MCB Accepts, published online ahead of print on 13 October 2014 Mol. Cell. Biol. doi:10.1128/MCB.00794-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Mice Without MacroH2A Histone Variants

2 3 John R. Pehrson1,2, Lakshmi N. Changolkar1,3, Carl Costanzi1,4 and N. Adrian Leu1,5

4 5 6

1

7

Philadelphia, PA 19104, USA

Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania,

8 9

3

Current address: Department of Pathology and Lab Medicine, University of Pennsylvania,

10

Philadelphia PA 19104, USA

11

4

Current address: Western Maine Health, Norway ME 04268, USA

12

5

Current address: Department of Human Genetics, The University of Utah, Salt Lake City UT

13

84112, USA

14 15

2

Corresponding author: [email protected]

16 17

Running title: Mice Without MacroH2A Histone Variants

18 19

Key words: chromatin; histone variants; gene regulation; metabolism

20 21

Introduction, Results and Discussion word count (characters excluding spaces): 29,919

22

Material and Methods word count (characters excluding spaces): 6592

23

1

24

Abstract

25

MacroH2A core histone variants have a unique structure that includes a C-terminal nonhistone

26

domain. They are highly conserved in vertebrates, and are thought to regulate gene expression.

27

However the nature of genes regulated by macroH2As and their biological significance remain

28

unclear. Here we examine macroH2A function in vivo by knocking out both macroH2A1 and

29

macroH2A2 in the mouse. While macroH2As are not required for early development, the absence

30

of macroH2As impaired prenatal and postnatal growth, and can significantly reduce reproductive

31

efficiency. The distribution of macroH2A.1- and macroH2A.2-containing nucleosomes shows

32

substantial overlap, as do their effects on gene expression. Our studies in fetal and adult liver

33

indicate that macroH2As can exert large positive or negative effects on gene expression, with

34

macroH2A.1 and macroH2A.2 acting synergistically on the expression of some genes and

35

apparently having opposing effects on others. These effects are very specific, and in the adult liver

36

preferentially involve genes related to lipid metabolism including the leptin receptor. MacroH2A-

37

dependent gene regulation changes substantially in postnatal development and can be strongly

38

affected by fasting. We propose that macroH2As produce adaptive changes to gene expression,

39

which in the liver focus on metabolism.

40

2

41

Introduction

42

MacroH2A core histone variants have a unique structure in which an N-terminal H2A domain is

43

connected to a C-terminal nonhistone domain [1]. The H2A domain can substitute for conventional

44

H2A in the nucleosome, and we estimated that ~ 1 in 30 nucleosomes contains a macroH2A in

45

adult rat liver [1, 2], an organ with relatively high macroH2A content. Most of the nonhistone

46

region consists of an evolutionarily conserved domain [3] called a macrodomain. Macrodomains

47

are found in a variety of proteins and as stand-alone proteins in many bacteria [3, 4]. Some, but not

48

all macrodomains bind ADP-ribose with high affinity [5, 6].

49

MacroH2As show a complex distribution in metazoan organisms [7]. The basal metazoan

50

species Trichoplax adhaerens and the sponge Amphimedon queenslandica have a macroH2A gene,

51

as does Hydra magnipapillata, clearly indicating its presence in early metazoan evolution. While

52

some other invertebrate genomes contain a macroH2A gene, many complete or nearly complete

53

invertebrate genomes lack macroH2A; e.g., macroH2A is present in a sea urchin

54

(Strongylocentrotus purpuratus), a tick (Ixodes scapularis) and an annelid (Capitella teleta), but is

55

absent in sequenced insects, a nematode (Caenorhabditis elegans) and a tunicate (Ciona

56

intestinalis). Two highly conserved macroH2A genes, macroH2A1 and macroH2A2, are found in

57

mammalian genomes, and clearly homologous genes can be found in birds, reptiles and fish. To

58

our knowledge, macroH2A genes have not been detected in fungal, protozoan, or plant genomes.

