original article liver kinase b1 is required for white ... · liver kinase b1 is required for white...

12
Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang, 1 Qilong Wang, 1 Ping Song, 1 and Ming-Hui Zou 1,2 White adipose tissue (WAT) is not only a lipogenic and fat storage tissue but also an important endocrine organ that regulates energy homeostasis, lipid metabolism, appetite, fertility, and immune and stress responses. Liver kinase B1 (LKB1), a tumor suppressor, is a key regulator in energy metabolism. However, the role of LKB1 in adipogenesis is unknown. The current study aimed to determine the contributions of LKB1 to adipogenesis in vivo. Using the Fabp4- Cre/loxP system, we generated adipose tissuespecic LKB1 knockout (LKB1 ad2/2 ) mice. LKB1 ad2/2 mice exhibited a reduced amount of WAT, postnatal growth retardation, and early death be- fore weaning. Further, LKB1 deletion markedly reduced the levels of insulin receptor substrate 1 (IRS1), peroxisome proliferatoractivated receptor g, CCAAT/enhancer-binding protein a, and phos- phorylated AMP-activated protein kinase (AMPK). Consistent with these results, overexpression of constitutively active AMPK par- tially ablated IRS1 degradation in LKB1-decient cells. LKB1 deletion increased the levels of F-box/WD repeat-containing pro- tein (Fbw) 8, the IRS1 ubiquitination E3 ligase. Silencing of Fbw8 increased IRS1 levels. Finally, promoter analysis and DNA chroma- tin immunoprecipitation analysis identied three sterol regulatory element (SRE) sites in the Fbw8 promoter, where SRE-binding pro- tein 1c binds and induces the expression of Fbw8. Taken together, these data indicate that LKB1 controls IRS1-dependent adipogenesis via AMPK in WAT. Diabetes 62:23472358, 2013 W hite adipose tissue (WAT) is not only an ac- tively lipogenic and fat storage tissue, but is also an important endocrine organ that secrets critical hormones and factors such as leptin, adiponectin, and tumor necrosis factor-a that reg- ulate energy homeostasis, lipid metabolism, appetite, and fertility, as well as immune and stress responses (1). In addition, dysfunctional WAT is reported to cause severe metabolic diseases such as obesity, type 2 diabetes, cancer cachexia, and lipodystrophies (2). WAT generation is controlled by a complex network of transcription factors, including numerous transcriptional activators, coactivators, and repressors (3). Two transcription factors, peroxisome proliferator-activated receptor g (PPARg) and CCAAT/ enhancer-binding protein a (C/EBPa), are considered master regulators of adipogenesis (3). Deletion of PPARg or C/EBPa in mice results in an almost complete absence of WAT (1,4). PPARg and C/EBPa positively regulate each others expression and cooperate to control adipogenesis (5). However, the factors and the underlying mechanisms that regulate the induction of PPARg and C/EBPa ex- pression during adipogenesis remain poorly understood. The insulin receptor substrate (IRS)/Akt signaling cas- cade is critical for adipogenesis. The loss of individual IRS proteins inhibits adipogenesis, with the order of im- portance being IRS1 . IRS3 . IRS2 . IRS4 (6). IRS1 knockout (KO) mice have signicant lipodystrophy, al- though brown adipose tissue (BAT) is relatively unaffected (7), and IRS1-transgenic mice show increased fat mass (8). Moreover, a novel spontaneous mutation in Irs1 in mice results in impaired adipogenesis (9), further conrming the important role of IRS1 in adipogenesis. Akt regulates the process of adipocyte differentiation through the induction of PPARg expression (10). Mice that lack both Akt1 and Akt2 are lipoatrophic, and mouse embryonic broblasts (MEFs) from these animals cannot undergo differentiation (10). The insulin/Akt-resistant state is partly caused by the loss of the IRS1 protein due to its enhanced degradation by the ubiquitin proteasome system (1113). Recently, IRS1 was reported to be degraded by the Cullin 7 (CUL7)/E3 ubiquitin ligase complex containing the F-box/WD repeat- containing protein (Fbw) 8-substrate-targeting subunit, Skp1, and the really interesting new gene nger protein regulator of cullins 1 (ROC1). Overexpression of CUL7 or Fbw8 could therefore lead to IRS1 degradation (14). However, the role of IRS1 E3 ligase in adipogenesis is unknown. The liver kinase B1 (LKB1) protein (also known as Stk11) is a serine/threonine protein kinase that was rst described as a tumor suppressor gene mutated in Peutz- Jeghers syndrome, a familial form of cancer (15,16). LKB1 is ubiquitously expressed in mammalian cells and is acti- vated in a complex with two scaffolding proteins: STE20- related adaptor (STRAD) protein and mouse protein 25 (MO25) (17). Currently, LKB1 is also implicated as a cen- tral regulator of cell polarity and energy metabolism in a variety of systems. These functions of LKB1 are thought to be achieved via the direct phosphorylation of the AMP- activated protein kinase (AMPK) family of proteins, con- sisting of 14 members (18). The roles of LKB1 in many tissues have been researched in the last decade using tissue-specic LKB1 KO mice. For example, deletion of LKB1 in smooth muscle produced benign gastrointestinal hamartomas (16). This result indicates that LKB1 has a role as a tumor suppressor gene. KO of LKB1 in skeletal muscle prevented AMPK activation and glucose uptake in response to contraction and phenformin (19), indicating its important function in energy metabolism. Loss of LKB1 in adult b-cells increased b-cell mass and abnormal polarity (20), conrming its signicance in cell polarity. However, the role of LKB1 in adipogenesis has not been previously demonstrated. Lkb1 KO embryos die before embryonic day 11 because of severe developmental defects, including From the 1 Section of Molecule Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and the 2 Department of Biochemistry and Molecular Biology, University of Okla- homa Health Sciences Center, Oklahoma City, Oklahoma. Corresponding author: Ming-Hui Zou, [email protected]. Received 6 September 2012 and accepted 3 February 2013. DOI: 10.2337/db12-1229 This article contains Supplementary Data online at http://diabetes .diabetesjournals.org/lookup/suppl/doi:10.2337/db12-1229/-/DC1. Ó 2013 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for prot, and the work is not altered. See http://creativecommons.org/licenses/by -nc-nd/3.0/ for details. diabetes.diabetesjournals.org DIABETES, VOL. 62, JULY 2013 2347 ORIGINAL ARTICLE

Upload: others

Post on 16-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

Liver Kinase B1 Is Required for White Adipose TissueGrowth and DifferentiationWencheng Zhang,

1Qilong Wang,

1Ping Song,

1and Ming-Hui Zou

1,2

White adipose tissue (WAT) is not only a lipogenic and fat storagetissue but also an important endocrine organ that regulates energyhomeostasis, lipid metabolism, appetite, fertility, and immune andstress responses. Liver kinase B1 (LKB1), a tumor suppressor, isa key regulator in energy metabolism. However, the role of LKB1 inadipogenesis is unknown. The current study aimed to determinethe contributions of LKB1 to adipogenesis in vivo. Using the Fabp4-Cre/loxP system, we generated adipose tissue–specific LKB1knockout (LKB1ad2/2) mice. LKB1ad2/2 mice exhibited a reducedamount of WAT, postnatal growth retardation, and early death be-fore weaning. Further, LKB1 deletion markedly reduced the levelsof insulin receptor substrate 1 (IRS1), peroxisome proliferator–activated receptor g, CCAAT/enhancer-binding protein a, and phos-phorylated AMP-activated protein kinase (AMPK). Consistent withthese results, overexpression of constitutively active AMPK par-tially ablated IRS1 degradation in LKB1-deficient cells. LKB1deletion increased the levels of F-box/WD repeat-containing pro-tein (Fbw) 8, the IRS1 ubiquitination E3 ligase. Silencing of Fbw8increased IRS1 levels. Finally, promoter analysis and DNA chroma-tin immunoprecipitation analysis identified three sterol regulatoryelement (SRE) sites in the Fbw8 promoter, where SRE-binding pro-tein 1c binds and induces the expression of Fbw8. Taken together,these data indicate that LKB1 controls IRS1-dependent adipogenesisvia AMPK in WAT. Diabetes 62:2347–2358, 2013

