Depletion of suppressor of cytokine signaling-1a causes hepatic ...

3 downloads 0 Views 1MB Size Report
Mar 10, 2015 - 274 hif1αl (Fig. 5A), max interactor 1(mxi-1), hypoxia inducible domain family, member 1A. 275. (higd1a), angiopoietin-like 4 (Angptl4, or pgar), ...
Articles in PresS. Am J Physiol Endocrinol Metab (March 10, 2015). doi:10.1152/ajpendo.00540.2014

1

Depletion of suppressor of cytokine signaling-1a causes hepatic

2

steatosis and insulin resistance in zebrafish

3 4 5

Ziru Dai1, 2, ¶, Hualin Wang3, ¶, Xia Jin2, Houpeng Wang3, Jiangyan He2, Mugen Liu1, Zhan

6

Yin1,2, Yonghua Sun3,*, Qiyong Lou2,*

7 8

1

9

and Technology, Center for Human Genome Research, Huazhong University of Science and

Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science

10

Technology, Wuhan, Hubei 430074, P. R. China

11

2

12

3

13

Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China

Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, State Key Laboratory of Freshwater Ecology and Biotechnology, Wuhan, Hubei, 430072, China

14 15

Running title: Lipodystrophy caused by socs1a depletion in zebrafish

16



17

*

18

0086-27-68780069), Yonghua Sun, Email: [email protected] (Tel. 0086-27-68780235, Fax:

19

0086-27-68780123).

These authors contribute equally to this work. Corresponding

authors:

Qiyong

Lou,

Email:

[email protected]

(Tel.

20 21

Key words: zebrafish, socs1a, gene targeting, hepatic steatosis, insulin resistance

22

Word count: Abstract: 245 words; Main text: 4847 words; Figure: 6

23

Copyright © 2015 by the American Physiological Society.

and

Fax:

24

Abstract

25

Suppressor of cytokine signaling-1a (SOCS1a) is a member of the suppressor of cytokine

26

signaling family, a group of related molecules that mediate the negative regulation of the

27

JAK-STAT pathway. Here, we depleted SOCS1a using the transcription activator-like (TAL)

28

effector nucleases (TALENs) technique to understand its physiological roles in zebrafish.

29

Although elevated levels of JAK-STAT5 activation and erythropoiesis have been observed in

30

socs1a-deficient zebrafish, these animals exhibited normal growth during the early stages.

31

Socs1a-deficient zebrafish began to grow slowly with certain mortalities after 20 days post

32

fertilization (dpf), while the heterozygous socs1a-deficient zebrafish exhibited enhanced

33

somatic growth. Decreased adiposity, hepatic steatosis, and insulin resistance were observed

34

in our socs1a-deficient adult zebrafish, which is similar to the lipodystrophy phenotypes

35

observed in mammals. Comparative transcriptomic analyses revealed elevated levels of

36

gluconeogenesis, lipolysis and hypoxia-inducible response and decreased activities of

37

lipogenesis and glycolysis in the hepatocytes of socs1a-deflicient adult zebrafish. Evident

38

mitochondrial dysfunction has also been observed in hepatocytes from socs1a-deficient

39

zebrafish. Taken together, our results suggest that the negative regulatory roles of SOCS1a on

40

JAK-STAT5 signaling may be involved in the suppression of the erythropoiesis and growth

41

hormone activities, which was also reflected with the fact of the enhanced somatic growth

42

performance observed in the heterozygous socs1a-deficient fish. The differences in the effects

43

caused by SOCS1a depletion on insulin sensitivity, lipid metabolism and inflammatory

44

responses between zebrafish and mammalian models observed here may reflect differences

45

between the functional mechanisms of SOCS members in terrestrial mammals and aquatic

46

teleosts.

47

48

INTRODUCTION

49

The emergency of various secreted signals and their receptors, including various cytokines

50

and hormones, enable cells in multicellular organisms to respond to distinct cues. The

51

JAK-STAT signaling pathway, which is one of the core transmission systems that mediate the

52

extracellular signals for the activation of many key signal cascades involved in various

53

physiological processes. Many cytokines or hormones that involves in many inflammatory

54

cytokine, GH, and prolactin signaling pathways have been proposed (32). The JAK-STAT

55

pathway can be negatively regulated at multiple levels. First, protein tyrosine phosphatases

56

can remove phosphate from receptors and activated STAT molecules. More recently, members

57

from a small protein family identified as suppressors of cytokine signaling (SOCS) have been

58

found to form negative feedback loops to inhibit JAK-STAT signaling (1). Based on mouse

59

genetic studies, distinguishable physiological roles for several members of the SOCS family

60

have been delineated. Among the mouse SOCS proteins, SOCS1 has been shown to be mainly

61

involved in IFNγ responses and prolactin signaling, whereas SOCS3 plays regulatory roles in

62

leukemia inhibitory factor and IL-6 signaling and placental development (15). The

63

observation of gigantism in SOCS2-deficient mice provides clear evidence that SOCS2 is a

64

negative regulator of GH signaling (29). Moreover, there is a growing body of evidence

65

suggesting a role for SOCS proteins in insulin signaling (4, 17, 24, 42).

66

Somatic growth is a tightly regulated process that is dependent on GH signaling and

67

action. Related to its role on postnatal growth promotion, GH has many other potent effects,

68

including lipid, glucose, and mineral metabolism (35). GH binds to its cognate receptor and

69

activates a number of transcription factors, including STAT5, mediated through intracellular

70

JAK2 activation (25). In mammals, the activation of JAK2-STAT5 signaling via GH leads to

71

the transcriptional activation of a wide range of target genes, including IGF-I, anti-apoptotic

72

genes, and SOCS2. Insulin is a peptide hormone produced by β-cells in the pancreas. Its main

73

function is to promote the absorption of glucose from the blood to other tissues and the

74

storage of fat for use as energy (17). Normally, GH also influences insulin signaling by

75

antagonizing the action of insulin on glucose and lipid homeostasis in diverse tissues (7). In

76

mammal animal models, chronic GH excess may promote insulin resistance by increasing

77

hepatic glucose production and triglyceride storage (10, 21).

78

The zebrafish SOCS family contains at least 12 members (http://zfin.org/). Additional

79

genome duplication also occurs in the stem lineage of teleost fishes to yield duplicate

80

members of SOCS genes in the zebrafish genome corresponding to some of the mammal

81

SOCS members (34). Zebrafish socs1a has been found to exhibit significant expression in the

82

liver (20). The expression of zebrafish socs1a may be induced by LPS and poly I:C,

83

suggesting its potential roles as an inhibitor of IFN signaling (30). However, it has also been

84

reported that socs1a may also be stimulated significantly in the livers of GH- or GHR-

85

overexpressing transgenic zebrafish, indicating its involvement in teleost GH signaling

86

pathways (13, 39).

87

Utilizing a dozen of powerful genetically modified mouse models, the physiological

88

functions of each SOCS family member have been carefully defined (32). However, there is

89

no similar genetic model for non-mammalian species. The specificity within the SOCS family

90

of proteins in an in vivo teleost model has not yet been elucidated. To address these questions,

91

we have generated zebrafish that lack the SOCS1a protein. These animals exhibited a

92

relatively healthy juvenile growth. However, the socs1a-null zebrafish begin to grow slowly

93

once they reach adulthood with certain casualties. The loss of socs1a in zebrafish was also

94

found to result in high phosphorylation levels of STAT5, high plasma levels of insulin, hepatic

95

steatosis, and decreased adipose tissue in adults. In additional, elevated levels of

96

gluconeogenesis and lipolysis in the socs1a-deficient liver have been revealed through

97

comparative transcriptomic studies of the zebrafish liver. In contrast, there are no significant

98

variations in the expression levels of the typical inflammatory cytokines, such as IFN-γ, IL-1,

99

IL-6, and TNFα, between the wild-type control and their socs1a-deficient siblings. Taken

100

together, our studies demonstrate that socs1a gene deficiency in zebrafish results in decreased

101

insulin sensitivity and hepatic steatosis with chronic excess-activated GH signaling.

