Evidence for MicroRNA-Mediated Regulation of Steroidogenesis by

School of Environmental and Life Sciences, The University of Newcastle, Callaghan, New South Wales 2308, Australia. ‡ Department of Biology and Chem...
0 downloads 0 Views 2MB Size
Subscriber access provided by Mount Allison University | Libraries and Archives

Article

Evidence for microRNA-mediated regulation of steroidogenesis by hypoxia Man Kit Richard YU, Gayathri Chaturvedi, Steve Kwan Hok Tong, Suraia Nusrin, John Paul Giesy, Rudolf S.S. Wu, and Richard Kong Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/es504676s • Publication Date (Web): 12 Dec 2014 Downloaded from http://pubs.acs.org on December 16, 2014

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Environmental Science & Technology is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 33

1

Environmental Science & Technology

ORIGINAL RESEARCH ARTICLE

2 3

Evidence for microRNA-mediated regulation of steroidogenesis by hypoxia

4

Richard Man Kit Yu1, Gayathri Chaturvedi2, Steve Kwan Hok Tong2, Suraia Nusrin2,

5

John Paul Giesy2,3,4, Rudolf Shiu Sun Wu4,5, Richard Yuen Chong Kong*,2,4

6 7 1

8

School of Environmental and Life Sciences, The University of Newcastle, Callaghan, New

9

South Wales, Australia; 2

10

Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue,

11

Kowloon, Hong Kong Special Administrative Region, China; 3

12 13 14

Saskatchewan, Saskatoon, Saskatchewan, Canada; 4

State Key Laboratory in Marine Pollution, City University of Hong Kong, Tat Chee Avenue,

15 16

Department of Veterinary Biomedical Sciences and Toxicology Centre, University of

Kowloon, Hong Kong Special Administrative Region, China; 5

School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong

17

Special Administrative Region, China.

18 19

*To whom correspondence should be addressed: Richard Y. C. Kong, Department of Biology

20

and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong

21

Special Administrative Region, China. Tel.: 852-3442-7794; Fax: 852-3442-0522; E-mail:

22

[email protected] .

23 24 25 1

ACS Paragon Plus Environment

Environmental Science & Technology

26

ABSTRACT

27

Environmental hypoxia can occur in both natural and occupational environments. Over the

28

recent years, the ability of hypoxia to cause endocrine disruption via perturbations in steroid

29

synthesis (steroidogenesis) has become increasingly clear. To further understand the

30

molecular mechanism underlying hypoxia-induced endocrine disruption, the steroid-

31

producing human cell line H295R was used to identify microRNAs (miRNAs) affecting

32

steroidogenic gene expression under hypoxia. Hypoxic treatment of H295R cells resulted in

33

the downregulation of seven steroidogenic genes and one of these, CYP19A1 (aromatase),

34

was shown to be regulated by the transcription factor hypoxia-inducible factor-1 (HIF-1).

35

Using bioinformatic and luciferase reporter analyses, miR-98 was identified to be a

36

CYP19A1-targeting miRNA from a subset of HIF-1-inducible miRNAs. Gain- and loss-of-

37

function analysis suggested that under hypoxia, the increased expression of miR-98 led to the

38

downregulation of CYP19A1 mRNA and protein expression and that it may have contributed

39

to a reduction in estradiol (E2) production. Intriguingly, luciferase reporter assays using

40

deletion constructs of a proximal 5′-flanking region of miR-98 did not reveal a hypoxia-

41

responsive element (HRE)-containing promoter. Overall, this study provided evidence for the

42

role of miRNAs in regulating steroidogenesis and novel insights into the molecular

43

mechanisms of hypoxia-induced endocrine disruption.

44 45 46 47 48 49 50 2

ACS Paragon Plus Environment

Page 2 of 33

Page 3 of 33

51

Environmental Science & Technology

ABSTRACT ART

52 53 54 55 56 57 58 59 60 61 62 63

3

ACS Paragon Plus Environment

Environmental Science & Technology

64

INTRODUCTION

65

Environmental hypoxia is a state of reduced oxygen availability. Typically, it occurs in

66

eutrophic waters (aquatic hypoxia), at high altitudes (hypobaric hypoxia) or in low-oxygen

67

workplace environments, such as fire-protected rooms (normobaric hypoxia). Modulation of

68

steroid synthesis (steroidogenesis) by hypoxia has been widely observed in various tissues,

69

such as the gonads,1‒6 placenta7‒9 and adrenal glands.10,11 Because the steroidogenesis

70

pathway is catalyzed by a number of oxygen-sensitive cytochrome P450 enzymes (CYPs),

71

hypoxia is often thought to hinder steroidogenesis through enzymatic inhibition. However,

72

recent evidence has suggested that it can also modulate the expression of steroidogenic genes,

