Gremlin 2 inhibits adipocyte differentiation through activation of Wnt/β-catenin signaling

  • Authors:
    • Qing Wu
    • Shi-Guo Tang
    • Zhong‑Ming Yuan
  • View Affiliations

  • Published online on: July 27, 2015     https://doi.org/10.3892/mmr.2015.4117
  • Pages: 5891-5896
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The primary function of white adipose tissues is to store excess energy. The current study aimed to investigate the roles of Gremlin 2 (Grem2), a glycoprotein in adipogenesis. Using polymerase chain reaction‑based microarrays, it was determined that Grem2 was markedly downregulated in adipose tissues from obese animals and humans. In addition, 3T3‑L1 cells were used to investigate the details of the mechanisms underlying the anti‑adipogenic effects of Grem2. Grem2 expression was markedly decreased upon the induction of adipocyte differentiation, as demonstrated by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. Notably, Grem2 overexpression inhibited adipogenesis, while knockdown of Grem2 led to an increase in adipogenesis. At the molecular level, Grem2 promotes nuclear translocation of β‑catenin, an integral Wnt signaling component. Consistently, inhibition of Wnt/β‑catenin signaling using a retrovirus targeting the β‑catenin coding region attenuated the anti‑adipogenic effects of Grem2. Therefore, to the best of our knowledge, the current study shows for the first time that Grem2 may be an important regulator of adipocyte differentiation.

Introduction

When dietary nutrient intake chronically exceeds energy expenditure, obesity occurs due to an increase in the size and/or number of white adipocytes (1). Obesity is closely associated with type 2 diabetes, fatty liver, hyperlipidemia, cardiovascular diseases and tumorigenesis (2,3). Thus, investigating the molecular and cellular mechanisms regulating adipocyte differentiation is crucial for designing potential therapies to prevent obesity and the associated diseases.

Decades of studies have identified a number of transcription factors involved in adipocyte differentiation, including peroxisome proliferator-activated receptor γ (PPARγ) and members of the C/EBP family (4,5). Additionally, a number of other factors or signaling molecules are involved in the control of adipocyte differentiation, such as Wnt/β-catenin, cAMP/CREB, and the family of kruppel-like factor (KLF) proteins (68). Furthermore, recent studies have indicated that modification of these proteins has an important role in adipogenesis. It has been shown that phosphorylation, acetylation and small ubiquitin-like modifier (SUMO)ylation of PPARγ affects adipocyte differentiation, though regulation of its transcriptional activity (911). In this regard, the process of adipogenesis is rather complex, suggesting that identification of the additional factors may provide novel insights into the mechanisms regulating the initiation of fat cell differentiation and obesity.

In the present study, polymerase chain reaction (PCR)-based microarrays were performed in obese and lean mice to screen potential genes involved in adipogenesis.

Materials and methods

Mice

Male C57BL/6, ob/ob and db/db mice, aged 10–12 weeks, were purchased from the Shanghai Laboratory Animal Company (Shanghai, China). All mice were housed and bred according to international standard conditions, with a 12-h dark/light cycle. For induction of obesity, mice aged 10 weeks were placed on a high-fat-diet (50 kcal% fat, 25 kcal% protein and 25 kcal% carbohydrate) for 12 weeks. The normal diet consisted of 10 kcal% fat, 15 kcal% protein and 75 kcal% carbohydrate. The animal protocol was reviewed and approved by the Animal Care Committee of Chongqing Ninth People's Hospital (Chongqing, China).

Human tissue samples

Human subcutaneous fat tissues from obese (BMI>25; n=25) and lean (BMI<23; n=20) adults were collected at the Department of Endocrinology at Chongqing Ninth People's Hospital. All subjects had a stable weight, defined as the absence of fluctuations of >1% of body weight for at least 2 months prior to surgery. Adipose tissue was immediately frozen in liquid nitrogen following extraction. All samples were obtained with informed consent and the study was approved by the Chongqing Ninth People's Hospital Institutional Review Board.

