Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Mammalian adipose tissue can be divided into two major types, namely, white adipose tissue (WAT) and brown adipose tissue (BAT). According to classical view, the main function of WAT is to store excess energy in the form of triglycerides, while BAT is a thermogenic tissue that acts a pivotal part in maintaining the core body temperature. White adipocytes display high plasticity and can transdifferentiate into beige adipocytes which have many similar morphological and functional properties with brown adipocytes under the stimulations of exercise, cold exposure and other factors. This phenomenon is also known as 'browning of WAT'. In addition to transdifferentiation, beige adipocytes can also come from de novo differentiation from tissue-resident progenitors. Activating BAT and inducing browning of WAT can accelerate the intake of glycolipids and reduce the insulin secretion requirement, which may be a new strategy to improve glycolipids metabolism and insulin resistance of obese and type 2 diabetes mellitus (T2DM) patients. This review mainly discusses the significance of brown and beige adipose tissues in the treatment of obesity and T2DM, and focuses on the effect of the browning agent on obesity and T2DM, which provides a brand-new theoretical reference for the prevention and treatment of obesity and T2DM.

Free full text 


Logo of adipocyteLink to Publisher's site
Adipocyte. 2021; 10(1): 48–65.
PMCID: PMC7801117
PMID: 33403891

Brown and beige adipose tissue: a novel therapeutic strategy for obesity and type 2 diabetes mellitus

ABSTRACT

Mammalian adipose tissue can be divided into two major types, namely, white adipose tissue (WAT) and brown adipose tissue (BAT). According to classical view, the main function of WAT is to store excess energy in the form of triglycerides, while BAT is a thermogenic tissue that acts a pivotal part in maintaining the core body temperature. White adipocytes display high plasticity and can transdifferentiate into beige adipocytes which have many similar morphological and functional properties with brown adipocytes under the stimulations of exercise, cold exposure and other factors. This phenomenon is also known as ‘browning of WAT’. In addition to transdifferentiation, beige adipocytes can also come from de novo differentiation from tissue-resident progenitors. Activating BAT and inducing browning of WAT can accelerate the intake of glycolipids and reduce the insulin secretion requirement, which may be a new strategy to improve glycolipids metabolism and insulin resistance of obese and type 2 diabetes mellitus (T2DM) patients. This review mainly discusses the significance of brown and beige adipose tissues in the treatment of obesity and T2DM, and focuses on the effect of the browning agent on obesity and T2DM, which provides a brand-new theoretical reference for the prevention and treatment of obesity and T2DM.

KEYWORDS: Brown adipose tissue, beige adipose tissue, white adipose tissue, browning of white adipose tissue, obesity, type 2 diabetes mellitus

Introduction

Obesity is a chronic metabolic disease caused by genetic, environmental, psychological and social factors. It is characterized by the imbalance of white adipose tissue (WAT) and brown adipose tissue (BAT). The global obesity rate continues to rise, which must be the result of a long-term imbalance between energy intake and energy expenditure. At present, about 2.2 billion people are overweight in the world, accounting for about one third of the global population, of which about 712 million people (ten percent of the global population) are obese people [1]. Obesity has a profound impact on tissue insulin sensitivity, and therefore has an impact on systemic glucose homoeostasis. Obesity is prone to a variety of diseases, which have been considered as potential causes or contributing factors of many obesity-related diseases, including insulin resistance and type 2 diabetes mellitus (T2DM) [2,3]. T2DM is a serious chronic metabolic disease that occurs when the blood glucose levels increase as a consequence of peripheral insulin resistance, which can be accompanied by the subsequent development of beta-cell failure and insulin deficiency. At present, there are 463 million diabetics. According to this growth trend, there will be 700 million diabetics in the world by 2045 [4]. Although there are many treatment methods for obesity and T2DM, including drugs that protect pancreatic cell function, increase insulin sensitivity and improve glucose and lipid metabolism, many drugs will bring adverse reactions. In addition, only a few drugs have been approved in many countries/regions, and the therapeutic effect of these drugs is not as expected. Therefore, it is necessary to find a novel therapy that can reduce calorie accumulation or increase energy expenditure to improve insulin sensitivity, reduce weight and maintain the activity of pancreatic beta cells.

It is known to all that there are two main types of mammalian adipose tissue: WAT and BAT. In the 1960s and early 1970s, it was thought that BAT was abundant in infancy, but regressed in adulthood, and there was almost no BAT or active BAT in adults [5,6]. However, with the in-depth researches of adipose tissue, a large quantity of bioactive brown/beige adipocytes were found in the neck and shoulder regions of adults by 18F-FDG-PET/CT detection [7,8]. It is currently believed that white and brown adipocytes arise from distinct embryonic precursors. White adipocytes mainly originate from myogenic factor 5 negative cells (Myf5), while brown adipocytes originate from myogenic factor 5 positive cells (Myf5+) [9,10]. Apart from different origins, their biological functions are totally different. White adipocytes (Figure 1) are specialized in lipid storage and mobilization. In times of caloric need, white adipocytes provide a long-term metabolic fuel via lipolysis and the release of fatty acids. Besides, white adipocytes are also involved in functions such as hormone secretion, immune function. On the other hand, brown adipocytes are specialized in energy expenditure and utilize chemical energy for thermogenesis, which acts a pivotal part in maintaining the core body temperature. Brown adipocytes (Figure 1) are characterized by multilocular lipid droplets and abundant mitochondria which contain a unique protein called uncoupling protein 1 (UCP1). Residing in the inner mitochondrial membrane, UCP1, a very significant regulatory factor in thermogenesis, uncouples mitochondrial respiration from ATP synthesis resulting in thermogenesis [11].

Comparison of white adipocytes, beige adipocytes and brown adipocytes in location, morphology, UCP1 expression level, mitochondrial density, function and marker genes. In adults, the subcutaneous fat is characterized by classic WAT. The BAT on clavicle consists of white, beige and brown adipocytes, while the classic BAT can be found in the deep neck, armpit and adjacent to skeletal muscle tissue. The fat adjacent to gonads, groin, mesentery and peritoneum of adult mice has the characteristics of classical WAT. The beige adipocytes were found in the groin of mice, while the classical BAT was found in the scapular area. Transcription factor 21 (TCF21), Transducin-like enhancer of split 3 (TLE3), Tumour necrosis factor receptor superfamily, member 9(CD137), T-Box 1 (TBX1), Transmembrane protein 26 (TMEM26), LIM homeobox protein 8 (LHX8), zinc finger protein of the cerebellum 1 (ZIC1)

However, with further insight into adipose tissue, it has been observed that in the case of cold exposure or activation of β-adrenergic receptors (β-ARs), a distinct type of adipocyte named beige adipocyte (Figure 1) can be detected in WAT. This phenomenon is known as ‘browning of WAT’ [12,13]. The origin of beige adipocytes is still controversial. It mainly includes the following points: (a) Beige adipocytes are derived from the transformation of mature white adipocytes; (b) Beige adipocytes exist a distinct beige adipocyte precursor; (c) Beige adipocytes come from de novo differentiation from tissue-resident progenitors [14,15]. Recently, it has been found that the number of UCP1 positive cells in beige adipose tissue and classical BAT is controlled by different genetic factors [16]. Beige adipocytes, at least those in the mouse subcutaneous depot, do not originate from myogenic factor 5 (myf5)-positive embryonic precursors that give rise to brown adipocytes [16]. Importantly, the metabolic characteristics of both brown adipocytes and beige adipocytes in the body have aroused great interest. Both brown adipocytes and beige adipocytes can uptake glucose and fatty acids to produce heat, which play a decisive role in regulating glucose and lipid metabolism in the whole body. It has been confirmed that the activation of brown and beige adipose tissues may represent a valid strategy to treat obesity and T2DM [17–19]. Therefore, elucidating the regulatory mechanisms of brown and beige adipose tissues may provide new targets for treating obesity and diabetes.

Role of brown and beige adipose tissue in obesity and T2DM

As previously stated, BAT acts a pivotal role in thermogenesis, contributing to energy consumption and preventing obesity. The thermogenic energy of BAT mainly comes from β-oxidation of fatty acids, while only a small part of the energy comes from glucose metabolism. However, BAT has a substantial impact on glucose homoeostasis in vivo, as BAT has a high rate of glucose uptake after activation compared with other metabolically active tissues [20]. BAT is one of the most insulin-sensitive tissues, and insulin enhances BAT glucose uptake, and glucose uptake by BAT can produce ATP through anaerobic hydrolysis in the cytoplasm. On the one hand, it can be used for the activation of free fatty acid (FFA) before entering the mitochondria for β-oxidation; on the other hand, it can prevent ATP deficiency caused by the uncoupling of oxidative phosphorylation in mitochondria [21]. In addition, glucose can also be used in the synthesis of FFA and triglycerides [22]. It has been found that activated BAT can take up and utilize glucose and lipid, and thereby improving beta-cell function and reducing the demand of insulin secretion from pancreatic beta cells [17]. FDG-PET/CT detection confirmed that the fasting glucose concentration of individuals with BAT was lower than that of individuals without BAT [23]. Under the conditions of cold exposure and insulin stimulation, both glucose uptake rate and insulin sensitivity of obese patients were lower than those lean subjects, suggesting that the effects of cold and insulin on BAT activity are markedly blunted in obesity [22]. Similarly, cold exposure or BAT transplantation can also improve glucose tolerance and insulin sensitivity in mice [24–26]. However, when the insulin receptor of brown adipocytes was knocked out, mice showed age-dependent interscapular brown fat loss, but the expression of UCP1 and UCP2 increased [27]. Moreover, these mice developed defective insulin secretion, leading to progressive glucose intolerance without insulin resistance [27]. The above results suggest a role of insulin receptors in brown fat development, as well as a role for BAT in regulating insulin secretion and glucose homoeostasis.