59

These findings indicate that macroH2As first appeared in early metazoan evolution, were lost in

60

some invertebrate lineages, but were strongly retained in higher vertebrate species.

61

In mice, humans and many other vertebrates, macroH2A1 transcripts are alternately spliced

62

to produce two macroH2A.1 variants, macroH2A.1.1 and macroH2A.1.2 that differ in an ~30

63

amino acid region of the macrodomain [1, 8]. MacroH2A2 appears to encode a single protein,

64

macroH2A.2, that has the same size and general structure as the macroH2A.1 variants, but is only

3

65

about 70% identical in primary structure [9, 10]. The macrodomain of macroH2A.1.1 binds ADP-

66

ribose [6], although the functional significance of this interaction for macroH2A1.1 nucleosomes is

67

unclear. There is no identified small molecule that binds the macrodomains of macroH2A.1.2 or

68

macroH2A.2. MacroH2As are present in all organs we have examined including liver, brain, testis,

69

thymus, kidney and adrenal gland. However, the macroH2A content and composition of different

70

tissues can be significantly different [9, 11]. For example, macroH2A.1.1 appears to be present

71

mainly in non-proliferating cells of differentiated tissues [11, 12].

72

Although it is generally believed that regulation of gene expression is an important role for

73

macroH2As, there is no consensus on the nature of the genes that are targeted or the biological

74

significance of this regulation. A study of gene expression in MACROH2A1 knockdown MCF-7

75

cells indicated that macroH2A.1 impacts the expression of many genes, with 19% of 97 genes

76

examined showing significantly altered expression [14]. A role in gene silencing is suggested by

77

the preferential localization of macroH2As to domains of transcriptionally silent heterochromatin

78

including the inactive X chromosome [9, 10, 15], pericentromeric heterochromatin in some cells

79

[13, 16], the XY-body of spermatocytes [17] and transcriptionally silent senescence-associated

80

heterochromatic foci [18]. Cell-type specific macroH2A-dependent silencing of IL-8 was observed

81

in studies of cultured cell lines [19], suggesting that macroH2As could be important for

82

establishing or maintaining cell-type specific patterns of gene expression. Other studies indicate

83

that macroH2As have a critical role in early development and differentiation. A knockdown of

84

macroH2A2 in developing zebrafish embryos led to severe developmental defects [20], and some

85

studies of embryonic stem (ES) cells found that knockdown of macroH2A1 and/or macroH2A2

86

impaired the expression of differentiation genes and the inactivation of pluripotency genes during

87

in vitro differentiation [21, 22]. In contrast, our studies of macroH2A1 knockout (KO) mice

88

showed no obvious defects in development or X-inactivation [23]. Changes in liver gene

4

89

expression were seen for a few genes that functionally cluster in the area of metabolism. Most of

90

these genes showed macroH2A.1 enrichment [24], suggesting that they are direct targets of

91

macroH2A.1 nucleosomes. Our results seemed to be inconsistent with the studies that showed

92

dramatic effects on early development and stem cell differentiation. One potential contributor to

93

this apparent inconsistency is macroH2A.2, which could have overlapping functions with

94

macroH2A.1 variants and mask significant aspects of macroH2A function in macroH2A1 KO

95

mice. To address this possibility and expand our understanding of macroH2A function in vivo, we

96

knocked out macroH2A2 and generated mice that lack both macroH2A.1 and macroH2A.2 histone

97

variants.

98 99

Materials and Methods

100

Mice

101

The University of Pennsylvania Institutional Animal Care and Use Committee approved all animal

102

protocols. Mice were maintained on a 12 hr day/night cycle with the lights coming on at 8 AM. Mice

103

used for organ harvest were euthanized with CO2 between 9 and 11 AM. For DEXA body composition

104

analyses the mice were anesthetized (100mg/Kg Ketamine + 10mg/kg Xylazine) and scanned (PIXImus,

105

GE Lunar).