Cell culture

3T3-L1 fibroblasts were obtained from The Cell Bank of Type Culture Collection of Chinese Academy of Sciences (Shanghai, China), and cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco-BRL, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco-BRL), 100 IU/ml penicillin and 100 µg/ml streptomycin (Gibco-BRL). Adipocyte differentiation was induced by treating the cells at 80–90% confluency with standard differentiation inducers [100 µM IBMX (Beyotime Institute of Biotechnology, Nantong, China), 100 nM dexamethasone (Beyotime Institute of Biotechnology) and 10 µg/ml insulin (Eli Lilly & Co., Indianapolis, IN, USA)] for 48 h (from day 0 to day 2). Cells were treated with DMEM supplemented with 10 µg/ml insulin and 10% FBS for another 48 h (from day 2 to day 4). Subsequently, the medium was replaced by DMEM and 10% FBS, which was changed daily, and the cells were cultured for a further 4–6 days.

RNA extraction, cDNA microarrays, and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from tissues or cells using the TRIzol reagent (Invitrogen Life Technologies, Shanghai, China), and reverse transcription was performed using the RT kit from Promega Corporation (Madison, WI, USA), according to the manufacturer's instructions. Affymetrix arrays were performed using Mouse Genome 2.0 chips from Gene Tech Company Limited (Shanghai, China). In order to quantify the transcripts of the interest genes, qPCR was performed using a SYBR Green Premix Ex Taq kit (Takara Bio Inc., Dalian, China) on a Light Cycler 480 (Roche, Basel, Switzerland) with 5 mg RNA. The following primer sequences form Bioyare Biotechnology Company (Shanghai, China) were used: Mouse Grem2, forward 5′-TGTGCTGGTAAAGGTAGCTGA-3′ and reverse 5′-CCACCTCTCTGAGTTGTTGCT-3′; human Grem2, forward 5′-ATCCCCTCGCCTTACAAGGA-3′ and reverse 5′-TCTTGCACCAGTCACTCTTGA-3′; C/EBPa, forward 5′-CAAGAACAGCAACGAGTACCG-3′ and reverse 5′-GTCACTGGTCAACTCCAGCAC-3′; PPARg, forward 5′-CTCCAAGAATACCAAAGTGCGA-3′ and reverse 5′-GCCTGATGCTTTATCCCCACA-3′; LPL, forward 5′-ATGGATGGACGGTAACGGGAA-3′ and reverse 5′-CCCGATACAACCAGTCTACTACA-3′; PEPCK, forward 5′-ATGAGCCGCAAACTGGGTC-3′ and reverse 5′-AGAGCCGAACTCCACAATCTC-3′; CD36, forward 5′-AGCATTCAACGCCAGGTTC-3′ and reverse 5′-CGAGTCTGTCAGTTCAATACCAA-3′; aP2, forward 5′-AAGGTGAAGAGCATCATAACCCT-3′ and reverse 5′-TCACGCCTTTCATAACACATTCC-3′; mouse HPRT1, forward 5′-TCAGTCAACGGGGGACATAAA-3′ and reverse 5′-GGGGCTGTACTGCTTAACCAG-3′; human HPRT1, forward 5′-CCTGGCGTCGTGATTAGTGAT-3′ and reverse 5′-AGACGTTCAGTCCTGTCCATAA-3′. The PCR cycling conditions were as follows: Initial period at 94°C for 3 min, followed by a two-step PCR program consisting of 95°C for 5 sec and 60°C for 30 sec for 55 cycles. Gene expression was calculated relative to the mRNA expression of HPRT1, and relative quantitation analysis of gene expression data was performed using the 2−ΔΔCt method.

Retrovirus, small interfering (si)RNA and transfection

Retroviruses containing empty vector or Grem2 cDNA were constructed by Invitrogen Life Technologies. siRNA oligos targeting Grem2 or β-catenin were purchased from Gene Pharma (Shanghai, China) and transfected into 3T3-L1 cells using Lipofectamine 2000 (Invitrogen Life Technologies), according to the manufacturer's instructions. Histological analysis was conducted using Oil Red O staining. Cells were fixed with 4% paraformaldehyde for 30 min, followed by incubation with Oil Red O (Sigma-Aldrich, St. Louis, MO, USA) for 45 min.

