Skip to main content
Adipocyte logoLink to Adipocyte
. 2012 Jul 1;1(3):168–172. doi: 10.4161/adip.20574

New insights into adipocyte-specific leptin gene expression

Christiane D Wrann 1,2, Evan D Rosen 1,3,*
PMCID: PMC3609097  PMID: 23700528

Abstract

The adipocyte-derived hormone leptin is a critical regulator of many physiological functions, ranging from satiety to immunity. Surprisingly, very little is known about the transcriptional pathways that regulate adipocyte-specific expression of leptin. In a recent published study, we pursued a strategy integrating BAC transgenic reporter mice, in vitro reporter assays, and chromatin state mapping to locate an adipocyte-specific cis-element upstream of the LEP gene in human fat cells. Quantitative proteomics (stable isotope labeling by amino acids in cell culture, SILAC) with affinity enrichment of protein-DNA complexes identified the transcription factor FOSL2 as a specific binder to the identified region. We confirmed that FOSL2 is an important regulator of LEP gene expression in vitro and in vivo using cell culture models and genetic mouse models. In this commentary, we discuss the transcriptional regulation of LEP gene expression, our strategy to identify an adipocyte-specific cis-regulatory element and the transcription factor(s) responsible for LEP gene expression. We also discuss our data on FOSL2 and leptin levels in physiology and pathophysiology. We speculate on unanswered questions and future directions.

Keywords: ChIP-Seq, Fosl2, SILAC, adipose, diabetes, leptin, obesity, transcription

Introduction

Leptin is an adipocyte-derived hormone that regulates food intake and energy expenditure, acting primarily through the central nervous system.1-3 In addition to its well-established role in metabolism, leptin also affects physiological processes as wide-ranging as reproduction, thyroid function, bone density, and immune biology.4,5 Recombinant leptin has been developed as a drug for treating insulin resistance in patients with lipodystrophy.6 Although early studies using leptin as a weight loss agent in human obesity proved disappointing,7 recent trials using leptin in combination with the pancreatic polypeptide amylin show some promise.8 There has also been interest in using leptin to improve insulin action in a weight-loss independent fashion; however, recombinant methionyl human (r-Met hu) leptin did not enhance insulin sensitivity in obese subjects with type 2 diabetes.9

In the two decades since leptin was first identified, over 20,000 papers have been written describing leptin’s actions in physiology and pathophysiology. In stark contrast to the wealth of data regarding the functions of leptin, however, our knowledge about the mechanisms involved in the transcriptional regulation of leptin gene expression, particularly the basis for its adipocyte-specific expression, is rather limited. Leptin is regulated primarily at the transcriptional level, and a wide variety of physiological conditions and pharmacological agents have been shown to affect its expression, including fasting and feeding, insulin, glucocorticoids, thiazolidinediones and even leptin itself.4,10,11 These studies are summarized in Table 1.

Table 1. Summary of factors regulating leptin gene expression in adipocytes.

Physiological conditions References Direction of effect
Fasting
11, 26, 35

Feeding
11, 26, 35

Obesity
37

Adipocyte volume 34
Pharmacological agents References Direction of effect
Insulin
26, 38, 39

Glucose
40

Glucocorticoids
38, 39

Hexosamines
4143

Cytokines (TNF, IL-1 and LIF) and lipopolysaccharide (LPS)
44, 45

β3-adrenergic agonist (CL316,243)
35

Leptin
46

Thiazolidinediones (TZD) 10
Transcription factors References Direction of effect
C/EBPα
1719

ADD1/SREBP1
20

AP-2β
22

FOSL2 27

Transcriptional Regulation of LEP Gene Expression

Leptin (encoded by the LEP gene in humans, Lep in mice) is generally expressed in an adipocyte-specific fashion. Small amounts of leptin have been reported in brain, pituitary, trophoblast, stomach, mammary epithelial cells, liver, chondrocytes and muscle,4,12-14 but the physiological importance of this low level expression, if any, has not been determined.