In addition to classic BAT activation to treat obesity and T2DM, the recruitment of beige adipocytes has recently attracted much attention as a novel therapeutic target for obesity and T2DM. Beige adipocytes also play a paramount role in weight control, energy balance regulation and amelioration of glucose and lipid metabolism. Under external stimuli (such as cold exposure, β3-adrenergic agonists, etc.), beige adipocytes recruited by WAT accelerate the absorption of circulating glucose and lipids, and increase energy consumption and thermogenesis [28,29]. The consumption of glucose and lipids indirectly improves glucose tolerance, insulin sensitivity and beta-cell function. In fact, brown and beige adipose tissues are potential therapeutic targets to treat obesity and T2DM due to their inherent thermogenic capacity and their ability to improve glucose metabolism. The protective effects of brown and beige adipose tissues against obesity and T2DM rely on the recruitment of beige adipocytes and the enhancement of BAT amount and/or BAT activity, resulting in increased UCP1 expression, energy expenditure and thermogenesis (Figure 2). Activation of beige and brown adipocytes increases glucose uptake, fatty acid oxidation and lipolysis, which results in the increase of insulin sensitivity and reduction in blood glucose, blood lipids and fat mass, thus preventing obesity and T2DM [30]. Overall, brown fat and beige adipose tissue display a markedly high metabolic activity, which may be used as an underlying therapeutic target for the treatment of obesity and T2DM.

Protective effects of beige and brown adipose tissue against obesity and T2DM. The recruitment of beige adipocytes and the increase of BAT amount and/or BAT activity can promote the expression of UCP1 and increase heat production. Overexpression of UCP1 protein will enhance glucose uptake, fatty acid oxidation and lipolysis of both beige and brown adipocytes, which leads to increased insulin sensitivity and reduction in blood glucose, blood lipids and fat mass, thus preventing T2DM. Note: Upward pointing green arrows and downward pointing red arrows indicate ‘increase’ and ‘decrease’, respectively

Effect of browning agents on obesity and T2DM

To further insight into the mechanism of brown and beige adipose tissue on obesity and T2DM, the effect of browning agents on obesity and T2DM is discussed in the following paragraphs (Figure 3 and Figure 4)

Detailed mechanisms of anti-obesity and type 2 diabetes with browning agents such as exercise, cold exposure, and brown fat transplantation. Note: The green arrow (→) indicates activation or promotion

Detailed mechanisms of anti-obesity and T2DM with endogenous browning agents (miRNA, BMP, Irisin and IL-6). Note: The green arrow (→) indicates activation or promotion, and Inverted ‘T’ () indicates inhibition

Exercise

Exercise benefits improvement of glucose homoeostasis and offers protection against metabolic disorders such as obesity and diabetes [31]. It has been found that exercise training improves insulin resistance, enhances glucose disposal and muscle glycogen storage by increasing the expression of GLUT4 [32]. In sports training, in addition to using glucose as an energy source, skeletal muscles also use part of lipids as an energy source to accelerate the intake and utilization of free fatty acids (FFA) [33]. In addition, exercise has been reported to induce browning of WAT and enhance energy consumption by promoting hypothalamic brain-derived neurotrophic factor (BDNF) expression [34]. Recently, it has been proved that several novel factors can induce browning of WAT. During the exercise, PGC1-α expression in mouse muscles stimulated the expression of fibronectin type III domain containing 5 (FNDC5), which is cleaved and identified hormone known as irisin [35]. Irisin has been shown to promote the recruitment of brown adipocytes in the WAT of mice [35]. As a circulating factor, meteorin-like (Metrnl) induced in muscles after exercise increases energy expenditure and improves glucose tolerance and the expression of genes associated with beige fat thermogenesis by stimulating an eosinophil and promoting alternative activation of adipose tissue macrophages [31].

In a human experimental study, compared with lean sedentary men, male endurance training athletes had lower activity of BAT; meanwhile, there was no significant difference in the expression of the classical brown adipocyte marker gene in subcutaneous WAT between the two groups [36]. It is speculated that the reason of the aforementioned findings may be that exercise training activates thermogenesis of skeletal muscles, thereby reducing the metabolic activity of BAT in humans. It seems that thermogenic processes of BAT and muscle do not occur simultaneously during exercise. In another human experiment, subcutaneous adipocytes became smaller and fatty acid composition changed after long-term exercise, which was probably related to the preferential mobilization of palmitic acid and oleic acid, and the preferential retention of stearic acid and linoleic acid [37]. The decrease of palmitic acid and oleic acid content in adipose tissue after exercise may be considered as a favourable change. Moreover, lactic acid, the product of anaerobic exercise, is likely to induce browning of WAT [38]. In rodents, exercise seemed to be a weak stimulus for BAT thermogenesis. Surprisingly, exercise increases mitochondrial biogenesis and activity in both subcutaneous WAT and visceral WAT, it also increases the expression of the brown adipocyte marker UCP1 in both adipose tissue depots, although these effects are much more pronounced in subcutaneous WAT [39–41]. This is inconsistent with the results of human experiments, which may be related to species differences, different locations of adipose tissues in humans and animals, and different experimental conditions.

Cold exposure

Cold can activate the sympathetic nervous system (SNS) and stimulate the release of norepinephrine (NE) [42]. NE binds to and activates the β-adrenergic receptors (β-ARs) in adipocytes, which contributes to the lipolysis of triglycerides in white and brown adipocytes [43]. Activated β-ARs interact with GTP binding protein GS of heterotrimer to activate adenylate cyclase, thereby increasing the level of cAMP in cells and activating cAMP-dependent protein kinase A (PKA) [42]. In fat cells phosphorylated PKA modifies lipid droplet binding proteins (such as lipid droplet binding proteins A and B) and hormone-sensitive lipases, thus leading to the release of FFA and glycerol, which are exported into the circulation and used as fuel for other tissues [44,45]. The released fatty acids can activate the UCP1 protein and act as a substrate for UCP1-mediated thermogenesis [21]. Furthermore, these fatty acids also conduce to regulating transcription control through peroxisome-proliferator-activated receptors α (PPAR α) and PPAR γ [46]. PPAR γ is of great significance in the differentiation of adipocytes in both WAT and BAT [47,48]. It has been confirmed that PPAR γ is conducive to improving the mitochondrial biosynthesis and oxidation function of beige adipocytes in vivo and in vitro by regulating PGC-1α, thus promoting the browning of WAT [49–51]. PGC-1α seems to be necessary for cold exposure to induce beige adipocyte recruitment and survival in WAT [52].

BAT activity is sustained by the SNS. Both acute and chronic cold exposure enhance energy expenditure and thermogenesis of brown and beige adipose tissues [53,54]. It has been found that BAT activated by cold exposure contributes to enhancing the uptake of triglyceride-rich lipoproteins (TRLs) through lipoprotein lipase (LPL) and transmembrane receptor CD36, thereby accelerating the removal of serum triglycerides [55]. In addition, cold exposure can also increase glucose uptake and insulin sensitivity by enhancing the expression of glucose transporter protein 4 (GLUT4) [56]. Cold exposure can activate beige and brown adipose tissue, and promote the release of factors that induce the browning of WAT in both myocardium and skeletal muscle [57]. For example, the release of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) from cardiomyocytes after cold exposure can enhance the expression of both PGC-1α and UCP1, as well as mitochondrial biosynthesis and energy expenditure in WAT [57]. It has been shown that ANP increases BAT mitochondrial activity in a p38 mitogen-activated protein kinase (p38 MAPK)-dependent manner [58]. BNP can increase the expression of UCP1 and PGC-1α in WAT and BAT, as well as increase respiration and energy consumption [58].

BAT transplantation

BAT is a metabolically active organ that participates in maintaining energy homoeostasis. Therefore, functional BAT transplantation may bring therapeutic benefits in treating diabetes, obesity and other related metabolic diseases. It has been shown that BAT transplantation can significantly prevent weight gain, reduce the total fat weight and enhance oxygen consumption in obese mice, thereby ameliorating IR and liver steatosis [59]. It has been found that when the BAT used for transplantation was obtained from interleukin-6 (IL-6)-knockout mice, high-fat diet-induced recipient mice did not reverse insulin resistance, indicating that IL-6 could serve as the secreted factor at least partially mediating the beneficial effects observed in the BAT transplantation studies [60]. It has been confirmed that BAT transplantation can heighten the level of circulating adiponectin, increase expression of β-AR and fatty acid oxidation-related genes in WAT [61]. BAT transplantation models successfully demonstrated the improvements in glucose metabolism and insulin sensitivity, as well as reductions in body mass and decreased adiposity in transplant recipients [62]. BAT transplantation enhanced the uptake of glucose by endogenous BAT, WAT and myocardium, surprisingly, not by skeletal muscles [62]. Also, it has been shown that transplantation of embryonic BAT can correct type 1 diabetes mellitus (T1DM) in streptozotocin-treated mice and reverse symptoms of clinical diabetes such as polyuria, polydipsia and polyphagia [63]. Further research found that plasma insulin-like growth factor-1 (IGF-1) was the first hormone to increase after BAT transplantation in nonobese diabetic mice [64]. As with streptozotocin-treated mice, euglycemia was independent of insulin. Also, it has been suggested that IGF-1 may directly reduce blood glucose through the activation of insulin receptor. Altogether, these findings support the potential for insulin-independent reversal of T1DM with BAT transplantation and suggest IGF-1 as a potential mediator in the resulting equilibrium [64].

Gut microbiota

Gut microbiota is established with the development of the host, and its composition is affected by various factors such as obesity and pregnancy [65,66]. Some changes of gut microbiota composition are associated with T2DM and obesity. Gut microbiota can affect the metabolism and insulin sensitivity of the host, and it is a significant regulator of host metabolic homoeostasis and energy balance. In recent years, a close relationship between gut microbiota and thermogenesis process of adipose tissues has been found [67]. Gut microbiota can activate BAT and promote browning of WAT. It has been shown that the water extract of Caulis Spatholobi can ameliorate obesity through activating BAT and modulating the composition of gut microbiota, which induced an increase of anti-obesity and anti-diabetes related bacteria genus [68]. In a cold environment, the expression of UCP1 protein in BAT and subcutaneous WAT of mice treated with antibiotics was reduced, resulting in cold intolerance [69]. However, intragastric administration of butyrate heightened the thermogenic ability of mice treated with antibiotics and reversed this defect [69]. The abovementioned results indicate that the depletion of microbiota weakens the adaptive thermogenesis of BAT and subcutaneous WAT, and that gut microbiota contributes to the up-regulation of thermogenesis in cold environments, which may be mediated by butyrate. Gut microbiota transplantation technology has a vital effect on remodelling gut microbial composition. Interestingly, it has been demonstrated that a cold-conditioned gut microbiome does not influence adaptive thermogenesis [70]. Based on the above results, it is speculated that cold exposure may increase the content of short-chain fatty acids produced by gut microbiota, thereby increasing adaptive thermogenesis, but a cold-conditioned gut microbiome does not influence adaptive thermogenesis. The latest research demonstrated that resveratrol improved glucose homoeostasis by enhancing BAT activation and browning of WAT. In addition, when the gut microbiota of mice fed with resveratrol was transplanted into sterile mice, the glucose intolerance of sterile mice was improved. The research found that one of the underlying mechanisms might partially be mediated by the gut microbiota-bile acid-Takeda G-protein coupled receptor 5 (TGR5)/UCP1 pathway [71]. Moreover, resveratrol can regulate the composition of gut microbiota by regulating the ‘gut microbiota adipose tissue’ axis, thus increasing energy consumption and regulating glucose and lipid metabolism to combat obesity [72]. Therefore, regulating the intestinal flora to activate BAT and induce WAT browning is an effective strategy for protective effects against obesity and T2DM.

