Oxysterols and nuclear receptors

Oxysterols and nuclear receptors

Accepted Manuscript Oxysterols and nuclear receptors Liqian Ma, Erik R. Nelson PII: S0303-7207(19)30022-X DOI: https://doi.org/10.1016/j.mce.2019.0...

703KB Sizes 0 Downloads 94 Views

Accepted Manuscript Oxysterols and nuclear receptors Liqian Ma, Erik R. Nelson PII:

S0303-7207(19)30022-X

DOI:

https://doi.org/10.1016/j.mce.2019.01.016

Reference:

MCE 10378

To appear in:

Molecular and Cellular Endocrinology

Received Date: 5 November 2018 Revised Date:

8 January 2019

Accepted Date: 16 January 2019

Please cite this article as: Ma, L., Nelson, E.R., Oxysterols and nuclear receptors, Molecular and Cellular Endocrinology (2019), doi: https://doi.org/10.1016/j.mce.2019.01.016. 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

Title: Oxysterols and Nuclear Receptors

1

Authors: Liqian Ma and Erik R. Nelsona,b,c,d,e* a

Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign. Urbana, IL. University of Illinois Cancer Center. Chicago, IL. c Division of Nutritional Sciences, University of Illinois at Urbana-Champaign. Urbana, IL. d Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois at Urbana Champaign, Urbana, IL e Cancer Center at Illinois, University of Illinois at Urbana-Champaign, IL

M AN U

SC

RI PT

b

Abbreviated Title: Oxysterols and Nuclear Receptors. Keywords: cholesterol, oxysterol, 27-hydroxycholesterol; estrogen receptor; liver x receptor; breast cancer; selective estrogen receptor modulator, selective nuclear receptor modulator

EP

TE D

* Corresponding Author: Erik R. Nelson. University of Illinois at Urbana-Champaign. 407 S Goodwin Ave (MC114), Urbana, IL, 61801. Phone: 217-244-5477. Fax: 217-333-1133. E-mail: [email protected].

AC C

2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23

ACCEPTED MANUSCRIPT

RI PT

Abstract: Oxysterols are derivatives of cholesterol and an important regulator of cholesterol metabolism, in part due to their role as ligands for nuclear receptors, such as the liver x receptors. Oxysterols are also known to be ligands for the RAR-related orphan receptors, involved in normal T cell differentiation. However, increasing evidence supports a role for oxysterols in the progression of several diseases. Here, we review recent developments in oxysterol research, highlighting the biological functions that oxysterols exert through their target nuclear receptors: the liver x receptors, estrogen receptors RAR-related orphan receptor and the glucocorticoid receptor. We also bring the regulation of the immune system into the context of interaction between oxysterols and nuclear receptors, discussing the effect of such interaction on the pro-inflammatory function of macrophages and the development of T cells. Finally, we examine the impact that oxysterols have on various disease models, including cancer, Alzheimer’s disease and atherosclerosis, stressing the role of nuclear receptors if previously identified. This review underscores the need to consider the multifaceted roles of oxysterols in terms of multiple receptor engagements and selective modulation of these receptors.

EP

TE D

M AN U

SC

Funding: This work was funded in part by grants from the Department of Defense Breast Cancer Research Program (BC171214) and the American Institute of Cancer Research (Award 31284) to ERN.

AC C

24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

ACCEPTED MANUSCRIPT 42

Introduction:

43 Oxysterols are derivatives of cholesterol produced by the oxidation of cholesterol molecules by enzymatic

45

addition of hydroxyl, carbonyl or epoxide functional groups1. Endogenous oxysterols are commonly produced

46

by non-enzymatic mechanisms2. Specifically, reactive oxygen species (ROS), including alkoxy and peroxyl

47

radicals, can oxidize cholesterol molecules1,3. Cholesterol can also be oxidized by the leukocyte-H2O2-HOCl

48

system during inflammation3. Non-enzymatic oxidation is primarily responsible for the generation of certain

49

oxysterols such as 7-ketocholesterol (7KC) and 7β-hydroxycholesterol (7βHC)3. However, producing

50

oxysterols by the oxidation of cholesterol on the side-chain is typically catalyzed through enzymatic routes,

51

which are mediated by cytochrome p450 family of enzymes4,5. For instance, cholesterol can be metabolized

52

into 27-hydroxycholesterol (27HC) by CYP27A1, 24-hydroxycholesterol (24HC) by CYP46A1, 25-

53

hydroxycholesterol (25HC) by cholesterol-25-hydroxylase and 22-hydroxycholesterol by CYP11A13,4 (Figure 1).

M AN U

SC

RI PT

44

54

Many oxysterols have been described to bind to and modulate the activity of the liver x receptors (LXRs),

56

thereby completing an important homeostatic loop in cholesterol metabolism (described below). However,

57

several recent studies have implicated oxysterols in the modulation of other nuclear receptors such as the

58

retinoic acid receptor-related orphan receptors (RORs), estrogen receptors (ERs) and glucocorticoid receptors

59

(GRs), as well as non-nuclear receptor mechanisms. Furthermore, it is becoming increasingly apparent that

60

oxysterols play important roles in several pathologies, including cancers, cardiovascular diseases, and

61

neurodegenerative diseases6. This review will discuss our current understanding of oxysterol modulation of

62

nuclear receptors. We will also discuss various pathologies where oxysterols have been described to play a

63

role, and when known, highlight whether nuclear receptor involvement has been examined.

65

EP

AC C

64

TE D

55

Oxysterols and Nuclear Receptors

66 67

Nuclear Receptors

68

Nuclear receptors are a large superfamily consisting of 48 ligand-inducible transcription factors, acting as

69

intracellular receptors that bind to lipophilic ligands capable of crossing the plasma membrane (e.g. steroids)7.

ACCEPTED MANUSCRIPT 70

Conformational changes are commonly triggered upon ligand binding, allowing nuclear receptors to bind to

71

their respective DNA response elements, releasing corepressors and recruiting coactivators to promote

72

transcription of target genes involved in a variety of cellular processes, including cell proliferation and

73

metabolism7,8.

RI PT

74

Nuclear receptors share homology in their structures and functional domains. Five domains are generally

76

shared across typical nuclear receptors: a variable N-terminal region, a conserved DNA binding domain, a

77

variable hinge region, a conserved ligand binding domain and a variable C-terminal region9. After steroid

78

hormones were linked to the progression of prostate cancer and breast cancer, nuclear receptors have gained

79

increasing attention in terms of their roles in disease7,10,11.

M AN U

80

SC

75

Oxysterols and Liver X Receptor Signaling

82

Of the nuclear receptors, the liver X receptors (LXRs) are thought to be the major target of oxysterols,

83

especially in terms of the regulation of cholesterol metabolism. LXRα (NR1H3) and LXRβ (NR1H2) are

84

isoforms of LXR, which serve as cholesterol sensors for the regulation of cholesterol excess12. LXRβ is

85

expressed fairly ubiquitously, while LXRα expression is highest in the liver, with lower expression to various

86

degrees in other tissues13,14. The two isoforms share significant sequence identity, and although speculated,

87

clear evidence of specific biological differences have not been well described. The LXRs are known to

88

heterodimerize with the retinoid X receptors (RXRs) and sit on various DNA response elements, with a direct

89

repeat 4 (DR4) being recognized as the consensus sequence (5′-AGGTCA-NNNN-AGGTCA-3′)12,15,16. In an

90

unliganded state, it is thought that the heterodimer interacts with transcriptionally repressive complex, such as

91

with nuclear receptor corepressor (NCoR), thereby transreprepressing the transcriptional activity of LXR17–19.

92

The transrepression elicited by NCoR complex plays an important role in LXR-mediated anti-inflammatory

93

pathways, such as suppression of Toll-like receptor (TLR)-dependent pro-inflammatory signal expression in

94

macrophages20,21. Upon ligand binding, corepressor proteins are exchanged for coactivators, including NAD+-

95

dependent deacetylases Sirtuin 1 (SIRT1), which allows the RNA polymerase II complex to associate and

96

initiate transcription18,19.

97

AC C

EP

TE D

81

ACCEPTED MANUSCRIPT The first endogenous ligands to be described for the LXRs were 22(R)-hydroxycholesterol (22RHC), 24(S)-

99

hydroxycholesterol (24SHC), and 24(S),25-epoxycholesterol (24,25EC)22. Since then, it has been

100

demonstrated that several oxysterols and certain precursors in the cholesterol-biosynthetic pathway can

101

modulate the activity of the LXRs. These include 22RHC, 25HC, 27HC, 24SHC, 24,25EC and desmosterol23–27.

102

These metabolites have a wide range of affinities or potencies, with 24SHC having an EC50 of 4 µM and 3 µM

103

compared to 27HC at 85 nM and 71 nM for LXRα and LXRβ, respectively22,27. For reference, 27HC is the most

104

abundant oxysterol in circulation with concentrations ranging from 67 ng/mL (0.17 µM) to 199 ng/mL (0.5 µM)

105

while 24SHC circulates from 39 ng/mL (0.1 µM) to 91 ng/mL (0.23 µM)28.

SC

RI PT

98

106

Given the hepatic expression of CYP7A1, and more ubiquitous expression of CYP27A1 (with significant

108

expression observed in cells of the myeloid immune lineage), it would be expected when cholesterol levels are

109

high, so too is its metabolism into oxysterols. Indeed, there is a positive association between circulating

110

cholesterol levels and 27HC in both mice and humans, while statin therapy is associated with decreased

111

plasma 27HC29,30. Furthermore addition of cholesterol to macrophages resulted in increased secretion of 27HC,

112

while inhibiting cholesterol synthesis with simvastatin abolished the production of 27HC by macrophages31,32.

113

Although it is difficult to account for esterification, these studies suggest that increased level, of cholesterol

114

could lead to increased production of 27HC, which can then engage LXRs. Upon binding of an endogenous

115

ligand to the LXR-RXR heterodimer, the corepressor complex dissociates with commensurate recruitment of

116

coactivation complexes, ultimately leading to a transcriptional program aimed at restoring cholesterol

117

homeostasis. Specifically, LXR activation leads to the induction of target genes involved in cholesterol efflux

118

(ATP binding cassette (ABC) transporters A1/G1/G5/G8), apolipoproteins (APO A-1/E), cholesterol metabolism

119

into bile acids (CYP7A1) and decreased intestinal cholesterol absorption33. The removal of cholesterol by

120

reverse cholesterol transport is facilitated by ABC transporters. ABCA1, expressed in multiple cell types,

121

promotes the efflux of free cholesterol to apolipoprotein, in particular APOA1, leading to the formation of

122

discoid lipid-poor HDL15,34. ABCG5 and ABCG8 are half transporters expressed on the apical membrane of

123

enterocyte and on the canalicular membrane of hepatocytes35. The formation of heterodimer by ABCG5 and

124

ABCG8 on hepatocytes can promote the secretion of cholesterol into bile, while on enterocytes can facilitate

125

the excretion of cholesterol back into intestinal lumen12,35. LXR activation is also involved in regulating lipid

AC C

EP

TE D

M AN U

107

ACCEPTED MANUSCRIPT homeostasis by promoting triglyceride synthesis. Genes upregulated in this process are sterol regulatory

127

element-binding protein 1, acetyl-CoA carboxylase, fatty acid synthase and stearoyl-CoA desaturase-134,36,37.

