Accepted Manuscript MicroRNAs in brown and beige fat
Deborah Goody, Alexander Pfeifer PII: DOI: Reference:
S1388-1981(18)30095-7 doi:10.1016/j.bbalip.2018.05.003 BBAMCB 58292
To appear in: Received date: Revised date: Accepted date:
26 October 2017 5 February 2018 4 May 2018
Please cite this article as: Deborah Goody, Alexander Pfeifer , MicroRNAs in brown and beige fat. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Bbamcb(2018), doi:10.1016/j.bbalip.2018.05.003
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT MicroRNAs in brown and beige fat Deborah Goody and Alexander Pfeifer
Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127
RI
PT
Bonn, Germany
SC
Correspondence:
Alexander Pfeifer, Institute of Pharmacology and Toxicology, University Hospital Bonn; University of
NU
Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany; (Tel) +49-(0)228-287 51300; (Fax) +49-
AC
CE
PT E
D
MA
(0)228-287 51301; email:
[email protected]
1
ACCEPTED MANUSCRIPT Highlights
MicroRNA (miRNA) regulate brown and beige adipogenesis miRNA can promote or inhibit the brown adipogenesis We review the mechanisms behind miRNA-regulation of brown and beige adipose tissue Recent findings might lead to the development of miRNA-based treatments against obesity
PT
Abstract
PT E
D
MA
NU
SC
RI
Brown adipose tissue (BAT) dissipates energy as heat and its activity correlates with leanness in human adults. Understanding the mechanisms behind the activation of BAT and the process of "browning", i.e. the appearance of inducible brown adipocytes called beige or brite (brown-in-white) cells in white adipose tissue (WAT), is of great interest for developing novel therapies to combat obesity. MicroRNAs (miRNAs) are small transcriptional regulators that control gene expression in a variety of tissues, including WAT and BAT. Recently, miRNAs were reported to regulate browning. Nevertheless, further studies are needed to fully understand the miRNA networks that are involved in the control of brown and beige/brite adipocytes. Particularly, most miRNA have so far been studied in mice, underlining the importance of additional human studies. In this review, we focus on the regulation of brown fat by miRNAs including their role in promoting or inhibiting the browning process. In recent years, RNA-based therapeutical approaches have entered clinical trials for treatment of other diseases, thus miRNAs could potentially be used to enhance brown and beige fat mass and activity as novel therapies against overweight and its complications.
CE
Keywords: Brown adipocyte, Beige adipocyte, MicroRNA, Obesity, Metabolism, Energy Homeostasis
AC
Abbreviations: ADAM17, ADAM metallopeptidase domain 17; AMPK, AMP-activated protein kinase; aP2, fatty-acid binding protein 4; AGO, Argonaute protein; ASO, anti-sense oligonucleotide; Bace, amyloid precursor protein-cleaving enzyme 1; BAT, brown adipose tissue; Blnc1, brown fat lncRNA 1; BMP7, bone morphogenetic protein 7; cAMP, cyclic AMP; C/EBP, CCAAT/enhancer-binding protein; cGMP, cyclic GMP; Cidea, cell death-inducing DNA fragmentation factor alpha-like effector A; Cox7, cytochrome c oxidase subunit 7; DIO, diet induced obesity; Dgcr8, microprocessor complex subunit; FABP4, fatty-acid binding protein 4; FGF21, fibroblast growth factor 21; Fndc5, fibronectin type III domain containing 5; HDAC3, histone deacetylase 3; HIF1, hypoxia-inducible factor 1; hMADS, human multipotent adipose tissue-derived stem cells; HoxC8, Homeobox C8; iWAT, inguinal white adipose tissue; Insig1, insulin induced gene 1; LNA, locked nucleic acid; lncRNAs, long non-coding RNAs; MAPK, p38 mitogen activated protein kinase; Mef2, myocyte enhancer factor-2; miRNA, microRNA; Mstn, myostatin; NE, norepinephrine; NPs, natriuretic peptides; Pde, phosphodiesterase; PGC-1α, peroxisome-proliferator-activated receptor γ-coactivator 1α; PHB, prohibitin; PKA, protein kinase A; PPAR, peroxisome-proliferator-activated receptor; PRDM16, protein PR domain containing 16; pri-miRNA, primary miRNA; RIP140, receptor-interacting protein 140; RNAi, RNA interference; Runx1t1, Runt-related transcription factor 1, translocated to, 1; scWAT, subcutaneous white adipose tissue; SIRT1, sirtuin 1; T2D, type 2 diabetes; UCP1, uncoupling protein 1; vWAT, visceral white adipose tissue; WAT, white adipose tissue.
2
ACCEPTED MANUSCRIPT 1. Introduction
MA
NU
SC
RI
PT
Over the past decades the worldwide population suffering from obesity has severely increased [1]. According to the world health organization (WHO) 1.9 billion human adults are overweight and more people die of overweight and obesity than of underweight and famine (WHO, 2014). Excessive fat accumulation is a result of an imbalance in energy homeostasis caused by an increased calorie intake versus calories expenditure. Various studies show that obesity and overweight are major health problems as they increase the risk of developing other diseases such as type 2 diabetes, cardiovascular disease and certain types of cancer [2]. Therapeutic options to treat overweight and obesity are lacking and understanding the underlying molecular mechanisms would help develop new, more efficient anti-obesity therapies. Current therapeutic options include dietetic and exercise treatment as well as bariatric surgery. Available pharmacological therapy to date, such as Orlistat and Liraglutide, are only indicated in special cases and can cause severe side-effects [3, 4]. In previous studies, we and others have established microRNAs (miRNAs) as relevant biomarkers of metabolism [5, 6]. Furthermore, recent research underlines the critical role of miRNAs in the regulation of adipose tissue and metabolism [7]. miRNAs have been reported to directly or indirectly regulate signaling pathways essential for the development and differentiation as well as function of adipose tissue. 1.1. Brown and beige fat tissue
AC
CE
PT E
D
There are two kinds of adipose tissue in mammals: white adipose tissue (WAT) and brown adipose tissue (BAT) [8]. The main tissue responsible for storing energy in form of triglycerides is WAT. WAT accumulates excessive energy in form of fat and is distributed throughout the whole body. The major WAT depots are classified based on their location as visceral or gonadal WAT (vWAT, gWAT) versus subcutaneous or inguinal WAT (scWAT, iWAT). White adipocytes feature unilocular lipid droplets, and considerably vary in size depending on the lipid load [9]. Contrary to WAT, BAT utilizes stored chemical energy for the production of heat in a process called non-shivering thermogenesis (NST) [10]. The involvement of BAT in heat production has long been known [11], but it was believed to play a major role only in newborns. Although interscapular BAT is the major thermogenic depot in infants, BAT appears to be widely distributed throughout the body during the first decade of life [12]. Anatomical studies showed that interscapular BAT disappears with advancing years [12], however, brown adipocytes can be found also in adults in the deeper regions of the body [12]. It has only been in the last decade that metabolically active BAT was identified in adult humans using positron emission tomography (PET) coupled with computer tomography (CT) imaging of radioactive glucose uptake (FDG-PET/CT) [13-16]. FDG-PET/CT clearly detected active BAT in the supraclavicular and neck regions as well as in the mediastinum [11, 13, 14, 16-18]. Brown adipocytes contain a high abundance of mitochondria and small multilocular lipid droplets. The protein mainly responsible for NST is the uncoupling protein 1 (UCP1), which is located in the membrane of mitochondria and uncouples the proton gradient to generate heat [19, 20]. BAT is highly innervated by the sympathetic nervous system, which releases 3
ACCEPTED MANUSCRIPT norepinephrine (NE) and the cotransmitter ATP which is rapidly converted to adenosine upon cold stimulation [21]. NE activates G protein-coupled receptors, thereby inducing the production of cyclic AMP (cAMP) and activation of protein kinase A (PKA) in brown adipocytes [17]. Regarding the action of adenosine in BAT, there are important species differences: adenosine activates human and murine brown adipocytes, but inhibits activation of BAT from hamster or rat [17, 22-25].