73

leading to either stimulation or inhibition of steroid production.3‒9,12‒14

74

From invertebrates to mammals, the transcriptional response to hypoxia is primarily

75

mediated by the transcription factor hypoxia-inducible factor-1 (HIF-1). HIF-1 is a

76

heterodimeric protein that consists of an oxygen-sensitive HIF-1α subunit and a constitutively

77

expressed HIF-1β subunit.15 Under hypoxic conditions, HIF-1α accumulates and dimerizes

78

with HIF-1β to activate expression of HIF-1 target genes through binding to their hypoxia-

79

responsive elements (HREs). To date, there are over 100 genes that are known to be regulated

80

by HIF-1, including those involved in oxygen delivery (erythropoietin, EPO), angiogenesis

81

(vascular endothelial growth factor, VEGF) and glucose uptake (glucose transporter 1,

82

GLUT-1).16 The involvement of HIF-1 in the regulation of steroidogenic gene expression has

83

been previously suggested by a number of studies. For instance, exposure to cobalt chloride

84

(CoCl2, a chemical inducer of HIF-1) decreases CYP11A1 mRNA expression and hence

85

progesterone production in testicular Leydig cells.17 Other studies have demonstrated that

86

HIF-1 can bind to and activate the gene promoters of 3β-HSD1 and CYP19A1 in Leydig

87

cells3 and breast adenocarcinoma cells,14 respectively. Despite these new insights, it remains

4

ACS Paragon Plus Environment

Page 4 of 33

Page 5 of 33

Environmental Science & Technology

88

to be determined whether hypoxia or HIF-1 regulates steroidogenic expression via any post-

89

transcriptional mechanisms, such as microRNA (miRNA) regulation.

90

MiRNAs have emerged as a novel class of post-transcriptional regulators of gene

91

expression. They are small non-coding RNAs (20‒22 nucleotides) that bind imperfectly to

92

the 3′-untranslated regions (3′-UTRs) of their target mRNAs, causing mRNA degradation

93

and/or translational repression. It has been estimated that over one-third of human genes are

94

regulated by miRNAs.18 Previously, a genome-wide screen has implicated approximately 50

95

miRNAs in human ovarian steroidogenesis.19 Notably, most of the miRNAs have been shown

96

to inhibit, and not to stimulate, the release of steroids. This raises the possibility that under

97

hypoxia, some of these or other miRNAs may be induced by HIF-1 and be responsible for the

98

hypoxic repression of certain steroidogenic genes.

99

The main goals of this study were to examine the effects of hypoxia on the expression of a

100

subset of steroidogenic genes essential for estradiol (E2) synthesis, including HMGR, StAR,

101

CYP11A1, CYP17A1, 3β-HSD2, 17β-HSD1 and CYP19A1 (Figure 1), and to identify HIF-1-

102

mediated miRNAs that contribute to the observed differential gene expression. To

103

accomplish these goals, we employed the human adrenocortical carcinoma cell line H295R as

104

an in vitro steroidogenesis model. This cell line expresses all of the key enzymes involved in

105

adrenal and gonadal steroidogenesis, overcoming the limitation associated with the frequent

106

tissue- and developmental stage-specific expression of these genes in vivo. Its versatility is

107

exemplified by its use in the recently OECD validated H295R Steroidogenesis Assay for the

108

screening and detection of endocrine disrupting chemicals.20 Our results have indicated that

109

miR-98 is an HIF-1-regulated miRNA that attenuates CYP19A1 expression and E2

110

production in hypoxic H295R cells.

111 112 5

ACS Paragon Plus Environment

Environmental Science & Technology

113

MATERIALS AND METHODS

114

Cell Culture. H295R cells were obtained from the ATCC and cultured as described

115

previously.21 Exposure of H295R cells (1.2 × 106) to normoxia (20% O2) or hypoxia (1% O2)

116

was carried out at 37°C for 24 h. The normoxic or hypoxic conditions were created in a

117

CO2 incubator by mixing 20% O2, 75% N2 and 5% CO2 or 1% O2, 94% N2 and 5% CO2,

118

respectively, using a gas mixer (WITT). The O2 levels were monitored using a

119

GasAlertMax XT II gas detector (BW Technologies) throughout the experiment.

120

qRT-PCR. qRT-PCR for quantifying expression of steroidogenic genes was performed as

121

described previously.21 miR-98 expression was quantified using a TaqMan MiRNA Reverse

122

Transcription Kit and a TaqMan Gene Expression Assay (both from Applied Biosystems).

123

U6 snRNA was used as a normalization control. The relative expression levels of

124

steroidogenic genes and miR-98 were analyzed using the comparative threshold cycle (CT)

125

method.22

126

Western Blot Analysis—Western blot analysis was performed as previously described23

127

using anti-HIF1α (BD Biosciences, #610959), anti-CYP19A1 (Santa Cruz, #sc-30086) and

128

anti-β-actin (Sigma, #A2228) antibodies.

