BAT (brown adipose tissue) is specialized to burn fatty acids for heat generation and energy expenditure to defend against cold and obesity. Accumulating studies have demonstrated that manipulation of BAT activity through various strategies can regulate metabolic homoeostasis and lead to a healthy phenotype. Two classes of ncRNA (non-coding RNA), miRNA and lncRNA (long non-coding RNA), play crucial roles in gene regulation during tissue development and remodelling. In the present review, we summarize recent findings on regulatory role of distinct ncRNAs in brown/beige adipocytes, and discuss how these ncRNA regulatory networks contribute to brown/beige fat development, differentiation and function. We suggest that targeting ncRNAs could be an attractive approach to enhance BAT activity for protecting the body against obesity and its pathological consequences.

BAT (brown adipose tissue), a specialized mammalian organ, is distinguished from WAT (white adipose tissue) by its location, morphology and physiological function [1]. BAT promotes energy expenditure via uncoupled respiration mediated by mitochondrial UCP1 (uncoupling protein 1) [2]. Another type of adipocyte expressing UCP1 is referred as beige or brite (brown in white) adipocyte and regarded as a non-classical/inducible brown adipocyte [3,4]. Ablation of BAT can induce obesity and metabolic diseases [5]; in contrast, enhancing BAT activity and function through different strategies leads to increased energy expenditure against obesity [6]. Therefore manipulation of BAT mass and function is an attractive therapeutic approach for obesity-associated metabolic syndrome and its complications.

There has been an upsurge of interest in understanding the mechanism underlying brown/beige adipogenesis. The master regulator of brown fat development is PRDM16 (PR domain-containing 16) which specifies the brown fat lineage from white fat and muscle [7]. It drives brown adipocyte development by binding and activating the transcriptional cofactors PGC-1α [PPAR (peroxisome-proliferator-activated receptor) γ co-activator 1α], PGC-1β, PPARα and PPARγ to induce brown-fat-selective genes, and by interacting with transcriptional co-repressors such as CtBP1 (C-terminal binding protein 1) and CtBP2 to repress white-fat-cell-selective genes [8,9]. In addition, a variety of extrinsic or intrinsic protein factors have been identified as regulators for BAT adipogenesis and physiological function (Figure 1). These studies have been reviewed in other excellent articles [6,1013] and so we will not discuss these extensively in the present review.

Key protein/ncRNA regulatory networks regulate brown/beige adipogenesis

Figure 1
Key protein/ncRNA regulatory networks regulate brown/beige adipogenesis

Brown and beige adipocytes originate from distinct progenitors, and are differently influenced either by extrinsic factors indirectly regulating key transcription factors (such as PRDM16 and PPARγ), or through mobilizing intrinsic factors (such as ncRNA). BAT executes its metabolic function mainly by fuel oxidation and UCP1-mediated thermogenesis to defend against cold and obesity. For detailed descriptions of these proteins and ncRNAs, see the text. BMP, bone morphogenetic protein; NE, noradrenaline (norepinephrine); RBP, RNA-binding protein; T3, 3,3′,5-tri-iodothyroninel; T4, thyroxine; TFx, thyroid receptor; VEGF, vascular endothelial growth factor.

Figure 1
Key protein/ncRNA regulatory networks regulate brown/beige adipogenesis

Brown and beige adipocytes originate from distinct progenitors, and are differently influenced either by extrinsic factors indirectly regulating key transcription factors (such as PRDM16 and PPARγ), or through mobilizing intrinsic factors (such as ncRNA). BAT executes its metabolic function mainly by fuel oxidation and UCP1-mediated thermogenesis to defend against cold and obesity. For detailed descriptions of these proteins and ncRNAs, see the text. BMP, bone morphogenetic protein; NE, noradrenaline (norepinephrine); RBP, RNA-binding protein; T3, 3,3′,5-tri-iodothyroninel; T4, thyroxine; TFx, thyroid receptor; VEGF, vascular endothelial growth factor.

Close modal

In the present review, we focus on the function of ncRNA (non-coding RNA) in brown/beige fat development and consider two distinct subsets of ncRNAs: the 20–24-nt miRNA, and the lncRNA (long non-coding RNA), which is arbitrarily classified as RNA longer than 200 nt [14]. miRNA is evolutionarily conserved and recognizes its target mRNA by imperfect pairing between its seed sequence and mRNA. Paired miRNA–mRNA together with Argonaute protein form a RNA–protein complex, miRISC (miRNA-induced silencing complex), in which miRNA induces target mRNA translational repression and/or AGO2 (Argonaute 2)-mediated mRNA cleavage [15,16]. In contrast, lncRNA has poor sequence conservation, and through its primary, secondary and tertiary structures, interacts with RBPs (RNA-binding proteins) to regulate gene expression at transcriptional or post-transcriptional levels [14,17]. Transcriptome studies have revealed distinctive expression patterns of ncRNA in different adipose depots, a dynamic change in ncRNA expression during BAT and WAT adipogenesis and functional importance of these ncRNAs in these biological processes [14,1820]. In the present review, we aim to cover the recent advances in understanding the role of ncRNAs in brown/beige adipocyte biology (Table 1 and Figure 1) and point out that manipulating ncRNAs could be a novel therapeutic approach for combating metabolic diseases. Because of limitations of space, it is impossible to cover all articles published in this rapid growing field, so we only focus on a few seminal pioneering studies and some recent studies with physiological evidence. We apologize for many excellent studies that are not discussed in the present review.