Western blot analysis

Cells or tissues were harvested and lysed with lysis buffer (150 mM NaCl, 50 mM Tris-HCl and 1% NP-40; pH 7.5). Proteins were quantified and separated by 10% SDS-PAGE and transferred to polyvinylidine difluoride membranes (Amersham Bioscience, Buckinghamshire, UK). After blocking with 10% non-fat milk in phosphate-buffered saline, membranes were immunoblotted with the following rabbit antibodies from Abcam (Cambridge, MA, USA): Polyclonal anti-Grem2 (ab102563; 1:1,000), monoclonal C/EBPα (ab40764; 1:1,000), polyclonal PPARγ (ab19481; 1:1,000), monoclonal P-P38 (ab178867; 1:2,000), polyclonal P-AKT (ab66138; 1:1,000), monoclonal P-CREB (ab32096; 1:2,000), monoclonal CREB (ab32515; 1:2,000), polyclonal β-catenin (ab6302; 1:1,000), polyclonal lamin B (ab16048; 1:2,000) and monoclonal GAPDH (ab181602; 1:1,000). The blots were then incubated with horseradish peroxidase-linked secondary antibodies (Cell Signaling Technology, Inc. Danvers, MA, USA). The signals were detected using the SuperSignal West Pico Chemiluminescent Substrate kit (Pierce, Rockford, IL, USA) according to manufacturer's instructions.

Statistical analysis

The results are expressed as the mean ± standard error of the mean and analyzed using Student's t-test or one way analysis of variance with GraphPad Prism software, version 5.0.1 (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Downregulation of Grem2 in obese mice and humans

To identify potential genes involved in adipogenesis and obesity, male C57BL/6 mice aged 10 weeks were placed on high-fat or normal diets for 12 weeks. The mice were sacrificed by cervical dislocation and gene expression arrays were performed using epididymal adipose tissues. The clustering analysis of the Affymetrix arrays revealed that 1,174 genes were upregulated and 1,572 genes were downregulated in the adipose tissues of mice fed a high-fat-diet compared with those fed a normal diet (P<0.05; data not shown). Among these genes, it was determined that Grem2 was significantly downregulated (P<0.001). Its downregulation was further confirmed by RT-qPCR and western blot analysis (Fig. 1A and B).

To determine whether the inhibition of Grem2 represents a common feature of obesity, db/db and ob/ob mice were employed as models for monogenic-induced obesity. Indeed, Grem2 mRNA and protein levels were found to be reduced in the epididymal adipose tissues of these mice, as compared with lean controls (Fig. 1C–F). Notably, Grem2 mRNA and protein levels were significantly reduced in subcutaneous fat from obese humans (Fig. 1G and H), further suggesting that the downregulation of Grem2 expression may be a conserved feature in obese mice and humans.

Grem2 suppresses the differentiation of 3T3-L1 preadipocytes into adipocytes

To explore the functions of Grem2 in adipogenesis, its gene expression was investigated at numerous time points during 3T3-L1 differentiation. The RT-qPCR results revealed that Grem2 mRNA gradually reduced from day 0 post-induction (Fig. 2A). The protein levels for Grem2 were consistent with its mRNA levels (Fig. 2B).

The role of Grem2 in adipogenesis was tested using retroviruses carrying Grem2 cDNA or empty vector, which resulted in forced overexpression of Grem2 in 3T3-L1 preadipocytes (Fig. 3A). These cells were cultured to confluence and differentiated using the standard induction cocktail for 8 days. Cell morphology indicated that cells overexpressing Grem2 accumulated significantly less lipids than the control cells (Fig. 3B). In addition, the expression levels of adipogenic regulators such as C/EBPα and PPARγ were reduced (Fig. 3C and D), which was consistent with the Oil Red O staining. Furthermore, markers of mature adipocytes LPL, PEPCK, CD36 and aP2, which are downstream targets of C/EBPα and PPARγ, were repressed by Grem2 overexpression (Fig. 3E).

Grem2 knockdown promotes adipocyte differentiation

To further examine the roles of Grem2, its specific siRNA was transfected into 3T3-L1 cells (Fig. 4A). As a result, knockdown of Grem2 levels showed a clear increase in the adipogenic potential of the 3T3-L1 cells compared with that of the cells treated with the negative control siRNA, as illustrated by cell morphology (Fig. 4B). Furthermore, knockdown of Grem2 increased expression levels of the adipocyte markers including C/EBPα, PPARγ, LPL, PEPCK, CD36 and aP2 (Fig. 4C and D). Together, these results indicate that Grem2 is a critical negative regulator in the adipogenesis.