The proximal promoters of the LEP gene in mouse and human have been characterized15,16 and a classic TATA box has been identified, as well as binding sites for C/EBP, Sp-1, GR, and CREB, and an E box element that may bind SREBP1c.17-20 Although a transgenic reporter driven by a 762 bp leptin proximal promoter cassette was able to drive gene expression in vivo, it showed lower levels of expression in adipocytes than in a variety of non-adipose tissues and was not affected by fasting, suggesting that this region is not responsible for adipose tissue specificity of leptin expression or physiological regulation of leptin gene expression.21 Recently, the transcription factor activator protein-2β (AP-2 β) has been shown to inhibit LEP expression by direct binding to the promoter.22 Various SNPs have been reported in the porcine leptin promoter that correlate with Lep expression23,24 and demethylation of specific CpG islands occurs during adipogenesis, associated with the onset of Lep expression25

All these studies have been limited by the fact that they have focused on the proximal promoter and/or on pre-specified adipocyte transcription factors. In fact, several major obstacles have impeded progress in identifying transcriptional pathways responsible for regulation of adipocyte-specific leptin gene expression, including (1) unreliable methods for unbiased detection of distal cis-regulatory elements, (2) insufficient tools to identify an unknown transcription factor binding to a known cis-regulatory element, and (3) a lack of good cell culture models for studying leptin gene expression in vitro. It is worth noting that the workhorse model of adipocyte research, the 3T3-L1 cell line, expresses extremely low levels of Lep26 and is thus unsuitable for these studies. Recent advances in all of these areas encouraged us to pursue the mechanism of adipocyte-specific leptin gene expression once again.27

Identification of a cis-Regulatory Element for Adipocyte-Specific LEP Gene Expression

We utilized a multi-step approach to finding cis-regulatory elements that direct lep gene expression. First, we sought to map such regions grossly, using BAC-transgenic reporter mice that express EGFP from the Lep translational start site. Analyzing multiple transgenic founders carrying BACs with varying lengths of Lep flanking sequences allowed us to narrow down the region required for adipose-specific expression and demonstrated that the three Lep exons, both introns, and 5.2 kb of 5′ flanking sequence were sufficient to drive adipocyte-specific EGFP expression. Next, we performed fine-mapping in vitro, by “tiling” across the upstream flanking region and through both introns with PCR primers, to generate reporter constructs that we could test in cultured human adipocytes. These cells are derived from human adipose stromal cells (hASCs), which can be differentiated into mature adipocytes in vitro and produce high levels of leptin. This effort identified a single 1 kb region approximately 4.5 kb upstream of the LEP transcriptional start site (TSS) that acts as enhancer in mature adipocytes. Further deletion and point mutations allowed us to discover a 30 bp region that was required for enhancer activity. Interestingly, we had previously performed chromatin state mapping in human adipocytes in culture, part of which involved mapping histone modifications associated with active and poised enhancers.28 The region we identified by reporter assays overlay an active enhancer region29,30 in our chromatin state maps (the only such place in the vicinity), which provided significant reassurance that we had identified a region of bona fide importance.

Identification of the Transcription Factor(s) for Adipocyte-Specific LEP Gene Expression

Once a relevant cis-regulatory element is found, it can be very difficult to identify the cognate transcription factor(s) that bind and activate it. The most common approach is to use computational motif finding to find sequences that suggest a particular factor. We tried this and found several interesting motifs for the androgen receptor (AR), peroxisome proliferator-activated receptor α (PPARα) and nuclear factor erythroid 2-related factor 1 (NFE2L1). Unfortunately, functional studies (i.e., overexpression and RNAi experiments) failed to demonstrate that any of these predicted factors were important in leptin expression. We therefore used our identified 30 bp cis-regulatory element as bait in an unbiased mass spectrometry-based quantitative proteomics approach combining stable isotope labeling by amino acids in cell culture (SILAC) with affinity enrichment of protein-DNA complexes using biotinylated DNA as affinity baits.31 To add specificity, we performed the assay in cultured human adipocytes as well as murine adipocytes differentiated from embryonic fibroblasts, both of which express leptin. Several transcription factors were found in each model, but FOSL2 and JUND were the only ones to appear in both. FOSL2 and JUND are both members of the AP1-transcription factor family, which includes FOS, FOSB, FOSL1, JUN, JUNB and JUND. We demonstrated that FOSL2 binds to our enhancer in mature human adipocytes, but not in pre-adipocytes, using ChIP-PCR.

