20-weeks old male control (- PSOE) and PSOE (+

    PSOE) mice. Data are expressed as mean ± SEM (n = 6-9/group). *: p < 0.05 for the

    Page 28 of 60Diabetes

  • 29

    effect of PSOE group versus control group.

    Figure 4. PSOE mice are resistant to Dex-induced fat accumulation and

    decreased energy expenditure.

    (A) Body weight curve;

    (B) Total body fat mass;

    (C) Abdominal fat mass;

    (D) Daily food intake;

    (E) Daily energy expenditure;

    (F) Daily RER (respiratory exchange ratio, VCO2/VO2);

    (G) Daily locomotor activity;

    (H) Basal rectal temperature;

    (I) UCP1 western blot and densitometric quantification in BAT (brown adipose

    tissue);

    (J) Serum NE (norepinephrine).

    All studies were conducted in 19 to 20-weeks old male control (- PSOE) and PSOE (+

    PSOE) mice treated without (- Dex) or with Dex (+ Dex). Data are expressed as mean

    ± SEM (n = 6-12/group). *: p < 0.05 for the effect of PSOE group versus control

    group.

    Figure 5. Dex decreases hypothalamic α-MSH content via SGK1 and i.c.v

    administration of α-MSH reverses obese phenotype in PSKO mice.

    Page 29 of 60 Diabetes

  • 30

    (A and B) Immunofluorescence for α-MSH in PVN (paraventricular nucleus) sections

    (A) and integrated density quantification (B) in 14 to 15-weeks old male wild-type

    mice treated without (- Dex) or with Dex (+ Dex);

    (C-E) Immunofluorescence for α-MSH in PVN sections (C), integrated density

    quantification (D) and relative hypothalamic α-MSH content by ELISA (E) in 12 to

    14-weeks old male control and PSKO mice;

    (F-G) Immunofluorescence for α-MSH in PVN sections (F) and integrated density

    quantification (G) in 19 to 20-weeks old male control and PSOE mice treated without

    (- Dex) or with Dex (+ Dex);

    (H-K) Body weight (H), abdominal fat mass (I), basal rectal temperature (J) and

    UCP1 western blot and densitometric quantification in BAT (brown adipose tissue)

    (K) in 10 to 12-weeks old male control (- PSKO) and PSKO (+ PSKO) mice treated

    without (- α-MSH) or with α-MSH (+ α-MSH).

    Data are expressed as mean ± SEM (n = 6-8/group). *: p < 0.05 for any treatment

    compared with control group for A-E. *: p < 0.05 for the effect of any group versus

    control mice treated without Dex; #: p < 0.05 for the effect of PSOE mice versus

    control mice following Dex treatment for G. *: p < 0.05 for the effect of any group

    versus control mice treated without α-MSH; #: p < 0.05 for the effect of with versus

    without α-MSH in PSKO mice for H-K.

    Figure 6. Dex reduces α-MSH precursor POMC expression via SGK1/FOXO3

    dependent pathway and down-regulation of FOXO3 in ARC largely reverses the

    Page 30 of 60Diabetes

  • 31

    obesity phenotype in PSKO mice.

    (A-C) Neuropeptides expression in hypothalamus (A), immunofluorescence for

    POMC in ARC (arcuate nucleus) sections (B) and integrated density quantification (C)

    in 14 to 15-weeks old male wild-type (WT) mice treated without (- Dex) or with Dex

    (+ Dex);

    (D and E) Immunofluorescence for POMC in ARC sections (D) and integrated

    density quantification (E) in 19 to 20-weeks old male control and PSOE mice treated

    without (- Dex) or with Dex (+ Dex);

    (F) P-FOXO3 and FOXO3 western blot and densitometric quantification in

    hypothalamus of 14 to 15-weeks old male WT mice treated without (- Dex) or with

    Dex (+ Dex);

    (G and H) Immunofluorescence for POMC neurons (red), p-FOXO3 (green) and

    merge (yellow) in ARC sections (G) and integrated density quantification in POMC

    neurons and colocalization (H) in 19 to 20-weeks old male control and PSOE mice

    treated without (- Dex) or with Dex (+ Dex);

    (I-Q) The expression of Sgk1, Foxo3 and Pomc in ARC (arcuate nucleus) (I), body

    weight (J), total body fat mass (K), abdominal fat mass (L), daily energy expenditure

    (M), daily RER (respiratory exchange ratio, VCO2/VO2) (N), basal rectal temperature

    (O), UCP1 western blot and densitometric quantification in BAT (brown adipose

    tissue) (P) and serum NE (norepinephrine) (Q) in 16 to 18-weeks old male control (-

    PSKO) and PSKO (+ PSKO) mice injected with Ad-scrambled (- Ad-shFOXO3) or

    Ad-shFOXO3 (+ Ad-shFOXO3).