106 107

MacroH2A KO mice

108

MacroH2A1 KO mice inbred in either the C57Bl/6 or 129/S6 (Taconic) background for at least 10

109

generations were produced as previously described [23]. Genotyping was done with a small piece of the

110

tail that was digested with proteinase K. Primers: F1 - TCTTTGCAAGGGTCAGACG; R1 –

111

ACTGGAGACAGCGCATTACC; R2 – CAACATAACCACCATGATCTCG. Reactions used Taq

5

112

polymerase (New England Biolabs), annealing temperature 55o C (normal allele 148 bp, KO allele 238

113

bp).

114

The macroH2A2 (H2afy2) KO targeting vector was based on a ~6.8 kb BglII-KpnI fragment of

115

mouse genomic DNA cloned into the pPNT vector [25] (Fig. 1S). The Neo cassette was flanked by

116

LoxP recombination sites and inserted into an EaeI site located less than 300 bp upstream of exon 2. A

117

third LoxP site was inserted into an EaeI site located less than 150 bp downstream of exon 2. The

118

targeting vector was electroporated into V6.5 ES cells [26]. ES cells positive for homologous

119

recombination by southern blotting (Fig. 1S) were injected into blastocysts and the chimeric males were

120

mated to obtain germ line transmission. Recombinant mice were bred with an EIIa Cre mouse (Jackson

121

Laboratories) and offspring were screened by PCR to identify mice carrying KO alleles. The

122

macroH2A2 KO allele was backcrossed into the C57Bl/6 and 129/S6 backgrounds for 10 or more

123

generations. Genotyping primers: AF1 - CCCAGCCAATCCCAAGTATGAGG; CR1 -

124

CCCCAACTCACTCCATATGCAGC. Tail DNA PCR reactions used AccuPrime Taq polymerase

125

(Invitrogen), annealing temperature 55o C (normal allele 895 bp, KO allele 320 bp), (Fig. 1S).

126 127

Microarrays

128

Wild-type and macroH2A KO females were super-ovulated and mated with wild-type or macroH2A KO

129

males respectively. Wild-type and KO embryos were collected, mixed together and transferred to wild-

130

type pseudo-pregnant females. Livers were collected from 18.5 d.p.c. fetuses. In studies involving adult

131

mice, wild-type and KO mice were raised together following weaning, and were euthanized at 2 months

132

of age at ~ 10 AM. The liver was immediately dissected and frozen in liquid nitrogen. Total RNA was

133

isolated using Trizol (Invitrogen).

134

Labeled cDNA was prepared from 250 ng of total RNA using the Ambion WT Expression Kit

135

(Life Technologies). Labeled cDNA (5.5 g) was hybridized to Mouse Gene 2.0 ST GeneChips (adult

6

136

livers) or Mouse Gene 1.0 ST GeneChips (fetal livers) (Affymetrix Inc., Santa Clara CA). The

137

microarrays were stained with streptavidin-phycoerythrin. A GeneChip 3000 7G scanner was used to

138

collect the fluorescence signal.

139

Affymetrix Command Console and Expression Console were used to quantitate expression levels

140

for targeted genes; default values provided by Affymetrix were applied. Border pixels were removed,

141

and the average intensity of pixels within the 75th percentile was computed for each probe. The average

142

of the lowest 2% of probe intensities occurring in each microarray sector was subtracted as background

143

from all features in that sector. Affymetrix cel files (containing probe intensities) were imported into

144

Partek Genomics Suite (v6.6, Partek, Inc., St. Louis, MO) where RMA normalization was applied

145

yielding log2-transformed intensities for all Transcript IDs. Normalized intensities were exported from

146

Partek into R, where Significance Analysis of Microarrays (SAM, using the samr v2.0 package, [27])

147

was applied, yielding a fold change and q-value (False Discovery Rate, FDR) for each Transcript ID.

148

The normalized data and cel files were deposited in the NCBI GEO database, accession number

149

GSE57089.

150 151

Real-time PCR

152

Total RNA was prepared from mouse liver using Trizol (Invitrogen). We synthesized cDNA using

153

random hexamer primers and M-MuLV Reverse Transcriptase (RNase H–) (New England Biolabs)

154

under standard conditions. Real-time PCR was performed and quantitated using the LightCycler System

155

(Roche Applied Science) as previously described [28]. Primer sequences and annealing temperature are

156

listed in Table 5S.