FOSL2 is an Important Regulator of LEP Gene Expression In Vitro and In Vivo

To demonstrate the functional importance of FOSL2, we knocked it down with RNAi in human adipocytes; as predicted, this led to reduced LEP gene expression. Unfortunately, Fosl2 global knockout mice die shortly after birth and before most adipose development is complete, so leptin levels in these animals cannot be interrogated. We did look at Fosl2 heterozygous mice, which have normal adiposity however, and they show reduced circulating leptin, as do mice with adipocyte-specific deletion of Fosl2. In addition, osteoblastic precursor cells taken from the skulls of global Fosl2 knockout mice can be differentiated into adipocytes in vitro; these cells display lower Lep gene expression. Gain-of-function studies were a bit harder to perform, in that overexpression of Fosl2 in mature adipocytes did not affect leptin expression. However, forced expression of Fosl2 during the development process resulted in adipocytes with normal overall differentiation but elevated leptin expression. Similarly, osteoblastic precursors from Fosl2-transgenic mice showed higher Lep expression when differentiated into adipocytes in vitro. Since the Fosl2-transgenic mice suffered from systemic fibrosis and inflammation,32 metabolic studies of these animals could not be conducted. It appears that Fosl2 is both necessary and sufficient for leptin expression in adipocytes, but it must be present during the differentiation process to exert its effects. This implies that Fosl2 has both direct and indirect actions on the Lep gene.

Roles of FOSL2 in Regulating Leptin Levels in Physiology and Pathophysiology

As mentioned above, apart from being expressed in adipocytes, Lep mRNA synthesis in adipocytes is regulated by a variety of physiological and experimental stimuli. In mice, there are depot-specific differences in Lep mRNA content, with inguinal fat expressing higher leptin levels than retroperitoneal or epididymal adipose tissue.33 There also appears to be a direct relationship between adipocyte volume and Lep expression regardless of depot.34 In our study, a correlation of increased levels of LEP gene expression with increased levels of FOSL2 gene expression was observed during the course of differentiation of human adipocytes and in different adipose depots in mice. In addition, we showed that stimulation of mature human adipocytes with dexamethasone resulted in increased LEP expression and simultaneous rise in FOSL2 expression.

Fasting and Feeding Regulation of Leptin

One of the most important physiological functions of leptin is the regulation of fasting-feeding behavior.1 Fasting reduces Lep mRNA and feeding increases it.35 We measured Fosl2 mRNA during the fed state and after 24 h of fasting, but we did not detect any significant differences. It is very possible that the mechanisms regulating adipocyte-specific gene expression of Lep are different from the mechanisms which regulate other physiological functions of Lep, such as fasting and feeding. It is worth pointing out that our approach was geared toward the discovery of regulators of adipocyte-specific expression during differentiation, so it is perhaps unsurprising that Fosl2 might be not a regulator of the fasting-feeding response.

Roles of FOSL2 in Regulating Leptin Levels in Obesity

To determine if Fosl2 plays a role in the elevated Lep expression observed in obesity, we analyzed Fosl2 expression in different mouse models of obesity. In mice with diet-induced obesity as well as in mice with genetic obesity (db/db), we found that increased Lep gene expression was accompanied by increased Fosl2 levels. A gene expression analysis of samples of subcutaneous fat from a human cohort with varying BMIs confirmed this association in humans.

