Mineralocorticoid receptor in adipocytes and macrophages: A promising target to fight metabolic...

8
1 3 Mineralocorticoid receptor in adipocytes and macrophages: A promising 4 target to fight metabolic syndrome 5 6 7 Vincenzo Marzolla a Q1 , Andrea Armani a , Alessandra Feraco a , Massimo U. De Martino b , Andrea Fabbri c , 8 Giuseppe M.C. Rosano a , Massimiliano Caprio a,9 a Centre for Clinical and Basic Research, IRCCS San Raffaele Pisana, Rome, Italy 10 b Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy Q2 11 c Department of Internal Medicine, Endocrinology Unit, S. Eugenio & CTO A. Alesini Hospitals, University Tor Vergata, Rome, Italy 12 13 15 article info 16 Article history: 17 Received 1 February 2014 18 Received in revised form 29 April 2014 19 Accepted 2 May 2014 20 Available online xxxx 21 Keywords: 22 Mineralocorticoid receptor 23 Adipocyte 24 Adipose tissue 25 Macrophage 26 Insulin resistance 27 28 abstract 29 Aldosterone is the primary ligand for the mineralocorticoid receptor (MR) and has been considered long 30 time a ‘‘renal’’ hormone, acting at this site as a key regulator of plasma volume, electrolyte homeostasis 31 and blood pressure. A new exciting era of MR biology began with the identification of MR in different 32 non-epithelial tissues such as brain, heart, vessels, macrophages/monocytes, and adipose tissue. The dis- 33 tribution of MR in such a wide range of tissues has suggested novel and unexpected roles for MR, for 34 example in energy metabolism and inflammation. An increasing body of evidence suggests a detrimental 35 effect of aldosterone excess on the development of metabolic alterations. Disturbances in glucose metab- 36 olism due to inappropriate activation of MR are frequently observed in patients with primary aldosteron- 37 ism as well as in obese subjects. MR antagonists have beneficial effects on glucose tolerance and 38 metabolic parameters in experimental animals, whereas their role in humans remains unclear. The 39 aim of this review is to discuss the pathophysiology of MR activation in experimental models, particularly 40 at the level of adipocytes and macrophages, to discuss novel and sometimes contrasting insights from 41 emerging studies, and to highlight deficiencies in the field. 42 Ó 2014 Published by Elsevier Inc. 43 44 45 46 1. Introduction 47 Modern societies are plagued by obesity with its metabolic 48 complications. Together with autoimmunity and allergy disorders, 49 anxiety, insomnia, and depression, these diseases represent multi- 50 factorial polygenic disorders that are complex and influenced by 51 multiple mechanisms. Among them, it is not surprising that gluco- 52 corticoids (GCs), the main effectors of the stress system, have been 53 proposed to play a key role [1]. Glucocorticoid hormones are 54 secreted by the adrenal cortex under the strict control of the hypo- 55 thalamic–pituitary–adrenal (HPA) axis [1]. They exert their func- 56 tion in different target tissues by binding two intracellular 57 receptors: the glucocorticoid receptor (GR) and the mineralocorti- 58 coid receptor (MR) [2], and are able to regulate multiple metabolic 59 pathways, including glycolysis/gluconeogenesis, fatty acids and 60 cholesterol metabolism [3]. GC is potent inducers of hepatic gluco- 61 neogenesis [4] and through this activity they increase circulating 62 glucose and insulin, which results in insulin resistance, a hallmark 63 of metabolic syndrome [5]. 64 2. MR and GR: from a single corticosteroid receptor to the 65 acquisition of distinct functions 66 It is hypothesized that MR and GR descend from a single ances- 67 tral corticosteroid receptor, which gave rise to two distinct recep- 68 tors with different signaling functions [6]. This hypothesis may 69 explain the similar affinity of MR for aldosterone, its natural ligand, 70 and physiological glucocorticoids. While in most vertebrates GR is 71 specifically activated by the stress hormone cortisol to in turn reg- 72 ulate metabolism, inflammation, and immunity [7], the MR is acti- 73 vated by aldosterone to control electrolyte homeostasis and other 74 processes [8]. MR activation by cortisol is limited in epithelial tis- 75 sues by the presence of the cortisol-inactivating enzyme 11bHSD2 76 to allow selective aldosterone binding [8]. Interestingly, the capac- 77 ity to synthesize aldosterone has evolved relatively recently, and 78 the sensitivity of corticoid receptors to aldosterone is therefore 79 more ancient than the hormone itself. The evolution of an MR that 80 could be independently regulated by aldosterone enabled more http://dx.doi.org/10.1016/j.steroids.2014.05.001 0039-128X/Ó 2014 Published by Elsevier Inc. Corresponding author. Address: Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, Rome, Italy. Tel.: +39 06 5225 3419; fax: +39 06 5225 5668. E-mail address: [email protected] (M. Caprio). Steroids xxx (2014) xxx–xxx Contents lists available at ScienceDirect Steroids journal homepage: www.elsevier.com/locate/steroids STE 7562 No. of Pages 8, Model 5G 14 May 2014 Please cite this article in press as: Marzolla V et al. Mineralocorticoid receptor in adipocytes and macrophages: A promising target to fight metabolic syn- drome. Steroids (2014), http://dx.doi.org/10.1016/j.steroids.2014.05.001

Transcript of Mineralocorticoid receptor in adipocytes and macrophages: A promising target to fight metabolic...

1

3

4

5

6

7 Q1

8

910 Q211

1213

1 5

1617181920

21222324252627

2 8

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

Steroids xxx (2014) xxx–xxx

STE 7562 No. of Pages 8, Model 5G

14 May 2014

Contents lists available at ScienceDirect

Steroids

journal homepage: www.elsevier .com/locate /s teroids

Mineralocorticoid receptor in adipocytes and macrophages: A promisingtarget to fight metabolic syndrome

http://dx.doi.org/10.1016/j.steroids.2014.05.0010039-128X/� 2014 Published by Elsevier Inc.

⇑ Corresponding author. Address: Laboratory of Cardiovascular Endocrinology,IRCCS San Raffaele Pisana, Via di Val Cannuta 247, Rome, Italy. Tel.: +39 06 52253419; fax: +39 06 5225 5668.

E-mail address: [email protected] (M. Caprio).

Please cite this article in press as: Marzolla V et al. Mineralocorticoid receptor in adipocytes and macrophages: A promising target to fight metabodrome. Steroids (2014), http://dx.doi.org/10.1016/j.steroids.2014.05.001

Vincenzo Marzolla a, Andrea Armani a, Alessandra Feraco a, Massimo U. De Martino b, Andrea Fabbri c,Giuseppe M.C. Rosano a, Massimiliano Caprio a,⇑a Centre for Clinical and Basic Research, IRCCS San Raffaele Pisana, Rome, Italyb Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italyc Department of Internal Medicine, Endocrinology Unit, S. Eugenio & CTO A. Alesini Hospitals, University Tor Vergata, Rome, Italy

a r t i c l e i n f o

293031323334353637383940

Article history:Received 1 February 2014Received in revised form 29 April 2014Accepted 2 May 2014Available online xxxx

Keywords:Mineralocorticoid receptorAdipocyteAdipose tissueMacrophageInsulin resistance

414243

a b s t r a c t

Aldosterone is the primary ligand for the mineralocorticoid receptor (MR) and has been considered longtime a ‘‘renal’’ hormone, acting at this site as a key regulator of plasma volume, electrolyte homeostasisand blood pressure. A new exciting era of MR biology began with the identification of MR in differentnon-epithelial tissues such as brain, heart, vessels, macrophages/monocytes, and adipose tissue. The dis-tribution of MR in such a wide range of tissues has suggested novel and unexpected roles for MR, forexample in energy metabolism and inflammation. An increasing body of evidence suggests a detrimentaleffect of aldosterone excess on the development of metabolic alterations. Disturbances in glucose metab-olism due to inappropriate activation of MR are frequently observed in patients with primary aldosteron-ism as well as in obese subjects. MR antagonists have beneficial effects on glucose tolerance andmetabolic parameters in experimental animals, whereas their role in humans remains unclear. Theaim of this review is to discuss the pathophysiology of MR activation in experimental models, particularlyat the level of adipocytes and macrophages, to discuss novel and sometimes contrasting insights fromemerging studies, and to highlight deficiencies in the field.