128

The relatively longer-term actions of LXR-mediated homeostasis work in concert with the quicker actions of

129

cholesterol and oxysterols in maintaining sterol regulatory element-binding proteins (especially SREBP 2) in an

130

inactive state bound to the endoplasmic reticulum, thereby reducing the transcription of genes associated with

131

cholesterol metabolism and import38.

RI PT

126

132

Oxysterol and Retinoic acid receptor-related Orphan Receptor (ROR) signaling

134

RORs are nuclear receptors with three subtypes, RORα (NR1F1), RORβ (NR1F2) and RORγ (NR1F3)39.

135

RORs regulate gene transcription by binding to their respective response element as monomers upon ligand

136

binding. Each type of ROR has multiple isoforms expressed in humans and mice, with RORγ having two

137

isoforms, RORγ1 and RORγt40. RORα are involved in a variety of cellular processes, such as cerebellar

138

development, circadian rhythm and lipid metabolism, while RORγ is critical for lymphocyte development and

139

differentiation39,40. Oxysterols, including 22RHC, 24SHC, 25HC and 27HC, have been described as

140

endogenous ligands for both RORα and RORγ41. However, the activities of oxysterols differ, as they have the

141

ability to act as both agonists or inverse agonists. For example, it has been generally found that 24SHC and

142

25HC act as inverse agonists for both RORα and RORγ, while 27HC acts as an agonist for RORγ41. However,

143

a study also identified the ability of 25HC and 22HC to restore RORγ transcriptional activity from a synthetic

144

inhibitor in mammalian cells lines, suggesting some agonistic activity42. Cholesterol and cholesterol sulfate are

145

also the endogenous ligands of ROR, specifically RORα, with cholesterol sulfate having higher affinity to the

146

receptor due to its better fitting in the ligand binding pocket43,44.

M AN U

TE D

EP

AC C

147

SC

133

148

Similar to the LXRs, RORs also participate in cholesterol metabolism. One of the transcriptional targets of

149

RORα activation is CYP7B1, which codes for oxysterol 7α-hydroxylase, an enzyme involved in metabolizing

150

27HC and 25HC to bile acids45. However, RORs’ interaction with oxysterols, especially RORγ and RORγt play

151

very important roles in T helper 17 cell (Th17) differentiation46,47. Th17 cells were shown to preferentially

152

produce two types of oxysterols, 7β, 27-dihydroxycholesterol (7β, 27DHC) and 7α, 27DHC, and these

153

oxysterols rescued the inhibitory effect of ursolic acid on RORγt, while promoting coactivator recruitment to the

ACCEPTED MANUSCRIPT ligand binding domain of RORγt46. In addition, RNA sequencing analysis showed that in Th17 cells, interleukin

155

(IL)-17A, IL-17F, IL-23R, C-C ligand (CCL)20 and C-C chemokine receptor (CCR)6 were regulated by both

156

RORγt and RORα48. Although it is clear that the RORs are required for normal Th17 differentiation and function,

157

the physiological relevance and precise impact of oxysterols on this process remain to be elucidated.

158 159

RI PT

154

Oxysterols and Estrogen Receptor (ER) Signaling

161

Oxysterols were definitively found to bind to and modulate the activity of the estrogen receptors just over 10

162

years ago49,50. There are two main subtypes of nuclear estrogen receptors in mammalian cells, ERα (NR3A1)

163

and ERβ (NR3A2). A membrane-associated G protein-coupled estrogen receptor 1 (GPER1, or GPR30) has

164

also been described, but whether oxysterols can interact with this receptor remains unknown51,52. Nuclear ER

165

signaling comprises ligand binding to ER, followed by dissociation of the heat shock protein from ER, revealing

166

nuclear localization signal, and homo- or hetero-dimerization of two ER monomers51. ER dimers then bind to

167

the respective estrogen response element of the target gene, recruiting co-activators and/or co-repressors,

168

leading to target gene transcription, including trefoil factor 1 (TIFF1) and progesterone receptor (PR)51,52. So-

169

called ‘non-genomic’ or ‘rapid’ ER signaling also exists, where cytosolic ERs engage in kinase-mediated

170

signaling cascades including mobilization of intracellular calcium, stimulation of adenylate cyclase activity and

171

epidermal growth factor receptor activation, ultimately resulting in rapid cellular changes6,52.

M AN U

TE D

EP

172

SC

160

As early as in 2004, oxysterols, including β-epoxycholesterol, 20(S)-hydroxycholesterol and 22RHC, were

174

shown to have estrogenic activity in ERα-expressing HeLa cells53. More recently, a Gal4-ER co-transfection

175

screen of several cholesterol metabolites revealed that 22RHC, 24SHC, 25HC and 27HC significantly inhibited

176

estradiol (E2) activation of both ERα and β, with IC50s ranging from 1-5 µM50. 27HC was subsequently shown

177

to be a selective ER modulator (SERM), with the ability to induce distinct ER conformational changes and to

178

manifest either activating or inhibitory activities, depending on the tissue and cellular context49. 27HC was

179

found to activate the classic ERE-Luciferase reporter (3XERE-TATA-Luc) in transfected HeLa cells, with an

180

EC50 of approximately 1 µM, and when co-treated with estradiol had a Ki of 1.32 µM49. Demonstrating its

AC C

173

ACCEPTED MANUSCRIPT 181

selective-modulatory capabilities, it could activate the ER to promote the cellular proliferation of ER+ breast

182

cancer cell lines, tumors or uteri, but inhibited estrogen-dependent expression of vascular nitric oxide synthase,

183

thereby repressing carotid artery reendothelialization49,50,54,55.

184 Other oxysterols or cholesterol metabolites besides 27HC have also been found to modulate ER activities.

186

7KC was shown to decrease the cytotoxicity of doxorubicin in MCF7 cells, and this effect was dependent on

187

ERα, as fulvestrant inhibition or ERα knockdown restored doxorubicin accumulation in MCF7 cells56. 7KC also

188

induced the expression of TIFF1, an ER target gene56. Structurally similar to 27HC, 25HC upregulates ER

189

target genes, including TIFF1, PR, cathepsin D, cyclin A and cyclin D1 in MCF7 and BG1 cells57. Furthermore,

190

25HC promotes ER+ breast cancer cell proliferation and inhibits hypoxia-inducible factor-1α (HIF-1α)

191

expression induced by hypoxia in cardiomyocytes to prevent cell apoptosis57.

M AN U

SC

RI PT

185

192

Subsequent studies have further confirmed the selective modulator capacity of 27HC. In hematopoietic stem

194

cells (HSCs) during pregnancy, 27HC could induce ERα-dependent HSC mobilization and extramedullary

195

hematopoiesis58. In a cancer cell model, 27HC enhanced ER-mediated transcriptional activity and the

196

recruitment of ER to TIFF1 and growth regulating estrogen receptor binding 1 (GREB1) response elements in

197

an ER+ cancer cell line (MCF7) adapted to long-term estrogen deprivation59. However, in cardiovascular cells,

198

27HC reversed the cardioprotective effect of estrogen by inhibiting estrogen-induced nitric oxide production by

199

vascular cells, indicating an antagonistic activity against ER60. 27HC exhibited a partial-agonist effect on

200

osteoblasts, but ultimately exerted a negative impact on bone quality that could be attenuated by exogenous

201

estrogen61,62. The negative effects on bone are likely a combination of signaling through the ERs and LXRs,

202

and 27HC has been recently been reported to be associated with decreased bone mineral density in

203

postmenopausal women when circulating estrogens are taken into account63. Moreover, 27HC could serve as

204

a partial agonist to ER in ER+ breast cancer cell lines other than MCF7 cells, including T47D and BT48349,54. It

205

has also been suggested that 27HC antagonizes the neuroprotective effects of estrogen signaling through ERα,

206

as an increase in 27HC concentration in the brain is associated with increased neurodegeneration of the

207

hippocampus, decreased expression of ER and a decreased level of hippocampal mitochondria64. 27HC was

208

also shown to exert effects through ERβ: not only was ERβ expression upregulated by 27HC in prostate

AC C

EP

TE D

193

ACCEPTED MANUSCRIPT 209

cancer cell lines, LNCaP and PC3, but the 27HC-induced proliferation of prostate cancer cells was attenuated

210

by the ERβ-specific inhibitor, PHTPP65.

211

Oxysterols and Other Nuclear Receptors

213

The glucocorticoid receptor (NR3C1, GR) is another type of nuclear receptor shown to interact with

214

oxysterols66. Normally, upon ligand (glucocorticoid steroid) binding, GR can dimerize and translocate into the

215

nucleus67,68. In the nucleus, GR dimers bind to GR response elements, recruit coactivators and induce gene

216

transcription (transactivation)67,68. Activated GRs can also complex with other transcription factors, such as

217

nuclear factor-κB (NFκB) and activator protein 1 (AP1), thereby sequestering the transcription factor and

218

repressing the respective target gene transcription (transrepression)67,68. One oxysterol recently identified as a

219

GR ligand is 6-oxo-cholestan-3β,5α-diol (OCDO), which is generated from 5,6α-epoxycholesterol and 5,6β-

220

epoxycholesterol (5,6EC) by cholesterol epoxide hydrolase (ChEH) and 11β-hydroxysteroid-dehydrogenase-

221

type-2 (11βHSD2) in the scenario of breast cancer66,69. By binding and translocating GR into the cellular

222

nucleus, OCDO led to the expression of MMP1, but did not stimulate the expression of cortisol induced-classic

223

GR target genes, SGK1 and MKP1. These data suggest that even though both OCDO and glucocorticoids

224

signal through GR, their actions on GR are distinct66,69.

TE D

M AN U

SC

RI PT

212

225

The estrogen-related receptor (ERR) family is composed of ERRα (NR3B1), ERRβ (NR3B2) and ERRγ

227

(NR3B3), with ERRα being the most studied70. ERRα shares 70% homology with ERα in the DNA-binding

228

domain, but classic ERα ligands, such as 17β-estradiol, do not directly regulate the activity of ERRα70,71. ERRα

229

has been categorized as an orphan receptor due to a lack of endogenous ligand, and is thought to be primarily

230

regulated by other proteins, such as PGC1α72. Recently, Wei et al. identified cholesterol to be a molecule that

231

activates and regulates the transcriptional activity of ERRα70. In addition, ERRα, at least in part, mediates the

232

cholesterol-stimulated osteoclastogenesis and bone resorption70. Whether oxysterols can also regulate the

233

activity of ERRα are unknown, but modeling would suggest that they could interact with the ERRα in a similar

234

way to cholesterol.

235

AC C

EP

226

ACCEPTED MANUSCRIPT Non-Nuclear Receptor Actions of Oxysterols

237

Although the focus of this review is on the interaction between oxysterols and nuclear receptors, it is important

238

to note that the mechanisms of action of oxysterols are not limited to nuclear receptors. Furthermore, the

239

precise mechanisms of action are not known for several of the biological phenomena that oxysterols are

240

implicated in (highlighted below). Oxysterols can also act through GPCRs, including Epstein-Barr virus-induced

241

G-protein coupled receptor 2 (EBI2/GPR183), GPR17, C-X-C chemokine receptor (CXCR)2 and Smoothened

242

(SMO)73–75. EBI2, CXCR2 and GPR17, in particular, were shown to be promiscuously modulated by

243

oxysterols73. In addition, Nedelcu et al. reported that Hedgehog signaling requires the binding of oxysterol to

244

vertebrate SMO75.