102 103

MATERIALS AND METHODS

104

Zebrafish husbandry. AB-line zebrafish were maintained at a 14-h light/10-h dark

105

rhythm in circulated water at 28.5°C (22). The zebrafish were fed newly hatched brine shrimp

106

and TetraMin Tropical Fish food flakes (Tetra, Germany) three times a day. The fat

107

composition in both food supplies are 15% and 8%, respectively (8). Embryos were obtained

108

through natural spawning and cultured at 28.5°C in Ringer’s solution. Their developmental

109

stages were determined according to hours post fertilization (hpf) at 28.5°C or following the

110

morphological features previously described (22). All of the procedures for experimental

111

animal manipulation were approved by the Animal Research and Ethic Committee of the

112

Institute of Hydrobiology of the Chinese Academy of Sciences.

113

Socs1a knockout via TALENs. The construction and the sequence-specific TALEN

114

effector repeats were performed following the procedures described by Huang et al. (18). The

115

four basic single unit vectors NI, NG, NN and H that recognized ATGC were assembled using

116

NheI and SpeI. To generate capped mRNA containing DNA-binding TALEN repeats and the

117

FokI endonuclease domain, the TALEN expression vectors were linearized with NotI and

118

transcribed using a Sp6 mRNA kit (mMessage mMachine kit, Ambion USA, AM1340).

119

Capped mRNA was injected into wild-type embryos at the one- or two-cell stage.

120

Approximately 10 pooled F0 embryos were lysed for genomic DNA isolation, and the target

121

region was then amplified and digested with StuI. The primers are listed below in Table 1. The

122

remaining mutation-positive larvae were raised to adulthood and outcrossed with wild-type

123

fish. The F1 larvae were analyzed by StuI digestion and genomic DNA and cDNA sequencing.

124

The F1 transgenic larvae were raised to adulthood and self-crossed to obtain F2 homozygous

125

offspring. The fin genomic DNA was extracted for genotyping. Two independent socs1a

126

mutant lines were obtained (Fig. 1). However, since the second mutant line (mutant line 2,

127

M2) has been generated much later, so most of the assays were performed with the fish of the

128

mutant line 1 (M1) unless specifically stated.

129

Messenger RNA synthesis and microinjection. The full-length zebrafish socs1a cDNA

130

flanked with two restriction digestion sites in the primers was amplified and sub-cloned into

131

the Psp64 construct. The construct was linearized with EcoRI and transcribed using the

132

SP6mRNA kit (mMessage mMachine kit, Ambion USA, AM1340). Sos1a-deficient female

133

and socs1a-deficient male zebrafish were crossed to obtain socs1a-deficient embryos. Half of

134

the embryos were injected with socs1a mRNA in PBS solution at the one- or two-cell stage,

135

and the other half were injected with PBS saline solution as controls. Five days after injection,

136

specimens were collected for subsequent experimentation.

137

Histological analysis and Oil red-O staining. The frozen livers were cryosectioned, and

138

the sections were subjected to Hematoxylin and Eosin staining. The frozen sections were also

139

stained with Oil red O to visualize the fat deposits in the liver as previously described (2). The

140

stained sections were visualized under a light field, and images were captured at different

141

magnifications.

142

Blood sugar measurement in zebrafish. An OneTouch UltraVue (LifeScan) glucose

143

meter was used for the measurement of blood glucose, which needs 1 µl of sample for each

144

measurement. Blood collection was performed as previously described (11). The zebrafish

145

were fed a chow diet regularly, and the zebrafish were fasted as previously described (11). For

146

the glucose treatment assay, the glucose-treated zebrafish were placed in 5% glucose solution

147

for 8 h and then rinsed in Ringer’s solution for 10 min (11).

148

Triglyceride (TG) content Assay. Triglyceride assay Kit was purchased from BioAssay

149

Systems (ETGA-200, CA94545 USA). The liver samples from the adult zebrafish at 120 dpf

150

stage were solubilized in ethanoic KOH and then neutralization with MgCl2 as described

151

(31). The assay was performed following the instructions supplied by the manufacturer.

152

Optical density of the samples was measured using SPECTRAMAX M2 (Molecular Device,

153

USA). Each 5 individuals per gender type were used for each genotype. The experiment was

154

repeated once.

155

Western blot analysis. Protein extraction buffer was purchased from Thermo (Thermo

156

89990, USA), and protein extraction was performed according to the manufacturer’s

157

instructions. STAT5.1 antibody was purchased from Sigma (SAB2102320, Sigma; used at

158

1:1000 dilution). Phosphorylated Stat5.1 antibody was purchased from Millipore (05-495,

159

Millipore; used at 1:1000 dilution). Akt antibody was purchased from CST (#4691, Cell

160

Signaling Technology; used at 1:1000 dilution). Phosphorylated Akt antibody was purchased

161

from CST (#4060, Cell Signaling Technology; used at 1:1000 dilution). SDS-PAGE was

162

performed as described previously (27).

163

Total RNA extraction and gene expression levels quantified with real-time PCR analysis.

164

Total RNA was extracted using RNeasy Mini Kit (Qiagen, Germany). The quality of the total

165

RNA was verified with a microspectrophotometer (Eppendorf) and through electrophoresis.

166

Total RNA was reverse-transcribed with MMLV reverse transcriptase (Thermo).

167

Complementary DNA was diluted 1:50 prior to use. SYBR Green mix was purchased from

168

Toyobo Biotech (Japan). The primers used for real-time PCR are listed in Table 1. To analysis

169

the gene expression at larval stages or adult stages, total RNA samples were extracted from at

170

least 40 larvae each genotype, or at least 4 adults each genotype. The expression level of the

171

β-actin gene was used as the internal control for normalization. The experiments were

172

performed at least for 3 duplications.

173

Transcriptome analysis. The total RNA were extracted from the liver tissues of

174

socs1a-deficient and control zebrafish from the M1 mutant line at the 90-dpf stage. The

175

constructions of the cDNA library, and cDNA sequencing and comparison were performed by

176

BGI Co. Ltd. The expression analyses were performed as previously described (16).

177

Whole-mount in situ hybridization. The riboprobe of zebrafish gata1 was previously

178

described (33). Antisense RNA probes labeled with digoxigenin-UTP (Roche, Germany) were

179

synthesized and used for whole-mount in situ hybridization as previously described (27).

180

ELISA. The protein extraction buffer for ELISA was prepared as follows: 10 mM Tris-Cl

181

pH 7.4, 0.1 mM EDTA, 10 mM sucrose, and 0.8% NaCl. The volume of the protein extraction

182

buffer was added to 10 ml per gram of liver tissue (wet weight). The liver was homogenized

183

using an ultrasonic processor on ice and centrifuged at 2000×g for 10 min. ELISA was

184

performed following the instructions supplied by the manufacturer. ELISA kits for zebrafish

185

insulin (H203) and glucagon (H183) were purchased from Nanjing Jiancheng Bioengineering

186

Institute (Nanjing, China).

187

Statistical analysis. T tests were performed for each experiment. Each result represents

188

the mean of at least three independent experiments. The error bars represent the standard

189

deviations. The p values were calculated and are indicated in the figure legends.