� 2014 Published by Elsevier Inc.

44

62

63

64

65

66

67

68

69

70

71

72

73

74

75

1. Introduction

Modern societies are plagued by obesity with its metaboliccomplications. Together with autoimmunity and allergy disorders,anxiety, insomnia, and depression, these diseases represent multi-factorial polygenic disorders that are complex and influenced bymultiple mechanisms. Among them, it is not surprising that gluco-corticoids (GCs), the main effectors of the stress system, have beenproposed to play a key role [1]. Glucocorticoid hormones aresecreted by the adrenal cortex under the strict control of the hypo-thalamic–pituitary–adrenal (HPA) axis [1]. They exert their func-tion in different target tissues by binding two intracellularreceptors: the glucocorticoid receptor (GR) and the mineralocorti-coid receptor (MR) [2], and are able to regulate multiple metabolicpathways, including glycolysis/gluconeogenesis, fatty acids andcholesterol metabolism [3]. GC is potent inducers of hepatic gluco-neogenesis [4] and through this activity they increase circulating

glucose and insulin, which results in insulin resistance, a hallmarkof metabolic syndrome [5].

76

77

78

79

80

2. MR and GR: from a single corticosteroid receptor to theacquisition of distinct functions

It is hypothesized that MR and GR descend from a single ances-tral corticosteroid receptor, which gave rise to two distinct recep-tors with different signaling functions [6]. This hypothesis mayexplain the similar affinity of MR for aldosterone, its natural ligand,and physiological glucocorticoids. While in most vertebrates GR isspecifically activated by the stress hormone cortisol to in turn reg-ulate metabolism, inflammation, and immunity [7], the MR is acti-vated by aldosterone to control electrolyte homeostasis and otherprocesses [8]. MR activation by cortisol is limited in epithelial tis-sues by the presence of the cortisol-inactivating enzyme 11bHSD2to allow selective aldosterone binding [8]. Interestingly, the capac-ity to synthesize aldosterone has evolved relatively recently, andthe sensitivity of corticoid receptors to aldosterone is thereforemore ancient than the hormone itself. The evolution of an MR thatcould be independently regulated by aldosterone enabled more

lic syn-

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

2 V. Marzolla et al. / Steroids xxx (2014) xxx–xxx

STE 7562 No. of Pages 8, Model 5G

14 May 2014

specific endocrine responses, developing new functions for cortisolthat could be controlled separately to the GR-mediated responses[6]. In such context, it is possible that the mineralocorticoid systemmay have later developed important functions also in extra-renalsites, involving the regulation of energy homeostasis, tissueinflammation and fibrosis, and creating several overlaps withactions classically defined as GR-mediated.

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

3. Mineralocorticoids and glucocorticoids: two convergingsystems controlling glucose metabolism

MR and GR activation, despite major differences in the regula-tion of tissue inflammation [9], exerts converging effects in thecontrol of glucose homeostasis and insulin resistance. Importantly,chronic exposure to glucocorticoid excess in humans, as seen inCushing syndrome, leads to a clinical phenotype harboring all com-ponents of the metabolic syndrome, including hypertension, vis-ceral obesity and hyperglycemia (extensively reviewed by DiDalmazi et al.). Differently, chronic exposure to mineralocorticoids,as seen in primary aldosteronism, may also be associated with dia-betes mellitus and glucose intolerance [10], but displays a moresubtle and still controversial effect on glucose metabolism. Theexact mechanism that links glucose intolerance and aldosteroneexcess is still unclear. However, hypokalemia may be at least partlyresponsible and aldosterone may directly inhibit insulin secretionby the pancreas [11]. A study by Fallo et al. describes a higher prev-alence of metabolic syndrome in patients with primary aldosteron-ism compared to patients with essential hypertension [12], withrelevant differences in glucose metabolism between patientgroups. The association between hyperaldosteronism, insulin resis-tance and insulin deficiency has been recently reviewed [13], withclear evidence for a strict relationship between aldosterone and theincreased risk in metabolic syndrome [14]. Importantly, excessaldosterone has been shown also in obesity as an epiphenomenonof an increased fat mass, and weight loss after gastric banding hasbeen demonstrated to reduce aldosterone levels along with areduction in blood pressure and insulin resistance [15]. Such a rela-tionship has been confirmed in an experimental model of life-longobesity in rodents, where exposure to HF diet from weaning leadsto hypertension and hyperaldosteronism [16]. In summary, popu-lation-based observational studies suggest a clear correlationbetween obesity and aldosterone levels, while experimental stud-ies have shown that adipocyte-related factors are possible atypical‘‘triggers’’ for aldosterone release from adrenal cells [17,18], whichmight explain the hyperaldosteronism often observed in obesesubjects. Finally, recent reports seem to support the hypothesisthat adipocytes are able to produce and secrete per se modestamounts of aldosterone, which may contribute to enhance totalcirculating aldosterone levels [19] and the higher degree of MRactivation in obesity.

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

4. Are MR blockers protective in insulin resistance and obesity?

Beneficial effects of MR antagonists have been demonstrated inseveral animal models of genetic and diet-induced obesity, but stillremain to be proven in human insulin resistance states. Interest-ingly, Guo et al. reported that systemic MR antagonism witheplerenone reversed the adverse metabolic consequences ofgenetic obesity in db/db mice, improving glucose tolerance, insulinresistance and adipokine expression in adipose tissue [20]. Similareffects were obtained in a model of diet-induced obesity, whereMR blockade induced a marked reduction of hypertrophicadipocytes and macrophage infiltration with a concomitantdecrease of pro-inflammatory adipokine levels in adipose tissue[21]. Importantly, our group recently showed that pharmacological

Please cite this article in press as: Marzolla V et al. Mineralocorticoid receptor idrome. Steroids (2014), http://dx.doi.org/10.1016/j.steroids.2014.05.001

MR antagonism with either spironolactone (Spiro, 7a-acetiltio-3-osso-17a-pregn-4-ene-21,17-carbolattone) or drospirenone (DRSP,6b,7b,15b,16b-dimethylene-3-foxo-17a-pregn-4-ene-21,17-carbo-lactone, a potent synthetic antimineralocorticoid with progesto-genic and moderate antiandrogenic properties) was able tocounter metabolic dysfunctions in a model of high-fat diet inducedobesity in female mice, and that beneficial effects on body fat com-position and glucose tolerance were associated to ‘‘browning’’ ofadipose tissue [22]. It is worthy to remark that different MR block-ers display distinct pharmacokinetic profiles. Therefore, the effectof MR blockade on metabolic parameters may vary by the MRantagonist applied. In fact, Homma et al. demonstrated that Spiro,but not eplerenone, negatively affected parameters of glucosemetabolism in a rat model of metabolic syndrome. These dataare in contrast with most of the current literature, and were likelyexplained by the increased aldosterone levels induced by Spiro, aswell as by possible species differences between rat and mice [23].

Clinical studies addressing the potential impact of MR blockadeon metabolic parameters are limited, since the largest clinical trialswith MR blockers have been conducted in the clinical setting ofheart failure [24–27]. Particular attention has been raised by clin-ical trials of oral contraceptives containing estrogens conjugated toDRSP. The relative binding affinity of DRSP for the human MR wasdemonstrated to be two to five times higher than that of aldoste-rone [28]. Given these pharmacological properties, DRSP has beenshown to display favorable effects on blood pressure [29–31] andbody weight [32–34]. In addition to its effects on salt and waterretention, a study conducted in healthy postmenopausal womenshowed that DRSP, in combination with estradiol, caused a signif-icant decrease in central fat mass and central fat mass/peripheralfat mass, as measured by dual-energy X-ray absorptiometry [35].We recently demonstrated that DRSP strongly inhibits adipose dif-ferentiation in murine and in human primary preadipocytes,through specific antagonism on the MR [36], providing new mech-anistic insights that may explain its aforementioned favorablemetabolic effects in clinical settings.