    Page 31 of 60 Diabetes

  • 32

    Data are expressed as mean ± SEM (n = 6-11/group). *: p < 0.05 for the effect of with

    versus without Dex treatment group for A, C and F. *: p < 0.05 for the effect of any

    group versus control mice treated without Dex; #: p < 0.05 for the effect of PSOE

    mice versus control mice following Dex treatment for E and H. *: p < 0.05 for the

    effect of any group versus control mice injected without Ad-shFOXO3; #: p < 0.05

    for the effect of with versus without Ad-shFOXO3 injection in PSKO mice for I-Q.

    Page 32 of 60Diabetes

  • E F

    0

    50

    100

    150

    *

    SG

    K1

    (a

    rbit

    rary

    flu

    ore

    scen

    ce u

    nit

    s)

    0

    50

    100

    150

    *

    P-S

    GK

    1 (

    arb

    itra

    ry

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    C D

    + Dex Dex - Fig. 1

    * *

    0

    50

    100

    150

    SGK1 p-SGK1

    Arb

    itra

    ry

    un

    its

    A B Dex: - + - + - +

    p-SGK1

    Actin

    SGK1 *

    0

    50

    100

    150

    Re

    lati

    ve

    Sg

    k1

    mR

    NA

    (%

    )

    100 um

    SGK1

    - Dex + Dex

    ARC ARC

    100 um

    - Dex + Dex

    ARC

    p-SGK1

    ARC

    tdTomato SGK1

    100 um

    - D

    ex

    +

    De

    x *

    SG

    K1

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    0

    50

    100

    150

    *

    SG

    K1

    /td

    To

    ma

    to

    co

    loc

    alizati

    on

    (%

    )

    0

    50

    100

    150

    G H Merge

    Page 33 of 60 Diabetes

  • 0

    100

    200

    300

    Arb

    itra

    ry u

    nit

    s

    *

    SGK1

    Actin

    Dex: - + - + - +

    Re

    lati

    ve

    Sg

    k1

    mR

    NA

    (%

    )

    0

    100

    200

    300 *

    I J

    + Dex Dex -

    0

    100

    200

    SG

    K1

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    *

    0

    50

    100

    150

    SG

    K1

    /td

    To

    ma

    to

    co

    loc

    alizati

    on

    (%

    )

    Fig. 1

    K L

    - D

    ex

    +

    De

    x

    tdTomato

    100 um

    SGK1 Merge

    2 h 2 h

    2 h 2 h

    Page 34 of 60Diabetes

  • Fig. 2

    B

    Control PSKO

    0

    50

    100

    150

    Re

    lati

    ve

    Sg

    k1 m

    RN

    A (

    %)

    *

    D E

    C

    *

    15

    20

    25

    30

    35

    Bo

    dy w

    eig

    ht

    (g)

    Age (weeks)

    5 7 9 11 13 15 17

    Control

    PSKO

    *

    0

    1

    2

    3

    Fa

    t m

    as

    s (

    g)

    0

    0.2

    0.4

    0.6

    Ab

    do

    min

    al

    fat

    ma

    ss

    (g

    )

    *

    A

    F

    0

    2

    4

    6 F

    oo

    d In

    take

    (g/d

    ay)

    0

    500

    1000

    1500

    Light Dark Total Lo

    co

    mo

    tor

    ac

    tivit

    y

    (co

    un

    ts)

    I J K

    35

    36

    37

    14:00 17:00

    Re

    cta

    l

    Te

    mp

    era

    ture

    (℃

    )

    * *

    UCP1

    Actin

    PSKO: - + - + - +

    0

    50

    100

    150

    Arb

    itra

    ry

    un

    its

    *

    L

    Se

    rum

    NE

    (n

    g/l

    )

    0

    200

    400

    *

    H

    0

    0.4

    0.8

    1.2

    Light Dark Total

    RE

    R

    * * *

    G

    0 0.1

    0.2

    0.3

    0.4

    0.5

    EE

    (k

    ca

    l/h

    r)

    * * *

    Light Dark Total

    tdTomato SGK1 Merge

    100 um

    Co

    ntr

    ol

    PS

    KO

    Page 35 of 60 Diabetes

  • Control PSOE

    0

    0.2

    0.4

    0.6

    0.8

    Light Dark Total E

    E (

    kc

    al/h

    r)

