Subcellular localization and membrane topology of 17β-hydroxysteroid dehydrogenases

Subcellular localization and membrane topology of 17β-hydroxysteroid dehydrogenases

Accepted Manuscript Subcellular localization and membrane topology of 17β-hydroxysteroid dehydrogenases Maria Tsachaki, Alex Odermatt PII: S0303-7207...

878KB Sizes 0 Downloads 49 Views

Accepted Manuscript Subcellular localization and membrane topology of 17β-hydroxysteroid dehydrogenases Maria Tsachaki, Alex Odermatt PII:

S0303-7207(18)30217-X

DOI:

10.1016/j.mce.2018.07.003

Reference:

MCE 10270

To appear in:

Molecular and Cellular Endocrinology

Received Date: 22 August 2017 Revised Date:

18 June 2018

Accepted Date: 3 July 2018

Please cite this article as: Tsachaki, M., Odermatt, A., Subcellular localization and membrane topology of 17β-hydroxysteroid dehydrogenases, Molecular and Cellular Endocrinology (2018), doi: 10.1016/ j.mce.2018.07.003. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

1 2

Subcellular localization and membrane topology of 17β-hydroxysteroid dehydrogenases

3

RI PT

Maria Tsachakia and Alex Odermatta*

5

6

a

7

of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland

Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University

M AN U

8

SC

4

9

* Corresponding author: Prof. Alex Odermatt, Division of Molecular and Systems Toxicology,

10

Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel,

11

Switzerland. [email protected]

15 16 17 18 19 20 21

EP

14

AC C

13

TE D

12

22

1

ACCEPTED MANUSCRIPT

Abstract

24

The 17β-hydroxysteroid dehydrogenases (17β-HSDs) comprise enzymes initially identified by their ability

25

to interconvert active and inactive forms of sex steroids, a vital process for the tissue-specific control of

26

estrogen and androgen balance. However, most 17β-HSDs have now been shown to accept substrates

27

other than sex steroids, including bile acids, retinoids and fatty acids, thereby playing unanticipated roles

28

in cell physiology. This functional divergence is often reflected by their different subcellular localization,

29

with 17β-HSDs found in the cytosol, peroxisome, mitochondria, endoplasmic reticulum and in lipid

30

droplets. Moreover, a subset of 17β-HSDs are integral membrane proteins, with their specific topology

31

dictating the cellular compartment in which they exert their enzymatic activity. Here, we summarize the

32

present knowledge on the subcellular localization and membrane topology of the 17β-HSD enzymes and

33

discuss the correlation with their biological functions.

34 Keywords:

36

metabolism; steroid

37

subcellular

localization;

membrane

topology;

17β-hydroxysteroid

dehydrogenases;

TE D

35

M AN U

SC

RI PT

23

Abbreviations: 3α-adiol, 5α-androstan-3α,17β-diol; 11β-HSD1, 11β-hydroxysteroid dehydrogenase type

39

1; 17β-HSD, 17β-hydroxysteroid dehydrogenase; A4, ∆4-androsten-3,17-dione; Aβ, amyloid peptide β;

40

ACS3, acyl-CoA synthase 3; ADRP, adipose differentiation-related protein; AKR, aldo-keto reductase;

41

AR, androgen receptor; CBR4, carbonyl reductase type 4; DHEA, dehydroepiandrosterone, DHT, 5α-

42

dihydrotestosterone; ER, endoplasmic reticulum; ERAB, ER-associated Aβ peptide binding protein; FAS,

43

fatty acid synthesis; G6PD, glucose 6-phosphate dehydrogenase; KAR, 3-ketoacyl-acyl carrier protein

44

(ACP) reductase; LSS, lanosterol synthase; NAD, nicotinamide adenine dinucleotide; PAT, perlipin,

45

ADRP and tail-interacting protein; PPAR, peroxisome proliferator-activated receptor; PSA, prostate-

46

specific antigen; Ribonuclease P, RNase P; RODH, retinol dehydrogenase; roGFP, reduction-oxidation

47

sensitive green fluorescent protein; SDR, short-chain dehydrogenase/reductase.

AC C

EP

38

48

2

ACCEPTED MANUSCRIPT

49

1. Introduction The 17β-hydroxysteroid dehydrogenases (17β-HSDs) form an enzyme subfamily of the superfamily of

51

short-chain dehydrogenases-reductases (SDRs), with the exception of 17β-HSD5 that belongs to the aldo-

52

keto reductase (AKR) family (Moeller et al., 2009). At least 14 different 17β-HSDs have been described

53

and they were named after their capacity to interconvert 17-oxo and 17-hydroxy sex steroids. They are

54

numbered according to the chronological order of their discovery and the first three members (17β-HSD1,

55

2 and 3) were recognized for their ability to interconvert the weaker 17-ketosteroid and the more potent

56

17β-hydroxy forms of estrogens and/or androgens (Gast et al., 1989; Geissler et al., 1994; Wu et al.,

57

1993). Although these enzymes act bi-directionally in vitro, they predominantly function as either

58

oxoreductases or dehydrogenases in intact cells as well as in vivo (Khan et al., 2004), depending on the

59

availability of a co-factor, which is preferably either nicotinamide adenine dinucleotide NADP(H) or

60

NAD(H). Because of the intracellular abundance of these reduced and oxidized co-factors, typically,

61

reductive 17β-HSDs use NADPH and oxidative enzymes NAD+ as co-factor (Hedeskov et al., 1987). The

62

co-factor binding site is formed by a conserved structural motif called Rossmann fold (Buehner et al.,

63

1973), encompassing the conserved sequence GxGxxxG. The Rossmann fold is a common co-factor

64

binding element of SDRs. Notably, the AKR 17β-HSD5 does not contain a GxGxxxG motif in its

65

sequence.

66

That 17β-HSD enzymes are not exclusively dedicated to sex steroid metabolism was realized after the

67

discovery of 17β-HSD4, which was shown to participate in peroxisomal β-oxidation of fatty acids

68

(Breitling et al., 2001; Leenders et al., 1996; Markus et al., 1996; Seedorf et al., 1995). Since then, the

69

notion of a multifunctional 17β-HSD family was gradually established and experimentally supported. The

70

diversity in substrate specificity and in vivo function is also mirrored in the subcellular localization of the

71

different 17β-HSD enzymes. However, the presence of an enzyme in a specific organelle cannot always be

72

deduced through the knowledge of its substrate. Although signal peptides and targeting sequences are

73

present in some 17β-HSDs, definitive proof of the presence in a cell compartment requires the

AC C

EP

TE D

M AN U

SC

RI PT

50

3

ACCEPTED MANUSCRIPT

employment of biochemical methods (such as subcellular fractionation and immunoblotting) or indirect

75

immunofluorescence. A critical issue in these studies is the utilization in each method of proper markers,

76

in order to undoubtedly designate a certain protein to a cell organelle or the cytosol.

77

Since all organelles are encircled by lipid mono- or bilayers, integral membrane proteins can either

78

function in the lumen of an organelle or at its cytosolic side. Therefore, the topology (the orientation of

79

different parts of a protein in relation to the membrane) is a fundamental feature of membrane proteins.

80

All 17β-HSDs residing in the endoplasmic reticulum (ER) or lipid droplets are membrane-bound (17β-

81

HSD2, 3, 6, 7, 11, 12 and 13). Different methods have been employed for topology determination of

82

membrane-anchored 17β-HSDs, including selective permeabilization of cell membranes with digitonin

83

followed by immunofluorescence detection, proteinase K protection assay followed by western blotting

84

and, recently, live-cell measurements using reduction-oxidation sensitive green-fluorescent protein

85

(roGFP) fusions with the respective 17β-HSD enzyme (Tsachaki et al., 2015).

86

For subcellular localization and membrane topology determination, peptide tags, fusion or truncated

87

proteins are frequently used. This raises the question of how such interventions could affect intracellular

88

targeting. Given the complex nature of the above studies, erroneous initial allocation in a cell organelle or

89

membrane topology have been reported, which has led to misconceptions regarding the physiological role

90

of certain 17β-HSDs. In this review, we elaborate on current understanding of 17β-HSD subcellular

91

localization and membrane topology (summarized in Fig. 1 and 2, respectively), based on previous

92

investigations that include both in silico studies and experimental evidence. In addition, we elaborate on

93

how this aspect of 17β-HSD protein physiology is potentially linked to their enzymatic function and the

94

metabolic pathways in which they are involved.

AC C

EP

TE D

M AN U

SC

RI PT

74

95

96

97

2. Subcellular localization of 17β-HSDs 2.1 Cytosolic 17β-HSD1, 5 and 14 4

ACCEPTED MANUSCRIPT

17β-HSD1 was the first enzyme identified in this family and its main function is to reduce the weakly

99

active 17-keto estrogen estrone to the potent 17β-hydroxy estrogen estradiol and thereby stimulating the

100

activity of estrogen receptors, a process particularly important in tissues such as the placenta, the breast

101

and the ovaries (Luu The et al., 1989). An elevated expression of 17β-HSD1 has been associated with sex-

102

specific malignancies, including breast and endometrial carcinomas (Gunnarsson et al., 2001; Oduwole et

103

al., 2004), and inhibitors of this enzyme are considered for potential therapeutic applications to treat

104

hormone-dependent cancer (Brozic et al., 2008; Cornel et al., 2012; Hilborn et al., 2017; Zhang et al.,

105

2012). Although other 17β-HSDs (mainly 17β-HSD7 and 17β-HSD12) have been reported to be able to

106

reduce estrone to estradiol, 17β-HSD1 clearly has the highest affinity, and combined with its expression

107

pattern it is considered to be the main enzyme exerting this function (Laplante et al., 2009). It was

108

suggested that 17β-HSD1 dwells in the cytosol, since this enzyme has no predicted membrane helices.

109

Subsequently, immunofluorescence experiments using a specific antibody against human 17β-HSD1

110

confirmed the cytosolic distribution (Tsachaki et al., 2015).

111

The inactivation of estradiol to estrone can be performed by several 17β-HSDs (17β-HSD2, 4, 6, 8, 9, 10

112

and 14), although the relative in vivo contribution of each of these enzymes to estradiol inactivation

113

remains unclear. Among these enzymes, 17β-HSD14 is the only one residing in the cytosol, as shown by

114

co-localization with the cytoplasmic protein phalloidin in immunofluorescence labelling experiments

115

(Lukacik et al., 2007). In the same study, no co-localization with nuclei or mitochondria could be

116

observed. 17β-HSD14 likely plays an important role in the local inactivation of estradiol in tissues where

117

it is highly expressed, such as the breast, placenta and brain (Jansson et al., 2006; Lukacik et al., 2007;

118

Sivik et al., 2012). Importantly, low expression of 17β-HSD14 was correlated with better prognosis, and

119

its expression was shown to be predictive of response to tamoxifen treatment in estrogen-receptor positive

120

breast cancer patients (Jansson et al., 2006; Sivik et al., 2012). In this case, the presence of 17β-HSD1 and

121

17β-HSD14 in the cytosol could provide an efficient system for the intracellular regulation and

122

maintenance of high estradiol concentrations. 17β-HSD14 is also able to oxidize other substrates, at least

123

in vitro, including 5-androstene-3β,17β-diol and testosterone (Lukacik et al., 2007). Resolving the crystal

AC C

EP

TE D

M AN U

SC

RI PT

98

5

ACCEPTED MANUSCRIPT

structure of 17β-HSD14 revealed that the substrate binding pocket is wide enough to accommodate

125

substrates other than steroids, raising the possibility that new roles for 17β-HSD14 will be uncovered in

126

the future (Bertoletti et al., 2016; Lukacik et al., 2007).