Intermittent fasting and caloric restriction

Intermittent fasting (IF) is an effective weight control strategy, but its mechanism is still unclear. A new study demonstrated that isocaloric IF improves glucose tolerance and postprandial insulin levels, and increases incretin expression in leptin-deficient ob/ob mice, indicating that isocaloric IF is effective in ameliorating nutrient-stimulated insulin secretion [73]. It has been found that IF can selectively stimulate the development of beige adipocytes in WAT, and significantly improve obesity, insulin resistance and liver steatosis by regulating intestinal microbial composition to increase the biosynthesis of acetate and lactate [74]. Interestingly, it has been reported that acetate and lactate are inducers of WAT browning [75,76].

Caloric restriction (CR) is another negative energy balance state, which can stimulate selective WAT glucose uptake, enhance the expression of type 2 immune response and sirtuin type1 (SIRT1), and promote the browning of WAT [77]. Further research demonstrated that these metabolic changes were mediated by increased eosinophil infiltration, type 2 cytokine signalling, and M2 macrophage polarization in adipose tissues of CR of animals [77]. Type 2 cytokines are necessary for browning of WAT and metabolic improvement during CR. Research showed that reducing energy intake weakened insulin-mediated lipolysis and affected the ratio of NAD+/NADH and AMP/ATP [78]. The increased ratio of NAD+/NADH activated the SIRT signalling pathway, thereby remodelling adipocyte phenotype [78]. SIRT1 suppresses white adipocyte development mediated by PPAR γ, and enhances lipolysis and energy expenditure [79]. Enforced expression of SIRT3 in HIB1B brown adipocytes can enhance the expression of UCP1, PGC-1α and a series of mitochondria-related genes, and play a decisive role in the adaptive thermogenesis of BAT [80]. Similarly, SIRT5 is necessary for the differentiation of brown adipocytes, and increases the expression of BAT marker genes through an indirect effect consisting of histone modifications [81].

Noncoding RNAs

Noncoding RNAs (ncRNAs) are transcripts lacking protein-coding capabilities, but they are of prominent significance in silencing the gene expression connected with complex gene regulatory networks [82,83]. According to the nucleotide length, ncRNAs are subsequently classified into long ncRNAs (lncRNAs) (>200 nucleotides) and small ncRNAs (sncRNAs) (<200 nucleotides). sncRNAs include microRNA (miRNA), small nuclear RNA (snRNA), circular RNA (circRNA), among others [84]. miRNAs are endogenous RNA molecules approximately 21–23 nucleotides in length, which play a prominent role in the regulation of gene expression [85].

It has been reported that miRNAs play a bidirectional role in activating brown and beige adipose tissues [84]. On the one hand, miRNAs play a positive regulatory role in activating BAT and inducing browning of WAT. Among the positive regulators, miR-455 is a new regulator that activates BAT. Under the condition of cold and the browning inducer bone morphogenesis proteins 7 (BMP7), miR-455 specifically expressed in BAT to regulate the differentiation and thermogenesis of brown adipocytes. In addition, adipose-specific miR-455 transgenic mice displayed browning of subcutaneous WAT after cold exposure [86]. miR-455 activates AMP-activated protein kinase (AMPK) α1 by targeting hypoxia-inducible factor 1α subunit inhibitor (HIF1an), which can promote the formation of beige fat cells and mitochondrial heat production [86]. Cold exposure can enhance expression of miR-32 in BAT, which is related to BAT specific super enhancer. By directly inhibiting its downstream target gene Tob1, miR-32 activates p38 MAPK signalling to promote fibroblast growth factor 21 (FGF21) expression and increase serum FGF21 level, thereby inducing browning of WAT [87]. Besides, miR-26 [88], miR-30b/c [89] and miR-196a [90] also play central roles in inducing browning of WAT.

On the other hand, miRNAs play a negative role in activating BAT and inducing browning of WAT. As a key negative regulator of brown and beige fat development and heat production, the overexpression of miR-199a/214 can inhibit the differentiation, thermogenic gene expression and mitochondrial respiration of brown adipocytes, while the knockout of miR-199a/214 can increase thermogenic gene expression and mitochondrial function of brown adipocytes [91]. Further research found that miR-199a/214 directly targeted PRDM16 and PGC-1α to inhibit the differentiation of brown fat cells and the development of beige fat [91]. miR-155 prevents the browning of WAT by inhibiting the expression of UCP1, Cidea, PPAR γ [92]. However, inhibition of miR-155 expression enhances brown adipocyte differentiation [92]. Similarly, miR-327 targets fibroblast growth factor 10 (FGF10) gene to prevent brown adipocyte differentiation and thermogenesis [93]. Besides, overexpression of miR-19b [94], miR-34a [95] and miR-133 [96] can also suppress brown adipocyte activity and beige adipocyte recruitment.

Bone morphogenetic proteins

As a class of multifunctional growth factors, bone morphogenesis proteins (BMPs) belong to the superfamily of transforming growth factor β (TGFβ). They were initially considered to be significant factors in promoting bone formation, but they were later identified to participate in the development of adipose tissue, intestine, brain, etc. [97]. Some members of the BMP family are of great importance in maintaining energy homoeostasis and participating in the early stage of adipogenesis. It was found that BMP2, 4, 6, 7 and 9 effectively induced mesenchymal stem cells to form adipocytes in vitro and in vivo [98]. In addition, BMP4, BMP7 and BMP8 can promote the differentiation of white adipocytes to brown adipocytes. Overexpression of BMP4 gene enhances the number of white adipocytes with similar characteristics as brown adipocytes and ameliorates insulin sensitivity [99]. BMP7 participates in the whole process of brown adipocyte development and acts a pivotal part in inducing the browning of WAT. BMP7 increases the expression of the transcriptional regulators PRDM16, PGC-1α, PPARγ and C/EBPs and promotes thermogenesis through p38 MAPK and PGC-1α dependent pathways [100]. BMP8b is another member of the BMP family, which can directly regulate thermogenesis. In mature BAT, BMP8b enhances the response to norepinephrine by increasing p38 MAPK/cAMP response element binding protein (CREB) signal transduction and lipase activity [101].

Irisin

In recent years, irisin as an underlying novel target for the treatment of obesity and insulin resistance, has attracted great interest and attention in the scientific community. Irisin expression can be induced by exercise, it increases energy consumption and ameliorates insulin resistance induced by high-fat diet [102]. Irisin binds to unknown receptors of white adipocytes, thereby increasing both PPARα and UCP1 expression, and inducing the browning of WAT [103]. Irisin can also enhance lipolysis through the cAMP-PKA-HSL/perilipin signalling pathway [104]. In addition, irisin can promote browning of WAT through p38 MAPK and extracellular regulated protein kinase (ERK) signalling pathway, thereby promoting islet β cell proliferation and ameliorating glucose tolerance [105]. Irisin enhances the energy consumption and thermogenesis of both skeletal muscles and BAT [106]. In conclusion, irisin can increase insulin sensitivity in skeletal muscle and adipose tissue, ameliorate the disorders of glucose and lipid metabolism and the function of pancreatic beta cells.

Fibroblast growth factor 21

Fibroblast growth factor 21 (FGF21) is a member of FGF family, it is expressed in liver, WAT, BAT, skeletal muscle and pancreas, and plays a prominent part in energy balance, glucose and lipid metabolism [107]. FGF21 is considered to be a promising agent for the treatment of T2DM and obesity. Clinical trials have reported that FGF21 has beneficial effects on lipid and bile acid metabolism, with clinical improvement in dyslipidemia, steatosis, weight loss and liver damage [108]. It has been found that FGF21 can enhance energy consumption by regulating the expression of thermogenic genes in adipose tissues such as BAT and subcutaneous WAT, thus reducing blood glucose and lipid levels and ameliorating glucose tolerance and insulin resistance in obese and diabetic mice [109–111]. Under long-term cold exposure, FGF21 expression in both BAT and subcutaneous WAT of mice increased significantly [112,113]. Under cold exposure conditions, FGF21 significantly enhances PGC-1α protein levels, and enables PGC-1α to participate in thermogenesis of adipose tissue induced by FGF21 [114]. FGF21 can activate BAT and induce browning of WAT through autocrine and paracrine actions [115]. FGF21 has been shown to increase PGC-1α protein expression and mitochondrial activity of 3T3L1 cells through the AMPK-SIRT1-PGC-1α signalling pathway, thereby enhancing mitochondrial function and increasing oxygen consumption [116]. In addition, FGF21 stimulates islet autophagy by inhibiting AMPK mTOR signalling pathway, which further consolidates the evidence that FGF21 is a drug therapeutic target for obesity and related T2DM [117].

Thyroid hormones

Thyroid hormones (THs) have a vital effect on thermoregulation. The interaction of THs and sympathetic nervous system can increase the expression of UCP1 protein in BAT, thus increasing thermogenesis of BAT [118]. It has been confirmed that THs can regulate BAT and induce browning of WAT [119]. In mice, BAT activity was decreased by hypothyroidism; by contrast, it was increased by hyperthyroidism [120]. Multi-compartment adipocytes and thermogenic gene expression were found in the WAT of mice with hyperthyroidism and hypothyroidism. It was found that hyperthyroidism and hypothyroidism may promote WAT thermogenesis through enhanced adrenergic signalling or compensation for impaired BAT function [120]. MAPK kinase 6 (MKK6) is highly expressed in WAT of obese individuals. However, MKK6 deletion increases energy expenditure and thermogenic capacity of WAT, protecting mice against diet-induced obesity and development of diabetes [121]. The underlying mechanism may be that deletion of MKK6 increases triiodothyronine (T3)-stimulated UCP1 expression in adipocytes, thereby increasing their thermogenic capacity [121]. In addition, coordinated glucagon and thyroid hormone actions synergize to correct hyperlipidaemia, steatohepatitis, atherosclerosis, glucose intolerance and obesity in mice with impaired metabolism [122].

