Page 1 of 41 Diabetes

hZnT8 (slc30a8) Transgenic Mice Which Overexpress the R325W Polymorph Have

Reduced Islet Zn2+ and Proinsulin Levels, Increased Glucose Tolerance After a High-Fat

Diet, and Altered Levels of Pancreatic Binding 1

Li Li 2, Shi Bai 2, and Christian T. Sheline 2,3,*

2 Department of Ophthalmology and the Neuroscience Center of Excellence,

Louisiana State University Health Sciences Center, New Orleans, LA 70112

3 Department of Neurology, Stony Brook University Hospital, Stony Brook, NY 11794

1 Supplemental Figures 14, and Supplemental Tables 13 are available as Online Supporting

Material with the online posting of this paper.

RUNNING TITLE: ZnT8 and Diabetes

WORD COUNT; ABSTRACT: 200; WORD COUNT, INTRODUCTION through

DISCUSSION: 3990; NUMBER OF FIGURES: 6; NUMBER OF TABLES: 2;

SUPPLEMENTARY MATERIAL: Online Supporting Materials: 8

AUTHOR LIST FOR INDEXING: Sheline, Bai, Li

* To Whom Correspondence Should be Addressed: Department of Neurology; Stony Brook

Medicine; HSC T12020; Stony Brook, NY 11794; Telephone (631) 4449281, Fax (631) 444

1474; Email – [email protected]

Diabetes Publish Ahead of Print, published online November 29, 2016 Diabetes Page 2 of 41

4 Abbreviations used: GAPDH (glyceraldehyde3phosphate dehydrogenase), HFD (Highfat diet), HFDD (Highfat diet, Zn2+ deficient), IPGTT (Intraperitoneal glucose tolerance testing),

ND (Normal nonpurified diet), RTPCR (Reverse transcriptase polymerase chain reaction), RW

(R325W), SEM (Standard error measurement), T1DM (type1 diabetes), T2DM (type2

2+ 2+ 2+ diabetes), WT (Wildtype), [Zn ]i (intracellular Zn concentration), Zn (Zinc), ZnE (excess

Zn2+ diet), ZnR (reduced Zn2+ diet), ZnT5 (Zinc transporter 5), ZnT8 (), ZP1

(ZinPyr1) Page 3 of 41 Diabetes

Abstract

Zinc is involved in both type1 (T1DM) and type2 (T2DM) diabetes. The wildtype (WT)

form of the betacell specific Zn2+ transporter, ZNT8, is linked to T2DM susceptibility. ZnT8

null mice have a mild phenotype with a slight decrease in glucose tolerance, whereas patients

with the ZnT8 R325W polymorphism (rs13266634) have decreased proinsulin staining and

susceptibility to T2DM. We measured zinc, insulin, and proinsulin stainings, and IPGTT in

transgenic mice overexpressing hZnT8 WT or hZnT8 R325W fed a normal or high fat diet (HFD).

2+ The hZnT8 R325W transgenic line had lower pancreatic [Zn ]i and proinsulin, and higher insulin

and glucose tolerance compared to control littermates after 10 weeks of a HFD in male mice.

The converse was true for the hZnT8 WT transgenic line, and dietary Zn2+ supplementation also

induced glucose intolerance. Finally, pancreatic zinc binding proteins were identified by Zn2+

affinity chromatography and proteomics. Increasing pancreatic Zn2+ (hZnT8WT) induced

nucleoside diphosphate kinase B (NDPKB), and Zn2+ reduction (hZnT8RW) induced

carboxypeptidaseA1 (CpA1). These data suggest that pancreatic Zn2+ and proinsulin levels

covary, but are inversely variant with insulin or glucose tolerance in the HFD model of T2DM

suggesting novel therapeutic targets.

Keywords: Zinc transporter 8, rs13266634, pancreas, insulin, proinsulin, type2 diabetes Diabetes Page 4 of 41

Introduction

Zinc (Zn2+) is involved in the diabetic process, but little is known about its role, or homeostatic mechanisms within the pancreas. In animal models of type1 diabetes (T1DM), Zn2+ chelators, compounds that prevent Zn2+ toxicity, knockout of Zn2+ transporter 5 (ZnT5), and a chronic Zn2+ reduced diet attenuated diabetic incidence and mortality (13). However, other studies have suggested that acute Zn2+ supplementation could be beneficial (reviewed in (4)).

The wildtype form of ZnT8 was linked to susceptibility for type2 diabetes (T2DM), whereas the

ZnT8 R325W polymorphism (rs13266634, or RW) decreases susceptibility as demonstrated by genome wide analyses (5; 6). Patients homozygous for the ZnT8 WT at risk form have increased proinsulin levels (7) which is detrimental for type2 diabetics (8). hZNT8 WT is also an

important autoantigen in adult onset T1DM patients who lack other autoantigens, and the

R325W polymorphism removes one autoantigenic epitope of hZNT8 (9). Finally, ZnT8 null

mice have a mild phenotype with a slight change in granule morphology and a slight decrease in

glucose tolerance (10), suggesting redundancy in the mechanisms for providing Zn2+ required for insulin packaging (11).

An animal model of type2 diabetes is the highfat diet (HFD). Feeding mice a diet from

616 weeks of age in which 60% of their calories derive from fat induces hyperglycemia,

hyperinsulinemia, and glucose intolerance to intraperitoneal glucose tolerance testing (IPGTT) in

a manner similar to type2 diabetics (12). We propose that Zn2+ is transported into the golgi and endoplasmic reticulum of βcells for secretory granule incorporation by Zn2+ transporters 5 and 8.

Knockout of ZnT5 decreases free secretory Zn2+ (3), whereas knockout of ZnT8 decreases both free and some insulin bound Zn2+ inducing a mild reduction in insulin secretion (10). During

chronic inflammation induced by obesity and T2DM, secretory Zn2+ homeostasis is disrupted, Page 5 of 41 Diabetes

leading to Zn2+ mediated potentiation of βcell death, or improper processing and packaging of

insulin. We generated hZnT8 WT and hZnT8 R325W betacell specific transgenic mouse lines,

2+ and characterized them for expression levels, pancreatic [Zn ]i and zinc binding proteins

(ZBP’s), insulin and proinsulin levels, and IPGTT after 10 weeks of a HFD. We tested the

hypothesis that excess pancreatic Zn2+ induced by overexpression of hZnT8 WT in mice would

be detrimental in a model of T2DM, whereas overexpression of hZnT8 R325W in mice would be

beneficial. Diabetes Page 6 of 41

Materials and Methods

Generation of human ZnT8 transgenic Mice, Breeding, and Genotyping. Transgenic rather than knockin mice were undertaken because overexpression was deemed necessary because of the short duration of the experimental plan. pIns1 plasmid (13) containing the human insulin promoter (1.9 kB) fused to the rabbit βglobin intron (same construct used in (14)), was the

expression construct. Human pZnT8WT-EGFP and pZnT8RW-EGFP constructs (15) were used as templates for PCR reactions using the ZnT8 cloning primers (Supplemental Table 1) to introduce appropriate restriction enzyme sites for cloning.

Zn2+ Transporter Expression. Total RNA from harvested mouse pancreas was extracted with 0.5M guanidinium isothiocyanate. For details of the method see (16) and Supplemental

Materials.

Animal Trials and High Fat Diet. All studies were conducted according to the Institutional

Animal Care and Use Committee (LSUHSC, New Orleans), the PHS Guide for the Care and Use

of Laboratory Animals, USDA Regulations, and the AVMA Panel on Euthanasia.