SC

RI PT

236

M AN U

245

Besides GPCRs, regulatory proteins, such as SREBPs, are also targets of oxysterols76–78. Specifically,

247

oxysterols can bind to Insulin-induced proteins (INSIGs), prompting INSIGs to bind to the SREBP cleavage-

248

activating protein (SCAP), which then leads to a conformational change in the cytoplasmic loop 6 of SCAP76,77.

249

SCAP is an SREBP-escort protein, which, upon synthesis, promotes the clustering of SREBP in coat protein

250

complex II (COPII)-coated vesicles76,77,79. The SCAP:SREBP complex vesicles then travel to the Golgi complex

251

from endoplasmic reticulum (ER)76,77,79. At the Golgi complex, SREBP is cleaved before entering the nucleus to

252

promote cholesterol synthesis76,77,79. Conformational change of SCAP, induced by oxysterols through INSIGs,

253

prevents the COPII proteins from binding to the MELADL sequence on SCAP, ultimately blocking the transport

254

of SREBPs to the Golgi complex and promotion of cholesterol synthesis76,77. Although cholesterol is the

255

prototypical ligand for the INSIGs, oxysterols have also been shown to bind. Thus, oxysterols likely have two

256

roles in terms of cellular cholesterol homeostasis: inhibition of SREBPs and engagement of the LXRs. One of

257

the SREBP isoforms, SREBP-1a, is highly expressed in macrophages and plays important roles in regulating

258

the activity of the inflammasome, such as the production of pro-inflammatory cytokine IL-1β80.

AC C

EP

TE D

246

259 260

Oxysterols in Health and Disease

261

Oxysterols and the Immune System

262

The roles of oxysterols in the immune system have gained increasing appreciation over the last decade. The

263

interaction of oxysterols with the immune system has been recognized as one of the major ways that

ACCEPTED MANUSCRIPT oxysterols contribute to diseases, and such interaction can be complicated and multifaceted. Demonstrating

265

the complexity of oxysterol regulation of the immune system, some studies have described 25-

266

hydroxycholesterol (25HC) as being immunosuppressive by inhibiting cytotoxic T cell production and the

267

inflammasome activities, while others have found that 7βHC and 25HC induced the production of pro-

268

inflammatory chemokines and cytokines in U937 and THP-1 cells4,81,82. Although some effects of oxysterols

269

have been attributed to specific mechanisms (such as via nuclear receptors or SREBP signaling), many are yet

270

to be explored. Where possible, we have highlighted known mechanisms within this review.

RI PT

264

SC

271 272

LXR activation has been shown to play an important anti-inflammatory role in the immune system, due to the

274

inhibition of pro-inflammatory signal production downstream of NFκB83. Ito et al. showed that LXR activation

275

could disrupt the membrane lipid organization by hindering the recruitment of adaptor proteins, MyD88 and

276

TRAF6, through ABCA1, ultimately resulting in the inhibition of the TLR signaling to NFκB and MAPK

277

effectors84. Non-nuclear receptor-mediated mechanisms are also prevalently used by oxysterols. For example,

278

25HC was identified as one of the anti-inflammatory oxysterols that reduced Il1b transcription and IL1-

279

activating inflammasome activity by repressing SREBP in macrophages85. Studies suggesting an antiviral role

280

of 25HC prompted Cagno et al. to treat cells with 25HC and 27HC prior to Herpes simplex virus 1 (HSV1)

281

infection86. This pre-treatment promoted IL-6 secretion in HeLa cells and NFκB activation in Vero cells, linking

282

25HC and 27HC to a pro-inflammatory role86. It is not clear whether these activities required SREBP or LXR. In

283

the context of cerebral inflammation, Jang et al. identified that 25HC promoted robust nucleotide-binding

284

oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome assembly and

285

activation in macrophages in a potassium efflux/mitochondrial ROS/LXR-dependent manner82. These findings

286

suggest that the actions of oxysterols on the immune function may be selective, cell-type specific and cell-

287

location specific.

AC C

EP

TE D

M AN U

273

288 289

Oxysterols, Nuclear Receptors and Innate Immune System

290

The interaction between LXR and the immune system was first reported by Joseph et al. in that the loss of

291

functional LXRs in mice made them highly susceptible to infection by the intracellular bacteria Listeria

ACCEPTED MANUSCRIPT monocytogenes87. Also, expression of LXR promoted macrophage survival during the infection87. The

293

described effects of LXRs on innate immune cells range from regulation of cholesterol efflux to regulation of

294

inflammatory status. LXRα plays an important role in regulating the activities of Kupffer cells and natural killer T

295

cells88. Endo-Umeda et al. showed that knocking out LXRα leads to an increased level of bone marrow-derived

296

Kupffer cell population (F4/80+CD68+CD11b+) in the liver of mice when they are fed with a high cholesterol

297

diet88. Also, the lack of LXRα led to increased production of pro-inflammatory cytokines by those cells88.

298

Contrary to Kupffer cells, deficiency in LXRα reduced the frequency of natural killer cells and impaired those

299

cells functionally, as illustrated by the decreased capacity to produce IL-4 and interferon (IFN)-γ post α-GalCer

300

stimulation88. Whether specific oxysterols can modulate these LXR-associated activities is likely, given that

301

they are ligands for the LXRs.

M AN U

SC

RI PT

292

302

LXR is not the only nuclear receptor that is involved in innate immunity management. Studies have reported

304

innate immune cell functions regulated by oxysterols through RORα and ER. Tuong et al. identified a crosstalk

305

between RORα and 25HC in regulating macrophage function89. They found that the cholesterol 25 hydroxylase

306

expression was attenuated in RORα-deficient staggerer (sg/sg) mice, along with macrophages exhibiting

307

dysregulated lipid phenotype. Also, knocking down cholesterol 25 hydroxylase with siRNA in wildtype

308

macrophages resulted in lipid dysregulation89. However, treating sg/sg macrophages with 25HC significantly

309

decreased lipid accumulation into lipid droplets by approximately 1.7-fold and rescued the lipid dysregulation

310

phenotype89. ER is another receptor involved in the oxysterol-immunity axis. During pregnancy, 27HC

311

promotes ERα-dependent hematopoietic stem cell mobilization58, and estrogens themselves have been shown

312

to recruit myeloid cells to the tumor niche90. As will be discussed below, 27HC also increases myeloid cell

313

recruitment to metastatic lesions, although it is not yet known whether this is mediated by the ERs or LXRs91.

EP

AC C

314

TE D

303

315

Many studies have suggested an anti-inflammatory effect of oxysterols on innate immune cells, although

316

several of these effects are either LXR/nuclear receptor-independent or have mechanisms that remain

317

unknown. For example, in glial cells, while the oxysterols oxidized on the sterol backbone generally had no

318

effect on pro-inflammatory cytokine production after lipopolysaccharide (LPS) induction, oxysterols oxidized on

319

the side chain generally decreased LPS-induced mRNA expression of IL-1β and macrophages inflammatory

ACCEPTED MANUSCRIPT protein (MIP)-1α, suppressing the inflammatory phenotype of macrophages92. In the same study, it was further

321

shown that 24,25EC, 25HC, and 27HC decreased LPS-induced expression of IL-6 and tumor necrosis factor

322

(TNF)-α92. This finding is further supported by Marengo et al., indicating that an oxysterol mixture decreased

323

the level of reactive oxygen species (ROS) production in human macrophages and increased the level of

324

immunoregulatory cytokine IL-10 secretion93. The relative contributions of LXRs or SREBPs in these processes

325

are not currently known. Besides cytokines, LXR ligands, such as 25HC, can downregulate LPS-induced

326

expression of cyclooxygenase (COX)2 and mPGE2 synthase 1 (mPGES1), both of which are involved in

327

producing prostaglandins, a group of physiologically active and pro-inflammatory lipid compounds94.

SC

RI PT

320

328

7-oxygenated cholesterols have been associated with the development of atherosclerosis, suggesting a pro-

330

inflammatory effect in foamy macrophages during the formation of the atherosclerotic lesion. Similar to those

331

effects of oxysterols that are anti-inflammatory, the mechanisms of oxysterols’ pro-inflammatory effects on the

332

innate immunity largely remain to be elucidated. Proteomics revealed that a mixed treatment of 7βHC and

333

7KC reduced the expressions of glyoxalase 1 and adenylyl cyclase-associated protein 1 (CAP1) in human

334

THP1 cells; both proteins are associated with anti-inflammatory activities95–97. Specifically, glyoxalase 1 inhibits

335

the accumulation of oxidative stress in diabetes mellitus97. Overexpressing CAP1 in monocytes exacerbated

336

adipose tissue inflammation in mice, while suppressing CAP1 expression abolished the resistin-mediated

337

inflammatory activity both in vitro and in vivo96. Another type of pro-inflammatory cytokine, IL-8, was also

338

shown to be upregulated in human macrophages via PI3K and MEK pathway upon 7KC and 7α-

339

hydroxycholesterol (7αHC) treatment98. In this study, the GPCR complement receptor, C5a was implicated in

340

mediating the upregulation of IL-8.

342

TE D

EP

AC C

341

M AN U

329

343 344

Oxysterol, Nuclear Receptors and Adaptive Immune System

345

The interaction between oxysterols and the adaptive immune system is mediated by both LXR and RORγt,

346

with RORγt specifically involved in Th17 differentiation and functionality. LXR activation and signaling usually

347

negatively regulates T cell proliferation99,83. It was shown by Bensinger et al. that LXRβ knockout mice

ACCEPTED MANUSCRIPT displayed unregulated T cell activity, including lymphoid hyperplasia and enhanced responses to antigen

349

challenge100. Also, Ma et al. showed that Tc9 cells, a polarized population of CD8+ T cells with strong anti-

350

tumor activity, are regulated by cholesterol through its derivative, oxysterols, such as 22RHC101. 22RHC

351

inhibited IL-9 expression by activating LXRs, which then resulted in LXR sumoylation and decreased p65

352

binding to the promoter region of Il9 gene, while IL-9 is essential for the survival of Tc9 in vivo101. LXR

353

activation by 25HC and 22RHC also takes part in T regulatory cell (Treg) control102,103. Mice lacking cholesterol

354

25 hydroxylase exhibited increased production of IL-10, while 25HC treatment impaired IL-27-induced Treg

355

differentiation by downregulating IL-10 expression in an LXR-dependent manner102.

SC

RI PT

348

356

Soroosh et al. first described the most potent selective activator for RORγt to be 7β, 27DHC46. They showed

358

that in vivo administration of 7β, 27DHC promoted IL-17 production and in vitro treatment of this oxysterol

359

enhanced IL-17-producing Th17 cell differentiation in a RORγt-dependent manner46. On the flip side, knocking

360

out the enzyme CYP27A1, which is responsible for the production of 7β, 27DHC, significantly reduced IL-17-

361

producing CD4+ and γδ+ T cell population46. Santori et al. also identified “cholesterol biosynthetic intermediate”

362

(CBI) downstream of lanosterol and upstream of zymosterol to be natural RORγt ligand42. In the study,

363

knocking out CYP51 led to smaller lymph node anlagen and stunted development of lymph node structure in

364

embryos, developmental processes mediated by RORγt42. In addition, breeding Sc4molf/f mice to CD4-cre and

365

RORγt-cre mice generated the deletion of Sc4mol in TCRαβ T cells and RORγt-expressing cells. Sc4mol is

366

responsible for modifying the C4-methyl groups of CBIs, and its deletion led to a significant decrease in IL-17-

367

producing CD4+ T cells42. The requirement of CYP51 and Sc4mol in RORγt-mediated adaptive immunity

368

developmental processes dictates the indispensable role of CBI in RORγt signaling.