MA
NU
SC
RI
PT
In addition to WAT and BAT, inducible brown adipocytes also called beige or brite (“brownin-white”) adipocytes exist in WAT depots, mainly iWAT/scWAT [26]. These cells are induced upon cold acclimatization and can dissipate energy as heat, a process also known as “browning” [27, 28]. Morphologically, beige adipocytes share major characteristics with classical brown adipocytes, including multilocular fat droplets, a high mitochondrial content and expression of brown fat-specific genes, including UCP1, cell death-inducing DNA fragmentation factor alpha-like effector A (Cidea), peroxisome-proliferator-activated receptor γ-coactivator 1α (PGC-1α), protein PR domain containing 16 (PRDM16) and CCAAT/enhancer-binding protein β (C/EBPβ) [28]. Several studies indicate that brown adipocytes have a different origin than white and beige adipocytes (Seale et al., 2008). Nonetheless, it is still under debate whether mature white adipocytes have the ability to transdifferentiate, or "un-mask" into beige adipocytes, or whether beige cells derive from a separate precursor cell line, which shares the same origin as white adipocytes, by de-novo differentiation [29-31]. Interestingly, Long and colleagues showed that the origin of beige adipocytes is very heterogenous depending on the location of WAT depots [32].
D
2. Brown fat: Function of miRNAs in brown adipogenesis
AC
CE
PT E
The development and function of brown adipocytes is tightly regulated by various hormones and proteins factors [28, 33, 34]. We and others have previously demonstrated that miRNAs also play a critical role in brown adipose tissue as transcriptional regulators and biomarkers [5, 7]. Extracellular miRNAs can be found in fluids, including blood and urine, packaged in extracellular vesicles or in microparticle-free form [35-38]. Their level has been linked to the status and progression of various diseases, including cancer [36, 39, 40]. Circulating miR-122 has been shown to be associated with type 2 diabetes and exosomal miR-92a inversely correlates with murine and human BAT activity [5, 41]. MiRNAs belong to the family of small non-coding RNAs, which comprise approximately 22 nucleotides and mediate RNA interference (RNAi) [42]. They are produced from short hairpin structures by two RNase III proteins, Drosha and Dicer, and associate with Argonaute (AGO) proteins [43]. These small RNAs decrease protein synthesis via interaction with partially complementary sites in the 3′untranslated region (3´UTR) of target messenger RNAs (mRNAs) [44]. MiRNA exerts its function as a guide by recognizing mRNAs, whereas AGO proteins recruit factors that induce translational repression and mRNA destabilization [45]. In this way, miRNAs may positively or negatively regulate classical white, brown and beige fat development. Although the involvement of miRNA in brown/beige adipogenesis is still not completely understood [4648], several studies have identified miRNAs that have specific functions in brown (Fig. 1) and/or beige (Fig. 2) adipocytes. 4
ACCEPTED MANUSCRIPT 2.1. Positive regulators of brown fat miR-328
NU
SC
RI
PT
Recent studies demonstrated the coincidence of ageing- and obesity-associated dysfunction of brown fat with impaired expression of the miRNA processing protein Dicer [49, 50]. Analysis of miRNA-expression identified miR-328 as a potential regulator of BAT differentiation [51]. MiR-328 was upregulated during brown adipocyte differentiation and decreased in BAT of DIO mice [51]. Inhibition of miR-328 in primary immortalized brown adipocytes resulted in decreased expression of thermogenic genes such as Cidea, Ucp1, Pgc-1α and Prdm16 (Fig. 1) [51]. Additionally, overexpression of miR-328 resulted in increased levels of C/EBPβ, UCP1 as well as oxygen consumption rates in brown adipocytes. Interestingly, there is a link between miR-328 and amyloid precursor protein-cleaving enzyme 1 (Bace1) [52]. Luciferase reporter assay revealed the direct downregulation of Bace1 by miR-328. Overexpression of miR-328 in brown adipocytes reduced Bace expression, while inhibition of miR-328 increased Bace protein levels. Moreover, Bace1 knockout mice demonstrate increased energy expenditure, elevated UCP1 expression in brown fat and are resistant against DIO [53]. miR-182/miR-203
AC
CE
PT E
D
MA
To address the question on how miRNAs contribute to maintenance of brown adipocytes, Kim and colleagues specifically inhibited Dgcr8 in mice [54]. Drosha, the key regulator of miRNA biogenesis, together with its key cofactor Dgcr8 (also known as Pasha in D. melanogaster) form the microprocessor complex, which further processes primary miRNA (pri-miRNA) in pre-miRNA in the cell nucleus [55]. Adipose tissue-specific Dgcr8 knockout mice showed "whitened" interscapular BAT with decreased expression of thermogenic genes as well as enlarged scWAT and epididymal WAT [54]. Furthermore, miRNA expression profiling identified miR-182 and miR-203 as new regulators of brown adipocyte development. The treatment of brown adipocytes with miR-182 and miR-203 inhibitors in vitro caused a significant reduction of brown fat markers, including UCP1, PGC-1α and Cidea, as well as mitochondrial markers (e.g. cytochrome c oxidase subunit 7, Cox7) (Fig. 1). Interestingly, the mRNA levels of classical adipogenic markers, such as peroxisome proliferator-activated receptor gamma (Pparγ) and fatty acid binding protein 4 (Fabp4) did not change upon inhibition of these miRNAs. A potential mechanism that might explain the effects of miR-182 and miR-203 in brown adipocytes are their targets, including the plateletderived growth factor alpha (PDGFRα) and insulin-induced gene 1 (INSIG1), which have been described as disruptors of adipocyte differentiation [56, 57]. miR-193b-365 In 2011, Sun and colleagues demonstrated that miR-193b and miR-365 form a cluster (miRNA-193b-365), which is highly expressed in BAT [52]. Moreover, they showed that the miRNA-193b-365 cluster significantly promotes murine brown adipocyte adipogenesis by inhibiting runt-related transcription factor 1 translocated to 1 (Runx1t1) expression - an inhibitor of white and brown adipogenesis (see also miR-455) [52]. Contradictory to these 5
ACCEPTED MANUSCRIPT findings, later in vivo studies showed that blocking miR-193b-365 cluster does not change the morphology of BAT and the expression of brown markers (UCP1, PRDM16) [58]. Therefore, further studies are necessary to understand the potential role of the miR-193b-365 cluster in differentiation and function of BAT. 2.2. Negative regulators of brown fat miR-106b-93
MA
NU
SC
RI
PT
Various members of the miR-17 family, including miR-106 and miR-93, control differentiation of stem cells in developing mouse embryos [59]. High levels of miR-106b and miR-93 were found in brown adipose tissue of obese mice [60]. In brown adipocytes, knockdown of miR-106b and miR-93 using locked nucleic acid (LNA) miRNA inhibitors markedly increased the expression of brown fat-specific genes, including Ucp1, Prdm16 and Cidea, while overexpression of miR-106b and miR-93 suppressed UCP1 expression (Fig. 1) [60]. Based on these findings, the miR-106b-93 cluster might play a significant role in energy homeostasis through negative regulation of brown adipocytes. Based on preliminary in silico target-analysis PPARα may be a target of miR-106b/-93 [60]. Nevertheless, further in vitro and in vivo studies are required to identify the exact mechanism of this miRNA regulator.
3. Beige fat-specific miRNAs: Role of miRNAs in browning
CE
PT E
D
In addition to being critical regulators of brown adipocyte development and function, recent studies revealed that miRNAs potentially play a central role in beige adipocytes [61]. Understanding the regulatory mechanism of miRNAs during this process may help developing new therapeutic approaches to increase energy expenditure and combat the obesity. Thus, we present here the current research on miRNAs specifically involved in the browning process (Fig. 2).
miR-196a
AC
3.1. Regulator that specifically enhance browning
Homeobox genes (Hox) genes, which are important for differentiation (e.g. hematopoiesis, myogenesis) [62, 63], have been shown to be differentially regulated in WAT and BAT [64]. Hoxc8 has been characterized as a marker gene for white fat, which is targeted by miR-196a [64, 65]. In 2012, Mori et al. observed an upregulation of miR-196a in iWAT from mice exposed to cold or adrenergic stimulation [65]. Moreover, miR-196a induces brown adipogenesis in human WAT. Regarding the mechanism, Mori et al showed that HOXC8 represses the brown fat-regulator C/EBPβ in cooperation with histone deacetylase 3 (HDAC3) in subcutaneous white adipocytes (Fig. 2) [65]. In addition, mice overexpressing miR-196a showed resistance to obesity and improved glucose metabolism [65]. 3.2. Negative regulator of browning 6
ACCEPTED MANUSCRIPT miR-125-5p
PT
As described earlier, the precise origin of beige adipocytes has been controversial [31, 66]. Nevertheless, miR-125-5p was identified as a negative regulator of beige adipocyte formation and function [67]. This miRNA was shown to be expressed at lower levels in BAT than in WAT, and miR-125-5p levels decreased during browning, being inverse proportional to UCP1 expression (Fig. 2) [67]. Importantly, injection of miR-125-5p into iWAT inhibited beige adipocyte formation and mitochondrial biogenesis, whereas injection of LNA inhibitors resulted in increased browning [67]. However, the mechanism behind the effect of miR-125b5p on beige adipocytes remains unknown.