At 6 weeks of age, the animals were randomly divided into 8 groups containing 6 mice in

each group such that researchers were blinded to genotype and diet. Groups 1 and 2 were hZnT8

WT-X40 Tg and Tg+, each fed with normal diet (2019, Harlan) (3), groups 3 and 4 were hZnT8

WT-X40 Tg and Tg+ fed with 60% high fat diet (Harlan TD.06414, Supplemental Table 2), and groups 5 and 6 and groups 7 and 8 were the same but with hZnT8 R325W-T12 transgenic mice. Additional groups of hZnT8 WT-X28, hZnT8 R325W-T8, and hZnT8 R325W-T16 lines

were similarly performed. At 16 weeks of age, IPGTT was performed, and plasma, serum and Page 7 of 41 Diabetes

pancreas were collected for further investigation. In addition, C57/Bl6 mice were fed with

normal diet (ND), High Fat Diet (HFD), and High Fat Diet Zn2+ Deficient (HFDD, 60/Fat, 1 mg

Zn2+/kg diet EWS) (Harlan TD.10872, Supplemental Table 2) plus 1, 60, or 260 mg Zn2+/kg diet

using ZnSO4 drinking water from 616 weeks, IPGTT performed, and tissues collected.

Glucose Tolerance Test and insulin measurements. Glucose tolerance was assessed by IPGTT at

16 weeks of age. Mice were fasted for 56h, weighed, and Dglucose (1.5 mg glucose/g body

weight) was injected intraperitoneally. Glucose in saphenous vein blood was measured at 0, 15,

30, 45, 60, and 120 minutes using a blood glucose monitoring system (One Touch, glucose

oxidase). Insulin was measured in duplicate from plasma using enzymelinked immunosorbent

assays (EMD Millipore, Darmstadt, Germany).

Insulin/proinsulin immunohistochemistry and Zn2+ staining with Zp1. Mice were anesthetized,

plasma and serum were generated, and they were killed 24h after IPGTT. Freshfrozen pancreata

were sectioned and sequentially stained for zinc (5 M Zinpyr1), and antiinsulin, or anti

proinsulin as previously described (3). Islet images were captured at identical exposures using a

Nikon TS100, and SIS software. The adjacent slide was fixed in 2% paraformaldehyde/2%

glutaraldehyde and developed using a monoclonal antibody to proinsulin (G39A8, DSHB, U

Iowa; 1:200), and goat anti mouse Cy3. Islet fluorescence intensity was measured by NIH

software Image J fromsix to eight slides per animal, and 6 animals per group. Identical threshold

intensities were calculated and integrated by area, and compared between each experimental

group.

Diabetes Page 8 of 41

Zinc-agarose chromatography. A 50 mg piece of pancreas from each of 3 animals fed a HFD of

each group (hZnT8WTX40, hZnT8RWT12, and C57/Bl6/J control) were rapidly homogenized

into 2 mls of lysis buffer (50 mM Tris (pH=7.5), 125 mM NaCl, 5% glycerol, 0.4% IGEPAL

CA630, 10 µM MgCl2, 1mM PMSF and fresh 1x protease inhibitor without EDTA), and cleared. The supernatant was dialyzed in lysis buffer, cleared (Input), and incubated for 2 hrs with 200 µL of highaffinity Zn2+agarose beads (Lambda Biotech, Saint Louis, MO) at 4 °C.

The slurry was poured into a minicolumn and the unbound (FT) fraction collected. The column

was washed with lysis buffer, eluted (Eluate) with 3 x 400 µL 500 mM imidazole in lysis buffer;

the beads were boiled in SDS buffer, each fraction was collected, and aliquots saved.

PEI/SDS-PAGE. The input, FT, and eluate proteins from C57/Bl6/J controls were precipitated, and 50 ug of each fraction labeled with Cy5, Cy3, and Cy2 respectively, for Figure 5. Eluates from ZnT8 WT and ZnT8 RW were labeled with Cy2 and Cy3, and the FT from ZnT8 WT was labeled with Cy5 for Figure 6 (17). Samples were first run on isoelectric focusing gel strips (pH

310 nonlinear, 24 cm, GE Healthcare, Piscataway, NJ); equilibrated, and run in the second dimension on 10% SDSPAGE gels. These 2D gels were scanned using a Typhoon 9400

Variable Mode Imager, and spot detection and quantification performed using DeCyder differential analysis software DIA, Version 5.0 (GE Healthcare, Piscataway, NJ, USA). Spots with at least 5fold greater abundance in the elution than the FT, and with sufficient Sypro staining (~2050ng) were excised and trypsin digested using the automated Spot

Handling Workstation from GE Healthcare.

Liquid chromatography-mass spectrometry (LC-MS). The trypsin digested peptides were resuspended in 0.1% formic acid/2% acetonitrile, loaded into a Dionex PepMap C18 trap Page 9 of 41 Diabetes

column, and separated by a New Objective reversed phase C18 Picofrit column/emitter (Woburn,

NJ). 0.1% formic acid aqueous solution and 0.1% formic acid in acetonitrile were used as Buffer

A and Buffer B, respectively. The gradient elution at 250 nL/min starts at 5%, to 25% buffer B in

20 minutes, to 60% buffer B in 10 minutes, to 80% buffer B in 15 minutes plus 10 minutes. A

blank run was inserted between sample runs to reduce cross contamination. All spectra were

acquired on a ThermoFisher LTQXL linear ion trap mass spectrometer (Waltham, MA)

coupling with an Eksigent nanoLC 2D (Dublin, CA).

Statistical Analysis. Values are the mean ± SEM. Differences in RTPCR, ZnT8 levels, and

blood glucose were analyzed by oneway analysis of variance between transgenic positive and

negative animals followed by a Bonferroni test, with P ≤0.05 defined as statistically significant.

For determination of significance of the stained pancreatic sections, we used the

Statistical Analysis Software program to analyze the data by applying the ThreeWay ANOVA

test to determine the effect of three nominal predictor variables on a continuous outcome

variable. Here the three predictor variables are strain (ZnT8R325W vs ZnT8WT), treatment

(HFD vs ND), and transgene (Tg+ Vs Tg). The continuous variable is raw integrated density of

the insulin, Zp1, or proinsulin stained pancreatic sections (n=36), with P ≤0.05 defined as

statistically significant. Diabetes Page 10 of 41

Results

RT-PCR was performed on each of the hZnT8 WT and hZnT8 RW transgenic mouse lines.

Nine hZnT8 WT and nine hZnT8 RW transgenic mouse lines were generated, and total pancreatic

RNA was isolated from each, reverse transcribed, and 25 cycles of PCR performed to determine

relative ZnT8 RNA expression between lines (top panel, Supplemental Figure. 1). GAPDH

expression was used as an internal control (bottom panel, Supplemental Fig. 1). Several high

and low expressing lines were determined.

Mice expressed the hZnT8 transgene predominantly in the pancreas. ZnT8 WT 549+ (X40), and

ZnT8 R325W 8038+ (T12) were chosen as the high expressing lines for further study, and were backcrossed to C57/Bl6/J for 10 generations. Pancreas, thymus, brain, and liver from these lines

were collected and RNA made. RTPCR was performed using 25 cycles, and the products run

on agarose gels as shown in Supplemental Figure 2. The expression was highest in pancreas,

with thymus and brain expressing much lower levels, and no detectable expression in liver as

expected for the insulin promoter (18).

qRT-PCR was performed on lines chosen for high or low expression. Realtime quantitative

PCR was performed using GAPDH as an internal control, and the quantitation is presented in

Supplemental Table 3 for hZnT8, mZIP4, and mZnT8. Lines X40, and T12 were chosen for

further study of high expressing lines for ZnT8 WT and ZnT8 RW, respectively. ZnT8 RW lines

T16, and T8 were chosen for low expressing lines, and ZnT8 WT X28 line was an intermediate

expressor. The expression of mZIP4 was reduced in the X40 pancreas, and increased in the T12 pancreas, whereas mZnT8 was unaffected (Supplemental Table 3). mZIP4 was studied because it

is preferentially expressed in the pancreas, and varies inversely with zinc levels (G. Andrews, Page 11 of 41 Diabetes

personal communication), confirming the ZP1 staining results in these transgenic lines. Protein

levels of hZnT8 could not be determined because the commercially available antibody did not

recognize protein in a Western blot (data not shown). Low levels of an appropriately sized

pancreatic protein were present in a highresolution 520% gradient SDSPAGE gel from

transgenic positive mice, and were not present in transgenic negative mice (data not shown).