Table 1
miRNAs that regulate brown/beige fat cell adipogenesis and physiological function
miRNAExpression profileFunctionPhenotype of miRNA transgenic or knockout mouse
miR-193b-365 BAT, WAT, other Promotes brown adipogenesis No transgenic mice 
miR-133 Muscle, BAT, SAT Inhibits brown/beige adipogenesis Knockout mice: SAT browning, increased insulin sensitivity and glucose tolerance 
miR-196a BAT, SAT Promotes beige adipogenesis Transgenic mice: enhanced energy expenditure, resistance to obesity and the metabolic syndrome 
miR-155 BAT, WAT Inhibits brown/beige adipogenesis Transgenic mice: reduced BAT mass; knockout mice: increased ability to adapt to cold exposure 
miR-27 BAT, WAT Inhibits brown/beige adipogenesis No transgenic mice 
miR-378 Muscle, BAT, WAT Promotes brown adipogenesis Transgenic mice: increased BAT mass, prevent both genetic and HFD-induced obesity 
miR-34a Liver, BAT, WAT Inhibits brown/beige adipogenesis No transgenic mice 
miRNAExpression profileFunctionPhenotype of miRNA transgenic or knockout mouse
miR-193b-365 BAT, WAT, other Promotes brown adipogenesis No transgenic mice 
miR-133 Muscle, BAT, SAT Inhibits brown/beige adipogenesis Knockout mice: SAT browning, increased insulin sensitivity and glucose tolerance 
miR-196a BAT, SAT Promotes beige adipogenesis Transgenic mice: enhanced energy expenditure, resistance to obesity and the metabolic syndrome 
miR-155 BAT, WAT Inhibits brown/beige adipogenesis Transgenic mice: reduced BAT mass; knockout mice: increased ability to adapt to cold exposure 
miR-27 BAT, WAT Inhibits brown/beige adipogenesis No transgenic mice 
miR-378 Muscle, BAT, WAT Promotes brown adipogenesis Transgenic mice: increased BAT mass, prevent both genetic and HFD-induced obesity 
miR-34a Liver, BAT, WAT Inhibits brown/beige adipogenesis No transgenic mice 

DGCR8 or Dicer adipose-knockout impairs the function of BAT

To determine the role of miRNA, as a whole, in adipose tissue, several mouse strains of adipose-specific knockout of DGCR8 (DiGeorge syndrome critical region 8) or Dicer, two key regulators of miRNA biogenesis pathways, have been generated [18,21]; in addition, a tamoxifen-inducible Dicer-knockout strain has also been generated [21]. Both DGCR8- and Dicer-knockout mice display enlarged but pale interscapular brown fat with reduced BAT marker expression, accompanied by an impairment of insulin sensitivity. Although deletion of DGCR8 or Dicer results in different defects in WAT, both of them impair WAT browning upon exposure to cold [18,21]. These results demonstrate that miRNA plays a critical role in BAT development, WAT browning and brown/beige fat physiology.

It is interesting to introduce key miRNAs into DGCR8- or Dicer-knockout brown adipocytes and determine their sole contribution to brown adipocyte biology. Because global miRNA biogenesis has been blocked in DGCR8- or Dicer-knockout mice, these strains provide a clean background to identify the core set of miRNAs that are sufficient to support brown fat development. It has been reported that, in the Dicer-knockout brown adipocytes, reintroduction of miR-346 and miR-362 partially reverses the decrease in C/EBPβ (CCAAT/enhancer-binding protein β), whereas re-expression of miR-365 increases UCP1 mRNA expression 20-fold [21]. Another advantage of these platforms is to facilitate the identification of miRNA targets. Because mRNA targets are normally repressed by multiple miRNAs, the effect of a single miRNA is often masked by other miRNAs targeting the same mRNA. However, in the absence of other miRNAs, the effect of a given miRNA will be more evident when it is introduced back to DGCR8- or Dicer-knockout cells.

Critical functional miRNAs in brown/beige adipocytes

miRNAs execute their regulatory function by participating in signalling networks and fine-tuning or switching signalling output levels [22]. Recent studies have identified several miRNAs functioning in different stages of brown/beige fat adipogenesis [23]; these regulatory networks are summarized as follows (Figure 2).

Key miRNA regulatory networks in brown/beige adipogenesis

Figure 2
Key miRNA regulatory networks in brown/beige adipogenesis

(A) Under the control of PRDM16, miR-193b-365 promotes brown adipogenesis by targeting Runx1t1, Cdon and Igfbp5. (B) miR-133, transcriptionally controlled by MEF2, serves as a major negative regulator of brown/beige adipogenesis by directly targeting PRDM16. (C) miR-196a targets Hoxc8 to up-regulate C/EBPβ for beige adipogenesis. HDAC3, histone deacetylase 3. (D) miR-155 forms a bistable feedback loop with its target C/EBPβ and maintains the balance between proliferation and differentiation of brown pre-adipocytes. TGFR, TGF receptor; (E) miR-378 promotes BAT differentiation and expansion partly through targeting Pde1b to elevate cAMP content. (F) miR-27 targets several key brown transcription factors in response to both glucocorticoid (GC) signal and adrenergic-stimuli-induced brown/beige adipogenesis. CREB, cAMP-response-element-binding protein; GCR, glucocorticoid receptor. (G) miR-34a exerts roles in multiple tissues through targeting FGF signalling to up-regulate thermogenic genes such as those encoding PGC-1α and UCP1. Ac, acetyl; βKL, β-Klotho; SIRT1, sirtuin 1; TFx, thyroid receptor.