127

As mentioned above, all 17β-HSDs belong to the SDR superfamily with the exception of 17β-HSD5 (also

128

known as AKR1C3) that is a member of the AKR superfamily (Penning, 2015). AKRs are dedicated in the

129

reduction of carbonyl-containing substrates, including sugar aldehydes, keto-prostaglandins and keto-

130

steroids. 17β-HSD5 catalyzes the reduction of the prostaglandins PGH2 and PGD2 to PGF2a and 9α,11β-

131

PGF2, respectively, both products shown to promote cancer cell proliferation (Matsuura et al., 1998;

132

Suzuki-Yamamoto et al., 1999). Importantly, 17β-HSD5 participates in metabolic cascades through which

133

the most potent human androgen 5α-dihydrotestosterone (DHT) is synthesized, and has been considered as

134

a potential target against prostate cancer (Adeniji et al., 2013). Upon binding of DHT to its cognate

135

androgen receptor (AR) in the cytosol, it translocates into the nucleus where it regulates the expression of

136

a plethora of target genes. Immunofluorescence experiments showed that in the absence of ligand

137

activation 17β-HSD5 co-localizes with the AR in the cytosol, whereas treatment with the 17β-HSD5

138

substrate ∆4-androsten-3,17-dione (A4) or the AR agonist R1881 led to translocation of both AR and 17β-

139

HSD5 to the nucleus (Yepuru et al., 2013). Intriguingly, chromatin immunoprecipitation experiments

140

demonstrated the binding of 17β-HSD5 to the promoter of the AR-target gene prostate specific antigen

141

(PSA), when it was suggested to act as a coactivator of the AR. Nuclear localization has not been shown

142

for any other 17β-HSD enzyme, and the specific function of a reductase binding to a gene regulatory

143

region remains enigmatic.

SC

M AN U

TE D

EP

AC C

144

RI PT

124

145

2.2 Peroxisomal 17β-HSD4

146

The only enzyme of the 17β-HSDs localized in the peroxisomes is 17β-HSD4 (Adamski et al., 1995).

147

Evidence for peroxisomal localization of 17β-HSD4 was provided by immunogold electron microscopy,

6

ACCEPTED MANUSCRIPT

where 17β-HSD4 co-localized with the peroxisomal-resident enzymes catalase and acyl-CoA oxidase

149

(Markus et al., 1995). Later, the subcellular localization of 17β-HSD4 was also deduced from the presence

150

at its C-terminus of the peroxisomal targeting tripeptide Ala-Lys-Leu (AKL) (Moller et al., 1999), which

151

represents an alternate version of the classical Ser-Lys-Leu (SKL) peroxisomal targeting signal. In the

152

porcine enzyme this sequence is varied as Ala-Lys-Ile, and its deletion from the protein causes cytosolic

153

mislocalization (Moller et al., 1999).

154

Although the N-terminal part of the protein is reminiscent of other SDRs, the amino acid sequence of 17β-

155

HSD4 points towards a multi-functional enzyme. The diverse roles of 17β-HSD4 in peroxisomes have

156

been well-documented and are attributed to distinct protein domains (Breitling et al., 2001). A sterol and

157

phospholipid transfer domain present at the C-terminus of 17β-HSD4 is related to sterol carrier protein 2

158

(SCP2), and such activity has been clearly demonstrated for the enzyme (Leenders et al., 1996; Markus et

159

al., 1996; Seedorf et al., 1995). Additionally, based on sequence similarity to the yeast FOX2 proteins,

160

17β-HSD4 was suggested to participate in peroxisomal β-oxidation of fatty acids and bile acids (Leenders

161

et al., 1996). Indeed, acyl-CoA dehydrogenase and 2-enoyl-CoA hydratase activities of 17β-HSD4 are

162

supported by strong experimental evidence (Caira et al., 1998; Leenders et al., 1996; Markus et al., 1996;

163

Qin et al., 1997). Knockout of 17β-HSD4 in mice showed severe impairments in β-oxidation of 2-methyl-

164

branched fatty acids, the bile acid intermediates di- and trihydroxycoprostanic acid, and very long-chain

165

fatty acids (Baes et al., 2000). The importance of 17β-HSD4 for peroxisomal function is further

166

underlined by reports for mutations of the HSD17B4 gene, leading to conditions reminiscent of the

167

Zellweger syndrome (Suzuki et al., 1997; van Grunsven et al., 1998; van Grunsven et al., 1999).

168

Despite the above functions attributed to 17β-HSD4, it has also been considered important for estrogen

169

metabolism, since it was first described as an estradiol-inactivating enzyme in porcine uterus (Adamski et

170

al., 1992). Luteneization of equine ovarian follicles, a process accompanied by a decrease in estradiol and

171

increase in progesterone levels, was linked to increase in the expression of 17β-HSD4 (Brown et al.,

172

2004). Additionally, single nucleotide polymorphisms in the HSD17B4 gene were well-correlated with

AC C

EP

TE D

M AN U

SC

RI PT

148

7

ACCEPTED MANUSCRIPT

endometrial cancer risk (Karageorgi et al., 2011). According to its amino acid sequence, 17β-HSD4 is

174

unlikely to contain any transmembrane helices and is thus expected to be a soluble protein within the

175

peroxisomal matrix. Since estrogen metabolism in this compartment has not been demonstrated, and the

176

presence of 17β-HSD4 in the cytosol has never been shown, the actual involvement of 17β-HSD4 in

177

estrogen metabolism remains obscure.

RI PT

173

178

2.3 Mitochondrial 17β-HSD8 and 17β-HSD10

180

Fatty acid synthesis (FAS) in mitochondria is responsible for producing a number of long-chain fatty

181

acids, including the respiratory complex I component 3-hydroxymyristoyl-ACP and lipoic acid (Hiltunen

182

et al., 2010). The human 3-ketoacyl-acyl carrier protein (ACP) reductase (KAR) has been recently

183

discovered to be 17β-HSD8. The presence of 17β-HSD8 in mitochondria was demonstrated by co-

184

localization with the MitoTracker Red dye in immunofluorescence labeling experiments (Chen et al.,

185

2009). The primary sequence of 17β-HSD8 indicates that it is a soluble enzyme and should thus reside in

186

the mitochondrial matrix. Moreover, it has been shown to form a heteroterameric complex with carbonyl

187

reductase type 4 (HsCBR4), where it acts as a scaffold to the functional subunit CBR4 (Venkatesan et al.,

188

2014). Based on these findings and the fact that it likely acts as an oxidase in vivo, it was suggested that

189

17β-HSD8 could function in channeling 3R-hydroxyacyl-CoA intermediates to β-oxidation in

190

mitochondria. Three-dimensional models of 17β-HSD8 in complex with NAD+ showed that it accepts

191

substrates similar to those of the β-ketoacyl-ACP reductases of Brassica napus, further supporting a role

192

in fatty acid metabolism (Pletnev et al., 2005).

193

The gene encoding 17β-HSD10 is located on the X chromosome and several mutations of the enzyme are

194

linked with mental retardation (Yang et al., 2011). The function of this enzyme remains to be further

195

investigated. Elevated 17β-HSD10 levels have been reported in Alzheimer’s patients (He et al., 2002). It

196

has been shown to bind the amyloid β (Aβ) peptide, which accumulates in the brain of Alzheimer’s

AC C

EP

TE D

M AN U

SC

179

8

ACCEPTED MANUSCRIPT

patients and is thought to play a major role in the disease progression (Kissinger et al., 2004). 17β-HSD10

198

is also important for isoleucine metabolism and oxidative inactivation of the potent neurosteroid

199

allopregnanolone (He et al., 2005; Ofman et al., 2003). Besides, 17β-HSD10 was shown to catalyze the

200

conversion of the inactive androgen 5α-androstan-3α,17β-diol (3α-adiol) to the potent androgen DHT,

201

with a potential role in the backdoor pathway of androgen generation that is thought to promote prostate

202

cancer (Auchus, 2004; Mohler, 2014; Penning, 2010).

203

Initially, 17β-HSD10 was reported to reside in the ER in HeLa cells (giving rise to the suggested name

204

ER-associated Aβ peptide binding protein-ERAB), and addition of Aβ peptide led to its re-distribution to

205

the plasma membrane in the human neuroblastoma cell line SK-N-SH (Yan et al., 1997), an observation

206

that was supported by confocal microscopy and subcellular fractionation studies. However, in the

207

immunofluorescence experiments presented in this work no plasma membrane marker was used to

208

confirm such localization. Additionally, no compartment-specific marker was demonstrated to validate

209

proper separation of all cell fractions. In a follow-up work by the same group, this localization could be

210

reproduced, however the additional presence of 17β-HSD10 in the mitochondria of the same cells was

211

shown (Yan et al., 1999). Although it cannot be excluded that 17β-HSD10 also localizes in the ER or at

212

mitochondria/ER contact sites in the cell lines tested above, all subsequent studies using specific markers

213

for both immunofluorescence labeling and subcellular fractionation experiments have reported exclusively

214

mitochondrial localization (He et al., 2001; He et al., 1999; He et al., 2000). Interestingly, an early study

215

identified bovine 17β-HSD10 as the 3-hydroxyacyl-CoA dehydrogenase of the mitochondrial fatty acid β-

216

oxidation (Furuta et al., 1997). It was also demonstrated that its protein sequence contains a 16-amino acid

217

N-terminal non-cleaved mitochondrial signal peptide, similar to that of human and rat thiolases.

218

Nevertheless, fusion of the 15 first amino acids of human 17β-HSD10 to GFP failed to result in

219

mitochondrial localization of the fusion protein, whereas fusion of the first 34 amino acids was sufficient

220

(Shafqat et al., 2003). This implies that additional elements following this signal peptide are necessary for

221

mitochondrial targeting.

AC C

EP

TE D

M AN U

SC

RI PT

197

9

ACCEPTED MANUSCRIPT

An important role of 17β-HSD10, directly linked to its subcellular localization, is in mitochondrial tRNA

223

processing, as an essential component of Ribonuclease P (RNase P) (Holzmann et al., 2008; Vilardo et al.,

224

2012). The role of 17β-HSD10 in mitochondrial tRNA processing is believed to underlie the etiology of

225

the HSD10 disease, which is characterized by severe neurological defects and cardiomyopathy

226

(Deutschmann et al., 2014; Falk et al., 2016; Vilardo et al., 2015). The plethora of substrates and functions

227

attributed to 17β-HSD10 points towards a multifunctional enzyme (Yang et al., 2014; Yang et al., 2005),

228

and the precise mechanisms by which its malfunction or absence leads to disease remain to be more

229

clearly determined.