Zn2+, proinsulin, and insulin staining were determined for each of the chosen lines on a HFD or

a normal diet. Groups of male mice (n=6 for each transgenic line) were exposed to a HFD, or a

ND from 616 weeks of age. Zinpyr1 (ZP1) staining was performed to determine the levels of

pancreatic, stainable Zn2+. Insulin and proinsulin immunohistochemistry were performed on

adjacent sections after postfixation. hZnT8 RW had significantly decreased Zn2+ and proinsulin

staining and increased insulin staining compared to transgenic negative littermates. hZnT8 WT

had significantly increased Zn2+ and proinsulin staining and decreased insulin staining compared

to transgenic negative littermates (Figures 1 and 2 for HFD and Supplemental Figures 3 and

4 for ND). The threshold integrated density of the staining intensity from all of the

photomicrographs was determined and is summarized in Table 1. There was a significant

difference in Zn2+, insulin, or proinsulin levels in hZnT8 RW fed a HFD compared to those fed a

ND. In hZnT8 WT, there was a significant difference in Zn2+ and insulin levels but not in

proinsulin levels dependent on dietary fat. There was a significant difference in Zn2+, insulin, and

proinsulin levels between hZnT8 RW and hZnT8 WT transgenic positive lines and negative

littermates fed a HFD. There was a significant difference in Zn2+ and insulin, but not in

proinsulin staining between the RW and WT lines fed a ND.

Diabetes Page 12 of 41

Glucose tolerance in male ZnT8 WT, ZnT8RW, and transgenic negative littermates after a HFD

or a ND. IPGTT was performed at 16 weeks of age, and the summarized data is presented in

Figure 3. There was a significant decrease in glucose tolerance for the ZnT8 WT line, X40, in both ND and HFD conditions compared to negative littermates. Conversely, there was a

significant increase in glucose tolerance for the ZnT8 RW line, T12, only in HFD conditions.

Low expressing ZnT8 RW lines had no effect on glucose tolerance. An alternate ZnT8 WT line

with slightly lower expression levels (X28) had a less significant decrease in glucose tolerance

(data not shown).

Plasma insulin levels. Insulin levels were measured (in ng/ml) at 0 min and 30 min (fasting), and

24hrs (fed) after IPGTT. ND control mice had 0.19 ± 0.02*, 0.39 ± 0.05*, and 2.1 ± 0.31*

respectively. HFD control mice had 1.1 ± 0.26, 2.58 ± 0.31, and 6.2 ± 0.55 respectively. HFD

ZnT8 WT mice had 1.02 ± 0.21, 1.7 ± 0.22*, and 5.5 ± 0.5 respectively. HFD ZnT8 RW mice

had 1.15 ± 0.28, 3.25 ± 0.42*, and 6.75 ± 0.61, respectively. HFDD + 260 ppm zinc had 1.0 ±

0.19, 1.65 ± 0.20*, and 5.45 ± 0.48, respectively. * indicates a significant difference from insulin

levels in HFD control mice at the same time point at P<0.05 (n=6).

Increasing Zn2+ through diet decreases glucose tolerance. C57/Bl6/J male mice (same genetic background as ZnT8 transgenic negative mice) were exposed to a HFD, a HFDreduced Zn2+ diet (2 mg Zn2+/kg diet), a HFDnormal Zn2+ diet (61 mg Zn2+/kg diet), and to a HFDexcess

Zn2+ diet (261 mg Zn2+/kg diet) from 616 weeks and compared to normal diet for IPGTT. There was a trend towards an increase in glucose tolerance for the HFDreduced Zn2+ compared to the

HFDnormal Zn2+, with a single time point achieving significance. Conversely, there was a

significant decrease in glucose tolerance for the HFDexcess Zn2+ compared to the HFDnormal Page 13 of 41 Diabetes

Zn2+ (Figure 4). This decreased glucose tolerance for the HFDexcess Zn2+ diet was

accompanied by a significant decrease in insulin levels and staining and an increase in zinc

staining (3) and proinsulin staining (data not shown and above).

Pancreatic Zn2+ binding proteins are secretory, mitochondrial and energy metabolic proteins

and some of their levels are modulated by pancreatic zinc concentrations. Pancreatic extracts

from control mice on HFD were analyzed for ZBP’s using zincagarose affinity chromatography

and proteomics, and proteins were identified (Figure 5 and Table 2). Only soluble proteins

between 10150 kD with intermediate zinc affinity can be identified using this method.

Carboxypeptidase A1 (CpA1) and αamylase bind Zn2+ showing that the technique is working.

Additional proteins identified play roles in mitochondrial and cellular energy metabolism

(succinylCoA: 3ketoacidCoA transferase (SCOT), methyl malonate semialdehyde

dehydrogenase (MMSDH), nucleoside diphosphate kinase A & B (NDPKA&B), and alpha

aminoadipic semialdehyde dehydrogenase). ZBP’s from pancreatic extracts of ZnT8WT versus

ZnT8RW were analyzed by proteomics. Of these identified pancreatic ZBP’s, NDPKB was

significantly increased (>5 fold) in the pancreas of ZnT8 WT (high zinc) transgenic mice as was

actin. CpA1 was significantly increased in the pancreas of ZnT8 RW (low zinc) transgenic mice

(Figure 6).

Discussion

We have generated and characterized transgenic mouse lines that overexpress hZnT8 WT

and hZnT8 RW under the control of the insulin promoter for: 1) expression at the RNA level; 2)

expression at the protein level; 3) organspecific expression; 4) stainable Zn2+ levels; 5) insulin Diabetes Page 14 of 41

and proinsulin levels; 6) IPGTT after ND or HFD; 7) the effects of dietary Zn2+ on HFDinduced

glucose intolerance in C57/Bl6 mice; and 8) ZBP’s and their differential expression.

Overexpression of the hZnT8 WT gene increases pancreatic Zn2+ and proinsulin levels, and decreases insulin and glucose tolerance. Conversely, overexpression of the hZnT8 RW gene decreases pancreatic Zn2+ and proinsulin levels, and increases insulin and glucose tolerance.

Altering dietary Zn2+ has similar effects on glucose tolerance after a HFD. We identified pancreatic ZBP’s and show that CpA1 is induced by low zinc, and NDPKB is induced by

increased zinc.

There are two superfamilies of mammalian Zn2+ transporters (19; 20) and members from each family function in βcells. Solute carrier 39A (Slc39A) family members, named ZIPs, import Zn2+ into cells, and solute carrier 30A (Slc30a) family members, named ZnTs, function in

Zn2+ efflux and transport into vesicular compartments (20). These transport proteins function as multimers (21). ZnT3 knockout mice don’t load Zn2+ in synaptic vesicles and therefore are resistant to neuronal injury after light damage, global ischemia or hypoglycemia (in submission, and (22; 23)). The lethal milk mouse has a mutation in the ZnT4 gene causing toxic Zn2+

deficient milk (24). Deletion of ZnT5 results in poor growth, osteopenia, low body fat, muscle

weakness, and malespecific cardiac death (25). ZNT5 functions as a heterodimer putting Zn2+ into the general secretory pathway. ZNT8 is βcellspecific and overexpression of ZNT8 in INS

1E cells increases cellular Zn2+ content, increases sensitivity to Zn2+ toxicity, and enhances basal

glucosestimulated insulin secretion (26). Thus, ZNT8 functions to sequester Zn2+ into the secretory granules of βcells analogous to ZNT3 in presynaptic secretory granules in the brain.