However, major concerns for the use of DRSP were raised aftercareful evaluation of the risk of non-fatal idiopathic venous throm-boembolism in women using oral contraceptives containing DRSP:use of the DRSP contraceptive was associated with a threefoldhigher risk of non-fatal idiopathic venous thromboembolism com-pared with levonorgestrel, in a study based on data from the UKGeneral Practice Research Database [37]. Similar results emergedfrom a study based on United States claims data [38], and discour-aged any further attempt to investigate the potential beneficialeffects of DRSP in clinical trials.

Divergent conclusions were suggested by the CHARM study, inwhich spironolactone therapy showed significant positive associa-tions with the development of diabetes in a population affected bychronic heart failure [39]. However, interventional studies specifi-cally designed to test the effects of MR antagonists on glucose andlipid metabolism are limited.

A recent work [40] conducted in healthy adult males did notshow any significant effect of a low, non-blood pressure-modifyingdose of eplerenone, on basal and postprandial glucose and lipidlevels. This study suggests that it may be necessary to study pop-ulations where the MR is inappropriately activated, in order toevaluate the potential favorable effects of MR antagonists on met-abolic function. However, recent work from the same group con-firmed the lack of beneficial effects of spironolactone on insulinsensitivity and endothelial function in a small population of nor-motensive obese patients with no other comorbidities [41], sug-gesting that excess MR activation does not significantlycontribute to changes in insulin resistance or endothelial dysfunc-tion induced by early obesity. Finally, a nicely designed study byDerosa et al. evaluated the effects of canrenoate on metabolic

n adipocytes and macrophages: A promising target to fight metabolic syn-

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

V. Marzolla et al. / Steroids xxx (2014) xxx–xxx 3

STE 7562 No. of Pages 8, Model 5G

14 May 2014

parameters of patients affected by metabolic syndrome, but with-out overt diabetes or cardiovascular diseases. In this work, treat-ment with MR antagonist did not affect body mass index orwaist circumference, but significantly reduced insulin resistance,as measured by fasting plasma insulin and HOMA index, and sys-temic inflammation (C-reactive protein; CRP and tumor necrosisfactor-a; TNF-a) [42]. To date, evidence from available studies inhumans are divergent, due to the heterogeneity of the populationsexamined and the different treatment protocols. There is indeed anurgent need for interventional studies conducted on carefullyselected populations, in order to understand the real potential ofMR antagonists for the prevention or amelioration of insulin resis-tance and other metabolic disturbances in different clinicalconditions.

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

5. Pathophysiological role of MR in the adipose

5.1. MR in white adipose tissue (WAT) metabolism and plasticity

Since the discovery of MR expression in murine and human adi-pose tissue, significant progress has been made in the understand-ing of the role of MR in adipocyte biology. Several reports indicatethat MR affects adipocyte function. In vitro studies showed thataldosterone as well as glucocorticoids promote murine and humanwhite preadipocyte differentiation through MR [36,43–45]. In par-ticular, we recently demonstrated that pharmacological blockadeof MR strongly inhibits 3T3-L1 adipocyte clonal expansion anddifferentiation by reducing the expression of peroxisome prolifer-ator-activated receptor gamma (PPARc), the master regulator ofadipogenesis [36]. Moreover, knock-down of MR by specific siRNAin primary human preadipocyte markedly inhibited adipocyte dif-ferentiation in vitro [46].

Obese individuals develop insulin resistance, characterized by areduced ability of insulin to inhibit the production of glucose fromthe liver and to promote glucose uptake in adipose tissue andskeletal muscle [47,48]. In obesity, impaired insulin sensitivity is

Hypertrophic White Adipocyte Functional White Adip

Brown-like Adipo

MR BLOCKA

White Adipocyte

Fig. 1. Schematic representation of the effects of MR blockade on adipocyte and macropreduce hypertrophic white adipocytes and increase small white and brown-like adipoadipokines and adipogenesis-related genes resulting in reduction of adipocyte size. (B) Ibrown adipocyte transcriptional profile, favoring a phenotypic change into a brown-likgenetic deletion and pharmacological antagonism in vitro are able to induce a switchinterpretation of the references to color in this figure legend, the reader is referred to th

Please cite this article in press as: Marzolla V et al. Mineralocorticoid receptor idrome. Steroids (2014), http://dx.doi.org/10.1016/j.steroids.2014.05.001

associated with adipocyte hypertrophy [49]. Hypertrophic adipo-cytes are characterized by increased expression of adipokines withpro-inflammatory activity such as leptin, interleukin-6 (IL-6), plas-minogen activator inhibitor-1 (PAI-1), TNF-a and monocyte che-moattractant protein 1 (MCP-1). On the other hand, theexpression of anti-inflammatory adipokines i.e. adiponectin andinterleukin-10 (IL-10) is reduced in enlarged adipocytes [50–52].It is well known that adiponectin is an insulin-sensitizing factorwith major role in controlling glucose and lipid homeostasis[53,54]. Interestingly, Guo et al. reported that aldosteroneincreases mRNA levels of TNF-a, MCP-1, and IL-6 and decreasesmRNA and protein levels of adiponectin in 3T3-L1 preadipocytes[20]. Local increase in pro-inflammatory adipokines, in particularMCP-1, contributes to macrophage infiltration within adipose tis-sue. Accordingly, in mouse models of genetic or diet-induced obes-ity, MR blockade induced a marked reduction of hypertrophicadipocytes and macrophage infiltration with a concomitantdecrease of pro-inflammatory adipokine levels in white adiposetissue (WAT) [20,21] (Figs. 1 and 2). Such effects suggest that spe-cific MR blockade in adipocytes could explain, at least in part,improvements in glucose homeostasis observed in mice treatedwith MR antagonists.

In contrast to the above-mentioned literature, Kuhn et al. haverecently described a paradoxical response to high fat diet in trans-genic mice globally over-expressing human MR. Global overexpression of human MR had beneficial metabolic effects in termsof decreased fat mass and concomitant improved glucose tolerancecompared to wild type mice. White preadipocytes derived frommice over-expressing human MR did not display impairedadipogenesis in vitro, suggesting that reduced adipocyte size thatwas observed in vivo did not depend on intrinsic defects in thecapability to differentiate of MR over-expressing adipocytes. Theauthors showed that although adipose tissue macrophage numberdid not differ between genotypes, transgenic mice displayed adistinct macrophage polarization phenotype, and demonstratedthat transgenic mice macrophage-conditioned media impairedpreadipocyte differentiation. This suggests that diffusible signaling

ocyte

cyte

Macrophage M2

Macrophage M1

DE

A

B

C

hage phenotypic transition. (A) In murine models of obesity, MR blockade is able tocytes abundance in fat depots. Adipocyte MR blockade modulates expression ofn white adipocytes, MR antagonism promotes gene expression transition toward ae adipocyte. (C) MR modulates macrophage polarization: macrophage-specific MR

from a pro-inflammatory (M1) into an anti-inflammatory (M2) phenotype. (Fore web version of this article.)

n adipocytes and macrophages: A promising target to fight metabolic syn-

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

MCP-1IL-6TNF-αIL-10

MCP-1IL-6TNF-αPAILEPTINADIPONECTIN

INSULIN RESISTANCEINFLAMMATION

MR ACTIVATION

MONOCYTES

MR BLOCKADE

MCP-1IL-6TNF-αPAILEPTINADIPONECTIN

INSULIN SENSITIVITYINFLAMMATION

MCP-1IL-6TNF-αIL-10

ICAM-1VCAM-1

ICAM-1VCAM-1

A B

Fig. 2. Schematic overview summarizing the role of MR in the adipose organ. (A) In obesity, adipocyte MR activation causes an increase of hypertrophic adipocytes and amarked infiltration of macrophages (M1, red cells). Adipose tissue macrophages and hypertrophic adipocytes produce an increased release of pro-inflammatory cytokines(TNF-a, IL-6, and MCP-1) leading to insulin resistance. The increase in leptin accompanied by a reduction in anti-inflammatory mediators (IL-10 and adiponectin) contributesto a state of chronic low-grade inflammation in the adipose organ. The increase in macrophage infiltration is also due to the increased expression of adhesion molecules(ICAM-1, VCAM-1 and E-SELECTIN) of the blood vessels supplying the adipose organ due to the activation of the MR. (B) In mouse models of genetic or diet-induced obesity,MR blockade counteracts the development of hypertrophic adipocytes and increases the presence of brown-like adipocytes (gray cells) in WAT. Moreover, MR antagonisminduces a switch in the polarization of resident tissue macrophages from M1 (red cells) to M2 (cyan cells), resulting in reduced release of pro-inflammatory molecules andincreased production of anti-inflammatory molecules (adiponectin, IL-10) by macrophages and adipocytes. (For interpretation of the references to color in this figure legend,the reader is referred to the web version of this article.)