TE D

EP

AC C

369

M AN U

357

370

Oxysterols and Cancer

371

Since 27HC was first described to be a selective estrogen receptor modulator, the effect of this oxysterol on

372

breast cancer has now been extensively studied in pre-clinical models, which is not surprising given that

373

approximately 60-75% of all breast cancers are ERα+. CYP27A1 protein expression was associated with

374

higher breast tumor grade, while CYP7B1 mRNA expression is associated with a better prognosis, indicating a

375

clinically relevant role for 27HC in breast cancer6,54 . As expected, 27HC promoted the in vitro proliferation of

ACCEPTED MANUSCRIPT cell lines in an ER-dependent manner49,54,55. 25HC has also been shown to promote the proliferation of breast

377

cancer cells through activation of the ER57. 27HC increased the growth of ER+ mammary tumors, while

378

inhibiting the production of 27HC attenuated hypercholesterolemia-driven tumor growth in vivo54,55. Intriguingly,

379

27HC could also promote the transition from epithelial-like to mesenchymal-like (EMT) in breast cancer

380

cells54,104. The synthetic LXR agonist, GW3965, could also promote EMT, indicating that this phenotype is likely

381

LXR mediated54. 7KC also plays a role in breast cancer in that 7KC decreased the cytotoxicity of doxorubicin in

382

ER+ breast cancer cell line MCF7 by upregulating P-glycoprotein expression, while this effect was not

383

observed in ER- cell line, MDA-MB-23156. The requirement of the ERα in 7KC’s effect was further illustrated by

384

knocking down ERα with siRNA, which restored the doxorubicin accumulation in MCF756.

SC

RI PT

376

M AN U

385

While the effects of 27HC and 7KC on breast cancer cells required ERα, ERβ may also play an important role

387

in 27HC signaling in cancer types. Raza et al. demonstrated that 27HC promoted prostate cancer cell

388

proliferation in vitro in LNCaP and PC3 cells through the ER. Specifically, 27HC induced proliferation was

389

significantly reduced in the presence of the ER inhibitor, ICI 182,780 (fulvestrant)65. Likewise, the ERβ-

390

selective antagonist PHTPP reversed the proliferative effect of 27HC on prostate cancer cells, indicating that

391

27HC signals through ERβ to promote their proliferation65. However, these results were contradicted by

392

another study, indicating that 27HC actually decreased the proliferation of prostate cancer cells, via an

393

SREBP2 mediated mechanism105. Furthermore, the expression of CYP27A1 was downregulated in prostate

394

cancers compared to normal tissue, and elevated CYP27A1 expression is associated with a better prognosis

395

for prostate cancer patients. Further work will be required to better characterize the impact of oxysterols on

396

prostate cancer.

AC C

EP

TE D

386

397 398

In addition to breast and prostate cancer, 27HC was shown to affect lung cancer development. Using H1395

399

cell line as a lung cancer model, Hiramitsu et al. found that 27HC advanced ERβ+ lung cancer cell proliferation

400

via phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling106. Chen et al. described the pro-metastatic

401

activity of 25HC in lung adenocarcinoma107. In A549 cells and NCL-H1975 cells, 25HC treatment enhanced

402

snai1 expression, a marker of EMT, in an LXR-dependent manner, which promoted invasion of those cancer

403

cells107.

ACCEPTED MANUSCRIPT 404 Although many oxysterols have been reported to contribute to cancer progression, cholestane-3β, 5α, 6β-triol

406

(triol) was found to have anti-cancer activity108. Not only was the triol treatment able to inhibit the growth of

407

LNCaP CDXR3, DU145 and PC3 prostate cancer cell lines, but also oral administration of this oxysterol was

408

able to slow the progression of xenograft PC3 in NUDE mice108. Triol treatment also inhibited the invasive

409

capacity of DU145 and PC3 cells and downregulated the epithelial to mesenchymal transition-related genes,

410

including N-cadherin, vimentin, Slug, focal adhesion kinase (FAK), phospho-FAK Ser722 and phospho-FAK

411

Tyr861108. Another LXR modulator, dendrogenin A, was reported by de Medina et al. to possess anti-tumor

412

property in vivo109. This cholesterol metabolite was produced by 5,6αEC reacting with histamine and was

413

significantly downregulated in breast tumor, compared to normal tissues109,110. Treating cancer cells with

414

dendrogenin A promoted cancer cell re-differentiation, inhibited OCDO production and induced lethal cancer

415

cell autophagy109,110.

M AN U

SC

RI PT

405

416

Besides the direct effects of oxysterols on cancer cells, these molecules impart an indirect effect on cancer by

418

interplaying with the immune system and creating a microenvironment that promotes cancer invasion and

419

metastasis16,91. Following the study that first identified the pro-metastatic effect of 27HC on breast cancer,

420

Baek et al. reported that 27HC required myeloid immune cell population to promote metastasis54,91. 27HC

421

treatment led to increased levels of polymorphonuclear-neutrophils (PMNs) and γδ+ T cells at distal metastatic

422

sites91. PMNs were further shown to be required for the enrichment of γδ+ T cells, and that both cell

423

populations were required for the full metastatic effect of 27HC91. Specifically, immune depletion of either

424

PMNs or γδ+ T cells reversed the 27HC-driven Met1 breast cancer metastasis to the lungs91. Gene expression

425

revealed that 27HC likely polarizes PMNs into an immune suppressive ‘N2’ phenotype, thereby explaining the

426

observed decreases in CD8+ cytotoxic T cells post 27HC treatment91. Moresco et al. also suggested that

427

oxysterols likely promote breast tumor metastasis by recruiting tumor-promoting neutrophils to the lung

428

metastatic niche, as oxysterol-inactivating enzyme sulfotransferase 2B1b (SULT2B1b)-transduced 4T1 tumor

429

had lower levels of metastasis and infiltration by tumor-promoting neutrophils in vivo111. Raccosta et al.

430

reported an oxysterol-dependent, LXR-independent mechanism by which oxysterols recruit tumor-promoting

431

neutrophils. They showed that bone marrow-derived CD11bhighGr1high myeloid cells migrate toward natural and

AC C

EP

TE D

417

ACCEPTED MANUSCRIPT tumor-released oxysterols112. Specifically, 22RHC led to the activation of CXCR2 on CD11bhighGr1high cells,

433

whose activation led to the production of pro-angiogenic factor, MMP9112. Inactivating CXCR2 inhibited AB1

434

and LLC tumor growth by reducing angiogenesis and immunosuppression112. An LXR-dependent mechanism

435

by which oxysterols create immunosuppressive microenvironment was also illustrated by the same group: by

436

activating LXRα on dendritic cells (DCs), oxysterols inhibit CCR7 expression on DCs, thereby preventing them

437

from migrating to lymph nodes to facilitate T cell activation against tumor cells113. Given that the pro-metastatic

438

effects of oxysterols involve the immune system, it is likely these effects are similar across solid tumors. Indeed,

439

27HC was shown to promote metastasis in pre-clinical models of breast, colon, pancreatic and lung cancers91.

440

Therefore, when examining the role of oxysterols in cancer progression, it is critical that one considers both

441

their direct effects on cancer cells as well as their cancer-cell extrinsic effects.

M AN U

SC

RI PT

432

442 Neurodegenerative Diseases

444

The brain contains approximately 25% of total cholesterol in our body114. In the central nervous system,

445

cholesterol metabolism is tightly controlled and separated from the rest of the body by the blood-brain barrier

446

(BBB)114. Oxysterols such as 27HC can cross the BBB, and play an important role in maintaining cholesterol

447

homeostasis in the central nervous system115,116. However, dysregulated cholesterol homeostasis in the brain,

448

especially along the oxysterol-LXR axis, is implicated in many neurodegenerative diseases, such as

449

Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD)6,114. Besides regulating

450

cholesterol and lipid metabolism, oxysterol signaling through the LXRs also participates in other cellular

451

processes. Among them, 24SHC, 25HC and 27HC have been shown to induce adaptive responses in neuronal

452

cells against cytotoxic oxidative stress induced by 7KC117. This protective effect requires the downstream

453

expression of ABCG1, but not ABCA1117. Additionally, 25HC induces the expression of cholesterol 25

454

hydroxylase via the activation of the LXR, independent of cell type118. In the nervous system, 22HC can

455

upregulate the expression of MAP kinase phosphatase (MKP)-1, which then suppresses the JNK-mediated

456

inflammation in brain astrocytes119. The mechanism of this suppression by 22RHC is via the phosphorylation of

457

Hu antigen R through PKCα, which in turn stabilizes MKP-1 mRNA119. Although the metabotropic glutamate

458

receptor 5 is required for the cytosolic increase in Ca2+, an event upstream of the phosphorylation of Hu, it is

AC C

EP

TE D

443

ACCEPTED MANUSCRIPT 459

not clear whether 22RHC directly binds to this receptor, or whether these activities require interaction with the

460

more traditional oxysterol binding proteins (such as the LXRs or INSIG).

461 24HC is the most abundant oxysterol synthesized in the brain, whereas 27HC is the primary oxysterol

463

produced by peripheral cells, which crosses the BBB114,115,120. While 7KC is associated with a neuro-damaging

464

role, 24HC was shown to have a protective effect during the development of neurodegenerative disorders. For

465

instance, Testa et al. demonstrated that, in SK-N-BE neuroblastoma cells, 24HC could modulate the SIRT1,

466

which then led to the prevention of hyperphosphorylated tau protein accumulation induced by amyloid β (Aβ)

467

monomers121. Okabe et al. also found that sub-lethal concentrations of 24SHC induced adaptive responses

468

and protected the human neuroblastoma SH-SY5Y cells against cytotoxic stress induced by 7KC, dependent

469

of activating LXRβ transcriptional pathway117. However, high concentrations of 24SHC could induce

470

“necroptosis-like” cell death in neuronal cells due to the formation of 24SHC esters120,122. Also, 24HC was

471

shown to increase Aβ accumulation, which in turn contributes to neuronal cell death. Reported by Gamba et al.,

472

24HC promoted not only the binding of Aβ to human neuronal cell lines, SK-N-BE and NT2, but also the pro‐

473

apoptotic and pro‐necrogenic effects of Aβ1–42 peptide on SK-N-BE and NT2, by locally increasing ROS levels

474

through NADPH oxidase123. These works together suggest that 24HC at low concentrations might be protective,

475

but as its levels increase due to dysregulated cholesterol metabolism, this oxysterol could then act to induce

476

cell death.

SC

M AN U

TE D

EP AC C

477

RI PT

462

478

27HC, as the most abundant peripheral oxysterol, can cross the BBB and is therefore also implicated in

479

neuronal cell death. An increased level of 27HC was reported in the brain of patients diagnosed with AD124–126.