ZNT8 may also function in heterodimers with ZNT’s 5, 6, and 7 to put Zn2+ into the secretory granules of βcells during their formation in the golgi. Page 15 of 41 Diabetes

A common single nucleotide polymorphism in ZnT8, which changes R325 to W, is

associated with a reduced risk for type2 diabetes (5; 6). This nonconservative substitution of a

charged basic amino acid for a hydrophobic polar amino acid occurs at the predicted transporter

dimer interface in an important modulatory region (10). The R325W protein causes reduced

Zn2+ staining when overexpressed in βcells suggesting that it acts as a dominant negative,

dimerizing with itself or other ZnTs and reducing their transport function. A knockout of ZnT8

demonstrates a small reduction in glucose tolerance, and a change in secretory granule

morphology (10; 27). These results suggest a functional redundancy in secretory Zn2+

transporters, and this redundancy is supported by the recently described importance of ZNT3 for

insulin secretion (28), and ZNT5 in models of type1 diabetes (3).

ZnT8 null mice (global or βcell) have a substantial reduction in Zn2+, but little effect on

glucose tolerance and no effect on insulin (10; 27). However in βcell ZnT8 KO’s, proinsulin

levels are increased while proinsulin transcription, and its transcription factors (pdx1, mafA) and

processing enzyme RNA’s (proconvertases 1/3 and 2, carboxypeptidase E) are decreased (27).

We suspect that as with T1DM, too little or too much Zn2+ are detrimental for T2DM, with a

partial Zn2+ reduction being optimal (3). ZnT8 global or βcell knockouts reduce Zn2+ most,

followed by ZnT8 RW overexpression (dominant negative) and ZnT8 knockdown which have

modestly reduced Zn2+ levels, versus ZnT8 WT overexpression which has high Zn2+ levels.

Varying the pancreatic ZnT8 WT/Zn2+ levels affect insulin staining and may be mediated in part

by effects of Zn2+ to inhibit processing of proinsulin to insulin as demonstrated by proinsulin and

insulin IHC.

Elevated proinsulin levels are associated with increased risk of T2DM in humans (8), and

homozygous carriers of the ZnT8 WT risk allele had elevated proinsulin/insulin ratios in humans Diabetes Page 16 of 41

(7). Overexpression of ZnT8 WT in transgenic mice results in an increase in Zn2+ and proinsulin staining and a decrease in insulin staining and glucose tolerance. hZNT8 RW significantly reduces pancreatic Zn2+ and proinsulin levels, and increases insulin and glucose tolerance

(Figures 3 and 4, and Table 2) which differentiates this mechanism from others that effect insulin

sensitivity. We hypothesize that hZNT8 RW forms hypoactive heterodimers with mZNT8, or perhaps with mZNT5, mZNT6, or mZNT7, which are all expressed in the ER and golgi.

Generally, a HFD demonstrated more substantial variations in Zn2+, insulin or proinsulin

stainings compared to a ND, suggesting an interaction between zinc levels and the effects of

highfat. Reducing Zn2+ in the diet is less effective than the RW transgenic mouse models to

increase glucose tolerance. However, increasing Zn2+ in the diet is at least as effective as the hZNT8 WT transgenic mouse model to decrease glucose tolerance after HFD (Figure 4).

Our data show that the levels of proinsulin and insulin are inversely modulated by Zn2+,

suggesting the conversion of proinsulin to insulin is inhibited by Zn2+. The processing enzymes

which convert proinsulin to insulin (proconvertases 1/2, 3, and carboxypeptidase E) have been

shown to be Ca2+ dependent (29), and therefore Zn2+ levels could affect proinsulin processing by

competition with Ca2+. Alternatively, proinsulin transcription could be Zn2+modulated, but this

should cause covariance of proinsulin and insulin. Proinsulin processing enzyme transcription

could be Zn2+modulated, and cytoplasmic/nuclear Zn2+ activates various kinase cascades impinging on transcription (3032). Also the insulin degrading enzyme proteinase III requires

Zn2+ for activity (33), and membrane associated insulin degrading activity is activated by Zn2+

(34). We postulate that ZNT8 RW mediates a moderate reduction in pancreatic golgi Zn2+ levels,

reducing competition for required Ca2+ binding sites on proinsulin processing enzymes, and

inducing less Zn2+dependent insulin degradation, thereby decreasing proinsulin levels, and

increasing insulin levels and glucose tolerance. Page 17 of 41 Diabetes

To understand this process, we identified pancreatic Zn2+ binding proteins. The role of α

amylase as a ZBP is interesting because it is a major pancreatic protein that requires Zn2+ for

activity, may be secreted in the apo form from αcells, and load with Zn2+ that is released from

βcells (35). This would provide a crosstalk/signaling mechanism between α and βcells, and

suggests a mechanism for excessive Zn2+ excretion in T1 and T2DM (36). Isolating αamylase

and CpA1 provides confidence that the technique is working, and inclusion of excess zinc

prevented purification of these ZBP’s (data not shown). Several of the ZBP’s identified were

energy metabolic enzymes, and/or mitochondrial proteins, confirming a role of energy

metabolism in diabetes. Glutathione Stransferase Mu is one of the ZBP’s identified in the

control pancreas. Another ZBP identified was MMSDH which is mitochondrial in origin and

displays diabetes and enalapril sensitive tyrosine nitration (37). By catalysing the CoA and

NADP+dependent oxidative decarboxylation of malonatesemialdehyde to acetylCoA,

MMSDH provides a nonglycolytic means for producing acetylCoA. Alpha amino adipic

semialdehyde dehydrogenase is involved in the metabolism of lysine. SuccinylCoA: 3ketoacid

CoA transferase is a mitochondrial enzyme that catalyzes the reversible transfer of coenzymeA

from acetoacetylCoA to succinate to form acetoacetate and succinylCoA, a reaction in the

gluconeogenesis pathway. Physiologically its enzymatic activity is involved in maintaining the

blood glucose level (38).

CpA1 is a soluble zincdependent enzyme synthesized in the pancreas and secreted into

the gastrointestinal tract where its activity is modulated by dietary zinc (39). CpA1 is a known

Zn2+ metallopeptidase and belongs to a family of enzymes which process proinsulin. CpA1 is

differentially increased in low pancreatic Zn2+ (ZnT8 RW), supporting our hypothesis that in the

HFD model of T2DM, reduced pancreatic Zn2+ could be beneficial by inducing proinsulin

maturation by CpA1 processing of enzymes involved. Diabetes Page 18 of 41

NDPKB is critically involved in the diabetic process, by modulating its isoenzyme activities and those of AMPK. NDPKA has been shown to be phosphorylated and inactivated by

AMPK and to also modulate AMPK function (40). AMPK is the major checkpoint for energy metabolism inducing ATP synthetic proteins and reducing pathways which consume ATP, and is critically involved in the diabetic process (reviewed in (41)). NDPKB is differentially increased in high pancreatic zinc conditions (Figure 6). NDPKB also has many functions related to cellular proliferation of immune cells (4244).

These results show that pancreatic Zn2+ and proinsulin levels covary, but are inversely variant with insulin and glucose tolerance in the HFD model of T2DM. We identified pancreatic

ZBP’s and determined those differentially affected by zinc concentrations as therapeutic targets.

We also suggest that ZNT8 RW acts in a dominant negative manner to reduce pancreatic Zn2+ levels, and that proinsulin and insulin processing are likely involved in the beneficial effects of this polymorphism, which we will test in future studies. Page 19 of 41 Diabetes

Acknowledgements

C.T.S. is the guarantor of this manuscript and had primary responsibility for research

design and conduct, writing, and final content; S.B. performed some of the Zn2+, insulin, and

proinsulin stainings, and analyzed data; L.L. performed most of the Zn2+ and insulin stainings,

and IPGTT measurements and analyzed data. Each author reviewed and approved the final

manuscript. We would like to thank AiLi Cai, and Chunxiao Shi of Washington University for

expert technical help in generating the DNA constructs, and in animal breeding and care. We

would like to thank Nirmala Tumarada of LSUHSC for expert technical assistance with the

imaging and statistical analyses. This publication was supported by DK 073446 (CTS), and

NCRR: 5P20 RR018766/ NIGMS: 8P20 GM103514 (CTS and proteomics core), and by NIH

P60 DK20579 to the DRTC at Washington University of which CTS is a member and which

subsidized the generation of the transgenic mice. Author disclosure: C.T. Sheline, S. Bai, and L.