190 191

RESULTS

192

Generation of socs1a-deficient zebrafish. The TALEN-based gene-targeting procedure

193

was used for the depletion of the socs1a gene zebrafish according to a previously published

194

method (18). Based on sequence information of zebrafish socs1a (NM_001003467), the

195

targeting region designed for socs1a was located at the second exon of the gene locus, and

196

both arms were 18 bp in length. The spacer length between the recognition arms was a 17-bp

197

fragment with a StuI site (Fig. 1A), which could be efficiently used for testing the mutation

198

within the spacer region. To test the efficiency of the depletion, the targeting region of the

199

socs1a locus was amplified from the genomic DNA of F0 embryos extracted at 3 dpf and then

200

subjected to digestion with StuI. Part of the amplified fragment could not be cut with StuI,

201

which indicates the existence of an indel within the targeting region after application of the

202

depletion procedure. When the F0 zebrafish reached adulthood, positive individuals were

203

identified by tail genomic DNA amplification and StuI digestion as a F0 founders. Subsequent

204

StuI digestion screening for individual juveniles revealed that the offspring of heterozygous

205

parents included fish of each of the three expected genotypes in M1 individuals (Fig. 1B). The

206

sequence analysis of the tail genomic DNA and transcripts of socs1a in homozygous mutant

207

fish confirmed that two independent mutant lines have been successfully generated as M1 and

208

M2 lines. In these mutants, the deletion of a single nucleotide, namely cytosine (C), or an

209

extra cytosine insertion in the StuI restriction site were found in M1 or M2 mutants

210

respectively (Fig. 1C), which results in premature termination of the mutant SOCS1a

211

containing 63 (M1) or 118 (M2) amino acids, of which only the first 22 amino acid residues

212

are identical to those of native zebrafish (Fig. 1D). After extraction of the total RNA from the

213

whole larvae body, the expression levels of the mutant and native forms of socs1a were

214

increased in socs1a-deficient fish than those of the wild-type control fish (Fig. 1E). This

215

might be results from the feedback compensatory regulation on socs1a transcription due to

216

the loss of the functional SOCS1a in vivo.

217

Conserved functions of zebrafish SOCS1a in Jak-Stat and GH signaling. SOCS proteins

218

have been well recognized as negative regulators of JAK-STAT signaling (1, 32). To confirm

219

the physiological effects of socs1a depletion in zebrafish in vivo, the status of the activation of

220

JAK-STAT signaling was analyzed. Elevated levels of STAT5 phosphorylation were observed

221

in socs1a-deficient zebrafish compared with wild-type control fish at 5 days post fertilization

222

(dpf, Fig. 2A). The application of AG490, an inhibitor of JAK, attenuated the excessive

223

activation of STAT5.1 phosphorylation in the mutant fish, suggesting that the superactivation

224

of STAT5.1 is mediated through JAK (Fig. 2A). It has been suggested that zebrafish SOCS1a

225

plays a regulatory role in erythropoiesis and inflammatory gene expression (30, 33).

226

Consistent with this notion, obvious elevated levels of erythropoiesis have been found in

227

socs1a-deficient fish during the early stages, as determined through an assay of gata1

228

expression at the 24-hpf stage (Fig. 2B) and the Hbbe1.1 (hemoglobin beta embryonic-1.1)

229

protein presence at the 5 dpf stage (Fig. 2C). In addition, elevated levels of erythropoiesis in

230

socs1a-deficient fish may be prevented by the application of AG490, suggesting the

231

involvement of JAK-STAT signaling in the function of zebrafish SOCS1a on erythropoiesis.

232

However, the excessive levels of erythropoiesis in socs1a-deficient fish tend to be moderate

233

during adulthood because only modestly increased numbers of red blood cells were observed

234

in the mutants compared with their wild-type control counterparts at 90 dpf stage (Fig. 2D).

235

To determine the inflammatory status in the socs1a-deficient larvae, the transcriptional

236

expression levels of the several typical inflammatory cytokines have been quantified with the

237

real-time PCR analyses. No significant elevation of the expression levels in socs1a-deficient

238

larvae compared with those of the control fish has been observed at 5 dpf stage (Fig. 2E).

239

At the hatchery, socs1a-deficient fish were undistinguishable from their normal siblings

240

(Fig. 3A), but by the 20-dpf stage, socs1a-deficient fish began to grow slowly compared with

241

their normal siblings (Fig. 3B). The socs1a-deficient fish became ill and died between 40 and

242

120 dpf (Fig. 3B). Most of the socs1a-deficient adult fish exhibited a smaller body size

243

compared with their wild-type control siblings raised in the same tank (Fig. 3D).

244

Hepatic steatosis and decreased adipose tissue in socs1a-deficient zebrafish. For the

245

purpose of healthy examination, the socs1a-deficient zebrafish and their wild-type siblings

246

were dissected. Decreased adipose tissue in the viscera was detected in the socs1a-deficient

247

zebrafish compared with the wild-type fish (Fig. 4A, C, and 4D). Because the liver is the

248

major organ involved in lipid metabolism and homeostasis (36), the pathological features of

249

the liver were examined by Oil red-O staining. Severe lipid accumulation and hepatic

250

steatosis were observed in the liver of socs1a-deficient zebrafish (Fig. 4E-H). Furthermore,

251

the TG content of the liver tissue was measured. As expected, the TG contents in both male

252

and female liver of socs1a-deficient zebrafish from both M1 and M2 were increased

253

compared with their wild control at 120 dpf stage (Table 2).

254

Comparative liver transcriptome analysis through RNA-Seq analysis. Comparative

255

transcriptomics of liver tissues from socs1a-deficient zebrafish and wild-type control fish at

256

90 dpf was performed using the RNA-Seq technique. Based on the RNA-Seq analysis results,

257

some of the transcripts encoding key molecules involved in metabolism were observed to

258

exhibit significantly differential expression (Table 3). We then selected 11 of these key genes

259

as representatives to confirm the expression patterns reflected by the RNA-Seq analysis.

260

These representative transcripts are insulin, insulin receptor b (insrb), phosphoenolpyruvate

261

carboxykinase (pck), glucose-6-phosphatase c family a1 (g6pca.1), hexokinase domain

262

containing 1 (shxk1), glucokinase (gk), O family member of forkhead transcription factors

263

(foxO1a), apolipoprotein A-IV (apoA4), low-density lipoprotein receptor (ldlr), stearoyl-CoA

264

desaturase (scd) and hypoxia induced factor, 1-alpha like (hif1αl). Our real-time RT-PCR

265

results confirmed that all of the 11 transcripts exhibited similar tendencies to those observed

266

in their expression patterns obtained through RNA-Seq analysis (Fig. 5A, Table 3). Among

267

the list of differentially expressed transcripts obtained based on the RNA-Seq analysis and our

268

real-time PCR results, Pck and g6pca1, rate-limiting enzymes of gluconeogenesis, were

269

significantly up-regulated in the liver from socs1a-deficient zebrafish compared with

270

wild-type control fish (Fig. 5A, Table 3). The expression level of a key molecule that

271

promotes the activity of lipolysis, foxO1a, was elevated in the socs1a-deficient liver (Fig. 5A).

272

In contrast, the transcription levels of shxk1, gk, and scd, which are key enzymes involved in

273

glycolysis and lipogenesis, were downregulated in the socs1a-deficient livers (Fig. 5A, Table

274

3). In addition, several genes involved in the hypoxia-induced responsive cascade, such as

275

hif1αl (Fig. 5A), max interactor 1(mxi-1), hypoxia inducible domain family, member 1A

276

(higd1a), angiopoietin-like 4 (Angptl4, or pgar), and heme oxygenase 1a (hmox1, or HO-1),

277

were also increased significantly in the socs1a-deficient liver (Table 3). In addition, the

278

transcriptional levels of many typical inflammatory cytokines, such as IFNγ, IL1b, IL6,

279

TNF-α and IL10, were either unchanged or downregulated in the socs1a-deficient liver

280

compared with the wild-type liver tissues (Table 3, or data not shown).

281

Insulin resistance in socs1a-deficient hepatocytes. The expression levels of insulin and

282

its receptor, insrb, have been found to be upregulated in the socs1a-deficient liver (Fig. 5A,

283

Table 3). The protein levels of insulin in the hepatocytes and plasma were measured. ELISA

284

of hepatic and plasma samples revealed significantly elevated levels of insulin in the

285

socs1a-deficient plasma and hepatocytes compared with those of the wild-type control fish

286

(Fig. 5B, 5C). However, the levels of glucagon in the liver and plasma were similar between

287

socs1a-deficient and wild-type control fish (Fig. 5D, 5E). However, under normal feeding,

288

starvation, and high-glucose conditions, the plasma levels of glucose in the socs1a-dificient

289

fish and wild-type control fish were similar (Fig. 5F). Furthermore, the levels of the

290

phosphorylated AKT levels decreased significantly in the hepatic tissue of socs1a-deficient

291

zebrafish compared with those of the control fish determined by the Western Blot analysis

292

(Fig. 5G), which might be associated with the insulin resistance in socs1a-deficient fish.