Interleukin-6

Interleukin-6 (IL-6) is a cytokine secreted by skeletal muscle, T helper cells, white adipocytes and brown adipocytes [123–125]. IL-6 is an independent predictor of T2DM and is considered to be involved in the development of inflammation, insulin resistance and beta-cell dysfunction [126]. Also, increased plasma IL-6 levels have been observed in obese patients [127]. However, the release of central IL-6 can suppress weight gain and visceral obesity, and also enhance UCP1 protein expression in BAT by stimulating the sympathetic nervous system [127]. It has been reported that during acute cold exposure, IL-6 in the serum of mice increased significantly, which can maintain the core body temperature. The abovementioned results indicate that IL-6 may partly act on the central nervous system [128]. It has been found that IL-6 lost fat weight by promoting fat lysis and inducing browning of WAT [129]. Besides, burn promoted browning of WAT and increased lipolysis in mice, and IL-6 was significant regulator in this process [130]. IL-6 can be secreted when skeletal muscles respond to exercise and exerts insulin-sensitizing effects. It was found that BAT transplantation ameliorated glucose tolerance and insulin sensitivity, reduced body weight, increased FGF21 levels, and completely reversed insulin resistance in high fat diet-induced obese mice [60]. Importantly, when obtaining BAT for transplantation from IL-6-knockout mice, the improved metabolic profile was lost, which clearly demonstrated that IL-6 participated in the process by which BAT regulates glucose homoeostasis and insulin sensitivity [60].

Macrophage polarization

In adipose tissue, a pro-inflammatory response promotes insulin resistance, while an anti-inflammatory response enhances insulin sensitivity. The degree of inflammation of adipose tissue depends on the dynamic interaction between various immune cells. According to their immune and metabolic functions, the immune cells in fat can be divided into two categories: neutrophils, mast cells, M1 macrophages, etc., which mainly promote inflammation and reduce insulin sensitivity and glucose metabolism; the other category includes regulatory T cells, alternatively activated (M2) macrophages, eosinophils, etc., which mainly exert anti-inflammatory effects and can promote tissue repair and improve insulin sensitivity [131–134]. Macrophages can be used as regulators to activate BAT and promote browning of WAT, and regulate the thermogenesis process, so as to link the immune response with thermogenesis and energy consumption. Under cold exposure, IL-4 selectively activates M2 macrophages in adipose tissue, and the activated M2 macrophages secrete catecholamines to increase thermogenic gene expression in BAT and promote lipolysis in WAT [135]. Furthermore, it was found that the browning of WAT caused by burn was related to the increase of macrophage infiltration, and more M2 macrophages were observed in the fat of burn patients. Similarly, the increase in M2 macrophages was also observed in the fat of burn mice [136]. Macrophages in burn stressed subcutaneous WAT undergo alternate activation to enhance tyrosine hydroxylase expression and promote catecholamine production [136]. Therefore, macrophages with anti-inflammatory effects contribute to inducing browning of WAT, and enhance systemic thermogenesis and energy expenditure.

Current status of research on browning agents for the treatment of obesity and T2DM

Based on the great plasticity of adipose tissue in thermogenesis, activating BAT and inducing browning of WAT has become a new way of treating obesity and T2DM. The development of drugs that activate BAT and/or induce browning of WAT has become a research hotspot in recent years. Commonly used drugs include: β3 adrenergic receptor agonists (Mirabegron), thiazolidinediones (Rosiglitazone), Glucagon-like peptide-1 receptor agonists (GLP-1RAs) (Liraglutide) and Berberine etc. (Figure 5).

Detailed mechanisms of anti-obesity and T2DM with browning agents such as β3 AR agonists, TZDs, GLP-1RAs and Berberine etc. Note: The green arrow (→) indicates activation or promotion

By activating β-adrenergic receptor, β3-adrenergic receptor agonists can reduce the weight of both subcutaneous and epididymal WAT, promote the expression of UCP1 gene in subcutaneous WAT and BAT, increase thermogenesis and insulin sensitivity, and effectively ameliorate hyperglycaemia in diabetic animal models [137–139]. Unfortunately, β3-adrenergic receptor agonist drugs (L-796,568) failed in human trials [140]. Importantly, mirabegron, a new β3- adrenergic receptor agonist, acutely stimulates human BAT thermogenesis and increases resting metabolic rate [141]. Rosiglitazone, as a PPARγ agonist, can heighten the expression of mitochondrion thermogenic gene and UCP1 in 3T3-L1 adipocytes, increase the calcium inflow and glucose uptake mediated by α1-adrenoceptor in the subcutaneous groin and epididymal WAT of mice, and promote browning of WAT in mice [142,143]. As a GLP-1RAs, liraglutide can reduce visceral fat weight and enhance fatty acid oxidation. In addition, liraglutide can increase the expression of BAT marker genes in subcutaneous WAT of diabetic rats, promote the browning of subcutaneous WAT and ameliorate metabolic abnormalities in rats [144]. Retinoic acid plays an important role in adipocyte differentiation, adaptive thermogenesis and fatty acid oxidation. It can reduce body weight, improve insulin sensitivity and promote browning of WAT [145].

Ginseng extract can inhibit the obesity of db/db mice, and its mechanism is related to the induction of Enterococcus faecalis, which can produce an unsaturated long-chain fatty acid, myristoleic acid (MA), which can reduce adiposity by BAT activation and beige adipocyte formation [146]. The ethanol extract of Platycodon grandiflorum root can increase the intake of fatty acid in epididymal WAT by inducing the expression of related transporters, resulting in decreased blood FFA concentrations. Moreover, the ethanol extract of Platycodon grandiflorum root can increase the expression of browning related genes, the level of overall transcriptome (Bmp4, Ucp3, Sirt3, etc.) and enzyme activity of carnitine palmitoyltransferase, thereby enhancing energy expenditure and promoting the browning of epididymal WAT and energy consumption [147]. Berberine is the main bioactive component of Coptis chinensis, a traditional Chinese herbal medicine, which has a significant effect on regulating glucose and cholesterol levels, anti-obesity and anti-diabetic [148,149]. It was found that berberine enhanced the activity of BAT and the expression of the thermogenic protein in subcutaneous WAT of leptin receptor-deficient db/db mice through the AMPK signalling pathway [150]. Resveratrol, a kind of natural polyphenol substance with strong activity, has a high content in many traditional Chinese medicines such as Polygonum cuspidatum and Veratrum. It has been demonstrated that resveratrol can reduce lipogenesis, improve the rate of lipolysis, increase the expression of both PGC-1α protein and UCP1 protein, and promote the browning of WAT by activating the SIRT1 signalling pathway in adipose tissue [151]. Pentamethylquercetin can heighten mitochondrial biogenesis and upregulate UCP-1 expression in WAT of high-fat-diet fed mice, and it contributes to promoting the browning of WAT [152]. Curcumin has been shown to enhance the expression of β3-adrenergic receptor gene in subcutaneous WAT of mice, thereby increasing plasma levels of noradrenaline and thermogenesis [153]. Genipin as a product of gardenoside hydrolysed by β-glucosidase, ameliorated diet-induced obesity via promoting lipid mobilization and browning of WAT in rats [154]. Nicotine is the main bioactive substance in tobacco, which can affect food intake and energy consumption. Moreover, it can induce browning of WAT through central action, which depends on the κ opioid receptor secreted by the lateral hypothalamus [155]. In addition, menthol [156], cardamom [157], allicin [158] and capsaicin [159] can also promote browning of WAT to counteract obesity and T2DM.

Conclusion and prospects

In the past decade, scientists have learnt new information about the origin of BAT and WAT, and gained new insights about the plasticity of adipocytes. Activated BAT plays a prominent role in resisting cold and maintaining body temperature, meanwhile, it has the effects of ameliorating glucose homoeostasis and insulin sensitivity. Under the stimulation of certain conditions, based on the plasticity of WAT, beige adipocytes with similar characteristics and functions as those of brown adipocytes can originate from WAT. Beige and brown adipocyte-mediated thermogenesis may be a significant factor in determining human energy balance. Activating BAT and inducing WAT browning-mediated thermogenesis is an attractive strategy for improving glucose homoeostasis and insulin resistance, which can be used as a new option for the treatment of obesity and T2DM. However, in parallel with these exciting developments, there are still many important issues to be addressed. It has been found that both UCP1-dependent and UCP1-independent mechanisms play a role in diet-induced thermogenesis [160]. Beige adipocytes of abdominal fat are mostly UCP1 negative but possess thermogenic capacity associated with a futile creatine cycle [161]. Importantly, Kazak et al. found that creatine enhances energy expenditure through stimulation of mitochondrial ATP turnover, and supports cold- and adrenergic-mediated adaptive thermogenesis [162,163]. This requires us to further study the related thermogenic mechanism of thermogenic fat. As most of the conclusions are based on animal or in vitro studies, the specific mechanisms underlying the browning of WAT in humans need to be better elucidated. Enhanced BAT metabolic rate will require more oxygen and energy, which may increase the load on other organs of the body. In addition, although some drugs (CL 316,243, rosiglitazone, etc.) have shown efficacy in activating BAT and inducing browning of WAT, their use is not devoid of possible serious side effects. How to control BAT activation and browning of WAT, as well as a series of problems such as safety, efficiency and specificity are all unanswered questions that need to be addressed.

Traditional Chinese medicine has the characteristics of multi-component, multi-target and high safety. At present, some of the most promising browning agents (plant extracts, Chinese medicine) to activate BAT and induce browning of WAT, including resveratrol, berberine, and curcumin, have made great progress in the experimental stage. For example, resveratrol in Polygonum cuspidatum can enhance the rate of fat decomposition, mitochondrial heat production and UCP1 protein expression by activating the SIRT1 signalling pathway. In addition, resveratrol can also activate BAT and induce browning of WAT through gut microbiota-bile acid-TGR5/UCP1 pathway to ameliorate glucose homoeostasis. Berberine activates WAT and induces browning of WAT by activating AMPK signalling pathway. But their overall role in metabolism and other aspects of physiology needs further study. Therefore, it is imperative for scientists to discover safe and effective Chinese medicine with the function of activating BAT and/or induce browning of WAT to treat obesity and T2DM. With the development of mechanisms research of BAT activation and browning of WAT induced by safe and effective Chinese medicine, a new avenue for the treatment of obesity and T2DM will be built, and the prospects of Chinese medicine in the treatment of diabetes will be much brighter and broader.