White adipose tissue (WAT) is not only an ac-tively lipogenic and fat storage tissue, but isalso an important endocrine organ thatsecrets critical hormones and factors such as

leptin, adiponectin, and tumor necrosis factor-a that reg-ulate energy homeostasis, lipid metabolism, appetite, andfertility, as well as immune and stress responses (1). Inaddition, dysfunctional WAT is reported to cause severemetabolic diseases such as obesity, type 2 diabetes, cancercachexia, and lipodystrophies (2). WAT generation iscontrolled by a complex network of transcription factors,including numerous transcriptional activators, coactivators,and repressors (3). Two transcription factors, peroxisomeproliferator-activated receptor g (PPARg) and CCAAT/enhancer-binding protein a (C/EBPa), are consideredmaster regulators of adipogenesis (3). Deletion of PPARgor C/EBPa in mice results in an almost complete absenceof WAT (1,4). PPARg and C/EBPa positively regulate each

other’s expression and cooperate to control adipogenesis(5). However, the factors and the underlying mechanismsthat regulate the induction of PPARg and C/EBPa ex-pression during adipogenesis remain poorly understood.

The insulin receptor substrate (IRS)/Akt signaling cas-cade is critical for adipogenesis. The loss of individual IRSproteins inhibits adipogenesis, with the order of im-portance being IRS1 . IRS3 . IRS2 . IRS4 (6). IRS1knockout (KO) mice have significant lipodystrophy, al-though brown adipose tissue (BAT) is relatively unaffected(7), and IRS1-transgenic mice show increased fat mass (8).Moreover, a novel spontaneous mutation in Irs1 in miceresults in impaired adipogenesis (9), further confirming theimportant role of IRS1 in adipogenesis. Akt regulates theprocess of adipocyte differentiation through the inductionof PPARg expression (10). Mice that lack both Akt1 andAkt2 are lipoatrophic, and mouse embryonic fibroblasts(MEFs) from these animals cannot undergo differentiation(10). The insulin/Akt-resistant state is partly caused by theloss of the IRS1 protein due to its enhanced degradation bythe ubiquitin proteasome system (11–13). Recently, IRS1was reported to be degraded by the Cullin 7 (CUL7)/E3ubiquitin ligase complex containing the F-box/WD repeat-containing protein (Fbw) 8-substrate-targeting subunit,Skp1, and the really interesting new gene finger proteinregulator of cullins 1 (ROC1). Overexpression of CUL7 orFbw8 could therefore lead to IRS1 degradation (14).However, the role of IRS1 E3 ligase in adipogenesis isunknown.

The liver kinase B1 (LKB1) protein (also known asStk11) is a serine/threonine protein kinase that was firstdescribed as a tumor suppressor gene mutated in Peutz-Jeghers syndrome, a familial form of cancer (15,16). LKB1is ubiquitously expressed in mammalian cells and is acti-vated in a complex with two scaffolding proteins: STE20-related adaptor (STRAD) protein and mouse protein 25(MO25) (17). Currently, LKB1 is also implicated as a cen-tral regulator of cell polarity and energy metabolism ina variety of systems. These functions of LKB1 are thoughtto be achieved via the direct phosphorylation of the AMP-activated protein kinase (AMPK) family of proteins, con-sisting of 14 members (18). The roles of LKB1 in manytissues have been researched in the last decade usingtissue-specific LKB1 KO mice. For example, deletion ofLKB1 in smooth muscle produced benign gastrointestinalhamartomas (16). This result indicates that LKB1 has arole as a tumor suppressor gene. KO of LKB1 in skeletalmuscle prevented AMPK activation and glucose uptake inresponse to contraction and phenformin (19), indicating itsimportant function in energy metabolism. Loss of LKB1 inadult b-cells increased b-cell mass and abnormal polarity(20), confirming its significance in cell polarity. However,the role of LKB1 in adipogenesis has not been previouslydemonstrated. Lkb1 KO embryos die before embryonic day11 because of severe developmental defects, including

From the 1Section of Molecule Medicine, Department of Medicine, Universityof Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and the2Department of Biochemistry and Molecular Biology, University of Okla-homa Health Sciences Center, Oklahoma City, Oklahoma.

Corresponding author: Ming-Hui Zou, [email protected] 6 September 2012 and accepted 3 February 2013.DOI: 10.2337/db12-1229This article contains Supplementary Data online at http://diabetes

.diabetesjournals.org/lookup/suppl/doi:10.2337/db12-1229/-/DC1.� 2013 by the American Diabetes Association. Readers may use this article as

long as the work is properly cited, the use is educational and not for profit,and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.

diabetes.diabetesjournals.org DIABETES, VOL. 62, JULY 2013 2347

ORIGINAL ARTICLE

Page 2: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

impaired neural tube closure and somitogenesis, mesen-chymal tissue cell death, and defective vasculature (21),therefore limiting research into the role of LKB1 in adiposedifferentiation. In the current study, we used the Cre/LoxP

system to generate an adipocyte-specific LKB1 deletion inmice (LKB1ad2/2), and we report that LKB1ad2/2 miceshowed impaired WAT differentiation and postnatal growthretardation.

FIG. 1. Severe growth retardation in adipose tissue–specific LKB1 KO mice. A: RT-PCR to detect LKB1 mRNA levels in WAT and BAT of WT mice(n = 3). B: Western blot analysis to detect LKB1 protein levels in WAT and BAT of mice. C and D: Western blot to detect p-AMPKa T172 levels inWT and LKB1-deficient WAT and BAT. The blot is representative of three blots obtained from three independent experiments. E: Photographs ofrepresentative 18-day-old male WT and LKB1ad2/2

mice. F: A typical growth curve for male mice from postnatal days 1–17 (n = 5). G: Kaplan-Meiersurvival plot of WT and LKB1ad2/2

mice. H: Blood glucose levels were measured at different ages for WT and LKB1ad2/2mice (n = 5). *P < 0.05

compared with WAT from WT. AU, arbitrary units. (A high-quality color representation of this figure is available in the online issue.)

LKB1 AND ADIPOGENESIS

2348 DIABETES, VOL. 62, JULY 2013 diabetes.diabetesjournals.org

Page 3: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

RESEARCH DESIGN AND METHODS

Reagents. Antibodies to C/EBPa (2295S), PPARg (2295S), phosphorylated (p-)AMPKa T172 (2535 L), Fabp4 (3544S), p-Akt Thr308 (9275S), p-Akt Ser473(9271S), Akt (9272S), p-S6K1 T389 (9234S), S6K1 (2708S) and p-FoxO1 Ser256(9461S) were from Cell Signaling Technology (Danvers, MA). Antibodies toLKB1 (sc-32245), GAPDH (sc-137179), and sterol regulatory element (SRE)–binding protein 1c (SREBP-1; sc-8984) were purchased from Santa Cruz Bio-technology (Santa Cruz, CA). Antibodies to uncoupling protein 1 (AB1426),p-IRS1 Tyr612 (07-846), and IRS1 (06-248) were purchased from Millipore(Temecula, CA). Anti-Fbw8 (ab85647) and p-IRS1 S636/639 (ab53038) were fromAbcam (Cambridge, U.K.). Anti-fatty acid synthetase (NB400114) was fromNovus Biologicals (Littleton, CO). Anti-CUL7 (13738–1-AP) was from Pro-teintech (Chicago, IL). Anti-FLAG (F7425) antibody was from Sigma-Aldrich (St.Louis, MO). The vectors expressing FLAG-tagged SREBP-1a, SREBP-1c, andSREBP-2 were purchased from Addgene (Cambridge, MA). FuGene HD trans-fection reagent was from Roche Applied Science (Indianapolis, IN). The Dual-Luciferase Assay Kit was purchased from Promega (Madison, WI).Generation of adipocyte-specific LKB1-deleted (LKB1ad2/2