SC

RI PT

222

M AN U

230

2.4 Lipid droplet-associated 17β-HSD11 and 17β-HSD13

232

Lipid droplets are cytoplasmic organelles dedicated to lipid storage and are regarded to be formed by

233

budding off from ER membranes (Pol et al., 2014). They contain a lipid monolayer and they are mostly

234

found in adipocytes, although they are also present in metabolically active tissues, such as the liver and the

235

muscle. 17β-HSD11 was the first enzyme in the 17β-HSD family shown to be located in lipid droplets. In

236

the first study describing its subcellular localization, N-terminally myc-tagged 17β-HSD11 transiently

237

transfected into the mouse adrenal Y1 cell line was regarded to be in the cytoplasm based on

238

immunofluorescence labeling (Chai et al., 2003). However, no organelle or cytoplasmic markers were

239

employed in this study, and the microscopic image was likely misinterpreted. Also, the N-terminal tag

240

might have affected the localization of the protein due to its close proximity to the membrane spanning

241

helix. Later, it was shown that 17β-HSD11 is one of the most abundant proteins in the lipid droplet

242

fraction of the human hepatoma cell line HuH7, together with Adipose differentiation-related protein

243

(ADRP) and acyl-CoA synthase 3 (ACS3). This was demonstrated by mass spectrometric analysis and

244

confirmed by immunoblotting using a specific antibody (Fujimoto et al., 2004). When cells were treated

245

with oleic acid, which promotes lipid droplet formation, the lipid droplet localization became more

246

evident. In this study, 17β-HSD11 exhibited an exclusive lipid droplet localization; it could not be

AC C

EP

TE D

231

10

ACCEPTED MANUSCRIPT

detected by immunoblotting in the ER cellular protein fraction nor did it show any ER labeling in

248

fluorescence labeling experiments. Interestingly, 17β-HSD11 expression was found to be upregulated in

249

the mouse intestine upon activation of peroxisome proliferator-activated receptor alpha (PPARα), which

250

has major roles in lipid metabolism (Motojima, 2004). In subcellular fractionation studies 17β-HSD11 was

251

present in both the ER and the lipid droplet fraction when mice were fed normal diet, and specifically

252

localized in the lipid droplet fraction when they were treated with a PPARα agonist (Yokoi et al., 2007).

253

Moreover, in the Chinese hamster ovary cell line 17β-HSD11 was found to localize in the ER under

254

normal conditions and to be transferred from the ER to the lipid droplets after oleic acid treatment.

255

The N-terminal hydrophobic part of mouse 17β-HSD11 (aminoacids 4-16) seems to be important for

256

correct intracellular targeting, since deletion of this region leads to mitochondrial localization (Horiguchi

257

et al., 2008). Based on in silico predictions this should be part of a transmembrane region, which also

258

contains the lipid droplet-targeting motif of the PAT family (perilipin, ADRP and tail-interacting protein).

259

Deletion of this motif or of two of its conserved amino acids causes cytosolic mislocalization of mouse

260

17β-HSD11 (Horiguchi et al., 2008). Analysis of the membrane topology of 17β-HSD11 suggested that it

261

contains a membrane helix close to the N-terminus, followed by the largest part of the protein, containing

262

the Rossmann fold and active sites, which faces the cytosol (Tsachaki et al., 2015).

263

Although 17β-HSD11 has been shown to be able to convert several substrates, including estradiol and 3α-

264

adiol (Brereton et al., 2001; Li et al., 1998; Lundova et al., 2016), the in vivo enzymatic activity is far from

265

being resolved and has surely not been correlated with its subcellular localization. Even less is known

266

about the other lipid droplet-associated enzyme 17β-HSD13 (Liu et al., 2007), which has a surprisingly

267

similar primary amino acid sequence to 17β-HSD11 (65% identical amino acids). The N-terminus of 17β-

268

HSD13, which is also highly hydrophobic and contains a motif similar to the PAT of 17β-HSD11, has

269

been shown to be sufficient for correct targeting of the protein in lipid droplets (Horiguchi et al., 2008).

270

Topology studies showed similar results than those obtained for 17β-HSD11, suggesting that the enzyme

271

transverses the membrane once and its active site protrudes into the cytosol (Tsachaki et al., 2015). The

AC C

EP

TE D

M AN U

SC

RI PT

247

11

ACCEPTED MANUSCRIPT

recent DiscovEHR human genetics study revealed an association of a 17β-HSD13 splice variant

273

(rs72613567:TA) resulting in an unstable truncated protein with a reduced risk of nonalcoholic and

274

alcoholic liver disease (Abul-Husn et al., 2018). This splice variant was associated with a lower risk of

275

nonalcoholic steatohepatitis, but not with simple steatosis, suggesting that this variant inhibits disease

276

progression. Wild-type 17β-HSD13 and the truncated variant were found to be expressed on the surface of

277

the lipid droplet membrane. This study also provided evidence for several bioactive lipids, including

278

leukotriene B3 and B4, as well as eicosanoids, as potential physiologically relevant substrates.

279

Interestingly, ablation of 17β-HSD13 in mice led to hepatic steatosis and inflammation, with evidence for

280

impaired mitochondrial β-oxidation, without alterations in circulating steroid concentrations or

281

reproductive performance (Adam et al., 2018). The above observations should stimulate further research

282

to uncover the physiologically most relevant substrates of 17β-HSD11 and 17β-HSD13, as well as the role

283

of these two enzymes in lipid metabolism, control of lipid storage and energy homeostasis.

M AN U

SC

RI PT

272

TE D

284

2.5 17β-HSD2, 3, 6, 7, 9 and 12 at the ER membrane

286

Besides 17β-HSD11 and 17β-HSD13, both located at the ER and in lipid droplets, six other 17β-HSDs

287

have been shown to localize at the ER. All are transmembrane proteins, with their substrate and co-factor

288

binding site facing either the ER lumen or the cytosol. Until recently, attempts to resolve the membrane

289

topology of these enzymes involved mainly proteinase K digestion of isolated microsomes followed by

290

immunoblotting to determine whether the exposed protein part is protected (indicating luminal

291

orientation) or not (supporting cytosolic orientation). This biochemical method relies on the proper

292

microsomal preparation, yielding >90% inside-out microsomes (ER luminal part protected, cytoplasmic

293

side facing the solution). The activity and purity of the proteinase K batch needs to be carefully evaluated.

294

Proteinase K is still active in the presence of detergent and at 4°C, thus proper inactivation to terminate the

295

reaction is essential. Another method is the selective permeabilization of plasma membranes using

296

detergents such as digitonin, followed by indirect immunofluorescence to determine whether a specific

AC C

EP

285

12

ACCEPTED MANUSCRIPT

epitope of the protein of interest faces the cytosolic or luminal side of the ER. Similarly, the plasma

298

membrane-specific pore forming agent streptolysin-O allows selective access to the cytosolic

299

compartment. Importantly, suitable positive and negative controls must be included in the above

300

mentioned methods.

301

Recently, we employed a method for membrane topology determination of SDRs in living cells, based on

302

fusing part or the SDR sequence with the redox-sensitive roGFP2 (Tsachaki et al., 2015). Depending on

303

the oxidation conditions of the compartment, the emission intensity of roGFP2 when excited at two

304

different wavelengths (405 nm and 480 nm) changes. The intensity ratio at the two different wavelengths

305

allows calculating the degree of roGFP2 oxidation. A high oxidation rate of roGFP2 indicates ER luminal

306

localization and a low oxidation rate a cytoplasmic one. By fusing roGFP2 either to the very C-terminus

307

of the different 17β-HSD enzymes or immediately upstream of the co-factor binding and active sites, we

308

could solve the topology of 17β-HSD2, 17β-HSD6, 17β-HSD7 and 17β-HSD12, along with the ER/lipid

309

droplet-localized 17β-HSD11 and 17β-HSD13. These results were corroborated with selective

310

permeabilization of cell membranes using digitonin.

311

For the above methods, fusion proteins of the respective 17β-HSD enzyme with GFP or other epitope tags

312

(for instance myc or flag) at either the C- or N-terminus can be used. Given that many proteins contain at

313

the N-terminus signal sequences for proper targeting, modifications at the C-terminus are usually

314

preferred. Proper localization of the fusion protein at the expected cellular compartment has to be

315

confirmed to avoid artifacts. Unfortunately, even with careful experimental design and validation,

316

structural changes introduced by protein modifications that could inverse proper membrane insertion can

317

never be fully excluded. Below, we revise our present understanding on the membrane topology of ER-

318

resident 17β-HSDs.

AC C

EP

TE D

M AN U

SC

RI PT

297

319

320

2.5.1 17β-HSD2 and 17β-HSD3: ER proteins with cytosolic orientation of their active site 13

ACCEPTED MANUSCRIPT

17β-HSD2 is responsible for oxidizing the active 17β-hydroxy forms of many sex steroids to their inactive

322

or weakly active 11-keto forms, exerting a protective function in peripheral tissues to avoid excessive

323

action of these steroids (Moghrabi et al., 1997). Importantly, it exhibits opposite effects to 17β-HSD1 by

324

inactivating estradiol to estrone (Miettinen et al., 1996), and a decreased expression has been associated

325

with breast cancer (Gunnarsson et al., 2001). Additionally, it has a role in androgen inactivation by

326

mediating the conversion of testosterone to A4, androstenediol to dehydroepiandrosterone (DHEA), DHT

327

to androstanedione and 3α-adiol to androsterone (Elo et al., 1996; Wu et al., 1993), and decreased

328

expression has been associated with colon cancer (Oduwole et al., 2003).

329

When first cloned, a putative C-terminal ER retention signal was identified and led to the assumption that

330

the part of the protein containing the active site is luminal (Wu et al., 1993). A subsequent study

331

conducting immunofluorescence experiments demonstrated that 17β-HSD2 co-localized at the ER

332

membrane with the ER-luminal protein BiP, and the presence of 17β-HSD2 in the ER lumen was

333

suggested (Puranen et al., 1999). However, given the size of the ER lipid bilayer, the diffraction limit of

334

the microscope does not allow from double immunofluorescence labeling experiments to distinguish

335

between proteins facing the ER lumen from proteins facing the cytoplasm. To solve this issue, we fused

336

roGFP2 to the C-terminus of 17β-HSD2, and also immediately downstream of the two putative N-terminal

337

transmembrane helices. These experiments revealed a cytosolic localization in both cases based on

338

roGFP2 oxidation. This was additionally supported by selective permeabilization experiments using

339

digitonin (Tsachaki et al., 2015). Taken together, these results strongly support a topology of 17β-HSD2

340

where the substrate and co-factor binding sites are oriented toward the cytosol.

341

Another microsomal enzyme initially suggested to have a luminal orientation is 17β-HSD3, whose

342

principal function is the reduction of A4 to testosterone in the testicular Leydig cells (Geissler et al.,

343

1994). Hu et al. hypothesized a coupling of 17β-HSD3 and 11β-hydroxysteroid dehydrogenase type 1

344

(11β-HSD1) activities, where 17β-HSD3 would utilize ER luminal NADPH to generate testosterone (Hu

345

et al., 2008). The formed NADP+ would then serve as co-factor for 11β-HSD1 to catalyze the inactivation

AC C

EP

TE D

M AN U

SC

RI PT

321

14

ACCEPTED MANUSCRIPT

of the potent glucocorticoid cortisol to cortisone under stress conditions. However, although an

347

interrelationship between the activity of 11β-HSD1 as an oxoreductase or dehydrogenase and the A4

348

conversion to testosterone was proposed (Latif et al., 2011), a direct functional link between the two

349

enzymes and competition for NADPH co-factor in the ER lumen was not unequivocally shown.