SC
RI
4. Involvement of miRNAs in the regulation of both brown adipocytes and browning process
MA
NU
Recent studies demonstrated that several miRNAs play important roles in both brown adipocytes and the browning process. Some miRNAs, such as miR-30 and miR-27, have identical effects in both brown and beige adipocytes, respectively [68-72]. In contrast miR378 remains the only characterized miRNA to have opposite functions in brown and beige adipogenesis [73]. Here, we present the current research on miRNAs involved in both brown adipocytes and the browning process (Fig. 3).
D
4.1. Positive regulators of brown and beige adipocytes
PT E
miR-30
AC
CE
The miR-30 family was first identified in adipogenesis through deep sequencing of human multipotent adipose tissue-derived stem (hMADS) cells [68]. MiR-30 family members were upregulated during adipogenic differentiation and their inhibition impaired adipogenesis markedly, partially via regulation of the transcription factor RUNX2 [68]. Their role in the regulation of thermogenesis was later described in vitro and in vivo [69]. MiR-30b/c levels were induced by cold exposure, and positively regulated the expression of thermogenic genes, including UCP1 and Cidea, in brown adipocytes (Fig. 3). Moreover, overexpression of miR30b/c greatly increased thermogenic gene expression in primary subcutaneous white adipocytes [69]. The promoting effect of these miRNAs on the development of beige fat was further supported by the finding that miR-30b/c represses the receptor-interacting protein 140 (RIP140), which functions as a corepressor of thermogenic genes [74]. miR-26 The miR-26a and miR-26b were the first miRNAs identified that regulate both adipocyte development as well as the acquisition of brown adipocyte characteristics in humans [75]. Inhibition of both miRNAs in the human adipocyte cell line hMADS prevented lipid accumulation and the expression of adipogenic genes such as FABP4, while ectopic miRexpression increased the differentiation towards a brown phenotype (Fig. 3) [75]. The positive 7
ACCEPTED MANUSCRIPT effect of miR-26a/b on browning of white adipocytes is due to its repressor activity on the sheddase ADAM17. Mice lacking ADAM17 were previously shown to be resistant to dietinduced obesity, further underlining the physiological importance of the link between miR26a/b and ADAM17 [76, 77]. miR-455
MA
NU
SC
RI
PT
MiR-455 was first identified as a new key regulator of brown adipogenesis in multipotent mesenchymal cells after treatment with bone morphogenetic protein 7 (BMP7) [78]. BMP7 had previously been shown to promote brown differentiation of preadipocytes and increase brown fat mass [79, 80]. By analyzing rodent and human tissue samples, the authors established miR-455 as a genuine BAT marker for both species [78]. Further in vitro experiments characterized miR-455 as a downstream effector of BMP7- and cold-mediated browning [78]. Adipose-specific gain-of-function mice (transgenic mice expressing miR-455 under the control of the Fabp4 promoter) exhibited increased browning of iWAT upon cold exposure compared to control [78]. Mechanistically, miR-455 inhibits the adipogenic suppressors Runx1t1 and Necdin, thus activating the expression of adipogenic markers (e.g. PPARγ and PGC1α) (Fig. 3) [78]. Furthermore, miR-455 activates AMPK1 by targeting HIF1, promoting adipogenic differentiation and mitochondrial biogenesis [78, 81-83]. miR-32
AC
CE
PT E
D
MiR-32 has been showed to be involved in lipid metabolism, but its role in BAT function was not identified until recently [84]. Ng et al. employed chromatin immunoprecipitation sequencing (ChIP-seq) and identified miR-32 as a BAT-specific super-enhancer [85]. Moreover, miR-32 levels were significantly increased in BAT during cold exposure promoting BAT thermogenesis and iWAT browning. The in vivo inhibition of miR-32 via injection of miR-32 anti-sense oligonucleotide (ASO) led to impaired thermogenic response and lower core body temperatures after cold exposure [85]. The BAT of cold-exposed mice injected with miR-32-ASO showed no significant morphologically changes but markedly lower levels of UCP1 compared to control (Fig. 3). Interestingly, inhibition of miR-32 resulted in less browning of iWAT after cold exposure, including significantly fewer multilocular cells and downregulation of BAT markers: Cidea, PPAR, PGC1, and UCP1. miR-32 represses the tumor suppressor Tob1, thus activating the p38/MAPK pathway, which is stimulated during cold exposure and induces the expression of fibroblast growth factor 21 (FGF21) [86-88]. FGF21 is a secreted factor that has multiple physiological roles including enhanced browning of iWAT [89]. 4.2. Negative regulators of brown and beige adipocytes miR-27 miR-27 was first characterized as a negative regulator of adipogenesis in hMADS cells partially through suppression of PPARγ and C/EBPα [46, 70, 90]. The group of Dong Liu 8
ACCEPTED MANUSCRIPT later described a novel mechanism of miR-27 in the inhibition of adipogenesis by targeting prohibitin (PHB), which is highly expressed in cells that rely heavily on mitochondrial function [71, 91]. Furthermore, miR-27 loss-of-function experiments in iWAT revealed an upregulation of thermogenic markers including UCP1, PRDM16 and PGC1 (Fig. 3) [72]. Concomitantly, another group showed that miR-27b functions as a negative regulator of PRDM16 to control browning of WAT [92]. miR-155
D
MA
NU
SC
RI
PT
Previous studies identified adipogenic transcription factor C/EBPβ as a target gene of miR155 in inflammation and in vitro models of white adipogenesis [93-95]. Subsequently, miR155 was characterized by our group as a negative regulator of brown adipogenesis by targeting C/EBPβ resulting in impaired UCP1 and PGC1 expression (Fig. 3) [7]. Surprisingly, promoter sequence analysis revealed putative C/EBP-binding sequences in the miR-155 promoter region [7]. The authors propose a bistable loop of adipocyte commitment, where high levels of miR-155 inhibit C/EBPβ expression and premature differentiation, while C/EBPβ induction by pro-adipogenic hormones inhibits transcription of miR-155. At the same time, miRNA-155 is positively regulated by transforming growth factor-β1 (TGFβ1), which has been described as an inhibitor of adipogenesis [96]. Inhibition of miR-155 augmented brown adipocyte differentiation and promoted a browning phenotype in white adipocytes in vitro and in vivo [7]. MiRNA-155 deficient mice showed a higher cellular respiration and increased number of “brown-like” adipocytes in iWAT after cold exposure [7].