) mice. Togenerate adipose tissue-specific disruption of the LKB1 gene (LKB1ad2/2),LKB1flox/flox mice were crossbred with transgenic mice expressing the Crerecombinase under the control of the Fabp4 promoter/enhancer. PCR-basedgenotyping of LKB1ad2/2 mice was performed with the following primers:Fabp4-Cre, 59-TCTCACGTACTGACGGTGG-39 and 59-CCAGCTTGCATGATCTCC-39;for the upstream 59 loxP site, 59-TCTAACAATGCGCTCATCGTCATCCTCGGC-39;and for the downstream 39 loxP site, 59-GAGATGGGTACCAGGAGTTGGGGC-39.The deletion of exons 3–6 of LKB1 was verified as described previously (22).The animals were housed in a controlled environment (20 6 2°C, 12-h/12-hlight/dark cycle) and had free access to water and standard chow diet. Allexperiments were conducted in accordance with the Animal Care and UseCommittee of the University of Oklahoma Health Science Center.

Preparation of primary MEFs and induction of adipocyte differentiation.

Primary MEFs were isolated from embryos of LKB1flox/flox mice at 13.5 dayspostcoitum. Cells were cultured at 37°C in high-glucose Dulbecco’s modifiedEagle’s medium (GIBCO BRL, Carlsbad, CA) supplemented with 10% (volumefor volume [v/v]) heat-inactivated FBS (Sigma-Aldrich) and 100 U/ml penicillin/streptomycin (GIBCO BRL). LKB1flox/flox MEF cells were infected with adeno-virus expressing lacZ (wild-type [WT]) or Cre recombinase (LKB1 KO). Two dayslater, the medium was replaced with the standard differentiation induction me-dium containing 0.5 mmol/L methylisobutylxanthine, 1 mmol/L dexamethasone,10 mg/mL insulin, 10 mmol/L troglitazone (all from Sigma), and 10% (v/v) FBS.This medium was changed every other day. For the preparation of proteins forWestern blot analysis, cells were harvested at 0, 2, 5, and 8 days of differentia-tion. AMPKa1- and -a2–deficient MEFs were prepared using the same procedure.Oil Red O staining. Oil Red O staining was performed at 8 days after celldifferentiation. Briefly, cells were washed twice with PBS and fixed with 10%formalin in PBS for 1 h. They were washed three times with water and stainedwith Oil Red O (six parts of 0.6% Oil Red O dye in isopropanol and four parts ofwater) for 1 h. Excess stain was removed by washing with water, and thenmicroscopic images were recorded.Cellular triglyceride levels. Triglycerides (TGs) were extracted from culturedcells using hexane/isopropanol (3:2 [v/v]). The lipid extracts were dried undera gentle stream of nitrogen and dissolved in cold isopropanol and TG levelsmeasured using a commercially available kit (Thermo Scientific, Waltham, MA).Histology. All fat tissues were fixed in 10% neutral buffered formalin andembedded in paraffin. Five-micrometer sections were cut, stained with he-matoxylin and eosin (H&E), and photographed under a light microscope.Serum profiling.Blood was taken from anesthetized mice. The levels of leptin,adiponectin, IGF-1, and insulin were measured by an ELISA kit (R&D Systems,Minneapolis, MN). Serum TG and cholesterol levels were measured using thecorresponding kit from Thermo Scientific.

FIG. 2. Deletion of LKB1 in mice inhibits the formation of WAT. A: Appearance of the epidydimal fat pad and BAT of WT and LKB1ad2/2mice at the

age of 18 days. B: Weight of various organs relative to overall body weight in WT and LKB1ad2/2mice at 18 days after birth. C: WAT and BAT from

18-day-old male WT and its LKB1ad2/2littermate were stained with H&E. Scale bar, 50 mm. D: Adipocyte size was measured using images of WAT

sections (n > 100 cells/group). *P < 0.05 compared with WT. (A high-quality color representation of this figure is available in the online issue.)

W. ZHANG AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 62, JULY 2013 2349

Page 4: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

Western blot. Fresh fat tissues were collected and immediately frozen in liquidnitrogen. Proteins from whole-cell lysates were extracted in lysis buffer withsonication and analyzed using Western blot. The intensities (density three area)of individual bands were measured by densitometry (Model GS-700, ImagingDensitometer; Bio-Rad, Hercules, CA).Quantitative real-time RT-PCR. Total RNA was extracted from frozen tis-sues (kept at 280°C) using the Tri Reagent (Ambion, Austin, TX), and 1 mg ofRNA was reverse-transcribed into cDNA using iScript cDNA Synthesis Kits(Bio-Rad). PCR amplification was performed using the SYBR PCR mix (Bio-Rad). The oligonucleotides used in PCR analysis were as follows: LKB1,59-AGCTGCGCAGGATCCCCAAT-39 and 59-TGGCACACAGGGAAGCGCTT-39; IRS1,59-ACTATGCCAGCATCAGCTTC-39 and 59-AAGGAGGATTTGCTGAGGTC-39; Fbw8,59-GCCACGGAGCCCGAGCCGCTGG-39 and 59-GCACCTCGTCTTCAGCAATCAC-39;and GAPDH, 59-TTGTCAAGCTCATTTCCTGGTATG-39 and 59-GCCATGTAGGC-CATGAGGTC-39.Chromatin immunoprecipitation. Chromatin immunoprecipitation (ChIP)analyses of MEF cells were performed using the Magna ChIP A/G ChromatinImmunoprecipitation Kit (Millipore), following the manufacturer’s recom-mendations. LKB1flox/flox MEF cells were first infected with adenovirusexpressing lacZ (WT) or Cre recombinase (LKB1 KO) for 48 h and then cross-linked using formaldehyde; DNA was then sheared by sonication and in-cubated with 1 mg of normal rabbit IgG (sc-2027) or anti–SREBP-1 (sc-8984).

Immunoprecipitation was performed with the magnetic beads included in thekit. For PCR, 2 ml of the 50 ml total immunoprecipitated DNA was analyzedwith the following oligonucleotides: SRE1, 59-CTCAGAGAGGTCTTCATGA-CAGCT-39 and 59-TGGCGCACGCCTTTAATCCCAGCA-39; SRE2, 59-TTGCCTA-CATCAGCACCCTGTTCT-39 and 59-CTACTTGATATTTCCACCGATGTA-39; andSRE3, 59-CTGATGAAGGCTCAGGCCTCTGAC-39 and 59-TGATGTAGGCAAGGG-TGGCTAAGG-39.DNA constructs and luciferase reporter assays. The DNA fragment fromthe mouse Fbw8 promoter cloned into pGL3Basic (Promega, Madison, WI) togenerate the WT-luc construct was amplified by PCR using the following oli-gonucleotides: 59-GCTAGCCACAAAGGCATGTACACAGGTATA-39 and 59-AAG-CTTACTGTCCATCCGCCGGAAGTGTCC-39. Mutant constructs of the predictedSRE sites in the Fbw8 promoter were generated by site-directed mutagenesis(Promega) using the following primers: SRE1, 59-CCTGGCTGTCCTGGAAC-CAAATCCATAGACCAGGCTGG-39 and 59-CCAGCCTGGTCTATGGATTTGGTTC-CAGGACAGCCAGG-39; SRE2, 59-TCTGGGAACATGGACTTAGGCAAACCCAATCTCACGTCCCATTG-39 and 59-CAATGGGACGTGAGATTGGGTTTGCCTAAGTCCATGTTCCCAGA-39; and SRE3, 59-ACAGATGACATCACTGTTTACTTTTGGAAACCCAGGACAGCCAGCGACAGC-39 and 59-GCTGTCGCTGGCTGTCCTGGGTT-TCCAAAAGTAAACAGTGATGTCATCTGT-39. For the luciferase assay, the lu-ciferase reporter plasmids along with the SREBP-1c plasmid were transfectedinto MEF cells in 24-well plates using FuGene HD reagent (Roche Applied

FIG. 3. LKB1 KO in adipose tissue affects hormone secretion and TG levels. Serum adiponectin (A), leptin (B), IGF-1 (C), insulin (D), TG (E), andcholesterol (F) levels were measured from 18-day-old male WT and LKB1ad2/2

mice (n = 5). *P < 0.05 compared with WT.