Li have no conflicts of interest.

Diabetes Page 20 of 41

Table 1. Quantitation of thresholded integrated density of proinsulin, insulin, and Zn2+

staining in RW and WT transgenic positive and negative mice fed HFD or ND 1

ZP1 Insulin Proinsulin

RW HFD Tg+ 458±83 a1,c1,d1 479±52.6 a2,c2,d2 199±20 a3,c3

RW HFD Tg 782±87 343±30.6 284±78

RW ND Tg+ 804±120 b1 121.4±18 b2 116±8.9 b3

RW ND Tg 620±116 50.9±9.6 240±35

WT HFD Tg+ 828±138 e1, f1 48.1±9.6 e2, f2 204±21.3 e3

WT HFD Tg 466±57 80.5±9.2 137±16.4

WT ND Tg+ 301±56 31.2±6.4 205±8.5

WT ND Tg 646±78 31.2±11 206±33

1 Values are the average x 104 ± SEM in relative fluorescence units of thresholded integrated density from 34 independent experiments (n=1525). a1,a2,a3 indicate a significant difference between ZnT8RW Tg+ and Tg mice fed a HFD in pancreatic ZP1, insulin, and proinsulin staining, respectively. b1,b2,b3 indicate a significant difference between ZnT8RW and ZnT8WT Tg+ mice fed a ND in pancreatic ZP1, insulin, and proinsulin staining, respectively. c1,c2,c3 indicate a significant difference between ZnT8RW Tg+ mice fed a HFD versus a ND in pancreatic ZP1, insulin, and proinsulin staining, respectively. d1,d2,d3 indicate a significant difference between ZnT8RW and ZnT8WT Tg+ mice fed a HFD in pancreatic ZP1, insulin, and proinsulin staining, respectively. e1,e2,e3 indicate a significant difference between ZnT8WT Tg+ and Tg mice fed a HFD in pancreatic ZP1, insulin, and proinsulin staining, respectively. f1,f2 indicate a significant difference between ZnT8WT Tg+ and Tg mice fed a ND in pancreatic ZP1, and insulin staining, respectively.

Page 21 of 41 Diabetes

Table 2. Identification of the control pancreatic ZBP’s.

Protein Name Score 1 # of peptide MW 2 matches 1. Alpha amylase 656 29 58.1 kD

2. Nucleoside diphosphate kinase A 431 21 17.3 kD

3. Nucleoside diphosphate kinase B 419 25 17.5 kD

4. Methylmalonatesemialdehyde 328 8 58.3 kD dehydrogenase 5. Carboxypeptidase A1 182 6 47.5 kD

6. Glutathione Stransferase Mu 1 175 7 26.1 kD

7. Eukaryotic translation initiation factor 5A1 126 7 17.0 kD

8. Alphaaminoadipic semialdehyde dehydrogenase 96 1 59.3 kD 9. SuccinylCoA:3ketoacidcoenzyme A transferase 1 87 3 56.4 kD

1 A score > 70 gives > 95% confidence that protein was present in the picked protein spot

from the 2D gel. The score was based upon the number of peptides identified for the

given protein, and the intensity of that peptide.

2 Calculated size in kD from the protein sequence was given as molecular weight (MW). Diabetes Page 22 of 41

FIGURE LEGENDS

FIGURE 1. Zn2+, proinsulin, and insulin staining of hZnT8 RW male mice (line T12) after a

highfat diet (HFD) from 616 weeks of age. Pancreatic sections (10 microns) from three

different 16 week old HFD fed hZnT8 T12 transgenic (+) mice (Top Panel), and three different

HFD fed transgenic () mice (Bottom Panel) were stained with antiinsulin (exposure time

500ms), antiproinsulin (exposure time 100 ms), and Zp1 (exposure time 200ms) at

magnification x 100. Scale bar represents 400 microns.

FIGURE 2. Zn2+, proinsulin, and insulin staining of hZnT8 WT male mice (line X40) after a

HFD from 616 weeks of age. Pancreatic sections from three different 16 week old HFD fed hZnT8 X40 transgenic (+) mice (Top Panel), and three different HFD fed transgenic () mice

(Bottom Panel) were stained with antiinsulin (exposure time 500ms), antiproinsulin (exposure time 100 ms), and Zp1 (exposure time 200ms) at magnification x 100. Scale bar represents 400 microns.

FIGURE 3. Intraperitoneal glucose tolerance in ZnT8 RW T12 (A) and ZnT8 WT X40 (B) transgenic positive and negative mice fed either a HFD or a ND from 616 weeks of age. Six to ten mice per group were tested for glucose tolerance at 16 weeks (1.5 mg glucose/g body weight) and blood glucose was measured at the indicated times after injection. * indicates a significant difference from transgenic negative animals on the same diet at P < 0.05

Page 23 of 41 Diabetes

FIGURE 4. Intraperitoneal glucose tolerance in C57/Bl6 mice fed a HFD, ND, or HFDD/2,

HFDD/61, or HFDD/261 mg Zn2+/kg diet from 616 weeks of age. Six to ten mice per group

were tested for glucose tolerance at 16 weeks (1.5 mg glucose/g body weight) and blood glucose

was measured at the indicated times after injection. * indicates a significant difference from

HFDD/61 mg Zn2+/kg diet at P < 0.05

FIGURE 5. 2D gels of ZBP’s from control pancreas. Fifty g of the input, flow through, and

eluate from Zn2+agarose columns of pancreatic extracts from control animals was precipitated,

labeled with Cy5, Cy3, and Cy2 respectively, and loaded onto PEI tube gels before running on

SDSPAGE gels in the second dimension. The left panel shows the Cy2labeled eluate proteins.

The right panel shows the Cy3labeled proteins from the flow through fraction. Protein spots 5

fold enriched in eluate and containing sufficient mass were picked (faint colored circles) and

identified using LCMS (Table 1).

FIGURE 6. 2D PEI/SDSPAGE gels of the zincagarose eluates of highzinc (hZnT8WT)

pancreas, versus lowzinc (hZnT8RW) pancreas. Fifty µg of the highzinc E1, highzinc FT, and

lowzinc E1 fractions were processed. Protein spots 5fold enriched in an eluate and containing

sufficient mass were picked and identified using LCMS. Blue circles were increased in low

zinc, and the red circle was increased in high zinc conditions. The right panel shows the three

circled spots at high magnification, and with histograms.