129 130

Quantification of Hormones. Extraction and quantification of E2 and testosterone (T) in the cell culture medium were performed as described by Hecker et al.24

131

Overexpression and Knockdown of miR-98. Overexpression and knockdown of miR-98

132

were conducted using precursor (pre-miR-98) and inhibitor (anti-miR-98) molecules

133

(Ambion), respectively. These molecules were transfected into H295R cells using a Neon

134

electroporation system (Invitrogen; 3 × 1600 V, 10 ms) at final concentrations of 100 and 150

135

nM for the pre-miR-98 and anti-miR-98, respectively. After 24 h, fresh medium was added to

136

the electroporated cells, and they were incubated under hypoxic conditions for an additional

137

24 h. The cells were then harvested for qRT-PCR and Western blot analyses. 6

ACS Paragon Plus Environment

Page 6 of 33

Page 7 of 33

Environmental Science & Technology

138

Preparation of Lentiviral Constructs for Overexpression and Knockdown of HIF-1α.

139

Under normoxia, the HIF-1α protein is rapidly degraded via the ubiquitin-proteasome

140

pathway that is initiated by the hydroxylation of two conserved proline residues (P402 and P564)

141

present in the oxygen-dependent degradation (ODD) domain of HIF-1α.25,26 To express the

142

normoxia-resistant HIF-1α protein, we generated an HIF-1α mutant construct (HIF1α:∆ODD),

143

in which P402 and P564 were substituted with alanine residues using a GeneArt Site-Directed

144

Mutagenesis System (Invitrogen). The mutated HIF-1α ORF was then cloned into a

145

pENTR/D-TOPO entry vector (Invitrogen) and subsequently transferred to a pLenti6/V5-

146

DEST lentiviral destination vector (Invitrogen) to produce an expression construct, pLenti-

147

HIF1α. To accomplish the siRNA-mediated knockdown of HIF-1α, two complementary

148

ssDNA oligonucleotides targeting HIF-1α were designed using BLOCK-iT RNAi Express

149

software (Invitrogen). The ssDNA oligonucleotides were annealed to generate dsDNA

150

oligonucleotides (HIF-1αi 956), which were then cloned into a pcDNA 6.2-GW/EmGFP

151

vector (Invitrogen). Subsequently, an entry clone was generated via a pDONR221 donor

152

vector (Invitrogen). The target sequence was finally transferred to a pLenti6/V5-DEST vector

153

(Invitrogen) to generate an expression construct, pLenti-HIF1αi956, which was used for HIF-

154

1α knockdown.

155

Lentivirus Production and Transduction. Lentiviruses for overexpression and

156

knockdown of HIF-1α were produced using a ViraPower Lentiviral Gateway Expression Kit

157

(Invitrogen). Briefly, 3 x 106 293FT cells were seeded 1 day before transfection. A total of 3

158

μg of lentiviral constructs and 9 μg of packing mix were cotransfected into the 293FT cells

159

using Lipofectamine 2000 (Invitrogen). The transfected cells were then incubated at 37°C for

160

48 h. Next, the cells were incubated with Lentivirus Concentrator (Clontech) at 4°C for 4 h to

161

concentrate the viruses. Viral particles were recovered by centrifuging the medium at 3,000 

162

g for 5 min at 4°C, and the pellet was resuspended in ice-cold DMEM/F12 complete medium 7

ACS Paragon Plus Environment

Environmental Science & Technology

Page 8 of 33

163

and stored at ‒80°C until use. H295R cells (1 × 106) were seeded 1 day prior to transduction.

164

A viral suspension containing either pLenti-HIF1α, pLenti-HIF1αi956 or empty pLenti6/V5-

165

DEST (control) was applied to the H295R cells in the presence of polybrene (6 μg/ml;

166

Sigma). The transduced cells were incubated at 37°C for 24 h. After incubation, the cells

167

were fed with fresh medium and kept under normoxia or hypoxia at 37°C for another 24 h

168

before harvesting.