480

Also, hypercholesterolemia was linked to accelerated AD pathology and the accumulation of Aβ127,128. One

481

mechanism of cholesterol-linked AD development could be attributed to 27HC accumulation in the brain. Ismail

482

et al. showed that 27HC reduced LXR-mediated brain glucose uptake, GLUT4 expression and spatial memory

483

in mouse model129. The reduced glucose metabolic activity in AD patients has been well-identified in the

484

angular gyrus and some parietotemporal regions130. Moreover, 27HC promoted NFκB binding to the promoter

485

of aspartyl protease BACE1, leading to an increased BACE1 activity and BACE1-mediated cleavage of

ACCEPTED MANUSCRIPT amyloid precursor protein, in a gadd153-dependent manner131. Also, 27HC-induced ROS production could

487

promote IL-6/STAT3 signaling, which was implicated in the induction of nerve cell senescence132. However, it

488

was unclear whether this effect was due to the selective modulatory activity of 27HC on ER. Taken together,

489

limiting dietary cholesterol intake would likely reduce the level of peripheral 27HC, and thus may exhibit clinical

490

value in preventing AD and other related neurodegenerative disorder development29,133. In this regard, there is

491

a positive correlation between plasma cholesterol and 27HC levels in humans, including after dietary

492

consumption of an extra 750mg/day cholesterol for 4 weeks29,133. In mice, a high cholesterol diet results in

493

significantly increased 27HC levels, although dietary restriction experiments have yet to be reported50,54,62.

SC

RI PT

486

494

25HC was reported to be involved in the progression of amyotrophic lateral sclerosis (ALS). Clinical analysis

496

showed that 25HC level was elevated in both cerebral spinal fluid and serum of ALS patients134. Also, the level

497

of 25HC was associated with the level of disease severity in ALS patients134. Mechanistically, 25HC-mediated

498

ALS pathogenesis is likely due to the induction of motor neuronal death, a process dependent of glycogen

499

synthase kinase-3β and LXR, which was shown by Kim et al. in the motor neuron-like cell line (NSC34)

500

expressing mutant G93A superoxide dismutase 1 gene134.

501

TE D

M AN U

495

Atherosclerosis

503

Atherosclerosis is the underlying pathology of many acute cardiovascular syndromes. Atherosclerosis

504

development is characterized by the narrowing of the artery, which results from the accumulation of lipids and

505

inflammatory cells within the intima of arteries. Progression of atherosclerosis is thought to be initiated by

506

endothelial cell dysfunction, leading to the accumulation of low-density lipoprotein-cholesterol (LDL-cholesterol)

507

in the extracellular matrix (ECM). LDL-cholesterol buildup at the extracellular matrix becomes a target of

508

oxidation, and LDLs can be oxidized into oxidized LDL (ox-LDL). Ox-LDL promotes the recruitment of

509

macrophages and the subsequent engulfment of ox-LDL by macrophages transforms macrophages into foam

510

cells6,33,135,136. This transformation is the hallmark of atherosclerotic lesion development137. However, recent

511

reports are questioning whether immune dysregulation may actually precede plaque formation138,139.

512

AC C

EP

502

ACCEPTED MANUSCRIPT Being a byproduct of cholesterol, oxysterols are formed in atherosclerotic plaques both non-enzymatically by

514

LDL oxidation and enzymatically during cholesterol catabolism by cytochrome p450 enzymes that are highly

515

expressed in macrophages33. Due to their prevalence in the atherosclerotic plaques, the role of oxysterols in

516

plaque formation has been heavily investigated. However, the pro-atherogenic properties of oxysterols have

517

been in a long debate largely due to the multifold effects of oxysterols. Macrophages are present in a spectrum

518

of so-called polarities. On the extremes of this polarization spectrum are classically activated, inflammatory M1

519

macrophages, and alternatively activated, anti-inflammatory macrophages. Macrophages across this spectrum

520

are localized within different types of atherosclerotic lesions. Macrophages localized in rupture-prone

521

atherosclerotic plaques more closely reflect the biology of M1 macrophages, while the macrophages that

522

situate in stable plaque more closely resemble the phenotype of M2 macrophages6,93. It has been reported by

523

Marengo et al. that 27HC contributes to the M2 polarization of human macrophages by upregulating the

524

expression of CD36 and CD204 and the production of IL-10, leading to the plaque stabilization93. On the

525

contrary, Gargiulo et al. found that 27HC promoted TLR4 and NFκB activation in U937 cells, which led to the

526

release of pro-inflammatory cytokines, IL-8, IL-1β and TNF-α140. The production of pro-inflammatory cytokines

527

induced by 27HC then contributed to the production of matrix metallopeptidase 9. Although these studies were

528

performed in vitro, the presence of matrix metallopeptidase 9 has the potential to promote plaque instability

529

and rupture140. The pro-inflammatory role of 27HC is likely due to the activation of ERα, as 27HC-induced pro-

530

inflammatory cytokine mRNA expression (IL-1β, IL-6 and TNF-α) was attenuated by ER deficiency60. Ishikawa

531

et al. reported a novel mechanism by which the activation of LXR exerts an atheroprotective role in endothelial

532

cells141. Specifically, GW3965-, T0901317- or 22RHC-induced LXR signaling promotes ER-dependent eNOS

533

activation141. By co-localizing with ERα in the lipid raft and thus functionally coupling with ERα, LXR stimulation

534

could

535

reendothelialization141.

lead

AC C

EP

TE D

M AN U

SC

RI PT

513

to

ERα

Ser118

phosphorylation

by

PI3K/Akt,

eventually

promoting

eNOS-induced

536 537

Structurally similar to 27HC, 25HC was also identified with an atheroprotective role. Li et al. found that Krüppel-

538

like factor 4 (KLF4), a master transcription factor that regulates the anti-inflammatory activity of macrophages,

539

could induce the expression of cholesterol 25 hydroxylase, LXRα and LXRβ in macrophages (RAW264.7)142.

540

Further, the activation of the KLF4-cholesterol 25 hydroxylase-LXR axis promoted M1 to M2 transition in

ACCEPTED MANUSCRIPT 541

macrophages, as both treating macrophages with 25HC and overexpressing KLF4 in macrophages led to a

542

decreased inflammatory activity in macrophages142. Knocking down cholesterol 25 hydroxylase in RAW264.7

543

cells also stimulated the expression of the pro-inflammatory genes and M1 phenotype142.

544 Non-nuclear receptor-mediated mechanisms by which oxysterols contribute to atherosclerosis development

546

have also reported. The β1-Subunit of BKCa channel (KCNMB1) is linked to atheroprotective, in that it

547

modulates vascular contractility143. Interestingly, 7KC has previously been reported to be a competitive inhibitor

548

of the aryl hydrocarbon receptor (AhR)144. In a subsequent report, 7KC was found to contribute to vascular

549

rigidity, and decreases the expression of KCNMB1 protein levels143. The decrease in KCNMB1 was attributed

550

to a decrease in the protein expression of the AhR, although the requirement of the AHR was not specifically

551

tested143. While the AhR is not a member of the nuclear receptor superfamily, it has a very similar mechanism

552

of action in that it is a ligand-activated transcription factor145,146. It will be of interest to evaluate whether other

553

oxysterols can modulate the AhR and to determine the physiological relevance of such engagement.

M AN U

SC

RI PT

545

554 Conclusion

556

Oxysterols play important roles in cholesterol metabolism and the regulation of cholesterol homeostasis.

557

Binding to LXRs, oxysterols promote cholesterol efflux and bile acid synthesis. However, an elevated level of

558

oxysterols has been implicated in the pathophysiology of many age-related diseases, such as breast cancer,

559

atherosclerosis and AD. The main target nuclear receptors mediating the actions of oxysterols are the LXRs,

560

ERs and RORs, although recent reports for the GR and potentially ERRα reveal the possibility for other

561

nuclear receptors being involved in oxysterol biology. The action of oxysterols on each type of receptor is

562

multifaceted. Being a SERM, 27HC can activate or inhibit ER in different cell types. For ROR, oxysterols’

563

actions range from agonist to inverse agonist. It is therefore possible that the multilevel effect of oxysterols on

564

the target receptors leads to diverse phenotypes in cells upon oxysterol stimulation. Also, desmosterol has now

565

been formally demonstrated to be a selective modulator of the LXRs in macrophages147. Specifically,

566

desmosterol activated LXR target gene, ABCA1, while suppressing the SREBP target gene, 24-

567

dehydrocholesterol reductase (DHCR24), and had no effect on fatty acid synthase in mouse and human

568

macrophages147. Indeed, phenotypically, there is also some debate regarding the inflammatory role of

AC C

EP

TE D

555

ACCEPTED MANUSCRIPT oxysterols. While they are generally seen as pro-inflammatory in the innate immune system, an increasing

570

number of studies reported the anti-inflammatory behaviors of oxysterols. The direct effect of oxysterols on T

571

cells are oxysterol- and T cell-type- specific. While 7β, 27DHC promotes Th17 differentiation, 25HC negatively

572

regulates Treg. Though 27HC is recognized as a SERM, other oxysterols’ selective modulatory effects are less

573

clear, although highly likely. Given the evidence that many oxysterols exert variable effects on cells, it is

574

important for future studies to investigate whether other oxysterols are involved in selectively modulating

575

nuclear receptors, such as LXR, in disease conditions.

SC

576

RI PT

569

There is now significant evidence that oxysterols play exacerbating roles across several diseases. Not only can

578

they have direct effects on cancer cells to promote cancer growth, but also oxysterols can modulate pro-tumor

579

host cells, such as myeloid cells, to promote disease progression. Specific mechanisms for many of the

580

described effects of oxysterols in physiology or disease are still unknown. Still, several of the roles of

581

oxysterols are mediated through the LXRs, and thus they have been proposed to be a therapeutic target.

582

However, challenges in targeting this receptor have emerged, as LXRs play an essential role in cholesterol and

583

lipid homeostasis148. While it may be beneficial to target this receptor, its activation leads to

584

hypertriglyceridemia148, which could increase the risk of developing cardiovascular-related diseases or

585

steatosis. Given the potential for LXR ligands to have differential effects depending on the tissue and cellular

586

context, targeting these receptors should proceed with caution. Therefore, it will be of importance for future

587

studies to consider the specific downstream targets upon oxysterol signaling in each of the disease models.

588 589 590 591

References

592

1. Griffiths, W. J. et al. Current trends in oxysterol research. Biochem. Soc. Trans. 44, 652–658 (2016).

593

2. Brzeska, M., Szymczyk, K. & Szterk, A. Current Knowledge about Oxysterols: A Review. J. Food Sci. 81,

594 595 596

AC C

EP

TE D

M AN U

577

R2299–R2308 (2016). 3. Poli, G., Biasi, F. & Leonarduzzi, G. Oxysterols in the pathogenesis of major chronic diseases. Redox Biol. 1, 125–130 (2013).

ACCEPTED MANUSCRIPT 597

4. Vejux, A. & Lizard, G. Cytotoxic effects of oxysterols associated with human diseases: Induction of cell

598

death (apoptosis and/or oncosis), oxidative and inflammatory activities, and phospholipidosis. Mol.

599

Aspects Med. 30, 153–170 (2009).

603 604 605

RI PT

602

218 (2007).

6. He, S. & Nelson, E. R. 27-Hydroxycholesterol, an endogenous selective estrogen receptor modulator. Maturitas 104, 29–35 (2017).

SC

601

5. Norlin, M. & Wikvall, K. Enzymes in the conversion of cholesterol into bile acids. Curr. Mol. Med. 7, 199–

7. Dhiman, V. K., Bolt, M. J. & White, K. P. Nuclear receptors in cancer - uncovering new and evolving roles through genomic analysis. Nat. Rev. Genet. 19, 160–174 (2018).

