Top Banner
Cytokines Interleukin-1 and Tumor Necrosis Factor- Regulate Different Transcriptional and Alternative Splicing Networks in Primary -Cells Fernanda Ortis, 1 Najib Naamane, 1 Daisy Flamez, 1 Laurence Ladrie `re, 1 Fabrice Moore, 1 Daniel A. Cunha, 1 Maikel L. Colli, 1 Thomas Thykjaer, 2 Kasper Thorsen, 3 Torben F. Ørntoft, 2,3 and Decio L. Eizirik 1 OBJECTIVE—Cytokines contribute to pancreatic -cell death in type 1 diabetes. This effect is mediated by complex gene networks that remain to be characterized. We presently utilized array analysis to define the global expression pattern of genes, including spliced variants, modified by the cytokines interleukin (IL)-1 interferon (IFN)- and tumor necrosis factor (TNF)- IFN- in primary rat -cells. RESEARCH DESIGN AND METHODS—Fluorescence-acti- vated cell sorter–purified rat -cells were exposed to IL-1 IFN- or TNF- IFN- for 6 or 24 h, and global gene expression was analyzed by microarray. Key results were confirmed by RT-PCR, and small-interfering RNAs were used to investigate the mechanistic role of novel and relevant transcription factors identified by pathway analysis. RESULTS—Nearly 16,000 transcripts were detected as present in -cells, with temporal differences in the number of genes modulated by IL-1 IFN or TNF- IFN-. These cytokine combinations induced differential expression of inflammatory response genes, which is related to differential induction of IFN regulatory factor-7. Both treatments decreased the expression of genes involved in the maintenance of -cell phenotype and growth/regeneration. Cytokines induced hypoxia-inducible fac- tor-, which in this context has a proapoptotic role. Cytokines also modified the expression of 20 genes involved in RNA splicing, and exon array analysis showed cytokine-induced changes in alternative splicing of 50% of the cytokine-modified genes. CONCLUSIONS—The present study doubles the number of known genes expressed in primary -cells, modified or not by cytokines, and indicates the biological role for several novel cytokine-modified pathways in -cells. It also shows that cyto- kines modify alternative splicing in -cells, opening a new avenue of research for the field. Diabetes 59:358–374, 2010 T ype 1 diabetes is an autoimmune disease char- acterized by a progressive and selective destruc- tion of the pancreatic -cells. During insulitis, activated macrophages and T-cells release cyto- kines such as interleukin (IL)-1, tumor necrosis factor (TNF)-, and interferon (IFN)- in the vicinity of the -cells, contributing for -cell dysfunction and apoptosis (1,2). Expression of TNF- and IL-1 was observed in pancreas of patients with recent type 1 diabetes onset and in animal models of the disease (1–3), prompting clinical trials based on the use of blockers of TNF- (4) or IL-1 (5) to prevent type 1 diabetes. In vitro exposure of rodent or human -cells to IL-1 IFN- or TNF- IFN-, but not to any of these cytokines alone, triggers -cell apoptosis (1,6). IL-1 IFN- affects the expression of several gene networks in -cells, modu- lating pro- and antiapoptotic pathways, expression of cytokines and chemokines, and decreasing expression of genes involved in -cell function (2,6 –10). Less is known about the genes induced by TNF-; both cytokines induce the key transcription factor nuclear factor (NF)-B (11), but they affect kinase cascade pathways differently, such as IB kinase, with the potential to trigger a differential gene expression outcome (11,12). We have previously addressed this issue by using a target microarray, the Apochip (13), to compare IL-1– and TNF-–induced genes. The findings obtained indicated some differences between these cytokines, mostly related to intensity of gene expression (12). These observations, however, were biased by the choice and limited number of probes in- cluded in the Apochip. Moreover, neither the Apochip nor usually utilized cDNA arrays (7–9) have the ability to identify splice variants of genes. This is a significant limitation, since recent data suggest that regulation of alternative splicing is of major importance for regulation of proteomic diversity and for cell physiology/pathology (14 –16). Cytokine composition and its respective concentrations may vary during insulitis, depending on the timing, degree of islet infiltration, immune cells present, and the pancre- atic -cell responses to the immune assault (10). This may explain why blocking TNF- or IL-1 at different stages of the pre-diabetic period may be more or less effective in preventing diabetes in rodent models (1,17), suggesting that the contribution of the different cytokines and their downstream signaling pathways may also vary between individual type 1 diabetic patients. This reinforces the need for understanding separately and in detail the gene From the 1 Laboratory of Experimental Medicine, Universite ´ Libre de Brux- elles, Brussels, Belgium; 2 CMO Aros Applied Biotechnology A/S, Science Park Skejby Brendstrupgaardsvej, Aarhus, Denmark; and the 3 Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark. Corresponding author: Decio L. Eizirik, [email protected]. Received 5 August 2009 and accepted 28 October 2009. Published ahead of print at http://diabetes.diabetesjournals.org on 23 November 2009. DOI: 10.2337/db09-1159. F.O. and N.N. contributed equally to this article. © 2010 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by -nc-nd/3.0/ for details. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. See accompanying commentary, p. 335. ORIGINAL ARTICLE 358 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org
17

Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

Feb 05, 2023

Download

Documents

Frank Vermeulen
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

Cytokines Interleukin-1� and Tumor Necrosis Factor-�Regulate Different Transcriptional and AlternativeSplicing Networks in Primary �-CellsFernanda Ortis,

1Najib Naamane,

1Daisy Flamez,

1Laurence Ladriere,

1Fabrice Moore,

1

Daniel A. Cunha,1

Maikel L. Colli,1

Thomas Thykjaer,2

Kasper Thorsen,3

Torben F. Ørntoft,2,3

and Decio L. Eizirik1

OBJECTIVE—Cytokines contribute to pancreatic �-cell deathin type 1 diabetes. This effect is mediated by complex genenetworks that remain to be characterized. We presently utilizedarray analysis to define the global expression pattern of genes,including spliced variants, modified by the cytokines interleukin(IL)-1� � interferon (IFN)-� and tumor necrosis factor (TNF)-� �IFN-� in primary rat �-cells.

RESEARCH DESIGN AND METHODS—Fluorescence-acti-vated cell sorter–purified rat �-cells were exposed to IL-1� �IFN-� or TNF-� � IFN-� for 6 or 24 h, and global gene expressionwas analyzed by microarray. Key results were confirmed byRT-PCR, and small-interfering RNAs were used to investigate themechanistic role of novel and relevant transcription factorsidentified by pathway analysis.

RESULTS—Nearly 16,000 transcripts were detected as presentin �-cells, with temporal differences in the number of genesmodulated by IL-1� � IFN� or TNF-� � IFN-�. These cytokinecombinations induced differential expression of inflammatoryresponse genes, which is related to differential induction of IFNregulatory factor-7. Both treatments decreased the expression ofgenes involved in the maintenance of �-cell phenotype andgrowth/regeneration. Cytokines induced hypoxia-inducible fac-tor-�, which in this context has a proapoptotic role. Cytokinesalso modified the expression of �20 genes involved in RNAsplicing, and exon array analysis showed cytokine-inducedchanges in alternative splicing of �50% of the cytokine-modifiedgenes.

CONCLUSIONS—The present study doubles the number ofknown genes expressed in primary �-cells, modified or not bycytokines, and indicates the biological role for several novelcytokine-modified pathways in �-cells. It also shows that cyto-kines modify alternative splicing in �-cells, opening a newavenue of research for the field. Diabetes 59:358–374, 2010

Type 1 diabetes is an autoimmune disease char-acterized by a progressive and selective destruc-tion of the pancreatic �-cells. During insulitis,activated macrophages and T-cells release cyto-

kines such as interleukin (IL)-1�, tumor necrosis factor(TNF)-�, and interferon (IFN)-� in the vicinity of the�-cells, contributing for �-cell dysfunction and apoptosis(1,2). Expression of TNF-� and IL-1� was observed inpancreas of patients with recent type 1 diabetes onset andin animal models of the disease (1–3), prompting clinicaltrials based on the use of blockers of TNF-� (4) or IL-1�(5) to prevent type 1 diabetes.