Conclusions and Unanswered Questions

Several issues, however, remain unresolved. First of all, we do not yet understand how FOSL2, which is not tissue restricted to adipocytes, can regulate adipose-specific gene expression. There are several intriguing possible mechanisms, many of which are known to be employed by members of the AP-1 transcription factor family, to which FOSL2 belongs. These include differential expression, composition and orientation of the heterodimeric binding partners, post-translational modifications, and interaction with ancillary proteins.36 Interestingly, although we identified JUND as a potential binding partner on our 30 bp cis-regulatory element, we could not demonstrate a specific requirement for this or any other Jun isoform in Lep expression. Perhaps there is greater functional redundancy among the Jun proteins than between FOS, FOSL1 and FOSB, which apparently cannot compensate for the loss of FOSl2 in leptin expression. Other factors may also be acting in concert with FOSL2. For example, there is a bona fide PPARγ binding site at the 5′ end of the 1 kb cis-regulatory region we discovered. Perhaps PPARγ plays a role in enabling FOSL2 binding during adipogenesis.

Our results should by no means be taken to exclude a possible role for other cis-regulatory regions important for Lep expression. Our BAC transgenic results suggest that multiple regions, especially at the 3′ end of the Lep gene, may be involved as well. For example, truncation of the 3′ flanking sequence in our BAC transgenic reporter mouse model led to a significant decrease of EGFP expression in adipose tissue and increased EGFP expression in other tissues. Taken together, these data suggest that many genomic regions and transcription factors will eventually be shown to regulate leptin expression.

Our study provided one of the first clues into the transcriptional regulation of the medically important adipokine leptin. Despite the fact that leptin levels are primarily regulated at the transcriptional level, our understanding of the factors at play has been poorly developed. We utilized an integrated strategy that combined BAC transgenesis, human reporter assays, epigenomics, and unbiased quantitative proteomics to identify FOSL2. We believe that a similar multi-pronged approach will be required to tackle many of the remaining problems in tissue-specific transcriptional regulation.

Wrann CD, Eguchi J, Bozec A, Xu Z, Mikkelsen T, Gimble J, et al. FOSL2 promotes leptin gene expression in human and mouse adipocytes. J Clin Invest. 2012;122:1010–21. doi: 10.1172/JCI58431.

