1 loss of egr-1 uncouples compensatory responses of ...pancreatic 6 cells undergo proliferation and...

47
1 Loss of EGR-1 uncouples compensatory responses of pancreatic cells 1 2 Sy-Ying Leu 1* , Li-Hua Kuo 1,2* , Wen-Tsan Weng 1* , I-Chia Lien 1* , Ching-Chun Yang 1 , Tai- 3 Tzu Hsieh 1 , Yi-Ning Cheng 1 , Po-Hsiu Chien 3 , Li-Chun Ho 1,4 , Shun-Hua Chen 5 , Yan-Shen 4 Shan 1,6,7 , Yun-Wen Chen 8 , Pei-Chun Chen 3 , Pei-Jane Tsai 9 , Junne-Ming Sung 10 , and Yau- 5 Sheng Tsai 1,3,6 6 1 Institute of Clinical Medicine, 2 Institute of Basic Medical Sciences, 3 Department of 7 Physiology, 5 Department of Microbiology and Immunology, 8 Department of 8 Pharmacology, and 9 Department of Medical Laboratory Science and Biotechnology, 9 College of Medicine, National Cheng Kung University, Tainan; 4 Division of Nephrology, 10 Department of Internal Medicine, E-DA Cancer Hospital, Kaohsiung; 6 Research Center 11 of Clinical Medicine, 7 Department of Surgery, and 10 Division of Nephrology, Department 12 of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan, ROC. 13 *contributed equally to this work 14 Address correspondence to: Yau-Sheng Tsai, PhD, 1 University Rd, National Cheng 15 Kung University, Tainan 701, Taiwan, R.O.C. Tel: +886-6-2353535 ext. 4242; Fax: +886- 16 6-2758781 17 E-mail: [email protected] 18 ORCID iD: 0000-0002-5560-2661 19 Running title: EGR-1 in β-cell compensation 20 Word count: abstract, 220; main text, 7416 words 21 22

Upload: others

Post on 20-Feb-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

1

Loss of EGR-1 uncouples compensatory responses of pancreatic cells 1

2

Sy-Ying Leu1*, Li-Hua Kuo1,2*, Wen-Tsan Weng1*, I-Chia Lien1*, Ching-Chun Yang1, Tai-3

Tzu Hsieh1, Yi-Ning Cheng1, Po-Hsiu Chien3, Li-Chun Ho1,4, Shun-Hua Chen5, Yan-Shen 4

Shan1,6,7, Yun-Wen Chen8, Pei-Chun Chen3, Pei-Jane Tsai9, Junne-Ming Sung10, and Yau-5

Sheng Tsai1,3,6 6

1Institute of Clinical Medicine, 2Institute of Basic Medical Sciences, 3Department of 7

Physiology, 5Department of Microbiology and Immunology, 8Department of 8

Pharmacology, and 9Department of Medical Laboratory Science and Biotechnology, 9

College of Medicine, National Cheng Kung University, Tainan; 4Division of Nephrology, 10

Department of Internal Medicine, E-DA Cancer Hospital, Kaohsiung; 6Research Center 11

of Clinical Medicine, 7Department of Surgery, and 10Division of Nephrology, Department 12

of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan, ROC. 13

*contributed equally to this work 14

Address correspondence to: Yau-Sheng Tsai, PhD, 1 University Rd, National Cheng 15

Kung University, Tainan 701, Taiwan, R.O.C. Tel: +886-6-2353535 ext. 4242; Fax: +886-16

6-2758781 17

E-mail: [email protected] 18

ORCID iD: 0000-0002-5560-2661 19

Running title: EGR-1 in β-cell compensation 20

Word count: abstract, 220; main text, 7416 words 21

22

Page 2: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

2

Abstract 1

Rationale: Subjects unable to sustain β-cell compensation develop type 2 diabetes. Early 2

growth response-1 protein (EGR-1), implicated in the regulation of cell differentiation, 3

proliferation, and apoptosis, is induced by diverse metabolic challenges, such as glucose 4

or other nutrients. Therefore, we hypothesized that deficiency of EGR-1 might influence 5

β-cell compensation in response to metabolic overload. 6

Methods: Mice deficient in EGR-1 (Egr1-/-) were used to investigate the in vivo roles of 7

EGR-1 in regulation of glucose homeostasis and beta-cell compensatory responses. 8

Results: In response to a high-fat diet, Egr1−/− mice failed to secrete sufficient insulin to 9

clear glucose, which was associated with lower insulin content and attenuated 10

hypertrophic response of islets. High-fat feeding caused a dramatic impairment in 11

glucose-stimulated insulin secretion and downregulated the expression of genes encoding 12

glucose sensing proteins. The cells co-expressing both insulin and glucagon were 13

dramatically upregulated in islets of high-fat-fed Egr1−/− mice. EGR-1-deficient islets 14

failed to maintain the transcriptional network for β-cell compensatory response. In human 15

pancreatic tissues, EGR1 expression correlated with the expression of β-cell 16

compensatory genes in the non-diabetic group, but not in the diabetic group. 17

Conclusion: These results suggest that EGR-1 couples the transcriptional network to 18

compensation for the loss of β-cell function and identity. Thus, our study highlights the 19

early stress coupler EGR-1 as a critical factor in the development of pancreatic islet 20

failure. 21

Keywords: β-cell compensation, β-cell identity, immediate genes, islet failure, stimulus-22

response coupling 23

Page 3: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

3

1

2

Page 4: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

4

Introduction 1

The pathogenesis of type 2 diabetes reflects an unbalanced dynamic interaction 2

between environmental factors, insulin-responsive tissues, and insulin-producing cells. 3

When insulin-responsive tissues become less sensitive to insulin or circulating nutrient 4

levels are increased, the demand for insulin grows to maintain normoglycemia. Pancreatic 5

cells undergo proliferation and increase output of secreted insulin. A coordinated 6

increase in -cell mass, insulin biosynthesis, and secretion comprises the -cell 7

compensatory response to increased level of nutrients (particularly glucose and fatty acid) 8

or growth hormones, e.g., incretin [1]. Individuals who fail to respond to the increased 9

insulin demand develop hyperglycemia, a hallmark of type 2 diabetes [1]. Thus, -cell 10

compensatory response is considered a critical determinant of whether glucose 11

intolerance will advance to diabetes. 12

13

Reduction in -cell number and progressive impairment of -cell function attenuate 14

-cell compensatory response [1]. However, Talchai et al. recently proposed that it is the 15

loss of -cell identity, rather than -cell death, that largely accounts for the significant 16

impairment of -cell function during diabetes progression [2], suggesting the importance 17

of sustaining the fully differentiated functional state of cells. Beta-cell identity is 18

defined by the presence of a specific gene expression signature and insulin [3]. 19

Maintenance of -cell identity in pancreatic islets requires a dedicated transcriptional 20

network [4]. For example, PDX1 deletion in postnatal islets resulted in a loss of cells 21

[5], whereas cells with NKX6.1 deletion assumed characteristics of cells [6]. In 22

addition, PAX4 is essential for the differentiation of - and -cell lineages, whereas ARX 23

is involved in the specification of - and pancreatic polypeptide-cell fates [7]. Moreover, 24

Page 5: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

5

other transcription factors, including NKX2.2, PAX6, and MAFA, have been linked to 1

the maintenance and development of functional cells [2, 8-10]. Therefore, transcription 2

factors involved in the pancreatic lineage development play a vital role in the mechanism 3

that allows cells to maintain identity and adapt to changing metabolic demands that 4

occur throughout life [11]. 5

6

It is conceivable that there is a mechanism that senses hostile environment and detects 7

toxic effects of nutrients, switching the transcription cascade that maintains -cell 8

function and identity. Early growth response-1 (EGR-1), an immediate-early transcription 9

factor, is the earliest downstream nuclear target sensitive to changes in the extracellular 10

environment. EGR-1 is barely expressed in the basal state. Many different stimulations, 11

including those with growth factors, hormones, cytokines, and stress inducers, promptly 12

induced EGR-1 expression [12]. The newly synthesized EGR-1 couples early 13

extracellular stimuli to long-term responses by changing expression of the target genes. 14

Our previous study showed that EGR-1 is rapidly and transiently induced by fatty acids, 15

leading to the rescue of β cells from fatty acid-induced ER stress and apoptosis [13]. 16

Those results indicated that EGR-1 is a critical early sensor in pancreatic cells that 17

enables cellular adaptation in response to fatty acid toxicity. Thus, inhibition of this 18

stimulus-transcription coupling machinery would impair maintenance and development 19

of cells, and underlie the development of diabetes in conditions of the metabolic stress. 20

21

Although EGR-1 expression is low in most tissues, it is highly enriched in pancreatic 22

islets [14]. Glucose and other secretagogues have been shown to rapidly induce EGR-1 23

expression in insulinoma and in primary islet cells [15, 16]. Functional studies revealed 24

Page 6: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

6

that EGR-1 controls insulin biosynthesis via regulation of PDX1 expression [17-19]. 1