PT E
miR-133
AC
CE
MiR-133 was previously shown to be expressed in BAT and downregulated after cold exposure, accompanied by the upregulation of the key thermogenic regulator PRDM16 [52, 97, 98]. Furthermore, miR-133 demonstrates a putative binding motif within the 3´UTR of PRDM16 [98]. Ectopic expression of miR-133 greatly impaired PRDM16 expression and decreased expression of brown genes, including Ucp1, Ppar and Pparγ (Fig. 3). In contrast, inhibition of miR-133 or its transcriptional regulator myocyte enhancer factor-2 (Mef2) enhanced differentiation of brown adipocytes and browning of white adipocytes [98]. Remarkably, double knockout of miR-133a (a1 and a2) in mice increased the thermogenic gene program in iWAT and shifted the morphology of these cells to a beige phenotype [99]. miR-34a Fu et al. found that high levels of miR-34a in obesity inhibit fat browning and brown fat formation partially by suppressing FGF21 and sirtuin 1 (SIRT1) [100]. Mice with dietinduced obesity, which were systemically injected with lentivirus encoding antisense miR-34a showed an increased mitochondrial function and expression of thermogenic markers in all types of WATs [100]. Mechanistically, miR-34a inhibition promotes the expression of FGF21 and SIRT1, and consequently FGF21/SIRT1-dependent deacetylation of PGC1, resulting in the induction of the browning genes Ucp1, and Prdm16 (Fig. 3). Furthermore, miR-34a has 9
ACCEPTED MANUSCRIPT been showed to be involved in myostatin (Mstn) mediated regulation of the protein fibronectin type III domain containing 5 (Fndc5) and browning of white adipocytes [101]. The cleaved version of Fndc5, a hormone called Irisin, was postulated as an inducer of WAT browning via activation of UCP1 expression [102]. 4.3. Regulator with opposing functions in brown and beige fat miR-378
D
5. Summary and Perspectives
MA
NU
SC
RI
PT
The miR-378 has been described as the first and only miRNA to have opposite functions in brown and beige adipocytes [73]. Transgenic mice overexpressing miR-378 specifically in adipose tissue exhibit an increased brown adipogenesis and increased BAT mass, but not inguinal or gonadal WAT mass. The authors hypothesized that BAT expansion, rather than miR-378 per se, leads to inhibition of beige adipocytes in iWAT. Moreover, miR-378 overexpression in iWAT decreased UCP1 and PRDM16 levels, whereas concentration of classical adipogenic markers (PPARγ and aP2) remained normal [73]. Regarding the mechanism behind this dual regulation, miR-378 targets the phosphodiesterase Pde1b, which catalyses the turnover of cAMP and cGMP, in BAT but not in WAT [73, 103]. In this way, miRNA-378 selectively promotes brown adipogenesis, while impairing beige adipogenesis in WAT.
AC
CE
PT E
In the last few years, BAT has been linked to amelioration of overweight and gained more interest as potential target of novel therapeutic tools for the fight of obesity [17]. Recently, inducible brown adipocytes in WAT depots, called beige or brite cells, have gained more interest due to their capability for energy expenditure and their positive effect on diet-induced obesity [104]. Besides hormones and protein factors, miRNAs are also key regulators of brown adipogenesis and the commitment of beige adipocytes to a brown-phenotype, in the “browning” process [5]. This regulation is based on the tight control of transcriptional regulators and important mediators of adipocyte development, including C/EBPβ and PGC1α/β, through miRNAs [7, 72]. MiRNA levels have been shown to be dysregulated in metabolic diseases, including obesity [105]. Because miRNA can be transported in body fluids, e.g. blood and urine, e.g. packaged in extracellular vesicles, they might play a central role in metabolic crosstalk between different cell-types in adipose tissues and/or between different organs. Interestingly, adipose tissue represents a major source of circulating exosomal miRNAs, and could therefore regulate gene expression in distant tissues [5, 106]. Consequently, circulating miRNAs may serve as useful biomarkers of brown/beige fat function, and may be potential therapeutic tool against overweight [5]. Although many studies have described circulating miRNAs as potential biomarkers of metabolic disorder [107], exosomal miRNAs were only recently described as indicators for BAT activity in the context of metabolism (Table 1) [5, 106]. Chen et al. demonstrated the inverse correlation of exosomal miR-92a with murine and human BAT activity [5]. In a recent study, Thomou et al. 10
ACCEPTED MANUSCRIPT showed that adipose tissue represents an important source of circulating exosomal miRNAs, which can function as regulators of secondary organs [106]. Table 1: Summary of circulating miRNAs involved in metabolism disorder.
MA
NU
SC
RI
PT
Role in metabolism inverse correlation with BAT activity correlation with BAT and WAT abundance positive correlation with obesity positive correlation with obesity Inverse correlation with obesity and T2D inverse correlation with obesity inverse correlation with obesity positive correlation with obesity inverse correlation with obesity inverse correlation with obesity inverse correlation with obesity inverse correlation with obesity inverse correlation with obesity inverse correlation with obesity positive correlation with obesity positive correlation with obesity positive correlation with obesity inverse correlation with T2D and obesity inverse correlation with T2D and obesity positive correlation with T2D inverse correlation with T2D inverse correlation with T2D inverse correlation with T2D inverse correlation with T2D inverse correlation with T2D inverse correlation with T2D inverse correlation with T2D inverse correlation with T2D inverse correlation with T2D inverse correlation with T2D positive correlation with T2D positive correlation with T2D positive correlation with T2D positive correlation with T2D positive correlation with T2D positive correlation with T2D positive correlation with T2D
D
PT E
Organism human and mouse human and mouse human human human human human human human human human human human human human human human human human human human human human human human human human human human human human human human human human human human
CE
miRNA exosomal miR-92a exosomal miR-99b miR-152 miR-17 miR-593 miR-138 miR-376a miR-15b miR-520c-3p miR-423-5p miR-532-5p miR-125b miR-130b miR-221 miR-140-5p miR-142-3p miR-222 miR-15a miR-503 miR-326 let-7a/f miR-20b miR-21 miR-24 miR-126 miR-191 miR-197 miR-223 miR-320 miR-486 miR-9 miR-29a miR-30d miR-34a miR-124a miR-146a miR-375
Reference [5] [106] [107] [107] [107] [38, 107] [38] [38] [108] [108] [108] [108] [108] [108] [108] [108] [108] [108-110] [38] [111] [111] [110] [110] [110] [110] [110] [110] [110] [110] [110] [112] [112] [112] [112] [112] [112] [112]
AC
In vitro and/or in vivo studies showed that miRNAs can positively and negatively impact brown/beige adipogenesis. In this review, we summarize positive regulators (brown-specific: miR-328, miR-182, miR-203, beige-specific: miR-196a) and negative regulators (brownspecific: miR-106b-93; beige-specific: miR-125-5p) of brown/beige adipocyte development. Interestingly, various miRNAs are involved in the regulation of both brown and beige adipocytes (miR-30, miR-26, miR-455, miR-32, miR-27, miR-155, miR-133, miR-34a, miR378). Another class of non-coding RNAs are long non-coding RNAs (lncRNAs), which are no longer than 200 nucleotides and play crucial biological roles [113]. Although their function in white adipocyte differentiation has been studied, their contribution to the browning process remains largely unknown [114, 115]. Recent studies showed that brown fat lncRNA 1 (Blnc1) promotes brown/beige adipogenesis and is required for the positive transcriptional regulation of thermogenic genes through recruitment of transcription factors of brown fat [116, 117]. 11
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
Despite growing knowledge on the function of non-coding RNAs in fat, further studies are needed to fully discern the complex mechanisms behind miRNA-mediated adipose tissue regulation, in order to develop therapeutic approaches to treat overweight.