4 V. Marzolla et al. / Steroids xxx (2014) xxx–xxx

STE 7562 No. of Pages 8, Model 5G

14 May 2014

molecules derived from macrophages may control adipocyte dif-ferentiation in vivo, although no information is provided aboutthe precise nature of the interactions between macrophages andadipocytes resulting in these unexpected effects on adipose tissue[55]. Such paradoxical effects on adipose tissue and metabolichomeostasis are indeed difficult to interpret, and probably relyon non-specific effects due to global human MR over-expression,which may lead to altered metabolism in these mice under basalcondition [56]. Increased expression of MR in the central nervoussystem might also affect responses to metabolic stress.

It is well established that obesity and insulin resistance areassociated to the hyperactivity of the renin-angiotensin-aldoste-rone system (RAAS), resulting in elevated plasma aldosterone lev-els [57] that could activate adipocyte MR and promote adiposedifferentiation and expansion [58]. Adipose tissue itself has beenshown to stimulate aldosterone production from adrenal glandsby releasing soluble bioactive factors [18,59–61]. Moreover, ithas been suggested that the hypertrophied adipocyte producesaldosterone which can stimulate adipose differentiation throughan autocrine loop [19]. Notably, increased aldosterone levels areable to activate endothelial MR promoting expression of adhesionmolecules such as intracellular adhesion molecule 1 (ICAM1) [62]and vascular cell adhesion molecule 1 (VCAM1) [63], resulting inincreased endothelial adhesion of monocytes that leave the bloodstream and migrate into adipose depots, hence contributing toinflammation and dysfunction of adipose tissue (Fig. 2).

Please cite this article in press as: Marzolla V et al. Mineralocorticoid receptor idrome. Steroids (2014), http://dx.doi.org/10.1016/j.steroids.2014.05.001

In summary, the major lines of evidences suggest that hyper-activation of MR due to glucocorticoids or aldosterone results indeleterious effects on adipose tissue function and glucose homeo-stasis. Further studies with mice lacking adipocyte MR expressionare deemed necessary to elucidate the role of adipocyte MR inmodulating metabolism of adipose cells and other types of cellspresent in adipose tissue (macrophages, endothelial cells, etc.) aswell as glucose and lipid metabolism.

6. MR antagonism in adipocytes and browning of WAT

Despite many studies on MR function in WAT, there are onlyfew reports focusing on the role of MR in brown adipose tissue(BAT) physiology. In mammals, WAT and BAT display different his-tological structure as well as distinct physiological roles. WAT hasnot only the capacity to store energy but it is capable of influencingmetabolic activity of the whole body secreting a large number ofadipokines [64]. Conversely, BAT evolved in mammals to dissipatechemical energy in the form of heat, a process termed not-shiver-ing thermogenesis which is required to maintain stable body tem-perature in a cold environment [65].

Cold exposure is the main stimulus for BAT activity. Such effectsare mediated by activation of the sympathetic nervous system,resulting in the stimulation of b-adrenergic receptors present inbrown adipocytes with increase in uncoupling protein 1 (UCP1)

n adipocytes and macrophages: A promising target to fight metabolic syn-

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

V. Marzolla et al. / Steroids xxx (2014) xxx–xxx 5

STE 7562 No. of Pages 8, Model 5G

14 May 2014

activity [66,67]. Apart from classic brown adipocytes found ininterscapular adipose tissue, thermogenic adipocytes (calledbrown-like or beige or brite adipocytes) [68,69] are also presentin WAT depots and their abundance in mice is strain- and loca-tion-dependent and increases in response to cold or b3-adrenergicstimuli [70]. Increase of thermogenic adipocytes under specificstimuli in WAT is known as ‘‘browning’’. Stanford and collaboratorshave shown that BAT transplantation, in mice fed either a high fatdiet or a normal chow, results in improved glucose metabolism andreduction in body weight and fat mass [71]. Importantly, recentreports indicate that functional BAT is present in adult humansand that its activity positively correlates with body mass index,suggesting a protective role against obesity [72–75].

Brilliant studies using transgenic mice have shown that MRgene is expressed and active in BAT [76]. Subsequent investigationsdemonstrated that MR activation in T37i cells, derived from ahibernoma expressing the SV40 large T antigen under the controlof the P1 promoter of the human MR, up-regulates general markersof adipocyte differentiation [(PPAR-c and adipocyte-specific fattyacid binding protein 2 (aP2)], but represses expression of UCP1,the hallmark of brown adipocytes [77,78].

Notably, recent data from our laboratory [22] have shown thatpharmacological MR antagonism, in female mice fed a high fat diet,induces browning of WAT through a direct control of autophagicrate in adipose tissue, hence promoting an increase in metabolicactivity of WAT depots and interscapular BAT; such effects couldaccount for the improvement in glucose homeostasis alreadyobserved in other studies [20,21,79]. Additional in vivo studiesusing adipocyte MR null-mice are mandatory to better understandthe role of MR in the context of adipocyte metabolism and brown-ing of WAT.

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

7. Role of MR in macrophages

7.1. Inflammation and macrophage polarization

It is well established that macrophage-mediated inflammationin adipose tissue contributes to the development of insulin resis-tance [80]. The number of macrophages in adipose tissue of obesemice is markedly higher compared to lean animals, with a preva-lent pro-inflammatory (M1) polarization. M1 polarized macro-phages release pro-inflammatory cytokines [TNF-a, interleukin 1beta (IL-1b), etc.] that inhibit insulin signaling through phosphory-lation of insulin receptor substrate (IRS) proteins [81]. In contrast,anti-inflammatory (M2) polarized macrophages are predominantin fat tissue of lean mice and release interleukin 10 (IL-10), whichprevents the detrimental effects of TNF-a on insulin signaling [82].Notably, under metabolic stress, the presence of pro-inflammatorymacrophages has also been detected in skeletal muscle and liverwhere M1 polarization leads to insulin resistance [83,84]. In suchcontext, a deeper knowledge of pathways affecting polarizationof the macrophage could help to design pharmacological strategiesaimed at preventing M1 activation and subsequent impairment ofinsulin sensitivity.

Interestingly, mice with macrophage-specific deletion of theMR gene displayed protection against cardiac fibrosis induced bypharmacological treatment [85–87], with reduction in M1 macro-phage markers [87] and parallel increase in M2 macrophage mark-ers in cardiac tissue [86]. These data confirmed a role for MR inregulating macrophage polarization and, in turn, inflammatoryresponse and cardiac remodeling [85–87]. In vitro treatment ofhuman and murine macrophages with MR antagonists confirmedthat MR blockade represses expression of M1 markers andpromotes M2 polarization [86,88] (Fig. 1). Accordingly, stimulationof MR function with aldosterone has been shown to increase

Please cite this article in press as: Marzolla V et al. Mineralocorticoid receptor idrome. Steroids (2014), http://dx.doi.org/10.1016/j.steroids.2014.05.001

lipopolysaccharide (LPS)-induced TNF-a expression, while co-treatment with eplerenone prevented LPS-induced expression ofTNF-a [89] in primary cultures of rat hepatic macrophages (knownas Kupffer cells) and mouse bone-marrow-derived macrophages.Inhibition of Kupffer cell and macrophage MR with eplerenonewas suggested to prevent the development of non-alcoholic steato-hepatitis in a mouse model of metabolic syndrome [89].