Grem2 activates Wnt/β-catenin signaling in adipocytes

Finally, the molecular basis for the observed phenotypic changes in 3T3-L1 cells with Grem2 overexpression was investigated. As shown in Fig. 5A, Wnt signaling was highly activated by Grem2 overexpression, as evidenced by nuclear accumulation of β-catenin, while other signaling molecules, including p38, AKT and cAMP/CREB, remained unaffected (Fig. 5B). Subsequently, it was investigated whether ablation of β-catenin reversed the roles of Grem2. A retrovirus targeting a β-catenin coding region (sh-catenin) was generated using pAD_BLOCK_IT_DEST vectors (Invitrogen Life Technologies, Grand Island, NY, USA) to knockdown endogenous β-catenin expression in 3T3-L1 cells (Fig. 5C). As a result, sh-catenin largely attenuated the inhibitory roles of Grem2 on the adipogenesis, as evidenced by gene expression of adipogenic markers (Fig. 5D), suggesting that the role of Grem2 in the adipogenesis, at least in part, relies on Wnt/β-catenin activation.

Discussion

In the present study, it was determined that the Grem2 gene was significantly downregulated in white adipose tissue from obese mice and humans. Using 3T3-L1 cells cultured in vitro, it was demonstrated that Grem2 inhibited adipocyte differentiation. Thus, for the first time to the best of our knowledge, these results identify Grem2 as a key regulator in adipose formation and obesity.

Members of the Gremlin family, including Grem2, have been shown to be bone morphogenetic protein (BMP) antagonists (12). Indeed, Grem2 regulates BMP signaling in cardiomyocyte and osteoblast differentiation (13,14). Furthermore, a recent human study revealed that genetic variants in Grem2 are associated with bone mineral density (15). Since certain members of the family of BMPs support white and brown adipocyte differentiation (16,17), whether Grem2 regulates BMP-induced adipogenesis requires further investigation.

At the molecular level, it was revealed that Grem2 activates Wnt/β-catenin signaling, as evidenced by its nuclear accumulation. Initial studies have demonstrated that ectopic expression of Wnt1, an activator of Wnt/β-catenin signaling, may inhibit adipogenesis (6,18,19). Concurrently, pharmacological compounds that activate Wnt/β-catenin signaling or transfection of stable β-catenin block preadipocyte differentiation (18). However, inhibition of Wnt signaling promotes cell differentiation (18,20), indicating that Wnt signaling has a negative role in the adipocyte differentiation. In accordance with this conclusion in vitro, transgenic mice overexpressing Wnt10b, an endogenous factor for Wnt signaling, have a 50% reduction in adiposity under a normal chow diet (21). These mice resist expansion of adipose tissue under conditions of high-fat-diet-induced and genetic-induced obesity (21). Furthermore, recent genetic research indicated the importance of Wnt/β-catenin signaling in human obesity. Polymorphisms of the Wnt10b gene may be associated with obesity in human subjects of European origin (22). Together, these data suggest that modulators of Wnt/β-catenin pathway may represent a useful therapeutic target for obesity (23).

In conclusion, the results of the current study highlight a pivotal role for Grem2 in white fat-cell differentiation by controlling Wnt/β-catenin signaling. Further studies are required to investigate the functions and mechanisms of Grem2 in vivo, using knockout or transgenic animals.

References

1 

Rueda-Clausen CF, Padwal RS and Sharma AM: New pharmacological approaches for obesity management. Nat Rev Endocrinol. 9:467–478. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Fenske W, Athanasiou T, Harling L, Drechsler C, Darzi A and Ashafian H: Obesity-related cardiorenal disease: the benefits of bariatric surgery. Nat Rev Nephol. 9:539–551. 2013. View Article : Google Scholar