These studies implicate a role of EGR-1 in the transcriptional cascade that regulates -2

cell identity. In addition, EGR-1 was shown to regulate -cell proliferation [14] and 3

rescue cells from ER stress and apoptosis by enhancing insulin/AKT signaling in our 4

previous study [13]. Although expressing a dominant-negative mutant of EGR-1 in 5

pancreatic -cells has been shown to impair insulin synthesis, glucose homeostasis and 6

islet size in vivo [19], the bona fide nature of EGR-1 in the stimulus-transcription 7

coupling of -cell compensatory response and identity has remained unclear. In this 8

study, we hypothesized that the loss of EGR-1 uncouples metabolic stress from the 9

transcriptional cascade, which is essential for augmenting -cell compensatory response 10

and maintenance of -cell identity. We tested whether EGR-1 directly regulates glucose 11

homeostasis and -cell compensatory responses using EGR-1-deficient (Egr1−/−) mice. 12

13

14

15

16

17

18

19

Page 7: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

7

Materials and Methods 1

Animals Egr1−/− mice, kindly provided by J. Milbrandt, Washington University in St. 2

Louis, St. Louis, Missouri, USA [20], were maintained on a C57BL/6 genetic 3

background. Studies were carried out using male mice and age-matched littermate wild-4

type (WT) mice. Mice were fed ad libitum either a regular chow (RC; Laboratory Rodent 5

Diet 5001, Purina, St. Louis, MO, USA) or a high-fat diet (HF; D12492, Research Diets, 6

New Brunswick, NJ, USA) for two to three months starting at 2 months of age. Animals 7

were handled following procedures approved by the Institutional Animal Care and Use 8

Committees of National Cheng Kung University. 9

Glucose metabolic assays Mice were fasted for 5 h and given an oral glucose bolus (2 10

g/kg body weight) or intraperitoneally injected with human regular insulin (0.4 U/kg 11

body weight, Eli Lilly, Indianapolis, IN, USA). Blood samples were collected before and 12

at 15, 30, 60, and 120 min after injections. Plasma glucose concentration was determined 13

by a glucose colorimetric test (Autokit Glucose, Wako, Osaka, Japan). Insulin was 14

measured using mouse insulin ELISA (Ultrasensitive Mouse Insulin ELISA, Mercodia, 15

Uppsala, Sweden). The insulin resistance (IR) index was calculated as the product of the 16

areas under glucose and insulin curves in glucose tolerance tests as previously described 17

[21]. For acute insulin secretion assay, mice were overnight fasted, and intraperitoneally 18

injected with glucose (3 or 4 g/kg). Blood samples were collected at 0, 2, 5, 15, and 30 19

min after the injection. 20

Morphological analysis Serial paraffin sections with 5 μm in thickness from four 21

different levels of pancreas (100 μm apart) were stained with H&E. The cross sectional 22

Page 8: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

8

area of islet and total tissue in four overlapping images were analyzed by ImageJ 1

software [22]. The minimum size that is considered an islet is a diameter above 50 μm. 2

Mean islet size is calculated by dividing the total islet area by the islet number. The ratio 3

of islet area versus total tissue area was calculated and then multiplied by the weight of 4

the pancreas to represent the endocrine mass. Similarly, insulin-stained section was 5

used to determine relative cross-sectional area of β cells (insulin-positive) and calculated 6

for β-cell mass [23]. Quantification of β-cell proliferation and apoptosis was done by 7

counting the insulin-positive cells with Ki67 and caspase-3 signals, respectively. At least 8

500 β cells were counted for each experimental condition. 9

Pancreatic islet isolation and dispersion Pancreatic islets were isolated using a 10

modified protocol [24]. Briefly, ampulla was clamped and collagenase XI (1 mg/mL, 11

Sigma-Aldrich, St. Louis, MO, USA) was injected into the common bile duct. The 12

pancreata were removed and incubated, and islets were separated by sedimentation and 13

hand-pick. To obtain isolated single islet cells, islets were dispersed with 0.1% trypsin. 14

Isolated cells were seeded onto glass coverslips pre-coated with poly-L-lysine (Sigma-15

Aldrich, St. Louis, MO, USA) for treatment and immunofluorescence staining. 16

Islet RNA analysis All islets from individual animals were collected and extracted using 17

the TRIzol Reagent (Invitrogen, Carlsbad, CA, USA). mRNA were analyzed with SYBR 18

green-based real-time quantitative RT-PCR (Applied Biosystems, Foster City, CA, USA) 19

with Actb as the reference gene in each reaction. Sequences of the primers used for RT-20

PCR assays are shown in Table S1. 21

Glucose-stimulated insulin secretion Batches of ten isolated islets were transferred to 22

Page 9: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

9

incubation vials containing pre-gassed Krebs–Ringer HEPES bicarbonate buffer in the 1

presence of 2.8 mM glucose. After incubation for 1 h, batch triplicates were transferred to 2

a solution containing 16.7 mM glucose for 1 h, and supernatant fractions were collected 3

and assayed for insulin ELISA. 4

Western blot analysis Animal tissue lysates in RIPA lysis buffer were subjected to 5

electrophoresis and transferred onto the polyvinylidene difluoride membranes (Millipore, 6

Bedford, MA, USA). Insulin, p35, CDK5, PCNA, CDC2, pS473-AKT, AKT, pT202/204-7

ERK1/2, ERK1/2, pS217/221-MEK1/2, MEK1/2, pS21/9-GSK3α/β, GSK3α/β (Cell 8

Signaling, Danvers, MA,USA) on the membrane were probed with respective primary 9

antibodies and horseradish peroxidase (HRP)-conjugated secondary antibodies (Vector 10

Laboratories, Burlingame CA, USA). Immunoreactivity was visualized by ECL plus 11

luminal solution (GE Healthcare, Pittsburgh, PA, USA). 12

Immunofluorescence staining Paraffin-section containing pancreas tissue were 13

deparaffinized, dehydrated, and boiled for epitope retrieval using an antigen retrieval 14

buffer at pH = 6.0 (Opal 4-color IHC Kit, PerkinElmer, Waltham, MA). Tissue sections 15

were blocked for 10 min and incubated with primary antibodies overnight at 4℃. Primary 16

antibodies include: MAFA, PDX1 (Cell Signaling, Danvers, MA, USA), pancreatic 17

polypeptide, somatostatin, NKX2.2, PAX4, PAX6, ARX, MAFA (Santa Cruz 18

Biotechnology, Dallas, TX, USA), Ki67, PDX1 (Abcam, Cambridge, UK), glucagon and 19

insulin (Sigma-Aldrich, St. Louis, MO, USA). The sections were incubated with Polymer 20

HRP-conjugated secondary antibodies for 10 min at room temperature. The primary 21

antibodies were stained with repeat procedures including antigen stripping and blocking 22

Page 10: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

10

steps. Cell nuclei was stained via adding mounting media containing fluoroshield with 1

DAPI. The images were visualized by confocal microscopy (C1-Si, Nikon). To quantify 2

colocalization, we used the Pearson's correlation coefficient (Rr). The images were 3

analyzed by using PSC Colocalization plug-in (ImageJ) [25, 26]. R ranges between -1 4

(perfect negative correlation) to +1 (perfect positive correlation) with 0 meaning no 5

correlation. 6

Bioinformatics analysis The PROMO database was used to identify proteins that contain 7

potential EGR-1 binding site within endocrine lineage developmental program. 8

Functional analysis of interacting proteins was determined using a commercial System 9

Biology package, Ingenuity Pathways Analysis (IPA 5.0, Ingenuity Systems), following 10

the application protocols. The results from the quantitative PCR were loaded into the IPA 11

program to visualize the associated changes by EGR-1 deficiency or HF feeding. 12

Cell culture MIN6 cells were maintained in high-glucose DMEM supplemented with 10 13

% FBS. For EGR-1 overexpression, cells were transfected with plasmids expressing 14

EGR-1 or control pCMV vector using a transfection reagent (Lipofectamine 3000, 15

Invitrogen). For EGR-1 knockdown, EGR-1 short hairpin RNA (shEGR-1, 16

TRCN0000301750) and control shLUC (TRCN0000072249) constructs from National 17

RNAi Core Facility were integrated into a lentiviral delivery system as described 18

previously [13]. 19

Chromatin immunoprecipitation (ChIP) assay The procedure for ChIP was described 20

previously [27]. In brief, the interaction between protein and DNA was fixed by using 1% 21

formaldehyde for 10 min. Cells were harvested and sonicated to fragment DNA (average 22

Page 11: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

11

size of 200–500 bp). EGR-1 antibody (#4154S; Cell signaling) was used to 1

immunoprecipitate the EGR-1 protein and DNA complexes. Potential EGR-1 binding 2

sites were amplified by specific primers (Table S2) after reversing cross-linking. 3