Footnotes

References

  • 1.Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425–32. doi: 10.1038/372425a0. [DOI] [PubMed] [Google Scholar]
  • 2.Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, et al. Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 1995;269:543–6. doi: 10.1126/science.7624777. [DOI] [PubMed] [Google Scholar]
  • 3.Belgardt BF, Brüning JC. CNS leptin and insulin action in the control of energy homeostasis. Ann N Y Acad Sci. 2010;1212:97–113. doi: 10.1111/j.1749-6632.2010.05799.x. [DOI] [PubMed] [Google Scholar]
  • 4.Margetic S, Gazzola C, Pegg GG, Hill RA. Leptin: a review of its peripheral actions and interactions. Int J Obes Relat Metab Disord. 2002;26:1407–33. doi: 10.1038/sj.ijo.0802142. [DOI] [PubMed] [Google Scholar]
  • 5.Friedman JM. Leptin at 14 y of age: an ongoing story. Am J Clin Nutr. 2009;89:973S–9S. doi: 10.3945/ajcn.2008.26788B. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ebihara K, Kusakabe T, Hirata M, Masuzaki H, Miyanaga F, Kobayashi N, et al. Efficacy and safety of leptin-replacement therapy and possible mechanisms of leptin actions in patients with generalized lipodystrophy. J Clin Endocrinol Metab. 2007;92:532–41. doi: 10.1210/jc.2006-1546. [DOI] [PubMed] [Google Scholar]
  • 7.Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R, Hunt T, et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA. 1999;282:1568–75. doi: 10.1001/jama.282.16.1568. [DOI] [PubMed] [Google Scholar]
  • 8.Ravussin E, Smith SR, Mitchell JA, Shringarpure R, Shan K, Maier H, et al. Enhanced weight loss with pramlintide/metreleptin: an integrated neurohormonal approach to obesity pharmacotherapy. Obesity (Silver Spring) 2009;17:1736–43. doi: 10.1038/oby.2009.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mittendorfer B, Horowitz JF, DePaoli AM, McCamish MA, Patterson BW, Klein S. Recombinant human leptin treatment does not improve insulin action in obese subjects with type 2 diabetes. Diabetes. 2011;60:1474–7. doi: 10.2337/db10-1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kallen CB, Lazar MA. Antidiabetic thiazolidinediones inhibit leptin (ob) gene expression in 3T3-L1 adipocytes. Proc Natl Acad Sci U S A. 1996;93:5793–6. doi: 10.1073/pnas.93.12.5793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Harris RB, Ramsay TG, Smith SR, Bruch RC. Early and late stimulation of ob mRNA expression in meal-fed and overfed rats. J Clin Invest. 1996;97:2020–6. doi: 10.1172/JCI118637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Goïot H, Laigneau JP, Devaud H, Sobhani I, Bado A. Similarities and differences in the transcriptional regulation of the leptin gene promoter in gastric and adipose cells. FEBS Lett. 2005;579:1911–6. doi: 10.1016/j.febslet.2005.02.031. [DOI] [PubMed] [Google Scholar]
  • 13.Moreno-Aliaga MJ, Swarbrick MM, Lorente-Cebrín S, Stanhope KL, Havel PJ, Martínez JA. Sp1-mediated transcription is involved in the induction of leptin by insulin-stimulated glucose metabolism. J Mol Endocrinol. 2007;38:537–46. doi: 10.1677/JME-06-0034. [DOI] [PubMed] [Google Scholar]
  • 14.Simopoulou T, Malizos KN, Iliopoulos D, Stefanou N, Papatheodorou L, Ioannou M, et al. Differential expression of leptin and leptin’s receptor isoform (Ob-Rb) mRNA between advanced and minimally affected osteoarthritic cartilage; effect on cartilage metabolism. Osteoarthritis Cartilage. 2007;15:872–83. doi: 10.1016/j.joca.2007.01.018. [DOI] [PubMed] [Google Scholar]
  • 15.de la Brousse FC, Shan B, Chen JL. Identification of the promoter of the mouse obese gene. Proc Natl Acad Sci U S A. 1996;93:4096–101. doi: 10.1073/pnas.93.9.4096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gong DW, Bi S, Pratley RE, Weintraub BD. Genomic structure and promoter analysis of the human obese gene. J Biol Chem. 1996;271:3971–4. doi: 10.1074/jbc.271.8.3971. [DOI] [PubMed] [Google Scholar]