    * * *

    0

    0.5

    1.0

    Light Dark Total

    RE

    R

    * * * F E G D

    0

    1

    2

    3

    4

    5

    Fo

    od

    in

    tak

    e

    (g/d

    ay) *

    0

    0.2

    0.4

    0.6

    Ab

    do

    min

    al

    fat

    ma

    ss

    (g

    )

    Fig. 3

    B A C

    0

    1

    2

    Fa

    t m

    as

    s (

    g)

    * 100 um

    mCherry

    mCherry

    p-NDRG1 Merge

    Merge p-NDRG1

    Co

    ntr

    ol

    PS

    OE

    Control

    PSOE

    * 20

    25

    30

    35

    0 2 4 6 8 10

    Time (weeks)

    Bo

    dy w

    eig

    ht

    (g)

    0

    500

    1000

    Light Dark Total Lo

    co

    mo

    tor

    ac

    tivit

    y

    (co

    un

    ts)

    H

    35

    36

    37

    14:00 17:00

    Re

    cta

    l

    tem

    pe

    ratu

    re (℃

    )

    * *

    I J K

    *

    0

    200

    400

    Se

    rum

    NE

    (n

    g/l

    )

    UCP1

    Actin

    PSOE: - + - + - +

    0

    100

    200

    300

    Arb

    itra

    ry

    un

    its

    *

    Page 36 of 60Diabetes

  • Control + Dex PSOE + Dex

    0

    2

    4

    6

    Fo

    od

    in

    tak

    e

    (g/d

    ay)

    14:00 17:00

    Re

    cta

    l

    tem

    pe

    ratu

    re (℃

    )

    35

    36

    37

    * *

    0

    0.4

    0.8

    1.2

    Light Dark Total

    RE

    R

    * * *

    0

    0.2

    0.4

    0.6

    0.8

    Light Dark Total

    EE

    (k

    ca

    l/h

    r)

    * * *

    0

    500

    1000

    1500

    Light Dark Total Lo

    co

    mo

    tor

    ac

    tivit

    y

    (co

    un

    ts)

    0

    0.5

    1.0

    Ab

    do

    min

    al

    fat

    ma

    ss

    (g

    )

    *

    Fig. 4

    Dex

    * 20

    25

    30

    35

    0 1 2 3 4 5 6 7 8 9 10 11

    Time (weeks)

    Bo

    dy w

    eig

    ht

    (g)

    Control

    PSOE

    A B C D

    E F G

    H

    0

    200

    400

    Se

    rum

    NE

    (n

    g/l

    )

    * J I

    0

    100

    200

    300

    Arb

    itra

    ry u

    nit

    s

    *

    0

    1

    2

    3

    4

    5

    Fa

    t m

    as

    s (

    g)

    *

    UCP1

    Actin

    PSOE: - + - + - +

    Dex: + + + + + +

    Page 37 of 60 Diabetes

  • Fig. 5

    E

    100 um

    a- MSH

    Control PSKO

    100 um

    a- MSH

    - Dex + Dex

    100 um

    a- MSH

    Control PSOE

    G F

    D C B A

    0

    50

    100

    150

    *

    a-

    MS

    H (

    arb

    itra

    ry

    flu

    ore

    sc

    en

    ce

    un

    its)

    Dex: - + 0

    50

    100

    150

    *

    a-

    MS

    H (

    arb

    itra

    ry

    flu

    ore

    sc

    en

    ce

    un

    its)

    H I J

    15

    20

    25

    Bo

    dy w

    eig

    ht

    (g)

    *

    * #

    0

    0.1

    0.2

    Ab

    do

    min

    al

    fat

    ma

    ss

    (g

    )

    *

    *

    #

    K

    34

    35

    36

    37

    Rec

    tal

    tem

    pe

    ratu

    re (℃

    ) #

    *

    *

    PSKO a- MSH Control a- MSH

    Control + a- MSH PSKO + a- MSH

    - -

    0

    50

    100

    150

    Rela

    tive

    a-

    MS

    H

    Co

    nte

    nt

    (%)

    *

    0

    100

    200

    300

    *

    *

    #

    a-

    MS

    H (

    arb

    itra

    ry

    flu

    ore

    sc

    en

    ce

    un

    its)