M AN U

600

8. Sever, R. & Glass, C. K. Signaling by Nuclear Receptors. Cold Spring Harb. Perspect. Biol. 5, (2013).

607

9. Mangelsdorf, D. J. et al. The Nuclear Receptor Superfamily: The Second Decade. Cell 83, 835–839 (1995).

608

10. Huggins, C. Endocrine methods of treatment of cancer of the breast. J. Natl. Cancer Inst. 15, 1–25 (1954).

609

11. Huggins, C. & Hodges, C. V. Studies on prostatic cancer. I. The effect of castration, of estrogen and

TE D

606

610

androgen injection on serum phosphatases in metastatic carcinoma of the prostate. CA. Cancer J. Clin. 22,

611

232–240 (1972).

614

EP

613

12. Maqdasy, S. et al. Once and for all, LXRα and LXRβ are gatekeepers of the endocrine system. Mol. Aspects Med. 49, 31–46 (2016).

AC C

612

13. Auboeuf, D. et al. Tissue distribution and quantification of the expression of mRNAs of peroxisome

615

proliferator-activated receptors and liver X receptor-alpha in humans: no alteration in adipose tissue of

616

obese and NIDDM patients. Diabetes 46, 1319–1327 (1997).

617 618 619 620

14. Heine, G. et al. Liver X receptors control IgE expression in B cells. J. Immunol. Baltim. Md 1950 182, 5276– 5282 (2009). 15. Lee, S. D. & Tontonoz, P. Liver X Receptors at the Intersection of Lipid Metabolism and Atherogenesis. Atherosclerosis 242, 29–36 (2015).

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

RI PT

626

18. Wang, C., Tian, L., Popov, V. M. & Pestell, R. G. Acetylation and Nuclear Receptor Action. J. Steroid Biochem. Mol. Biol. 123, 91–100 (2011).

19. Li, X. et al. SIRT1 deacetylates and positively regulates the nuclear receptor LXR. Mol. Cell 28, 91–106

SC

625

OMEGA 3 FATTY ACIDS. Cell 155, 200–214 (2013).

(2007).

20. Glass, C. K. & Saijo, K. Nuclear receptor transrepression pathways that regulate inflammation in

M AN U

624

17. Li, P. et al. NCoR REPRESSION OF LXRs RESTRICTS MACROPHAGE BIOSYNTHESIS OF INSULIN-SENSITIZING

macrophages and T cells. Nat. Rev. Immunol. 10, 365–376 (2010).

21. Treuter, E. New wrestling rules of anti-inflammatory transrepression by oxysterol receptor LXR revealed. Cell Res. 21, 711–714 (2011).

22. Janowski, B. A. et al. Structural requirements of ligands for the oxysterol liver X receptors LXRα and LXRβ.

TE D

623

Metab. TEM 28, 485–496 (2017).

Proc. Natl. Acad. Sci. U. S. A. 96, 266–271 (1999).

23. Morello, F. et al. LXR-activating oxysterols induce the expression of inflammatory markers in endothelial cells through LXR-independent mechanisms. Atherosclerosis 207, 38–44 (2009).

EP

622

16. Kloudova, A., Guengerich, F. P. & Soucek, P. The role of oxysterols in human cancer. Trends Endocrinol.

24. Lehmann, J. M. et al. Activation of the nuclear receptor LXR by oxysterols defines a new hormone

AC C

621

ACCEPTED MANUSCRIPT

response pathway. J. Biol. Chem. 272, 3137–3140 (1997). 25. Yoshikawa, T. et al. Identification of liver X receptor-retinoid X receptor as an activator of the sterol regulatory element-binding protein 1c gene promoter. Mol. Cell. Biol. 21, 2991–3000 (2001). 26. Chen, W., Chen, G., Head, D. L., Mangelsdorf, D. J. & Russell, D. W. Enzymatic Reduction of Oxysterols Impairs LXR Signaling in Cultured Cells and the Livers of Mice. Cell Metab. 5, 73–79 (2007). 27. Fu, X. et al. 27-hydroxycholesterol is an endogenous ligand for liver X receptor in cholesterol-loaded cells. J. Biol. Chem. 276, 38378–38387 (2001).

ACCEPTED MANUSCRIPT

647

648

649

650

651

plasma by high-performance liquid chromatography-mass spectrometry. J. Lipid Res. 45, 776–781 (2004). 29. Karuna, R. et al. Plasma levels of 27-hydroxycholesterol in humans and mice with monogenic disturbances of high density lipoprotein metabolism. Atherosclerosis 214, 448–455 (2011).

RI PT

646

28. Burkard, I., Rentsch, K. M. & von Eckardstein, A. Determination of 24S- and 27-hydroxycholesterol in

30. Thelen, K. M. et al. Effect of pravastatin on plasma sterols and oxysterols in men. Eur. J. Clin. Pharmacol. 62, 9–14 (2006).

31. Hansson, M., Ellis, E., Hunt, M. C., Schmitz, G. & Babiker, A. Marked induction of sterol 27-hydroxylase

SC

645

activity and mRNA levels during differentiation of human cultured monocytes into macrophages. Biochim.

653

Biophys. Acta 1593, 283–289 (2003).

654

M AN U

652

32. Babiker, A. et al. Elimination of cholesterol in macrophages and endothelial cells by the sterol 27-

655

hydroxylase mechanism. Comparison with high density lipoprotein-mediated reverse cholesterol transport.

656

J. Biol. Chem. 272, 26253–26261 (1997).

33. Brown, A. J. & Jessup, W. Oxysterols and atherosclerosis. Atherosclerosis 142, 1–28 (1999).

658

34. Bełtowski, J. Liver X receptors (LXR) as therapeutic targets in dyslipidemia. Cardiovasc. Ther. 26, 297–316

659

TE D

657

(2008).

35. Baldán, Á., Bojanic, D. D. & Edwards, P. A. The ABCs of sterol transport. J. Lipid Res. 50, S80–S85 (2009).

661

36. Rong, S. et al. Expression of SREBP-1c Requires SREBP-2-mediated Generation of a Sterol Ligand for LXR in

663 664 665 666

AC C

662

EP

660

Livers of Mice. eLife 6, (2017).

37. Calkin, A. & Tontonoz, P. LXR signaling pathways and atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 30, (2010).

38. Baek, A. E. & Nelson, E. R. The Contribution of Cholesterol and Its Metabolites to the Pathophysiology of Breast Cancer. Horm. Cancer 7, 219–228 (2016).

667

ACCEPTED MANUSCRIPT 39. Wang, Y., Kumar, N., Crumbley, C., Griffin, P. R. & Burris, T. P. A Second Class of Nuclear Receptors for

668

Oxysterols: Regulation of RORα and RORγ activity by 24S-Hydroxycholesterol (Cerebrosterol). Biochim.

669

Biophys. Acta 1801, 917–923 (2010).

673

674

675

676

RI PT

672

lipid/glucose metabolism, inflammation, and insulin sensitivity. Front. Endocrinol. 4, (2013).

41. Mutemberezi, V., Guillemot-Legris, O. & Muccioli, G. G. Oxysterols: From cholesterol metabolites to key mediators. Prog. Lipid Res. 64, 152–169 (2016).

SC

671

40. Jetten, A. M., Kang, H. S. & Takeda, Y. Retinoic acid-related orphan receptors α and γ: key regulators of

42. Santori, F. R. et al. Identification of Natural RORγ Ligands that Regulate the Development of Lymphoid Cells. Cell Metab. 21, 286–297 (2015).

M AN U

670

43. Kallen, J. A. et al. X-ray structure of the hRORalpha LBD at 1.63 A: structural and functional data that

677

cholesterol or a cholesterol derivative is the natural ligand of RORalpha. Struct. Lond. Engl. 1993 10, 1697–

678

1707 (2002).

44. Kallen, J., Schlaeppi, J.-M., Bitsch, F., Delhon, I. & Fournier, B. Crystal structure of the human RORalpha

TE D

679

680

Ligand binding domain in complex with cholesterol sulfate at 2.2 A. J. Biol. Chem. 279, 14033–14038

681

(2004).

45. Wada, T., Kang, H. S., Jetten, A. M. & Xie, W. The Emerging Role Of Nuclear Receptor RORα And Its

EP

682

Crosstalk With LXR In Xeno- And Endobiotic Gene Regulation. Exp. Biol. Med. Maywood NJ 233, 1191–1201

684

(2008).

685

686

AC C

683

46. Soroosh, P. et al. Oxysterols are agonist ligands of RORγt and drive Th17 cell differentiation. Proc. Natl. Acad. Sci. U. S. A. 111, 12163–12168 (2014).

687

47. Bird, L. T cells: Endogenous agonists for orphan RORγ. Nat. Rev. Immunol. 15, 70–71 (2015).

688

48. Castro, G. et al. RORγt and RORα signature genes in human Th17 cells. PLoS ONE 12, (2017).

689

49. DuSell, C. D., Umetani, M., Shaul, P. W., Mangelsdorf, D. J. & McDonnell, D. P. 27-hydroxycholesterol is an

690

endogenous selective estrogen receptor modulator. Mol. Endocrinol. Baltim. Md 22, 65–77 (2008).

ACCEPTED MANUSCRIPT

691

692

50. Umetani, M. et al. 27-Hydroxycholesterol is an endogenous SERM that inhibits the cardiovascular effects of estrogen. Nat. Med. 13, 1185–1192 (2007). 51. Imai, Y. et al. Nuclear Receptors in Bone Physiology and Diseases. Physiol. Rev. 93, 481–523 (2013).

694

52. Wardell, S. E., Nelson, E. R. & McDonnell, D. P. From empirical to mechanism-based discovery of clinically

700

701 702 703 704 705 706 707 708 709

SC

699

54. Nelson, E. R. et al. 27-Hydroxycholesterol Links Hypercholesterolemia and Breast Cancer Pathophysiology. Science 342, 1094–1098 (2013).

M AN U

698

transcriptional activation assay system using HeLa cells. Biosci. Biotechnol. Biochem. 68, 1790–1793 (2004).

55. Wu, Q. et al. 27-Hydroxycholesterol promotes cell-autonomous, ER-positive breast cancer growth. Cell Rep. 5, 637–645 (2013).

56. Wang, C.-W. et al. 7-ketocholesterol and 27-hydroxycholesterol decreased doxorubicin sensitivity in breast cancer cells: estrogenic activity and mTOR pathway. Oncotarget 8, 66033–66050 (2017).

TE D

697

53. Sato, H. et al. Oxysterol regulation of estrogen receptor alpha-mediated gene expression in a

57. Lappano, R. et al. The Cholesterol Metabolite 25-Hydroxycholesterol Activates Estrogen Receptor αMediated Signaling in Cancer Cells and in Cardiomyocytes. PLoS ONE 6, (2011). 58. Oguro, H. et al. 27-Hydroxycholesterol induces hematopoietic stem cell mobilization and extramedullary

EP

696

useful Selective Estrogen Receptor Modulators (SERMs). Steroids 90, 30–38 (2014).

hematopoiesis during pregnancy. J. Clin. Invest. 127, 3392–3401

AC C

695

RI PT

693

59. Simigdala, N. et al. Cholesterol biosynthesis pathway as a novel mechanism of resistance to estrogen deprivation in estrogen receptor-positive breast cancer. Breast Cancer Res. BCR 18, (2016).