3 

Tao W and Lagergren J: Clinical management of obese patients with cancer. Nat Rev Clin Oncol. 10:519–533. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Rosen ED and MacDougald OA: Adipocyte differentiation from the inside out. Nat Rev Mol Cell Biol. 7:885–896. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Kawai M and Rosen CJ: PPARy: A circadian transcription factor in adipogenesis and osteogenesis. Nat Rev Endocrinol. 6:629–636. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Cristancho AG and Lazar MA: Forming functional fat: a growing understanding of adipocyte differentiation. Nat Rev Mol Cell Biol. 12:722–734. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Oishi Y, Manabe I, Tobe K, et al: Kruppel-like transcription factor klf5 is a key regulator of adipocyte differentiation. Cell Metab. 1:27–39. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Birsoy K, Chen Z and Friedman J: Transcriptional regulation of adipogenesis by klf4. Cell Metab. 7:339–347. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Choi JH, Banks AS, Estall JL, et al: Anti-diabetic drugs inhibit obesity-linked phosphorylation of PPARgamma by cdk5. Nature. 466:451–456. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Qiang L, Wang L, Kon N, et al: Brown remodeling of white adipose tissue by sirt1-dependent deacetylation of ppargamma. Cell. 150:620–632. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Dutchak PA, Katafuchi T, Bookout AL, et al: Fibroblast growth factor-21 regulates PPARgamma activity and the antidiabetic actions of thiazolidinediones. Cell. 148:556–567. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Zuniga E, Rippen M, Alexander C, Schilling TF and Crump JG: Gremlin 2 regulates distinct roles of bmp and endothelin 1 signaling in dorsoventral patterning of the facial skeleton. Development. 138:5147–5156. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Suzuki D, Yamada A, Aizawa R, et al: bmp2 differentially regulates the expression of gremlin1 and gremlin2, the negative regulators of bmp function, during osteoblast differentiation. Calcif Tissue Int. 91:88–96. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Muller II, Melville DB, Tanwar V, et al: Functional modeling in zebrafish demonstrates that the atrial-fibrillation-associated gene GREM2 regulates cardiac laterality, cardiomyocyte differentiation and atrial rhythm. Dis Models Mech. 6:332–341. 2013. View Article : Google Scholar

15 

Paternoster L, Lorentzon M, Lehtimaki T, et al: Genetic determinants of trabecular and cortical volumetric bone mineral densities and bone microstructure. PLoS Genet. 9:e10032472013. View Article : Google Scholar : PubMed/NCBI

16 

Macotela Y, Emanuelli B, Mori MA, et al: Intrinsic differences in adipocyte precursor cells from different white fat depots. Diabetes. 61:1691–1699. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Takahashi A, Morita M, Yokoyama K, Suzuki T and Yamamoto T: Tob2 inhibits peroxisome proliferator-activated receptor gamma2 expression by sequestering smads and c/ebpalpha during adipocyte differentiation. Mol Cell Biol. 32:5067–5077. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Ross SE, Hemati N, Longo KA, et al: Inhibition of adipogenesis by wnt signaling. Science. 289:950–953. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Laudes M: Role of WNT signalling in the determination of human mesenchymal stem cells into preadipocytes. J Mol Endocrinol. 46:R65–R72. 2011.PubMed/NCBI

20 

Prestwich TC and Macdougald OA: Wnt/beta-catenin signaling in adipogenesis and metabolism. Curr Opin Cell Biol. 19:612–617. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Wright WS, Longo KA, Dolinsky VW, et al: WNT10b inhibits obesity in ob/ob and agouti mice. Diabetes. 56:295–303. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Chistodoulides C, Scarda A, Granzotto M, et al: WNT10B mutations in human obesity. Diabetologia. 49:678–684. 2006. View Article : Google Scholar

23 

Takada I, Kouzmenko AP and Kato S: Wnt and PPARgamma signaling in osteoblastogenesis and adipogenesis. Nat Rev Rheumatol. 5:442–447. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2015
Volume 12 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu Q, Tang S and Yuan ZM: Gremlin 2 inhibits adipocyte differentiation through activation of Wnt/β-catenin signaling. Mol Med Rep 12: 5891-5896, 2015
APA
Wu, Q., Tang, S., & Yuan, Z. (2015). Gremlin 2 inhibits adipocyte differentiation through activation of Wnt/β-catenin signaling. Molecular Medicine Reports, 12, 5891-5896. https://doi.org/10.3892/mmr.2015.4117
MLA
Wu, Q., Tang, S., Yuan, Z."Gremlin 2 inhibits adipocyte differentiation through activation of Wnt/β-catenin signaling". Molecular Medicine Reports 12.4 (2015): 5891-5896.
Chicago
Wu, Q., Tang, S., Yuan, Z."Gremlin 2 inhibits adipocyte differentiation through activation of Wnt/β-catenin signaling". Molecular Medicine Reports 12, no. 4 (2015): 5891-5896. https://doi.org/10.3892/mmr.2015.4117