Interestingly, the effects of macrophage-specific deletion of MRhave not been explored in animal models of obesity. In obesity andmetabolic syndrome, increased levels of aldosterone and glucocor-ticoids are able to stimulate MR activity of resident macrophages inadipose tissue, hence inducing M1 activation and, in turn, insulinresistance. Considering that MR antagonists find wide applicationin clinical practice in patients with heart failure [90], it could bechallenging to investigate the metabolic effects of MR antagonistson macrophage polarization and glucose metabolism also in obesesubjects.

Surprisingly, the recent data by Kuhn et al. [55] show that adi-pose tissue macrophages derived from mice overexpressing thehuman MR display reduced M1 pro-inflammatory polarization,without any significant changes in M2 markers, and propose thatsuch effect, although of modest entity, could be responsible forthe protection against high fat-induced obesity. Elegant co-cultureexperiments in this study indicated that macrophages derivedfrom MR over-expressing mice are able to repress adipocyte differ-entiation in preadipocytes extracted from wild-type mice, througha modest reduction of the adipogenic factor PPAR-c and the lipo-genic enzyme fatty acid synthase (FAS).

Again in contrast with this work, most available studies indicatethat MR blockade in macrophages suppresses the pro-inflamma-tory M1 phenotype with beneficial effects in terms of attenuatedfibrotic responses and protection from high blood pressure. Furtherstudies will be required to better elucidate the complex role playedby macrophage MR in modulating adipocyte metabolism, plasticityand proinflammatory profile. Considering that macrophage polari-zation affects inflammation of adipose tissue which, in turn, repre-sents a hallmark of obesity, the design of new antagonists specificfor macrophage MR may represents a potentially valid tool to fightadipocyte dysfunction (Fig. 2).

8. Macrophage polarization and ‘‘browning’’ of WAT

Elegant data by Nguyen et al. have showed that macrophagepolarization affects metabolic changes occurring in fat depots dur-ing adaptive thermogenesis in mice [91]. In mice exposed to cold,increased expression of alternative/M2 polarization markers (Arg1,Mrc1and Clec10a) was detected in BAT and WAT, whereas classi-cal/M1 polarization markers were not modulated. Notably, coldexposure did not induce alternative macrophage polarization inskeletal muscle and liver [91].

Interleukin-4 (IL-4) and interleukin-13 (IL-13) are known toinduce M2 polarization [92–95]. whereas Signal Transducer andActivator of Transcription 6 (STAT6) mediates interleukin 4 and13 (IL-4 and IL-13) effects on macrophage polarization [96]. Ngu-yen et al. observed that cold exposure was not able to induce alter-native macrophage polarization in IL-4/IL-13–/– as well as inSTAT6–/– mice [91]. Importantly, both mouse models exposed tocold displayed a reduced capacity to induce thermogenic genesin BAT and reduced mobilization of fatty acids in WAT. Moreover,the authors showed that alternative activation of macrophagespresent in BAT and WAT leads to release of catecholamines bymacrophages themselves, which in turn activates a thermogenicgene program and lipolysis in fat depots [91]. These data demon-strate for the first time that macrophages play a major role in theregulation of thermogenic adaptation. Modulation of macrophage

n adipocytes and macrophages: A promising target to fight metabolic syn-

452

453

454

455

456

457

458

459

460

461

462

463

464

465

466

467

468

469

470

471

472

473

474

475

476

477

478

479

480 Q3Q4

481

482

483

484

485486487488489490491492493494495496497498499500501502503504505506507508509510511512513514515516517518

519520521522

6 V. Marzolla et al. / Steroids xxx (2014) xxx–xxx

STE 7562 No. of Pages 8, Model 5G

14 May 2014

MR may then regulate such complex phenomenon, given its abilityto affect M1/M2 polarization, and in particular to control theiralternative activation.

523524525526527528529530531532533534535536537538539540541542543544545546547548549550551552553554555556557Q5558559560561562563564565566567568569570571572573574575576577578579580581582583584585586587588589590591592593594595596597598599600601602603604

9. Conclusions

In humans, modest or contradictory effects of pharmacologicalMR antagonism have been observed on adipose metabolic param-eters. In contrast, MR antagonists clearly improve glucose toler-ance and lipid parameters in different mouse models of obesity.The reasons for these differences remain obscure and difficult tointerpret. Appropriate and properly designed metabolic studiesfocused on insulin resistance and weight gain in obese humans,using different type, dose and duration of treatment with MRantagonists, will be necessary to better characterize the effects ofMR antagonists and explain the discrepancies between humanand murine models. The specific contribution of MR in adipocytesand macrophages in the regulation of adipose tissue adaptation tometabolic distress, represents a central issue that needs furtherinvestigation. Comparative studies with macrophage- or adipo-cyte-specific MR null mice may provide precious information forbetter understanding the role of MR in the development of dys-functional adipocytes.

Indeed, MR modulates the inflammatory status of the macro-phage, which in turn markedly affects adipose tissue metabolism(Fig. 2). For this reason, a better understanding of the mechanismsby which adipocyte-macrophage MR inhibition improves meta-bolic function, could identify new molecular targets to treat adipo-cyte dysfunction, obesity and its metabolic complications.

Acknowledgments

This work was supported by a grant from Ministero della Salute(Bando Giovani Ricercatori 2009, to MC) and by institutional fun-dings from University Tor Vergata (Progetti Ricerca Interesse Naz-ionale Ministero dell’Università e della Ricerca 2009, to AF).

References

[1] Chrousos GP. Stress and disorders of the stress system. Nat Rev Endocrinol2009;5:374–81.

[2] Funder JW. Glucocorticoid and mineralocorticoid receptors: biology andclinical relevance. Annu Rev Med 1997;48:231–40.

[3] Di Dalmazi G, Pagotto U, Pasquali R, Vicennati V. Glucocorticoids and type 2diabetes: from physiology to pathology. J Nutr Metab 2012;2012:525093.

[4] Pilkis SJ, Granner DK. Molecular physiology of the regulation of hepaticgluconeogenesis and glycolysis. Annu Rev Physiol 1992;54:885–909.

[5] Boscaro M, Giacchetti G, Ronconi V. Visceral adipose tissue: emerging role ofgluco-and mineralocorticoid hormones in the setting of cardiometabolicalterations. Ann N Y Acad Sci 2012;1264:87–102.

[6] Bridgham JT, Carroll SM, Thornton JW. Evolution of hormone-receptorcomplexity by molecular exploitation. Science 2006;312:97–101.

[7] Chrousos GP, Kino T. Glucocorticoid signaling in the cell. Expanding clinicalimplications to complex human behavioral and somatic disorders. Ann N YAcad Sci 2009;1179:153–66.

[8] Farman N, Rafestin-Oblin ME. Multiple aspects of mineralocorticoid selectivity.Am J Physiol Renal Physiol 2001;280:F181–92.

[9] Chantong B, Kratschmar DV, Nashev LG, Balazs Z, Odermatt A.Mineralocorticoid and glucocorticoid receptors differentially regulate NF-kappaB activity and pro-inflammatory cytokine production in murine BV-2microglial cells. J Neuroinflammation 2012;9:260.

[10] Conn JW. Hypertension, the potassium ion and impaired carbohydratetolerance. N Engl J Med 1965;273:1135–43.

[11] Shimamoto K, Shiiki M, Ise T, Miyazaki Y, Higashiura K, Fukuoka M, Hirata A,Masuda A, Nakagawa M, Iimura O. Does insulin resistance participate in animpaired glucose tolerance in primary aldosteronism? J Hum Hypertens1994;8:755–9.