Figure 2
Key miRNA regulatory networks in brown/beige adipogenesis

(A) Under the control of PRDM16, miR-193b-365 promotes brown adipogenesis by targeting Runx1t1, Cdon and Igfbp5. (B) miR-133, transcriptionally controlled by MEF2, serves as a major negative regulator of brown/beige adipogenesis by directly targeting PRDM16. (C) miR-196a targets Hoxc8 to up-regulate C/EBPβ for beige adipogenesis. HDAC3, histone deacetylase 3. (D) miR-155 forms a bistable feedback loop with its target C/EBPβ and maintains the balance between proliferation and differentiation of brown pre-adipocytes. TGFR, TGF receptor; (E) miR-378 promotes BAT differentiation and expansion partly through targeting Pde1b to elevate cAMP content. (F) miR-27 targets several key brown transcription factors in response to both glucocorticoid (GC) signal and adrenergic-stimuli-induced brown/beige adipogenesis. CREB, cAMP-response-element-binding protein; GCR, glucocorticoid receptor. (G) miR-34a exerts roles in multiple tissues through targeting FGF signalling to up-regulate thermogenic genes such as those encoding PGC-1α and UCP1. Ac, acetyl; βKL, β-Klotho; SIRT1, sirtuin 1; TFx, thyroid receptor.

Close modal

miR-193b-365

Using microarray methods, researchers have identified a list of miRNAs enriched in BAT compared with WAT, among which miR-193b and miR-365 are a bicistronic transcript [24]. The level of miR-193b-365 is significantly up-regulated during brown adipogenesis. This miRNA cluster is needed for lineage-specific brown adipogenesis, as established by both overexpression and inhibition experiments. Blocking miR-193b-365 causes a remarkable reduction in lipid accumulation during adipogenesis, whereas ectopic expression of miR-193b decreases mRNA levels of myogenic markers on C2C12 myoblasts and up-regulates mRNA levels of two master adipogenic factors, PPARγ and C/EBPα, thus inducing a transition of myoblasts to brown adipocytes under adipogenic differentiation conditions. miR-193b executes these function at least by directly repressing Cdon (cell adhesion-associated, oncogene-regulated), Igfbp5 (insulin-like growth factor-binding protein 5) (two pro-myogenic factors) and Runx1t1 (runt-related transcription factor 1 translocated to 1) (an inhibitor of brown adipogenesis) [25]. This regulatory network is initiated by the brown lineage determination factor PRDM16, as PRDM16 induces miR-193b-365 expression at least partially by inducing expression of its transcription factor PPARα. Therefore miR-193b-365 represents a new part of the PRDM16-induced programme. However, blocking miR-193b-365 with miRNA inhibitors in mature brown adipocytes does not affect the expression of key brown adipocyte genes, indicating that this gene cluster carries out its primary function during development. Screening for miRNAs essential for maintenance of BAT features requires further investigation [24].

miR-133

On the basis of profiling analysis of miRNAs in BAT before and after cold exposure, RNA sequencing for differentially expressed miRNAs between satellite cells and brown pre-adipocytes, and through bioinformatic analysis predicting PRDM16 targets, several groups have successively revealed miR-133 as a critical regulator of adipocyte browning [2628].

miR-133 belongs to a group of myomiRs (muscle-specific miRNAs) and is a potent repressor of non-muscle genes, thereby playing a role in embryonic stem cell differentiation and muscle specification [29,30]. miR-133 serves as a major negative regulator of brown/beige adipogenesis. It has a distinct expression pattern among myogenic and brown adipogenic lineages, and their progenitors. The level of miR-133 is down-regulated during the induction of brown adipocyte differentiation and in BAT after cold exposure [26,27]. The down-regulation of miR-133 is inversely correlated with PRDM16 and UCP1 expression levels. Functional assays for miRNA target identification reveal that both miR-133a and miR-133b directly target the conserved 8-nt seed sequences in the 3′UTR of PRMD16 mRNA [2628]. Moreover, overexpression of miR-133 in brown pre-adipocytes dramatically decreases the mRNA level of PRDM16, as well as other brown adipogenic markers and general adipogenic markers [27], but increases myogenic markers [26]. By contrast, inhibition of miR-133 causes an increase in BAT transcription factors PPARγ and PPARα, and a decrease in myogenic markers [26]. Therefore the brown lineage commitment is influenced directly by miR-133 through targeting PRDM16. miR-133 is regulated directly by MEF2 (myocyte enhancer factor 2) during adrenergic-stimuli-induced BAT development and differentiation. Adrenergic stimulation upon cold exposure leads to a decrease in MEF2, resulting in a reduced expression of miR-133 and an increased expression of PRDM16, which facilitates brown lineage commitment [26].

Inhibition of miR-133 during BAT differentiation causes enhanced mitochondrial activity and BAT programme, leading to an increase in the basal oxygen consumption rate [26]. Similar modulations are observed in the progression of frozen injury-induced muscle regeneration in which miR-133 antagonism induces an increase in uncoupled respiration, glucose uptake and thermogenesis in intramuscular brown adipocytes [27]. The bioenergetic effects of miR-133 antagonism on muscle cells make it a critical modulator of whole-body energy expenditure: antagonism of miR-133 in mice fed with either a normal diet or an HFD (high-fat diet) display a significant increase in total energy expenditure, without much change in physical activity and food intake. Moreover, antagonism of miR-133 leads to improved glucose tolerance and decreased infiltration of inflammatory cells in eWAT (epididymal WAT) [27]. Consistent with this, knockout of miR-133 in mice leads to SAT (subcutaneous adipose tissue) browning and increased insulin sensitivity [28]. All of these studies indicate that miR-133 can serve as a therapeutic target to defend against diet-induced obesity.

miR-196a

miR-196a expression is induced in SVF (stromal-vascular fraction) cells during brown adipogenesis by an adenylate cyclase activator, forskolin, or cold exposure [31]. The miR-196a gene is located within the gene clusters encoding the Hox (homeobox) family, which plays a role in system differentiation including adipogenesis [31,32]. One of the Hox genes, Hoxc8, is categorized as a white-fat gene and a repressor of brown adipogenesis [33]. Hoxc8 targets the C/EBPβ regulatory sequence and represses C/EBPβ, a positive regulator of the BAT programme. During the brown adipogenesis of white fat progenitors, the expression of Hoxc8 is suppressed directly by miR-196a, which facilitates the browning progression. In iBAT (interscapular BAT)-SVF cells, miR-196a is barely expressed, and its target Hoxc8 is even absent, which implies that miR-196a does not play a role in conventional brown adipogenesis [31]. Fat-specific miR-196a transgenic mice generate metabolically functional beige cells in WAT, and exhibit enhanced energy expenditure, improved glucose metabolism and resistance to obesity. Therefore manipulation of miR-196a–Hoxc8–C/EBPβ signalling in WAT progenitors to initiate browning can be a therapeutic strategy for obesity-associated metabolic consequences.