350

Importantly, it was later demonstrated that the active site of 17β-HSD3 protrudes into the cytosol, based

351

on both proteinase K protection and selective permeabilization experiments (Legeza et al., 2013).

352

Moreover, knockdown of glucose 6-phosphate dehydrogenase (G6PD), the enzyme producing NADPH in

353

the cytosol, significantly inhibited 17β-HSD3 activity, while modulation of hexose-6-phosphate

354

dehydrogenase (H6PD), generating NADPH in the ER lumen, had no effect. Conversely and as expected,

355

knockdown of H6PD decreased 11β-HSD1 oxoreduction activity, while knockdown of G6PD had no

356

effect.

M AN U

SC

RI PT

346

357

2.5.2 17β-HSD6, 7, 9 and 12: ER proteins with luminal orientation of their active site

359

A common feature of all 17β-HSDs located at the ER (including 17β-HSD11 and 17β-HSD13) is the

360

presence of a hydrophobic domain, predicted to form transmembrane helices, starting at amino acids 1-10

361

(Fig. 2). The co-factor binding site typically follows immediately downstream of this hydrophobic region.

362

An exception to this rule constitutes 17β-HSD7 (Krazeisen et al., 1999), which only contains a highly

363

hydrophobic region closer to the C-terminus (amino acids 238-258 out of 341). Addition of roGFP2 at the

364

C-terminus of 17β-HSD7 led to low oxidation of the sensor indicative of cytosolic localization, which was

365

supported by selective permeabilization experiments of a C-terminally tagged protein (Tsachaki et al.,

366

2015). Furthermore, an N-terminal FLAG-tag on 17β-HSD7 was protected in selective permeabilization

367

experiments, suggesting luminal orientation. The addition of the epitope at the N-terminus did not change

368

ER localization of the enzyme. Taken together, these results indicate that 17β-HSD7 is an ER membrane

369

protein with its substrate and co-factor binding site oriented toward the ER lumen, although the retention

370

signal that keeps it in the ER remains obscure.

AC C

EP

TE D

358

15

ACCEPTED MANUSCRIPT

17β-HSD7 is a reductive enzyme able to convert estrone to estradiol similar to 17β-HSD1 albeit with

372

lower efficiency (Laplante et al., 2009). Its relative in vivo contribution to the estradiol pool is unclear, but

373

elevated expression of the enzyme has been shown in breast cancer tissue (Shehu et al., 2011; Wang et al.,

374

2017). The focus on the function of 17β-HSD7 was shifted away from sex steroid metabolism when it was

375

identified as the homolog of the yeast 3-ketosteroid reductase Erg27p, which participates in cholesterol

376

synthesis (Marijanovic et al., 2003). It was shown that 17β-HSD7 is able to convert zymosterone to

377

zymosterol in vitro, and mice deficient of the murine enzyme die at embryonic day 10.5 (Jokela et al.,

378

2010). Analysis of knockout embryos preceding this stage showed severe developmental abnormalities,

379

which were correlated with defects in the cholesterol synthesis pathway. The part of cholesterol

380

biosynthesis in which 17β-HSD7 acts (namely, the post-squalene phase) takes place at the ER. However,

381

the membrane topology of the enzymes participating in this pathway is insufficiently investigated.

382

Intriguingly, according to the hidden Markov algorithm TMHMM and TOPCONS predictions, lanosterol

383

synthase (LSS), which converts squalene epoxide to lanosterol in this series of reactions, is not predicted

384

to contain any transmembrane domains. This implies that it is a soluble enzyme and supports that at least

385

part of the post-squalene cholesterol synthesis takes place in the ER lumen. Further complications come

386

from the fact that the yeast homologue of lanosterol synthase Erg7p was found to localize mainly in lipid

387

droplets, and to be occasionally distributed between the ER and lipid droplets (Milla et al., 2002; Mullner

388

et al., 2004). Thus, further studies are need to shed more light on this aspect of cholesterol metabolism.

389

Another ER enzyme that has been linked with reductive conversion of estrone to estradiol is 17β-HSD12

390

(Luu-The et al., 2006), although with very low efficiency compared to 17β-HSD1 (Laplante et al., 2009)

391

(own observations). Interestingly, it was shown that 17β-HSD12 is the 3-ketoacyl-coA reductase (KAR) in

392

the four-step cascade leading to elongation of long-chain fatty acids in the ER (Moon et al., 2003).

393

Although only one multifunctional enzyme is required for elongation of fatty acids with less than 16

394

carbon atoms in the cytosol, four different enzymatic entities are responsible for elongating longer fatty

395

acids in the ER (Kihara, 2012). Through this process, vital fatty acids are produced, including arachidonic

396

acid, eicosapentaenoic acid and docosahexaenoic acid. Arachidonic acid is the precursor of a variety of

AC C

EP

TE D

M AN U

SC

RI PT

371

16

ACCEPTED MANUSCRIPT

biologically active lipids, including prostaglandins, thromboxanes, leukotriens, eicosanoids and lipoxins.

398

The importance of 17β-HSD12 function was demonstrated in mice, where knockout led to lethality at

399

embryonic day E9.5, with severely impaired organogenesis (Rantakari et al., 2010). Embryonic stem cells

400

heterozygous for 17β-HSD12 showed decreased arachidonic acid levels, suggesting an important role of

401

this enzyme in the synthesis of arachidonic acid and other biologically active lipids and in neuronal

402

development. Additionally, observations from 17β-HSD12 knockout mice revealed a role in female

403

reproduction with impaired meiosis in the ovaries of heterozygous mice and failure in oogenesis and

404

ovulation (Kemilainen et al., 2016). Importantly, arachidonic acid and several important metabolites such

405

as prostaglandins D2, E2 and F2α as well as thromboxane B2 were decreased in the ovaries, while steroid

406

hormones were not altered. High expression of 17β-HSD12 was correlated with poor prognosis in ovarian

407

and breast cancer (Nagasaki et al., 2009; Song et al., 2006; Szajnik et al., 2012) and knockdown of 17β-

408

HSD12 in breast and ovarian carcinoma cell lines led to reduced cell proliferation. This effect could not be

409

rescued by estradiol, but rather by arachidonic acid supplementation (Nagasaki et al., 2009; Szajnik et al.,

410

2012). Recently, 17β-HSD12 expression has been associated with COX-2 expression in ovarian cancer,

411

further emphasizing its function in arachidonic acid production and cell proliferation (Kemilainen et al.,

412

2018). All these findings underline the important role of 17β-HSD12 in long-chain fatty acid elongation.

413

In an early study, protection of activity after protease treatment of microsomes was used in order to assess

414

the membrane topology of the enzymes involved in rat long-chain fatty acid elongation (Osei et al.,

415

1989). Based on the results, the first condensing enzyme in this cascade as well as the last trans-2-enoyl-

416

CoA reductase of the elongation are facing the cytosolic part of the ER membrane, whereas the 3-

417

hydroxyacyl-CoA dehydratase had an activity embedded in the ER membrane. The yeast 3-hydroxyacyl-

418

CoA dehydratase (Phs1) was later determined to be a protein that transverses the ER membrane six times,

419

and the critical residues for its enzymatic activity were found to be located within its transmembrane

420

domains, which has not been reported for any other hydratase (Kihara et al., 2008). 17β-HSD12 is

421

predicted to possess up to two transmembrane domains, one close to the N- and one upstream of the C-

422

terminus. Fusion of its C-terminus to roGFP2 showed that this part of the protein is cytosolic, whereas

AC C

EP

TE D

M AN U

SC

RI PT

397

17

ACCEPTED MANUSCRIPT

addition of roGFP2 upstream of its substrate and co-factor binding site, suggested that this part is luminal

424

(Tsachaki et al., 2015). The above results suggest a complicated mechanism for long-chain fatty acid

425

elongation, with enzymatic catalysis taking place on two different sides of the ER membrane, as well as

426

within the membrane.

427

The membrane topology of 17β-HSD9 (Su et al., 1999) has been elaborately investigated using the

428

proteinase K protection assay and deletion constructs (Simon et al., 1999). It was demonstrated that 17β-

429

HSD9 contains a transmembrane domain starting at the beginning of the protein, followed by the main

430

luminal part and a C-terminal transmembrane domain that leaves six amino acids in the cytosolic

431

compartment. Although the native protein does not seem to be glycosylated, introduction of an N-

432

glycosylation site at the proposed luminal region induced glycosylation, further supporting the luminal

433

localization of the main part of the enzyme. A similar topology was demonstrated for 17β-HSD6 using

434

roGFP2 fusions at the C-terminus, revealing cytoplasmic orientation of the very C-terminus and the part

435

following the first transmembrane region, which indicates luminal orientation of the main part of the

436

enzyme (Tsachaki et al., 2015).

437

The two enzymes also share functional similarity. Both enzymes are involved in the metabolism of

438

retinoids, hence their alternative names retinol dehydrogenase (RODH) for 17β-HSD6 and 11-cis retinol

439

dehydrogenase for 17β-HSD9. Their substrate preference differs though, with 17β-HSD6 accepting

440

mostly all-trans retinol and 17β-HSD9 9-cis retinol or 11-cis retinol (Napoli, 2001; Wang et al., 1999).

441

The membrane topology of these enzymes indicates that at least a part of the complex retinoid metabolism

442

takes place in the lumen of the ER and implies the existence of an unknown transport mechanism of

443

retinoids from the cytosol to the ER. Besides their role in retinoid metabolism, both enzymes are able to

444

oxidize estradiol to estrone, as well as androgens at the 3α or 17β position. Both 17β-HSD6 and 17β-

445

HSD9 can catalyze the conversion of 3α-adiol to DHT, although based on their tissue distribution 17β-

446

HSD6 is considered the biological relevant enzyme for the oxidation of 3a-adiol (Bauman et al., 2006;

447

Biswas et al., 1997; Napoli, 2001; Su et al., 1999; Wang et al., 1999). These reactions are considered to be

AC C

EP

TE D

M AN U

SC

RI PT

423

18

ACCEPTED MANUSCRIPT

of paramount importance for DHT synthesis/inactivation through the alternative or ‘backdoor’ pathway,

449

bypassing testosterone production (Biason-Lauber et al., 2013; Fukami et al., 2013). This mechanism of

450

regulating local DHT levels seems to be central in the pathology of prostate cancer. Given the topology of

451

17β-HSD6 it remains unclear why DHT should be formed within the ER since unliganded AR resides in

452

the cytoplasm. Conversely, the enzymes that reduce androsterone to 3a-adiol (17β-HSD3) and DHT to 3α-

453

adiol (AKR1C2) are active in the cytosolic compartment, which may allow intracellular control of DHT

454

concentrations through subcellular compartmentalization. Further research is needed to understand the

455

combined action of several enzymes participating in the same metabolic pathway and located in different

456

compartments.