293

These observations suggest that the typical phenomenon of insulin resistance was observed in

294

the socs1a-deficient fish.

295

Enhanced somatic growth in the heterozygous socs1a-deficient zebrafish. Contrast to the

296

homozygous socs1a-deficient mutant zebrafish, the heterozygous socs1a mutant zebrafish

297

from both independent lines seems to be healthy. Interestingly, their somatic growth

298

performances have been enhanced compared with their wild-type control siblings (Fig. 6A,

299

6B). Moderated increased levels of the phosphorylated Stat5.1 in the heterozygous

300

socs1a-deficient zebrafish liver have been also observed through Western Blot analyses

301

(Figure 6C, D), which indicating an enhanced GH signaling activation in the heterozygous

302

socs1a-deficient zebrafish as well. However, unlike their homozygous individuals, a partially

303

depletion of the socs1a allele could promote the somatic growth which results an integrated

304

gigantism effects.

305 306

DISCUSSION

307

SOCS1-null mice develop complex fatal neonatal defects featuring fatty degeneration

308

and necrosis of the multiorgan failure as a result of inflammatory infiltration, which appear to

309

be largely due to hyperresponsiveness to IFN-γ (38). The typical hepatosteatosis phenotype in

310

adult SOCS1-null mice could only be induced by a high-fat diet (HFD) after the early death

311

of SOCS1-null mice rescued with a RAG2-deficient background (12). The neutralization of

312

IFNγ in SOCS1/IFNγ-double-null mice improved their whole-body insulin sensitivity due to

313

enhanced insulin action in the liver and greater suppression of hepatic glucose production (19).

314

The zebrafish SOCS1a has been suggested to play roles in hematopoiesis and the IFN

315

signaling pathway similar to those of its mammalian counterpart. It has been demonstrated

316

that the functions of SOCS1a could be mediated by the JAK2-STAT5 pathway (30, 33). The

317

results of this study demonstrated elevated levels of phosphorylated STAT5 and hemoglobin

318

mediated via JAK activation in socs1a-null zebrafish compared with the wild-type control

319

fish during their early and adult stages, which confirmed the roles of SOCS1a in activation of

320

the JAK-STAT5 pathway and hematopoiesis (Fig. 2B, 2C). Based on our RNA-seq analyses,

321

although a moderate upregulation of IFNγ expression was observed in socs1a-deficient fish

322

during the early stages (Fig. 2E), no significant changes of the numbers of the adult

323

erythrocytes, and the expression levels of typical inflammatory cytokines, such as ifn-γ, il-1b,

324

il-6, and tnfα, in adult hepatocytes were observed between socs1a-deficient and control

325

zebrafish (Fig. 2D; Table 3).

326

The up-regulated expression levels of socs1a have been reported in GH over-expressing

327

transgenic zebrafish previously, suggesting that SOCS1a could be also a negative modulator

328

of the somatotrophic axis in zebrafish (13, 39). GH signaling is known to stimulate

329

gluconeogenesis, lipolysis and adipose tissue mobilization in mammals and teleosts (26, 28,

330

36). A central role of the JAK2-STAT5 pathway in mediating the growth-stimulating action of

331

GH signaling has been reported in teleosts (23). The expression levels of Foxo1a, pck and

332

G6Pase, a key regulator and two enzymes controlling the gluconeogenesis, were significantly

333

increased in the liver of socs1a-deficient zebrafish compared with those of the control fish

334

(Fig. 5A). Moreover, a significant decrease in the size of adipose tissue was observed in the

335

socs1a-deficient zebrafish compared with the control fish (Fig. 4A, C and D), which is

336

opposite against the phenotype of increased adiposity in gh-deficient zebrafish (28). In

337

addition, the enhanced somatic growth performance has been observed in the heterozygous

338

socs1a-deficient fish (Fig. 6A). Taken together, all these findings revealed that socs1a

339

deficiency results in a chronic excess activation of GH signaling in zebrafish. In contrast, the

340

expression levels of pck and G6Pase either remained unchanged in SOCS1-null mice or

341

significantly decreased in SOCS1/IFNγ-double-null mice (12, 19), providing further evidence

342

regarding the differences in the physiological functions and signaling mediated by zebrafish

343

SOCS1a and mouse SOCS1.

344

Energy metabolism and somatic growth are closely coordinated in animals, and the liver

345

is a key metabolic organ that governs body energy metabolism (36). With an evident defect in

346

somatic growth in the homozygous socs1a-deficient adult zebrafish (Fig. 3), the dynamical

347

status of the liver was our focus in this study. At 90 dpf, liver steatosis was observed in

348

socs1a-deficient fish (Fig. 4E-H; Table 2), as could also be observed in SOCS1-, SOCS2-,

349

and SOCS3-deficient mice only in the presence of HFD (12, 37, 42). Moreover, significantly

350

increased plasma insulin levels were observed in socs1a-null fish, and the mutant fish did not

351

exhibit improved plasma glucose clearance compared with the control fish (Fig. 5). However,

352

the increased levels of insulin in socs1a-deficient fish did not stimulate lipogenesis and

353

glycolysis in the liver, which was reflected through comparative transcriptomics of liver

354

tissues from control and mutant fish (Fig. 5F). This finding suggests that socs1a depletion

355

impairs hepatic insulin sensitivity in socs1a-deficient zebrafish (5, 36). However, the

356

impaired insulin sensitivity could be achieved by the over-expression of SOCS1 or SOCS3 in

357

the liver transgenic mouse models (40). Mouse SOCS1 deficiency does not prevent

358

HFD-induced insulin resistance in combination with SOCS1/RAG2 deficiency (12). SOCS2

359

deletion in mice can protect against hepatic steatosis but worsens insulin resistance only with

360

HFD feeding (42). Notably, the phenotype observed in socs1a-deficient zebrafish is clearly

361

different from the SOCS1-, SOCS2-, and SOCS3- knockout mouse phenotypes (12, 37, 40,

362

42), with enhanced liver steatosis and impaired insulin sensitivity under normal feeding

363

conditions, suggesting the differences in the regulation of hepatic metabolism between these

364

two vertebrate models.

365

The liver lies at the crossroad of lipid metabolism by actively taking up lipids associated

366

with remnant particles as well as FFA and NEFA originating from adipose tissue lipolysis. As

367

a result of enhanced lipolysis and gluconeogenesis activities in the socs1a-deficient liver,

368

these activities should consume a large amount of oxygen to produce energy (14). It is

369

conceivable that lipid metabolism is related to hypoxia sensing, particularly in aquatic

370

vertebrates. Unlike SOCS1-, SOCS2-, and SOCS3-deficient mouse models, the expression

371

level of hif1al was increased significantly in the liver of the socs1a mutant fish (Fig. 5A).

372

Hif-mediated metabolic approaches appear to be important for the maintenance of local

373

oxygen homeostasis in the liver by limiting either oxygen consumption or be promoting fatty

374

acid oxidation to glycolysis and blocking mitochondrial biogenesis (14). Due to the highly

375

activated GH signaling in socs1a mutant fish, glycolysis activity is inhibited. In fact, the

376

transcriptional expression of several key enzymes involved in glycolysis, such as shxk1 and

377

gk, were decreased in the socs1a mutant liver (Fig. 5A). In addition, up-regulated levels of

378

several key hypoxia-inducible genes, such as mxi-1, higd1a, Angptl4, and hmox1, other than

379

hif1al (Fig. 5A), have been observed in the socs1a-deficient liver (Table 3). These molecules

380

have been reported to be involved in hypoxia-inducible responses, and important regulators

381

for mitochondrial biogenesis (43), physiological stress (3), LPL activity in peripheral tissues

382

(9), and cytoprotection against oxidative stress during liver ischemia (41). Thus, our results

383

above reveal that the depletion of socs1a causes a complex metabolic syndrome, including

384

locally hypoxia stress, progressive hepatic steatosis, and insulin resistance in fish. Based on

385

the observed loss of visceral and subcutaneous fat, hepatic steatosis and hepatic insulin

386

resistance, our socs1a-deficient zebrafish display a phenotype similar to that observed in

387

mammalian models with lipodystrophy (6).