miR-155

TGFβ1 (transforming growth factor β1) potently inhibits adipogenesis, and knockout of Smad3 induces the WAT to BAT phenotypic transition [34,35]. A recent study demonstrated that the inhibitory effect of TGFβ1/Smad signalling on brown fat adiposeness is partly exerted through induction of miR-155 [36]. miR-155 is highly expressed in pre-adipocytes, but decreases dramatically after induction of differentiation. It forms a bistable feedback loop with its target C/EBPβ to maintain the balance between proliferation and differentiation of brown pre-adipocytes. miR-155 transgenic mice display reduced BAT mass, whereas miR-155-knockout mice show a better adaption to cold exposure by recruiting brite fat cells (inducible beige cells) and enhancing differentiation of BAT [36]. These data demonstrate that miR-155 serves as a negative regulator for brown fat differentiation.

miR-27

A single miRNA family can regulate a large number of targets, some of which may be involved in the same biological process or signalling pathway [16,23]. miR-27a/miR-27b is such a miRNA family, which serves as a upstream regulator of brown/beige adipogenesis by directly targeting several key transcription factors such as PRDM16, PPARα, CREB (cAMP-response-element-binding protein) and PGC-1β [37]. Moreover, antagonizing miR-27b function prevents dexamethasone (one type of glucocorticoid) induced inhibition of BAT differentiation in primary adipocytes, and improves glucocorticoid treatment-induced central fat accumulation and metabolic dysfunction in mice. Glucocorticoid induces the expression of miR-27b through glucocorticoid receptor-mediated transcriptional regulation [38]. Therefore miR-27 is a responder of glucocorticoid signal, as well as a central regulator of brown/beige adipogenesis.

miR-378

miR-378 resides in the gene locus of PGC-1β and is induced during brown adipogenesis. miR-378 promotes BAT differentiation and expansion, but suppresses the formation of beige adipocytes in subcutaneous WAT. miR-378 directly targets Pde1b (phosphodiesterase 1b) that catalyses the turnover of cAMP [39]. This may be one of the regulators in BAT activity and expansion, as single nucleotide polymorphisms in the Pde1b gene are associated with cattle back-fat thickness, and cAMP can potentiate adipocyte differentiation [40,41]. Indeed, miR-378 transgenic mice display elevated cAMP levels which leads to increased lipolysis and activity in BAT, accompanied with enlarged organ mass and improved metabolic profile.

miR-34a

Upon cold exposure, 3T3-L1 fat cells display a marked decrease in miR-34a and an increase in browning-related genes, such as those encoding UCP1, PGC-1α and PRDM16. This inverse correlation is also observed in adipose tissue, which suggests that miR-34a inhibits fat browning [42]. Mechanistically, miR-34a directly targets FGFR1 (fibroblast growth factor receptor 1) to attenuate FGF21 (fibroblast growth factor 21)/SIRT1 (sirtuin 1)-dependent deacetylation of PGC-1α, which controls the browning transcriptional programme [42,43]. Circulating FGF21, an endocrine mitokine, enhances fatty acid β-oxidation in liver and adipose tissue, induces WAT browning, and promotes protection from diet-induced obesity and insulin resistance [44]. The integrated actions from multiple tissues induced by FGF21 signalling makes its upstream miR-34a a potent modulator in metabolic regulatory networks. Importantly, beneficial effects mediated by inhibition of miR-34a in HFD-fed mice include several aspects, including reduced adiposity and improved serum profiles [42]; miR-34a inhibition induces browning in all types of WAT, and promotes a more robust BAT programme [42]. These distinctive effects imply that miR-34a exerts roles in multiple tissues through targeting FGF signalling.

The function of lncRNAs in BAT is poorly understood despite existing reports on their expression pattern in BAT [45,46]. Using massively parallel sequencing of polyadenylation-selected RNAs, researchers have identified 175 lncRNAs which are regulated during adipogenesis. Further RNAi-mediated loss-of-function experiments have characterized ten lncRNAs that are required for adipogenesis and are referred to as lnc-RAP-n (lncRNA Regulated in AdiPogenesis) [47]. One of them, lnc-RAP-1 (or Firre, functional intergenic repeating RNA element) interacts with the nuclear matrix factor hnRNPU (heterogeneous nuclear ribonucleoprotein U) to mediate trans-chromosomal interactions, and brings loci encoding known regulators of adipogenesis into close proximity. This hnRNPU–Firre-mediated interaction may be required for proper function of multiple biological processes including adipogenesis [48].

This study is mainly based on the WAT system, but the role of lncRNA in brown adipocyte differentiation is not well understood. A recent research has identified Blnc1 (brown fat lncRNA 1) as a BAT-enriched lncRNA, which promotes brown and beige adipocyte differentiation through a ribonucleoprotein complex formed with EBF2 (early B-cell factor 2) to enhance expression of thermogenic genes such as UCP1. This regulation pattern is feedforward because Blnc1 itself is positively regulated by EBF2 [49].