Abbreviations

White adipose tissue (WAT), Brown adipose tissue (BAT), Beige adipocytes, Type 2 diabetes mellitus (T2DM), Type 1 diabetes mellitus (T1DM), Uncoupling protein 1 (UCP1), Peroxisome-proliferator-activated receptor γ coactivator-1α (PGC-1α), Peroxisome proliferator-activated receptors γ (PPAR γ), Positive regulatory domain-containing 16 (PRDM16), Fibronectin type III domain containing 5 (FNDC5), cAMP-dependent protein kinase A (PKA), p38 mitogen-activated protein kinase (p38 MAPK), Extracellular regulated protein kinases (ERK), Hypoxia-inducible factor 1, α subunit inhibitor (HIF1an), AMP-activated protein kinase (AMPK), Fibroblast growth factor 10 (FGF10), Fibroblast growth factor 21 (FGF21), CCAAT/enhancer-binding protein β (C/EBPβ), Insulin-like growth factor-1 (IGF-1), Insulin receptor substrate 1 (IRS1), Takeda G-protein coupled receptor 5 (TGR5), Norepinephrine (NE), Sirtuin1 (SIRT1), Bone morphogenesis proteins (BMPs), Thyroid hormones (THs), Intermittent fasting (IF), Caloric restriction (CR), Interleukin-6 (IL-6), MAPK kinase 6 (MKK6), β-adrenergic receptor agonists (β-ARs agonists), Thiazolidinediones (TZDs), Glucagon-like peptide-1 receptor agonists (GLP-1RAs).

Authors’ contributions

Long Cheng, Jingkang Wang, Quantao Ma, Yaqi Li, Pengfei Li, Yongcheng An, Hongyu Dai, Yuhui Duan, Lu Shi, Yinglan Lv, Huimin Li, Chen Wang, Haifeng Du and Baosheng Zhao participated in drafting, editing, and writing the manuscript. Long Cheng and Baosheng Zhao approved the final version of the manuscript. Long Cheng Quantao Ma, Yaqi Li and Baosheng Zhao designed the figures.

Disclosure statement

The authors declare that they have no competing interest.