    PSOE

    Control

    Dex: - - + +

    Control PSOE

    - Dex + Dex

    0

    100

    200

    Arb

    itra

    ry

    un

    its

    * *

    *

    #

    a- MSH: - +

    UCP1

    Actin

    PSKO: - - + +

    + -

    Page 38 of 60Diabetes

  • E D

    100 um

    POMC

    Control PSOE

    F Dex: - + - + - +

    p-FOXO3

    FOXO3

    Actin

    Arb

    itra

    ry

    un

    its

    0

    50

    100

    150

    * *

    + Dex Dex -

    Control PSOE

    - Dex + Dex

    0

    100

    200 *

    *

    PO

    MC

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    #

    Dex:- - + +

    PSOE Control

    0

    50

    100

    150

    Pc1/3 Pc2 Cpe Pam Prcp Re

    lati

    ve

    mR

    NA

    (%

    )

    Pomc

    *

    Fig. 6 + Dex Dex -

    0

    50

    100

    150

    *

    - Dex + Dex B C

    Dex:- +

    A

    PO

    MC

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    100 um

    POMC

    H G

    100 um

    mCherry

    p-FOXO3

    Control PSOE

    - D

    ex

    + D

    ex

    0

    100

    200

    P-F

    OX

    O3 (

    arb

    itra

    ry

    flu

    ore

    scen

    ce u

    nit

    s)

    PSOE Control

    *

    *

    #

    Dex:- - + +

    0

    50

    100

    150

    p-F

    OX

    O3

    /mC

    he

    rry

    co

    localizati

    on

    (%

    )

    Dex:- - + +

    *

    *

    #

    Merge

    Page 39 of 60 Diabetes

  • Fig. 6

    35

    36

    37

    14:00 17:00

    Re

    cta

    l

    tem

    pa

    ratu

    re (℃

    )

    *

    *

    *

    * # #

    *

    #

    0

    100

    200

    NE

    (n

    g/l

    )

    *

    Q P O Ad-shFOXO3: - + -

    UCP1

    Actin

    PSKO: - - + + +

    0

    100

    200

    Arb

    itra

    ry

    un

    its

    *

    * #

    Control Ad-shFOXO3

    Control + Ad-shFOXO3

    PSKO Ad-shFOXO3

    PSKO + Ad-shFOXO3

    - -

    0

    0.5

    1.0

    Ab

    do

    min

    al

    fat

    ma

    ss

    (g

    )

    *

    *

    #

    0

    0.2

    0.4

    0.6

    0.8

    Light Dark Total

    EE

    (k

    ca

    l/h

    r)

    * * #

    # #

    * * * * *

    *

    0

    0.4

    0.8

    1.2

    Light Dark Total

    RE

    R

    * * * * * * * #

    #

    #

    L M N

    0

    100

    200

    Sgk1 Foxo3 Pomc Re

    lati

    ve

    mR

    NA

    (%

    )

    * * * *

    *

    *

    * #

    #

    0

    2

    4

    6

    8

    Fa

    t m

    as

    s (

    g)

    *

    *

    #

    0

    10

    20

    30

    40

    Bo

    dy w

    eig

    ht

    (g)

    * # I J K

    Page 40 of 60Diabetes

  • SUPPLEMENTAL MATERIAL

    FIGURE LEGENDS

    Figure S1. Chronic Dex treatment increases adiposity and decreases energy

    expenditure.

    (A) Body weight curve;

    (B) Total body fat mass;

    (C) Lean mass;

    (D) Abdominal fat mass;

    (E) Daily food intake;

    (F) Daily energy expenditure;

    (G) Daily locomotor activity;

    (H) Basal rectal temperature;

    (I) UCP1 western blot and densitometric quantification in BAT (brown adipose

    tissue);

    (J) Serum NE (norepinephrine).

    All studies were conducted in 14 to 15-weeks old male wild-type mice treated without

    (- Dex) or with Dex (+ Dex) for 6 weeks. Data are expressed as mean ± SEM (n =

    6-16/group). *: p < 0.05 for the effect of with versus without Dex treatment group.

    Figure S2. SGK1 in POMC neurons and metabolic parameters in PSKO mice.

    (A and B) Immunofluorescence for tdTomato (red), POMC (green) and merge

    (yellow) in ARC (arcuate nucleus) sections (A) and colocalization (B);

    Page 41 of 60 Diabetes

  • 2

    (A) Integrated density quantification in POMC neurons and colocalization for SGK1

    in ARC sections;

    (D and E) Immunofluorescence for SGK1 in PVN (paraventricular nucleus) sections

    (D) and integrated density quantification (E);

    (F and G) Immunofluorescence for SGK1 in VMH (ventromedial nucleus) sections (F)

    and integrated density quantification (G);

    (H) Lean mass.