These previous studies suffer from an incomplete annotation of lncRNA in adipose tissue. To address this obstacle, our group performed deep RNA-seq from BAT, inguinal WAT and eWAT, which provides us with approximately half a billion of reads for transcriptome reconstruction. We developed a computational pipeline and identified more than 1500 lncRNAs expressed in adipose tissue. Of these, 127 are BAT-restricted, and many of them are targeted by key adipogenic regulators such as PPARγ and C/EBPα [50]. Since they are BAT-enriched, we refer to them as lnc-BATEs. lnc-BATE1 is up-regulated 30-fold during adipogenesis and is required for brown adipogenesis and the maintenance of brown adipocyte features. lnc-BATE1 specifically binds the nuclear matrix factor hnRNPU; both of them are required for proper brown adipogenesis [48,50], but how this ribonucleotide complex exerts its functions is not well understood and requires further investigation.

Manipulation of BAT and WAT browning has been linked to a healthy metabolic phenotype and a defence against obesity and the metabolic syndrome. Several recent studies have identified new BAT-specific lncRNAs and revealed that some of them, such as lnc-BATE1 and Blnc1, promote brown and/or beige adipocyte differentiation and physiological function (Figure 3). Although the detailed regulatory networks and mechanisms are still not fully understood, lncRNAs are promising therapeutic targets for obesity and associated metabolic diseases.

lncRNAs in BAT adipogenesis

Figure 3
lncRNAs in BAT adipogenesis

The research route for exploiting lncRNAs in BAT adipogenesis contains four steps: 1, transcriptional profiling; 2, verifying lncRNAs; 3, functional study; and 4, mechanism exploring. Two examples for regulatory mechanisms of lncRNAs in BAT are presented (upper panel: Firre–hnRNPU interplay-mediated trans-chromosomal interactions promotes adipogenesis; lower panel: Blnc1 cis-regulates EBF2, and Blnc1–EBF2 forms a feedforward loop to promote brown/beige adipogenesis). iWAT, inguinal WAT.

Figure 3
lncRNAs in BAT adipogenesis

The research route for exploiting lncRNAs in BAT adipogenesis contains four steps: 1, transcriptional profiling; 2, verifying lncRNAs; 3, functional study; and 4, mechanism exploring. Two examples for regulatory mechanisms of lncRNAs in BAT are presented (upper panel: Firre–hnRNPU interplay-mediated trans-chromosomal interactions promotes adipogenesis; lower panel: Blnc1 cis-regulates EBF2, and Blnc1–EBF2 forms a feedforward loop to promote brown/beige adipogenesis). iWAT, inguinal WAT.

Close modal

BAT protects against obesity by promoting energy expenditure via uncoupled respiration, and is regarded as an attractive therapeutic target organ for obesity and its pathological consequences [1]. In the present review, we have summarized the functional role of two types of intrinsic factor, miRNA and lncRNA, in brown/beige adipogenesis and physiological function. Increased expression of miR-193b-365, miR-196a and miR-378 or inhibition of miR-133, miR-155, miR-27 and miR-34a can promote brown fat adipogenesis. Although it is still insufficient to construct a whole miRNA regulatory network in BAT adipogenesis, these studies display explicit correlations among miRNAs themselves and their targeted signalling pathways. On this basis, further investigations will probably reveal the core set of miRNAs responsible for reconstructing BAT with higher activities.

Comparing with the relatively unifying molecular mechanism of miRNAs, the mechanisms used by lncRNAs are more diverse. Firre and lnc-BATE1 interact with nuclear matrix factor hnRNPU to make a spatial reconstruction of chromosome to bring a few metabolic genes into closer proximity which may lead to co-regulation [48]. EBF2 and its cis-regulator Blnc1 form a feedforward loop to promote brown/beige adipogenesis [49]. More functional lncRNAs with different mechanisms are expected to emerge in the near future. These lncRNA studies, together with the miRNA study, will depict a new regulatory layer governing brown/beige fat plasticity under physiological and pathological conditions.

This work was supported by a Singapore National Research Foundation (NRF) fellowship [grant number NRF-2011NRF-NRFF 001-025 (to L.S.)]. This research is also supported by the Singapore National Research Foundation (NRF) under its Co-operative Basic Research Grant (CBRG) [grant number NMRC/CBRG/0070/2014] and administrated by the Singapore Ministry of Health's National Medical Research Council.

BAT

brown adipose tissue

Blnc1

brown fat lncRNA 1

C/EBP

CCAAT/enhancer-binding protein

CtBP

C-terminal binding protein

DGCR8

DiGeorge syndrome critical region 8

EBF2

early B-cell factor 2

eWAT

epididymal WAT

FGF

fibroblast growth factor

Firre

functional intergenic repeating RNA element

HFD

high-fat diet

hnRNPU

heterogeneous nuclear ribonucleoprotein U

Hox

homeobox

lnc-BATE

lncRNA BAT-enriched

lnc-RAP-n

lncRNA Regulated in AdiPogenesis

lncRNA

long non-coding RNA

MEF2

myocyte enhancer factor 2

ncRNA

non-coding RNA

PGC

PPARγ co-activator

PPAR

peroxisome-proliferator-activated receptor

PRDM16

PR domain-containing 16

SAT

subcutaneous adipose tissue

SVF

stromal-vascular fraction

TGFβ1

transforming growth factor β1

UCP1

uncoupling protein 1

WAT

white adipose tissue

1
Rosen
 
E.D.
Spiegelman
 
B.M.
 
What we talk about when we talk about fat
Cell
2014
, vol. 
156
 (pg. 
20
-
44
)
[PubMed]
2
Krauss
 
S.
Zhang
 
C.Y.
Lowell
 
B.B.
 
The mitochondrial uncoupling-protein homologues
Nat. Rev. Mol. Cell Biol.
2005
, vol. 
6
 (pg. 
248
-
261
)
[PubMed]
3
Guerra
 
C.
Koza
 
R.A.
Yamashita
 
H.
Walsh
 
K.
Kozak
 
L.P.
 