12
ACCEPTED MANUSCRIPT References
AC
CE
PT E
D
MA
NU
SC
RI
PT
[1] M. Ng, T. Fleming, M. Robinson, B. Thomson, N. Graetz, C. Margono, E.C. Mullany, S. Biryukov, C. Abbafati, S.F. Abera, et al., Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013, Lancet, 384 (2014) 766-781. [2] S.M. Grundy, H.B. Brewer, Jr., J.I. Cleeman, S.C. Smith, Jr., C. Lenfant, Definition of metabolic syndrome: Report of the National Heart, Lung, and Blood Institute/American Heart Association conference on scientific issues related to definition, Circulation, 109 (2004) 433-438. [3] J. Zhi, A.T. Melia, R. Guerciolini, J. Chung, J. Kinberg, J.B. Hauptman, I.H. Patel, Retrospective population-based analysis of the dose-response (fecal fat excretion) relationship of orlistat in normal and obese volunteers, Clin Pharmacol Ther, 56 (1994) 82-85. [4] G.A. Bray, G. Fruhbeck, D.H. Ryan, J.P. Wilding, Management of obesity, Lancet, 387 (2016) 1947-1956. [5] Y. Chen, J.J. Buyel, M.J. Hanssen, F. Siegel, R. Pan, J. Naumann, M. Schell, A. van der Lans, C. Schlein, H. Froehlich, et al., Exosomal microRNA miR-92a concentration in serum reflects human brown fat activity, Nat Commun, 7 (2016) 11420. [6] J.A. Deiuliis, MicroRNAs as regulators of metabolic disease: pathophysiologic significance and emerging role as biomarkers and therapeutics, Int J Obes (Lond), 40 (2016) 88-101. [7] Y. Chen, F. Siegel, S. Kipschull, B. Haas, H. Frohlich, G. Meister, A. Pfeifer, miR-155 regulates differentiation of brown and beige adipocytes via a bistable circuit, Nat Commun, 4 (2013) 1769. [8] B.B. Lowell, J.S. Flier, Brown adipose tissue, beta 3-adrenergic receptors, and obesity, Annu Rev Med, 48 (1997) 307-316. [9] S. Cinti, M. Cigolini, G. Gazzanelli, O. Bosello, An ultrastructural study of adipocyte precursors from epididymal fat pads of adult rats in culture, J Submicrosc Cytol, 17 (1985) 631636. [10] D.O. Foster, M.L. Frydman, Nonshivering thermogenesis in the rat. II. Measurements of blood flow with microspheres point to brown adipose tissue as the dominant site of the calorigenesis induced by noradrenaline, Can J Physiol Pharmacol, 56 (1978) 110-122. [11] D. Hull, M.M. Segall, The contribution of brown adipose tissue to heat production in the new-born rabbit, J Physiol, 181 (1965) 449-457. [12] J.M. Heaton, The distribution of brown adipose tissue in the human, J Anat, 112 (1972) 3539. [13] A.M. Cypess, S. Lehman, G. Williams, I. Tal, D. Rodman, A.B. Goldfine, F.C. Kuo, E.L. Palmer, Y.H. Tseng, A. Doria, et al., Identification and importance of brown adipose tissue in adult humans, N Engl J Med, 360 (2009) 1509-1517. [14] M. Saito, Y. Okamatsu-Ogura, M. Matsushita, K. Watanabe, T. Yoneshiro, J. NioKobayashi, T. Iwanaga, M. Miyagawa, T. Kameya, K. Nakada, et al., High incidence of metabolically active brown adipose tissue in healthy adult humans: effects of cold exposure and adiposity, Diabetes, 58 (2009) 1526-1531. [15] W.D. van Marken Lichtenbelt, J.W. Vanhommerig, N.M. Smulders, J.M. Drossaerts, G.J. Kemerink, N.D. Bouvy, P. Schrauwen, G.J. Teule, Cold-activated brown adipose tissue in healthy men, N Engl J Med, 360 (2009) 1500-1508. [16] K.A. Virtanen, M.E. Lidell, J. Orava, M. Heglind, R. Westergren, T. Niemi, M. Taittonen, J. Laine, N.J. Savisto, S. Enerback, et al., Functional brown adipose tissue in healthy adults, N Engl J Med, 360 (2009) 1518-1525. [17] B. Cannon, J. Nedergaard, Brown adipose tissue: function and physiological significance, Physiol Rev, 84 (2004) 277-359. [18] W.D. van Marken Lichtenbelt, P. Schrauwen, Implications of nonshivering thermogenesis for energy balance regulation in humans, Am J Physiol Regul Integr Comp Physiol, 301 (2011) R285-296. [19] D.G. Nicholls, V.S. Bernson, G.M. Heaton, The identification of the component in the inner membrane of brown adipose tissue mitochondria responsible for regulating energy dissipation, Experientia Suppl, 32 (1978) 89-93. 13
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[20] S. Cinti, C. Zancanaro, A. Sbarbati, M. Cicolini, P. Vogel, D. Ricquier, S. Fakan, Immunoelectron microscopical identification of the uncoupling protein in brown adipose tissue mitochondria, Biol Cell, 67 (1989) 359-362. [21] T. Gnad, S. Scheibler, I. von Kugelgen, C. Scheele, A. Kilic, A. Glode, L.S. Hoffmann, L. Reverte-Salisa, P. Horn, S. Mutlu, et al., Adenosine activates brown adipose tissue and recruits beige adipocytes via A2A receptors, Nature, 516 (2014) 395-399. [22] L.M. Dickson, S. Gandhi, B.T. Layden, R.N. Cohen, B. Wicksteed, Protein kinase A induces UCP1 expression in specific adipose depots to increase energy expenditure and improve metabolic health, Am J Physiol Regul Integr Comp Physiol, 311 (2016) R79-88. [23] R.J. Schimmel, L. McCarthy, Role of adenosine as an endogenous regulator of respiration in hamster brown adipocytes, Am J Physiol, 246 (1984) C301-307. [24] D. Szillat, L.J. Bukowiecki, Control of brown adipose tissue lipolysis and respiration by adenosine, Am J Physiol, 245 (1983) E555-559. [25] J.A. Woodward, E.D. Saggerson, Effect of adenosine deaminase, N6phenylisopropyladenosine and hypothyroidism on the responsiveness of rat brown adipocytes to noradrenaline, Biochem J, 238 (1986) 395-403. [26] B. Cousin, S. Cinti, M. Morroni, S. Raimbault, D. Ricquier, L. Penicaud, L. Casteilla, Occurrence of brown adipocytes in rat white adipose tissue: molecular and morphological characterization, J Cell Sci, 103 ( Pt 4) (1992) 931-942. [27] M. Harms, P. Seale, Brown and beige fat: development, function and therapeutic potential, Nat Med, 19 (2013) 1252-1263. [28] A. Pfeifer, L.S. Hoffmann, Brown, beige, and white: the new color code of fat and its pharmacological implications, Annu Rev Pharmacol Toxicol, 55 (2015) 207-227. [29] A. Vitali, I. Murano, M.C. Zingaretti, A. Frontini, D. Ricquier, S. Cinti, The adipose organ of obesity-prone C57BL/6J mice is composed of mixed white and brown adipocytes, J Lipid Res, 53 (2012) 619-629. [30] J. Nedergaard, B. Cannon, The browning of white adipose tissue: some burning issues, Cell Metab, 20 (2014) 396-407. [31] Q.A. Wang, C. Tao, R.K. Gupta, P.E. Scherer, Tracking adipogenesis during white adipose tissue development, expansion and regeneration, Nat Med, 19 (2013) 1338-1344. [32] J.Z. Long, K.J. Svensson, L. Tsai, X. Zeng, H.C. Roh, X. Kong, R.R. Rao, J. Lou, I. Lokurkar, W. Baur, et al., A smooth muscle-like origin for beige adipocytes, Cell Metab, 19 (2014) 810-820. [33] S. Kajimura, P. Seale, T. Tomaru, H. Erdjument-Bromage, M.P. Cooper, J.L. Ruas, S. Chin, P. Tempst, M.A. Lazar, B.M. Spiegelman, Regulation of the brown and white fat gene programs through a PRDM16/CtBP transcriptional complex, Genes Dev, 22 (2008) 1397-1409. [34] P. Seale, B. Bjork, W. Yang, S. Kajimura, S. Chin, S. Kuang, A. Scime, S. Devarakonda, H.M. Conroe, H. Erdjument-Bromage, et al., PRDM16 controls a brown fat/skeletal muscle switch, Nature, 454 (2008) 961-967. [35] M. Arrese, A. Eguchi, A.E. Feldstein, Circulating microRNAs: emerging biomarkers of liver disease, Semin Liver Dis, 35 (2015) 43-54. [36] M.A. Javidi, A.H. Ahmadi, B. Bakhshinejad, N. Nouraee, S. Babashah, M. Sadeghizadeh, Cell-free microRNAs as cancer biomarkers: the odyssey of miRNAs through body fluids, Med Oncol, 31 (2014) 295. [37] J. Zhang, S. Li, L. Li, M. Li, C. Guo, J. Yao, S. Mi, Exosome and exosomal microRNA: trafficking, sorting, and function, Genomics Proteomics Bioinformatics, 13 (2015) 17-24. [38] N. Pescador, M. Perez-Barba, J.M. Ibarra, A. Corbaton, M.T. Martinez-Larrad, M. Serrano-Rios, Serum circulating microRNA profiling for identification of potential type 2 diabetes and obesity biomarkers, PLoS One, 8 (2013) e77251. [39] J.R. Chevillet, Q. Kang, I.K. Ruf, H.A. Briggs, L.N. Vojtech, S.M. Hughes, H.H. Cheng, J.D. Arroyo, E.K. Meredith, E.N. Gallichotte, et al., Quantitative and stoichiometric analysis of the microRNA content of exosomes, Proc Natl Acad Sci U S A, 111 (2014) 14888-14893. [40] S. Vienberg, J. Geiger, S. Madsen, L.T. Dalgaard, MicroRNAs in metabolism, Acta Physiol (Oxf), 219 (2017) 346-361.