Diabetes Page 24 of 41

Literature Cited

1. Kim BJ, Kim YH, Kim S, Kim JW, Koh JY, Oh SH, Lee MK, Kim KW, Lee MS: Zinc as a paracrine effector in pancreatic islet cell death. Diabetes 49:367372., 2000 2. Priel T, ArichaTamir B, Sekler I: Clioquinol attenuates zincdependent betacell death and the onset of insulitis and hyperglycemia associated with experimental type I diabetes in mice. Eur J Pharmacol 565:232239, 2007 3. Sheline CT, Shi C, Takata T, Zhu J, Zhang W, Sheline PJ, Cai AL, Li L: Dietary Zinc Reduction, Pyruvate Supplementation, or Zinc Transporter 5 Knockout Attenuates betaCell Death in Nonobese Diabetic Mice, Islets, and Insulinoma Cells. J Nutr 142:21192127, 2012 4. Taylor CG: Zinc, the pancreas, and diabetes: insights from rodent studies and future directions. Biometals 18:305312, 2005 5. Sladek R, Rocheleau G, Rung J, Dina C, Shen L, Serre D, Boutin P, Vincent D, Belisle A, Hadjadj S, Balkau B, Heude B, Charpentier G, Hudson TJ, Montpetit A, Pshezhetsky AV, Prentki M, Posner BI, Balding DJ, Meyre D, Polychronakos C, Froguel P: A genomewide association study identifies novel risk loci for type 2 diabetes. Nature 445:881885, 2007 6. Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos MR, Stringham HM, Chines PS, Jackson AU, ProkuninaOlsson L, Ding CJ, Swift AJ, Narisu N, Hu T, Pruim R, Xiao R, Li XY, Conneely KN, Riebow NL, Sprau AG, Tong M, White PP, Hetrick KN, Barnhart MW, Bark CW, Goldstein JL, Watkins L, Xiang F, Saramies J, Buchanan TA, Watanabe RM, Valle TT, Kinnunen L, Abecasis GR, Pugh EW, Doheny KF, Bergman RN, Tuomilehto J, Collins FS, Boehnke M: A genomewide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316:13411345, 2007 7. Majithia AR, Jablonski KA, McAteer JB, Mather KJ, Goldberg RB, Kahn SE, Florez JC, Group DPPR: Association of the SLC30A8 missense polymorphism R325W with proinsulin levels at baseline and after lifestyle, metformin or troglitazone intervention in the Diabetes Prevention Program. Diabetologia 54:25702574, 2011 8. Wareham NJ, Byrne CD, Williams R, Day NE, Hales CN: Fasting proinsulin concentrations predict the development of type 2 diabetes. Diabetes Care 22:262270, 1999 9. Wenzlau JM, Moua O, Liu Y, Eisenbarth GS, Hutton JC, Davidson HW: Identification of a major humoral epitope in Slc30A8 (ZnT8). Ann N Y Acad Sci 1150:252255, 2008 10. Nicolson TJ, Bellomo EA, Wijesekara N, Loder MK, Baldwin JM, Gyulkhandanyan AV, Koshkin V, Tarasov AI, Carzaniga R, Kronenberger K, Taneja TK, da Silva Xavier G, Libert S, Froguel P, Scharfmann R, Stetsyuk V, Ravassard P, Parker H, Gribble FM, Reimann F, Sladek R, Hughes SJ, Johnson PR, Masseboeuf M, Burcelin R, Baldwin SA, Liu M, Lara Lemus R, Arvan P, Schuit FC, Wheeler MB, Chimienti F, Rutter GA: Insulin storage and glucose homeostasis in mice null for the granule zinc transporter ZnT8 and studies of the type 2 diabetesassociated variants. Diabetes 58:20702083, 2009 11. Huang XF, Arvan P: Intracellular transport of proinsulin in pancreatic betacells. Structural maturation probed by disulfide accessibility. J Biol Chem 270:2041720423., 1995 12. Chung J, Nguyen AK, Henstridge DC, Holmes AG, Chan MH, Mesa JL, Lancaster GI, Southgate RJ, Bruce CR, Duffy SJ, Horvath I, Mestril R, Watt MJ, Hooper PL, Kingwell BA, Vigh L, Hevener A, Febbraio MA: HSP72 protects against obesityinduced insulin resistance. Proc Natl Acad Sci U S A 105:17391744, 2008 13. Miyazaki J, Araki K, Yamato E, Ikegami H, Asano T, Shibasaki Y, Oka Y, Yamamura K: Establishment of a pancreatic beta cell line that retains glucoseinducible insulin secretion: Page 25 of 41 Diabetes

special reference to expression of isoforms. Endocrinology 127:126132, 1990 14. Moynihan KA, Grimm AA, Plueger MM, BernalMizrachi E, Ford E, CrasMeneur C, Permutt MA, Imai S: Increased dosage of mammalian Sir2 in pancreatic beta cells enhances glucosestimulated insulin secretion in mice. Cell Metab 2:105117, 2005 15. Chimienti F, Devergnas S, Favier A, Seve M: Identification and cloning of a betacell specific zinc transporter, ZnT8, localized into insulin secretory granules. Diabetes 53:2330 2337, 2004 16. Han JH, Stratowa C, Rutter WJ: Isolation of fulllength putative rat lysophospholipase cDNA using improved methods for mRNA isolation and cDNA cloning. Biochemistry 26:16171625, 1987 17. Alban A, David SO, Bjorkesten L, Andersson C, Sloge E, Lewis S, Currie I: A novel experimental design for comparative twodimensional gel analysis: twodimensional difference gel electrophoresis incorporating a pooled internal standard. Proteomics 3:3644, 2003 18. Kojima H, Fujimiya M, Terashima T, Kimura H, Chan L: Extrapancreatic proinsulin/insulin expressing cells in diabetes mellitus: is history repeating itself? Endocr J 53:715722, 2006 19. Taylor KM, Nicholson RI: The LZT proteins; the LIV1 subfamily of zinc transporters. Biochim Biophys Acta 1611:1630, 2003 20. Palmiter RD, Huang L: Efflux and compartmentalization of zinc by members of the SLC30 family of solute carriers. Pflugers Arch 447:744751, 2004 21. Suzuki T, Ishihara K, Migaki H, Nagao M, YamaguchiIwai Y, Kambe T: Two different zinc transport complexes of cation diffusion facilitator proteins localized in the secretory pathway operate to activate alkaline phosphatases in vertebrate cells. J Biol Chem 280:3095630962, 2005 22. Bai S, Sheline CR, Zhou Y, Sheline CT: A reduced zinc diet or zinc transporter 3 knockout attenuate light induced zinc accumulation and retinal degeneration. Exp Eye Res 108:5967, 2013 23. Suh SW, Hamby AM, Gum ET, Shin BS, Won SJ, Sheline CT, Chan PH, Swanson RA: Sequential release of nitric oxide, zinc, and superoxide in hypoglycemic neuronal death. J Cereb Blood Flow Metab 28:16971706, 2008 24. Huang L, Gitschier J: A novel gene involved in zinc transport is deficient in the lethal milk mouse. Nat Genet 17:292297, 1997 25. Inoue K, Matsuda K, Itoh M, Kawaguchi H, Tomoike H, Aoyagi T, Nagai R, Hori M, Nakamura Y, Tanaka T: Osteopenia and malespecific sudden cardiac death in mice lacking a zinc transporter gene, Znt5. Hum Mol Genet 11:17751784., 2002 26. Chimienti F, Devergnas S, Pattou F, Schuit F, GarciaCuenca R, Vandewalle B, KerrConte J, Van Lommel L, Grunwald D, Favier A, Seve M: In vivo expression and functional characterization of the zinc transporter ZnT8 in glucoseinduced insulin secretion. J Cell Sci 119:41994206, 2006 27. Wijesekara N, Dai FF, Hardy AB, Giglou PR, Bhattacharjee A, Koshkin V, Chimienti F, Gaisano HY, Rutter GA, Wheeler MB: Beta cellspecific Znt8 deletion in mice causes marked defects in insulin processing, crystallisation and secretion. Diabetologia 53:16561668, 2010 28. Smidt K, Jessen N, Petersen AB, Larsen A, Magnusson N, Jeppesen JB, Stoltenberg M, Culvenor JG, Tsatsanis A, Brock B, Schmitz O, Wogensen L, Bush AI, Rungby J: SLC30A3 Diabetes Page 26 of 41