EGR-1 overexpression in mice We injected a single dose of EGR-1 plasmid (35 μg 4

pCMV-EGR-1 plasmid/1.6 ml saline/mouse) into HF-fed WT mice by hydrodynamics-5

based gene delivery through the tail vein as described previously [28]. Mice received the 6

empty vector pCMV (35 μg pCMV plasmid/1.6 ml saline/mouse) as controls. The 7

animals were euthanized at days 0, 1, 3, 7, or 10 after plasmid administration. Liver and 8

pancreas tissues were analyzed for protein abundance. Because expression of EGR-1 was 9

rapidly increased at day 3 after a single injection of EGR-1 plasmid, we then performed 10

acute in vivo insulin secretion assay at day 3 in the HF-fed WT mice. 11

Collection of human pancreas tissues Normal pancreas specimens were obtained from 12

58 patients who underwent pancreatic surgery for pancreatitis or pancreatic cancer at the 13

Department of Surgery of National Cheng Kung University Hospital from Jan. 2013 to 14

Apr. 2014. Normal structure containing islets from pancreatic tissues was confirmed by 15

microscopic examination. Ethics approval for human studies was obtained from the 16

Institutional Review Board of National Cheng Kung University Hospital, and informed 17

consent was given by each patient. 18

Data analysis Values are reported as mean ± SEM. Statistical analyses were conducted 19

by Student’s t-test or one-way ANOVA followed by Bonferroni multiple comparison test. 20

Correlation was evaluated using Spearman’s rank correlation coefficient. Differences 21

were considered to be statistically significant at P<0.05. 22

Page 12: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

12

Results 1

Impaired glucose homeostasis in Egr1−/− mice after high-fat diet feeding. The body 2

weight was similar in wild-type (WT) and Egr1−/− mice in both basal (regular chow, RC) 3

and enhanced metabolic demand (high-fat, HF) conditions (Figures 1A and S1). Except 4

for the lower liver weight in Egr1−/− mice, organ weights of white adipose tissue, 5

including gonadal and inguinal fat, brown adipose tissue, and kidney did not differ 6

between genotypes regardless of diets (Figure 1B). To assess the effect of EGR-1 7

deficiency on whole-body glucose utilization, we performed an oral glucose tolerance 8

test. Although Egr1−/− mice fed RC cleared glucose as efficiently as WT mice (Figure 1C, 9

upper panel), we found a slight reduction in insulin levels of RC-fed Egr1−/− mice during 10

the oral glucose tolerance test (Figure 1D, upper panel). When mice were placed on the 11

HF diet, Egr1−/− mice could not efficiently clear glucose and had significantly lower 12

insulin levels (lower panels of Figure 1C and D). These results suggest that Egr1−/− mice 13

responded normally to glucose challenge in the basal condition, but were incapable of 14

handling glucose challenge when insulin demand was increased. Consistent with the data 15

from previous studies [29, 30], the insulin tolerance test showed that Egr1−/− mice were 16

protected from diet-induced insulin resistance (Figure S2A). Moreover, compared to HF-17

fed WT mice, HF-fed Egr1−/− mice had higher phosphorylation levels of IRS1 Tyr896 18

and Akt Ser473 in the liver (Figure S2B), and higher energy expenditure, as measured by 19

oxygen consumption (VO2) and carbon dioxide production (VCO2) during both dark and 20

light phases (Figure S2C). 21

22

Reduced islet size and insulin content in Egr1−/− mice after HF diet consumption. 23

Given that increases in -cell mass and insulin biosynthesis are important in islet 24

Page 13: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

13

compensatory response, we first determined the involvement of EGR-1 in the increase of 1

-cell mass. Pancreas weight in Egr1−/− mice did not differ from that of WT mice 2

regardless of diets (Figure 2A and B). Islet morphometric analysis revealed normal islet 3

size and endocrine mass of RC-fed Egr1−/− mice (Figure 2C and D). HF feeding doubled 4

pancreas weight as well as tripled islet size and endocrine mass in WT mice, reflecting a 5

compensatory islet hypertrophic response. Although Egr1−/− mice increased pancreas 6

weight in response to HF-feeding, the increases in the islet size and endocrine mass in 7

Egr1−/− mice were attenuated. Thus, attenuation of the increases of islet size and 8

endocrine mass may explain the failure to handle glucose challenge in HF-fed Egr1−/− 9

mice. 10

11

We next determined insulin content in the pancreas lysate, and found that insulin 12

protein levels were reduced regardless of diet (Figure 2E and F). Insulin intensity within 13

islets of RC-fed Egr1−/− mice was slightly lower than that in islets of RC-fed WT mice 14

(Figure 2G). HF feeding increased islet size and insulin intensity in WT mice, but these 15

increases was attenuated in Egr1−/− mice (Figure 2G). Consistently, the levels of -cell 16

mass and C-peptide were significantly reduced in HF-fed Egr1−/− mice (Figure 2H and I). 17

We further examined insulin secretory vesicle in cells from HF-fed WT and Egr1−/− 18

mice by transmission electron microscopy (Figure 2J). Ultrastructural analysis according 19

to Orci et al [31] showed that the proportion of mature vesicles, which contain electron-20

dense granules, was lower in HF-fed Egr1−/− mice (Figure 2K). The proportion of 21

immature vesicles, which contain diffused gray granules, was increased in HF-fed Egr1−/− 22

mice. Together, these results indicate that EGR-1 deficiency impaired insulin biosynthesis 23

and affected the formation of insulin secretory vesicles. 24

Page 14: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

14

1

Reduced glucose-stimulated insulin secretion in Egr1−/− mice after HF diet 2

consumption. Given that -cell compensation also involves its responsiveness of 3

nutrient-secretion coupling, we then tested its sensitivity to the nutrient stimuli. We first 4

measured expression of genes involved in glucose sensing and secretion coupling from 5

isolated islets. Expression of genes encoding proteins necessary for glucose entry, such as 6

glucose transporter (Glut2) and glucokinase (Gck), and secretion coupling, such as ATP-7

sensitive potassium channels KIR6.2 (Kcnj11) and SUR1 (Abcc8), did not differ between 8

RC-fed Egr1−/− and WT mice (Figure 3A, upper panel). However, expression levels of 9

these genes were significantly lower in HF-fed Egr1−/− mice than in HF-fed WT mice 10

(Figure 3A, lower panel). We next performed acute in vivo insulin secretion assay to 11

examine the machinery of glucose sensing and insulin secretion in Egr1−/− mice. Whereas 12

RC-fed Egr1−/− mice secreted normal amount of insulin in response to exogenous 13

glucose, HF-fed Egr1−/− mice showed attenuated glucose-stimulated insulin secretion 14

(GSIS) (Figure 3B). However, insulin secretion in response to L-arginine was similar in 15

Egr1−/− and WT mice regardless of diets (Figure 3C).We then isolated pancreatic islets 16

and performed GSIS ex vivo. Exposure of islets to 16.7 mmol/L glucose resulted in 17

insulin secretion into the culture medium. Islets from RC-fed Egr1−/− mice tended to 18

secrete less insulin, and islets from HF-fed Egr1−/− mice showed significantly attenuated 19

insulin secretion (Figure 3D). Taken together, EGR-1 deficiency, in conjunction with HF 20

diet consumption, impaired the nutrient-secretion coupling, which was likely mediated 21

through the downregulation of glucose-sensing machinery. 22

23

Loss of EGR-1 attenuates the survival pathway in the islet. Because we previously 24

Page 15: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

15

found that EGR-1 deficiency enhanced palmitic acid-induced apoptosis in β cells [13], we 1

speculated that the impairment of -cell compensation during Egr1 deficiency is 2

attributed to compromised survival of islet cells. We found that Ki67-positive cells 3

were significantly reduced in the islets of HF-fed Egr1−/− mice compared with their 4

number in the islets of HF-fed WT mice (Figure 4A, B, and C). Moreover, HF-fed Egr1−/− 5

mice showed an increased cleaved caspase-3 signal in cells (Figure 4B). The regulation 6

of -cell survival is mediated by CDK5/p35 activation and PI3K/AKT signaling 7

pathways [32]. AKT has also been proven to modulate -cell proliferation by regulating 8

cyclin D1 and cyclin D2 levels [33]. We observed that expression levels of proliferation-9

related genes, including PCNA (Pcna), Ki67 (Mki67), p35 (Cdk5r1), CDC2 (Cdk1), 10

cyclin A2 (Ccna2), cyclin D1 (Ccnd1), and cyclin D2 (Ccnd2), were decreased in HF-fed 11

Egr1−/− mice compared to those in HF-fed WT mice (Figure 4C). CDK5, p35, PCNA and 12

CDC2 protein levels were consistently reduced in the pancreas of HF-fed Egr1−/− mice 13

(Figure 4D). Furthermore, phosphorylation of AKT Ser473, which is crucial for -cell 14

growth and survival [32], as well as Ser21/9 phosphorylation of its substrate GSK-3α/β, 15

was significantly decreased in the pancreas of HF-fed Egr1−/− mice. In addition, 16

phosphorylation of MEK1/2 and ERK1/2, which are implicated in the regulation of 17

expression of survival genes and prevention of apoptosis [34], was also decreased in the 18

pancreas of HF-fed Egr1−/− mice. Together, these results suggest that EGR1 deficiency 19

attenuates islet cell proliferation possibly through the downregulation of multiple 20

signaling pathways, including CDK5/p35-mediated AKT and MEK/ERK pathways, in 21

the HF-induced compensatory response. 22

23

Loss of -cell identity in the islets of HF-fed Egr1−/− mice. During prolonged metabolic 24