14
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[41] P. Willeit, P. Skroblin, A.R. Moschen, X. Yin, D. Kaudewitz, A. Zampetaki, T. Barwari, M. Whitehead, C.M. Ramirez, L. Goedeke, et al., Circulating MicroRNA-122 Is Associated With the Risk of New-Onset Metabolic Syndrome and Type 2 Diabetes, Diabetes, 66 (2017) 347-357. [42] R.C. Lee, V. Ambros, An extensive class of small RNAs in Caenorhabditis elegans, Science, 294 (2001) 862-864. [43] M. Ha, V.N. Kim, Regulation of microRNA biogenesis, Nat Rev Mol Cell Biol, 15 (2014) 509-524. [44] W. Filipowicz, S.N. Bhattacharyya, N. Sonenberg, Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight?, Nat Rev Genet, 9 (2008) 102-114. [45] E. Huntzinger, E. Izaurralde, Gene silencing by microRNAs: contributions of translational repression and mRNA decay, Nat Rev Genet, 12 (2011) 99-110. [46] M. Karbiener, C. Fischer, S. Nowitsch, P. Opriessnig, C. Papak, G. Ailhaud, C. Dani, E.Z. Amri, M. Scheideler, microRNA miR-27b impairs human adipocyte differentiation and targets PPARgamma, Biochem Biophys Res Commun, 390 (2009) 247-251. [47] M. Karbiener, C. Neuhold, P. Opriessnig, A. Prokesch, J.G. Bogner-Strauss, M. Scheideler, MicroRNA-30c promotes human adipocyte differentiation and co-represses PAI-1 and ALK2, RNA Biol, 8 (2011) 850-860. [48] C. Hilton, M.J. Neville, F. Karpe, MicroRNAs in adipose tissue: their role in adipogenesis and obesity, Int J Obes (Lond), 37 (2013) 325-332. [49] N.H. Rogers, A. Landa, S. Park, R.G. Smith, Aging leads to a programmed loss of brown adipocytes in murine subcutaneous white adipose tissue, Aging Cell, 11 (2012) 1074-1083. [50] M.A. Mori, T. Thomou, J. Boucher, K.Y. Lee, S. Lallukka, J.K. Kim, M. Torriani, H. YkiJarvinen, S.K. Grinspoon, A.M. Cypess, et al., Altered miRNA processing disrupts brown/white adipocyte determination and associates with lipodystrophy, J Clin Invest, 124 (2014) 3339-3351. [51] M. Oliverio, E. Schmidt, J. Mauer, C. Baitzel, N. Hansmeier, S. Khani, S. Konieczka, M. Pradas-Juni, S. Brodesser, T.M. Van, et al., Dicer1-miR-328-Bace1 signalling controls brown adipose tissue differentiation and function, Nat Cell Biol, 18 (2016) 328-336. [52] L. Sun, H. Xie, M.A. Mori, R. Alexander, B. Yuan, S.M. Hattangadi, Q. Liu, C.R. Kahn, H.F. Lodish, Mir193b-365 is essential for brown fat differentiation, Nat Cell Biol, 13 (2011) 958965. [53] P.J. Meakin, A.J. Harper, D.L. Hamilton, J. Gallagher, A.D. McNeilly, L.A. Burgess, L.M. Vaanholt, K.A. Bannon, J. Latcham, I. Hussain, et al., Reduction in BACE1 decreases body weight, protects against diet-induced obesity and enhances insulin sensitivity in mice, Biochem J, 441 (2012) 285-296. [54] H.J. Kim, H. Cho, R. Alexander, H.C. Patterson, M. Gu, K.A. Lo, D. Xu, V.J. Goh, L.N. Nguyen, X. Chai, et al., MicroRNAs are required for the feature maintenance and differentiation of brown adipocytes, Diabetes, 63 (2014) 4045-4056. [55] A.M. Denli, B.B. Tops, R.H. Plasterk, R.F. Ketting, G.J. Hannon, Processing of primary microRNAs by the Microprocessor complex, Nature, 432 (2004) 231-235. [56] S. Fitter, K. Vandyke, S. Gronthos, A.C. Zannettino, Suppression of PDGF-induced PI3 kinase activity by imatinib promotes adipogenesis and adiponectin secretion, J Mol Endocrinol, 48 (2012) 229-240. [57] J. Li, K. Takaishi, W. Cook, S.K. McCorkle, R.H. Unger, Insig-1 "brakes" lipogenesis in adipocytes and inhibits differentiation of preadipocytes, Proc Natl Acad Sci U S A, 100 (2003) 9476-9481. [58] Y. Feuermann, K. Kang, O. Gavrilova, N. Haetscher, S.J. Jang, K.H. Yoo, C. Jiang, F.J. Gonzalez, G.W. Robinson, L. Hennighausen, MiR-193b and miR-365-1 are not required for the development and function of brown fat in the mouse, RNA Biol, 10 (2013) 1807-1814. [59] K.M. Foshay, G.I. Gallicano, miR-17 family miRNAs are expressed during early mammalian development and regulate stem cell differentiation, Dev Biol, 326 (2009) 431-443. [60] Y. Wu, J. Zuo, Y. Zhang, Y. Xie, F. Hu, L. Chen, B. Liu, F. Liu, Identification of miR-106b93 as a negative regulator of brown adipocyte differentiation, Biochem Biophys Res Commun, 438 (2013) 575-580. [61] N. Arias, L. Aguirre, A. Fernandez-Quintela, M. Gonzalez, A. Lasa, J. Miranda, M.T. Macarulla, M.P. Portillo, MicroRNAs involved in the browning process of adipocytes, J Physiol Biochem, 72 (2016) 509-521. 15
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[62] B. Argiropoulos, R.K. Humphries, Hox genes in hematopoiesis and leukemogenesis, Oncogene, 26 (2007) 6766-6776. [63] I. Naguibneva, M. Ameyar-Zazoua, A. Polesskaya, S. Ait-Si-Ali, R. Groisman, M. Souidi, S. Cuvellier, A. Harel-Bellan, The microRNA miR-181 targets the homeobox protein Hox-A11 during mammalian myoblast differentiation, Nat Cell Biol, 8 (2006) 278-284. [64] T.J. Schulz, T.L. Huang, T.T. Tran, H. Zhang, K.L. Townsend, J.L. Shadrach, M. Cerletti, L.E. McDougall, N. Giorgadze, T. Tchkonia, et al., Identification of inducible brown adipocyte progenitors residing in skeletal muscle and white fat, Proc Natl Acad Sci U S A, 108 (2011) 143148. [65] M. Mori, H. Nakagami, G. Rodriguez-Araujo, K. Nimura, Y. Kaneda, Essential role for miR-196a in brown adipogenesis of white fat progenitor cells, PLoS Biol, 10 (2012) e1001314. [66] S. Cinti, Transdifferentiation properties of adipocytes in the adipose organ, Am J Physiol Endocrinol Metab, 297 (2009) E977-986. [67] M. Giroud, D.F. Pisani, M. Karbiener, V. Barquissau, R.A. Ghandour, D. Tews, P. FischerPosovszky, J.C. Chambard, U. Knippschild, T. Niemi, et al., miR-125b affects mitochondrial biogenesis and impairs brite adipocyte formation and function, Mol Metab, 5 (2016) 615-625. [68] L.E. Zaragosi, B. Wdziekonski, K.L. Brigand, P. Villageois, B. Mari, R. Waldmann, C. Dani, P. Barbry, Small RNA sequencing reveals miR-642a-3p as a novel adipocyte-specific microRNA and miR-30 as a key regulator of human adipogenesis, Genome Biol, 12 (2011) R64. [69] F. Hu, M. Wang, T. Xiao, B. Yin, L. He, W. Meng, M. Dong, F. Liu, miR-30 promotes thermogenesis and the development of beige fat by targeting RIP140, Diabetes, 64 (2015) 20562068. [70] S.Y. Kim, A.Y. Kim, H.W. Lee, Y.H. Son, G.Y. Lee, J.W. Lee, Y.S. Lee, J.B. Kim, miR-27a is a negative regulator of adipocyte differentiation via suppressing PPARgamma