responds to glucose and zinc variations in betacells and is critical for insulin production and in vivo glucosemetabolism during betacell stress. PLoS One 4:e5684, 2009 29. Davidson HW, Rhodes CJ, Hutton JC: Intraorganellar calcium and pH control proinsulin cleavage in the pancreatic beta cell via two distinct sitespecific endopeptidases. Nature 333:9396, 1988 30. Park JA, Koh JY: Induction of an immediate early gene egr1 by zinc through extracellular signalregulated kinase activation in cortical culture: its role in zincinduced neuronal death. J Neurochem 73:450456, 1999 31. Kim S, Jung Y, Kim D, Koh H, Chung J: Extracellular zinc activates p70 S6 kinase through the phosphatidylinositol 3kinase signaling pathway. J Biol Chem 275:2597925984, 2000 32. Manzerra P, Behrens MM, Canzoniero LM, Wang XQ, Heidinger V, Ichinose T, Yu SP, Choi DW: Zinc induces a Src family kinasemediated upregulation of NMDA receptor activity and excitotoxicity. Proc Natl Acad Sci U S A 98:1105511061., 2001 33. Becker AB, Roth RA: Identification of glutamate169 as the third zincbinding residue in proteinase III, a member of the family of insulindegrading enzymes. Biochem J 292 ( Pt 1):137142, 1993 34. Bellomo G, Nicotera PL, Travaglino F, Palma Martino A, Mirabelli F, Fratino P: Insulin degradation in human erythrocyte: effects of cations. Acta Diabetol Lat 22:6369, 1985 35. Lu J, Combs GF, Jr.: Effect of excess dietary zinc on pancreatic exocrine function in the chick. J Nutr 118:681689, 1988 36. Chausmer AB: Zinc, insulin and diabetes. J Am Coll Nutr 17:109115, 1998 37. Ishii N, Carmines PK, Yokoba M, Imaizumi H, Ichikawa T, Ikenagasa H, Kodera Y, OhIshi M, Aoki Y, Maeda T, Takenaka T, Katagiri M: converting enzyme inhibition curbs tyrosine nitration of mitochondrial proteins in the renal cortex during the early stage of diabetes mellitus in rats. Clin Sci (Lond) 124:543552, 2013 38. Berry GT, Fukao T, Mitchell GA, Mazur A, Ciafre M, Gibson J, Kondo N, Palmieri MJ: Neonatal hypoglycaemia in severe succinylCoA: 3oxoacid CoAtransferase deficiency. J Inherit Metab Dis 24:587595, 2001 39. Fernandez D, Boix E, Pallares I, Aviles FX, Vendrell J: Structural and Functional Analysis of the Complex between Citrate and the Zinc Peptidase Carboxypeptidase A. Enzyme Res 2011:128676 40. Crawford RM, Treharne KJ, ArnaudDabernat S, Daniel JY, Foretz M, Viollet B, Mehta A: Understanding the molecular basis of the interaction between NDPKA and AMPK alpha 1. In Mol Cell Biol United States, 2006, p. 59215931 41. Rutter GA, Leclerc I: The AMPregulated kinase family: enigmatic targets for diabetes therapy. In Mol Cell Endocrinol Ireland, 2009, p. 4149 42. Di L, Srivastava S, Zhdanova O, Sun Y, Li Z, Skolnik EY: Nucleoside diphosphate kinase B knockout mice have impaired activation of the K+ channel KCa3.1, resulting in defective T cell activation. J Biol Chem 285:3876538771, 2010 43. Srivastava S, Zhdanova O, Di L, Li Z, Albaqumi M, Wulff H, Skolnik EY: Protein histidine phosphatase 1 negatively regulates CD4 T cells by inhibiting the K+ channel KCa3.1. Proc Natl Acad Sci U S A 105:1444214446, 2008 44. Postel EH, Wohlman I, Zou X, Juan T, Sun N, D'Agostin D, Cuellar M, Choi T, Notterman DA, La Perle KM: Targeted deletion of Nm23/nucleoside diphosphate kinase A and B reveals their requirement for definitive erythropoiesis in the mouse embryo. Dev Dyn 238:775787, 2009 Page 27 of 41 Diabetes

Proinsulin Insulin Zp1 Diabetes Page 28 of 41

Proinsulin Insulin Zp1 Page 29 of 41 Diabetes

A 8 7

6 * * * 5

4 * 3 Tg(-)-ND Glucose Level (g/L) 2 Tg(+)-ND Tg(-)-HFD 1 Tg(+)-HFD 0 0 15 30 45 60 120 Time (min) B 8

7 * * * 6 * 5 * 4

3 Tg(-)-ND

Glucose Level (g/L) Tg(+)-ND 2 Tg(-)-HFD 1 Tg(+)-HFD

0 0 15 30 45 60 120 Time (min) Diabetes Page 30 of 41

8

7 ** * * 6

5 * 4 ND 3 HFD HFDD/2 mg/kg diet Blood Glucose (g/L) Glucose Blood 2 HFDD/61 mg Zn/kg diet 1 HFDD/261 mg Zn/kg diet

0 0 15 30 45 60 120 Time (min) Page 31 of 41 Diabetes

Eluate Flowthrough Diabetes Page 32 of 41

A B

High Low Zinc Zinc

NDPKB

High Pancreatic Zinc

Actin CpA1

Low Pancreatic Zinc Page 33 of 41 Diabetes

Generation of human ZnT8 transgenic Mice, Breeding, and Genotyping. pIns 1 plasmid (26) containing the

human insulin promoter (1.9 kB) fused to the rabbit βglobin intron (same construct used in 27), was used as

the expression construct. Human pZnT8WT-EGFP and pZnT8RW-EGFP constructs (28) were used as

templates for PCR reactions using the ZnT8 cloning primers whose sequence is given in Supplemental Table

1. These primers generated new EcoRI sites to allow cloning, and removed the EGFP fusion protein. The

PCR products were inserted into the EcoRI site in the pIns1 plasmid and verified by sequencing, and

expression was tested in MIN6 cells. Constructs were linearized, gel purified, and injected into CBA X B6

blastocysts by the Washington University Transgenic Core Facility. These mice were bred with C57BL/6J to

syngeneity (Jackson Lab, Bar Harbor, ME). hZnT8 genotype of mouse lines was performed on tail DNA by

PCR amplification using the T8hum RTF and RTR human specific primers (Supplemental Table 1). The hZnT8

and mZnT8 primers were chosen to cover regions of sequence divergence to differentiate the human and

mouse .

Zn2+ Transporter Gene Expression. Total RNA from harvested mouse pancreas was extracted with 0.5M

guanidinium isothiocyanate. For details of the method see (29). The RNA concentration was measured, and

RNA integrity was verified by electrophoresis in a 1% agarose gel. Reverse transcription (RT) was performed

using the iScript cDNA synthesis kit (BioRad, Hercules, CA, USA) and 1g total pancreatic RNA in 20 L.

Realtime RTPCR specific primers for human ZnT8 R325W (RW) and ZnT8 wildtype (WT) were the same as

above (T8hum RTF and RTR). Mouse GAPDH, ZIP4, and ZnT8 forward and reverse primers were also used

(Supplemental Table 1)

The reactions used iQSYBR green Supermix (BioRad) and 1 L of the RT reaction with a final

concentration of 100 nmol/L of the primer. The PCR cycle for ZnT8 was as follows: 95°C 5 min, 40 cycles of

95°C/30 s, 62°C/30 s, and 72°C/ 30 s. The PCR cycle for mZIP4 was as described (30). The melt curve was

65°C to 95°C in increments of 0.5°C every 5 s to demonstrate a single PCR product, and the correct size

product was verified by agarose gel electrophoresis. The meltcurve analyses were performed using an iCycler

iQ Multicolor RealTime PCR Detection System (BioRad). Samples were compared using the relative CT (The

cycle number at the threshold level of logbased fluorescence) method. The fold increase or decrease

(=Power) was determined relative to a transgenic negative control after normalizing to the GAPDH Diabetes Page 34 of 41

housekeeping gene using the 2∆∆CT(Livak) method, where ∆CT is (gene of interest CT) (GAPDH CT) and

∆∆CT is (∆CT ZnT8RW+ or ZnT8WT+) (∆CT ZnT8RW or ZnT8WT ).