388

Our data provide strong evidence that SOCS1a acts as an essential negative regulator of

389

GH signaling mediated by JAK-STAT5 in zebrafish. However, due to differences in

390

physiological and metabolic features between the terrestrial mammal model and the aquatic

391

vertebrate model, the gigantism phenotype of the SOCS2-null mouse has not been achieved in

392

SOCS1a-deficient fish, which actually progressively develop a complex metabolic syndrome.

393

Although changes in the expression patterns of typical inflammatory cytokines are not

394

observed in the socs1a mutants, several inflammation-responsive genes have been found to be

395

differentially expressed in the livers between wild-type and socs1a-deficient fish (Table 3).

396

Therefore, chronic, low-grade inflammation may not be excluded as a cause of the hepatic

397

steatosis and insulin resistance observed in the mutant fish. Zebrafish SOCS1a has been

398

suggested to function in immune response and GH signaling, which is consistent with our

399

observation that socs1a-deficient fish exhibit characteristics of both GH and hepatic steatosis

400

phenotypes. However, compared with mouse genetic models, socs1a depletion in zebrafish

401

did not recapitulate the phenotypes of SOCS1-, SOCS2-, or SOCS3-deficient mice. In this

402

aspect, our existing data also cannot rule out the compensatory roles of zebrafish SOCS1b in

403

our socs1a-deficient models. More detailed analyses of the unique metabolic features of

404

teleosts from mammal animal models and of the major targeting signaling pathway for each

405

teleost SOCS member will further clarify the metabolism principles applied in fishery and the

406

physiological roles of teleost SOCS members.

407 408

GRANTS

409

This study was supported by funds obtained from the National Basic Research Program of

410

China (973 Program, 2014CB138602) and Hi-Tech Research and Development Program of

411

China (863 Program, 2012AA022402) to ZY, Natural Science Foundation of China (No.

412

31222052) to YHS.

413 414

DISCLOSURES

415

The authors declare no conflict of interest.

416 417

REFERENCES

418

Primary Sources

419

Secondary Sources

420

Uncategorized References

421 422 423 424

1.

Ahmed SF, and Farquharson C. The effect of GH and IGF1 on linear growth and skeletal

development and their modulation by SOCS proteins. J Endocrinol 206: 249-259, 2010. 2.

Amali AA, Rekha RD, Lin CJ, Wang WL, Gong HY, Her GM, and Wu JL. Thioacetamide induced

liver damage in zebrafish embryo as a disease model for steatohepatitis. J Biomed Sci 13: 225-232,

425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468

2006. 3.

Ameri K, Rajah AM, Nguyen V, Sanders TA, Jahangiri A, DeLay M, Donne M, Choi HJ, Tormos KV,

Yeghiazarians Y, Jeffrey SS, Rinaudo PF, Rowitch DH, Aghi M, and Maltepe E. Nuclear Localization of the Mitochondrial Factor HIGD1A during Metabolic Stress. Plos One 8: 2013. 4.

Banks AS, Li J, McKeag L, Hribal ML, Kashiwada M, Accili D, and Rothman PB. Deletion of SOCS7

leads to enhanced insulin action and enlarged islets of Langerhans. J Clin Invest 115: 2462-2471, 2005. 5.

Chen G, Zhang Y, Lu D, Li NQ, and Ross AC. Retinoids synergize with insulin to induce hepatic Gck

expression. Biochem J 419: 645-653, 2009. 6.

Cortes VA, Cautivo KM, Rong SX, Garg A, Horton JD, and Agarwal AK. Leptin ameliorates insulin

resistance and hepatic steatosis in Agpat2(-/-) lipodystrophic mice independent of hepatocyte leptin receptors. J Lipid Res 55: 276-288, 2014. 7.

Davidson MB, Shen DC, Venkatesan N, and Sladen G. In vivo insulin antagonism but evanescent

in vitro tissue effect in rats with growth hormone-secreting tumors. J Endocrinol Invest 10: 569-574, 1987. 8.

De Clercq P, Arijs Y, Van Meir T, Van Stappen G, Sorgeloos P, Dewettinck K, Rey M, Grenier S,

and Febvay G. Nutritional value of brine shrimp cysts as a factitious food for Orius laevigatus (Heteroptera: Anthocoridae). Biocontrol Science and Technology 15: 467-479, 2005. 9.

Dijk W, and Kersten S. Regulation of lipoprotein lipase by AngptI4. Trends Endocrin Met 25:

146-155, 2014. 10. Dominici FP, Argentino DP, Munoz MC, Miquet JG, Sotelo AI, and Turyn D. Influence of the crosstalk between growth hormone and insulin signalling on the modulation of insulin sensitivity. Growth Horm IGF Res 15: 324-336, 2005. 11. Eames SC, Philipson LH, Prince VE, and Kinkel MD. Blood Sugar Measurement in Zebrafish Reveals Dynamics of Glucose Homeostasis. Zebrafish 7: 205-213, 2010. 12. Emanuelli B, Macotela Y, Boucher J, and Ronald Kahn C. SOCS-1 deficiency does not prevent diet-induced insulin resistance. Biochem Biophys Res Commun 377: 447-452, 2008. 13. Figueiredo MA, Mareco EA, Silva MDP, and Marins LF. Muscle-specific growth hormone receptor (GHR) overexpression induces hyperplasia but not hypertrophy in transgenic zebrafish. Transgenic Res 21: 457-469, 2012. 14. Goda N, and Kanai M. Hypoxia-inducible factors and their roles in energy metabolism. Int J Hematol 95: 457-463, 2012. 15. Greenhalgh CJ, Rico-Bautista E, Lorentzon M, Thaus AL, Morgan PO, Willson TA, Zervoudakis P, Metcalf D, Street I, Nicola NA, Nash AD, Fabri LJ, Norstedt G, Ohlsson C, Flores-Morales A, Alexander WS, and Hilton DJ. SOCS2 negatively regulates growth hormone action in vitro and in vivo. J Clin Invest 115: 397-406, 2005. 16. He W, Dai X, Chen X, He J, and Yin Z. Zebrafish pituitary gene expression before and after sexual maturation. J Endocrinol 221: 429-440, 2014. 17. Howard JK, and Flier JS. Attenuation of leptin and insulin signaling by SOCS proteins. Trends Endocrinol Metab 17: 365-371, 2006. 18. Huang P, Xiao A, Zhou M, Zhu Z, Lin S, and Zhang B. Heritable gene targeting in zebrafish using customized TALENs. Nat Biotechnol 29: 699-700, 2011. 19. Jamieson E, Chong MM, Steinberg GR, Jovanovska V, Fam BC, Bullen DV, Chen Y, Kemp BE, Proietto J, Kay TW, and Andrikopoulos S. Socs1 deficiency enhances hepatic insulin signaling. J Biol Chem 280: 31516-31521, 2005.

469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512

20. Jin HJ, Xiang LX, and Shao JZ. Identification and characterization of suppressor of cytokine signaling 1 (SOCS-1) homologues in teleost fish. Immunogenetics 59: 673-686, 2007. 21. Kim YD, Li T, Ahn SW, Kim DK, Lee JM, Hwang SL, Kim YH, Lee CH, Lee IK, Chiang JY, and Choi HS.