LKB1 AND ADIPOGENESIS

2350 DIABETES, VOL. 62, JULY 2013 diabetes.diabetesjournals.org

Page 5: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

Science). The pRL-TK plasmid carrying the Renilla luciferase under the con-trol of the thymidine kinase promoter was also cotransfected as an internalcontrol for transfection efficiency. Cells were harvested 48 h after trans-fection, and luciferase activities were then analyzed using the Dual-LuciferaseAssay Kit (Promega).

Statistics.Mean6 SEM values were analyzed using GraphPad Prism (version3; GraphPad Software Inc., San Diego, CA). Statistical comparisons betweengroups were performed using the Student t test. Curves were compared usingtwo-way ANOVA and Bonferroni posttests. Groups were considered significantlydifferent at P values #0.05.

FIG. 4. Impaired adipogenesis in LKB1-deficient WAT. A: Western blot analysis of adipocyte-specific genes and adipogenic transcription factors forWAT and BAT from WT and 18-day-old male LKB1ad2/2

mice. B and C: Western blot analysis of IRS1/Akt signaling in WAT from WT and LKB1ad2/2

mice. The blot is a representative of three blots obtained from three independent experiments. D: RT-PCR to detect IRS1 mRNA levels in WT andLKB1-deficient WAT (n = 3). E: Western blot analysis to detect Fbw8 and Cul7 protein levels in WT and LKB1-deficient WAT. F: RT-PCR to detectFbw8 mRNA level in WT and LKB1-deficient WAT (n = 3). *P < 0.05 compared with WT.

W. ZHANG AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 62, JULY 2013 2351

Page 6: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

FIG. 5. LKB1 regulates adipogenesis through the IRS1/Akt signaling pathway. A: LKB1flox/flox MEF cells were infected with adenovirus expressinglacZ (WT) or Cre recombinase (LKB1 KO) for 2 days and then induced to differentiate into adipocytes. Oil Red O staining was performed after 8days. B: TG levels were measured on day 8 of differentiation in WT and LKB1-deficient cells (n = 4). C and D: Cells were harvested at 0, 2, 5, and 8days of differentiation, and Western blot analysis was performed to detect adipogenic proteins and IRS1/Akt signaling. E: LKB1flox/flox MEF cellswere first infected with adenovirus expressing lacZ (WT) or Cre recombinase (LKB1 KO) for 24 h and then infected with CA-Akt or GFP virus foranother 24 h. After that, cells were induced to differentiate into adipocytes. Oil Red O staining was performed after 8 days. F: Cells were harvested

LKB1 AND ADIPOGENESIS

2352 DIABETES, VOL. 62, JULY 2013 diabetes.diabetesjournals.org

Page 7: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

RESULTS

Severe growth retardation in adipose tissue–specificLKB1 KO mice. We first investigated the mRNA andprotein levels of LKB1 in WAT and BAT from WT mice. Asdepicted in Fig. 1A, the mRNA levels of LKB1 in WAT werefourfold higher than those in BAT. Compared with theweak staining seen for LKB1 in BAT, higher expression ofLKB1 proteins was found in WAT (Fig. 1B), suggesting thatLKB1 is mainly present in WAT.

To further elucidate the role of LKB1, we generatedmice containing an adipose tissue–specific LKB1 deletionusing mice containing the floxed LKB1 allele (22). BecauseFabp4, like LKB1, is mainly expressed in WAT, we crossedLKB1flox/flox with Fabp4-Cre mice to generate WAT-specificLKB1 KO (LKB1ad2/2) mice. The LKB1flox/flox/Cre2 micewere used as WT controls. As expected, LKB1 was notdetectable in the WAT and remained low in the BAT, ofLKB1ad2/2 mice (Fig. 1C). Consistent with this, the level ofphospho-AMPKa (Thr172), one of the most important sub-strates of LKB1, was barely detectable in the WAT ofLKB1ad2/2 mice, in contrast to the unaltered levels of p-AMPK seen in the BAT of LKB1ad2/2 mice (Fig. 1C and D).Taken together, our results suggest that LKB1 is essential formaintaining AMPK Thr172 phosphorylation in WAT in vivo.

Compared with their WT counterparts (Fig. 1E), LKB1ad2/2

mice exhibited severe postnatal growth retardation, start-ing at day 4 after birth. The body weights of LKB1ad2/2

mice at birth and for the first 3 days after birth were in-distinguishable from their littermates (Fig. 1F). However,from day 5 after birth, all LKB1ad2/2 mice had significantlyreduced body weights relative to those of their WT coun-terparts (Fig. 1F). At day 17, male LKB1ad2/2 mice weighed49.5% less than their WT littermates (5.15 6 0.45 vs.10.20 6 0.52 g, n = 5; P , 0.001). Importantly, LKB1ad2/2

mice began to die at day 3 after birth. None of theLKB1ad2/2 mice survived beyond postnatal day 19 (Fig.1G), whereas all WT pups survived. The blood glucoselevels of LKB1ad2/2 mice measured at days 3, 9, and 18were markedly lower than those of WT mice (Fig. 1H).Deletion of LKB1 in mice inhibits the formation ofWAT. Next, we examined the effect of LKB1 deletion onWAT and BAT. Necropsy analysis revealed that theamount of WAT, including subcutaneous, visceral, or go-nadal fat tissue, was severely reduced in LKB1ad2/2 micecompared with their age-matched littermates. Fig. 2Ashows epididymal fat from 18-day-old WT and LKB1ad2/2

mice. The 18-day-old male WT mice had a mean value of9.5% white fat of body weight, whereas LKB1ad2/2 micehad 1.27% white fat of body weight (Fig. 2B). The amountof BAT was also smaller in LKB1ad2/2 mice than in WT(Fig. 2A); however, the ratio of BAT to body weight wasnot lower in the LKB1ad2/2 mice (Fig. 2B). Histologicalanalyses of WAT and BAT using H&E staining showed thatwhite adipocytes were smaller in LKB1ad2/2 mice than inWT mice (Fig. 2C and D). In contrast, there was no dif-ference in the morphology of BAT between WT andLKB1ad2/2 mice, further confirming that deletion of LKB1affects only WAT but not BAT. Interestingly, the ratios ofliver, kidney, and heart tissue to body weight were similarbetween WT and LKB1ad2/2 mice (Fig. 2B). Histological

examination confirmed normal structures in both heartand liver in LKB1ad2/2 mice (Supplementary Fig. 1).LKB1 KO in adipose tissue affects hormone secretionand TG levels. In addition to its primary function as anenergy reservoir, WAT is an endocrine organ, secretingadipocytokines that regulate glucose and lipid metabolism(23). We therefore examined whether the defective adi-pogenesis seen in LKB1ad2/2 mice affected the secretionof critical hormones such as adiponectin, leptin, and IGF-1.Serum adiponectin levels were lower in LKB1ad2/2 mice(3.88 6 1.17 mg/mL) than in WT mice (6.08 6 1.28 mg/mL)(Fig. 3A). Circulating levels of leptin were barely detectablein 18-day-old LKB1ad2/2 mice (Fig. 3B). Similarly, serum IGF-1 levels were reduced by 89.6% in LKB1ad2/2 mice (Fig. 3C).However, insulin levels were similar between WT andLKB1ad2/2 mice (Fig. 3D).