References

[1] Afshin A, Forouzanfar MH, Reitsma MB, et al. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377(1):13–27. [Europe PMC free article] [Abstract] [Google Scholar]
[2] Kohlgruber A, Lynch L.. Adipose tissue inflammation in the pathogenesis of type 2 diabetes. Curr Diab Rep. 2015;15(11):92. [Abstract] [Google Scholar]
[3] Duclos M. Osteoarthritis, obesity and type 2 diabetes: the weight of waist circumference. Ann Phys Rehabil Med. 2016;59(3):157–160. [Abstract] [Google Scholar]
[4] International Diabetes Federation . IDF diabetes Atlas. 9th ed. Brussels, Belgium: International Diabetes Federation; 2019. [Google Scholar]
[5] Aherne W, Hull D. Brown adipose tissue and heat production in the newborn infant. J Pathol Bacteriol. 1966;91(1):223–234. [Abstract] [Google Scholar]
[6] Heaton J. The distribution of brown adipose tissue in the human. J Anat. 1972;112(Pt 1):35–39. [Europe PMC free article] [Abstract] [Google Scholar]
[7] Cypess AM, Lehman S, Williams G, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med. 2009;360(15):1509–1517. [Europe PMC free article] [Abstract] [Google Scholar]
[8] Virtanen KA, Lidell ME, Orava J, et al. Functional brown adipose tissue in healthy adults. N Engl J Med. 2009;360(15):1518–1525. [Abstract] [Google Scholar]
[9] Seale P, Bjork B, Yang WL, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature. 2008;454(7207):961–967. [Europe PMC free article] [Abstract] [Google Scholar]
[10] Sanchez-Gurmaches J, Hung CM, Sparks CA, et al. PTEN loss in the Myf5 lineage redistributes body fat and reveals subsets of white adipocytes that arise from Myf5 precursors. Cell Metab. 2012;16(3):348–362. [Europe PMC free article] [Abstract] [Google Scholar]
[11] Lidell ME, Betz MJ, Enerbäck S. Brown adipose tissue and its therapeutic potential. J Intern Med. 2014;276(4):364–377. [Abstract] [Google Scholar]
[12] Lim S, Honek J, Xue Y, et al. Cold-induced activation of brown adipose tissue and adipose angiogenesis in mice. Nat Protoc. 2012;7(3):606–615. [Abstract] [Google Scholar]
[13] Xiao C, Goldgof M, Gavrilova O, et al. Anti-obesity and metabolic efficacy of the β3-adrenergic agonist, CL316243, in mice at thermoneutrality compared to 22°C. Obesity. 2015;23(7):1450–1459. [Europe PMC free article] [Abstract] [Google Scholar]
[14] Wu J, Boström P, Sparks LM, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell. 2012;150(2):366–376. [Europe PMC free article] [Abstract] [Google Scholar]
[15] Kaisanlahti A, Glumoff T. Browning of white fat: agents and implications for beige adipose tissue to type 2 diabetes. J Physiol Biochem. 2019;75(1):1–10. [Europe PMC free article] [Abstract] [Google Scholar]
[16] Harms M, Seale P. Brown and beige fat: development, function and therapeutic potential. Nat Med. 2013;19(10):1252–1263. [Abstract] [Google Scholar]
[17] Singh AK, Aryal B, Chaube B, et al. Brown adipose tissue derived ANGPTL4 controls glucose and lipid metabolism and regulates thermogenesis. Mol Metab. 2018;11:59–69. [Europe PMC free article] [Abstract] [Google Scholar]
[18] Orava J, Nuutila P, Noponen T, et al. Blunted metabolic responses to cold and insulin stimulation in brown adipose tissue of obese humans. Obesity. 2013;21(11):2279–2287. [Abstract] [Google Scholar]
[19] Kubo N, Kawahara M, Okamatsu-Ogura Y, et al. Evaluation of glucose uptake and uncoupling protein 1 activity in adipose tissue of diabetic mice upon β-adrenergic stimulation. Mol Imaging Biol. 2019;21(2):249–256. [Abstract] [Google Scholar]
[20] Loyd C, Obici S. Brown fat fuel use and regulation of energy homeostasis. Curr Opin Clin Nutr Metab Care. 2014;17(4):368–372. [Abstract] [Google Scholar]
[21] Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84(1):277–359. [Abstract] [Google Scholar]
[22] Orava J, Nuutila P, Lidell ME, et al. Different metabolic responses of human brown adipose tissue to activation by cold and insulin. Cell Metab. 2011;14(2):272–279. [Abstract] [Google Scholar]
[23] Lee P, Greenfield JR, Ho KK, et al. A critical appraisal of the prevalence and metabolic significance of brown adipose tissue in adult humans. Am J Physiol Endocrinol Metab. 2010;299(4):E601–6. [Abstract] [Google Scholar]
[24] Wang X, Wang L, Sun Y, et al. DNA methylation and histone deacetylation regulating insulin sensitivity due to chronic cold exposure. Cryobiology. 2017;74:36–42. [Abstract] [Google Scholar]
[25] Villarroya F, Giralt M. The beneficial effects of brown fat transplantation: further evidence of an endocrine role of brown adipose tissue. Endocrinology. 2015;156(7):2368–2370. [Abstract] [Google Scholar]
[26] Blumenfeld NR, Kang HJ, Fenzl A, et al. A direct tissue-grafting approach to increasing endogenous brown fat. Sci Rep. 2018;8(1):7957. [Europe PMC free article] [Abstract] [Google Scholar]
[27] Guerra C, Navarro P, Valverde AM, et al. Brown adipose tissue-specific insulin receptor knockout shows diabetic phenotype without insulin resistance. J Clin Invest. 2001;108(8):1205–1213. [Europe PMC free article] [Abstract] [Google Scholar]
[28] Yao L, Cui X, Chen Q, et al. Cold-inducible SIRT6 regulates thermogenesis of brown and beige fat. Cell Rep. 2017;20(3):641–654. [Abstract] [Google Scholar]
[29] Jong JMAD, Wouters RTF, Boulet N, et al. The β3-adrenergic receptor is dispensable for browning of adipose tissues. AJP Endocrinology and Metabolism. 2017;312(6):E508–E518. [Abstract] [Google Scholar]
[30] Desjardins EM, Steinberg GR. Emerging role of AMPK in brown and beige adipose tissue (BAT): implications for obesity, insulin resistance, and type 2 diabetes. Curr Diab Rep. 2018;18(10):80. [Abstract] [Google Scholar]
[31] Rao R, Long J, White J, et al. Meteorin-like is a hormone that regulates immune-adipose interactions to increase beige fat thermogenesis. Cell. 2014;157(6):1279–1291. [Europe PMC free article] [Abstract] [Google Scholar]
[32] Richter EA, Hargreaves HM. Exercise, GLUT4, and skeletal muscle glucose uptake. Physiol Rev. 2013;93(3):993–1017. [Abstract] [Google Scholar]
[33] Wolfe RR, Klein S, Carraro F, et al. Role of triglyceride-fatty acid cycle in controlling fat metabolism in humans during and after exercise. Am J Physiol. 1990;258(2 Pt1):E382–E389. [Abstract] [Google Scholar]
[34] Cao L, Choi EY, Liu X, et al. White to brown fat phenotypic switch induced by genetic and environmental activation of a hypothalamic-adipocyte axis. Cell Metab. 2011;14(3):324–338. [Europe PMC free article] [Abstract] [Google Scholar]
[35] Boström P, Wu J, Jedrychowski MP, et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature. 2013;481(4):463–468. [Europe PMC free article] [Abstract] [Google Scholar]
[36] Vosselman MJ, Hoeks J, Brans B, et al. Low brown adipose tissue activity in endurance-trained compared with lean sedentary men. Int J Obes. 2015;39(12):1696–1702. [Abstract] [Google Scholar]
[37] Sutherland WHF, Woodhouse SP, Heyworth MR. Physical training and adipose tissue fatty acid composition in men. Metabolism. 1981;30(9):839–844. [Abstract] [Google Scholar]
[38] Lizcano F. The beige adipocyte as a therapy for metabolic diseases. Int J Mol Sci. 2019;20(20):5058. [Europe PMC free article] [Abstract] [Google Scholar]
[39] De Matteis R, Lucertini F, Guescini M, et al. Exercise as a new physiological stimulus for brown adipose tissue activity. Nutr Metab Cardiovasc Dis. 2013;23(6):582–590. [Abstract] [Google Scholar]
[40] Segawa M, Oh-Ishi S, Kizaki T, et al. Effect of running training on brown adipose tissue activity in rats: A reevaluation. Res Commun Mol Pathol Pharmacol. 1998;100(1):77–82. [Abstract] [Google Scholar]
[41] Stanford KI, Middelbeek RJ, Goodyear LJ. Exercise effects on white adipose tissue: beiging and metabolic adaptations. Diabetes. 2015;64(7):2361–2368. [Europe PMC free article] [Abstract] [Google Scholar]
[42] Collins S. β-adrenoceptor signaling networks in adipocytes for recruiting stored fat and energy expenditure. Front Endocrinol. 2012;2:102. [Europe PMC free article] [Abstract] [Google Scholar]
[43] Vitali A, Murano I, Zingaretti MC, et al. The adipose organ of obesity-prone C57BL/6J mice is composed of mixed white and brown adipocytes. J Lipid Res. 2012;53(4):619–629. [Europe PMC free article] [Abstract] [Google Scholar]
[44] Lass A, Zimmermann R, Oberer M, et al. Lipolysis-A highly regulated multi-enzyme complex mediates the catabolism of cellular fat stores. Prog Lipid Res. 2011;50(1):14–27. [Europe PMC free article] [Abstract] [Google Scholar]
[45] Bickel PE, Tansey JT, Welte MA. PAT proteins, an ancient family of lipid droplet proteins that regulate cellular lipid stores. Biochim Biophys Acta. 2009;1791(6):419–440. [Europe PMC free article] [Abstract] [Google Scholar]
[46] Villarroya F, Iglesias R, Giralt M. PPARs in the control of uncoupling proteins gene expression. PPAR Res. 2007;2007:74364. [Europe PMC free article] [Abstract] [Google Scholar]
[47] Park HS, Ju UI, Park JW, et al. PPAR γ neddylation essential for adipogenesis is a potential target for treating obesity. Cell Death Differ. 2016;23(8):1296–1311. [Europe PMC free article] [Abstract] [Google Scholar]
[48] Pu Y, Veiga-Lopez A. PPARγ agonist through the terminal differentiation phase is essential for adipogenic differentiation of fetal ovine preadipocytes. Cell Mol Biol Lett. 2017;22(1):6. [Europe PMC free article] [Abstract] [Google Scholar]
[49] Ohno H, Shinoda K, Spiegelman BM, et al. PPARγ agonists induce a white-to-brown fat conversion through stabilization of PRDM16 protein. Cell Metab. 2012;15(3):395–404. [Europe PMC free article] [Abstract] [Google Scholar]
[50] Park SS, Lee YJ, Kang H, et al. Lactobacillus amylovorus KU4 ameliorates diet-induced obesity in mice by promoting adipose browning through PPARγ signaling. Sci Rep. 2019;9(1):20152. [Europe PMC free article] [Abstract] [Google Scholar]
[51] Zhou Y, Yang J, Huang J, et al. The formation of brown adipose tissue induced by transgenic over-expression of PPAR γ2. Biochem Biophys Res Commun. 2014;446(2014):959–964. [Abstract] [Google Scholar]
[52] Jankovic A, Golic I, Markelic M, et al. Two key temporally distinguishable molecular and cellular components of white adipose tissue browning during cold acclimation. J Physiol. 2015;593(15):3267–3280. [Abstract] [Google Scholar]
[53] Castellani JW, Young AJ. Human physiological responses to cold exposure: acute responses and acclimatization to prolonged exposure. Auton Neurosci. 2016;196:63–74. [Abstract] [Google Scholar]
[54] Chondronikola M, Volpi E, Børsheim E, et al. Brown adipose tissue is linked to a distinct thermoregulatory response to mild cold in people. Front Physiol. 2016;7:129. [Europe PMC free article] [Abstract] [Google Scholar]
[55] Bartelt A, Bruns OT, Reimer R, et al. Brown adipose tissue activity controls triglyceride clearance. Nat Med. 2011;17(2):200–205. [Abstract] [Google Scholar]
[56] Nedergaard J, Bengtsson T, Cannon B. New powers of brown fat: fighting the metabolic syndrome. Cell Metab. 2011;13(3):238–240. [Abstract] [Google Scholar]
[57] Liang X, Pan J, Cao C, et al. Transcriptional response of subcutaneous white adipose tissue to acute cold exposure in mice. Int J Mol Sci. 2019;20(16):3968. [Europe PMC free article] [Abstract] [Google Scholar]
[58] Bordicchia M, Liu D, Amri EZ, et al. Cardiac natriuretic peptides act via p38 MAPK to induce the brown fat thermogenic program in mouse and human adipocytes. J Clin Invest. 2012;122(3):1022–1036. [Europe PMC free article] [Abstract] [Google Scholar]