Orphan

nuclear

receptor

small

heterodimer

partner

negatively

regulates

growth

hormone-mediated induction of hepatic gluconeogenesis through inhibition of signal transducer and activator of transcription 5 (STAT5) transactivation. J Biol Chem 287: 37098-37108, 2012. 22. Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, and Schilling TF. Stages of embryonic development of the zebrafish. Dev Dyn 203: 253-310, 1995. 23. Kittilson JD, Jones E, and Sheridan MA. ERK, Akt, and STAT5 are Differentially Activated by the Two Growth Hormone Receptor Subtypes of a Teleost Fish (Oncorhynchus Mykiss). Front Endocrinol (Lausanne) 2: 30, 2011. 24. Krebs DL, Uren RT, Metcalf D, Rakar S, Zhang JG, Starr R, De Souza DP, Hanzinikolas K, Eyles J, Connolly LM, Simpson RJ, Nicola NA, Nicholson SE, Baca M, Hilton DJ, and Alexander WS. SOCS-6 binds to insulin receptor substrate 4, and mice lacking the SOCS-6 gene exhibit mild growth retardation. Mol Cell Biol 22: 4567-4578, 2002. 25. Lanning NJ, and Carter-Su C. Recent advances in growth hormone signaling. Rev Endocr Metab Disord 7: 225-235, 2006. 26. Leena S, Shameena B, and Oommen OV. Studies on the effect of growth hormone in vivo and in vitro on lipogenic enzymes and transaminases in a teleost Anabas testudineus (Bloch). Endocr Res 25: 341-355, 1999. 27. Lou Q, He J, Hu L, and Yin Z. Role of lbx2 in the noncanonical Wnt signaling pathway for convergence and extension movements and hypaxial myogenesis in zebrafish. Biochimica et biophysica acta 1823: 1024-1032, 2012. 28. McMenamin SK, Minchin JE, Gordon TN, Rawls JF, and Parichy DM. Dwarfism and increased adiposity in the gh1 mutant zebrafish vizzini. Endocrinology 154: 1476-1487, 2013. 29. Metcalf D, Greenhalgh CJ, Viney E, Willson TA, Starr R, Nicola NA, Hilton DJ, and Alexander WS. Gigantism in mice lacking suppressor of cytokine signalling-2. Nature 405: 1069-1073, 2000. 30. Nie L, Xiong R, Zhang YS, Zhu LY, Shao JZ, and Xiang LX. Conserved inhibitory role of teleost SOCS-1s in IFN signaling pathways. Dev Comp Immunol 43: 23-29, 2014. 31. Norris AW, Chen L, Fisher SJ, Szanto I, Ristow M, Jozsi AC, Hirshman MF, Rosen ED, Goodyear LJ, Gonzalez FJ, Spiegelman BM, and Kahn CR. Muscle-specific PPARγ-deficient mice develop increased adiposity and insulin resistance but respond to thiazolidinediones. The Journal of Clinical Investigation 112: 608-618, 2003. 32. O'Sullivan LA, Liongue C, Lewis RS, Stephenson SEM, and Ward AC. Cytokine receptor signaling through the Jak-Stat-Socs pathway in disease. Mol Immunol 44: 2497-2506, 2007. 33. O'Sullivan LA, Noor SM, Trengove MC, Lewis RS, Liongue C, Sprigg NS, Nicholson SE, and Ward AC. Suppressor of cytokine signaling 1 regulates embryonic myelopoiesis independently of its effects on T cell development. J Immunol 186: 4751-4761, 2011. 34. Opazo JC, Butts GT, Nery MF, Storz JF, and Hoffmann FG. Whole-Genome Duplication and the Functional Diversification of Teleost Fish Hemoglobins. Mol Biol Evol 30: 140-153, 2013. 35. Press M. Growth hormone and metabolism. Diabetes Metab Rev 4: 391-414, 1988. 36. Rui LY. Energy Metabolism in the Liver. Compr Physiol 4: 177-197, 2014. 37. Sachithanandan N, Fam BC, Fynch S, Dzamko N, Watt MJ, Wormald S, Honeyman J, Galic S, Proietto J, Andrikopoulos S, Hevener AL, Kay TW, and Steinberg GR. Liver-specific suppressor of

513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534

cytokine signaling-3 deletion in mice enhances hepatic insulin sensitivity and lipogenesis resulting in fatty liver and obesity. Hepatology 52: 1632-1642, 2010. 38. Starr R, Metcalf D, Elefanty AG, Brysha M, Willson TA, Nicola NA, Hilton DJ, and Alexander WS. Liver degeneration and lymphoid deficiencies in mice lacking suppressor of cytokine signaling-1. Proc Natl Acad Sci U S A 95: 14395-14399, 1998. 39. Studzinski ALM, Almeida DV, Lanes CFC, Figueiredo MD, and Marins LF. SOCS1 and SOCS3 are the main negative modulators of the somatotrophic axis in liver of homozygous GH-transgenic zebrafish (Danio rerio). Gen Comp Endocr 161: 67-72, 2009. 40. Ueki K, Kondo T, Tseng YH, and Kahn CR. Central role of suppressors of cytokine signaling proteins in hepatic steatosis, insulin resistance, and the metabolic syndrome in the mouse. Proc Natl Acad Sci U S A 101: 10422-10427, 2004. 41. Yun N, Cho HI, and Lee SM. Impaired autophagy contributes to hepatocellular damage during ischemia/reperfusion: heme oxygenase-1 as a possible regulator. Free Radic Biol Med 68: 168-177, 2014. 42. Zadjali F, Santana-Farre R, Vesterlund M, Carow B, Mirecki-Garrido M, Hernandez-Hernandez I, Flodstrom-Tullberg M, Parini P, Rottenberg M, Norstedt G, Fernandez-Perez L, and Flores-Morales A. SOCS2 deletion protects against hepatic steatosis but worsens insulin resistance in high-fat-diet-fed mice. FASEB J 26: 3282-3291, 2012. 43. Zhang H, Gao P, Fukuda R, Kumar G, Krishnamachary B, Zeller KI, Dang CV, and Semenza GL. HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity. Cancer Cell 11: 407-420, 2007.

535

Figure legends

536

Figure 1. Disruption of zebrafish socs1a gene by TALENs. A: the endogenous socs1a gene is

537

shown (top), and the exons are depicted as boxes. The coding region is shown in black. The

538

targeting arms aimed at the coding region are underlined with a red line and are shown in red

539

font, and the flanking region in the middle is shown in black font. Two independent mutant

540

lines have been obtained. One nucleotide deletion or one nucleotide insertion in the targeting

541

region has been found in mutant line 1 (M1) or line 2 (M2) respectively. B: a representative

542

genotyping for StuI digestion patterning of the PCR products amplified from the genomic

543

DNA samples from M1 socs1a-/-, socs1a+/- and socs1a+/+ individuals. C: a C deletion in the

544

targeting region of the M1 mutant socs1a site (arrow) and a C insertion in the targeting region

545

of the M2 mutant socs1a site (arrow). D: the diagram shows a truncated protein, in which the

546

first 22 amino acids (shown in green) are identical to those of the wild-type SOCS1a protein

547

and which contains 41 (M1) or 96 (M2) miscoding amino acids (shown in red) predicted from

548

the sequences of the mutant forms of the socs1a transcripts in the two independent mutant

549

lines. E: the relative levels of the wild-type and mutant socs1a transcripts from socs1a+/+ and

550

socs1a-/- larvae at 5 dpf were assayed by quantitative RT-PCR.

551 552

Figure 2. Effects of zebrafish socs1a disruption on JAK-STAT5 activation, erythropoiesis,

553

and inflammatory gene expression. A: elevated levels of phosphorylated STAT5.1 in socs1a-/-

554

fish compared with wild-type control fish at 5 dpf stage. This highly elevated level of

555

STAT5.1 phosphorylation could be attenuated with the addition of native socs1a mRNA or

556

AG490, an inhibitor of JAK kinases. B: an upregulated expression level of gata1, an early

557

erythroid marker, was found in socs1a-/- embryos (bottom) compared with wild-type embryos

558

(upper) at 24 hpf. C: elevated levels of Hbbe1 in socs1a-/- fish compared with the wild-type

559

larvae were detected through western blot at 5 dpf. The enhanced erythropoiesis could also be

560

attenuated by the addition of AG490. D: a modest increase in the numbers of erythrocytes was

561

observed in socs1a-/- adults compared with wild-type adults, as determined through the red

562

blood cell counts at 90 dpf. E: no significantly elevated expression levels of the inflammatory

563

genes ifnγ, il1b, nfkb2, nkkbiab, il6 and tnfα were observed in socs1a-/- larvae compared with

564

their control siblings at 5 dpf.