In vitro exposure of rodent or human �-cells to IL-1� �IFN-� or TNF-� � IFN-�, but not to any of these cytokinesalone, triggers �-cell apoptosis (1,6). IL-1� � IFN-� affectsthe expression of several gene networks in �-cells, modu-lating pro- and antiapoptotic pathways, expression ofcytokines and chemokines, and decreasing expression ofgenes involved in �-cell function (2,6–10). Less is knownabout the genes induced by TNF-�; both cytokines inducethe key transcription factor nuclear factor (NF)-�B (11),but they affect kinase cascade pathways differently, suchas I�B kinase, with the potential to trigger a differentialgene expression outcome (11,12). We have previouslyaddressed this issue by using a target microarray, theApochip (13), to compare IL-1�– and TNF-�–inducedgenes. The findings obtained indicated some differencesbetween these cytokines, mostly related to intensity ofgene expression (12). These observations, however, werebiased by the choice and limited number of probes in-cluded in the Apochip. Moreover, neither the Apochip norusually utilized cDNA arrays (7–9) have the ability toidentify splice variants of genes. This is a significantlimitation, since recent data suggest that regulation ofalternative splicing is of major importance for regulationof proteomic diversity and for cell physiology/pathology(14–16).

Cytokine composition and its respective concentrationsmay vary during insulitis, depending on the timing, degreeof islet infiltration, immune cells present, and the pancre-atic �-cell responses to the immune assault (10). This mayexplain why blocking TNF-� or IL-1� at different stages ofthe pre-diabetic period may be more or less effective inpreventing diabetes in rodent models (1,17), suggestingthat the contribution of the different cytokines and theirdownstream signaling pathways may also vary betweenindividual type 1 diabetic patients. This reinforces theneed for understanding separately and in detail the gene

From the 1Laboratory of Experimental Medicine, Universite Libre de Brux-elles, Brussels, Belgium; 2CMO Aros Applied Biotechnology A/S, SciencePark Skejby Brendstrupgaardsvej, Aarhus, Denmark; and the 3Departmentof Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.

Corresponding author: Decio L. Eizirik, [email protected] 5 August 2009 and accepted 28 October 2009. Published ahead of

print at http://diabetes.diabetesjournals.org on 23 November 2009. DOI:10.2337/db09-1159.

F.O. and N.N. contributed equally to this article.© 2010 by the American Diabetes Association. Readers may use this article as

long as the work is properly cited, the use is educational and not for profit,and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.

The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked “advertisement” in accordance

with 18 U.S.C. Section 1734 solely to indicate this fact.

See accompanying commentary, p. 335.

ORIGINAL ARTICLE

358 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org

Page 2: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

networks downstream of IL-1� � IFN-� and TNF-� �IFN-�, with the ultimate goal of devising targeted andindividualized therapies to preserve �-cells in early type 1diabetes. We have presently addressed this question byusing primary rat �-cells treated for 6 or 24 h withcombinations of IL-1� � IFN-� and TNF-� � IFN-� andperforming array analysis using first the latest Affymetrixmicroarray, covering �28,000 genes, and then the Af-fymetrix exon-array, covering �850,000 exons and havingthe potential to identify most splice variants present in acell. This was followed by global analysis of gene expres-sion using Ingenuity Pathway Analysis (IPA) software,which indicated networks of special interest for subse-quent studies. The data obtained doubles the number ofknown genes expressed in primary rat �-cells, modified ornot by cytokines, and identifies several novel cytokine-modified pathways in �-cells, including cytokines/chemo-kines, Krebs cycle genes, hormone receptors, and hypoxia-inducible factor (HIF)-1�–regulated genes. It alsoindicates that cytokines modify alternative splicing in�-cells, opening a new avenue of research in the field.