Next, we detected whether reduced WAT altered thelevels of TG in LKB1ad2/2 mice. As shown in Fig. 3E, LKB1deletion in adipose tissue resulted in decreased serum TG,indicating that LKB1 deficiency leads to impaired adipo-genesis and lipogenesis in mice. However, LKB1 deficiencyin WAT did not change serum cholesterol levels (Fig. 3F)or the mRNA level of hydroxymethylglutaryl (HMG) CoAreductase, the rate-limiting enzyme in cholesterol synthe-sis, in the livers (Supplementary Fig. 2). Our results sup-port the notion that liver but not WAT is the central organin maintaining whole-body cholesterol homeostasis (24),and WAT is poor at de novo cholesterol biosynthesis (25).Impaired adipogenesis in LKB1-deficient WAT. Wefurther investigated how LKB1 deficiency affects adipo-genesis. We first monitored the levels of C/EBPa andPPARg, two known transcription factors for adipogenesis.As shown in Fig. 4A, the levels of C/EBPa and PPARg weresignificantly lower in LKB1-deficient WAT, but not in BAT,compared with WT. As expected, the levels of Fabp4 weresignificantly reduced in LKB1-deficient WAT, but not inBAT. The levels of uncoupling protein 1 (UCP1) were re-duced in LKB1-deficient WAT but not in BAT (Fig. 4A).

Insulin/Akt signaling is known to be essential in theprocess of adipogenesis; therefore, we determined whetherLKB1 deficiency altered the insulin/Akt pathway. As depictedin Fig. 4B, total IRS1 levels were lower in LKB1-deficientWAT. Consistent with this, the levels of phospho-Akt (T308and S473) were also lower upon LKB1 deletion (Fig. 4Band C).

To elucidate how LKB1 deletion causes a decrease inIRS1, we detected IRS1 mRNA levels using RT-PCR. Asshown in Fig. 4D, IRS1 mRNA levels were similar betweenWT and LKB1ad2/2 adipose tissues, suggesting that LKB1deletion may lower IRS1 levels by increasing its degrada-tion. Since IRS1 was reported to be degraded by the E3ubiquitin complex containing CUL7 and the Fbw8-substrate-targeting subunit (14), we tested whether LKB1deletion altered the levels of CUL7 and Fbw8. The level ofCUL7, which is the scaffolding protein in the complex,remained unchanged in LKB1-deficient WAT, whereas thelevel of Fbw8, the IRS1-target subunit, was higher (Fig.4E). Consistent with this, Fbw8 mRNA levels were eight-fold higher in LKB1-deficient adipose tissue than in WTadipose tissue (Fig. 4F).

after differentiation, and Western blot analysis was performed to detect adipogenic proteins. The blot is representative of three blots obtainedfrom three independent experiments. G: LKB1flox/flox MEF cells were infected with adenovirus expressing lacZ (WT) or Cre recombinase (LKB1 KO)for 2 days and then treated with insulin (100 nmol/L) for 15 min. Western blot analysis was performed to detect Akt pathway targets. H: Westernblot analysis was performed to detect p-S6K1 T389 and p–IRS1 S636/639 in WT and LKB1-deficient WAT. *P < 0.05 compared with WT.

W. ZHANG AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 62, JULY 2013 2353

Page 8: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

FIG. 6. LKB1 regulates Fbw8 expression in an AMPK-dependent manner. A and B: Time course of LKB1, Fbw8, and IRS1 expression and AMPKphosphorylation. LKB1flox/flox MEF cells were infected with adenovirus expressing Cre recombinase for different times, and then Western blot wasperformed to detect the protein levels of LKB1, p-AMPK, Fbw8, and IRS1 at the indicated times. The blot is representative of three blots obtainedfrom three independent experiments. C: LKB1flox/flox MEF cells were first transfected with control (CTR) siRNA or Fbw8 siRNA and then infectedwith adenovirus expressing lacZ (WT) or Cre (LKB1 KO) for 2 days. The level of IRS1 was detected using Western blot. D: LKB1flox/flox MEF cellswere first infected with CA-AMPK or control virus for 24 h and then infected with Cre virus to delete LKB1; Western blot was then performed to

LKB1 AND ADIPOGENESIS

2354 DIABETES, VOL. 62, JULY 2013 diabetes.diabetesjournals.org

Page 9: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

LKB1 regulates adipogenesis through the IRS1/Aktsignaling pathway. We further researched how LKB1altered adipogenesis in cultured cells. We culturedLKB1flox/flox MEFs, and infection with Ad-Cre virus led toLKB1 deletion in these cultured cells. LKB1flox/flox MEFswere first infected with the lacZ or Cre virus and then in-duced to differentiate into adipocytes for 8 days. Oil Red Ostaining showed that the number of adipocytes obtainedfrom LKB1-deficient MEFs was significantly lower thanthat obtained from WT (Fig. 5A). As expected, the TGlevels were also lower upon LKB1 deletion (Fig. 5B).Western blot results indicated that the levels of the adi-pogenesis markers Fabp4 and fatty acid synthetase (FAS),as well as those of C/EBPa and PPARg, were significantlydecreased in LKB1-deficient cells (Fig. 5C), consistent withthe results from LKB-deficient WAT (Fig. 4A).

To confirm the presence of impaired IRS1/Akt signalingin LKB1-deficient cells, total IRS1 was detected by Westernblot. As seen in Fig. 5D, the IRS1 level was significantlydecreased in LKB1-deficient cells. A phospho-specific an-tibody directed at Tyr612 of IRS1 (a putative binding do-main for the p85 subunit of phosphoinositide-3-kinase)was used to examine the insulin-stimulated activation ofIRS1 (26,27). As expected, the level of p-IRS1 (Tyr612) wasalso decreased in LKB1-deficient cells (Fig. 5D). Consistentwith this, the levels of phospho-Akt (Thr308 and Ser473)and p-FoxO1 (Ser256) were also decreased (Fig. 5D).

To investigate if Akt reduction contributed to reducedadipogenesis by LKB1, LKB1-deficient cells were infectedwith adenoviruses encoded with constitutively active (CA)-Akt or GFP. As shown in Fig. 5E, adenoviral overexpressionof CA-Akt but not GFP restored the adipogenesis in LKB1-deficient cells. Consistently, overexpression of CA-Akt butnot GFP normalized the expression of Fabp4, PPARg, andC/EBPa in LKB1-deficient cells (Fig. 5F).

To further confirm impaired insulin/Akt signaling inLKB1-deficient cells, WT or LKB1-deficient cells weretreated with insulin (100 nmol/L) for 15 min, and Aktphosphorylation was detected by Western blots. As shownin Fig. 5G, both phospho-Akt S473 and T308 were de-creased in LKB1-deficient cells, suggesting impaired Aktphosphorylation in LKB1-deficient cells. Since IRS1 wasreported to be phosphorylated by S6K1 with consequentAkt inhibition (28), we further probed for p-S6K1 inLKB12/2 adipocytes. As shown in Fig. 5H, p-S6K1 (T389)was increased in LKB1-deficient WAT whereas the levelsof p-IRS1 (Ser636/639) in WAT from LKB12/2 were re-duced when compared with WT, suggesting that increasedAkt inhibition in LKB1-deficient WAT was not due to in-creased phosphorylation of IRS1 by S6K1. Consistently,the ratios of p-IRS1 (Ser636/639) to total IRS1 were notaltered in LKB1-deficient WAT (data not shown). Since thetotal IRS1 was significantly lower in LKB1-deficient WATthan WT (Fig. 4B), our data suggest that LKB1 deletionresults in decreased IRS1 levels with consequent Akt re-duction, thereby leading to impaired adipogenesis.LKB1 regulates Fbw8 expression in an AMPK-dependent manner. Our results above indicated thatthe expression of Fbw8 is increased in LKB1-deficientWAT. To further confirm that LKB1 deletion leads to Fbw8overexpression, LKB1flox/flox MEFs were infected with

virus encoding the Cre recombinase for different times,and Western blotting was performed to detect the Fbw8level. As shown in Fig. 6A, Cre-mediated LKB1 loss led todecreased p-AMPK, increased Fbw8, and decreased IRS1levels. Fig. 6B depicts the time course of the relationshipsamong LKB1, p-AMPK, Fbw8, and IRS1. LKB1 levels de-creased within 12 h, the phosphorylation of AMPK wassignificantly decreased and the Fbw8 level elevated at 24 h,and the IRS1 level began to decrease at 36 h. These datasuggest that LKB1 loss decreased AMPK phosphorylation.