Page 16: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

16

stress, cells lose expression of functional markers and convert to other endocrine cell 1

types [2]. We next examined whether EGR-1 deficiency altered morphology and 2

composition of the pancreatic islet. In contrast to the decreased percentage of insulin-3

positive cells, we found increased percentages of glucagon-positive cells, pancreatic 4

polypeptide-positive PP cells, and somatostatin-positive cells in the islets of HF-fed 5

Egr1−/− mice (Figure 5A, B, C, and D). The decreased percentage of cells and increased 6

percentages of , PP, and cells in the islets were observed in RC–fed Egr1−/− mice 7

(Figure S3), and even in the neonatal Egr1−/− mice (Figure S4). However, we did not 8

detect Ki67-positive PP- and δ-cells (Figure S5). Interestingly, the cells co-expressing 9

insulin and glucagon, which are rarely found in the islets of HF-fed WT mice, were 10

dramatically upregulated in the islets of HF-fed Egr1−/− mice (Figure 5A). In addition, the 11

islets of Egr1−/− mice also contained other bi-hormonal cells, including those co-12

expressing insulin and pancreatic polypeptide as well as those co-expressing insulin and 13

somatostatin (Figure 5B and C). Analysis of the correlation of colocalizing hormones, 14

reflected by the Pearson's correlation coefficient, showed that the colocalizations of 15

insulin and glucagon as well as insulin and somatostatin were increased in the islets of 16

Egr1−/− mice (Figure 5E). Further characterization of the islets of Egr1−/− mice revealed 17

that transcription factors maintaining -cell identity, including NKX2.2, MAFA, and 18

PAX6, were downregulated (Figure 5F). Interestingly, we noticed that glucagon-labeled 19

cells expressed typical -cell transcription markers, such as PDX1, PAX4, and PAX6, 20

in the islets of HF-fed Egr1−/− mice (Figure 6A, B, and C). However, expression of α-cell 21

transcription marker ARX was limited to glucagon-labeled α-cells in the islets of HF-fed 22

Egr1−/− mice (Figure 6D). These results suggest that EGR-1 deficiency results in the loss 23

of clear identification between α and β cells during enhanced metabolic demand. 24

Page 17: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

17

1

Decreased transcriptional network in the islets of HF-fed Egr1−/− mice. Given that 2

EGR-1 deficiency attenuated several key components of -cell compensatory response, 3

we hypothesized that EGR-1 sets an early checkpoint for the compensatory network. To 4

test this hypothesis, we first performed the bioinformatics analysis to screen for the 5

transcriptional factors and endocrine molecules that contain EGR-1 binding site. The 6

prediction results showed that 20 (out of 38) proteins within the developmental program 7

of islet lineages [35] contain an EGR-1 binding site (Figure 7A). Most of the 8

transcriptional factors carrying EGR-1 binding site are involved in the development of 9

posterior foregut, pancreatic endoderm, and endocrine cell maturation. We next 10

determined expression levels of genes in pancreatic islets by using quantitative PCR. 11

Expression levels of genes encoding members of the hepatocyte nuclear factor family 12

(HNF1A, HNF1B, HNF3B, and HNF4A), which are required for proper pancreatic 13

development [11, 36], were significantly downregulated in the islets of HF-fed Egr1−/− 14

mice (Figure 7B). The genes encoding endocrine lineage-specified transcription factors, 15

such as PDX1, NEUROG3, and NEUROD1 [11], were also downregulated in the islets of 16

HF-fed Egr1−/− mice. Furthermore, expression levels of genes encoding transcription 17

factors that maintain -cell identity, such as NKX2.2, NKX6.1, MAFA, PAX4, and PAX6 18

[37, 38], as well as insulins, INS1 and INS2, were downregulated in islets of HF-fed 19

Egr1−/− mice. 20

21

Finally, to gain further insight into the putative pathways associated with EGR-1 in 22

the compensatory network, we performed the Ingenuity Pathways Analysis (IPA). We 23

first examined the response of high-fat feeding on the islets of WT and Egr1−/− mice. 24

Page 18: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

18

High-fat feeding up-regulated (red) several transcription factors, including PDX1, 1

NKX6.1, NEUROD1, NEUROG3, PAX6, and MAFA, in the islets of WT mice (Figure 2

7C). Although high-fat feeding also up-regulated PDX1, NKX6.1, NEUROD1, and 3

NEUROG3 in the islets of Egr1−/− mice, it did not up-regulate PAX6 and MAFA (Figure 4

7D). Therefore, high-fat feeding could normally bring up several transcription factors, but 5

the deficiency of EGR-1 causes a different response in upregulation of these transcription 6

factors. We further examined the effect of EGR-1 deficiency on the transcription network 7

in both regular and HF diet feeding. In regular diet, EGR-1 deficiency down-regulated 8

(green) transcription factors, including HNF4A, PDX1, NKX2.2, NKX6.1, NEUROG3, 9

PAX4, PAX6, and MAFA (Figure 7E). EGR-1 knockout combined with HF diet leads to 10

further down-regulation in transcription factors FOXA2, HNF1B, HNF1A, and 11

NEUROD1 (Figure 7F). Thus, EGR-1 deficiency alone has an impact on transcription 12

network, and EGR-1 knockout combined with HF diet leads to a synergistic effect. 13

14

To solidify the effect of EGR-1 knockout in β-cells, we conducted a series of 15

experiments in MIN6 mouse pancreatic β cells. Firstly, knockdown of EGR-1 by short 16

hairpin RNA (shRNA) significantly decreased HNF4A, PDX1, NEUROD1, NKX6.1, 17

MAFA, and PAX6, whereas it significantly increased ARX (Figure 7G). While EGR-1 18

knockdown modestly decreased INS1, it did not affect expression of INS2. Secondly, we 19

further demonstrated the binding of EGR-1 to the genomic sequence of these potential 20

target genes. Chromatin immunoprecipitation (ChIP)-PCR in MIN6 cells showed that 21

EGR-1 bound to the promoter region of Pdx1 and Arx (Figure 7H and Figure S6). These 22

results suggest that EGR-1 affects β-cell function in a cell-intrinsic mechanism. Finally, 23

we overexpressed EGR-1 in the HF-fed WT mice by hydrodynamics-based gene delivery. 24

Page 19: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

19

Expression of EGR-1 was rapidly increased at day 3 after a single injection of EGR-1 1

plasmid (Figure S7A). We then performed acute in vivo insulin secretion assay at day 3 in 2

the HF-fed WT mice, and found that EGR-1 overexpression increased the second phase 3

of GSIS (Figure S7B). Thus, EGR-1 plays a key role in regulation of the production of 4

insulin and glucose homeostasis in HF-fed mice. 5

6

Positive correlation between expression levels of EGR-1 and -cell compensatory 7

genes in human pancreatic tissues. To investigate the involvement of EGR-1 in human 8

pancreatic -cell compensatory responses, we collected 58 human pancreas samples and 9

divided them into type 2 diabetic and non-diabetic groups (Figure 8A). Interestingly, we 10

found that EGR1 expression correlated with the expression of its downstream gene PDX1 11

and -cell compensatory genes, including CCND1 (cyclin D1 for -cell proliferation), 12

INS (insulin), and GCK (glucokinase for nutrition-secretion coupling) in the non-diabetic 13

group (Figure 8A, upper panels). However, these correlations were not found in the 14

diabetic group (Figure 8A, lower panels). These results suggest a role of EGR-1 in the 15

regulation of -cell compensation, particularly in non-diabetic subjects. Moreover, we 16

further divided our study population into 2 groups (normal weight, BMI < 23 vs. 17

overweight/obese, BMI ≧ 23) using the Asian criteria of the World Health 18

Organization. Although EGR1 expression correlated with the expression of PDX1, 19

CCND1, INS, and GCK in both groups, the correlation tends to be more significant with 20

higher r-value and smaller p-value in the overweight/obese group than in normal weight 21

group (Figure 8B). 22

23

24

Page 20: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

20

Discussion 1

This study was designed to examine -cell compensatory response under metabolic 2

stress and to test the hypothesis that EGR-1 plays a significant role in this stimulus-3

transcription coupling. Dissecting the regulation of this immediate-early gene in 4

adaptation to metabolic stress is crucial for our understanding of type 2 diabetes 5

development. We found that Egr1−/− mice fed a chow diet displayed normal glucose 6

tolerance and GSIS. Upon metabolic stress, EGR-1 deficiency impaired -cell 7

compensatory responses, as was evidenced by impairments in insulin biosynthesis, -cell 8

mass expansion, and nutrient-secretion coupling. Furthermore, EGR-1 deficiency also 9

downregulated pro-survival pathways and elicited a population of glucagon+ cells 10

carrying β-cell characteristics. These defects were linked to the reductions in a number of 11

transcription factors and signaling cascades essential to maintain the compensatory 12

network and -cell identity. Thus, our study identifies EGR-1 as a regulator of the 13

transcriptional network that compensates for the loss of -cell mass and function. 14