565 566

Figure 3. General phenotypic abnormality observed in socs1a-deficient zebrafish. A: normal

567

morphological features of socs1a+/+ (upper) and socs1a-/- (lower) larvae at the 3-dpf stage. B:

568

the obvious somatic growth retardation of socs1a-/- zebrafish began at the juvenile stage.

569

Asterisk (*) indicated significant difference. C: elevated mortality observed after 40 dpf. D:

570

the socs1a-/- adults (bottom) were significantly smaller in size compared with the wild-type

571

zebrafish adults (upper) raised in the same tank at the 90 dpf stage.

572 573

Figure 4. Hepatic steatosis and abnormal adiposity observed in socs1a-deficient adults. A:

574

representative fluorescent image of adipose tissue from adult zebrafish (90 dpf) stained with

575

Nile red obtained using an excitation wave length of 470 nm. Socs1a+/+ (Upper) and socs1a-/-

576

(lower) zebrafish were set in the same field. A marked decrease in the size of the

577

subcutaneous adipose tissue (white arrows) was observed in the socs1a-/- adults (bottom)

578

compared with the wild-type control fish (upper). B: diagram of the region used for sectioning.

579

C and D: Oil red O staining of adult zebrafish trunk sections. A socs1a+/+ sample is shown in

580

(C), and a socs1a-/- sample is shown in (D). E and F: representative images of Oil Red

581

O-stained sections from adult wild-type liver tissue at low (20×, E) and high (100×, F)

582

magnification. The high-magnification image observed in (F) is boxed in (E). G and H:

583

representative images of Oil Red O-stained sections from adult wild-type liver tissue at low

584

(20×, G) and high (100×, H) magnification. The high-magnification image observed in (H)

585

is shown as in panel (G). A markedly higher number of lipid drops are observed in the liver

586

tissue from socs1a-deficient adults compared with that from the control adults at the 90 dpf

587

stage. All of the assays were performed with at least three pairs of size-matched socs1a-/- and

588

socs1a+/+ adults, and similar results were obtained.

589 590

Figure 5. Real-time RT-PCR confirmation of the representative key signaling transcripts

591

identified from the comparative liver RNA-Seq transcriptomics analyses. RNA samples were

592

extracted from 4 livers of socs1a-/- zebrafish or control siblings at 90 dpf respectively. The

593

real-time RT-PCR measurements were performed with specific primers designed for insulin

594

(ins),

595

glucose-6-phosphatase c family a1 (g6pca.1), hexokinase domain containing 1 (shxk1),

596

glucokinase (gk), O family member of Forkhead transcription factors (foxO1a),

597

apolipoprotein A-IV (apoA4), low density lipoprotein receptor (ldlr), stearoyl-CoA desaturase

598

(scd), and hypoxia induced factor 1-alpha like (hif1αl). The relative transcript levels were

599

determined by real-time RT-PCR using β-actin as the internal standard. The expression levels

600

of some of the genes in the livers were assayed with the samples from both M1 and M2

insulin

receptor

b

(insrb),

phosphoenolpyruvate

carboxykinase

(pck),

601

mutant lines. B and C: the levels of insulin in the liver (B) and plasma (C) from socs1a-/- and

602

wild-type control adults were determined by ELISA. D and E: the levels of glucagon in the

603

liver (D) and plasma (E) samples from socs1a-/- and wild-type control adults were determined

604

by ELISA. F: blood glucose concentration in socs1a-/- and socs1a+/+ zebrafish under different

605

conditions at 90 dpf. The detailed conditions are described in the “Materials and Methods”

606

section. G: decreased levels of phosphorylated AKT in socs1a-/- fish compared with wild-type

607

control fish in hepatic tissue at 90 dpf. The data are expressed as the means ± SE from three

608

separate measurements. The asterisk (*) and double asterisks (**) indicate significant

609

differences at a (P < 0.05) and very significant differences (P< 0.01) with at least a one-fold

610

difference. The primers designed for the real-time PCR analysis are listed in Table 1.

611 612

Figure 6. Improved performance of somatic growth observed in the heterozygous

613

socs1a-deficient zebrafish. A and B: the measurements of the body weight (A) and body

614

length (B) of the socs1a+/- fish compared with their control siblings respectively. The

615

measurements for the socs1a+/- fish were assayed at 120 dpf stage for M1 (n=15 for the

616

control siblings, n=13 for the socs1a+/-) or at 185 dpf stage for M2 (n=9 for the control

617

siblings, n=21 for the socs1a+/-). The socs1a+/- fish and their control siblings from each mutant

618

line were raised in the same tank respectively. The slower overall growth of the individuals of

619

M2 than those of M1 observed might be due to different densities and culture conditions in

620

different aquariums. C: the levels of the phosphorylated-STAT5.1 in the adult hepatic samples

621

of the socs1a+/+, socs1a+/-, and their control sibling fish were quantified with Western Blot

622

analyses. D: Gray values of western blot signal were read by Image J.

623

624

Table 1. Primers used in the experiments product symbol

Gene ID

length

forward

reverse

genotyping

NM_001003467

378

GCCGGTTGGTCTTTCCTGTA

TTAATCCGGATGCTGACGGG

socs1a

NM_001003467

704

AACGACCGATGAGTCTCATC

TAATGCAACAAGTCTCGTCT

g6pca1

NM_001003512.1

104

ATCGCTGCACCTTACGAGAT

ACCCAGTGAAACACGCTCTC

foxO1a

NM_001077257.2

107

GCGGCAAAGAAAAAGCTGGC

TCATTGCTGTGGGAGTTCGG

Apof

XM_003198822.3

132

TTCTAGAAGAGCCAGTGAGTGC

TGAGGGAATGGAGCTTTGCC

ldlra

NM_001030283.1

117

ATACATACGCGTCATCCCCC

GCGCCCCTGATGCTGTAT

scd

NM_198815.2

143

CTCACACTCCTCTGGGCTTTT

AAGGCCATGGAGTTTCCGAT

igfbp1a

NM_173283.3

102

AGCGAGACAGCACCAGATCA

GTCGAATGGCTTTCCGTGC

insulin

NM_131056.1

147

GGTCGTGTCCAGTGTAAGCA

GGAAGGAAACCCAGAAGGGG

insulinR

NM_001123229.1

106

ACTGCTGGTCTTTCGGAGTG

CCTCCGTCCATGACGAACTT

pck

NM_214751.1

106

GGTCAACAACTGGCCCTGTA

CAGCAGTGAGTTTCCTCCGT

shxk1

NM_001115125.1

136

TCAGCTAATCTGGTGGCTGC

GCCTTTTGGGGTACTGTGGA

gk

NM_001045385.2

102

CACCGCTGACCTGCTATGAT

AGTCGGCCACTTCACATACG

hif1al

NM_200405

94

CGTCGAAAAGGCCCAGTTTG

CGAGCTCATAACACCTCCGT

il1b

NM_212844.2

150

TGGACTTCGCAGCACAAAATG

GTTCACTTCACGCTCTTGGATG

nfkb

NM_001001840.3

133

AAACAAGACGCAAGGAGCCC

GCTGAAGGAAACGTCATAGGC

nfkbiab

NM_199629.1

142

TGCACAGGAGCAGTGTAACG

GGTCAGGTGATAAGGCGTGT

il6

NM_001261449.1

98

ATGACGGCATTTGAAGGGGT

CGCGTTAGACATCTTTCCGTG

β-actin

NM_131031.1

99

GCCACCTTAAATGGCCTAGCA

GCCATACAGAGCAGAAGCCA

ifnγ

NM_212864.1

133

TGGGCGATCAAGGAAAACGA

TTGATGCTTTAGCCTGCCGT

tnfa

NM_212859.2

99

ATCATTTTGGCTGTGGGCCT

TGTGAGTCTCAGCACACTTCC

socs1a

625 626

Table 2. Comparison of liver triglyceride content between socs1a+/+ and socs1a-/- fish Mutant lines M1 M1 M1 M1 M2 M2 M2 M2

627 628 629

Genotypes socs1a+/+ socs1a-/socs1a+/+ socs1a-/socs1a+/+ socs1a-/socs1a+/+ socs1a-/-

Genders male male female female male male female female

Triglyceride content in the liver (μmol/g) 17.23±0.63 25.65±0.51 * 18.62±0.70g 24.80±0.69 * 20.32±0.49 39.25±0.53* 17.06±0.61 25.38±0.46*

*represents significant difference between socs1a+/+ and socs1a-/-(P value less than 0.05).