  • 17.He Y, Chen H, Quon MJ, Reitman M. The mouse obese gene. Genomic organization, promoter activity, and activation by CCAAT/enhancer-binding protein alpha. J Biol Chem. 1995;270:28887–91. doi: 10.1074/jbc.270.48.28887. [DOI] [PubMed] [Google Scholar]
  • 18.Hwang CS, Mandrup S, MacDougald OA, Geiman DE, Lane MD. Transcriptional activation of the mouse obese (ob) gene by CCAAT/enhancer binding protein alpha. Proc Natl Acad Sci U S A. 1996;93:873–7. doi: 10.1073/pnas.93.2.873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Miller SG, De Vos P, Guerre-Millo M, Wong K, Hermann T, Staels B, et al. The adipocyte specific transcription factor C/EBPalpha modulates human ob gene expression. Proc Natl Acad Sci U S A. 1996;93:5507–11. doi: 10.1073/pnas.93.11.5507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kim JB, Sarraf P, Wright M, Yao KM, Mueller E, Solanes G, et al. Nutritional and insulin regulation of fatty acid synthetase and leptin gene expression through ADD1/SREBP1. J Clin Invest. 1998;101:1–9. doi: 10.1172/JCI1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chen XL, Hartzell DL, McGraw RA, Hausman GJ, Dean RG. Analysis of a 762-bp proximal leptin promoter to drive and control regulation of transgene expression of growth hormone receptor in mice. Biochem Biophys Res Commun. 1999;262:187–92. doi: 10.1006/bbrc.1999.1176. [DOI] [PubMed] [Google Scholar]
  • 22.Fuke T, Yoshizaki T, Kondo M, Morino K, Obata T, Ugi S, et al. Transcription factor AP-2beta inhibits expression and secretion of leptin, an insulin-sensitizing hormone, in 3T3-L1 adipocytes. Int J Obes (Lond) 2010;34:670–8. doi: 10.1038/ijo.2009.295. [DOI] [PubMed] [Google Scholar]
  • 23.Stachowiak M, Mackowski M, Madeja Z, Szydlowski M, Buszka A, Kaczmarek P, et al. Polymorphism of the porcine leptin gene promoter and analysis of its association with gene expression and fatness traits. Biochem Genet. 2007;45:245–53. doi: 10.1007/s10528-006-9070-x. [DOI] [PubMed] [Google Scholar]
  • 24.Liu D, Hu Y, Yang X, Liu Y, Wei S, Jiang Y. Identification and genetic effects of a novel polymorphism in the distal promoter region of porcine leptin gene. Mol Biol Rep. 2011;38:2051–7. doi: 10.1007/s11033-010-0330-9. [DOI] [PubMed] [Google Scholar]
  • 25.Melzner I, Scott V, Dorsch K, Fischer P, Wabitsch M, Brüderlein S, et al. Leptin gene expression in human preadipocytes is switched on by maturation-induced demethylation of distinct CpGs in its proximal promoter. J Biol Chem. 2002;277:45420–7. doi: 10.1074/jbc.M208511200. [DOI] [PubMed] [Google Scholar]
  • 26.MacDougald OA, Hwang CS, Fan H, Lane MD. Regulated expression of the obese gene product (leptin) in white adipose tissue and 3T3-L1 adipocytes. Proc Natl Acad Sci U S A. 1995;92:9034–7. doi: 10.1073/pnas.92.20.9034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wrann CD, Eguchi J, Bozec A, Xu Z, Mikkelsen T, Gimble J, et al. FOSL2 promotes leptin gene expression in human and mouse adipocytes. J Clin Invest. 2012;122:1010–21. doi: 10.1172/JCI58431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Mikkelsen TS, Xu Z, Zhang X, Wang L, Gimble JM, Lander ES, et al. Comparative epigenomic analysis of murine and human adipogenesis. Cell. 2010;143:156–69. doi: 10.1016/j.cell.2010.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bernstein BE, Stamatoyannopoulos JA, Costello JF, Ren B, Milosavljevic A, Meissner A, et al. The NIH Roadmap Epigenomics Mapping Consortium The NIH Roadmap Epigenomics Mapping Consortium. Nat Biotechnol. 2010;28:1045–8. doi: 10.1038/nbt1010-1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Creyghton MP, Cheng AW, Welstead GG, Kooistra T, Carey BW, Steine EJ, et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci U S A. 2010;107:21931–6. doi: 10.1073/pnas.1016071107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ong SE, Schenone M, Margolin AA, Li X, Do K, Doud MK, et al. Identifying the proteins to which small-molecule probes and drugs bind in cells. Proc Natl Acad Sci U S A. 2009;106:4617–22. doi: 10.1073/pnas.0900191106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Eferl R, Hasselblatt P, Rath M, Popper H, Zenz R, Komnenovic V, et al. Development of pulmonary fibrosis through a pathway involving the transcription factor Fra-2/AP-1. Proc Natl Acad Sci U S A. 2008;105:10525–30. doi: 10.1073/pnas.0801414105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Guo KY, Halo P, Leibel RL, Zhang Y. Effects of obesity on the relationship of leptin mRNA expression and adipocyte size in anatomically distinct fat depots in mice. Am J Physiol Regul Integr Comp Physiol. 2004;287:R112–9. doi: 10.1152/ajpregu.00028.2004. [DOI] [PubMed] [Google Scholar]