[59] Liu X, Zheng Z, Zhu X, et al. Brown adipose tissue transplantation improves whole-body energy metabolism. Cell Res. 2013;23(6):851–854. [Europe PMC free article] [Abstract] [Google Scholar]
[60] Stanford KI, Middelbeek RJ, Townsend KL, et al. Brown adipose tissue regulates glucose homeostasis and insulin sensitivity. J Clin Invest. 2013;123(1):215–223. [Europe PMC free article] [Abstract] [Google Scholar]
[61] Liu X, Wang S, You Y, et al. Brown adipose tissue transplantation reverses obesity in Ob/Ob mice. Endocrinology. 2015;156(7):2461–2469. [Abstract] [Google Scholar]
[62] White JD, Dewal RS, Stanford KI. The beneficial effects of brown adipose tissue transplantation. Mol Aspects Med. 2019;68:74–81. [Europe PMC free article] [Abstract] [Google Scholar]
[63] Gunawardana SC, Piston DW. Reversal of type 1 diabetes in mice by brown adipose tissue transplant. Diabetes. 2012;61(3):674–682. [Europe PMC free article] [Abstract] [Google Scholar]
[64] Gunawardana SC, Piston DW. Insulin-independent reversal of type 1 diabetes in nonobese diabetic mice with brown adipose tissue transplant. Am J Physiol Endocrinol Metab. 2015;308(12):E1043–E55. [Europe PMC free article] [Abstract] [Google Scholar]
[65] Ridaura VK, Faith JJ, Rey FE, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341(6150):1241214. [Europe PMC free article] [Abstract] [Google Scholar]
[66] Koren O, Goodrich JK, Cullender TC, et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell. 2012;150(3):470–480. [Europe PMC free article] [Abstract] [Google Scholar]
[67] Moreno-Navarrete JM, Fernandez-Real JM. The gut microbiota modulates both browning of white adipose tissue and the activity of brown adipose tissue. Rev Endocr Metab Disord. 2019;20(4):387–397. [Abstract] [Google Scholar]
[68] Zhang CH, Liu JY, He XY, et al. Caulis spatholobi ameliorates obesity through activating brown adipose tissue and modulating the composition of gut microbiota. Int J Mol Sci. 2019;20(20):5150. [Europe PMC free article] [Abstract] [Google Scholar]
[69] Li B, Li L, Li M, et al. Microbiota depletion impairs thermogenesis of brown adipose tissue and browning of white adipose tissue. Cell Rep. 2019;26(10):2720–2737.e5. [Abstract] [Google Scholar]
[70] Krisko TI, Nicholls HT, Bare CJ, et al. Dissociation of adaptive thermogenesis from glucose homeostasis in microbiome-deficient mice. Cell Metab. 2020;31(3):592–604.e9. [Europe PMC free article] [Abstract] [Google Scholar]
[71] Hui S, Liu Y, Huang L, et al. Resveratrol enhances brown adipose tissue activity and white adipose tissue browning in part by regulating bile acid metabolism via gut microbiota remodeling. Int J Obes. 2020;44(8):1678–1690. [Abstract] [Google Scholar]
[72] Zhou LY, Xiao XH, Zhang Q, et al. Deciphering the anti-obesity benefits of resveratrol: the “gut microbiota-adipose tissue” axis. Front Endocrinol. 2019;10:413. [Europe PMC free article] [Abstract] [Google Scholar]
[73] Kim YH, Lee JH, Yeung JL, et al. Thermogenesis-independent metabolic benefits conferred by isocaloric intermittent fasting in ob/ob mice. Sci Rep. 2019;9(1):2479. [Europe PMC free article] [Abstract] [Google Scholar]
[74] Li G, Xie C, Lu S, et al. Intermittent fasting promotes white adipose browning and decreases obesity by shaping the gut microbiota. Cell Metab. 2017;26(4):672–685.e4. [Europe PMC free article] [Abstract] [Google Scholar]
[75] Sahuri-Arisoylu M, Brody LP, Parkinson JR, et al. Reprogramming of hepatic fat accumulation and browning of adipose tissue by the short-chain fatty acid acetate. Int J Obes. 2016;40(6):955–963. [Abstract] [Google Scholar]
[76] Petersen C, Nielsen MD, Andersen ES, et al. MCT1 and MCT4 expression and lactate flux activity increase during white and brown adipogenesis and impact adipocyte metabolism. Sci Rep. 2017;7(1):13101. [Europe PMC free article] [Abstract] [Google Scholar]
[77] Fabbiano S, Suárez-Zamorano N, Rigo D, et al. Caloric restriction leads to browning of white adipose tissue through type 2 immune signaling. Cell Metab. 2016;24(3):434–446. [Abstract] [Google Scholar]
[78] Qiang L, Wang L, Kon N, et al. Brown remodeling of white adipose tissue by SirT1-dependent deacetylation of Pparγ. Cell. 2012;150(3):620–632. [Europe PMC free article] [Abstract] [Google Scholar]
[79] Picard F, Kurtev M, Chung N, et al. Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-γ. Nature. 2004;429(6993):771–776. [Europe PMC free article] [Abstract] [Google Scholar]
[80] Shi T, Wang F, Stieren E, et al. SIRT3, a mitochondrial sirtuin deacetylase, regulates mitochondrial function and thermogenesis in brown adipocytes. J Biol Chem. 2005;280(14):13560–13567. [Abstract] [Google Scholar]
[81] Shuai L, Zhang L, Li B, et al. SIRT5 regulates brown adipocyte differentiation and browning of subcutaneous white adipose tissue. Diabetes. 2019;68(7):1449–1461. [Abstract] [Google Scholar]
[82] Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annu Rev Biochem. 2012;81(1):145–166. [Europe PMC free article] [Abstract] [Google Scholar]
[83] Shukla GC, Singh J, MicroRNAs: BS. Processing, maturation, target recognition and regulatory functions. Mol Cell Pharmacol. 2011;3(3):83–92. [Europe PMC free article] [Abstract] [Google Scholar]
[84] Lorente-Cebrián S, González-Muniesa P, Milagro FI, et al. MicroRNAs and other non-coding RNAs in adipose tissue and obesity: emerging roles as biomarkers and therapeutic targets. Clin Sci. 2019;133(1):23–40. [Abstract] [Google Scholar]
[85] Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–233. [Europe PMC free article] [Abstract] [Google Scholar]
[86] Zhang H, Guan M, Townsend KL, et al. MicroRNA-455 regulates brown adipogenesis via a novel HIF1an-AMPK-PGC1α signaling network. EMBO Rep. 2015;16(10):1378–1393. [Europe PMC free article] [Abstract] [Google Scholar]
[87] Ng R, Hussain NA, Zhang Q, et al. miRNA-32 drives brown fat thermogenesis and trans-activates subcutaneous white fat browning in mice. Cell Rep. 2017;19(6):1229–1246. [Europe PMC free article] [Abstract] [Google Scholar]
[88] Karbiener M, Pisani DF, Frontini A, et al. MicroRNA-26 family is required for human adipogenesis and drives characteristics of brown adipocytes. Stem Cells. 2014;32(6):1578–1590. [Abstract] [Google Scholar]
[89] Hu F, Wang M, Xiao T, et al. miR-30 promotes thermogenesis and the development of beige fat by targeting RIP140. Diabetes. 2015;64(6):2056–2068. [Europe PMC free article] [Abstract] [Google Scholar]
[90] Mori M, Nakagami H, Rodriguez-Araujo G, et al. Essential role for miR-196a in brown adipogenesis of white fat progenitor cells. PLoS Biol. 2012;10(4):e1001314. [Europe PMC free article] [Abstract] [Google Scholar]
[91] He L, Tang M, Xiao T, et al. Obesity associated miR-199a/214 cluster inhibits adipose browning via PRDM16-PGC-1α transcriptional network. Diabetes. 2018;67(12):2585–2600. [Abstract] [Google Scholar]
[92] Gaudet AD, Fonken LK, Gushchina LV, et al. miR-155 deletion in female mice prevents diet-induced obesity. Sci Rep. 2016;6(1):22862. [Europe PMC free article] [Abstract] [Google Scholar]
[93] Fischer C, Seki T, Lim S, et al. A miR-327-FGF10-FGFR2-mediated autocrine signaling mechanism controls white fat browning. Nat Commun. 2017;8(1):2079. [Europe PMC free article] [Abstract] [Google Scholar]
[94] Lv YF, Yu J, Sheng YL, et al. Glucocorticoids suppress the browning of adipose tissue via miR-19b in male mice. Endocrinology. 2018;159(1):310–322. [Abstract] [Google Scholar]
[95] Fu T, Seok S, Choi S, et al. MicroRNA 34a inhibits beige and brown fat formation in obesity in part by suppressing adipocyte fibroblast growth factor 21 signaling and SIRT1 function. Mol Cell Biol. 2014;34(22):4130–4142. [Europe PMC free article] [Abstract] [Google Scholar]
[96] Kim S, Park JW, Lee MG, et al. Reversine promotes browning of white adipocytes by suppressing miR-133a. J Cell Physiol. 2018;234(4):3800–3813. [Abstract] [Google Scholar]
[97] Hogan BL. Bone morphogenetic proteins: multifunctional regulators of vertebrate development. Genes Dev. 1996;10(13):1580–1594. [Abstract] [Google Scholar]
[98] Kang QW, Song WX, Luo Q, et al. A comprehensive analysis of the dual roles of BMPs in regulating adipogenic and osteogenic differentiation of mesenchymal progenitor cells. Stem Cells Dev. 2009;18(4):545–559. [Europe PMC free article] [Abstract] [Google Scholar]
[99] Qian SW, Tang Y, Li X, et al. BMP4-mediated brown fat-like changes in white adipose tissue alter glucose and energy homeostasis. Proc Natl Acad Sci U S A. 2013;110(9):E798–E807. [Europe PMC free article] [Abstract] [Google Scholar]
[100] Tseng YH, Kokkotou E, Schulz TJ, et al. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature. 2008;454(7207):1000–1004. [Europe PMC free article] [Abstract] [Google Scholar]
[101] Whittle AJ, Carobbio S, Martins L, et al. BMP8B increases brown adipose tissue thermogenesis through both central and peripheral actions. Cell. 2012;149(4):871–885. [Europe PMC free article] [Abstract] [Google Scholar]
[102] Castillo-Quan JI. From white to brown fat through the PGC-1α-dependent myokine irisin: implications for diabetes and obesity. Dis Model Mech. 2012;5(3):293–295. [Europe PMC free article] [Abstract] [Google Scholar]
[103] Mahajan RD, Patra SK. Irisin, a novel myokine responsible for exercise induced browning of white adipose tissue. Indian J Clin Biochem. 2013;28(1):102–103. [Europe PMC free article] [Abstract] [Google Scholar]
[104] Xiong X, Chen D, Sun H, et al. FNDC5 overexpression and irisin ameliorate glucose/lipid metabolic derangements and enhance lipolysis in obesity. Biochim Biophysica Acta. 2015;1852(9):1867–1875. [Abstract] [Google Scholar]
[105] Zhang Y, Li R, Meng Y, et al. Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling. Diabetes. 2014;63(2):514–525. [Abstract] [Google Scholar]
[106] Rodríguez A, Becerril S, Ezquerro S, et al. Cross-talk between adipokines and myokines in fat browning. Physiol. 2016;219(2):362–381. [Abstract] [Google Scholar]
[107] Lee P, Swarbrick MM, Greenfield JR. The sum of all browning in FGF21 therapeutics. Cell Metab. 2015;21(6):795–796. [Abstract] [Google Scholar]
[108] Struik D, Dommerholt MB, Jonker JW. Fibroblast growth factors in control of lipid metabolism: from biological function to clinical application. Curr Opin Lipidol. 2019;30(3):235–243. [Europe PMC free article] [Abstract] [Google Scholar]
[109] Kharitonenkov A, Shiyanova TL, Koester A, et al. FGF-21 as a novel metabolic regulator. J Clin Invest. 2005;115(6):1627–1635. [Europe PMC free article] [Abstract] [Google Scholar]
[110] Kolumam G, Chen MZ, Tong R, et al. Sustained brown fat stimulation and insulin sensitization by a humanized bispecific antibody agonist for fibroblast growth factor receptor 1/βKlotho complex. EBioMedicine. 2015;2(7):730–743. [Europe PMC free article] [Abstract] [Google Scholar]
[111] Fisher FM, Kleiner S, Douris N, et al. FGF21 regulates PGC-1α and browning of white adipose tissues in adaptive thermogenesis. Genes Deve. 2012;26(3):271–281. [Europe PMC free article] [Abstract] [Google Scholar]
[112] Vázquez P, Hérnandez-Sánchez C, Escalona-Garrido C, et al. Increased FGF21 in brown adipose tissue of tyrosine hydroxylase heterozygous mice: implications for cold adaptation. J Lipid Res. 2018;59(12):2308–2320. [Europe PMC free article] [Abstract] [Google Scholar]