expression, Biochem Biophys Res Commun, 392 (2010) 323-328. [71] T. Kang, W. Lu, W. Xu, L. Anderson, M. Bacanamwo, W. Thompson, Y.E. Chen, D. Liu, MicroRNA-27 (miR-27) targets prohibitin and impairs adipocyte differentiation and mitochondrial function in human adipose-derived stem cells, J Biol Chem, 288 (2013) 3439434402. [72] L. Sun, M. Trajkovski, MiR-27 orchestrates the transcriptional regulation of brown adipogenesis, Metabolism, 63 (2014) 272-282. [73] D. Pan, C. Mao, B. Quattrochi, R.H. Friedline, L.J. Zhu, D.Y. Jung, J.K. Kim, B. Lewis, Y.X. Wang, MicroRNA-378 controls classical brown fat expansion to counteract obesity, Nat Commun, 5 (2014) 4725. [74] J. Nautiyal, M. Christian, M.G. Parker, Distinct functions for RIP140 in development, inflammation, and metabolism, Trends Endocrinol Metab, 24 (2013) 451-459. [75] M. Karbiener, D.F. Pisani, A. Frontini, L.M. Oberreiter, E. Lang, A. Vegiopoulos, K. Mossenbock, G.A. Bernhardt, T. Mayr, F. Hildner, et al., MicroRNA-26 family is required for human adipogenesis and drives characteristics of brown adipocytes, Stem Cells, 32 (2014) 15781590. [76] M. Serino, R. Menghini, L. Fiorentino, R. Amoruso, A. Mauriello, D. Lauro, P. Sbraccia, M.L. Hribal, R. Lauro, M. Federici, Mice heterozygous for tumor necrosis factor-alpha converting enzyme are protected from obesity-induced insulin resistance and diabetes, Diabetes, 56 (2007) 2541-2546. [77] R.W. Gelling, W. Yan, S. Al-Noori, A. Pardini, G.J. Morton, K. Ogimoto, M.W. Schwartz, P.J. Dempsey, Deficiency of TNFalpha converting enzyme (TACE/ADAM17) causes a lean, hypermetabolic phenotype in mice, Endocrinology, 149 (2008) 6053-6064. [78] H. Zhang, M. Guan, K.L. Townsend, T.L. Huang, D. An, X. Yan, R. Xue, T.J. Schulz, J. Winnay, M. Mori, et al., MicroRNA-455 regulates brown adipogenesis via a novel HIF1anAMPK-PGC1alpha signaling network, EMBO Rep, 16 (2015) 1378-1393. [79] Y.H. Tseng, E. Kokkotou, T.J. Schulz, T.L. Huang, J.N. Winnay, C.M. Taniguchi, T.T. Tran, R. Suzuki, D.O. Espinoza, Y. Yamamoto, et al., New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure, Nature, 454 (2008) 1000-1004. [80] T.J. Schulz, P. Huang, T.L. Huang, R. Xue, L.E. McDougall, K.L. Townsend, A.M. Cypess, Y. Mishina, E. Gussoni, Y.H. Tseng, Brown-fat paucity due to impaired BMP signalling induces compensatory browning of white fat, Nature, 495 (2013) 379-383. 16
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[81] J.J. Rochford, R.K. Semple, M. Laudes, K.B. Boyle, C. Christodoulides, C. Mulligan, C.J. Lelliott, S. Schinner, D. Hadaschik, M. Mahadevan, et al., ETO/MTG8 is an inhibitor of C/EBPbeta activity and a regulator of early adipogenesis, Mol Cell Biol, 24 (2004) 9863-9872. [82] Y.H. Tseng, A.J. Butte, E. Kokkotou, V.K. Yechoor, C.M. Taniguchi, K.M. Kriauciunas, A.M. Cypess, M. Niinobe, K. Yoshikawa, M.E. Patti, et al., Prediction of preadipocyte differentiation by gene expression reveals role of insulin receptor substrates and necdin, Nat Cell Biol, 7 (2005) 601-611. [83] N. Zhang, Z. Fu, S. Linke, J. Chicher, J.J. Gorman, D. Visk, G.G. Haddad, L. Poellinger, D.J. Peet, F. Powell, et al., The asparaginyl hydroxylase factor inhibiting HIF-1alpha is an essential regulator of metabolism, Cell Metab, 11 (2010) 364-378. [84] D. Shin, S.Y. Howng, L.J. Ptacek, Y.H. Fu, miR-32 and its target SLC45A3 regulate the lipid metabolism of oligodendrocytes and myelin, Neuroscience, 213 (2012) 29-37. [85] R. Ng, N.A. Hussain, Q. Zhang, C. Chang, H. Li, Y. Fu, L. Cao, W. Han, W. Stunkel, F. Xu, miRNA-32 Drives Brown Fat Thermogenesis and Trans-activates Subcutaneous White Fat Browning in Mice, Cell Rep, 19 (2017) 1229-1246. [86] M.W. Helms, D. Kemming, C.H. Contag, H. Pospisil, K. Bartkowiak, A. Wang, S.Y. Chang, H. Buerger, B.H. Brandt, TOB1 is regulated by EGF-dependent HER2 and EGFR signaling, is highly phosphorylated, and indicates poor prognosis in node-negative breast cancer, Cancer Res, 69 (2009) 5049-5056. [87] Y. Jiao, K.K. Sun, L. Zhao, J.Y. Xu, L.L. Wang, S.J. Fan, Suppression of human lung cancer cell proliferation and metastasis in vitro by the transducer of ErbB-2.1 (TOB1), Acta Pharmacol Sin, 33 (2012) 250-260. [88] D.V. Chartoumpekis, I.G. Habeos, P.G. Ziros, A.I. Psyrogiannis, V.E. Kyriazopoulou, A.G. Papavassiliou, Brown adipose tissue responds to cold and adrenergic stimulation by induction of FGF21, Mol Med, 17 (2011) 736-740. [89] F.M. Fisher, S. Kleiner, N. Douris, E.C. Fox, R.J. Mepani, F. Verdeguer, J. Wu, A. Kharitonenkov, J.S. Flier, E. Maratos-Flier, et al., FGF21 regulates PGC-1alpha and browning of white adipose tissues in adaptive thermogenesis, Genes Dev, 26 (2012) 271-281. [90] Q. Lin, Z. Gao, R.M. Alarcon, J. Ye, Z. Yun, A role of miR-27 in the regulation of adipogenesis, FEBS J, 276 (2009) 2348-2358. [91] P.J. Coates, R. Nenutil, A. McGregor, S.M. Picksley, D.H. Crouch, P.A. Hall, E.G. Wright, Mammalian prohibitin proteins respond to mitochondrial stress and decrease during cellular senescence, Exp Cell Res, 265 (2001) 262-273. [92] X. Kong, J. Yu, J. Bi, H. Qi, W. Di, L. Wu, L. Wang, J. Zha, S. Lv, F. Zhang, et al., Glucocorticoids transcriptionally regulate miR-27b expression promoting body fat accumulation via suppressing the browning of white adipose tissue, Diabetes, 64 (2015) 393-404. [93] M. He, Z. Xu, T. Ding, D.M. Kuang, L. Zheng, MicroRNA-155 regulates inflammatory cytokine production in tumor-associated macrophages via targeting C/EBPbeta, Cell Mol Immunol, 6 (2009) 343-352. [94] J. Worm, J. Stenvang, A. Petri, K.S. Frederiksen, S. Obad, J. Elmen, M. Hedtjarn, E.M. Straarup, J.B. Hansen, S. Kauppinen, Silencing of microRNA-155 in mice during acute inflammatory response leads to derepression of c/ebp Beta and down-regulation of G-CSF, Nucleic Acids Res, 37 (2009) 5784-5792. [95] M. Skarn, H.M. Namlos, P. Noordhuis, M.Y. Wang, L.A. Meza-Zepeda, O. Myklebost, Adipocyte differentiation of human bone marrow-derived stromal cells is modulated by microRNA-155, microRNA-221, and microRNA-222, Stem Cells Dev, 21 (2012) 873-883. [96] R.A. Ignotz, J. Massague, Type beta transforming growth factor controls the adipogenic differentiation of 3T3 fibroblasts, Proc Natl Acad Sci U S A, 82 (1985) 8530-8534. [97] T.B. Walden, J.A. Timmons, P. Keller, J. Nedergaard, B. Cannon, Distinct expression of muscle-specific microRNAs (myomirs) in brown adipocytes, J Cell Physiol, 218 (2009) 444-449. [98] M. Trajkovski, K. Ahmed, C.C. Esau, M. Stoffel, MyomiR-133 regulates brown fat differentiation through Prdm16, Nat Cell Biol, 14 (2012) 1330-1335. [99] W. Liu, P. Bi, T. Shan, X. Yang, H. Yin, Y.X. Wang, N. Liu, M.A. Rudnicki, S. Kuang, miR133a regulates adipocyte browning in vivo, PLoS Genet, 9 (2013) e1003626. [100] T. Fu, S. Seok, S. Choi, Z. Huang, K. Suino-Powell, H.E. Xu, B. Kemper, J.K. Kemper, MicroRNA 34a inhibits beige and brown fat formation in obesity in part by suppressing 17