  • 34.Zhang Y, Guo KY, Diaz PA, Heo M, Leibel RL. Determinants of leptin gene expression in fat depots of lean mice. Am J Physiol Regul Integr Comp Physiol. 2002;282:R226–34. doi: 10.1152/ajpregu.00392.2001. [DOI] [PubMed] [Google Scholar]
  • 35.Zhang Y, Matheny M, Zolotukhin S, Tumer N, Scarpace PJ. Regulation of adiponectin and leptin gene expression in white and brown adipose tissues: influence of beta3-adrenergic agonists, retinoic acid, leptin and fasting. Biochim Biophys Acta. 2002;1584:115–22. doi: 10.1016/s1388-1981(02)00298-6. [DOI] [PubMed] [Google Scholar]
  • 36.Chinenov Y, Kerppola TK. Close encounters of many kinds: Fos-Jun interactions that mediate transcription regulatory specificity. Oncogene. 2001;20:2438–52. doi: 10.1038/sj.onc.1204385. [DOI] [PubMed] [Google Scholar]
  • 37.Frederich RC, Löllmann B, Hamann A, Napolitano-Rosen A, Kahn BB, Lowell BB, et al. Expression of ob mRNA and its encoded protein in rodents. Impact of nutrition and obesity. J Clin Invest. 1995;96:1658–63. doi: 10.1172/JCI118206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bradley RL, Cheatham B. Regulation of ob gene expression and leptin secretion by insulin and dexamethasone in rat adipocytes. Diabetes. 1999;48:272–8. doi: 10.2337/diabetes.48.2.272. [DOI] [PubMed] [Google Scholar]
  • 39.Buyse M, Viengchareun SAY, Bado A, Lombès M. Insulin and glucocorticoids differentially regulate leptin transcription and secretion in brown adipocytes. FASEB J. 2001;15:1357–66. doi: 10.1096/fj.00-0669com. [DOI] [PubMed] [Google Scholar]
  • 40.Mueller WM, Gregoire FM, Stanhope KL, Mobbs CV, Mizuno TM, Warden CH, et al. Evidence that glucose metabolism regulates leptin secretion from cultured rat adipocytes. Endocrinology. 1998;139:551–8. doi: 10.1210/en.139.2.551. [DOI] [PubMed] [Google Scholar]
  • 41.Wang J, Liu R, Hawkins M, Barzilai N, Rossetti L. A nutrient-sensing pathway regulates leptin gene expression in muscle and fat. Nature. 1998;393:684–8. doi: 10.1038/31474. [DOI] [PubMed] [Google Scholar]
  • 42.Considine RV, Cooksey RC, Williams LB, Fawcett RL, Zhang P, Ambrosius WT, et al. Hexosamines regulate leptin production in human subcutaneous adipocytes. J Clin Endocrinol Metab. 2000;85:3551–6. doi: 10.1210/jc.85.10.3551. [DOI] [PubMed] [Google Scholar]
  • 43.Zhang P, Klenk ES, Lazzaro MA, Williams LB, Considine RV. Hexosamines regulate leptin production in 3T3-L1 adipocytes through transcriptional mechanisms. Endocrinology. 2002;143:99–106. doi: 10.1210/en.143.1.99. [DOI] [PubMed] [Google Scholar]
  • 44.Grunfeld C, Zhao C, Fuller J, Pollack A, Moser A, Friedman J, et al. Endotoxin and cytokines induce expression of leptin, the ob gene product, in hamsters. J Clin Invest. 1996;97:2152–7. doi: 10.1172/JCI118653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sarraf P, Frederich RC, Turner EM, Ma G, Jaskowiak NT, Rivet DJ, 3rd, et al. Multiple cytokines and acute inflammation raise mouse leptin levels: potential role in inflammatory anorexia. J Exp Med. 1997;185:171–5. doi: 10.1084/jem.185.1.171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wang J, Liu R, Liu L, Chowdhury R, Barzilai N, Tan J, et al. The effect of leptin on Lep expression is tissue-specific and nutritionally regulated. Nat Med. 1999;5:895–9. doi: 10.1038/10577. [DOI] [PubMed] [Google Scholar]

Articles from Adipocyte are provided here courtesy of Taylor & Francis

RESOURCES