To establish a causative role for Fbw8-induced IRS1degradation, Fbw8 small interfering RNA (siRNA) wasused to knockdown Fbw8, and then the Cre virus was usedto delete LKB1. LKB1 deletion increased IRS1 degradation(Fig. 6C), and as expected, knockdown of Fbw8 blockedLKB1 deletion-induced IRS1 degradation. Taken together,these data indicate that LKB1 deletion increases IRS1degradation via Fbw8 overexpression.

Because the level of p-AMPKa was significantly de-creased in LKB1-deficient WAT (Fig. 1C), we next de-termined whether LKB1 loss-induced IRS1 degradationwas AMPK-mediated. Overexpression of a CA-AMPK vi-ral construct increased AMPK activity, as evidenced bythe increased phosphorylation of acetyl-CoA carboxylase(ACC), one of the downstream enzymes of AMPK (Fig. 6D).As shown in Fig. 6D, transfection with CA-AMPK ablatedboth Fbw8 expression and the IRS1 reduction caused byLKB1 deletion. These data indicate that LKB1 regulatesFbw8 expression in an AMPK-dependent manner.

To further confirm the role of AMPK in adipogenesis,primary AMPKa1- and -a2–deficient and WT MEF cellswere cultured for 25 days. Both AMPKa1- and -a2–deficient MEFs exhibited dramatically impaired adipocytedifferentiation compared with WT MEFs (Fig. 6E). West-ern blot results showed that the levels of PPARg andFabp4 were decreased in differentiated AMPKa1- and -a2–deficient cells (Fig. 6F). Thus, LKB1 deletion has a pheno-type similar to that of AMPKa loss in adipogenesis, whichfurther confirms the importance of the LKB1–AMPK–Fbw8–IRS1 axis in adipogenesis.LKB1 deletion induces SREBP-1c to regulate Fbw8expression. We next dissected how LKB1/AMPK regu-lates Fbw8 expression. Promoter analysis of Fbw8 wasperformed using the Transcription Factor Database (http://www.gene-regulation.com) internet-based transcriptionfactor binding site program. Three SRE sites were found inthe Fbw8 promoter (Fig. 7A), suggesting that SREBP maybind to the Fbw8 promoter to regulate its expression.

There are three SREBP isoforms, and they have differ-ent distributions and target genes. To determine whichSREBP isoform regulated Fbw8 expression, flag-taggedactive SREBP isoforms were transfected into cells, andWestern blots were performed to detect Fbw8 expression.As shown in Fig. 7B, SREBP1c, but not SREBP1a orSREBP2, induced the overexpression of Fbw8.

AMPK is reported to phosphorylate and inhibit SREBP-1cactivity (29). Next, we investigated whether LKB1 deletioninduced the maturation of SREBP-1c and its translocationinto the nucleus. Cell fractionation was performed toseparate cytoplasmic and nuclear proteins. As shown inFig. 7C, LKB1 deletion not only increased the conversion

detect Fbw8 and IRS1 levels. E: Primary AMPKa1- and -a2–deficient MEFs and WT MEFs were cultured for 25 days, and images were recordedunder a light microscope, original magnification 3100. F: Western analysis for PPARg and Fabp4 in differentiated WT and AMPKa1- and -a2–deficient cells. (A high-quality color representation of this figure is available in the online issue.)

W. ZHANG AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 62, JULY 2013 2355

Page 10: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

FIG. 7. LKB1 deletion induces SREBP-1c to regulate Fbw8 expression. A: The Fbw8 promoter was analyzed using Transcription Factor Databasesoftware, revealing three SRE sites within the Fbw8 promoter. The WT and three mutant Fbw8 promoter luciferase constructs are shown.B: Protein expression from flag-labeled SREBP-1c, SREBP-1a, SREBP-2, or control plasmids transfected into MEFs, and Fbw8 protein levels weredetected by Western blot. C: The cytoplasmic and nuclear fractions were isolated from WT and LKB1-deficient cells, and then SREBP-1 proteinswere detected using Western blot. D: ChIP for different SREBP-1 binding sites in the Fbw8 promoter. The PCR was performed using primersaccording to different SRE sites. E: WT or mutant Fbw8 promoter luciferase constructs were transfected together with SREBP-1c or the controlplasmid into MEFs, and luciferase activity was measured after 24 h. Experiments were performed in triplicate, and results of the luciferasereporter assay are presented as fold changes 6 SEM of the Firefly/Renilla luciferase activities. F: LKB1flox/flox MEF cells were first transfectedwith control (CTR) siRNA or SREBP-1c siRNA and then infected with adenovirus expressing lacZ (WT) or Cre recombinase (LKB1 KO). Western

LKB1 AND ADIPOGENESIS

2356 DIABETES, VOL. 62, JULY 2013 diabetes.diabetesjournals.org

Page 11: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

of the SREBP-1 precursor (125 kDa) into active SREBP-1(65 kDa) but also promoted the translocation of SREBP-1into the nucleus.

To further confirm that SREBP-1c can bind to the SREsites in the Fbw8 promoter, different primers were designedfor a DNA ChIP experiment. Deletion of LKB1 inducedbinding of SREBP-1c to all of the three SRE sites in theFbw8 promoter (Fig. 7D). To further analyze the functionalrole of SREBP-1c binding sites on the activity of the Fbw8promoter, we mutated the core SREBP-1c binding sites inthree SREs and then inserted these into luciferase plasmids.As shown in Fig. 7E, SREBP-1c transfection significantlyincreased luciferase activity from the WT Fbw8 promoter.In contrast, the SRE mutants showed a reduction in SREBP-1c–mediated Fbw8 promoter activation.

Finally, SREBP-1c siRNA was used to knockdownSREBP-1c, and then the Cre virus was used to deleteLKB1. As shown in Fig. 7F, LKB1 deletion resulted in Fbw8overexpression, as observed in an earlier experiment.Moreover, SREBP-1c knockdown ablated LKB1 deletion-induced Fbw8 overexpression (Fig. 7F).

DISCUSSION

In this study, we have demonstrated that the LKB1-IRS1pathway is indispensable for adipogenesis. The impairedadipogenesis seen in mice lacking LKB1 in adipose tissue(LKB1ad2/2 mice) results in reductions in WAT and earlydeath. Thus, our results establish that LKB1 is essential forthe development and metabolism of adipose tissue.