Emergence of brown adipocytes in white fat in mice is under genetic control: effects on body weight and adiposity
J. Clin. Invest.
1998
, vol. 
102
 (pg. 
412
-
420
)
[PubMed]
4
Wu
 
J.
Bostrom
 
P.
Sparks
 
L.M.
Ye
 
L.
Choi
 
J.H.
Giang
 
A.H.
Khandekar
 
M.
Virtanen
 
K.A.
Nuutila
 
P.
Schaart
 
G.
, et al 
Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human
Cell
2012
, vol. 
150
 (pg. 
366
-
376
)
[PubMed]
5
Lowell
 
B.B.
S-Suselic
 
V.
Hamann
 
A.
Lawitts
 
J.A.
Himms-Hagen
 
J.
Boyer
 
B.B.
Kozak
 
L.P.
Flier
 
J.S.
 
Development of obesity in transgenic mice after genetic ablation of brown adipose tissue
Nature
1993
, vol. 
366
 (pg. 
740
-
742
)
[PubMed]
6
Harms
 
M.
Seale
 
P.
 
Brown and beige fat: development, function and therapeutic potential
Nat. Med.
2013
, vol. 
19
 (pg. 
1252
-
1263
)
[PubMed]
7
Seale
 
P.
Bjork
 
B.
Yang
 
W.
Kajimura
 
S.
Chin
 
S.
Kuang
 
S.
Scime
 
A.
Devarakonda
 
S.
Conroe
 
H.M.
Erdjument-Bromage
 
H.
, et al 
PRDM16 controls a brown fat/skeletal muscle switch
Nature
2008
, vol. 
454
 (pg. 
961
-
967
)
[PubMed]
8
Kajimura
 
S.
Seale
 
P.
Kubota
 
K.
Lunsford
 
E.
Frangioni
 
J.V.
Gygi
 
S.P.
Spiegelman
 
B.M.
 
Initiation of myoblast to brown fat switch by a PRDM16-C/EBP-β transcriptional complex
Nature
2009
, vol. 
460
 (pg. 
1154
-
1158
)
[PubMed]
9
Kajimura
 
S.
Seale
 
P.
Tomaru
 
T.
Erdjument-Bromage
 
H.
Cooper
 
M.P.
Ruas
 
J.L.
Chin
 
S.
Tempst
 
P.
Lazar
 
M.A.
Spiegelman
 
B.M.
 
Regulation of the brown and white fat gene programs through a PRDM16/CtBP transcriptional complex
Genes Dev.
2008
, vol. 
22
 (pg. 
1397
-
1409
)
[PubMed]
10
Peirce
 
V.
Carobbio
 
S.
Vidal-Puig
 
A.
 
The different shades of fat
Nature
2014
, vol. 
510
 (pg. 
76
-
83
)
[PubMed]
11
Kajimura
 
S.
Saito
 
M.
 
A new era in brown adipose tissue biology: molecular control of brown fat development and energy homeostasis
Annu. Rev. Physiol.
2014
, vol. 
76
 (pg. 
225
-
249
)
[PubMed]
12
Villarroya
 
F.
Vidal-Puig
 
A.
 
Beyond the sympathetic tone: the new brown fat activators
Cell Metab.
2013
, vol. 
17
 (pg. 
638
-
643
)
[PubMed]
13
Lo
 
K.A.
Sun
 
L.
 
Turning WAT into BAT: a review on regulators controlling the browning of white adipocytes
Biosci. Rep.
2013
, vol. 
33
 pg. 
art:e00065
 
14
Knoll
 
M.
Lodish
 
H.F.
Sun
 
L.
 
Long non-coding RNAs as regulators of the endocrine system
Nat. Rev. Endrocrinol.
2015
, vol. 
11
 (pg. 
151
-
160
)
15
Valencia-Sanchez
 
M.A.
Liu
 
J.
Hannon
 
G.J.
Parker
 
R.
 
Control of translation and mRNA degradation by miRNAs and siRNAs
Genes Dev.
2006
, vol. 
20
 (pg. 
515
-
524
)
[PubMed]
16
Guo
 
H.
Ingolia
 
N.T.
Weissman
 
J.S.
Bartel
 
D.P.
 
Mammalian microRNAs predominantly act to decrease target mRNA levels
Nature
2010
, vol. 
466
 (pg. 
835
-
840
)
[PubMed]
17
Turner
 
M.
Galloway
 
A.
Vigorito
 
E.
 
Noncoding RNA and its associated proteins as regulatory elements of the immune system
Nat. Immunol.
2014
, vol. 
15
 (pg. 
484
-
491
)
[PubMed]
18
Kim
 
H.J.
Cho
 
H.
Alexander
 
R.
Patterson
 
H.C.
Gu
 
M.
Lo
 
K.A.
Xu
 
D.
Goh
 
V.J.
Nguyen
 
L.N.
Chai
 
X.
, et al 
MicroRNAs are required for the feature maintenance and differentiation of brown adipocytes
Diabetes
2014
, vol. 
63
 (pg. 
4045
-
4056
)
[PubMed]
19
Xie
 
H.
Sun
 
L.
Lodish
 
H.F.
 
Targeting microRNAs in obesity
Expert Opin. Ther. Targets
2009
, vol. 
13
 (pg. 
1227
-
1238
)
[PubMed]
20
Alexander
 
R.
Lodish
 
H.
Sun
 
L.
 
MicroRNAs in adipogenesis and as therapeutic targets for obesity
Expert Opin. Ther. Targets
2011
, vol. 
15
 (pg. 
623
-
636
)
[PubMed]
21
Mori
 
M.A.
Thomou
 
T.
Boucher
 
J.
Lee
 
K.Y.
Lallukka
 
S.
Kim
 
J.K.
Torriani
 
M.
Yki-Jarvinen
 
H.
Grinspoon
 
S.K.
Cypess
 
A.M.
Kahn
 
C.R.
 