M AN U

SC

RI PT

448

457

458

3. Conclusion remarks

The subcellular localization of all 17β-HSD enzymes has now been determined. This effort required in

460

most cases confirmation by both indirect immunofluorescence and biochemical methods, such as

461

subcellular fractionation. Erroneous intracellular distribution has been initially assumed, for example in

462

case of 17β-HSD10, mainly due to misinterpretation of experimental data in the absence of suitable

463

positive and negative controls or appropriate markers for each cell compartment. For instance, it was

464

noted that cytochrome C oxidase rather than cytochrome C should be used as mitochondrial marker, since

465

upon mitochondrial damage cytochrome C is released into the cytosol, thus not representing an ideal

466

mitochondria-specific marker (Yang et al., 2014).

467

Since the 17β-HSDs that localize to the ER and/or lipid droplets are integral membrane proteins, unveiling

468

their membrane topology has been the focus of several studies. Topology determination is a laborious

469

process often requiring mutagenesis, protein tagging or the use of fusion proteins. These manipulations

470

may disturb proper topology, making the application of different methods indispensable. The most

471

commonly used method for membrane topology determination involves the proteinase K protection assay

AC C

EP

TE D

459

19

ACCEPTED MANUSCRIPT

using isolated microsomes, which reveals whether a part of the protein is protected from digestion, i.e.

473

luminal. This is a multi-step, indirect method, which can provide reliable results if appropriate controls are

474

employed. Another approach involves the selective permeabilization of the plasma membrane with

475

detergents such as digitonin or the use of pore-forming agents like streptolysin-O, and subsequent

476

immunofluorescence detection. This method requires extensive optimization of experimental conditions,

477

as well as the use of suitable luminal and cytosolic control proteins. Another recently added technique

478

includes the fusion of the protein of interest with roGFP2 and ratiometric calculation of the sensor

479

oxidation status, indicative of luminal (more oxidized) or cytosolic localization (Tsachaki et al., 2015).

480

Ideally, at least two independent methods are applied for membrane topology determination.

481

Subcellular localization and membrane topology dictates the compartment in which an enzyme is active,

482

and may provide insight into its biological function. This is of particularly interest for 17β-HSDs, as many

483

of these enzymes accept several common substrates in vitro that often do not represent genuine in vivo

484

substrates. A typical example is the interconversion of estrone and estradiol, where 17β-HSD1, 7 and 12

485

can perform the reductive and 17β-HSD2, 4, 6, 8, 9 and 10 the oxidative reaction. The relative

486

contribution of these enzymes in estrogen metabolism is far from being fully understood and may depend

487

on specific cellular conditions. However, given the cytoplasmic/nuclear localization of estrogen receptors

488

one might anticipate that mitochondrial or peroxisomal localization of a given 17β-HSD indicates a

489

different major physiological role for these enzymes. Determination of the subcellular localization

490

supports the hypothesis generation process when limited information exists regarding the substrates or

491

function of an enzyme. Therefore, resolving the subcellular localization and membrane topology of the

492

17β-HSD enzymes paved the way for understanding their biological roles and will continue to be

493

important for future functional studies.

AC C

EP

TE D

M AN U

SC

RI PT

472

494

495

Acknowledgements

496

This work was supported by the Swiss National Science Foundation No 31003A-179400. 20

ACCEPTED MANUSCRIPT

497

498

AC C

EP

TE D

M AN U

SC

RI PT

499

21

ACCEPTED MANUSCRIPT

Figure Legends

501

Figure 1. Schematic representation of the subcellular localization of the 17β-HSD enzymes. 17β-

502

HSD1, 5 and 14 are cytosolic. According to one study, 17β-HSD5 translocates into the nucleus in complex

503

with the androgen receptor. 17β-HSD11 and 17β-HSD13 have been reported to localize to both the lipid

504

droplets and the ER. 17β-HSD2, 17β-HSD3, 17β-HSD6, 17β-HSD7, 17β-HSD9 and 17β-HSD12 localize

505

at the ER. 17β-HSD8 and 10 are present in mitochondria, and 17β-HSD4 in peroxisomes.

506

Figure 2. Membrane topology of 17β-HSD enzymes of the ER and lipid droplets. For every protein

507

we illustrate the GXXXGXG motif (which is part of the co-factor binding site) and the active site, along

508

with the amino acid number at which these consensus sequences start. At the C-terminus the total amino

509

acid number is shown. 17β-HSD2, 17β-HSD3, 17β-HSD11 and 17β-HSD13 exhibit cytosolic, and 17β-

510

HSD6, 17β-HSD7, 17β-HSD9 and 17β-HSD12 luminal localization of the GXXXGXG motif and active

511

site.

M AN U

SC

RI PT

500

TE D

512

513

References

514

Abul-Husn, N.S., Cheng, X., Li, A.H., Xin, Y., Schurmann, C., Stevis, P., Liu, Y., Kozlitina, J., Stender, S., Wood, G.C., Stepanchick, A.N., Still, M.D., McCarthy, S., O'Dushlaine, C., Packer, J.S.,

516

Balasubramanian, S., Gosalia, N., Esopi, D., Kim, S.Y., Mukherjee, S., Lopez, A.E., Fuller, E.D.,

517

Penn, J., Chu, X., Luo, J.Z., Mirshahi, U.L., Carey, D.J., Still, C.D., Feldman, M.D., Small, A.,

518

Damrauer, S.M., Rader, D.J., Zambrowicz, B., Olson, W., Murphy, A.J., Borecki, I.B., Shuldiner,

520 521 522 523

AC C

519

EP

515

A.R., Reid, J.G., Overton, J.D., Yancopoulos, G.D., Hobbs, H.H., Cohen, J.C., Gottesman, O., Teslovich, T.M., Baras, A., Mirshahi, T., Gromada, J. and Dewey, F.E., 2018. A ProteinTruncating HSD17B13 Variant and Protection from Chronic Liver Disease. N Engl J Med. 378, 1096-1106.

Adam, M., Heikela, H., Sobolewski, C., Portius, D., Maki-Jouppila, J., Mehmood, A., Adhikari, P.,

524

Esposito, I., Elo, L.L., Zhang, F.P., Ruohonen, S.T., Strauss, L., Foti, M. and Poutanen, M., 2018.

525

Hydroxysteroid (17beta) dehydrogenase 13 deficiency triggers hepatic steatosis and inflammation

526

in mice. FASEB J. 32, 3434-3447. 22

ACCEPTED MANUSCRIPT

527

Adamski, J., Husen, B., Marks, F. and Jungblut, P.W., 1992. Purification and properties of oestradiol 17

528

beta-dehydrogenase extracted from cytoplasmic vesicles of porcine endometrial cells. Biochem J.

529

288 ( Pt 2), 375-81. Adamski, J., Normand, T., Leenders, F., Monte, D., Begue, A., Stehelin, D., Jungblut, P.W. and de

531

Launoit, Y., 1995. Molecular cloning of a novel widely expressed human 80 kDa 17 beta-

532

hydroxysteroid dehydrogenase IV. Biochem J. 311 ( Pt 2), 437-43.

534 535 536 537

Adeniji, A.O., Chen, M. and Penning, T.M., 2013. AKR1C3 as a target in castrate resistant prostate cancer. J Steroid Biochem Mol Biol. 137, 136-49.

Auchus, R.J., 2004. The backdoor pathway to dihydrotestosterone. Trends in endocrinology and metabolism: TEM. 15, 432-8.

SC

533

RI PT

530

Baes, M., Huyghe, S., Carmeliet, P., Declercq, P.E., Collen, D., Mannaerts, G.P. and Van Veldhoven, P.P., 2000. Inactivation of the peroxisomal multifunctional protein-2 in mice impedes the

539

degradation of not only 2-methyl-branched fatty acids and bile acid intermediates but also of very

540

long chain fatty acids. J Biol Chem. 275, 16329-36.

M AN U

538

541

Bauman, D.R., Steckelbroeck, S., Williams, M.V., Peehl, D.M. and Penning, T.M., 2006. Identification of

542

the major oxidative 3alpha-hydroxysteroid dehydrogenase in human prostate that converts 5alpha-

543

androstane-3alpha,17beta-diol to 5alpha-dihydrotestosterone: a potential therapeutic target for

544

androgen-dependent disease. Mol Endocrinol. 20, 444-58.

Bertoletti, N., Braun, F., Lepage, M., Moller, G., Adamski, J., Heine, A., Klebe, G. and Marchais-

TE D

545 546

Oberwinkler, S., 2016. New Insights into Human 17beta-Hydroxysteroid Dehydrogenase Type

547

14: First Crystal Structures in Complex with a Steroidal Ligand and with a Potent Nonsteroidal

548

Inhibitor. J Med Chem. 59, 6961-7.

Biason-Lauber, A., Miller, W.L., Pandey, A.V. and Fluck, C.E., 2013. Of marsupials and men:

EP

549 550

"Backdoor" dihydrotestosterone synthesis in male sexual differentiation. Mol Cell Endocrinol.

551

371, 124-32.

553 554 555 556

Biswas, M.G. and Russell, D.W., 1997. Expression cloning and characterization of oxidative 17beta- and

AC C

552

3alpha-hydroxysteroid dehydrogenases from rat and human prostate. J Biol Chem. 272, 15959-66.

Breitling, R., Marijanovic, Z., Perovic, D. and Adamski, J., 2001. Evolution of 17beta-HSD type 4, a multifunctional protein of beta-oxidation. Mol Cell Endocrinol. 171, 205-10.

Brereton, P., Suzuki, T., Sasano, H., Li, K., Duarte, C., Obeyesekere, V., Haeseleer, F., Palczewski, K.,

557

Smith, I., Komesaroff, P. and Krozowski, Z., 2001. Pan1b (17betaHSD11)-enzymatic activity and

558

distribution in the lung. Mol Cell Endocrinol. 171, 111-7.

559 560

Brown, K.A., Boerboom, D., Bouchard, N., Dore, M., Lussier, J.G. and Sirois, J., 2004. Human chorionic gonadotropin-dependent regulation of 17beta-hydroxysteroid dehydrogenase type 4 in 23

ACCEPTED MANUSCRIPT

561

preovulatory follicles and its potential role in follicular luteinization. Endocrinology. 145, 1906-

562

15.

564 565

Brozic, P., Lanisnik Risner, T. and Gobec, S., 2008. Inhibitors of 17beta-hydroxysteroid dehydrogenase type 1. Curr Med Chem. 15, 137-50. Buehner, M., Ford, G.C., Moras, D., Olsen, K.W. and Rossman, M.G., 1973. D-glyceraldehyde-3-

RI PT

563

566

phosphate dehydrogenase: three-dimensional structure and evolutionary significance. Proc Natl

567

Acad Sci U S A. 70, 3052-4.

568

Caira, F., Clemencet, M.C., Cherkaoui-Malki, M., Dieuaide-Noubhani, M., Pacot, C., Van Veldhoven, P.P. and Latruffe, N., 1998. Differential regulation by a peroxisome proliferator of the different

570

multifunctional proteins in guinea pig: cDNA cloning of the guinea pig D-specific multifunctional

571

protein 2. Biochem J. 330 ( Pt 3), 1361-8.

Chai, Z., Brereton, P., Suzuki, T., Sasano, H., Obeyesekere, V., Escher, G., Saffery, R., Fuller, P.,

M AN U

572

SC

569

573

Enriquez, C. and Krozowski, Z., 2003. 17 beta-hydroxysteroid dehydrogenase type XI localizes to

574

human steroidogenic cells. Endocrinology. 144, 2084-91.