    All studies were conducted in 12 to 14-weeks old male control and PSKO mice. Data

    are expressed as mean ± SEM (n = 4-6/group). *: p < 0.05 for the effect of PSKO

    group versus control group.

    Figure S3. POMC neuron anatomy and pituitary-adrenal axis function in PSKO

    mice.

    (A) Relative POMC neuron area;

    (B) POMC neuron number and distribution throughout ARC (arcuate nucleus);

    (C) Serum corticosterone content;

    (D) Serum growth hormone content.

    All studies were conducted in 12 to 14-weeks old male control and PSKO mice. Data

    are expressed as mean ± SEM (n = 6-8/group). *: p < 0.05 for the effect of PSKO

    group versus control group.

    Figure S4. Metabolic phenotypes in female PSKO mice.

    Page 42 of 60Diabetes

  • 3

    (A) Body weight curve;

    (B) Total body fat mass;

    (C) Daily food intake;

    (D) Daily energy expenditure;

    (E) Daily RER (respiratory exchange ratio, VCO2/VO2);

    (F) Daily locomotor activity;

    (G) Basal rectal temperature.

    All studies were conducted in 15 to 16-weeks old female control and PSKO mice.

    Data are expressed as mean ± SEM (n = 6-13/group). *: p < 0.05 for the effect of

    PSKO group versus control group.

    Figure S5. Metabolic phenotypes in PSKO-ER mice.

    (A) Body weight curve;

    (B) Total body fat mass;

    (C) Abdominal fat mass;

    (D) Daily food intake;

    (E) Daily energy expenditure;

    (F) Daily RER (respiratory exchange ratio, VCO2/VO2);

    (G) Daily locomotor activity;

    (H) Basal rectal temperature.

    All studies were conducted in 12 to 13-weeks old male control and PSKO-ER mice

    treated with tamoxifen (T) at 8 weeks of age. Data are expressed as mean ± SEM

    Page 43 of 60 Diabetes

  • 4

    (n=6-8/group). *: p < 0.05 for the effect of PSKO-ER group versus control group.

    Figure S6. Identification of POMC neurons and SGK1 expression in PSOE mice.

    (A and B) Immunofluorescence for mCherry (red), POMC (green) and merge (yellow)

    in ARC (arcuate nucleus) sections (A) and colocalization (B);

    (C and D) Immunofluorescence for POMC neurons (red), SGK1 (green) and merge

    (yellow) in ARC sections (C) and integrated density quantification in POMC neurons

    (D);

    (E and F) Immunofluorescence for SGK1 in PVN (paraventricular nucleus) sections

    (E) and integrated density quantification (F);

    (G and H) Immunofluorescence for SGK1 in VMH (ventromedial nucleus) sections

    (G) and integrated density quantification (H);

    (I) Lean mass;

    (J) Total body fat mass.

    Studies were conducted in 19 to 20-weeks old male POMC-Cre mice injected with

    AAV-null (for A and B). Studies were conducted in 19 to 20-weeks old male control

    and PSOE mice (for C-H). Studies were conducted in 13 to 14-weeks old male control

    and PSOE mice 5 weeks after ARC injection of AAV-null or AAV-CA SGK1 prior to

    Dex treatment (for I and J). Data are expressed as mean ± SEM (n = 4-12/group). *: p

    < 0.05 for the effect of PSOE group versus control group.

    Figure S7. The metabolic phenotypes of PSKO mice under Dex treatment.

    Page 44 of 60Diabetes

  • 5

    (A) Body weight;

    (B) Total body fat mass;

    (C) Abdominal fat mass;

    (D) Lean mass;

    (E) Daily food intake;

    (F) Daily energy expenditure.

    All studies were conducted in 14 to 16-weeks old male control and PSKO mice

    treated without (- Dex) or with Dex (+ Dex) for 6 weeks. Data are expressed as mean

    ± SEM (n =10-13/group). *: p < 0.05 for the effect of any group versus control mice

    treated without Dex; #: p < 0.05 for the effect of with versus without Dex treatment in

    PSKO mice.

    Figure S8. SGK1 regulates POMC expression.

    (A-C) Neuropeptide expression in hypothalamus (A), immunofluorescence for POMC

    in ARC (arcuate nucleus) sections (B) and integrated density quantification (C) in 12

    to 14-weeks old male control and PSKO mice;

    (D) Sgk1 and Pomc expression in primary hypothalamic neurons treated without (-

    Ad-shSGK1) or with Ad-shSGK1 (+ Ad-shSGK1);

    (E) Sgk1 and Pomc expression in primary hypothalamic neurons treated without (- CA

    SGK1) or with CA SGK1 plasmid (+ CA SGK1).