[12] Fallo F, Veglio F, Bertello C, Sonino N, Della MP, Ermani M, Rabbia F, FederspilG, Mulatero P. Prevalence and characteristics of the metabolic syndrome inprimary aldosteronism. J Clin Endocrinol Metab 2006;91:454–9.

[13] Ronconi V, Turchi F, Appolloni G, di T V, Boscaro M, Giacchetti G. Aldosterone,mineralocorticoid receptor and the metabolic syndrome: role of themineralocorticoid receptor antagonists. Curr Vasc Pharmacol 2012;10:238–46.

Please cite this article in press as: Marzolla V et al. Mineralocorticoid receptor idrome. Steroids (2014), http://dx.doi.org/10.1016/j.steroids.2014.05.001

[14] Ingelsson E, Pencina MJ, Tofler GH, Benjamin EJ, Lanier KJ, Jacques PF, Fox CS,Meigs JB, Levy D, Larson MG, Selhub J, D’Agostino Sr RB, Wang TJ, Vasan RS.Multi marker approach to evaluate the incidence of the metabolic syndromeand longitudinal changes in metabolic risk factors: the Framingham OffspringStudy. Circulation 2007;116:984–92.

[15] Dall’Asta C, Vedani P, Manunta P, Pizzocri P, Marchi M, Paganelli M, Folli F,Pontiroli AE. Effect of weight loss through laparoscopic gastric banding onblood pressure, plasma renin activity and aldosterone levels in morbid obesity.Nutr Metab Cardiovasc Dis 2009;19:110–4.

[16] Northcott CA, Fink GD, Garver H, Haywood JR, Laimon-Thomson EL, McClain JL,Pires PW, Rainey WE, Rigsby CS, Dorrance AM. The development ofhypertension and hyperaldosteronism in a rodent model of life-long obesity.Endocrinology 2012;153:1764–73.

[17] Marzolla V, Armani A, Zennaro MC, Cinti F, Mammi C, Fabbri A, Rosano GM,Caprio M. The role of the mineralocorticoid receptor in adipocyte biology andfat metabolism. Mol Cell Endocrinol 2012;350:281–8.

[18] Ehrhart-Bornstein M, Lamounier-Zepter V, Schraven A, Langenbach J,Willenberg HS, Barthel A, Hauner H, McCann SM, Scherbaum WA, BornsteinSR. Human adipocytes secrete mineralocorticoid-releasing factors. Proc NatlAcad Sci U S A 2003;100:14211–6.

[19] Briones AM, Nguyen Dinh CA, Callera GE, Yogi A, Burger D, He Y, Correa JW,Gagnon AM, Gomez-Sanchez CE, Gomez-Sanchez EP, Sorisky A, Ooi TC, RuzickaM, Burns KD, Touyz RM. Adipocytes produce aldosterone through calcineurin-dependent signaling pathways: implications in diabetes mellitus-associatedobesity and vascular dysfunction. Hypertension 2012;59:1069–78.

[20] Guo C, Ricchiuti V, Lian BQ, Yao TM, Coutinho P, Romero JR, Li J, Williams GH,Adler GK. Mineralocorticoid receptor blockade reverses obesity-relatedchanges in expression of adiponectin, peroxisome proliferator-activatedreceptor-gamma, and proinflammatory adipokines. Circulation2008;117:2253–61.

[21] Hirata A, Maeda N, Hiuge A, Hibuse T, Fujita K, Okada T, Kihara S, Funahashi T,Shimomura I. Blockade of mineralocorticoid receptor reverses adipocytedysfunction and insulin resistance in obese mice. Cardiovasc Res2009;84:164–72.

[22] Armani A, Cinti F, Marzolla V, Morgan J, Cranston GA, Antelmi A, Carpinelli G,Canese R, Pagotto U, Quarta C, Malorni W, Matarrese P, Marconi M, Rosano G,Fabbri A, Young MJ, Caprio M. Mineralocorticoid receptor antagonism inducesbrowning of white adipose tissue through impairment of autophagy andprevents adipocyte dysfunction in high fat diet-fed mice. FASEB J 2014.

[23] Homma T, Fujisawa M, Arai K, Ishii M, Sada T, Ikeda M. Spironolactone, but noteplerenone, impairs glucose tolerance in a rat model of metabolic syndrome. JVet Med Sci 2012;74:1015–22.

[24] Pitt B. Effect of aldosterone blockade in patients with systolic left ventriculardysfunction: implications of the RALES and EPHESUS studies. Mol CellEndocrinol 2004;217:53–8.

[25] Pitt B, Reichek N, Willenbrock R, Zannad F, Phillips RA, Roniker B, Kleiman J,Krause S, Burns D, Williams GH. Effects of eplerenone, enalapril, andeplerenone/enalapril in patients with essential hypertension and leftventricular hypertrophy: the 4E-left ventricular hypertrophy study.Circulation 2003;108:1831–8.

[26] Pitt B, Remme W, Zannad F, Neaton J, Martinez F, Roniker B, Bittman R, HurleyS, Kleiman J, Gatlin M. Eplerenone, a selective aldosterone blocker, in patientswith left ventricular dysfunction after myocardial infarction. N Engl J Med2003;348:1309–21.

[27] Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, Perez A, Palensky J, Wittes J.The effect of spironolactone on morbidity and mortality in patients withsevere heart failure. Randomized Aldactone Evaluation Study Investigators. NEngl J Med 1999;341:709–17.

[28] Fuhrmann U, Krattenmacher R, Slater EP, Fritzemeier KH. The novel progestindrospirenone and its natural counterpart progesterone: biochemical profileand antiandrogenic potential. Contraception 1996;54:243–51.

[29] White WB, Pitt B, Preston RA, Hanes V. Antihypertensive effects ofdrospirenone with 17beta-estradiol, a novel hormone treatment inpostmenopausal women with stage 1 hypertension. Circulation2005;112:1979–84.

[30] White WB, Hanes V, Chauhan V, Pitt B. Effects of a new hormone therapy,drospirenone and 17-beta-estradiol, in postmenopausal women withhypertension. Hypertension 2006;48:246–53.

[31] Preston RA, Norris PM, Alonso AB, Ni P, Hanes V, Karara AH. Randomized,placebo-controlled trial of the effects of drospirenone-estradiol on bloodpressure and potassium balance in hypertensive postmenopausal womenreceiving hydrochlorothiazide. Menopause 2007;14:408–14.

[32] Oelkers W, Foidart JM, Dombrovicz N, Welter A, Heithecker R. Effects of a neworal contraceptive containing an antimineralocorticoid progestogen,drospirenone, on the renin-aldosterone system, body weight, blood pressure,glucose tolerance, and lipid metabolism. J Clin Endocrinol Metab1995;80:1816–21.

[33] Archer DF, Thorneycroft IH, Foegh M, Hanes V, Glant MD, Bitterman P,Kempson RL. Long-term safety of drospirenone-estradiol for hormone therapy:a randomized, double-blind, multicenter trial. Menopause 2005;12:716–27.

[34] Foidart JM, Faustmann T. Advances in hormone replacement therapy: weightbenefits of drospirenone, a 17alpha-spirolactone-derived progestogen.Gynecol Endocrinol 2007;23:692–9.

[35] Tanko LB, Christiansen C. Effects of 17beta-oestradiol plus different doses ofdrospirenone on adipose tissue, adiponectin and atherogenic metabolites inpostmenopausal women. J Intern Med 2005;258:544–53.

n adipocytes and macrophages: A promising target to fight metabolic syn-

605606607608609610611612613614615616617618619620621622623624625626 Q6627628629630631632633634635636637638639640641642643644645646647648649650651652653654655656657658659660661662663664665666667668669670671672673674675676677678679680681682683684685686687688689

690691692693694695696697698699700701702703704705706707708709710711712713714715716717718719720721722723724725726727728729730731732733734735736737738739740741742743744745746747748749750751752753754755756757758759760761762763764765766767768769770771772773774

V. Marzolla et al. / Steroids xxx (2014) xxx–xxx 7

STE 7562 No. of Pages 8, Model 5G

14 May 2014

[36] Caprio M, Antelmi A, Chetrite G, Muscat A, Mammi C, Marzolla V, Fabbri A,Zennaro MC, Feve B. Anti adipogenic effects of the mineralocorticoid receptorantagonist drospirenone: potential implications for the treatment of metabolicsyndrome. Endocrinology 2011;152:113–25.