710

60. Umetani, M. et al. The Cholesterol Metabolite 27-Hydroxycholesterol Promotes Atherosclerosis via

711

Proinflammatory Processes Mediated by Estrogen Receptor Alpha. Cell Metab. 20, 172–182 (2014).

712

61. DuSell, C. D. et al. The Endogenous Selective Estrogen Receptor Modulator 27-Hydroxycholesterol Is a

713

Negative Regulator of Bone Homeostasis. Endocrinology 151, 3675–3685 (2010).

ACCEPTED MANUSCRIPT

714

62. Nelson, E. R. et al. The Oxysterol, 27-Hydroxycholesterol, Links Cholesterol Metabolism to Bone

715

Homeostasis Through Its Actions on the Estrogen and Liver X Receptors. Endocrinology 152, 4691–4705

716

(2011). 63. Chang, P.-Y. et al. 27-hydroxycholesterol, an endogenous SERM, and risk of fracture in postmenopausal

RI PT

717

718

women: A nested case-cohort study in the Women’s Health Initiative. J. Bone Miner. Res. Off. J. Am. Soc.

719

Bone Miner. Res. (2018). doi:10.1002/jbmr.3576

64. Brooks, S. W., Dykes, A. C. & Schreurs, B. G. A High-Cholesterol Diet Increases 27-Hydroxycholesterol and

SC

720

Modifies Estrogen Receptor Expression and Neurodegeneration in Rabbit Hippocampus. J. Alzheimers Dis.

722

JAD 56, 185–196 (2017).

724

725

726

65. Raza, S. et al. The cholesterol metabolite 27-hydroxycholesterol stimulates cell proliferation via ERβ in prostate cancer cells. Cancer Cell Int. 17, (2017).

66. Voisin, M. et al. Identification of a tumor-promoter cholesterol metabolite in human breast cancers acting through the glucocorticoid receptor. Proc. Natl. Acad. Sci. U. S. A. 114, E9346–E9355 (2017).

TE D

723

M AN U

721

727

67. Weikum, E. R., Knuesel, M. T., Ortlund, E. A. & Yamamoto, K. R. Glucocorticoid receptor control of

728

transcription: precision and plasticity via allostery. Nat. Rev. Mol. Cell Biol. 18, 159–174 (2017).

731

732

733

734

735

736

EP

730

68. Ratman, D. et al. How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering. Mol. Cell. Endocrinol. 380, 41–54 (2013).

AC C

729

69. Silvente-Poirot, S., Dalenc, F. & Poirot, M. The Effects of Cholesterol-Derived Oncometabolites on Nuclear Receptor Function in Cancer. Cancer Res. 78, 4803–4808 (2018). 70. Wei, W. et al. Ligand Activation of ERRα by Cholesterol Mediates Statin and Bisphosphonate Effects. Cell Metab. 23, 479–491 (2016). 71. Ascenzi, P., Bocedi, A. & Marino, M. Structure-function relationship of estrogen receptor alpha and beta: impact on human health. Mol. Aspects Med. 27, 299–402 (2006).

ACCEPTED MANUSCRIPT

737

72. Chang, C. & McDonnell, D. P. Molecular Pathways: The Metabolic Regulator Estrogen-Related Receptor

738

Alpha as a Therapeutic Target in Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 18, 6089–6095

739

(2012).

741

73. Sensi, C. et al. Oxysterols act as promiscuous ligands of class-A GPCRs: in silico molecular modeling and in vitro validation. Cell. Signal. 26, 2614–2620 (2014).

RI PT

740

74. Hannedouche, S. et al. Oxysterols direct immune cell migration through EBI2. Nature 475, 524–527 (2011).

743

75. Nedelcu, D., Liu, J., Xu, Y., Jao, C. & Salic, A. Oxysterol binding to the extracellular domain of Smoothened

745

in Hedgehog signaling. Nat. Chem. Biol. 9, 557–564 (2013).

76. Radhakrishnan, A., Ikeda, Y., Kwon, H. J., Brown, M. S. & Goldstein, J. L. Sterol-regulated transport of

M AN U

744

SC

742

746

SREBPs from endoplasmic reticulum to Golgi: Oxysterols block transport by binding to Insig. Proc. Natl.

747

Acad. Sci. U. S. A. 104, 6511–6518 (2007).

748

77. Sun, L.-P., Seemann, J., Goldstein, J. L. & Brown, M. S. Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: Insig renders sorting signal in Scap inaccessible to COPII proteins. Proc.

750

Natl. Acad. Sci. U. S. A. 104, 6519–6526 (2007).

753

754

755

756

757

mechanisms, both involving SCAP and Insigs. J. Biol. Chem. 279, 52772–52780 (2004).

EP

752

78. Adams, C. M. et al. Cholesterol and 25-hydroxycholesterol inhibit activation of SREBPs by different

79. Matsuda, M. et al. SREBP cleavage-activating protein (SCAP) is required for increased lipid synthesis in liver

AC C

751

TE D

749

induced by cholesterol deprivation and insulin elevation. Genes Dev. 15, 1206–1216 (2001). 80. Im, S.-S. et al. Linking Lipid Metabolism to the Innate Immune Response in Macrophages through Sterol Regulatory Element Binding Protein -1a. Cell Metab. 13, 540–549 (2011). 81. Erridge, C., Webb, D. J. & Spickett, C. M. 25-Hydroxycholesterol, 7beta-hydroxycholesterol and 7-

758

ketocholesterol upregulate interleukin-8 expression independently of Toll-like receptor 1, 2, 4 or 6

759

signalling in human macrophages. Free Radic. Res. 41, 260–266 (2007).

ACCEPTED MANUSCRIPT

764

765

766 767 768 769 770 771 772

(2013).

RI PT

763

83. Spann, N. J. & Glass, C. K. Sterols and oxysterols in immune cell function. Nat. Immunol. 14, 893–900

84. Ito, A. et al. LXRs link metabolism to inflammation through Abca1-dependent regulation of membrane composition and TLR signaling. eLife 4,

85. Reboldi, A. et al. 25-hydroxycholesterol suppresses interleukin-1-driven inflammation downstream of type

SC

762

adrenoleukodystrophy through activation of the NLRP3 inflammasome. Nat. Commun. 7, 13129 (2016).

I interferon. Science 345, 679–684 (2014).

86. Cagno, V. et al. Inhibition of herpes simplex-1 virus replication by 25-hydroxycholesterol and 27-

M AN U

761

82. Jang, J. et al. 25-hydroxycholesterol contributes to cerebral inflammation of X-linked

hydroxycholesterol. Redox Biol. 12, 522–527 (2017).

87. Joseph, S. B. et al. LXR-Dependent Gene Expression Is Important for Macrophage Survival and the Innate Immune Response. Cell 119, 299–309 (2004).

88. Endo-Umeda, K., Nakashima, H., Umeda, N., Seki, S. & Makishima, M. Dysregulation of Kupffer Cells/

TE D

760

773

Macrophages and Natural Killer T Cells in Steatohepatitis in LXRα Knockout Male Mice. Endocrinology

774

(2018). doi:10.1210/en.2017-03141

777

778 779

780 781 782

EP

776

89. Tuong, Z. K. et al. RORα and 25-Hydroxycholesterol Crosstalk Regulates Lipid Droplet Homeostasis in Macrophages. PLoS ONE 11, (2016).

AC C

775

90. Svoronos, N. et al. Tumor Cell–Independent Estrogen Signaling Drives Disease Progression through Mobilization of Myeloid-Derived Suppressor Cells. Cancer Discov. 7, 72–85 (2017). 91. Baek, A. E. et al. The cholesterol metabolite 27 hydroxycholesterol facilitates breast cancer metastasis through its actions on immune cells. Nat. Commun. 8, 864 (2017). 92. Mutemberezi, V. et al. Oxysterol levels and metabolism in the course of neuroinflammation: insights from in vitro and in vivo models. J. Neuroinflammation 15, 74 (2018).

ACCEPTED MANUSCRIPT

783 784 785

93. Marengo, B. et al. Oxysterol mixture and, in particular, 27-hydroxycholesterol drive M2 polarization of human macrophages. BioFactors Oxf. Engl. 42, 80–92 (2016). 94. Guillem-Llobat, P. & Íñiguez, M. A. Inhibition of lipopolysaccharide-induced gene expression by liver X receptor ligands in macrophages involves interference with early growth response factor 1. Prostaglandins

787

Leukot. Essent. Fatty Acids 96, 37–49 (2015).

RI PT

786

95. Ward, L. J., Ljunggren, S. A., Karlsson, H., Li, W. & Yuan, X.-M. Exposure to atheroma-relevant 7-oxysterols

789

causes proteomic alterations in cell death, cellular longevity, and lipid metabolism in THP-1 macrophages.

790

PLOS ONE 12, e0174475 (2017).

792 793

96. Lee, S. et al. Adenylyl Cyclase-Associated Protein 1(CAP1) is a Receptor for Human Resistin and Mediates

M AN U

791

SC

788

Inflammatory Actions of Human Monocytes. Cell Metab. 19, 484–497 (2014). 97. Kim, K. M., Kim, Y. S., Jung, D. H., Lee, J. & Kim, J. S. Increased glyoxalase I levels inhibit accumulation of oxidative stress and an advanced glycation end product in mouse mesangial cells cultured in high glucose.

795

Exp. Cell Res. 318, 152–159 (2012).

798 799 800 801 802 803 804 805

C5a receptor. PLOS ONE 12, e0173749 (2017).

99. Zhang, R., Liu, Z., Li, Y. & Wu, B. LXR agonist regulates the proliferation and apoptosis of human T-Cell

EP

797

98. Cho, H. et al. 7α-Hydroxycholesterol induces monocyte/macrophage cell expression of interleukin-8 via

acute lymphoblastic leukemia cells via the SOCS3 pathway. Int. J. Biochem. Cell Biol. 78, 180–185 (2016). 100.

AC C

796

TE D

794

Bensinger, S. J. et al. LXR signaling couples sterol metabolism to proliferation in the acquired immune

response. Cell 134, 97–111 (2008). 101.

Ma, X. et al. Cholesterol negatively regulates IL-9–producing CD8+ T cell differentiation and antitumor

activity. J. Exp. Med. 215, 1555–1569 (2018). 102.

Vigne, S. et al. IL-27-Induced Type 1 Regulatory T-Cells Produce Oxysterols that Constrain IL-10

Production. Front. Immunol. 8, 1184 (2017).

812

813

814

815

816

817

818

819

820

821

822

823

824

825

826

827

828

829

phenotype. Oncol. Rep. 26, 389–397 (2011).

RI PT

811

Torres, C. G. et al. 27-hydroxycholesterol induces the transition of MCF7 cells into a mesenchymal

105. Alfaqih, M. A. et al. CYP27A1 Loss Dysregulates Cholesterol Homeostasis in Prostate Cancer. Cancer Res. 77, 1662–1673 (2017). 106.

Hiramitsu, S. et al. Estrogen Receptor Beta-Mediated Modulation of Lung Cancer Cell Proliferation by

27-Hydroxycholesterol. Front. Endocrinol. 9, (2018). 107.

SC

810

104.

Chen, L. et al. 25-Hydroxycholesterol promotes migration and invasion of lung adenocarcinoma cells.

M AN U

809

e0184985 (2017).