    Data are expressed as mean ± SEM (n = 6-12/group). *: p < 0.05 for the effect of CA

    SGK1 group versus control group.

    Page 45 of 60 Diabetes

  • 6

    Figure S9. SGK1 regulates POMC expression by phosphorylating FOXO3.

    (A) P-FOXO3 and FOXO3 western blot and densitometric quantification in ARC

    (arcuate nucleus) of 12 to 14-weeks old male control (- PSKO) and PSKO (+ PSKO)

    mice;

    (B) P-FOXO3, FOXO3 and SGK1 western blot and densitometric quantification in

    primary hypothalamic neurons treated without (- Ad-shSGK1) or with Ad-shSGK1 (+

    Ad-shSGK1);

    (C) P-FOXO3, FOXO3 and SGK1 western blot and densitometric quantification in

    primary hypothalamic neurons treated without (- CA SGK1) or with CA SGK1

    plasmid (+ CA SGK1);

    (D) Sgk1, Foxo3 and Pomc expression in primary hypothalamic neurons treated with

    Ad-scrambled (- Ad-shSGK1) or Ad-shSGK1 (+ Ad-shSGK1) in the presence of

    control reagent (- siFOXO3) or siFOXO3 (+ siFOXO3).

    Data are expressed as mean ± SEM (n = 6-8/group). *: p < 0.05 for any treatment

    compared with control group for A-C. *: p < 0.05 for the effect of any group versus

    group without Ad-shSGK1 and siFOXO3; #: p < 0.05 for the effect of with versus

    without siFOXO3 in the presence of Ad-shSGK1 for D.

    Figure S10. Down-regulation of FOXO3 largely reverses hypothalamic α-MSH

    content and in PSKO mice.

    (A and B) Immunofluorescence for FOXO3 in ARC (arcuate nucleus), PVN

    Page 46 of 60Diabetes

  • 7

    (paraventricular nucleus) and VMH (ventromedial nucleus) sections (A) and

    integrated density quantification (B) in PSKO mice injected with Ad-scrambled (-

    Ad-shFOXO3) or Ad-shFOXO3 (+ Ad-shFOXO3);

    (C) Daily food intake;

    (D) Daily locomotor activity;

    (E and F) Immunofluorescence for α-MSH in PVN sections (E) and integrated density

    quantification (F).

    All studies were conducted in 16 to 18-weeks old male control and PSKO mice

    injected with Ad-scrambled (- Ad-shFOXO3) or Ad-shFOXO3 (+ Ad-shFOXO3).

    Data are expressed as mean ± SEM (n = 6-11/group). *: p < 0.05 for the effect of any

    group versus control mice injected without Ad-shFOXO3; #: p < 0.05 for the effect of

    with versus without Ad-shFOXO3 injection in PSKO mice for B and E.

    Figure S11. Levels of hypothalamic nuclear and cytoplasma GR, SGK1 and

    FOXO3 in mice treated with Dex.

    (A) Gr expression in hypothalamus;

    (B) GR and p-GR western blot and densitometric quantification in hypothalamus;

    (C) Immunofluorescence for POMC neurons (red), GR/SGK1/FOXO3 (green) and

    merge (yellow) in ARC (arcuate nucleus) sections from male POMC-tdTomato

    indicator mice;

    (D) Total and phosphorylated GR, SGK1 andFOXO3 western blot in nuclear and

    cytoplasm in hypothalamus.

    Page 47 of 60 Diabetes

  • 8

    Studies were conducted in 14 to 15-weeks old male wild-type mice treated without (-

    Dex) or with Dex (+ Dex) for 6 weeks (for A, B and D) or 9-weeks old male

    POMC-tdTomato indicator mice (for C). Data are expressed as ± SEM (n =

    6-11/group). *: p < 0.05 for the effect of with versus without Dex treatment group.