[37] Parkin L, Sharples K, Hernandez RK, Jick SS. Risk of venous thromboembolismin users of oral contraceptives containing drospirenone or levonorgestrel:nested case-control study based on UK General Practice Research Database.BMJ 2011;342:d2139.

[38] Jick SS, Hernandez RK. Risk of non-fatal venous thromboembolism in womenusing oral contraceptives containing drospirenone compared with womenusing oral contraceptives containing levonorgestrel: case-control study usingUnited States claims data. BMJ 2011;342:d2151.

[39] Preiss D, Zetterstrand S, McMurray JJ, Ostergren J, Michelson EL, Granger CB,Yusuf S, Swedberg K, Pfeffer MA, Gerstein HC, Sattar N. Predictors ofdevelopment of diabetes in patients with chronic heart failure in theCandesartan in Heart Failure Assessment of Reduction in Mortality andMorbidity (CHARM) program. Diabetes Care 2009;32:915–20.

[40] Krug AW, Stelzner L, Rao AD, Lichtman AH, Williams GH, Adler GK. Effect oflow dose mineralocorticoid receptor antagonist eplerenone on glucose andlipid metabolism in healthy adult males. Metabolism 2013;62:386–91.

[41] Garg R, Kneen L, Williams GH, Adler GK. Effect of mineralocorticoid receptorantagonist on insulin resistance and endothelial function in obese subjects.Diabetes Obes Metab 2013.

[42] Derosa G, Bonaventura A, Bianchi L, Romano D, D’Angelo A, Fogari E, Maffioli P.Effects of canrenone in patients with metabolic syndrome. Expert OpinPharmacother 2013;14:2161–9.

[43] Hauner H, Entenmann G, Wabitsch M, Gaillard D, Ailhaud G, Negrel R, PfeifferEF. Promoting effect of glucocorticoids on the differentiation of humanadipocyte precursor cells cultured in a chemically defined medium. J ClinInvest 1989;84:1663–70.

[44] Rondinone CM, Rodbard D, Baker ME. Aldosterone stimulated differentiationof mouse 3T3-L1 cells into adipocytes. Endocrinology 1993;132:2421–6.

[45] Caprio M, Feve B, Claes A, Viengchareun S, Lombes M, Zennaro MC. Pivotal roleof the mineralocorticoid receptor in corticosteroid-induced adipogenesis.FASEB J 2007;21:2185–94.

[46] Armani A, Marzolla V, Rosano G, Caprio M. Mineralocorticoid vs glucocorticoidreceptors: solo players or team mates in the control of adipogenesis? Int J Obes2014.

[47] Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipidmetabolism. Nature 2001;414:799–806.

[48] Hribal ML, Oriente F, Accili D. Mouse models of insulin resistance. Am J PhysiolEndocrinol Metab 2002;282:E977–81.

[49] Arner E, Ryden M, Arner P. Tumor necrosis factor alpha and regulation ofadipose tissue. N Engl J Med 2010;362:1151–3.

[50] Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest2005;115:1111–9.

[51] Jernas M, Palming J, Sjoholm K, Jennische E, Svensson PA, Gabrielsson BG, LevinM, Sjogren A, Rudemo M, Lystig TC, Carlsson B, Carlsson LM, Lonn M.Separation of human adipocytes by size: hypertrophic fat cells display distinctgene expression. FASEB J 2006;20:1540–2.

[52] Skurk T, Alberti-Huber C, Herder C, Hauner H. Relationship between adipocytesize and adipokine expression and secretion. J Clin Endocrinol Metab 2007;92:1023–33.

[53] Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum proteinsimilar to C1q, produced exclusively in adipocytes. J Biol Chem 1995;270:26746–9.

[54] Berg AH, Combs TP, Scherer PE. ACRP30/adiponectin: an adipokine regulatingglucose and lipid metabolism. Trends Endocrinol Metab 2002;13:84–9.

[55] Kuhn E, Bourgeois C, Keo V, Viengchareun S, Muscat A, Meduri G, Le MD, FeveB, Lombes M. Paradoxical resistance to high-fat diet-induced obesity andaltered macrophage polarization in mineralocorticoid receptor-overexpressingmice. Am J Physiol Endocrinol Metab 2014;306:E75–90.

[56] Le Menuet D, Viengchareun S, Muffat-Joly M, Zennaro MC, Lombes M.Expression and function of the human mineralocorticoid receptor: lessonsfrom transgenic mouse models. Mol Cell Endocrinol 2004;217:127–36.

[57] Tuck ML, Sowers J, Dornfeld L, Kledzik G, Maxwell M. The effect of weightreduction on blood pressure, plasma renin activity, and plasma aldosteronelevels in obese patients. N Engl J Med 1981;304:930–3.

[58] Feraco A, Armani A, Mammi C, Fabbri A, Rosano GM, Caprio M. Role ofmineralocorticoid receptor and renin-angiotensin-aldosterone system inadipocyte dysfunction and obesity. J Steroid Biochem Mol Biol 2013.

[59] Krug AW, Vleugels K, Schinner S, Lamounier-Zepter V, Ziegler CG, Bornstein SR,Ehrhart-Bornstein M. Human adipocytes induce an ERK1/2 MAP kinases-mediated upregulation of steroidogenic acute regulatory protein (StAR) and anangiotensin II-sensitization in human adrenocortical cells. Int J Obes (Lond)2007;31:1605–16.

[60] Jeon JH, Kim KY, Kim JH, Baek A, Cho H, Lee YH, Kim JW, Kim D, Han SH, Lim JS,Yoon DY, Kim SH, Oh GT, Kim E, Yang Y. A novel adipokine CTRP1 stimulatesaldosterone production. FASEB J 2008;22:1502–11.

[61] Vleugels K, Schinner S, Krause D, Morawietz H, Bornstein SR, Ehrhart-Bornstein M, Krug AW. ERK1/2 MAPKs and Wnt signaling pathways areindependently involved in adipocytokine-mediated aldosterone secretion. ExpClin Endocrinol Diabetes 2011;119:644–8.

[62] Caprio M, Newfell BG, La SA, Baur W, Fabbri A, Rosano G, Mendelsohn ME, JaffeIZ. Functional mineralocorticoid receptors in human vascular endothelial cells

Please cite this article in press as: Marzolla V et al. Mineralocorticoid receptor idrome. Steroids (2014), http://dx.doi.org/10.1016/j.steroids.2014.05.001

regulate intercellular adhesion molecule-1 expression and promote leukocyteadhesion. Circ Res 2008;102:1359–67.

[63] Deuchar GA, McLean D, Hadoke PW, Brownstein DG, Webb DJ, Mullins JJ,Chapman K, Seckl JR, Kotelevtsev YV. 11beta-hydroxysteroid dehydrogenasetype 2 deficiency accelerates atherogenesis and causes proinflammatorychanges in the endothelium in apoe–/– mice. Endocrinology 2011;152:236–46.

[64] Ahima RS, Flier JS. Adipose tissue as an endocrine organ. Trends EndocrinolMetab 2000;11:327–32.

[65] Seale P, Kajimura S, Spiegelman BM. Transcriptional control of brownadipocyte development and physiological function-of mice and men. GenesDev 2009;23:788–97.

[66] Lowell BB, Spiegelman BM. Towards a molecular understanding of adaptivethermogenesis. Nature 2000;404:652–60.

[67] Inokuma K, Ogura-Okamatsu Y, Toda C, Kimura K, Yamashita H, Saito M.Uncoupling protein 1 is necessary for norepinephrine-induced glucoseutilization in brown adipose tissue. Diabetes 2005;54:1385–91.