169

Preparation of Luciferase Reporter Constructs. To assess the interaction between miR-

170

98 and CYP19A1 mRNA, a 174-bp fragment of the CYP19A1 3′-UTR was amplified by PCR

171

using

172

GCAAAGCTTCATGACCCCAAAGCCAAG-3′

173

CAGACTAGTGCGAATTCCAAGGTGTGC-3′ (reverse), which contained HindIII and SpeI

174

sites (underlined), respectively. The PCR product was double-digested with HindIII and SpeI

175

and cloned into the HindIII-SpeI site of the pMIR-REPORT luciferase vector (Ambion)

176

immediately downstream of the CMV-driven firefly luciferase gene. A construct possessing

177

mutations disrupting the putative miR-98 binding sites in the 3′-UTR of CYP19A1 was

178

prepared using a GeneArt Site-Directed Mutagenesis System (Invitrogen) with the following

179

primers: 5-AAGTATTTTTTAATCCTAATTCAAAATTTAACAGTTAC-3′ (forward) and

180

5-GTAACTGTTAAATTTTGAATTAGGATTAAAAAATACTT-3′ (reverse). Both the

181

wild-type and mutated plasmid clones (designated as pMIR-19-WT and pMIR-19-MUT,

182

respectively) were verified by DNA sequencing.

genomic

DNA

from

H295R

cells

and

the

following

(forward)

primers: and

5′5′-

183

To assess the transactivation potential of HIF-1 on the miR-98 promoter, five deletion

184

constructs of the miR-98 5′-flanking region (with or without putative HREs) were generated

185

from genomic DNA by PCR using the following primers (restriction sites are underlined):

186

forward primers, 5′-ACGTTGCTAGCCGCTGGACAGAAGAATGCAA-3′ for construct

187

pGL-miR98p1 (‒2687/+17), 5′-AGGATGCTAGCTTGACTGTGTCTGCCGTAATGTG-3′ 8

ACS Paragon Plus Environment

Page 9 of 33

Environmental Science & Technology

5′-

188

for

189

TGAACCTCGAGAGTGAACCTAGCCTGTGACTGA-3′ for construct pGL-miR98p3 (‒

190

1473/+17), 5′-ATCCAGCTAGCAACAGAATCTCTCAGGTAAC-3′ for construct pGL-

191

miR98p4 (‒1055/+17), 5′-GTCTCGCTAGCTCCTAATTCTGTGGTACC-3′ for construct

192

pGL-miR98p5

193

GCATCAAGCTTGGCATGAGCAGAATCCTCTAA-3′. The PCR products were double-

194

digested with NheI (or XhoI) and HindIII and cloned into the NheI-HindIII or XhoI-HindIII

195

site of a pGL4.10 vector (Promega).

construct

pGL-miR98p2

(‒522/+17),

and

a

common

(‒2283/+17),

reverse

primer,

5′-

196

Transient Transfection and Luciferase Reporter Assay. Transfection into HeLa cells

197

was carried out using Lipofectamine 2000 (Invitrogen) in a 24-well plate (1 × 105 cells/well).

198

For the miRNA-mRNA interaction assay, cells were co-transfected with pMIR-REPORT

199

constructs with or without pre-miR-98 (100 nM) or scrambled oligos (Ambion). For the miR-

200

98 promoter assay, cells were co-transfected with pGL-miR98p1‒5 deletion constructs and

201

HIF-1α expression vector pCMV-HIF1α, which contained the full-length HIF1α:∆ODD

202

sequence inserted in pCMV-TNT (Promega). EPO-HRE-luciferase reporter plasmid (pGL-

203

Epo-HRE), which contained four copies of the HRE sequence from the human EPO promoter,

204

was used as a positive control. Cells were harvested at 24 h after transfection, and reporter

205

assays were performed using a Dual-Luciferase Reporter Assay Kit (Promega). Luciferase

206

expression was measured with a FLUOstar Optima plate reader (BMG Labtech) and

207

normalized using β-galactosidase expression as an internal control. Data are expressed as the

208

mean of the relative luciferase units (RLUs) measured in three independent experiments.

209

Statistical Analysis. Student’s t-test or ANOVA followed by Student-Newman-Keuls

210

post hoc test was used to test the null hypothesis that there was no significant difference

211

between the values in the control and treatment groups. The results are presented as the mean

212

± SD. Differences were considered to be statistically significant at a p-value of ≤0.05. 9

ACS Paragon Plus Environment

Environmental Science & Technology

213

Page 10 of 33

RESULTS

214

Hypoxia Downregulates CYP19A1 mRNA Expression via HIF-1. Exposure of H295R

215

cells to hypoxia (1% O2) significantly suppressed mRNA expression of the majority of the

216

steroidogenic genes examined with the exception of CYP11A1, which was not significantly

217

affected by hypoxia (Figure 2A). All other genes, including HMGR, StAR, CYP17A1, 3β-

218

HSD2, 17β-HSD1 and CYP19A1, showed 34-fold reductions in expression in the hypoxic

219

cells. The activation of HIF-1 in hypoxic cells was supported by an increase in the expression

220

of GLUT-1 (a HIF-1 target gene). To investigate the role of HIF-1 in regulating steroidogenic

221

gene expression, gain- and loss-of-function experiments were carried out. Overexpression of

222

HIF-1α under normoxia significantly decreased mRNA expression of StAR, CYP11A1 and

223

CYP19A1 and increased that of CYP17A1; however, all of the changes in expression were

224

present at low levels (