LKB1ad2/2 mice were significantly smaller and moreslender than the control littermates and typically diedwithin 3 weeks after birth. Some studies have reportedviable mouse models with reduced adipose deposition(30,31), indicating that the early lethality seen in LKB1ad2/2

mice may be unrelated to the inability of these mice to formWAT. Blood glucose levels are much lower in LKB1ad2/2

mice than in WT mice. Some reports showed that hypo-glycemia in Sirt6-KO mice or C/EBPa-KO mice caused theirearly death (32,33). Thus, the early lethality in LKB1ad2/2

mice may result from the hypoglycemic phenotype.In this study, we report for the first time, based on ChIP

and luciferase assays, that SREBP-1c binds to the Fbw8promoter and regulates its expression. Thus, LKB1 deletionled to SREBP-1c activation and Fbw8 overexpression,resulting in decreased IRS1 levels and impaired adipo-genesis. Our findings clarify the function of SREBP-1c inadipose tissue and further explain the phenotype of trans-genic mice with adipose tissue–specific expression ofSREBP-1c. SREBP-1s are considered to be profoundly in-volved in the transcriptional regulation of lipogenic enzymes(34), and the predominant SREBP-1 isoform in liver andadipose tissue is SREBP-1c rather than SREBP-1a (35).However, the involvement of SREBP-1c in fatty acid syn-thesis differs fundamentally between the liver and adiposetissue. Several lines of evidence, especially from transgenicand KO mouse models (36,37), have proved that maturehepatic SREBP-1c regulates lipogenic gene expression in theliver. In contrast, targeted disruption of the SREBP-1 genescarcely affected the dynamic changes in lipogenic geneexpression in the adipose tissue (37). Moreover, SREBP-1c

overexpression in adipocytes disrupted the differentiationprocesses of adipocytes, resulting in lipodystrophy intransgenic mice (38). Hence, the role of SREBP-1c in lipo-genic gene regulation in adipocytes is not well-understood.

Another important finding in this study is that LKB1regulates SREBP-1c activity through AMPK. This obser-vation is consistent with a previous report on the re-lationship between AMPK and SREBP-1c (29). However,the role of AMPK in adipogenesis is still unclear and con-troversial. Some studies have reported that treatment of3T3-L1 cells with AMPK agonists leads to inhibition of clonalexpansion (39,40); therefore, AMPK is thought to be anti-adipogenic. In our study, AMPK activity was decreased inLKB1-deficient WAT, and this was accompanied by impairedadipogenesis. This suggests that AMPK is adipogenic. Be-sides, AMPKa-deficient MEFs could not differentiate intoadipocytes. However, whole-body AMPKa1 or -a2 KO micedo not exhibit the lipodystrophic or obese phenotype. It istherefore possible that AMPKa1 and -a2 have compensatoryeffects in mice bearing KOs of the other gene. For example,AMPKa2 activity in BAT, WAT, and liver was significantlygreater in AMPKa12/2 than in WT mice (41). Besides,AMPK was reported to be required for brown adipocytedifferentiation (42). Thus, it is necessary to generate adi-pose tissue–specific AMPKa1 and -a2 double-KO mice tofurther confirm the role of AMPK in adipogenesis.

LKB1ad2/2 mice exhibit severely decreased WAT mass,which is linked to decreased adipocyte size. This model isuseful for studying the differentiation process of WAT.However, the role of LKB1 in BAT is not clear. BecauseFabp4 was found mainly in WAT, Fabp4-mediated LKB1deletion mainly occurred in WAT (Figs. 1A and B and 4A).In addition, we found that LKB1 is normally expressed inBAT and that LKB1ad2/2 mice have normal BAT (Figs. 1Aand B and 4A). Thus, our study has not addressed theimportance of LKB1 in BAT, and further studies on the roleof LKB1 in BAT are warranted. As expected based on thereduced fat storage, LKB1ad2/2 mice also show reducedlevels of circulating adiponectin, leptin, TGs, and IGF-I.IGF-1 plays an important role in skeletal development andnormal postnatal growth (43). IGF-1 deficiency in miceresults in marked skeletal dysplasia and growth re-tardation (44). Thus, the lower level of serum IGF-1 inLKB1ad2/2 mice may contribute to their small skeletonsize and growth retardation.

In summary, we report the identification of LKB1 as anessential player in adipocyte differentiation. Our workprovides a new mouse model that exhibits impaired adi-pogenesis, which will prove very useful in studies of themechanism of adipogenesis. As preadipocytes from elderlyhumans retain the capacity to differentiate in vitro (45), it ispossible that adipogenesis occurs throughout life at a lowrate until energy storage demands promote further differ-entiation. Our study opens up new avenues of research intohuman lipid metabolism. Our future studies will investigatethe relationship between LKB1 and obesity.

ACKNOWLEDGMENTS

This study was supported by National Institutes of Healthgrants HL-079584, HL-074399, HL-080499, HL-089920,

blot was performed to detect Fbw8 levels. *P < 0.05 compared with the WT construct plus control plasmid; #P < 0.05 compared with the WTconstruct plus SREBP-1c plasmid. IgG, ChIP with antibody against immunoglobulin G control; Input, positive control using sonicated chromatin asPCR template; NC, negative control without template; SREBP1, ChIP with antibody against SREBP-1. (A high-quality color representation of thisfigure is available in the online issue.)

W. ZHANG AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 62, JULY 2013 2357

Page 12: ORIGINAL ARTICLE Liver Kinase B1 Is Required for White ... · Liver Kinase B1 Is Required for White Adipose Tissue Growth and Differentiation Wencheng Zhang,1 Qilong Wang,1 Ping Song,1

HL-096032, HL-110488, HL-105157, and HL-110488 andby funds from the Warren Chair in Diabetes Research ofthe University of Oklahoma Health Science Center (all toM.-H.Z.). Part of this work was also supported by anAmerican Heart Association Scientist Development Grant(11SDG5560036 to P.S.). M.-H.Z. is a recipient of theNational Established Investigator Award from the Ameri-can Heart Association.

No potential conflicts of interest relevant to this articlewere reported.

W.Z. designed and performed the experiments, analyzeddata, and prepared the manuscript. Q.W. and P.S.researched the data. M.-H.Z. conceived the projects,designed the experiments, reviewed the data, and wrotethe manuscript. M.-H.Z. is the guarantor of this work and,as such, had full access to all the data in the study andtakes responsibility for the integrity of the data and theaccuracy of the data analysis.

REFERENCES

1. Koutnikova H, Cock TA, Watanabe M, et al. Compensation by the musclelimits the metabolic consequences of lipodystrophy in PPAR gammahypomorphic mice. Proc Natl Acad Sci USA 2003;100:14457–14462

2. Ahima RS, Flier JS. Adipose tissue as an endocrine organ. Trends Endo-crinol Metab 2000;11:327–332

3. Farmer SR. Transcriptional control of adipocyte formation. Cell Metab2006;4:263–273

4. Linhart HG, Ishimura-Oka K, DeMayo F, et al. C/EBPalpha is required fordifferentiation of white, but not brown, adipose tissue. Proc Natl Acad SciUSA 2001;98:12532–12537

5. Rosen ED, MacDougald OA. Adipocyte differentiation from the inside out.Nat Rev Mol Cell Biol 2006;7:885–896

6. Tseng YH, Kriauciunas KM, Kokkotou E, Kahn CR. Differential roles ofinsulin receptor substrates in brown adipocyte differentiation. Mol CellBiol 2004;24:1918–1929

7. Laustsen PG, Michael MD, Crute BE, et al. Lipoatrophic diabetes in Irs1(-/-)/Irs3(-/-) double knockout mice. Genes Dev 2002;16:3213–3222

8. Murata Y, Tsuruzoe K, Kawashima J, et al. IRS-1 transgenic mice showincreased epididymal fat mass and insulin resistance. Biochem BiophysRes Commun 2007;364:301–307

9. DeMambro VE, Kawai M, Clemens TL, et al. A novel spontaneous mutationof Irs1 in mice results in hyperinsulinemia, reduced growth, low bone massand impaired adipogenesis. J Endocrinol 2010;204:241–253

10. Peng XD, Xu PZ, Chen ML, et al. Dwarfism, impaired skin development,skeletal muscle atrophy, delayed bone development, and impeded adipo-genesis in mice lacking Akt1 and Akt2. Genes Dev 2003;17:1352–1365