157

7

158

Purification of macroH2A nucleosomes

159

MacroH2A-containing mono- and oligonucleosomes were purified from bulk chromatin by thio-affinity

160

chromatography as described [28]. Briefly, fifty mixed gender 18.5-d.p.c. fetal mouse livers were very

161

gently homogenized using a Potter Elvehjem tissue homogenizer, and nuclei were purified by

162

ultracentrifugation through a sucrose cushion; fetal liver nuclei must be handled with care to avoid

163

aggregation. An H1-stripped chromatin fraction was prepared and digested to mono- and

164

oligonucleosomes. H1 removal was done at 60 mM NaCl instead of the 50 mM previously used, which

165

improved removal. Nuclease digestions used recombinant micrococcal nuclease (New England Biolabs),

166

and the protease inhibitor cocktail cOmplete (Roche) was used throughout the procedure. The

167

solubilized nucleosomes were first passed through a column of Activated Thiol Sepharose (GE

168

Healthcare Life Sciences), which was connected in tandem to a column of Thiopropyl Sepharose (GE

169

Healthcare Life Sciences). The columns were disconnected, and washed and eluted as previously

170

described [28], except we first washed the Activated Thiol Sepharose column with 0.5 M NaCl, 1 mM

171

EDTA, 10 mM Tris pH 7.5 prior to elution with a 100 mM -mercaptoethanol, 100 mM NaCl, 1 mM

172

EDTA, 10 mM Tris pH 7.5. This elution procedure was also used for the Thiopropyl Sepharose column;

173

the 0.5 M NaCl wash removes nucleosomes bound to the column through cysteine-containing

174

nonhistone proteins. The same thio-affinity procedure was used for a control experiment done with ~50

175

double KO 18.5-d.p.c. livers.

176 177

High-throughput sequencing and mapping of sequences

178

Mononucleosomal DNA was purified from an agarose gel, and the DNA ends repaired using standard

179

procedures. An A was added to the 3’ ends using Klenow (exo-), and Illumina multiplex adaptors were

180

ligated to the fragments following standard procedures. Ligated fragments were gel purified and

181

amplified using Illumina multiplex primers. Libraries were sequenced to 50 bp using an Illumina HiSeq

8

182

2000 sequencer. Reads were aligned to the mouse genome (release mm9) using Bowtie, allowing up to 3

183

mismatches using the flags '-v 3 -k 1 -m 1 --best –strata’. The sequences were deposited in the NCBI

184

GEO database, accession number GSE57089. For the comparison of the macroH2A.1 track (Thiopropyl-

185

eluted) to the macroH2A.2 track (Activated Thiol-eluted) (Fig. 4C), the macroH2A over input (starting

186

material) ratios were computed by tabulating the expected center of fragments corresponding to each

187

read, then assigning them to sliding-window bins. The log2 ratio was computed with a pseudocount of 1

188

added to both macroH2A and input read totals. Bins that had 0 macroH2A and input reads were

189

discarded. The log2 ratios were normalized by subtracting the genome-wide median log2 ratio.

190 191

Western blots

192

Proteins were separated by SDS gel electrophoresis, transferred to polyvinylidene difluoride membranes

193

[11], and probed with rabbit polyclonal antibodies raised against the nonhistone regions of rat

194

macroH2A1.1, 1.2 and human macroH2A.2 [15]. The signal was detected by chemiluminescence. The

195

blot in Fig. 1 used a low bisacrylamide SDS polyacrylamide gel, which allows the resolution of all three

196

macroH2A variants [9]. The blots in Figs. 3, 4 and 2S used a gel with a standard

197

acrylamide:bisacrylamide ratio of 37.5:1, which does not resolve macroH2A.1.1 from macroH2A.1.2.