575

Chen, Z., Kastaniotis, A.J., Miinalainen, I.J., Rajaram, V., Wierenga, R.K. and Hiltunen, J.K., 2009.

576

17beta-hydroxysteroid dehydrogenase type 8 and carbonyl reductase type 4 assemble as a

577

ketoacyl reductase of human mitochondrial FAS. FASEB J. 23, 3682-91.

578

Cornel, K.M., Kruitwagen, R.F., Delvoux, B., Visconti, L., Van de Vijver, K.K., Day, J.M., Van Gorp, T., Hermans, R.J., Dunselman, G.A. and Romano, A., 2012. Overexpression of 17beta-

580

hydroxysteroid dehydrogenase type 1 increases the exposure of endometrial cancer to 17beta-

581

estradiol. J Clin Endocrinol Metab. 97, E591-601.

582

TE D

579

Deutschmann, A.J., Amberger, A., Zavadil, C., Steinbeisser, H., Mayr, J.A., Feichtinger, R.G., Oerum, S., Yue, W.W. and Zschocke, J., 2014. Mutation or knock-down of 17beta-hydroxysteroid

584

dehydrogenase type 10 cause loss of MRPP1 and impaired processing of mitochondrial heavy

585

strand transcripts. Hum Mol Genet. 23, 3618-28.

EP

583

Elo, J.P., Akinola, L.A., Poutanen, M., Vihko, P., Kyllonen, A.P., Lukkarinen, O. and Vihko, R., 1996.

587

Characterization of 17beta-hydroxysteroid dehydrogenase isoenzyme expression in benign and

590

AC C

586

591

HSD17B10 mutation impairing the activities of the mitochondrial RNase P complex causes X-

592

linked intractable epilepsy and neurodevelopmental regression. RNA Biol. 13, 477-85.

588 589

593

malignant human prostate. Int J Cancer. 66, 37-41.

Falk, M.J., Gai, X., Shigematsu, M., Vilardo, E., Takase, R., McCormick, E., Christian, T., Place, E., Pierce, E.A., Consugar, M., Gamper, H.B., Rossmanith, W. and Hou, Y.M., 2016. A novel

Fujimoto, Y., Itabe, H., Sakai, J., Makita, M., Noda, J., Mori, M., Higashi, Y., Kojima, S. and Takano, T.,

594

2004. Identification of major proteins in the lipid droplet-enriched fraction isolated from the

595

human hepatocyte cell line HuH7. Biochim Biophys Acta. 1644, 47-59. 24

ACCEPTED MANUSCRIPT

596

Fukami, M., Homma, K., Hasegawa, T. and Ogata, T., 2013. Backdoor pathway for dihydrotestosterone

597

biosynthesis: implications for normal and abnormal human sex development. Dev Dyn. 242, 320-

598

9.

599

Furuta, S., Kobayashi, A., Miyazawa, S. and Hashimoto, T., 1997. Cloning and expression of cDNA for a newly identified isozyme of bovine liver 3-hydroxyacyl-CoA dehydrogenase and its import into

601

mitochondria. Biochim Biophys Acta. 1350, 317-24.

RI PT

600

Gast, M.J., Sims, H.F., Murdock, G.L., Gast, P.M. and Strauss, A.W., 1989. Isolation and sequencing of a

603

complementary deoxyribonucleic acid clone encoding human placental 17 beta-estradiol

604

dehydrogenase: identification of the putative cofactor binding site. Am J Obstet Gynecol. 161,

605

1726-31.

SC

602

Geissler, W.M., Davis, D.L., Wu, L., Bradshaw, K.D., Patel, S., Mendonca, B.B., Elliston, K.O., Wilson,

607

J.D., Russell, D.W. and Andersson, S., 1994. Male pseudohermaphroditism caused by mutations

608

of testicular 17 beta-hydroxysteroid dehydrogenase 3. Nat Genet. 7, 34-9.

609

M AN U

606

Gunnarsson, C., Olsson, B.M., Stal, O. and Southeast Sweden Breast Cancer, G., 2001. Abnormal

610

expression of 17beta-hydroxysteroid dehydrogenases in breast cancer predicts late recurrence.

611

Cancer Res. 61, 8448-51.

612

He, X.Y., Merz, G., Chu, C.H., Lin, D., Yang, Y.Z., Mehta, P., Schulz, H. and Yang, S.Y., 2001. Molecular cloning, modeling, and localization of rat type 10 17beta-hydroxysteroid

614

dehydrogenase. Mol Cell Endocrinol. 171, 89-98.

615

TE D

613

He, X.Y., Merz, G., Mehta, P., Schulz, H. and Yang, S.Y., 1999. Human brain short chain L-3hydroxyacyl coenzyme A dehydrogenase is a single-domain multifunctional enzyme.

617

Characterization of a novel 17beta-hydroxysteroid dehydrogenase. J Biol Chem. 274, 15014-9.

618

He, X.Y., Merz, G., Yang, Y.Z., Pullakart, R., Mehta, P., Schulz, H. and Yang, S.Y., 2000. Function of

EP

616

619

human brain short chain L-3-hydroxyacyl coenzyme A dehydrogenase in androgen metabolism.

620

Biochim Biophys Acta. 1484, 267-77. He, X.Y., Wegiel, J., Yang, Y.Z., Pullarkat, R., Schulz, H. and Yang, S.Y., 2005. Type 10 17beta-

625

AC C

621

626

Alzheimer's disease model. Brain Res Mol Brain Res. 99, 46-53.

622 623 624

627

hydroxysteroid dehydrogenase catalyzing the oxidation of steroid modulators of gammaaminobutyric acid type A receptors. Mol Cell Endocrinol. 229, 111-7.

He, X.Y., Wen, G.Y., Merz, G., Lin, D., Yang, Y.Z., Mehta, P., Schulz, H. and Yang, S.Y., 2002. Abundant type 10 17 beta-hydroxysteroid dehydrogenase in the hippocampus of mouse

Hedeskov, C.J., Capito, K. and Thams, P., 1987. Cytosolic ratios of free [NADPH]/[NADP+] and

628

[NADH]/[NAD+] in mouse pancreatic islets, and nutrient-induced insulin secretion. Biochem J.

629

241, 161-7. 25

ACCEPTED MANUSCRIPT

630

Hilborn, E., Stal, O. and Jansson, A., 2017. Estrogen and androgen-converting enzymes 17beta-

631

hydroxysteroid dehydrogenase and their involvement in cancer: with a special focus on 17beta-

632

hydroxysteroid dehydrogenase type 1, 2, and breast cancer. Oncotarget. 8, 30552-30562.

633

Hiltunen, J.K., Autio, K.J., Schonauer, M.S., Kursu, V.A., Dieckmann, C.L. and Kastaniotis, A.J., 2010. Mitochondrial fatty acid synthesis and respiration. Biochim Biophys Acta. 1797, 1195-202.

635

Holzmann, J., Frank, P., Loffler, E., Bennett, K.L., Gerner, C. and Rossmanith, W., 2008. RNase P

RI PT

634

636

without RNA: identification and functional reconstitution of the human mitochondrial tRNA

637

processing enzyme. Cell. 135, 462-74.

639

Horiguchi, Y., Araki, M. and Motojima, K., 2008. 17beta-Hydroxysteroid dehydrogenase type 13 is a liver-specific lipid droplet-associated protein. Biochem Biophys Res Commun. 370, 235-8.

SC

638

Horiguchi, Y., Araki, M. and Motojima, K., 2008. Identification and characterization of the ER/lipid

641

droplet-targeting sequence in 17beta-hydroxysteroid dehydrogenase type 11. Arch Biochem

642

Biophys. 479, 121-30.

643 644 645

M AN U

640

Hu, G.X., Lian, Q.Q., Lin, H., Latif, S.A., Morris, D.J., Hardy, M.P. and Ge, R.S., 2008. Rapid mechanisms of glucocorticoid signaling in the Leydig cell. Steroids. 73, 1018-24. Jansson, A.K., Gunnarsson, C., Cohen, M., Sivik, T. and Stal, O., 2006. 17beta-hydroxysteroid

646

dehydrogenase 14 affects estradiol levels in breast cancer cells and is a prognostic marker in

647

estrogen receptor-positive breast cancer. Cancer Res. 66, 11471-7.

Jokela, H., Rantakari, P., Lamminen, T., Strauss, L., Ola, R., Mutka, A.L., Gylling, H., Miettinen, T.,

649

Pakarinen, P., Sainio, K. and Poutanen, M., 2010. Hydroxysteroid (17beta) dehydrogenase 7

650

activity is essential for fetal de novo cholesterol synthesis and for neuroectodermal survival and

651

cardiovascular differentiation in early mouse embryos. Endocrinology. 151, 1884-92.

652

Karageorgi, S., McGrath, M., Lee, I.M., Buring, J., Kraft, P. and De Vivo, I., 2011. Polymorphisms in

EP

TE D

648

653

genes hydroxysteroid-dehydrogenase-17b type 2 and type 4 and endometrial cancer risk. Gynecol

654

Oncol. 121, 54-8.

656 657 658 659 660

Kemilainen, H., Adam, M., Maki-Jouppila, J., Damdimopoulou, P., Damdimopoulos, A.E., Kere, J.,

AC C

655

Hovatta, O., Laajala, T.D., Aittokallio, T., Adamski, J., Ryberg, H., Ohlsson, C., Strauss, L. and Poutanen, M., 2016. The Hydroxysteroid (17beta) Dehydrogenase Family Gene HSD17B12 Is Involved in the Prostaglandin Synthesis Pathway, the Ovarian Function, and Regulation of Fertility. Endocrinology. 157, 3719-3730.

Kemilainen, H., Huhtinen, K., Auranen, A., Carpen, O., Strauss, L. and Poutanen, M., 2018. The

661

Expression of HSD17B12 Is Associated with COX-2 Expression and Is Increased in High-Grade

662

Epithelial Ovarian Cancer. Oncology. 94, 233-242.

26

ACCEPTED MANUSCRIPT

663

Khan, N., Sharma, K.K., Andersson, S. and Auchus, R.J., 2004. Human 17beta-hydroxysteroid

664

dehydrogenases types 1, 2, and 3 catalyze bi-directional equilibrium reactions, rather than

665

unidirectional metabolism, in HEK-293 cells. Arch Biochem Biophys. 429, 50-9.

667 668

Kihara, A., 2012. Very long-chain fatty acids: elongation, physiology and related disorders. J Biochem. 152, 387-95.

RI PT

666

Kihara, A., Sakuraba, H., Ikeda, M., Denpoh, A. and Igarashi, Y., 2008. Membrane topology and essential

669

amino acid residues of Phs1, a 3-hydroxyacyl-CoA dehydratase involved in very long-chain fatty

670

acid elongation. J Biol Chem. 283, 11199-209.

671

Kissinger, C.R., Rejto, P.A., Pelletier, L.A., Thomson, J.A., Showalter, R.E., Abreo, M.A., Agree, C.S., Margosiak, S., Meng, J.J., Aust, R.M., Vanderpool, D., Li, B., Tempczyk-Russell, A. and

673

Villafranca, J.E., 2004. Crystal structure of human ABAD/HSD10 with a bound inhibitor:

674

implications for design of Alzheimer's disease therapeutics. J Mol Biol. 342, 943-52.