    Page 48 of 60Diabetes

  • 20

    25

    30

    0 1 2 3 4 5 6

    Time (weeks)

    Bo

    dy w

    eig

    ht

    (g)

    0

    2

    4

    6

    Fo

    od

    in

    tak

    e

    (g/d

    ay)

    0

    0.2

    0.4

    0.6

    0.8

    Light Dark Total

    EE

    (k

    ca

    l/h

    r)

    * *

    0

    500

    1000

    1500

    Light Dark Total Lo

    co

    mo

    tor

    acti

    vit

    y

    (co

    un

    ts)

    A B C

    E F G

    35

    36

    37

    38

    14:00 17:00

    Re

    cta

    l

    Te

    mp

    era

    ture

    (℃

    )

    * *

    H

    Arb

    itra

    ry

    un

    its

    0 50

    100 150

    *

    UCP1

    Actin

    Dex: - + - + - + I

    0

    100

    200

    Se

    rum

    NE

    (n

    g/l

    )

    *

    J

    Fig. S1

    0

    0.2

    0.4

    0.6

    Ab

    od

    om

    ina

    l

    fat

    ma

    ss

    (g

    ) *

    *

    0

    1

    2

    3

    0 1 2 3 4 5 6

    Time (weeks) F

    at

    ma

    ss

    (g

    )

    - Dex + Dex + Dex Dex -

    D

    15

    20

    25

    Le

    an

    ma

    ss

    (g

    )

    Time (weeks)

    *

    0 1 2 3 4 5 6

    Page 49 of 60 Diabetes

  • A

    Fig. S2

    B

    0

    50

    100

    PO

    MC

    / t

    dTo

    ma

    to

    co

    -lo

    ca

    lisa

    tio

    n (

    %)

    tdTomato

    POMC

    100 um 50 um

    Merge

    C

    0

    50

    100

    150

    *

    SG

    K1

    /td

    To

    ma

    to

    co

    localizati

    on

    (%

    )

    *

    SG

    K1

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    0

    50

    100

    150 D E

    F G

    100 um

    SGK1

    Control PSKO

    SGK1

    Control PSKO

    100 um

    SG

    K1

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    0

    50

    100

    150

    SG

    K1 (

    arb

    itra

    ry

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    0

    50

    100

    150

    PV

    N

    VM

    H

    0

    10

    20

    30

    Le

    an

    ma

    ss

    (g

    ) H

    Control PSKO

    Page 50 of 60Diabetes

  • Fig. S3

    Control PSKO

    A B

    0

    100

    200

    300

    400

    1 2 3 4 5 6 7 8 9 10 11 12

    ARC section

    PO

    MC

    ne

    uro

    n

    co

    un

    ts

    0

    50

    100

    Co

    rtic

    os

    tero

    ne

    (ng

    /ml)

    0

    1

    2

    3

    4

    Gro

    wth

    ho

    rmo

    ne

    (ng

    /ml)

    C D

    0

    50

    100

    150

    Re

    lati

    ve

    PO

    MC

    ne

    uro

    n a

    rea

    (%

    )

    Page 51 of 60 Diabetes

  • A B C D

    G

    0

    1000

    2000

    Light Dark Total Lo

    co

    mo

    tor

    ac

    tivit

    y

    (co

    un

    ts)

    E F

    Control PSKO

    Fig. S4

    Re

    cta

    l

    tem

    pe

    ratu

    re (℃

    )

    35

    36

    37

    *

    *

    0

    1

    2

    3

    Fa

    t m

    as

    s (

    g)

    0

    2

    4

    6

    Fo

    od

    In

    tak

    e

    (g/d

    ay)

    0

    0.4

    0.8

    1.2

    Light Dark Total

    RE

    R

    * * *

    0

    0.2

    0.4

    0.6

    Light Dark Total

    EE

    (k

    ca

    l/h

    r)

    * * *

    Age (week)

    15

    20

    25

    30

    6 8 10 12 14 16

    Bo

    dy w

    eig

    ht

    (g)

    *

    Control

    PSKO

    Page 52 of 60Diabetes

  • Fig. S5

    Control + T PSKO-ER + T

    A B

    0

    0.4

    0.8

    1.2

    Light Dark Total

    RE

    R

    *

    0

    500

    1000

    1500

    Lo

    co

    mo

    tor

    ac

    tivit

    y

    (co

    un

    ts)

    Light Dark Total 35

    36

    37

    Re

    cta

    l

    tem

    pe

    ratu

    re (℃

    )

    *

    C D

    H F G

    0

    0.2

    0.4

    0.6

    Light Dark Total

    EE

    (k

    ca

    l/h

    r)

    * * *

    E

    0

    2

    4

    6

    Fo

    od

    in

    tak

    e

    (g/d

    ay)

    15

    20

    25

    30

    5 7 9 11 13

    Bo

    dy w

    eig

    ht

    (g) * * *

    Age (weeks)

    Tamoxifen

    PSKO-ER Control

    Age (weeks)

    * *

    0

    1

    2

    3

    7 9 10 11

    Fat

    mass (

    g)