15

EGR-1 has been revealed as an early sensor and regulator in both physiological and 16

pathological conditions [12]. In the absence of external stimuli, EGR-1 expression is 17

scarce. Upon stimulation by extracellular stimuli, the biosynthesis of EGR-1 is rapidly 18

induced. EGR-1 couples extracellular stimulations to long-term responses by changing 19

expression patterns of its target genes. In addition to sensing the hostile environment, 20

EGR-1 has also been shown to be a glucose-responsive gene in cells. The induction of 21

EGR-1 expression by glucose appears to correlate with insulin secretion [39]. EGR-1 22

expression is implicated in proliferation and transactivation of insulin gene in cells 23

[40]. Moreover, the attenuation of EGR-1 expression likely contributed to -cell 24

Page 21: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

21

dysfunction in diabetic rats [14]. These observations are consistent with our findings that 1

HF-fed Egr1−/− mice had reduced islet size, insulin content, and GSIS, as well as 2

impaired glucose homeostasis. These data suggest that EGR-1 plays important roles in 3

controlling proliferation, insulin biosynthesis, and glucose sensing of pancreatic cells in 4

HF-fed mice. To test whether EGR-1 serves as a candidate for clinical application, we 5

overexpressed EGR-1 in the HF-fed mice by hydrodynamics-based gene delivery. Our 6

results showed that of EGR-1 overexpression in the HF-fed mice increased the second 7

phase of GSIS. Thus, the induction of the early sensor EGR-1 may be a therapeutic 8

strategy for targeting pancreatic islet failure. 9

10

The specialized features of cells are determined by the expression of a subset of 11

genes controlled by several transcription factors. Many members of the hepatocyte 12

nuclear factor (HNF) family, such as HNF1A, HNF1B, HNF3A, HNF3B, and HNF4A, 13

are important in regulating early development of pancreas [11, 36]. Although some HNFs 14

are not predicted to contain an EGR-1 binding site, EGR-1 deficiency caused a general 15

reduction in the levels of these HNFs. Cell lineage is determined by feed-forward and 16

autocrine control of transcription factor gene expression [41]. It is likely that EGR-1 17

deficiency affects expression of HNFs through the feed-forward mechanism. After 18

pancreas formation, expression of NEUROG3 and NEUROD1 specifies endocrine 19

precursor cell formation. At the later stage, another set of transcriptional factors, 20

including NKX2.2, NKX6.1, MAFA, and PDX1, drive islet cell differentiation. These 21

transcription factors are critical for the expression of genes in a mature cell that enable 22

its specialized function, i.e. the expression, processing, storage, and regulated secretion of 23

insulin. These genes generally harbor binding sites for PDX1, NEUROD1, MAFA, but 24

Page 22: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

22

we also found that most of them contain EGR-1 binding sites (Figure 7A). Moreover, 1

EGR-1 deficiency caused a general reduction in expression levels of these transcriptional 2

factors when mice were placed in the condition of metabolic overload. These 3

transcriptional factors are crucial for the maintenance of -cell phenotype, and EGR-1 4

can be induced under special circumstances to coordinate expression of these 5

transcriptional factors, allowing β cells to respond to stressful conditions. 6

7

Many studies have identified external stimuli and intrinsic transcriptional networks 8

that are crucial for the generation of functional insulin-producing cells in vitro and in 9

vivo [42, 43]. Several transcriptional factors that are essential for pancreatic development 10

during embryogenesis also regulate the maintenance of -cell function in the adulthood 11

[44]. This implies that transcriptional networks that are necessary for the initial 12

specification of cells may be re-expressed in adult cells to maintain their identity and 13

function in a hostile environment. However, the role of EGR-1 in the regulation of -cell 14

identity has not been addressed. In this study, increased numbers of glucagon+, pancreatic 15

polypeptide+, and somatostatin+ cells were found in the islets of Egr1−/− mice. However, 16

we did not detect Ki67-positive PP- and δ-cells, suggesting that these increases are not 17

likely caused by increased proliferation of PP- and δ-cells. Thus, increased number of 18

other endocrine cell types in the islets of Egr1−/− mice might result from the conversion 19

of β cells. Another possibility is that the decrease of β cells in Egr1−/− mice may activate 20

some other endocrine cell types to acquire β-like cell characteristics. Nevertheless, the 21

origin of these non β cells with β-cell characteristics will await advanced lineage-tracing 22

experiments. These findings suggest that EGR-1 is a critical factor that turns on 23

transcriptional networks for the maintenance of -cell identity. This mechanism may be 24

Page 23: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

23

important during the development of pancreatic islet failure. 1

2

A previous study by Müller et al. showed reduced insulin biosynthesis and islet size 3

in transgenic mice expressing a dominant-negative mutant of EGR-1 in pancreatic β-cells 4

[19]. The reported model is based on a transgenic system impairing the biological 5

functions of the entire EGR family. When keeping these mice with a normal diet, they 6

already showed the evidence of reduced β-cell function. In contrast, a similar phenotype 7

occurs in the Egr1−/− mice with a high-fat diet. We then evaluated the levels of EGR-2 8

and EGR-3 in the islets of Egr1−/− mice. Our results showed that mRNA levels of EGR-2 9

and EGR-3 tended to be higher in Egr1−/− mice, but their levels were extremely low in the 10

islets (Egr2/Egr1= 0.0045 and Egr3/Egr1= 0.0002; Figure S8), suggesting a relatively 11

minor impact of EGR-2 and EGR-3 in the islets. Nevertheless, compensation of EGR-1 12

loss by homologous proteins of the EGR family cannot be excluded. However, we found 13

the discordance between two models. For instance, the expression of glucagon and 14

somatostatin and the proliferation of β-cells did not differ between their mutant and 15

control mice, which are likely due to low proliferation and turnover in the basal chow-fed 16

condition. In contrast, our study applied Egr1−/− mice, which specifically address the 17

physiological consequence of EGR-1 deficiency during metabolic stress. In addition, the 18

natural high-caloric diet feeding course allows us to unravel the possible transcription 19

network and signaling cascade affected by EGR-1 deficiency under metabolic stress. 20

Particularly, we found that WT mice fed with HF diet still retained β-cell identity. 21

However, Egr1−/− mice fed with HF diet lost this clear identification between α and β 22

cells. Therefore, EGR-1 deficiency results in a loss of cell identity between α and β cells. 23

Understanding how a β cell maintains its identity through EGR-1-mediated transcription 24

Page 24: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

24

network would help find a therapeutic strategy for targeting pancreatic islet failure. 1

2

It has been reported that EGR-1 could affect the insulin sensitivity and metabolism 3

in adipose tissue, liver, and skeletal muscle. For example, Yu et al. showed that EGR-1 4

decreases adipocyte insulin sensitivity by blocking PI3K/AKT and augmenting 5

ERK/MAPK signaling [29]. Inhibition of EGR-1 activity by injecting an adenovirus 6

carrying dominant-negative EGR-1 into epididymal fat of hyperphagic db/db mice 7

improved their sensitivity to systemic insulin. Chronic exposure to insulin causes 8

persistent ERK/MAPK activity in adipocytes and enhances insulin resistance in an EGR-9

1-dependent manner [45]. Moreover, deletion of EGR-1 in the liver and skeletal muscle 10

improves systemic insulin sensitivity [46, 47]. Consistently, our data and published 11

results of other groups showed that Egr1−/− mice were protected from diet-induced insulin 12

resistance [30]. Because the assessment of insulin secretion and -cell function could be 13

affected by the peripheral insulin sensitivity, the role of EGR-1 in regulating -cell mass 14

and function should be carefully interpreted and would require either a β-cell specific 15

EGR-1 knockout or the transplantation of wild-type β-cells into the whole body knockout 16

model. Nevertheless, we believe that providing the information regarding how a β cell 17

maintains its identity through EGR-1-mediated transcription network to the field is a 18

timely issue. 19

20

Although these results suggest that EGR-1 knockout affects β-cell function, it is not 21

clear whether this occurs through a cell-intrinsic or cell-extrinsic mechanism. It should be 22

noted that because we employed a whole-body knockout system, it is possible that the 23

role of EGR-1 in pancreatic β cells was masked by metabolic adaptation to EGR-1 24

Page 25: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

25

deficiency in other relevant tissues. Therefore, the decreased expression of potential 1

EGR-1 targets in islets does not rule out the possibility that EGR-1 deficiency in other 2

cell types produces cell-extrinsic effects on β-cell expression of these genes. To 3

strengthen a cell-intrinsic role of EGR-1 in β-cells, we conducted a series of experiments. 4