[68] Cousin B, Cinti S, Morroni M, Raimbault S, Ricquier D, Penicaud L, Casteilla L.Occurrence of brown adipocytes in rat white adipose tissue: molecular andmorphological characterization. J Cell Sci 1992;103(Pt 4):931–42.

[69] Young P, Arch JR, Ashwell M. Brown adipose tissue in the parametrial fat pad ofthe mouse. FEBS Lett 1984;167:10–4.

[70] Guerra C, Koza RA, Yamashita H, Walsh K, Kozak LP. Emergence of brownadipocytes in white fat in mice is under genetic control. Effects on body weightand adiposity. J Clin Invest 1998;102:412–20.

[71] Stanford KI, Middelbeek RJ, Townsend KL, An D, Nygaard EB, Hitchcox KM,Markan KR, Nakano K, Hirshman MF, Tseng YH, Goodyear LJ. Brown adiposetissue regulates glucose homeostasis and insulin sensitivity. J Clin Invest2013;123:215–23.

[72] Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, Kuo FC,Palmer EL, Tseng YH, Doria A, Kolodny GM, Kahn CR. Identification andimportance of brown adipose tissue in adult humans. N Engl J Med2009;360:1509–17.

[73] Saito M, Okamatsu-Ogura Y, Matsushita M, Watanabe K, Yoneshiro T, Nio-Kobayashi J, Iwanaga T, Miyagawa M, Kameya T, Nakada K, Kawai Y, TsujisakiM. High incidence of metabolically active brown adipose tissue in healthyadult humans: effects of cold exposure and adiposity. Diabetes2009;58:1526–31.

[74] van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JM,Kemerink GJ, Bouvy ND, Schrauwen P, Teule GJ. Cold-activated brown adiposetissue in healthy men. N Engl J Med 2009;360:1500–8.

[75] Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren R, Niemi T, TaittonenM, Laine J, Savisto NJ, Enerback S, Nuutila P. Functional brown adipose tissue inhealthy adults. N Engl J Med 2009;360:1518–25.

[76] Zennaro MC, Le MD, Viengchareun S, Walker F, Ricquier D, Lombes M.Hibernoma development in transgenic mice identifies brown adipose tissue asa novel target of aldosterone action. J Clin Invest 1998;101:1254–60.

[77] Penfornis P, Viengchareun S, Le MD, Cluzeaud F, Zennaro MC, Lombes M. Themineralocorticoid receptor mediates aldosterone-induced differentiation ofT37i cells into brown adipocytes. Am J Physiol Endocrinol Metab2000;279:E386–94.

[78] Viengchareun S, Penfornis P, Zennaro MC, Lombes M. Mineralocorticoid andglucocorticoid receptors inhibit UCP expression and function in brownadipocytes. Am J Physiol Endocrinol Metab 2001;280:E640–9.

[79] Wada T, Kenmochi H, Miyashita Y, Sasaki M, Ojima M, Sasahara M, Koya D,Tsuneki H, Sasaoka T. Spironolactone improves glucose and lipid metabolismby ameliorating hepatic steatosis and inflammation and suppressing enhancedgluconeogenesis induced by high-fat and high-fructose diet. Endocrinology2010;151:2040–9.

[80] Lee J. Adipose tissue macrophages in the development of obesity-inducedinflammation, insulin resistance and type 2 diabetes. Arch Pharm Res2013;36:208–22.

[81] Chawla A, Nguyen KD, Goh YP. Macrophage-mediated inflammation inmetabolic disease. Nat Rev Immunol 2011;11:738–49.

[82] Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch inadipose tissue macrophage polarization. J Clin Invest 2007;117:175–84.

[83] Odegaard JI, Ricardo-Gonzalez RR, Red EA, Vats D, Morel CR, Goforth MH,Subramanian V, Mukundan L, Ferrante AW, Chawla A. Alternative M2activation of Kupffer cells by PPAR delta ameliorates obesity-induced insulinresistance. Cell Metab 2008;7:496–507.

[84] Hevener AL, Olefsky JM, Reichart D, Nguyen MT, Bandyopadyhay G, Leung HY,Watt MJ, Benner C, Febbraio MA, Nguyen AK, Folian B, Subramaniam S,Gonzalez FJ, Glass CK, Ricote M. Macrophage PPAR gamma is required fornormal skeletal muscle and hepatic insulin sensitivity and full antidiabeticeffects of thiazolidinediones. J Clin Invest 2007;117:1658–69.

[85] Rickard AJ, Morgan J, Tesch G, Funder JW, Fuller PJ, Young MJ. Deletion ofmineralocorticoid receptors from macrophages protects againstdeoxycorticosterone/salt-induced cardiac fibrosis and increased bloodpressure. Hypertension 2009;54:537–43.

[86] Usher MG, Duan SZ, Ivaschenko CY, Frieler RA, Berger S, Schutz G, Lumeng CN,Mortensen RM. Myeloid mineralocorticoid receptor controls macrophagepolarization and cardiovascular hypertrophy and remodeling in mice. J ClinInvest 2010;120:3350–64.

[87] Bienvenu LA, Morgan J, Rickard AJ, Tesch GH, Cranston GA, Fletcher EK,Delbridge LM, Young MJ. Macrophage mineralocorticoid receptor signaling

n adipocytes and macrophages: A promising target to fight metabolic syn-

775776777778779780781782783784785786787788789

790791792793794795796797798799800801802803

804

8 V. Marzolla et al. / Steroids xxx (2014) xxx–xxx

STE 7562 No. of Pages 8, Model 5G

14 May 2014

plays a key role in aldosterone-independent cardiac fibrosis. Endocrinology2012;153:3416–25.

[88] Labuzek K, Liber S, Buldak L, Machnik G, Liber J, Okopien B. Eplerenonepromotes alternative activation in human monocyte-derived macrophages.Pharmacol Rep 2013;65:226–34.

[89] Wada T, Miyashita Y, Sasaki M, Aruga Y, Nakamura Y, Ishii Y, Sasahara M,Kanasaki K, Kitada M, Koya D, Shimano H, Tsuneki H, Sasaoka T. Eplerenoneameliorates the phenotypes of metabolic syndrome with NASH in liver-specific SREBP-1c Tg mice fed high-fat and high-fructose diet. Am J PhysiolEndocrinol Metab 2013;305:E1415–25.

[90] Messaoudi S, Azibani F, Delcayre C, Jaisser F. Aldosterone, mineralocorticoidreceptor, and heart failure. Mol Cell Endocrinol 2012;350:266–72.

[91] Nguyen KD, Qiu Y, Cui X, Goh YP, Mwangi J, David T, Mukundan L, BrombacherF, Locksley RM, Chawla A. Alternatively activated macrophages producecatecholamines to sustain adaptive thermogenesis. Nature 2011;480:104–8.

Please cite this article in press as: Marzolla V et al. Mineralocorticoid receptor idrome. Steroids (2014), http://dx.doi.org/10.1016/j.steroids.2014.05.001

[92] Stein M, Keshav S, Harris N, Gordon S. Interleukin 4 potently enhances murinemacrophage mannose receptor activity: a marker of alternative immunologicmacrophage activation. J Exp Med 1992;176:287–92.

[93] Doherty TM, Kastelein R, Menon S, Andrade S, Coffman RL. Modulation ofmurine macrophage function by IL-13. J Immunol 1993;151:7151–60.

[94] Doyle AG, Herbein G, Montaner LJ, Minty AJ, Caput D, Ferrara P, Gordon S.Interleukin-13 alters the activation state of murine macrophages in vitro:comparison with interleukin-4 and interferon-gamma. Eur J Immunol1994;24:1441–5.

[95] Van Dyken SJ, Locksley RM. Interleukin-4-and interleukin-13-mediatedalternatively activated macrophages: roles in homeostasis and disease. AnnuRev Immunol 2013;31:317–43.

[96] Takeda K, Kishimoto T, Akira S. STAT6: its role in interleukin 4-mediatedbiological functions. J Mol Med (Berl) 1997;75:317–26.

n adipocytes and macrophages: A promising target to fight metabolic syn-