SUPPLEMENTAL FIGURE 1. Expression levels of hZnT8 WT and RW transgenic mouse lines. Human ZnT8

mRNA expression for WT and R325W animal lines (upper panel) was assessed by RTPCR performed on

mouse pancreas RNA. Mouse GAPDH was used as an internal control (lower panel). The negative control (

CT) was performed without RNA, and the positive control (+CT) was performed using 1 ng of ZnT8-GFP

plasmid DNA. The names of the different animal lines are given below.

SUPPLEMENTAL FIGURE 2. RTPCR of tissues from ZnT8 WT and RW transgenic mouse lines. To

demonstrate the relative levels of expression of the transgene in different tissues, the pancreas (P), thymus

(T), brain (B), and liver (L) were isolated, RNA extracted, and RTPCR performed. The upper panel shows the

ZnT8 expression in each sample, whereas the lower panel shows the GAPDH loading control expression in each sample. The first lane is DNA size markers, and the last lane in the upper panel is the positive control for

ZnT8 (1 ng of ZnT8-GFP plasmid DNA).

SUPPLEMENTAL FIGURE 3. Zn2+, proinsulin, and insulin staining of hZnT8 RW male mice (line T12) after a normal diet (ND). Pancreatic sections from three different 16 week old hZnT8 T12 transgenic (+) mice fed ND

(Top Panel), and three different transgenic () mice fed ND (Bottom Panel) were stained with antiinsulin

(exposure time 500ms), antiproinsulin (exposure time 100 ms), and Zp1 (exposure time 200ms) at

magnification x 100. Scale bar represents 400 microns.

SUPPLEMENTAL FIGURE 4. Zn2+, proinsulin, and insulin staining of hZnT8 WT male mice (line X40) after a

ND. Pancreatic sections from three different 16 week old hZnT8 X40 transgenic (+) mice fed ND (Top Panel) , and three different transgenic () mice fed ND (Bottom Panel) were stained with antiinsulin (exposure time

500ms), antiproinsulin (exposure time 100 ms), and Zp1 (exposure time 200ms) at magnification x 100. Scale bar represents 400 microns.

Page 35 of 41 Diabetes

Online Supplemental Material

SUPPLEMENTAL TABLE 1. Primer sequences used in this manuscript.

1 hZnT8CloningF: 5’CCCCGGATCCCGAATTCATGGAGTTTCTTGAA3’

1 hZnT8CloningR: 5’CCCCGAATTCCGGATCCGTCACAGGGGTCTTC3’

2 hZnT8RTF: 5’CTCCTGGGCAACGTGCTGGTTGTTGTGCTG3’

2 hZnT8RTR: 5’TGGTCTCTCTCTGGGACACTGATCTTTATT3’ (190bp).

3 mZnT8RTF: 5’CAAGCCACCAAGATGTACGCCTTCCCTCTA3’

3 mZnT8RTR: 5’ TCGGTGCCCTGCTGTCTGTCCTTTGCATCT3’ (415bp)

3 mGAPDHRTF: 5’GGTGAAGGTCGGTGTGAACG3’

3 mGAPDHRTR: 5’CTCGCTCCTGGAAGATGGTG3’ (230 bp).

3 mZIP4longRTF: 5′AGAAGTCAGCACCTCTACAAGGAACGC3’

3 mZIP4longRTR: 5′AGTAGCTGGCTCAGACCCAGGGTC3′ (475bp).

1 Cloning primers were used to generate restriction enzyme sites, and to remove the GFP

portion of the fusion protein construct used as the template for the cDNA PCR reaction.

2 hZnT8 primers were used both for DNA genotyping of the mouse transgenic lines, and for

qRTPCR of pancreatic RNA levels.

3 The other three pairs of primers were used for qRTPCR of the specified pancreatic RNA. The

number given in parentheses is the size of the PCR product in base pairs. Diabetes Page 36 of 41

Online Supporting Material

SUPPLEMENTAL TABLE 2 Content of Harlan Teklad High Fat Diets

Ingredient High Fat Diet (60% of calories) 1 High Fat Diet (60% of calories) 2 (Harlan Teklad TD.06414, g/kg) (Harlan Teklad TD.10872, g/kg) Casein 265.0 NA Egg White Solids, spray-dried NA 266.0 L-Cystine 4.0 NA Dextrose, monohydrate NA 151.5 Maltodextrin 160.0 160.0 Sucrose 90.0 NA Lard 310.0 325.0 Soybean Oil 30.0 NA Corn Oil NA 20.0 Cellulose 65.5 30.0 Mineral Mix, AIN-93G-MX (3) 48.0 NA Mineral Mix, Zn Deficient NA 34.2 (81264) (31) Calcium Phosphate, dibasic 3.4 NA Biotin NA 0.005 Vitamin Mix, AIN-93G-VX (3) 21.0 NA Vitamin Mix, Teklad (40060) (31) NA 13.3 Choline Bitartrate 3.0 NA Chromium Potassium Sulfate, NA 0.027 dodecahydrate Blue Food Color 0.1 NA Ethoxyquin, antioxidant NA 0.004

1 This diet contained 50-60 mg Zn2+/kg of diet.

2 This diet contained 0.8-1.2 mg Zn2+/kg of diet.

NA specifies not added. Page 37 of 41 Diabetes

Supplemental Table 3. Quantitation of RT-PCR of the ZnT8 WT and ZnT8 RW transgenic mouse lines 1

Animal Sample hZnT8 mZIP4 mZnT8

Line ID Power Power Power

X-28-WT 8078+ 1076 ± 137 a NP NP

X-40-WT 549+ 1258 ± 94 a 0.5 ± 0.1 b 0.9 ± 0.2

T-12-RW 8038+ 1643 ± 160 a 1.8 ± 0.2 b 1.1 ± 0.2

T-16-RW 852+ 46 ± 15 NP NP

T-8-RW 8024+ 53 ± 12 NP NP

Negative 615- 1 ± 3 1.0 ± 0.15 1.0 ± 0.17

1 Values are the average ± SEM, from 4 independent experiments, normalized to GAPDH, and compared to

hZnT8 transgenic negative littermates (fold increase above negative control).

a Indicates a significant difference of hZnT8 RNA expression from negative littermates at P < 0.05

b Indicates a significant difference of mZIP4 RNA expression from negative littermates at P < 0.05 Page 38 of 41 of 38 Page ZnT8 190bp 230bp GAPDH –CT +CT Wtins-523wt Wtins-615wt Wtins-619wt Rwins-866+ Rwins-7111+

Wtins-8065+ Wtins-8053+

Diabetes Wtins-331+

Wtins-8078+ Wtins-549+ Rwins-1077/1078-2+ Rwins-8019+ Rwins-852+

Rwins-8024+ Rwins-8038+ Page 39 of 41 Diabetes

Negative ZnT8 ZnT8 WT Control R325W PTBLPTBL PTBL+

ZnT8

GAPDH Diabetes Page 40 of 41

Online Supplemental Material

Proinsulin Insulin Zp1

Supplemental Figure 3. Zn2+, proinsulin, and insulin staining of hZnT8 RW male mice (line 5 after a normal diet /%  Pancreatic sections from three different  week old hZnT8 5 transgenic mice fed ND (Top 1BOFM and three different transgenic  mice fed ND (Bottom 1BOFM were stained with BOUJJOTVMJO(exposure time NT BOUJQSPJOTVMJO (exposure time NT and ;Q (exposure time NT at magnification Y Scale bar represents 400 microns. Page 41 of 41 Diabetes

Online Supplemental Material

Proinsulin Insulin Zp1

Supplemental Figure 4. Zn2+, proinsulin, and insulin staining of hZnT8 WT male mice (line 9 after a ND. Pancreatic sections from three different  week old hZnT8 9 transgenic mice fed ND (Top 1BOFM and three different transgenic  mice fed ND (Bottom 1BOFM were stained with BOUJJOTVMJO(exposure time NT BOUJQSPJOTVMJO (exposure time NT and ;Q (exposure time NT at magnification Y Scale bar represents 400 microns.