198 199

Results

200

Knockout of macroH2A2

201

We knocked out macroH2A2 by removing the second exon (see Methods and Fig. 1S). This exon

202

contains the initiator methionine and encodes the first 57 amino acids of the histone region of

203

macroH2A.2. As in the case of the knockout of macroH2A1, the macroH2A2 KO did not produce

204

any obvious developmental defects or reproductive problems. We kept a small group of

9

205

macroH2A2 KO and wild-type mice in a mixed C57Bl/6 x 129/Sv background for more than 1.5

206

years and did not observe any obvious pathology in the KOs in comparison to controls.

207

The macroH2A2 KO allele was backcrossed into the C57Bl/6 and 129/S6 (Taconic)

208

backgrounds for 10 or more generations, which should eliminate most background effects except

209

for regions closely linked to macroH2A2 (H2afy2). We used an F1 (C57Bl/6 x 129/Sv) ES cell

210

line, and sequence polymorphisms around macroH2A2 showed that the KO allele is from the

211

129/Sv background. Our macroH2A1 KO allele is also in the 129/Sv background. The phenotypic

212

effects of the macroH2A double KO are different in the C57Bl/6 and 129/S6 backgrounds as

213

described below.

214 215

Normal development in the absence of macroH2A

216

We were able to produce macroH2A double KO males and females, and western blotting

217

confirmed the absence of macroH2A.1 and macroH2A.2 (Fig. 1). External and internal

218

examination of late stage fetuses, newborn pups and adults did not reveal any obvious structural

219

defects associated with the absence of macroH2As. We did a histological examination of major

220

organs and tissues from two young adult double KOs and an age-matched control and did not

221

observe any obvious anomalies or pathology in the double KOs. These results show that mouse

222

development can proceed in an apparently normal fashion in the absence of macroH2A variants.

223 224

Reproductive problems of C57Bl/6 macroH2A double KOs

225

We efficiently bred macroH2A double KOs with one another in the 129/S6 background, but

226

C57Bl/6 double KOs bred very poorly with one another. In some cases the female did not appear

227

to become pregnant, while in other cases many or all of the pups died or were eaten shortly after

228

birth. Young adult C57Bl/6 double KOs typically produced offspring when bred with wild type

10

229

mice or mice expressing at least one macroH2A gene, showing that these mice are not generally

230

infertile. We believe the effects of the macroH2A double KO exacerbate reproductive and

231

mothering problems that occur with wild-type C57Bl/6 mice [29]. In addition, double KOs appear

232

to be more susceptible to perinatal death (see below).

233 234

Reduced size of macroH2A double KOs

235

Double KO pups in both the 129/S6 and C57Bl/6 backgrounds were smaller on average than wild-

236

type pups and this difference persisted into adulthood. To demonstrate these effects we mixed

237

wild-type and double KO pre-implantation embryos and implanted them into wild-type recipient

238

females. An experiment tracking the weights of double KO and wild-type C57Bl/6 males is shown

239

in Fig. 2, and similar results were observed with females. We used dual-energy X-ray

240

absorptiometry (DEXA) to analyze the size and body composition of 129/S6 adult double KO and

241

control males (Table 1). The double KOs weighed less and were 9% shorter in length. Most of the

242

weight difference was accounted for by reduced lean body mass. The bone mineral content, bone

243

mineral density and fat content of the double KOs were not significantly different from their wild-

244

type littermates.

245

We used C57Bl/6 mice to examine whether the absence of macroH2As reduced the size of

246

late stage fetuses. We bred double KO males with females that were heterozygous for macroH2A1

247

and KO for macroH2A2. Fetuses were collected at 17.5 and 18.5 days post coitum (d.p.c.). We

248

obtained an equal number of double KO fetuses and fetuses that were heterozygous for

249

macroH2A1 and KO for macroH2A2, but the double KO fetuses were smaller, Table 2. A similar

250

result was obtained with a group of fetuses that were produced by breeding double KO males with

251

females that were KO for macroH2A1 and heterozygous for macroH2A2, Table 2.