Firstly, the decreased percentage of cells and increased percentages of , PP, and cells 5

in the islets were observed in RC–fed Egr1−/− mice, and even in the neonatal Egr1−/− 6

mice. These results suggest that the effect of EGR-1 knockout in β-cells is observed when 7

the influence by peripheral tissues is relatively minimal. Secondly, EGR-1 knockdown 8

decreased expression of a number of β-cell transcriptional factors, whereas it increased α-9

cell transcriptional factor in in vitro cell model. Thirdly, we further demonstrated the 10

binding of EGR-1 to the promoter region of potential target genes Pdx1 and Arx in MIN6 11

mouse pancreatic β cells. In summary, these results suggest that EGR-1 affects β-cell 12

function in a cell-intrinsic mechanism. Nevertheless, we believe that providing the 13

information regarding how β cell maintains its identity through EGR-1-mediated 14

transcription network to the field is a timely issue. 15

16

To examine the clinical significance of EGR-1 in -cell compensatory response, we 17

collected normal pancreatic tissues from 58 human subjects and divided them into 18

diabetic and non-diabetic groups. It has been suggested that -cell failure occurs after an 19

initial compensatory state of -cell expansion in type 2 diabetes. We then hypothesized 20

that the compensatory response would remain intact in non-diabetic subjects, but would 21

be impaired in diabetic subjects. Indeed, we found that EGR-1 expression positively 22

correlated with that of cyclin D1, insulin, glucokinase in non-diabetic subjects that 23

contained a spectrum of normal and pre-diabetic conditions. However, these correlations 24

Page 26: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

26

were absent in diabetic subjects. These results suggest that EGR-1 is able to execute its 1

role in governing the transcriptional network only when the compensatory machinery is 2

intact. 3

4

Interestingly, there seems to exist a discordance between the effects of EGR-1 on β 5

cells in mouse versus human islets. In mice, EGR-1 seems to have an effect on β-cell 6

transcription only after a metabolic challenge (HF-diet). However, in humans, the effect 7

of EGR-1 is observed in unchallenged healthy individuals and disappears in diabetic 8

subjects. This discordance may stem from the difference in the experimental design and a 9

physiological difference between mice and humans. For instance, in humans, we mainly 10

focused on the correlations between EGR-1 and compensatory genes. In mice, we 11

primarily tested the effect of EGR-1 loss on β-cell function and compensatory machinery. 12

We suggest that the regulation of compensatory machinery by EGR-1 may take place in 13

individuals with preserved β-cell function, but dissipate in those with impaired β-cell 14

function. Because the HF-diet feeding in mice is more correlated with obesity, we then 15

divided our subjects into normal weight and overweight/obese groups. The correlations of 16

EGR1 with PDX1, CCND1, INS, and GCK are stronger in the overweight/obese group 17

than in normal weight group (Figure 8B). These results suggest that, in humans, EGR-1 18

tends to have a stronger effect on β-cell transcription after a metabolic challenge. 19

20

In summary, our study demonstrated that the loss of EGR-1 uncouples metabolic 21

stress from the transcriptional cascade essential for -cell compensatory response and 22

identity. Thus, the early mediator EGR-1 is crucial for long-term adaptive effects during 23

the compensatory response in cells. We also identified EGR-1 as an important 24

Page 27: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

27

transcription factor that promotes -cell identity, maintains -cell characteristics, and 1

represses non--cell programs. These effects are likely mediated through a series of 2

transcriptional factors maintaining -cell identity during metabolic stress. Understanding 3

the molecular mechanism by which EGR-1 allows cells retain the stable identity will be 4

important for deriving fully differentiated, functional cells to meet the metabolic 5

demand. Furthermore, our study underscores the early stress coupler EGR-1 as a critical 6

factor in the pathogenesis of pancreatic islet failure. 7

8

Page 28: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

28

Abbreviations 1

EGR-1: early growth response-1; HF: high-fat; WT: wild-type; RC: regular chow; OGTT: 2

oral glucose tolerance test; GSIS: glucose-stimulated insulin secretion; ASIS: arginine-3

stimulated insulin secretion; PI3K: phosphoinositide 3-kinases; AKT: serine/threonine-4

specific protein kinase; MEK: mitogen-activated protein kinase kinase; ERK: 5

extracellular signal-regulated kinases; INS: insulin; GCK: glucokinase; HNF: hepatocyte 6

nuclear factor; IR: insulin resistance. 7

8

Acknowledgments 9

We thank Dr. Sheng-Hsiang Lin and Dr. Michael W. Hughes at the Institute of 10

Clinical Medicine, National Cheng Kung University for critical suggestions; Shun-Yun 11

Hou, Ya-Han Lin, and Chih-Yu Lin at the Institute of Clinical Medicine, National Cheng 12

Kung University and Dr. Chia-Ning Shen at Genomic Research Center, Academia Sinica 13

for technical assistance; and Editage for language editing. Y.S.T is the guarantor of this 14

work and, as such, had full access to all the data in the study and takes responsibility for 15

the integrity of the data and the accuracy of the data analysis. This work was supported 16

by grants from Ministry of Science and Technology (MOST-107-2320-B-006-063MY3 17

and MOST-107-2320-B-006-003), National Health Research Institutes (NHRI-EX104-18

10231SI), and National Cheng Kung University- E-DA Hospital Research Project. This 19

funding source had no role in the design of this study, analyses and interpretation of the 20

data, or decision to submit results. 21

22

23

24

Page 29: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

29

Author contributions 1

The author contribution is listed below: S.Y.L. researched data, contributed to 2

discussion, wrote manuscript. L.H.K. researched data, contributed to discussion, wrote 3

manuscript. W.T.W. researched data, contributed to discussion, wrote manuscript. I.C.L. 4

researched data, contributed to discussion, wrote manuscript. C.C.Y. researched data, 5

contributed to discussion. T.T.H. researched data. Y.N.C. collected biopsy, researched 6

data. P.H.C. researched data, wrote manuscript. L.C.H. contributed to discussion. S.H.C. 7

contributed to experimental design and discussion. Y.S.S. collected biopsy, contributed to 8

discussion. Y.W.C. contributed to experimental design and discussion. P.C.C. contributed 9

to experimental design and discussion. P.J.T. contributed to discussion, reviewed/edited 10

manuscript. J.M.S. contributed to discussion, reviewed/edited manuscript. Y.S.T. 11

contributed to discussion, wrote manuscript, reviewed/edited manuscript. 12

13

Competing interests 14

None. 15

16

17

Page 30: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

30

References 1

1. Prentki M, Nolan CJ. Islet beta cell failure in type 2 diabetes. J Clin Invest. 2006; 2

116: 1802–12. 3

2. Talchai C, Xuan S, Lin HV, Sussel L, Accili D. Pancreatic beta cell dedifferentiation 4

as a mechanism of diabetic beta cell failure. Cell. 2012; 150: 1223–34. 5

3. Spijker HS, Song H, Ellenbroek JH, Roefs MM, Engelse MA, Bos E, et al. Loss of 6

beta-Cell identity occurs in type 2 diabetes and is associated with islet amyloid deposits. 7

Diabetes. 2015; 64: 2928-38. 8

4. Katsumoto K, Kume S. The role of CXCL12-CXCR4 signaling pathway in 9

pancreatic development. Theranostics. 2013; 3: 11-17. 10

5. Guo L, Inada A, Aguayo-Mazzucato C, Hollister-Lock J, Fujitani Y, Weir GC, et al. 11

PDX1 in ducts is not required for postnatal formation of beta-cells but is necessary for 12

their subsequent maturation. Diabetes. 2013; 62: 3459-68. 13

6. Rutter GA, Pullen TJ, Hodson DJ, Martinez-Sanchez A. Pancreatic beta-cell identity, 14

glucose sensing and the control of insulin secretion. Biochem J. 2015; 466: 203-18. 15

7. Friedman-Mazursky O, Elkon R, Efrat S. Redifferentiation of expanded human islet 16

beta cells by inhibition of ARX. Sci Rep. 2016; 6: 20698. 17

8. Wang Z, York NW, Nichols CG, Remedi MS. Pancreatic beta cell dedifferentiation 18

in diabetes and redifferentiation following insulin therapy. Cell Metab. 2014; 19: 872–82. 19

9. Cinti F, Bouchi R, Kim-Muller JY, Ohmura Y, Sandoval PR, Masini M, et al. 20

Evidence of beta-Cell Dedifferentiation in Human Type 2 Diabetes. J Clin Endocrinol 21

Metab. 2016; 101: 1044-54. 22

10. Brereton MF, Iberl M, Shimomura K, Zhang Q, Adriaenssens AE, Proks P, et al. 23

Reversible changes in pancreatic islet structure and function produced by elevated blood 24

glucose. Nat Commun. 2014; 5: 4639. 25

11. Bernardo AS, Hay CW, Docherty K. Pancreatic transcription factors and their role in 26

the birth, life and survival of the pancreatic beta cell. Mol Cell Endocrinol. 2008; 294: 1–27

9. 28

12. Thiel G, Cibelli G. Regulation of life and death by the zinc finger transcription 29

factor Egr-1. J Cell Physiol. 2002; 193: 287-92. 30

13. Cheong MW, Kuo LH, Cheng YN, Tsai PJ, Ho LC, Tai HC, et al. Loss of Egr-1 31

sensitizes pancreatic beta-cells to palmitate-induced ER stress and apoptosis. J Mol Med 32