Altered miRNA processing disrupts brown/white adipocyte determination and associates with lipodystrophy
J. Clin. Invest.
2014
, vol. 
124
 (pg. 
3339
-
3351
)
[PubMed]
22
Herranz
 
H.
Cohen
 
S.M.
 
MicroRNAs and gene regulatory networks: managing the impact of noise in biological systems
Genes Dev.
2010
, vol. 
24
 (pg. 
1339
-
1344
)
[PubMed]
23
Trajkovski
 
M.
Lodish
 
H.
 
MicroRNA networks regulate development of brown adipocytes
Trends Endocrinol. Metab.
2013
, vol. 
24
 (pg. 
442
-
450
)
[PubMed]
24
Sun
 
L.
Xie
 
H.
Mori
 
M.A.
Alexander
 
R.
Yuan
 
B.
Hattangadi
 
S.M.
Liu
 
Q.
Kahn
 
C.R.
Lodish
 
H.F.
 
Mir193b-365 is essential for brown fat differentiation
Nat. Cell Biol.
2011
, vol. 
13
 (pg. 
958
-
965
)
[PubMed]
25
Timmons
 
J.A.
Wennmalm
 
K.
Larsson
 
O.
Walden
 
T.B.
Lassmann
 
T.
Petrovic
 
N.
Hamilton
 
D.L.
Gimeno
 
R.E.
Wahlestedt
 
C.
Baar
 
K.
, et al 
Myogenic gene expression signature establishes that brown and white adipocytes originate from distinct cell lineages
Proc. Natl. Acad. Sci. U.S.A.
2007
, vol. 
104
 (pg. 
4401
-
4406
)
[PubMed]
26
Trajkovski
 
M.
Ahmed
 
K.
Esau
 
C.C.
Stoffel
 
M.
 
MyomiR-133 regulates brown fat differentiation through Prdm16
Nat. Cell Biol.
2012
, vol. 
14
 (pg. 
1330
-
1335
)
[PubMed]
27
Yin
 
H.
Pasut
 
A.
Soleimani
 
V.D.
Bentzinger
 
C.F.
Antoun
 
G.
Thorn
 
S.
Seale
 
P.
Fernando
 
P.
van Ijcken
 
W.
Grosveld
 
F.
, et al 
MicroRNA-133 controls brown adipose determination in skeletal muscle satellite cells by targeting Prdm16
Cell Metab.
2013
, vol. 
17
 (pg. 
210
-
224
)
[PubMed]
28
Liu
 
W.
Bi
 
P.
Shan
 
T.
Yang
 
X.
Yin
 
H.
Wang
 
Y.X.
Liu
 
N.
Rudnicki
 
M.A.
Kuang
 
S.
 
miR-133a regulates adipocyte browning in vivo
PLoS Genet.
2013
, vol. 
9
 pg. 
e1003626
 
[PubMed]
29
Williams
 
A.H.
Liu
 
N.
van Rooij
 
E.
Olson
 
E.N.
 
MicroRNA control of muscle development and disease
Curr. Opin. Cell Biol.
2009
, vol. 
21
 (pg. 
461
-
469
)
[PubMed]
30
Chen
 
J.F.
Mandel
 
E.M.
Thomson
 
J.M.
Wu
 
Q.
Callis
 
T.E.
Hammond
 
S.M.
Conlon
 
F.L.
Wang
 
D.Z.
 
The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation
Nat. Genet.
2006
, vol. 
38
 (pg. 
228
-
233
)
[PubMed]
31
Mori
 
M.
Nakagami
 
H.
Rodriguez-Araujo
 
G.
Nimura
 
K.
Kaneda
 
Y.
 
Essential role for miR-196a in brown adipogenesis of white fat progenitor cells
PLoS Biol.
2012
, vol. 
10
 pg. 
e1001314
 
[PubMed]
32
Narendra
 
V.
Rocha
 
P.P.
An
 
D.
Raviram
 
R.
Skok
 
J.A.
Mazzoni
 
E.O.
Reinberg
 
D.
 
Transcription
CTCF establishes discrete functional chromatin domains at the Hox clusters during differentiation
Science
2015
, vol. 
347
 (pg. 
1017
-
1021
)
[PubMed]
33
Schulz
 
T.J.
Huang
 
T.L.
Tran
 
T.T.
Zhang
 
H.
Townsend
 
K.L.
Shadrach
 
J.L.
Cerletti
 
M.
McDougall
 
L.E.
Giorgadze
 
N.
Tchkonia
 
T.
, et al 
Identification of inducible brown adipocyte progenitors residing in skeletal muscle and white fat
Proc. Natl. Acad. Sci. U.S.A.
2011
, vol. 
108
 (pg. 
143
-
148
)
[PubMed]
34
Yadav
 
H.
Quijano
 
C.
Kamaraju
 
A.K.
Gavrilova
 
O.
Malek
 
R.
Chen
 
W.
Zerfas
 
P.
Zhigang
 
D.
Wright
 
E.C.
Stuelten
 
C.
, et al 
Protection from obesity and diabetes by blockade of TGF-β/Smad3 signaling
Cell Metab.
2011
, vol. 
14
 (pg. 
67
-
79
)
[PubMed]
35
Ignotz
 
R.A.
Massague
 
J.
 
Type β transforming growth factor controls the adipogenic differentiation of 3T3 fibroblasts
Proc. Natl. Acad. Sci. U.S.A.
1985
, vol. 
82
 (pg. 
8530
-
8534
)
[PubMed]
36
Chen
 
Y.
Siegel
 
F.
Kipschull
 
S.
Haas
 
B.
Frohlich
 
H.
Meister
 
G.
Pfeifer
 
A.
 
miR-155 regulates differentiation of brown and beige adipocytes via a bistable circuit
Nat. Commun.
2013
, vol. 
4
 pg. 
1769
 
[PubMed]
37
Sun
 
L.
Trajkovski
 
M.
 