M AN U

675

SC

672

Krazeisen, A., Breitling, R., Imai, K., Fritz, S., Moller, G. and Adamski, J., 1999. Determination of

676

cDNA, gene structure and chromosomal localization of the novel human 17beta-hydroxysteroid

677

dehydrogenase type 7(1). FEBS Lett. 460, 373-9.

678

Laplante, Y., Rancourt, C. and Poirier, D., 2009. Relative involvement of three 17beta-hydroxysteroid

679

dehydrogenases (types 1, 7 and 12) in the formation of estradiol in various breast cancer cell lines

680

using selective inhibitors. Mol Cell Endocrinol. 301, 146-53.

Latif, S.A., Shen, M., Ge, R.S., Sottas, C.M., Hardy, M.P. and Morris, D.J., 2011. Role of 11beta-OH-

TE D

681 682

C(19) and C(21) steroids in the coupling of 11beta-HSD1 and 17beta-HSD3 in regulation of

683

testosterone biosynthesis in rat Leydig cells. Steroids. 76, 682-9.

684

Leenders, F., Tesdorpf, J.G., Markus, M., Engel, T., Seedorf, U. and Adamski, J., 1996. Porcine 80-kDa protein reveals intrinsic 17 beta-hydroxysteroid dehydrogenase, fatty acyl-CoA-

686

hydratase/dehydrogenase, and sterol transfer activities. J Biol Chem. 271, 5438-42.

688 689 690 691 692

Legeza, B., Balazs, Z., Nashev, L.G. and Odermatt, A., 2013. The microsomal enzyme 17betahydroxysteroid dehydrogenase 3 faces the cytoplasm and uses NADPH generated by glucose-6-

AC C

687

EP

685

phosphate dehydrogenase. Endocrinology. 154, 205-13.

Li, K.X., Smith, R.E. and Krozowski, Z.S., 1998. Cloning and expression of a novel tissue specific 17beta-hydroxysteroid dehydrogenase. Endocr Res. 24, 663-7.

Liu, S., Huang, C., Li, D., Ren, W., Zhang, H., Qi, M., Li, X. and Yu, L., 2007. Molecular cloning and

693

expression analysis of a new gene for short-chain dehydrogenase/reductase 9. Acta Biochim Pol.

694

54, 213-8.

695

Lukacik, P., Keller, B., Bunkoczi, G., Kavanagh, K.L., Lee, W.H., Adamski, J. and Oppermann, U., 2007.

696

Structural and biochemical characterization of human orphan DHRS10 reveals a novel cytosolic

697

enzyme with steroid dehydrogenase activity. Biochem J. 402, 419-27. 27

ACCEPTED MANUSCRIPT

698

Lundova, T., Stambergova, H., Zemanova, L., Svobodova, M., Havrankova, J., Safr, M. and Wsol, V.,

699

2016. Human dehydrogenase/reductase (SDR family) member 8 (DHRS8): a description and

700

evaluation of its biochemical properties. Mol Cell Biochem. 411, 35-42. Luu-The, V., Tremblay, P. and Labrie, F., 2006. Characterization of type 12 17beta-hydroxysteroid

702

dehydrogenase, an isoform of type 3 17beta-hydroxysteroid dehydrogenase responsible for

703

estradiol formation in women. Mol Endocrinol. 20, 437-43.

RI PT

701

Luu The, V., Labrie, C., Zhao, H.F., Couet, J., Lachance, Y., Simard, J., Leblanc, G., Cote, J., Berube, D.,

705

Gagne, R. and et al., 1989. Characterization of cDNAs for human estradiol 17 beta-dehydrogenase

706

and assignment of the gene to chromosome 17: evidence of two mRNA species with distinct 5'-

707

termini in human placenta. Mol Endocrinol. 3, 1301-9.

708

SC

704

Marijanovic, Z., Laubner, D., Moller, G., Gege, C., Husen, B., Adamski, J. and Breitling, R., 2003. Closing the gap: identification of human 3-ketosteroid reductase, the last unknown enzyme of

710

mammalian cholesterol biosynthesis. Mol Endocrinol. 17, 1715-25.

711 712 713

M AN U

709

Markus, M., Husen, B., Leenders, F., Jungblut, P.W., Hall, P.F. and Adamski, J., 1995. The organelles containing porcine 17 beta-estradiol dehydrogenase are peroxisomes. Eur J Cell Biol. 68, 263-7. Markus, M., Husen, B., Leenders, F., Seedorf, U., Jungblut, P.W., Hall, P.H. and Adamski, J., 1996.

714

Peroxisomes contain an enzyme with 17 beta-estradiol dehydrogenase, fatty acid

715

hydratase/dehydrogenase, and sterol carrier activity. Ann N Y Acad Sci. 804, 691-3. Matsuura, K., Shiraishi, H., Hara, A., Sato, K., Deyashiki, Y., Ninomiya, M. and Sakai, S., 1998.

TE D

716 717

Identification of a principal mRNA species for human 3alpha-hydroxysteroid dehydrogenase

718

isoform (AKR1C3) that exhibits high prostaglandin D2 11-ketoreductase activity. J Biochem.

719

124, 940-6.

Miettinen, M.M., Mustonen, M.V., Poutanen, M.H., Isomaa, V.V. and Vihko, R.K., 1996. Human 17 beta-

EP

720 721

hydroxysteroid dehydrogenase type 1 and type 2 isoenzymes have opposite activities in cultured

722

cells and characteristic cell- and tissue-specific expression. Biochem J. 314 ( Pt 3), 839-45.

724 725 726 727 728

Milla, P., Athenstaedt, K., Viola, F., Oliaro-Bosso, S., Kohlwein, S.D., Daum, G. and Balliano, G., 2002.

AC C

723

Yeast oxidosqualene cyclase (Erg7p) is a major component of lipid particles. J Biol Chem. 277, 2406-12.

Moeller, G. and Adamski, J., 2009. Integrated view on 17beta-hydroxysteroid dehydrogenases. Mol Cell Endocrinol. 301, 7-19.

Moghrabi, N., Head, J.R. and Andersson, S., 1997. Cell type-specific expression of 17 beta-

729

hydroxysteroid dehydrogenase type 2 in human placenta and fetal liver. J Clin Endocrinol Metab.

730

82, 3872-8.

731 732

Mohler, J.L., 2014. Concept and viability of androgen annihilation for advanced prostate cancer. Cancer. 120, 2628-37. 28

ACCEPTED MANUSCRIPT

733

Moller, G., Luders, J., Markus, M., Husen, B., Van Veldhoven, P.P. and Adamski, J., 1999. Peroxisome

734

targeting of porcine 17beta-hydroxysteroid dehydrogenase type IV/D-specific multifunctional

735

protein 2 is mediated by its C-terminal tripeptide AKI. J Cell Biochem. 73, 70-8.

737 738

Moon, Y.A. and Horton, J.D., 2003. Identification of two mammalian reductases involved in the twocarbon fatty acyl elongation cascade. J Biol Chem. 278, 7335-43.

RI PT

736

Motojima, K., 2004. 17beta-hydroxysteroid dehydrogenase type 11 is a major peroxisome proliferator-

739

activated receptor alpha-regulated gene in mouse intestine. Eur J Biochem. 271, 4141-6.

740

Mullner, H., Zweytick, D., Leber, R., Turnowsky, F. and Daum, G., 2004. Targeting of proteins involved in sterol biosynthesis to lipid particles of the yeast Saccharomyces cerevisiae. Biochim Biophys

742

Acta. 1663, 9-13.

743

SC

741

Nagasaki, S., Suzuki, T., Miki, Y., Akahira, J., Kitada, K., Ishida, T., Handa, H., Ohuchi, N. and Sasano, H., 2009. 17Beta-hydroxysteroid dehydrogenase type 12 in human breast carcinoma: a prognostic

745

factor via potential regulation of fatty acid synthesis. Cancer Res. 69, 1392-9.

746

M AN U

744

Napoli, J.L., 2001. 17beta-Hydroxysteroid dehydrogenase type 9 and other short-chain

747

dehydrogenases/reductases that catalyze retinoid, 17beta- and 3alpha-hydroxysteroid metabolism.

748

Mol Cell Endocrinol. 171, 103-9.

749

Oduwole, O.O., Li, Y., Isomaa, V.V., Mantyniemi, A., Pulkka, A.E., Soini, Y. and Vihko, P.T., 2004. 17beta-hydroxysteroid dehydrogenase type 1 is an independent prognostic marker in breast

751

cancer. Cancer Res. 64, 7604-9.

752

TE D

750

Oduwole, O.O., Makinen, M.J., Isomaa, V.V., Pulkka, A., Jernvall, P., Karttunen, T.J. and Vihko, P.T., 2003. 17Beta-hydroxysteroid dehydrogenase type 2: independent prognostic significance and

754

evidence of estrogen protection in female patients with colon cancer. J Steroid Biochem Mol Biol.

755

87, 133-40.

756

EP

753

Ofman, R., Ruiter, J.P., Feenstra, M., Duran, M., Poll-The, B.T., Zschocke, J., Ensenauer, R., Lehnert, W., Sass, J.O., Sperl, W. and Wanders, R.J., 2003. 2-Methyl-3-hydroxybutyryl-CoA dehydrogenase

758

deficiency is caused by mutations in the HADH2 gene. Am J Hum Genet. 72, 1300-7.

759 760 761

AC C

757

Osei, P., Suneja, S.K., Laguna, J.C., Nagi, M.N., Cook, L., Prasad, M.R. and Cinti, D.L., 1989. Topography of rat hepatic microsomal enzymatic components of the fatty acid chain elongation system. J Biol Chem. 264, 6844-9.

762

Penning, T.M., 2015. The aldo-keto reductases (AKRs): Overview. Chem Biol Interact. 234, 236-46.

763

Penning, T.M., 2010. New frontiers in androgen biosynthesis and metabolism. Current opinion in

764 765

endocrinology, diabetes, and obesity. 17, 233-9. Pletnev, V.Z. and Duax, W.L., 2005. Rational proteomics IV: modeling the primary function of the

766

mammalian 17beta-hydroxysteroid dehydrogenase type 8. J Steroid Biochem Mol Biol. 94, 327-

767

35. 29

ACCEPTED MANUSCRIPT

768 769 770

Pol, A., Gross, S.P. and Parton, R.G., 2014. Review: biogenesis of the multifunctional lipid droplet: lipids, proteins, and sites. J Cell Biol. 204, 635-46. Puranen, T.J., Kurkela, R.M., Lakkakorpi, J.T., Poutanen, M.H., Itaranta, P.V., Melis, J.P., Ghosh, D., Vihko, R.K. and Vihko, P.T., 1999. Characterization of molecular and catalytic properties of

772

intact and truncated human 17beta-hydroxysteroid dehydrogenase type 2 enzymes: intracellular

773

localization of the wild-type enzyme in the endoplasmic reticulum. Endocrinology. 140, 3334-41.

774

RI PT

771

Qin, Y.M., Haapalainen, A.M., Conry, D., Cuebas, D.A., Hiltunen, J.K. and Novikov, D.K., 1997.