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
adipocyte fibroblast growth factor 21 signaling and SIRT1 function, Mol Cell Biol, 34 (2014) 4130-4142. [101] X. Ge, D. Sathiakumar, B.J. Lua, H. Kukreti, M. Lee, C. McFarlane, Myostatin signals through miR-34a to regulate Fndc5 expression and browning of white adipocytes, Int J Obes (Lond), 41 (2017) 137-148. [102] P. Bostrom, J. Wu, M.P. Jedrychowski, A. Korde, L. Ye, J.C. Lo, K.A. Rasbach, E.A. Bostrom, J.H. Choi, J.Z. Long, et al., A PGC1-alpha-dependent myokine that drives brown-fatlike development of white fat and thermogenesis, Nature, 481 (2012) 463-468. [103] S.H. Francis, M.A. Blount, J.D. Corbin, Mammalian cyclic nucleotide phosphodiesterases: molecular mechanisms and physiological functions, Physiol Rev, 91 (2011) 651-690. [104] P. Cohen, J.D. Levy, Y. Zhang, A. Frontini, D.P. Kolodin, K.J. Svensson, J.C. Lo, X. Zeng, L. Ye, M.J. Khandekar, et al., Ablation of PRDM16 and beige adipose causes metabolic dysfunction and a subcutaneous to visceral fat switch, Cell, 156 (2014) 304-316. [105] R.A. McGregor, M.S. Choi, microRNAs in the regulation of adipogenesis and obesity, Curr Mol Med, 11 (2011) 304-316. [106] T. Thomou, M.A. Mori, J.M. Dreyfuss, M. Konishi, M. Sakaguchi, C. Wolfrum, T.N. Rao, J.N. Winnay, R. Garcia-Martin, S.K. Grinspoon, et al., Adipose-derived circulating miRNAs regulate gene expression in other tissues, Nature, 542 (2017) 450-455. [107] L. Wu, X. Dai, J. Zhan, Y. Zhang, H. Zhang, S. Zeng, W. Xi, Profiling peripheral microRNAs in obesity and type 2 diabetes mellitus, APMIS, 123 (2015) 580-585. [108] F.J. Ortega, J.M. Mercader, V. Catalan, J.M. Moreno-Navarrete, N. Pueyo, M. Sabater, J. Gomez-Ambrosi, R. Anglada, J.A. Fernandez-Formoso, W. Ricart, et al., Targeting the circulating microRNA signature of obesity, Clin Chem, 59 (2013) 781-792. [109] G. Al-Kafaji, G. Al-Mahroos, N.A. Alsayed, Z.A. Hasan, S. Nawaz, M. Bakhiet, Peripheral blood microRNA-15a is a potential biomarker for type 2 diabetes mellitus and pre-diabetes, Mol Med Rep, 12 (2015) 7485-7490. [110] A. Zampetaki, S. Kiechl, I. Drozdov, P. Willeit, U. Mayr, M. Prokopi, A. Mayr, S. Weger, F. Oberhollenzer, E. Bonora, et al., Plasma microRNA profiling reveals loss of endothelial miR126 and other microRNAs in type 2 diabetes, Circ Res, 107 (2010) 810-817. [111] D. Santovito, V. De Nardis, P. Marcantonio, C. Mandolini, C. Paganelli, E. Vitale, F. Buttitta, M. Bucci, A. Mezzetti, A. Consoli, et al., Plasma exosome microRNA profiling unravels a new potential modulator of adiponectin pathway in diabetes: effect of glycemic control, J Clin Endocrinol Metab, 99 (2014) E1681-1685. [112] L. Kong, J. Zhu, W. Han, X. Jiang, M. Xu, Y. Zhao, Q. Dong, Z. Pang, Q. Guan, L. Gao, et al., Significance of serum microRNAs in pre-diabetes and newly diagnosed type 2 diabetes: a clinical study, Acta Diabetol, 48 (2011) 61-69. [113] L. Sun, L.A. Goff, C. Trapnell, R. Alexander, K.A. Lo, E. Hacisuleyman, M. Sauvageau, B. Tazon-Vega, D.R. Kelley, D.G. Hendrickson, et al., Long noncoding RNAs regulate adipogenesis, Proc Natl Acad Sci U S A, 110 (2013) 3387-3392. [114] Y. Huang, Y. Zheng, C. Jin, X. Li, L. Jia, W. Li, Long Non-coding RNA H19 Inhibits Adipocyte Differentiation of Bone Marrow Mesenchymal Stem Cells through Epigenetic Modulation of Histone Deacetylases, Sci Rep, 6 (2016) 28897. [115] M. Li, X. Sun, H. Cai, Y. Sun, M. Plath, C. Li, X. Lan, C. Lei, F. Lin, Y. Bai, et al., Long non-coding RNA ADNCR suppresses adipogenic differentiation by targeting miR-204, Biochim Biophys Acta, 1859 (2016) 871-882. [116] S. Li, L. Mi, L. Yu, Q. Yu, T. Liu, G.X. Wang, X.Y. Zhao, J. Wu, J.D. Lin, Zbtb7b engages the long noncoding RNA Blnc1 to drive brown and beige fat development and thermogenesis, Proc Natl Acad Sci U S A, 114 (2017) E7111-E7120. [117] X.Y. Zhao, S. Li, G.X. Wang, Q. Yu, J.D. Lin, A long noncoding RNA transcriptional regulatory circuit drives thermogenic adipocyte differentiation, Mol Cell, 55 (2014) 372-382.
18
ACCEPTED MANUSCRIPT Figure legends
PT
Figure 1: Molecular mechanisms of miRNAs that specifically regulate brown adipogenesis. miRNAs that have a positive effect on UCP1 in brown adipogenesis are depicted on the left; negative miRNA in brown adipocytes is shown on the right; exemplified here by their effect on UCP1 expression. Positive regulators block the inhibitory effect of theirs targets on thermogenic markers; the mechanism behind the negative regulator miR-106b-93 remains unknown.
NU
SC
RI
Figure 2: MiRNAs that regulate specifically the browning process. The positive miR-196a is shown on the left; negative miR-125-5p on the right; exemplified here by their effect on UCP1 expression. MiR-196a inhibits the negative effect of its target on thermogenic markers; the mechanism behind the negative regulator miR-125-5p is not clear.
AC
CE
PT E
D
MA
Figure 3: MiRNAs that are involved in the regulation of both brown adipocytes and browning process. Positive regulators are depicted on top (+); negative regulators are shown in the bottom (-). Positive regulators block the inhibitory effect of theirs targets on thermogenic markers; negative regulators target factors important for the browning process; exemplified here by their effect on UCP1 expression.
19
ACCEPTED MANUSCRIPT Highlights
PT RI SC NU MA D PT E CE
MicroRNA (miRNA) regulate brown and beige adipogenesis miRNA can promote or inhibit the brown adipogenesis We review the mechanisms behind miRNA-regulation of brown and beige adipose tissue Recent findings might lead to the development of miRNA-based treatments against obesity
AC
20
Figure 1
Figure 2
Figure 3