(Berl). 2015; 93: 807-18. 33

14. Garnett KE, Chapman P, Chambers JA, Waddell ID, Boam DS. Differential gene 34

expression between Zucker Fatty rats and Zucker Diabetic Fatty rats: a potential role for 35

Page 31: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

31

the immediate-early gene Egr-1 in regulation of beta cell proliferation. J Mol Endocrinol. 1

2005; 35: 13-25. 2

15. Josefsen K, Sorensen LR, Buschard K, Birkenbach M. Glucose induces early growth 3

response gene (Egr-1) expression in pancreatic beta cells. Diabetologia. 1999; 42: 195-4

203. 5

16. Frodin M, Sekine N, Roche E, Filloux C, Prentki M, Wollheim CB, et al. Glucose, 6

other secretagogues, and nerve growth factor stimulate mitogen-activated protein kinase 7

in the insulin-secreting beta-cell line, INS-1. J Biol Chem. 1995; 270: 7882-9. 8

17. Eto K, Kaur V, Thomas MK. Regulation of insulin gene transcription by the 9

immediate–early growth response gene Egr–1. Endocrinology. 2006; 147: 2923–35. 10

18. Ashcroft FM, Rorsman P. Diabetes mellitus and the beta cell: the last ten years. Cell. 11

2012; 148: 1160–71. 12

19. Muller I, Rossler OG, Wittig C, Menger MD, Thiel G. Critical role of Egr 13

transcription factors in regulating insulin biosynthesis, blood glucose homeostasis, and 14

islet size. Endocrinology. 2012; 153: 3040–53. 15

20. Lee SL, Tourtellotte LC, Wesselschmidt RL, Milbrandt J. Growth and differentiation 16

proceeds normally in cells deficient in the immediate early gene NGFI-A. J Biol Chem. 17

1995; 270: 9971-7. 18

21. Kuo LH, Tsai PJ, Jiang MJ, Chuang YL, Yu L, Lai KT, et al. Toll-like receptor 2 19

deficiency improves insulin sensitivity and hepatic insulin signalling in the mouse. 20

Diabetologia. 2011; 54: 168-79. 21

22. Tsai YS, Kim HJ, Takahashi N, Kim HS, Hagaman JR, Kim JK, et al. Hypertension 22

and abnormal fat distribution but not insulin resistance in mice with P465L PPARgamma. 23

J Clin Invest. 2004; 114: 240–9. 24

23. Xiao X, Gaffar I, Guo P, Wiersch J, Fischbach S, Peirish L, et al. M2 macrophages 25

promote beta-cell proliferation by up-regulation of SMAD7. Proc Natl Acad Sci U S A. 26

2014; 111: E1211-20. 27

24. Li DS, Yuan YH, Tu HJ, Liang QL, Dai LJ. A protocol for islet isolation from mouse 28

pancreas. Nat Protoc. 2009; 4: 1649–52. 29

25. Giulianelli S, Vaque JP, Soldati R, Wargon V, Vanzulli SI, Martins R, et al. Estrogen 30

receptor alpha mediates progestin-induced mammary tumor growth by interacting with 31

progesterone receptors at the cyclin D1/MYC promoters. Cancer Res. 2012; 72: 2416-27. 32

26. Dunn KW, Kamocka MM, McDonald JH. A practical guide to evaluating 33

colocalization in biological microscopy. Am J Physiol Cell Physiol. 2011; 300: C723-42. 34

27. Lin SC, Chien CW, Lee JC, Yeh YC, Hsu KF, Lai YY, et al. Suppression of dual-35

specificity phosphatase-2 by hypoxia increases chemoresistance and malignancy in 36

Page 32: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

32

human cancer cells. J Clin Invest. 2011; 121: 1905-16. 1

28. Ka SM, Hsieh TT, Lin SH, Yang SS, Wu CC, Sytwu HK, et al. Decoy receptor 3 2

inhibits renal mononuclear leukocyte infiltration and apoptosis and prevents progression 3

of IgA nephropathy in mice. Am J Physiol Renal Physiol. 2011; 301: F1218-30. 4

29. Yu X, Shen N, Zhang ML, Pan FY, Wang C, Jia WP, et al. Egr-1 decreases adipocyte 5

insulin sensitivity by tilting PI3K/Akt and MAPK signal balance in mice. EMBO J. 2011; 6

30: 3754-65. 7

30. Zhang J, Zhang Y, Sun T, Guo F, Huang S, Chandalia M, et al. Dietary obesity-8

induced Egr-1 in adipocytes facilitates energy storage via suppression of FOXC2. Sci 9

Rep. 2013; 3: 1476. 10

31. Orci L, Ravazzola M, Amherdt M, Perrelet A, Powell SK, Quinn DL, et al. The 11

trans-most cisternae of the Golgi complex: a compartment for sorting of secretory and 12

plasma membrane proteins. Cell. 1987; 51: 1039-51. 13

32. Daval M, Gurlo T, Costes S, Huang CJ, Butler PC. Cyclin-dependent kinase 5 14

promotes pancreatic beta-cell survival via Fak-Akt signaling pathways. Diabetes. 2011; 15

60: 1186-97. 16

33. Fatrai S, Elghazi L, Balcazar N, Cras-Meneur C, Krits I, Kiyokawa H, et al. Akt 17

induces beta-cell proliferation by regulating cyclin D1, cyclin D2, and p21 levels and 18

cyclin-dependent kinase-4 activity. Diabetes. 2006; 55: 318-25. 19

34. McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, Chang F, et al. 20

Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug 21

resistance. Biochim Biophys Acta. 2007; 1773: 1263–84. 22

35. Pagliuca FW, Melton DA. How to make a functional beta-cell. Development. 2013; 23

140: 2472-83. 24

36. Maestro MA, Cardalda C, Boj SF, Luco RF, Servitja JM, Ferrer J. Distinct roles of 25

HNF1beta, HNF1alpha, and HNF4alpha in regulating pancreas development, beta-cell 26

function and growth. Endocr Dev. 2007; 12: 33-45. 27

37. Murtaugh LC. Pancreas and beta-cell development: from the actual to the possible. 28

Development. 2007; 134: 427-38. 29

38. Kaneto H, Miyatsuka T, Kawamori D, Yamamoto K, Kato K, Shiraiwa T, et al. 30

PDX-1 and MafA play a crucial role in pancreatic beta-cell differentiation and 31

maintenance of mature beta-cell function. Endocr J. 2008; 55: 235-52. 32

39. Chang SY, Cho JM, Kim DB, Jang HJ, Ko SH, Jo YH, et al. Molecular mechanisms 33

of early growth response protein-1 (EGR-1) expression by quercetin in INS-1 beta-cells. J 34

Cell Biochem. 2012; 113: 1559-68. 35

40. Eto K, Kaur V, Thomas MK. Regulation of insulin gene transcription by the 36

Page 33: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

33

immediate-early growth response gene Egr-1. Endocrinology. 2006; 147: 2923-35. 1

41. Sasse SK, Gerber AN. Feed-forward transcriptional programming by nuclear 2

receptors: regulatory principles and therapeutic implications. Pharmacol Ther. 2015; 145: 3

85-91. 4

42. Gutierrez GD, Bender AS, Cirulli V, Mastracci TL, Kelly SM, Tsirigos A, et al. 5

Pancreatic beta cell identity requires continual repression of non-beta cell programs. J 6

Clin Invest. 2017; 127: 244-59. 7

43. Li X, Jia L, Chen X, Dong Y, Ren X, Dong Y, et al. Islet alpha-cell inflammation 8

induced by NF-kappaB inducing kinase (NIK) leads to hypoglycemia, pancreatitis, 9

growth retardation, and postnatal death in mice. Theranostics. 2018; 8: 5960-71. 10

44. Taylor BL, Liu FF, Sander M. Nkx6.1 is essential for maintaining the functional 11

state of pancreatic beta cells. Cell Rep. 2013; 4: 1262–75. 12

45. Shen N, Yu X, Pan FY, Gao X, Xue B, Li CJ. An early response transcription factor, 13

Egr-1, enhances insulin resistance in type 2 diabetes with chronic hyperinsulinism. J Biol 14

Chem. 2011; 286: 14508-15. 15

46. Wu J, Tao W, Bu D, Zhao Y, Zhang T, Chong D, et al. Egr-1 transcriptionally 16

activates protein phosphatase PTP1B to facilitate hyperinsulinemia-induced insulin 17

resistance in the liver in type 2 diabetes. FEBS Lett. 2019; 593: 3054-63. 18

47. Wu J, Tao WW, Chong DY, Lai SS, Wang C, Liu Q, et al. Early growth response-1 19

negative feedback regulates skeletal muscle postprandial insulin sensitivity via activating 20