775

Recombinant 2-enoyl-CoA hydratase derived from rat peroxisomal multifunctional enzyme 2:

776

role of the hydratase reaction in bile acid synthesis. Biochem J. 328 ( Pt 2), 377-82.

Rantakari, P., Lagerbohm, H., Kaimainen, M., Suomela, J.P., Strauss, L., Sainio, K., Pakarinen, P. and

SC

777

Poutanen, M., 2010. Hydroxysteroid (17{beta}) dehydrogenase 12 is essential for mouse

779

organogenesis and embryonic survival. Endocrinology. 151, 1893-901.

780

M AN U

778

Seedorf, U., Engel, T., Assmann, G., Leenders, F. and Adamski, J., 1995. Intrinsic sterol- and

781

phosphatidylcholine transfer activities of 17 beta-hydroxysteroid dehydrogenase type IV. J Steroid

782

Biochem Mol Biol. 55, 549-53.

783

Shafqat, N., Marschall, H.U., Filling, C., Nordling, E., Wu, X.Q., Bjork, L., Thyberg, J., Martensson, E., Salim, S., Jornvall, H. and Oppermann, U., 2003. Expanded substrate screenings of human and

785

Drosophila type 10 17beta-hydroxysteroid dehydrogenases (HSDs) reveal multiple specificities in

786

bile acid and steroid hormone metabolism: characterization of multifunctional

787

3alpha/7alpha/7beta/17beta/20beta/21-HSD. Biochem J. 376, 49-60.

TE D

784

Shehu, A., Albarracin, C., Devi, Y.S., Luther, K., Halperin, J., Le, J., Mao, J., Duan, R.W., Frasor, J. and

789

Gibori, G., 2011. The stimulation of HSD17B7 expression by estradiol provides a powerful feed-

790

forward mechanism for estradiol biosynthesis in breast cancer cells. Mol Endocrinol. 25, 754-66.

791

EP

788

Simon, A., Romert, A., Gustafson, A.L., McCaffery, J.M. and Eriksson, U., 1999. Intracellular localization and membrane topology of 11-cis retinol dehydrogenase in the retinal pigment

793

epithelium suggest a compartmentalized synthesis of 11-cis retinaldehyde. J Cell Sci. 112 ( Pt 4),

794 795 796 797 798 799 800 801

AC C

792

549-58.

Sivik, T., Gunnarsson, C., Fornander, T., Nordenskjold, B., Skoog, L., Stal, O. and Jansson, A., 2012. 17beta-Hydroxysteroid dehydrogenase type 14 is a predictive marker for tamoxifen response in oestrogen receptor positive breast cancer. PLoS One. 7, e40568.

Sivik, T., Vikingsson, S., Green, H. and Jansson, A., 2012. Expression patterns of 17beta-hydroxysteroid dehydrogenase 14 in human tissues. Horm Metab Res. 44, 949-56. Song, D., Liu, G., Luu-The, V., Zhao, D., Wang, L., Zhang, H., Xueling, G., Li, S., Desy, L., Labrie, F. and Pelletier, G., 2006. Expression of aromatase and 17beta-hydroxysteroid dehydrogenase types 30

ACCEPTED MANUSCRIPT

802

1, 7 and 12 in breast cancer. An immunocytochemical study. J Steroid Biochem Mol Biol. 101,

803

136-44.

804

Su, J., Lin, M. and Napoli, J.L., 1999. Complementary deoxyribonucleic acid cloning and enzymatic characterization of a novel 17beta/3alpha-hydroxysteroid/retinoid short chain

806

dehydrogenase/reductase. Endocrinology. 140, 5275-84.

807

RI PT

805

Suzuki-Yamamoto, T., Nishizawa, M., Fukui, M., Okuda-Ashitaka, E., Nakajima, T., Ito, S. and

808

Watanabe, K., 1999. cDNA cloning, expression and characterization of human prostaglandin F

809

synthase. FEBS Lett. 462, 335-40.

810

Suzuki, Y., Jiang, L.L., Souri, M., Miyazawa, S., Fukuda, S., Zhang, Z., Une, M., Shimozawa, N., Kondo, N., Orii, T. and Hashimoto, T., 1997. D-3-hydroxyacyl-CoA dehydratase/D-3-hydroxyacyl-CoA

812

dehydrogenase bifunctional protein deficiency: a newly identified peroxisomal disorder. Am J

813

Hum Genet. 61, 1153-62.

M AN U

814

SC

811

Szajnik, M., Szczepanski, M.J., Elishaev, E., Visus, C., Lenzner, D., Zabel, M., Glura, M., DeLeo, A.B.

815

and Whiteside, T.L., 2012. 17beta Hydroxysteroid dehydrogenase type 12 (HSD17B12) is a

816

marker of poor prognosis in ovarian carcinoma. Gynecol Oncol. 127, 587-94.

817

Tsachaki, M., Birk, J., Egert, A. and Odermatt, A., 2015. Determination of the topology of endoplasmic

818

reticulum membrane proteins using redox-sensitive green-fluorescence protein fusions. Biochim

819

Biophys Acta. 1853, 1672-82.

van Grunsven, E.G., van Berkel, E., Ijlst, L., Vreken, P., de Klerk, J.B., Adamski, J., Lemonde, H.,

TE D

820 821

Clayton, P.T., Cuebas, D.A. and Wanders, R.J., 1998. Peroxisomal D-hydroxyacyl-CoA

822

dehydrogenase deficiency: resolution of the enzyme defect and its molecular basis in bifunctional

823

protein deficiency. Proc Natl Acad Sci U S A. 95, 2128-33. van Grunsven, E.G., van Berkel, E., Mooijer, P.A., Watkins, P.A., Moser, H.W., Suzuki, Y., Jiang, L.L.,

EP

824

Hashimoto, T., Hoefler, G., Adamski, J. and Wanders, R.J., 1999. Peroxisomal bifunctional

826

protein deficiency revisited: resolution of its true enzymatic and molecular basis. Am J Hum

827

Genet. 64, 99-107.

828 829 830 831 832

AC C

825

Venkatesan, R., Sah-Teli, S.K., Awoniyi, L.O., Jiang, G., Prus, P., Kastaniotis, A.J., Hiltunen, J.K., Wierenga, R.K. and Chen, Z., 2014. Insights into mitochondrial fatty acid synthesis from the structure of heterotetrameric 3-ketoacyl-ACP reductase/3R-hydroxyacyl-CoA dehydrogenase. Nat Commun. 5, 4805.

Vilardo, E., Nachbagauer, C., Buzet, A., Taschner, A., Holzmann, J. and Rossmanith, W., 2012. A

833

subcomplex of human mitochondrial RNase P is a bifunctional methyltransferase--extensive

834

moonlighting in mitochondrial tRNA biogenesis. Nucleic Acids Res. 40, 11583-93.

835

Vilardo, E. and Rossmanith, W., 2015. Molecular insights into HSD10 disease: impact of SDR5C1

836

mutations on the human mitochondrial RNase P complex. Nucleic Acids Res. 43, 6649. 31

ACCEPTED MANUSCRIPT

837

Wang, J., Chai, X., Eriksson, U. and Napoli, J.L., 1999. Activity of human 11-cis-retinol dehydrogenase

838

(Rdh5) with steroids and retinoids and expression of its mRNA in extra-ocular human tissue.

839

Biochem J. 338 ( Pt 1), 23-7.

840

Wang, X.Q., Aka, J.A., Li, T., Xu, D., Doillon, C.J. and Lin, S.X., 2017. Inhibition of 17betahydroxysteroid dehydrogenase type 7 modulates breast cancer protein profile and enhances

842

apoptosis by down-regulating GRP78. J Steroid Biochem Mol Biol. 172, 188-197.

RI PT

841

Wu, L., Einstein, M., Geissler, W.M., Chan, H.K., Elliston, K.O. and Andersson, S., 1993. Expression

844

cloning and characterization of human 17 beta-hydroxysteroid dehydrogenase type 2, a

845

microsomal enzyme possessing 20 alpha-hydroxysteroid dehydrogenase activity. J Biol Chem.

846

268, 12964-9.

847

SC

843

Yan, S.D., Fu, J., Soto, C., Chen, X., Zhu, H., Al-Mohanna, F., Collison, K., Zhu, A., Stern, E., Saido, T., Tohyama, M., Ogawa, S., Roher, A. and Stern, D., 1997. An intracellular protein that binds

849

amyloid-beta peptide and mediates neurotoxicity in Alzheimer's disease. Nature. 389, 689-95.

850

M AN U

848

Yan, S.D., Shi, Y., Zhu, A., Fu, J., Zhu, H., Zhu, Y., Gibson, L., Stern, E., Collison, K., Al-Mohanna, F.,

851

Ogawa, S., Roher, A., Clarke, S.G. and Stern, D.M., 1999. Role of ERAB/L-3-hydroxyacyl-

852

coenzyme A dehydrogenase type II activity in Abeta-induced cytotoxicity. J Biol Chem. 274,

853

2145-56.

854

Yang, S.Y., He, X.Y., Isaacs, C., Dobkin, C., Miller, D. and Philipp, M., 2014. Roles of 17betahydroxysteroid dehydrogenase type 10 in neurodegenerative disorders. J Steroid Biochem Mol

856

Biol. 143, 460-72.

859 860 861 862 863

and disease. Mol Cell Endocrinol. 343, 1-6.

Yang, S.Y., He, X.Y. and Schulz, H., 2005. Multiple functions of type 10 17beta-hydroxysteroid

EP

858

Yang, S.Y., He, X.Y. and Miller, D., 2011. Hydroxysteroid (17beta) dehydrogenase X in human health

dehydrogenase. Trends Endocrinol Metab. 16, 167-75. Yepuru, M., Wu, Z., Kulkarni, A., Yin, F., Barrett, C.M., Kim, J., Steiner, M.S., Miller, D.D., Dalton, J.T. and Narayanan, R., 2013. Steroidogenic enzyme AKR1C3 is a novel androgen receptor-selective

AC C

857

TE D

855

coactivator that promotes prostate cancer growth. Clin Cancer Res. 19, 5613-25.

864

Yokoi, Y., Horiguchi, Y., Araki, M. and Motojima, K., 2007. Regulated expression by PPARalpha and

865

unique localization of 17beta-hydroxysteroid dehydrogenase type 11 protein in mouse intestine

866 867

and liver. FEBS J. 274, 4837-47.

Zhang, C.Y., Chen, J., Yin, D.C. and Lin, S.X., 2012. The contribution of 17beta-hydroxysteroid

868

dehydrogenase type 1 to the estradiol-estrone ratio in estrogen-sensitive breast cancer cells. PLoS

869

One. 7, e29835.

870 32

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Highlights •

The 17β-hydroxysteroid dehydrogenases (17β-HSDs) consists of enzymes with diverse functions, spanning from sex steroid and retinoid metabolism to fatty acid synthesis/oxidation. Investigations of subcellular localization have shown that different 17β-HSDs are present in the

RI PT



cytosol, endoplasmic reticulum, lipid droplets, peroxisomes or mitochondria. The 17β-HSDs that reside in the endoplasmic reticulum and lipid droplets are integral membrane proteins and their

Subcellular localization and membrane topology of the 17β-HSD enzymes is tightly correlated

EP

TE D

M AN U

with their biological function.

AC C



SC

topology has now been resolved.