11

252

The reduced size of late stage double KO fetuses and the slower growth of double KO pups

253

and young adults show that the absence of macroH2As impairs pre- and post-natal growth. This

254

suggests a role for macroH2As in regulating metabolic, neurologic and/or hormonal controls of

255

growth. Our previous discovery that macroH2A.1 variants regulate metabolic genes in the liver is

256

consistent with this idea, although the absence of macroH2A.1 by itself did not have any obvious

257

effect on growth.

258 259

Increased perinatal death of C57Bl/6 macroH2A double KOs

260

Our breeding experiences found a deficiency in the number of C57Bl/6 double KOs that survived

261

to the age of weaning (3 weeks). Table 3 summarizes the results we obtained when we bred mice

262

with different combinations of macroH2A KO alleles. Overall, the number of macroH2A double

263

KOs produced was ~ 29% less than expected based on the genotypes of the parents. We observed

264

significant numbers of mice dying shortly after birth, and genotype analyses of 25 dead newborns

265

found a strong bias toward double KOs (84%). Interestingly, we did not observe a significant

266

deficiency of double KOs in our studies of 71 late stage fetuses (Table 2). Taken together these

267

results indicate that the perinatal period is the important time when macroH2A double KOs were

268

lost. More double KO females survived to weaning than males, although the difference was small;

269

this indicates that issues related to X-inactivation are unlikely to be a major factor in the loss of

270

double KO females.

271 272

Additional phenotypic effects in 129/S6 macroH2A double KO mice

273

Double KO 129/S6 mice frequently showed signs of bilaterally increased palpebral fissure in

274

comparison to controls. Histological analysis of the globes did not reveal any abnormalities and

275

histological analysis of the heads found no orbital mass or cyst that could cause a protrusion of the

12

276

globes. We conclude that this is a true macroblepharon due to an abnormally increased length of

277

the eyelid margin. Some double KOs developed eyelid inflammation - typically between 6 and 12

278

months of age - possibly secondary to eye irritation caused by the wider eyelid margin.

279

Some 129/S6 macroH2A double KO mice showed increased darkening of the hair on their

280

back. This effect appeared at ~ 6 weeks of age and was typically more evident in young adults than

281

older mice.

282 283

MacroH2A-dependent gene regulation in development - fetal liver

284

Our current hypothesis is that gene regulation is an important function of macroH2A histones. Our

285

previous studies of macroH2A1 KO mice identified genes that appear to be regulated by

286

macroH2A.1 in adult liver, but not in neonatal liver [23]. To increase our understanding of the

287

developmental aspect of macroH2A function, we used microarrays to examine gene expression in

288

129/S6 double KO fetal liver (18.5 d.p.c.); six double KOs were compared to six controls, with the

289

control livers coming from wild-type fetuses that developed together with the double KOs in wild-

290

type recipient females. The macroH2A.1 content of late fetal liver is lower than adult liver and

291

consists almost entirely of macroH2A.1.2 [11, 23]. The macroH2A.2 content of late fetal liver is

292

relatively low, but is higher than in adult liver (Fig. 3).

293

Remarkably few genes showed altered expression in our microarray analysis of double KO

294

fetal liver. Twenty genes showed a 1.5-fold or more increase in expression in the double KOs, but

295

only Npy (encodes neuropeptide Y) and Serpina7 (encodes thyroxine binding globulin) showed a

296

low false discovery rate. Pip5k1b (encodes a phosphatidylinositol-4-phosphate kinase) showed a 2-

297

fold decrease in expression with a low false discovery rate, but no other genes showed more than a

298

2-fold decrease in expression. The increased expression of Npy and Serpina7, and the decreased

299

expression of Pip5k1b were confirmed by a real-time PCR analysis of a larger group of fetal livers

13

300

(Table 4). We also used real-time PCR to examine the expression of some genes that showed a

301

smaller change in expression and a false discovery rate of

Mice without macroH2A histone variants.

MacroH2A core histone variants have a unique structure that includes a C-terminal nonhistone domain. They are highly conserved in vertebrates and are ...
4MB Sizes 2 Downloads 9 Views