Biochem. Biophys. Res. Commun. 484, 857–863 (2017). 108.

Lin, C.-Y. et al. Cholestane-3β, 5α, 6β-triol Suppresses Proliferation, Migration, and Invasion of Human

Prostate Cancer Cells. PLOS ONE 8, e65734 (2013). 109.

de Medina, P. et al. Dendrogenin A arises from cholesterol and histamine metabolism and shows cell

TE D

808

Herold, M. et al. Liver X receptor activation promotes differentiation of regulatory T cells. PloS One 12,

differentiation and anti-tumour properties. Nat. Commun. 4, 1840 (2013). 110.

Poirot, M. & Silvente-Poirot, S. The tumor-suppressor cholesterol metabolite, dendrogenin A, is a new

class of LXR modulator activating lethal autophagy in cancers. Biochem. Pharmacol. 153, 75–81 (2018). 111.

EP

807

103.

Moresco, M. A. et al. Enzymatic Inactivation of Oxysterols in Breast Tumor Cells Constraints Metastasis

AC C

806

ACCEPTED MANUSCRIPT

Formation by Reprogramming the Metastatic Lung Microenvironment. Front. Immunol. 9, (2018). 112.

Raccosta, L. et al. The oxysterol–CXCR2 axis plays a key role in the recruitment of tumor-promoting

neutrophils. J. Exp. Med. 210, 1711–1728 (2013). 113.

Villablanca, E. J. et al. Tumor-mediated liver X receptor-alpha activation inhibits CC chemokine

receptor-7 expression on dendritic cells and dampens antitumor responses. Nat. Med. 16, 98–105 (2010). 114.

Courtney, R. & Landreth, G. E. LXR Regulation of Brain Cholesterol: From Development to Disease.

Trends Endocrinol. Metab. 27, 404–414 (2016).

ACCEPTED MANUSCRIPT 830 831 832

115.

Heverin, M. et al. Crossing the barrier: net flux of 27-hydroxycholesterol into the human brain. J. Lipid

Res. 46, 1047–1052 (2005). 116.

Björkhem, I., Leoni, V. & Svenningsson, P. On the fluxes of side-chain oxidized oxysterols across blood-

brain and blood-CSF barriers and origin of these steroids in CSF (Review). J. Steroid Biochem. Mol. Biol.

834

(2018). doi:10.1016/j.jsbmb.2018.12.009

837 838

839

Okabe, A. et al. Adaptive responses induced by 24S-hydroxycholesterol through liver X receptor

pathway reduce 7-ketocholesterol-caused neuronal cell death. Redox Biol. 2, 28–35 (2013). 118.

Liu, Y. et al. 25-Hydroxycholesterol activates the expression of cholesterol 25-hydroxylase in an LXR-

dependent mechanism. J. Lipid Res. 59, 439–451 (2018). 119.

SC

836

117.

M AN U

835

RI PT

833

Kim, H., Woo, J. H., Lee, J. H., Joe, E. & Jou, I. 22(R)-hydroxycholesterol induces HuR-dependent MAP

840

kinase phosphatase-1 expression via mGluR5-mediated Ca2+/PKCα signaling. Biochim. Biophys. Acta BBA -

841

Gene Regul. Mech. 1859, 1056–1070 (2016).

845 846

TE D

844

Noguchi, N., Urano, Y., Takabe, W. & Saito, Y. New aspects of 24(S)-hydroxycholesterol in modulating

neuronal cell death. Free Radic. Biol. Med. 87, 366–372 (2015). 121.

Testa, G. et al. A silver lining for 24-hydroxycholesterol in Alzheimer’s disease: The involvement of the

neuroprotective enzyme sirtuin 1. Redox Biol. 17, 423–431 (2018). 122.

EP

843

120.

Yamanaka, K., Saito, Y., Yamamori, T., Urano, Y. & Noguchi, N. 24(S)-Hydroxycholesterol Induces

AC C

842

847

Neuronal Cell Death through Necroptosis, a Form of Programmed Necrosis. J. Biol. Chem. 286, 24666–

848

24673 (2011).

849 850 851 852

123.

Gamba, P. et al. Interaction between 24-hydroxycholesterol, oxidative stress, and amyloid-β in

amplifying neuronal damage in Alzheimer’s disease: three partners in crime. Aging Cell 10, 403–417 (2011). 124.

Testa, G. et al. Changes in brain oxysterols at different stages of Alzheimer’s disease: Their involvement

in neuroinflammation. Redox Biol. 10, 24–33 (2016).

857

858

859

860

861

862

863

864

865

126.

Shafaati, M. et al. Marked accumulation of 27-hydroxycholesterol in the brains of Alzheimer’s patients

with the Swedish APP 670/671 mutation. J. Lipid Res. 52, 1004–1010 (2011). 127.

Refolo, L. M. et al. Hypercholesterolemia accelerates the Alzheimer’s amyloid pathology in a transgenic

mouse model. Neurobiol. Dis. 7, 321–331 (2000). 128.

Park, S. H. et al. Hypercholesterolemia accelerates amyloid β-induced cognitive deficits. Int. J. Mol.

Med. 31, 577–582 (2013). 129.

RI PT

856

Parkinson’s disease overlap? Exp. Gerontol. 68, 13–18 (2015).

SC

855

Marwarha, G. & Ghribi, O. Does the oxysterol 27-hydroxycholesterol underlie Alzheimer’s disease-

Ismail, M.-A.-M. et al. 27-Hydroxycholesterol impairs neuronal glucose uptake through an IRAP/GLUT4

M AN U

854

125.

system dysregulation. J. Exp. Med. 214, 699–717 (2017).

130. Teune, L. K. et al. Typical cerebral metabolic patterns in neurodegenerative brain diseases. Mov. Disord. 25, 2395–2404 (2010). 131.

Marwarha, G., Raza, S., Prasanthi, J. R. P. & Ghribi, O. Gadd153 and NF-κB Crosstalk Regulates 27-

TE D

853

ACCEPTED MANUSCRIPT

866

Hydroxycholesterol-Induced Increase in BACE1 and β-Amyloid Production in Human Neuroblastoma SH-

867

SY5Y Cells. PLOS ONE 8, e70773 (2013). 132.

Liu, J. et al. The ROS-mediated activation of IL-6/STAT3 signaling pathway is involved in the 27-

EP

868

hydroxycholesterol-induced cellular senescence in nerve cells. Toxicol. Vitro Int. J. Publ. Assoc. BIBRA 45,

870

10–18 (2017).

871

872

873

874

875

876

133.

AC C

869

Hirayama, T. et al. Serum concentration of 27-hydroxycholesterol predicts the effects of high-

cholesterol diet on plasma LDL cholesterol level. Hepatol. Res. Off. J. Jpn. Soc. Hepatol. 39, 149–156 (2009). 134.

Kim, S.-M. et al. 25-Hydroxycholesterol is involved in the pathogenesis of amyotrophic lateral sclerosis.

Oncotarget 8, 11855–11867 (2017). 135. Torres, N., Guevara-Cruz, M., Velázquez-Villegas, L. A. & Tovar, A. R. Nutrition and Atherosclerosis. Arch. Med. Res. 46, 408–426 (2015).

ACCEPTED MANUSCRIPT

883

884

885

886 887

888

889

890

891

892

893

894

895

896

897

898

atherosclerotic properties of oxysterols. Lipids Health Dis. 16, (2017). 138.

RI PT

882

Zmysłowski, A. & Szterk, A. Current knowledge on the mechanism of atherosclerosis and pro-

Sima, P., Vannucci, L. & Vetvicka, V. Atherosclerosis as autoimmune disease. Ann. Transl. Med. 6,

(2018). 139.

Libby, P. History of Discovery: Inflammation in Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 32,

SC

881

137.

2045–2051 (2012). 140.

Gargiulo, S. et al. Relation between TLR4/NF-κB signaling pathway activation by 27-hydroxycholesterol

M AN U

880

2 (2017).

and 4-hydroxynonenal, and atherosclerotic plaque instability. Aging Cell 14, 569–581 (2015). 141.

Ishikawa, T. et al. LXRβ/estrogen receptor-α signaling in lipid rafts preserves endothelial integrity. J.

Clin. Invest. 123, 3488–3497 (2013). 142.

Li, Z. et al. Krüppel-Like Factor 4 Regulation of Cholesterol-25-Hydroxylase and Liver X Receptor

TE D

879

Foks, A. C. & Bot, I. Preface: Pathology and Pharmacology of Atherosclerosis. Eur. J. Pharmacol. 816, 1–

Mitigates Atherosclerosis Susceptibility. Circulation 136, 1315–1330 (2017). 143.

Son, Y. et al. 7-Ketocholesterol induces the reduction of KCNMB1 in atherosclerotic blood vessels.

Biochem. Biophys. Res. Commun. 457, 324–327 (2015). 144.

EP

878

136.

Savouret, J. F. et al. 7-ketocholesterol is an endogenous modulator for the arylhydrocarbon receptor. J.

AC C

877

Biol. Chem. 276, 3054–3059 (2001). 145.

Larigot, L., Juricek, L., Dairou, J. & Coumoul, X. AhR signaling pathways and regulatory functions.

Biochim. Open 7, 1–9 (2018). 146.

Beischlag, T. V., Morales, J. L., Hollingshead, B. D. & Perdew, G. H. The Aryl Hydrocarbon Receptor

Complex and the Control of Gene Expression. Crit. Rev. Eukaryot. Gene Expr. 18, 207–250 (2008).

ACCEPTED MANUSCRIPT

899

147.

Muse, E. D. et al. Cell-specific discrimination of desmosterol and desmosterol mimetics confers

900

selective regulation of LXR and SREBP in macrophages. Proc. Natl. Acad. Sci. U. S. A. 115, E4680–E4689

901

(2018).

903

148.

Fessler, M. B. The challenges and promise of targeting the Liver X Receptors for treatment of

inflammatory disease. Pharmacol. Ther. 181, 1–12 (2018).

904

AC C

EP

TE D

M AN U

SC

905

RI PT

902

Figure Legends:

EP

TE D

M AN U

SC

RI PT

Figure 1: Chemical structures and metabolic pathways for several common oxysterols. ROS: reactive oxygen species. DHCR24: 24-dehydrocholesterol reductase. CYP46A1: cytochrome P450 46A1. CYP27A1: cytochrome P450 27A1. CYP11A1: cytochrome P450 11A1. 7βHC: 7β-hydroxycholesterol. 7KC: 7ketocholesterol. 24HC: 24-hydroxycholesterol. 27HC:27-hydroxycholesterol. 25HC: 25-hydroxycholesterol 22HC: 22-hydroxycholesterol.

AC C

906 907 908 909 910 911 912

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

H

RI PT

H

H

H

H

SC M AN U

HO

DHCR24

H

R OS

OH

EP

TE D

7βHC

H H

H

HO

H

AC C

H

H H

H OH

HO

H

H

O H

7KC HO

H

Cholesterol

Cho l

CYP11A1

S O R

H

H

Desmoste

HO

H

H

H

22HC

ACCEPTED MANUSCRIPT Highlights:

EP

TE D

M AN U

SC

RI PT

Oxysterols play important roles in cholesterol regulation and homeostasis Certain oxysterols serve as ligands for nuclear receptors There is increasing interest in the roles of oxysterols in different diseases Review of oxysterols in disease, with emphasis on the roles of nuclear receptors

AC C

• • • •