[113] Hua L, Zhuo Y, Jiang D, et al. Identification of hepatic fibroblast growth factor 21 as a mediator in 17β-estradiol-induced white adipose tissue browning. Faseb J. 2018;32(10):5602–5611. [Abstract] [Google Scholar]
[114] Hondares E, Iglesias R, Giralt A, et al. Thermogenic activation induces FGF21 expression and release in brown adipose tissue. J Biol Chem. 2011;286(15):12983–12990. [Europe PMC free article] [Abstract] [Google Scholar]
[115] Fisher F, Maratos-Flier E. Understanding the physiology of FGF21. Annu Rev Physiol. 2016;78(1):223–241. [Abstract] [Google Scholar]
[116] Chau MD, Gao J, Yang Q, et al. Fibroblast growth factor 21 regulates energy metabolism by activating the AMPK-SIRT1-PGC-1α pathway. Proc Natl Acad Sci U S A. 2010;107(28):12553–12558. [Europe PMC free article] [Abstract] [Google Scholar]
[117] Cheng STW, Li SYT, Leung PS. Fibroblast growth factor 21 stimulates pancreatic islet autophagy via inhibition of AMPK-mTOR signaling. Int J Mol Sci. 2019;20(10):2517. [Europe PMC free article] [Abstract] [Google Scholar]
[118] Krause K. Novel aspects of white adipose tissue browning by thyroid hormones. Exp Clin Endocrinol Diabetes. 2019;7. DOI:10.1055/a-1020-5354 [Abstract] [CrossRef] [Google Scholar]
[119] Martínez-Sánchez N, Moreno-Navarrete JM, Contreras C, et al. Thyroid hormones induce browning of white fat. J Endocrinol. 2017;232(2):351–362. [Europe PMC free article] [Abstract] [Google Scholar]
[120] Weiner J, Kranz M, Klöting N, et al. Thyroid hormone status defines brown adipose tissue activity and browning of white adipose tissues in mice. Sci Rep. 2016;6(1):38124. [Europe PMC free article] [Abstract] [Google Scholar]
[121] Matesanz N, Bernardo E, Acín-Pérez R, et al. MKK6 controls T3-mediated browning of white adipose tissue. Nature Commun. 2017;8(1):856–869. [Europe PMC free article] [Abstract] [Google Scholar]
[122] Finan B, Clemmensen C, Zhu Z, et al. Chemical hybridization of glucagon and thyroid hormone optimizes therapeutic impact for metabolic disease. Cell. 2016;167(3):843–857.e14. [Abstract] [Google Scholar]
[123] Pal M, Febbraio MA, Whitham M. From cytokine to myokine: the emerging role of interleukin-6 in metabolic regulation. Immunol Cell Biol. 2014;92(4):331–339. [Abstract] [Google Scholar]
[124] Mohamed-Ali V, Goodrick S, Rawesh A, et al. Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-alpha, in vivo. J Clin Endocrinol Metab. 1997;82(12):4196–4200. [Abstract] [Google Scholar]
[125] Abdullahi A, Samadi O, Auger C, et al. Browning of white adipose tissue after a burn injury promotes hepatic steatosis and dysfunction. Cell Death Dis. 2019;10(12):870–883. [Europe PMC free article] [Abstract] [Google Scholar]
[126] Akbari M, Hassan-Zadeh VIL-6. IL-6 signalling pathways and the development of type 2 diabetes. Inflammopharmacology. 2018;26(3):685–698. [Abstract] [Google Scholar]
[127] Cottam DR, Mattar SG, Barinas-Mitchell E, et al. The chronic inflammatory hypothesis for the morbidity associated with morbid obesity: implications and effects of weight loss. Obes Surg. 2004;14(5):589–600. [Abstract] [Google Scholar]
[128] Egecioglu E, Anesten F, Schéle E, et al. Interleukin-6 is important for regulation of core body temperature during long-term cold exposure in mice. Biomed Rep. 2018;9(3):206–212. [Europe PMC free article] [Abstract] [Google Scholar]
[129] Han J, Meng Q, Shen L, et al. Interleukin-6 induces fat loss in cancer cachexia by promoting white adipose tissue lipolysis and browning. Lipids Health Dis. 2018;17(1):14–21. [Europe PMC free article] [Abstract] [Google Scholar]
[130] Patsouris D, Qi P, Abdullahi A, et al. Burn induces browning of the subcutaneous white adipose tissue in mice and humans. Cell Rep. 2015;13(8):1538–1544. [Europe PMC free article] [Abstract] [Google Scholar]
[131] Mathis D. Immunological goings-on in visceral adipose tissue. Cell Metab. 2013;17(6):851–859. [Europe PMC free article] [Abstract] [Google Scholar]
[132] Ricardo-Gonzalez RR, Eagle AR, Odegaard JI, et al. IL-4/STAT6 immune axis regulates peripheral nutrient metabolism and insulin sensitivity. Proc Natl Acad Sci U S A. 2010;107(52):22617–22622. [Europe PMC free article] [Abstract] [Google Scholar]
[133] Odegaard JI, Ricardo-Gonzalez RR, Eagle AR, et al. Alternative M2 activation of kupffer cells by PPARδ ameliorates obesity-induced insulin resistance. Cell Metab. 2008;7(6):496–507. [Europe PMC free article] [Abstract] [Google Scholar]
[134] Rao RR, Long JZ, White JP, et al. Meteorin-like is a hormone that regulates immune-adipose interactions to increase beige fat thermogenesis. Cell. 2014;157(6):1279–1291. [Europe PMC free article] [Abstract] [Google Scholar]
[135] Nguyen KD, Qiu Y, Cui X, et al. Alternatively activated macrophages produce catecholamines to sustain adaptive thermogenesis. Nature. 2011;480(7375):104–108. [Europe PMC free article] [Abstract] [Google Scholar]
[136] Abdullahi A, Auger C, Stanojcic M, et al. Alternatively activated macrophages drive browning of white adipose tissue in burns. Ann Surg. 2019;269(3):554–563. [Europe PMC free article] [Abstract] [Google Scholar]
[137] Ghorbani M, Himms-Hagen J. Appearance of brown adipocytes in white adipose tissue during CL 316,243-induced reversal of obesity and diabetes in Zucker fa/fa rats. Int J Obes Relat Metab Disord. 1997;21(6):465–475. [Abstract] [Google Scholar]
[138] Emilsson V, Summers RJ, Hamilton SH, et al. The effects of the β3-adrenoceptor agonist BRL 35135 on UCP isoform mRNA expression. Biochem Biophys Res Commun. 1998;252(2):450–454. [Abstract] [Google Scholar]
[139] Park JW, Jung KH, Lee JH, et al. 18F-FDG PET/CT monitoring of β3 agonist-stimulated brown adipocyte recruitment in white adipose tissue. J Nucl Med. 2014;56(1):153–158. [Abstract] [Google Scholar]
[140] Cypess A, Weiner L, Roberts-Toler C, et al. Activation of human brown adipose tissue by a β3-adrenergic receptor agonist. Cell Metab. 2015;21(1):33–38. [Europe PMC free article] [Abstract] [Google Scholar]
[141] Larsen TM, Toubro S, van Baak MA, et al. Effect of a 28-d treatment with L-796568, a novel beta (3)-adrenergic receptor agonist, on energy expenditure and body composition in obese men. Am J Clin Nutr. 2002;76(4):780–788. [Abstract] [Google Scholar]
[142] Taylor D, Gottlieb RA. Parkin-mediated mitophagy is downregulated in browning of white adipose tissue. Obesity. 2017;25(4):704–712. [Europe PMC free article] [Abstract] [Google Scholar]
[143] Merlin J, Sato M, Nowell C, et al. The PPARγ agonist rosiglitazone promotes the induction of brite adipocytes, increasing β-adrenoceptor-mediated mitochondrial function and glucose uptake. Cell Signal. 2018;42:54–66. [Abstract] [Google Scholar]
[144] Zhao L, Zhu C, Lu M, et al. The key role of a glucagon-like peptide-1 receptor agonist in body fat redistribution. J Endocrinol. 2019;240(2):271–286. [Abstract] [Google Scholar]
[145] Bonet ML, Ribot J, Palou A. Lipid metabolism in mammalian tissues and its control by retinoic acid. Biochim Biophys Acta. 2012;1821(1):177–189. [Abstract] [Google Scholar]
[146] Quan L, Zhang C, Dong M, et al. Myristoleic acid produced by enterococci reduces obesity through brown adipose tissue activation. Gut. 2019;69(7):19. [Abstract] [Google Scholar]
[147] Kim YJ, Ryu R, Choi JY, et al. Platycodon grandiflorus root ethanol extract induces lipid excretion, lipolysis, and thermogenesis in diet-induced obese mice. J Med Food. 2019;22(11):1100–1109. [Abstract] [Google Scholar]
[148] Kim SH, Shin EJ, Kim ED, et al. Berberine activates GLUT1-mediated glucose uptake in 3T3-L1 adipocytes. Biol Pharm Bull. 2007;30(11):2120–2125. [Abstract] [Google Scholar]
[149] Hu X, Zhang Y, Xue Y, et al. Berberine is a potential therapeutic agent for metabolic syndrome via brown adipose tissue activation and metabolism regulation. Am J Transl Res. 2018;10(11):3322–3329. [Europe PMC free article] [Abstract] [Google Scholar]
[150] Zhang Z, Zhang H, Li B, et al. Berberine activates thermogenesis in white and brown adipose tissue. Nat Commun. 2014;5(5):5493–5507. [Abstract] [Google Scholar]
[151] Liao W, Yin X, Li Q, et al. Resveratrol-induced white adipose tissue browning in obese mice by remodeling fecal microbiota. Molecules. 2018;23(12):3356. [Europe PMC free article] [Abstract] [Google Scholar]
[152] Han Y, Wu JZ, Shen JZ, et al. Pentamethylquercetin induces adipose browning and exerts beneficial effects in 3T3-L1 adipocytes and high-fat diet-fed mice. Sci Rep. 2017;7(1):1123. [Europe PMC free article] [Abstract] [Google Scholar]
[153] Wang S, Wang X, Ye Z, et al. Curcumin promotes browning of white adipose tissue in a norepinephrine-dependent way. Biochem Biophys Res Commun. 2015;466(2):247–253. [Abstract] [Google Scholar]
[154] Guan L, Gong D, Yang S, et al. Genipin ameliorates diet-induced obesity via promoting lipid mobilization and browning of white adipose tissue in rats. Phytother Res. 2018;32(4):723–732. [Abstract] [Google Scholar]
[155] Seoane-Collazo P, Liñares-Pose L, Rial-Pensado E, et al. Central nicotine induces browning through hypothalamic κ opioid receptor. Nat Commun. 2019;10(1):4037. [Europe PMC free article] [Abstract] [Google Scholar]
[156] Sakellariou P, Valente A, Carrillo AE, et al. Chronic l-menthol-induced browning of white adipose tissue hypothesis: A putative therapeutic regime for combating obesity and improving metabolic health. Med Hypotheses. 2016;93:21–26. [Abstract] [Google Scholar]
[157] Seo YJ, Jin H, Lee K, et al. Cardamonin suppresses lipogenesis by activating protein kinase a-mediated browning of 3T3-L1 cells. Phytomedicine. 2019;65:153064. [Abstract] [Google Scholar]
[158] Lee CG, Rhee DW, Kim BO, et al. Allicin induces beige-like adipocytes via KLF15 signal cascade. J Nutr Biochem. 2019;64:13–24. [Abstract] [Google Scholar]
[159] Yoneshiro T, Saito M. Transient receptor potential activated brown fat thermogenesis as a target of food ingredients for obesity management. Curr Opin Clin Nutr Metab Care. 2013;16(6):625–631. [Abstract] [Google Scholar]
[160] Anunciado-Koza R, Ukropec J, Koza RA, et al. Inactivation of UCP1 and the glycerol phosphate cycle synergistically increases energy expenditure to resist diet-induced obesity. J Biol Chem. 2008;283(41):27688–27697. [Europe PMC free article] [Abstract] [Google Scholar]
[161] Bertholet AM, Kazak L, Chouchani ET, et al. Mitochondrial patch clamp of beige adipocytes reveals UCP1-positive and UCP1-negative cells both exhibiting futile creatine cycling. Cell Metab. 2017;25(4):811–822.e4. [Europe PMC free article] [Abstract] [Google Scholar]
[162] Kazak L, Chouchani ET, Lu GZ, et al. Genetic depletion of adipocyte creatine metabolism inhibits diet-induced thermogenesis and drives obesity. Cell Metab. 2017;26(4):660–671. [Europe PMC free article] [Abstract] [Google Scholar]
[163] Kazak L, Rahbani JF, Samborska B, et al. Ablation of adipocyte creatine transport impairs thermogenesis and causes diet-induced obesity. Nat Metab. 2019;1(3):360–370. [Europe PMC free article] [Abstract] [Google Scholar]

Articles from Adipocyte are provided here courtesy of Taylor & Francis

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/97259098
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/97259098

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1080/21623945.2020.1870060

Supporting
Mentioning
Contrasting
0
80
0

Article citations


Go to all (104) article citations

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

National Natural Science Foundation

    National Natural Science Foundation of China (2)