Ptp1b transcription. FASEB J. 2018; 32: 4370-9. 21

22

23

24

Page 34: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

34

Legends to figures 1

Figure 1. Glucose intolerance and reduced insulin levels in Egr1−/− mice. A: Body 2

weights of male mice fed RC at 2-month-old and a HF diet for 2 months starting 3

at 2 months of age. Numbers of mice are inside bars. B: Organ weight ratios in 4-4

month-old male mice fed RC (upper panel) and a HF diet (lower panel) for 2 5

months. Gon, gonadal; Ing, inguinal WAT, white adipose tissue; BAT, brown 6

adipose tissue. C and D: Plasma (C) glucose and (D) insulin levels during OGTT 7

in 4-month-old male mice fed RC (upper panels) and a HF diet (lower panels) for 8

2 months. WT RC, n=6; Egr1−/− RC, n=11; WT HF, n=16; Egr1−/− HF, n=15. 9

*P<0.05, **P<0.01 and ***P<0.001 compared to WT. 10

11

Figure 2. Failure to increase islet size and insulin content in response to HF 12

feeding in Egr1−/− mice. A: Morphology of pancreas from 5-month-old male mice 13

fed RC and a HF for 3 months. Scale bar: 200 μm. B: Pancreas weight (g), C: 14

mean islet size (μm2), D: endocrine mass (%) were measured in sections from 15

pancreas of RC- and HF-fed mice. E and F: Insulin protein levels in the pancreas 16

lysate determined by (E) ELISA or (F) immunoblot. The relative intensities of the 17

bands by densitometric quantification to WT are indicated. G: 18

Immunofluorescence images (Insulin, red; Nuclei, blue) in the pancreatic tissue 19

from 5-month-old male mice fed RC and a HF diet for 3 months. (Scale bars, 50 20

μm). H: β-cell mass (mg) in sections from pancreas of 5-month-old male mice fed 21

a HF diet for 3 months. I: Fasting plasma C-peptide levels from 4-month-old male 22

mice fed RC and a HF diet for 2 months. Numbers of mice are inside bars. J: 23

Representative transmission electron microscopic images of pancreatic β-cell of 24

Page 35: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

35

HF-fed WT and Egr1−/− mice. Scale bar, 2 μm. Photomicrograph at upper right 1

illustrates mature granules (asterisk) and immature granules (arrowhead) at a 2

higher magnification. K: Percentage of mature granules. Averages of granule 3

percentages inside bars were from 8 fields for each genotype (40 μm2 for each 4

field). *P<0.05, **P<0.01, and ***P<0.001. 5

6

Figure 3. Impaired insulin secretory response to glucose challenge in Egr1−/− 7

mice after HF feeding. A: Expression of genes for glucose sensing and secretion 8

coupling in isolated islets of 5-month-old male mice fed RC (upper panel) and a 9

HF diet (lower panel) for 3 months. B: In vivo glucose-stimulated insulin secretion 10

(GSIS) and C: arginine-stimulated insulin secretion (ASIS) in 4-month-old male 11

mice fed RC (upper panel) or a HF diet (lower panel) for 2 months after 12

intraperitoneal challenge of glucose (3 g/kg body weight) and arginine (0.3 g/kg 13

body weight), respectively. D: Ex vivo GSIS in isolated pancreatic islets from 14

male mice fed RC (upper panel) or a HF diet (lower panel). Numbers of mice are 15

inside bars. *P<0.05 and **P<0.01. 16

17

Figure 4. Egr1 deficiency exhibits reduced proliferation and neogenesis in the 18

islet upon HF feeding. A: Immunofluorescence staining for Ki67 (green) and B: 19

quantification of Ki67+ cells in the pancreas of 5-month-old male WT (n=4) and 20

Egr1−/− (n=4) mice fed a HF diet for 3 months. Numbers inside bars are 21

accumulated percentages of different categories of Ki67+ cell number per islet. C: 22

Quantification of Ki67-positive insulin-positive cells. Scale bar: 50 μm. D: 23

Immunofluorescence staining for cleaved caspase-3 (green) in the pancreatic 24

Page 36: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

36

islets. Images are co-stained for insulin (red) and nuclei (blue). Scale bar: 200 1

μm. E: Quantification of caspases-3-positive insulin-positive cells. F: mRNA 2

levels of proliferation markers in the isolated islets of HF-fed Egr1−/− (n=10) mice 3

relative to WT (n=6) mice. G: Immunoblot analysis on proliferation-related 4

markers and signaling molecules. The relative intensities of the bands by 5

densitometric quantification to WT are indicated. *P<0.05 and ***P<0.001. 6

7

Figure 5. Loss of β-cell identity in the islets of HF-fed Egr1−/− mice. Confocal 8

images of co-staining for the markers of β-cell (insulin) in red with (A) α-cell 9

(glucagon), (B) PP-cell (pancreatic polypeptide, PPY) or (C) δ-cell (somatostatin, 10

SST) in green in the pancreas of 5-month-old male Egr1−/− and WT mice fed a 11

HF diet for 3 months. The enlarged images highlight the representative co-12

localization with 5 magnification from white squares in the overlay images. The 13

co-localization coefficient is presented as the product of the differences from the 14

mean (PDM) image. Yellow or blue color pixels indicate co-localization or 15

segregation, respectively. D: Quantification of the markers for β-, α-, PP-, and δ-16

cells in the pancreas of HF-fed Egr1−/− and WT mice. E: Pearson’s coefficient (Rr) 17

of the co-localization between insulin and other hormones in the pancreas of HF-18

fed Egr1−/− and WT mice. The result ranges between -1 (perfect negative 19

correlation) to +1 (perfect positive correlation) with 0 meaning no correlation. F: 20

Immunofluorescence staining of the transcription factors critical for β-cell identity. 21

Images are co-stained for nuclei (blue). ***P<0.001. 22

23

Figure 6. Glucagon-positive α cells expressed typical -cell transcription factors 24

Page 37: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

37

in the islets of HF-fed Egr1−/− mice. Confocal images of co-staining for the 1

markers of β-cell (insulin; white) and α-cell (glucagon; green) with (A) PDX1, (B) 2

PAX4, (C) PAX6, and (D) ARX (red) in the pancreas of HF-fed Egr1−/− and WT 3

mice. The enlarged images highlight the representative co-localization with 5 4

magnification from white squares in the overlay images. The fluorescence 5

intensity profile from green, red, and white channels is shown. 6

7

Figure 7. Decreased transcriptional network in the islets of HF-fed Egr1−/− mice. 8

A: Bioinformatics analysis in identification of transcriptional factors and endocrine 9

molecules that contain potential EGR-1 binding site. Sequences for proteins 10

involved in the development program of islet lineages were analyzed for potential 11

EGR-1 binding site using the PROMO transcription factor binding site database. 12

B: Expression of genes for pancreatic development, endocrine lineage 13

specification and β-cell identity in the isolated islets of HF-fed Egr1−/− (n=10) 14

relative to WT (n=6) mice. *P<0.05 and **P<0.01 compared to WT mice. Pathway 15

analysis of the response of HF feeding on the islets of (C) WT and (D) Egr1−/− 16

mice using Ingenuity Pathways Analysis (IPA) software. Pathway analysis of the 17

effect of EGR-1 deficiency on the transcription network in (E) RC and (F) HF diet 18

feeding. Color shading corresponds to the type of dysregulation: red for up-19

regulated and green for down-regulated genes according to the results of 20

quantitative PCR. The shape of the node indicates the major function of the 21

protein: transcription regulators (dumbbell); kinases (three arrows); and others 22

(circle). G: Expression of genes in EGR-1 knockdowned (n=5) relative to control 23

(n=5) MIN6 cells. H: ChIP assay in EGR-1 overexpressed and control (pCMV) 24

Page 38: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

38

MIN6 cells. Sequences containing the potential EGR-1 binding sites in Pdx1 and 1

Arx were amplified by real-time PCR. n=3 in each group. *P<0.05, **P<0.01, and 2

***P<0.001. 3

4

Figure 8. The correlation between EGR-1 expression and compensatory genes in 5

human pancreatic tissues. Scatter plot illustrating the Spearman’s correlation of 6

normalized reads per patient between EGR1 and PDX1, as well as 7

compensatory genes, such as CCND1 (cyclin D1), INS (insulin), and GCK 8

(glucokinase), in (A) non-diabetic (upper panels) and diabetic (lower panels) 9

subjects; and (B) normal weight (BMI < 23, upper panels) and overweight/obese 10

(BMI ≧ 23, lower panels) subjects. Spearman’s rank correlation coefficients r 11

and P value are provided in each plot. *P<0.05, **P<0.01, and ***P<0.001. 12

13

Page 39: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

Figure 1

Page 40: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

A

B C D E

F G H

I J K

Figure 2

Page 41: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

Figure 3

Page 42: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

A

D

E F

B

G

C

Figure 4

Page 43: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

Figure 5A

E

B

C

D

F

Page 44: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

Figure 6

Page 45: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

Figure 7

G H

FE

A B

C D

Page 46: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin

A

B

Figure 8

Page 47: 1 Loss of EGR-1 uncouples compensatory responses of ...Pancreatic 6 cells undergo proliferation and increase output of secreted insulin. A coordinated 7 increase in -cell mass, insulin