11. Lee AV, Gooch JL, Oesterreich S, Guler RL, Yee D. Insulin-like growthfactor I-induced degradation of insulin receptor substrate 1 is mediated bythe 26S proteasome and blocked by phosphatidylinositol 39-kinase in-hibition. Mol Cell Biol 2000;20:1489–1496

12. Pederson TM, Kramer DL, Rondinone CM. Serine/threonine phosphoryla-tion of IRS-1 triggers its degradation: possible regulation by tyrosinephosphorylation. Diabetes 2001;50:24–31

13. Sun XJ, Goldberg JL, Qiao LY, Mitchell JJ. Insulin-induced insulin receptorsubstrate-1 degradation is mediated by the proteasome degradation path-way. Diabetes 1999;48:1359–1364

14. Xu X, Sarikas A, Dias-Santagata DC, et al. The CUL7 E3 ubiquitin ligasetargets insulin receptor substrate 1 for ubiquitin-dependent degradation.Mol Cell 2008;30:403–414

15. Giardiello FM, Trimbath JD. Peutz-Jeghers syndrome and managementrecommendations. Clin Gastroenterol Hepatol 2006;4:408–415

16. Katajisto P, Vallenius T, Vaahtomeri K, et al. The LKB1 tumor suppressorkinase in human disease. Biochim Biophys Acta 2007;1775:63–75

17. Alessi DR, Sakamoto K, Bayascas JR. LKB1-dependent signaling pathways.Annu Rev Biochem 2006;75:137–163

18. Shackelford DB, Shaw RJ. The LKB1-AMPK pathway: metabolism andgrowth control in tumour suppression. Nat Rev Cancer 2009;9:563–575

19. Sakamoto K, McCarthy A, Smith D, et al. Deficiency of LKB1 in skeletalmuscle prevents AMPK activation and glucose uptake during contraction.EMBO J 2005;24:1810–1820

20. Fu A, Ng AC, Depatie C, et al. Loss of Lkb1 in adult beta cells increasesbeta cell mass and enhances glucose tolerance in mice. Cell Metab 2009;10:285–295

21. Ylikorkala A, Rossi DJ, Korsisaari N, et al. Vascular abnormalities andderegulation of VEGF in Lkb1-deficient mice. Science 2001;293:1323–1326

22. Bardeesy N, Sinha M, Hezel AF, et al. Loss of the Lkb1 tumour suppressorprovokes intestinal polyposis but resistance to transformation. Nature2002;419:162–167

23. Wang P, Mariman E, Renes J, Keijer J. The secretory function of adipocytesin the physiology of white adipose tissue. J Cell Physiol 2008;216:3–13

24. Kovacs WJ, Shackelford JE, Tape KN, et al. Disturbed cholesterol ho-meostasis in a peroxisome-deficient PEX2 knockout mouse model. MolCell Biol 2004;24:1–13

25. Kovanen PT, Nikkilä EA, Miettinen TA. Regulation of cholesterol synthesisand storage in fat cells. J Lipid Res 1975;16:211–223

26. Esposito DL, Li Y, Cama A, Quon MJ. Tyr(612) and Tyr(632) in humaninsulin receptor substrate-1 are important for full activation of insulin-stimulated phosphatidylinositol 3-kinase activity and translocation ofGLUT4 in adipose cells. Endocrinology 2001;142:2833–2840

27. Sun XJ, Crimmins DL, Myers MG Jr, Miralpeix M, White MF. Pleiotropicinsulin signals are engaged by multisite phosphorylation of IRS-1. Mol CellBiol 1993;13:7418–7428

28. Tzatsos A, Kandror KV. Nutrients suppress phosphatidylinositol 3-kinase/Akt signaling via raptor-dependent mTOR-mediated insulin receptor sub-strate 1 phosphorylation. Mol Cell Biol 2006;26:63–76

29. Li Y, Xu S, Mihaylova MM, et al. AMPK phosphorylates and inhibits SREBPactivity to attenuate hepatic steatosis and atherosclerosis in diet-inducedinsulin-resistant mice. Cell Metab 2011;13:376–388

30. Asterholm IW, Halberg N, Scherer PE. Mouse Models of LipodystrophyKey reagents for the understanding of the metabolic syndrome. DrugDiscov Today Dis Models 2007;4:17–24

31. Luo J, Sladek R, Carrier J, Bader JA, Richard D, Giguère V. Reduced fatmass in mice lacking orphan nuclear receptor estrogen-related receptoralpha. Mol Cell Biol 2003;23:7947–7956

32. Wang ND, Finegold MJ, Bradley A, et al. Impaired energy homeostasis inC/EBP alpha knockout mice. Science 1995;269:1108–1112

33. Xiao C, Kim HS, Lahusen T, et al. SIRT6 deficiency results in severe hy-poglycemia by enhancing both basal and insulin-stimulated glucose uptakein mice. J Biol Chem 2010;285:36776–36784

34. Yokoyama C, Wang X, Briggs MR, et al. SREBP-1, a basic-helix-loop-helix-leucine zipper protein that controls transcription of the low densitylipoprotein receptor gene. Cell 1993;75:187–197

35. Shimomura I, Shimano H, Horton JD, Goldstein JL, Brown MS. Differentialexpression of exons 1a and 1c in mRNAs for sterol regulatory elementbinding protein-1 in human and mouse organs and cultured cells. J ClinInvest 1997;99:838–845

36. Shimano H, Horton JD, Shimomura I, Hammer RE, Brown MS, GoldsteinJL. Isoform 1c of sterol regulatory element binding protein is less activethan isoform 1a in livers of transgenic mice and in cultured cells. J ClinInvest 1997;99:846–854

37. Shimano H, Yahagi N, Amemiya-Kudo M, et al. Sterol regulatory element-binding protein-1 as a key transcription factor for nutritional induction oflipogenic enzyme genes. J Biol Chem 1999;274:35832–35839

38. Shimomura I, Hammer RE, Richardson JA, et al. Insulin resistance anddiabetes mellitus in transgenic mice expressing nuclear SREBP-1c in adi-pose tissue: model for congenital generalized lipodystrophy. Genes Dev1998;12:3182–3194

39. Habinowski SA, Witters LA. The effects of AICAR on adipocyte differen-tiation of 3T3-L1 cells. Biochem Biophys Res Commun 2001;286:852–856

40. Lee H, Kang R, Bae S, Yoon Y. AICAR, an activator of AMPK, inhibitsadipogenesis via the WNT/b-catenin pathway in 3T3-L1 adipocytes. Int JMol Med 2011;28:65–71

41. Bauwens JD, Schmuck EG, Lindholm CR, et al. Cold tolerance, cold-inducedhyperphagia, and nonshivering thermogenesis are normal in a₁-AMPK-/-mice. Am J Physiol Regul Integr Comp Physiol 2011;301:R473–R483

42. Vila-Bedmar R, Lorenzo M, Fernández-Veledo S. Adenosine 59-monophosphate-activated protein kinase-mammalian target of rapamycin cross talk regu-lates brown adipocyte differentiation. Endocrinology 2010;151:980–992

43. Baker J, Liu JP, Robertson EJ, Efstratiadis A. Role of insulin-like growthfactors in embryonic and postnatal growth. Cell 1993;75:73–82

44. Wang Y, Nishida S, Sakata T, et al. Insulin-like growth factor-I is essential forembryonic bone development. Endocrinology 2006;147:4753–4761

45. Hauner H, Entenmann G, Wabitsch M, et al. Promoting effect of gluco-corticoids on the differentiation of human adipocyte precursor cells cul-tured in a chemically defined medium. J Clin Invest 1989;84:1663–1670

LKB1 AND ADIPOGENESIS

2358 DIABETES, VOL. 62, JULY 2013 diabetes.diabetesjournals.org