630 631

Table 3. Some of differential expressed transcripts between the hepatic tissues of socs1a-/fish and their control siblings based on RNA-seq analyses Gene ID

Full name

Symbol

RPKM +/+

*

RPKM of -/-

log2

socs1a *

Ratio**

39.171289

93.342562

1.2527384

socs1a angptl4

of

ENSDARG00000035859

angiopoietin-like 4

ENSDARG00000086281

apolipoprotein A-IV a

APOA4

0.7967262

7.0850112

3.1526143

ENSDARG00000016773

cytokine

socs8

5.4190333

1.6627393

-1.704474

foxo1a

22.812925

53.834928

1.238691

g6pca

12.742255

25.232548

1.1302395

inducible

SH2-containing protein b ENSDARG00000063540 ENSDARG00000031616

forkhead box O1 a glucose-6-phosphatase

a,

catalytic ENSDARG00000068006

glucokinase (hexokinase 4)

gck

53.531774

0.6856726

-6.286732

ENSDARG00000027529

heme

hmox1

32.919674

77.315739

1.2318121

hkdc1

7.6791699

1.5390071

-2.318951

h6pd

65.751368

31.997154

-1.039077

hig1

9.860814

22.024901

1.1593569

hif1al

34.340549

100.65683

1.55146

oxygenase

(decycling) 1a ENSDARG00000038703

hexokinase

domain

containing 1 ENSDARG00000060153

hexose-6-phosphate

ENSDARG00000022303

HIG1

dehydrogenase hypoxia

inducible

domain family, member 1A ENSDARG00000041169

hypoxia-inducible factor 1, alpha subunit, like

ENSDARG00000071524

insulin receptor b

inrb

3.6456078

8.1925728

1.1681572

ENSDARG00000032768

interferon regulatory factor

irf1b

25.657533

12.24516

-1.067171

irf2bp2b

14.869047

7.2601528

-1.03424

irf7

2.6557538

16.457309

2.6315352

irf8

7.6835218

1.7315174

-2.149731

isg15

15.326053

98.491401

2.6840116

il1b

1.3716531

0.1920888

-2.83607

receptor,

il4r

7.8087661

3.8386256

-1.024505

10

receptor,

il10ra

1.8827663

0.7812326

-1.26903

2

receptor,

il2rga

3.163493

0.8667808

-1.867779

21

receptor,

ir21

2.8691533

1.3393382

-1.099105

lipoprotein

ldlr

105.53622

239.91985

1.1848143

1b ENSDARG00000006275

interferon regulatory factor 2 binding protein 2b

ENSDARG00000045661

interferon regulatory factor 7

ENSDARG00000056407

interferon regulatory factor 8

ENSDARG00000086374

interferon stimulated gene, 15 kDa

ENSDARG00000005419

interleukin 1, beta

ENSDARG00000031051

interleukin

4

tandem duplicate 1 ENSDARG00000041180

interleukin alpha

ENSDARG00000068858

interleukin gamma a

ENSDARG00000069961

interleukin

tandem duplicate 1 ENSDARG00000029476

low

density

receptor a ENSDARG00000040884

max

interactor

1,

mxi1

5.9676921

17.655519

1.5648742

pck1

63.775129

205.1901

1.6858953

ins

0.5725708

5.4124101

3.2407452

scd

389.23362

111.45556

-1.804168

tnfsf10

3.8694333

0.655503

-2.561448

tnfb

1.3868937

0.2158035

-2.684067

dimerization protein ENSDARG00000013522

phosphoenolpyruvate carboxykinase 1 (soluble)

ENSDARG00000035350

preproinsulin

ENSDARG00000033662

stearoyl-CoA

desaturase

(delta-9-desaturase) ENSDARG00000057241

tumor

necrosis

(ligand)

factor

superfamily,

member 10 ENSDARG00000013598

tumor necrosis factor b (TNF superfamily, member 2)

632 633 634 635

*RPKM: reads per kilobase of mRNA length per million mapped reads **socs1a mutant/socs1a control ***socs1a mutant/socs1a control

start codon

A

exon2

exon1

WT: atagaagatcgccaagtgagcactgaggcctcctcagacgtcttccaatcccaaagat M1: atagaagatcgccaagtgagcactgagg-ctcctcagacgtcttccaatcccaaagat (Δ1) M2: atagaagatcgccaagtgagcactgaggccctcctcagacgtcttccaatcccaaagat (+1)

B

-/-

+/-

+/-

+/+

+/+

M C

C deletion

M2

M1

D

E

22aa

SOCS1a +/+

2 201aa

SOCS1a -/- M1

63aa

SOCS1a -/- M2

118aa

C insertion

Relative expression of Socs1a

1.5 1 0.5

0 con

Figure 1

M1

M2

B

gata1

24 hpf

A P-STAT5.1

socs1a+/+

STAT5.1 β-actin

Numbers (106) cell counts(million)

D

Hbbe1 β-actin

E 4

Red blood cell counts

3 2 1 0

socs1a+/+ socs1a-/-

Figure 2

Expression levels (folds)

C

socs1a-/1.80 1.60 1.40 1.20 1.00 0.80 0.60 0.40 0.20 0.00

socs1a+/+ socs1a-/-

IFNγ

il1b

nfkb2 nfkbiab

IL6

TNFα

A

B *

0.5

socs1a+/+

socs1a-/-

50μm

C

100%

Body weight (gram)

3 dpf

0.45 0.35

*

0.3 0.25 0.2 0.15 0.1

socs1a+/+

0.05

socs1a-/-

80%

0

D

60%

20

40

60

90

dpf 120 socs1a+/+

40% socs1a+/+

20%

socs1a-/-

socs1a-/-

0% 0

20

Figure 3

40

50

60

70

80

90 100 110 120

dpf

5mm

90 dpf

Survival rate (%)

*

0.4

Socs1a+/+

A 60mm

Socs1a+/+

Socs1a-/-

C

D

Subcutaneous

Socs1a-/-

B 5mm

F

25um

3um

socs1a+/+

E

H

25um

3um

socs1a-/-

G

Figure 4

5mm

A Relative expression levels˄folds˅

18 16 14

**

12

socs1a+/+

**

**

10

M1

**

*

8

M2

**

6 4

*

*

*

2

*

** **

**

** **

0

Hepatic insulin concentration (mIU/g )

0.0400

*

Plasma Glucagon concentration(pg/g)

C

0.0300 0.0200 0.0100 0.0000 socs1a+/+

E

G6pca.1

F

3.500 3.000 2.500 2.000 socs1a+/+ socs1a-/-

GK

foxO1a

*

6.000 5.500 5.000 4.500

apoA4

LDLR

D

10

G

socs1a+/+ socs1a-/-

P-AKT

8 6

AKT

4 2

Β-actin

0

chow diet

fasted

glucose treated

Hif1al

3.00 2.00 1.00 0.00 socs1a+/+ socs1a-/-

socs1a+/+ socs1a-/-

12

SCD

4.00

4.000

socs1a-/-

4.000

Figure 5

shxk1

Hepatic glucagon concentration (pg/g)

pck

blood glucose˄mmol/L˅

B

insrb

Plasma insulin Concentration (mIu/mg)

ins

body weight(g)

A

*

0.5

socs1a+/+

0.4

socs1a+/-

0.3

*

2.9

B

*

socs1a+/+

2.8 body length(cm)

0.6

0.2

socs1a+/-

2.7

*

2.6 2.5 2.4

0.1

2.3

0 M1

M1

M2

M2

P-STAT5.1 STAT5.1 β-actin

Relative gray value

D

C

0.7 0.6 0.5 0.4 0.3 0.2 0.1 0 socs1a+/+ socs1a+/- socs1a-/-

Figure 6