MiR-27 orchestrates the transcriptional regulation of brown adipogenesis
Metabolism
2014
, vol. 
63
 (pg. 
272
-
282
)
[PubMed]
38
Kong
 
X.
Yu
 
J.
Bi
 
J.
Qi
 
H.
Di
 
W.
Wu
 
L.
Wang
 
L.
Zha
 
J.
Lv
 
S.
Zhang
 
F.
, et al 
Glucocorticoids transcriptionally regulate miR-27b expression promoting body fat accumulation via suppressing the browning of white adipose tissue
Diabetes
2015
, vol. 
64
 (pg. 
393
-
404
)
[PubMed]
39
Pan
 
D.
Mao
 
C.
Quattrochi
 
B.
Friedline
 
R.H.
Zhu
 
L.J.
Jung
 
D.Y.
Kim
 
J.K.
Lewis
 
B.
Wang
 
Y.X.
 
MicroRNA-378 controls classical brown fat expansion to counteract obesity
Nat. Commun.
2014
, vol. 
5
 pg. 
4725
 
[PubMed]
40
Shin
 
S.
Heo
 
J.
Yeo
 
J.
Lee
 
C.
Chung
 
E.
 
Genetic association of phosphodiesterase 1B (PDE1B) with carcass traits in Korean cattle
Mol. Biol. Rep.
2012
, vol. 
39
 (pg. 
4869
-
4874
)
[PubMed]
41
Cristancho
 
A.G.
Lazar
 
M.A.
 
Forming functional fat: a growing understanding of adipocyte differentiation
Nat. Rev. Mol. Cell Biol.
2011
, vol. 
12
 (pg. 
722
-
734
)
[PubMed]
42
Fu
 
T.
Seok
 
S.
Choi
 
S.
Huang
 
Z.
Suino-Powell
 
K.
Xu
 
H.E.
Kemper
 
B.
Kemper
 
J.K.
 
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
, vol. 
34
 (pg. 
4130
-
4142
)
[PubMed]
43
Qiang
 
L.
Wang
 
L.
Kon
 
N.
Zhao
 
W.
Lee
 
S.
Zhang
 
Y.
Rosenbaum
 
M.
Zhao
 
Y.
Gu
 
W.
Farmer
 
S.R.
Accili
 
D.
 
Brown remodeling of white adipose tissue by SirT1-dependent deacetylation of PPARγ
Cell
2012
, vol. 
150
 (pg. 
620
-
632
)
[PubMed]
44
Fisher
 
F.M.
Kleiner
 
S.
Douris
 
N.
Fox
 
E.C.
Mepani
 
R.J.
Verdeguer
 
F.
Wu
 
J.
Kharitonenkov
 
A.
Flier
 
J.S.
Maratos-Flier
 
E.
Spiegelman
 
B.M.
 
FGF21 regulates PGC-1α and browning of white adipose tissues in adaptive thermogenesis
Genes Dev.
2012
, vol. 
26
 (pg. 
271
-
281
)
[PubMed]
45
Chen
 
J.
Cui
 
X.
Shi
 
C.
Chen
 
L.
Yang
 
L.
Pang
 
L.
Zhang
 
J.
Guo
 
X.
Wang
 
J.
Ji
 
C.
 
Differential lncRNA expression profiles in brown and white adipose tissues
Mol. Genet. Genomics
2015
, vol. 
290
 (pg. 
699
-
707
)
[PubMed]
46
You
 
L.H.
Zhu
 
L.J.
Yang
 
L.
Shi
 
C.M.
Pang
 
L.X.
Zhang
 
J.
Cui
 
X.W.
Ji
 
C.B.
Guo
 
X.R.
 
Transcriptome analysis reveals the potential contribution of long noncoding RNAs to brown adipocyte differentiation
Mol. Genet. Genomics
2015
[PubMed]
47
Sun
 
L.
Goff
 
L.A.
Trapnell
 
C.
Alexander
 
R.
Lo
 
K.A.
Hacisuleyman
 
E.
Sauvageau
 
M.
Tazon-Vega
 
B.
Kelley
 
D.R.
Hendrickson
 
D.G.
, et al 
Long noncoding RNAs regulate adipogenesis
Proc. Natl. Acad. Sci. U.S.A.
2013
, vol. 
110
 (pg. 
3387
-
3392
)
[PubMed]
48
Hacisuleyman
 
E.
Goff
 
L.A.
Trapnell
 
C.
Williams
 
A.
Henao-Mejia
 
J.
Sun
 
L.
McClanahan
 
P.
Hendrickson
 
D.G.
Sauvageau
 
M.
Kelley
 
D.R.
, et al 
Topological organization of multichromosomal regions by the long intergenic noncoding RNA Firre
Nat. Struct. Mol. Biol.
2014
, vol. 
21
 (pg. 
198
-
206
)
[PubMed]
49
Zhao
 
X.Y.
Li
 
S.
Wang
 
G.X.
Yu
 
Q.
Lin
 
J.D.
 
A long noncoding RNA transcriptional regulatory circuit drives thermogenic adipocyte differentiation
Mol. Cell
2014
, vol. 
55
 (pg. 
372
-
382
)
[PubMed]
50
Alvarez-Dominguez
 
J.R.
Bai
 
Z.
Xu
 
D.
Yuan
 
B.
Lo
 
K.A.
Yoon
 
M.J.
Lim
 
Y.C.
Knoll
 
M.
Slavov
 
N.
Chen
 
S.
, et al 
De novo reconstruction of adipose tissue transcriptomes reveals long non-coding RNA regulators of brown adipocyte development
Cell Metab.
2015
, vol. 
21
 (pg. 
764
-
776
)
[PubMed]
This is an open access article published by Portland Press Limited and distributed under the Creative Commons Attribution Licence 3.0.