    0

    0.1

    0.2

    0.3

    Ab

    do

    min

    al

    fat

    ma

    ss

    (g

    )

    *

    Page 53 of 60 Diabetes

  • 0

    50

    100

    PO

    MC

    / m

    Ch

    err

    y

    co

    -lo

    ca

    lisa

    tio

    n (

    %)

    200 um 50 um

    mCherry

    POMC

    Fig. S6

    A B

    Merge

    Co

    ntr

    ol

    PS

    OE

    C D E F

    G H

    Co

    ntr

    ol

    PS

    OE

    100 um

    SGK1

    100 um

    SGK1

    Control PSOE

    SG

    K1

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    0

    50

    100

    150

    SG

    K1

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    0

    50

    100

    150

    mCherry SGK1

    100 um

    *

    SG

    K1

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    ) 0

    100

    200

    300

    Control PSOE

    Merge

    PVN

    VM

    H

    0

    1

    2

    3

    Fa

    t m

    as

    s (

    g)

    J I

    0

    10

    20

    30

    Le

    an

    ma

    ss

    (g

    )

    Page 54 of 60Diabetes

  • Control Dex

    Control + Dex

    PSKO Dex

    PSKO + Dex

    - -

    0

    10

    20

    30

    40

    0 6

    Time (weeks)

    Bo

    dy w

    eig

    ht

    (g) * * * *

    #

    0

    1

    2

    3

    4

    5

    0 6

    Time (weeks) F

    at

    ma

    ss

    (g

    )

    * * * * * #

    0

    10

    20

    30

    Le

    an

    ma

    ss

    (g

    )

    0 6

    Time (weeks)

    * #

    0

    0.5

    1.0

    Ab

    do

    min

    al

    fat

    ma

    ss

    (g

    )

    * * *

    0

    2

    4

    6

    Fo

    od

    in

    tak

    e

    (g/d

    ay)

    0

    0.2

    0.4

    0.6

    0.8

    Light Dark Total

    EE

    (k

    ca

    l/h

    r)

    * * * * * * * * * #

    A B C

    D E F

    Fig. S7 Page 55 of 60 Diabetes

  • Fig. S8

    B

    C

    100 um

    POMC

    Control PSKO

    *

    Pc1/3 Pc2 Cpe Pam Prcp 0

    50

    100

    150

    Pomc Re

    lati

    ve

    mR

    NA

    (%

    ) PSKO Control

    A

    0

    50

    100

    150

    *

    PO

    MC

    (a

    rbit

    rary

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    0

    50

    100

    150 R

    ela

    tive

    mR

    NA

    (%

    )

    Sgk1 Pomc

    * *

    D

    *

    0

    200

    400

    600

    800

    Sgk1 Pomc Re

    lati

    ve

    mR

    NA

    (%

    )

    *

    E Ad-shSGK1 + Ad-shSGK1

    - CA SGK1

    + CA SGK1

    -

    Page 56 of 60Diabetes

  • Fig. S9

    Actin

    FOXO3

    P-FOXO3

    Ad-shSGK1: - + - + - +

    SGK1

    B

    D

    Ad-shSGK1 siFOXO3

    + Ad-shSGK1 siFOXO3

    Ad-shSGK1 + siFOXO3

    + Ad-shSGK1 + siFOXO3

    0

    100

    200

    Sgk1 Foxo3 Pomc Re

    lati

    ve

    mR

    NA

    (%

    )

    * * * * *

    * # #

    0

    50

    100

    150

    Arb

    itra

    ry u

    nit

    s

    * * *

    p-FOXO3

    FOXO3

    SGK1

    Actin

    CASGK1: - + - + - +

    0

    100

    200

    Arb

    itra

    ry u

    nit

    s

    * *

    C

    *

    Ad-shSGK1

    + Ad-shSGK1

    -

    CA SGK1

    + CA SGK1

    -

    - -

    -

    -

    A

    FOXO3

    p-FOXO3

    PSKO: - + + - +

    Actin

    0

    50

    100

    150

    Arb

    itra

    ry

    un

    its

    * *

    PSKO Control

    -

    Page 57 of 60 Diabetes

  • *

    FO

    XO

    3 (

    arb

    itra

    ry

    flu

    ore

    sc

    en

    ce

    un

    its

    )

    * #

    B

    Fig. S10

    A

    FOXO3

    - Ad-shFOXO3 + Ad-shFOXO3 - Ad-shFOXO3 + Ad-shFOXO3

    Control PSKO Control PSKO