RESEARCH DESIGN AND METHODS

Cell culture and cytokine exposure; viability and Western blot assay; nitric oxideand chemokine (CC-motif) ligand (CCL) 5 measurement; sample preparation forarray analysis; real-time RT-PCR and normal PCR; immunofluorescence; pro-moter in silico analysis and promoter reporter assay are available at the onlineappendix Supplementary Methods at http://diabetes.diabetesjournals.org/cgi/content/full/db09-1159/DC1.Gene expression array data analysis. The GeneChip Rat Genome 230 2.0arrays (Affymetrix), containing 31,099 probesets representing �28,000 ratgenes was used in the study. The GC-Robust MultiChip Average (GCRMA) (18)was used, as part of the GCRMA package in the Bioconductor site (http://www.bioconductor.org), to preprocess the raw data (CEL files). For theanalysis, see Supplementary Methods. Pathway analysis was done by IPA 5.5software.Exon-array data analysis. The CEL files corresponding to the GeneChip RatExon 1.0 ST Arrays (Affymetrix) were imported and analyzed by the Array-Assist Exon software (Stratagene Software Solutions), as described in Sup-plementary Methods.RNA interference. Small-interfering RNA (siRNA) against activating factor(ATF) 4, HIF-1�, and IFN regulatory factor (IRF)-7 (supplementary Table 2),were used to knock down expression of the respective target genes. AllstarsNegative Control siRNA (Qiagen, Venlo, Netherlands) was used as a negativecontrol. Transfection using DharmaFECT1 (Thermo Scientific, Chicago, IL)was performed as previously described and validated (19).Statistical analysis. Comparisons between groups were carried out either bypaired t test or by ANOVA followed by t tests with Bonferroni correction asrequired. A P � 0.05 was considered as statistically significant. Arraystatistical analysis is described in Supplementary Methods.

RESULTS

Effect of IL-1� � IFN-� or TNF-� � IFN-� on theviability, nitric oxide production, and gene expres-sion of rat �-cells. �-Cells were exposed to IL-1� �IFN-� or TNF-� � IFN-� and collected at 6 and 24 h forarray analysis. Viability was not affected by the cytokinetreatment after 24 h (supplementary Fig. 1A), but therewas a twofold increase in apoptosis after 72 h (supplemen-tary Fig. 1A) without significant changes in the percentageof necrotic cells (data not shown). Both cytokine combi-nations increased nitric oxide (NO) production after 24 hof exposure (supplementary Fig. 1B), with higher induc-tion by IL-1� � IFN-� as compared with TNF-� � IFN-�.These results are similar to our previous observations(12), confirming biological activity of the cytokines. In thearray analysis, nearly 16,000 probe sets, corresponding to7,991 genes, were detected as present in control and/orcytokine-treated �-cells (supplementary Table 3). TNF-� �

IFN-� modified the expression of a higher number ofgenes compared with IL-1� � IFN-� at 6 h, while this wasinverted at 24 h, with higher number of IL-1� � IFN-�–modified genes (Fig. 1). At 6 and 24 h, 67 and 48%,respectively, of the total number of cytokine-modifiedgenes was differentially induced by IL-1� � IFN-� orTNF-� � IFN-�. Supplementart Tables 4–7 list all tran-scripts considered as modified by the different cytokinecombinations at 6 and 24 h and classified by IPA. In Table1, selected genes with a putative role in �-cell function/dysfunction and death were classified by one of theinvestigators (D.L.E.), using an adaptation of a previouslydescribed in-house classification (7,9).Analysis of gene networks and pathways regulated byIL-1� � IFN-� or TNF-� � IFN-� in rat �-cells. IPAanalysis identified 50 and 100 IL-1� � IFN-�–modified and50 and 86 TNF-� � IFN-�–modified networks containing�12 focus genes and representing key transcription fac-tors and their interactions with target genes after 6 and24 h, respectively (data not shown). The networks regu-lated by the transcription factors NF-�B (supplementaryFig. 2A) and Myc (supplementary Fig. 2B) were among thetop scores for both cytokines. Depending on the cytokinestested, however, these networks often contained differentgroups of genes regulated by the same transcription fac-tor. Different temporal patterns of transcription factoractivation may lead to a differential induction of down-stream genes (11). IL-1� induced an earlier and moresustained NF-�B activation, represented by nuclear p65, ascompared with TNF-� (supplementary Fig. 2C).

The canonical pathways regulated by IL-1� � IFN-� orTNF-� � IFN-� after 24 h were identified by IPA, and thetop 32 pathways are shown in supplementary Fig. 3.Among these, many were related to local inflammatoryresponses, such as IFN signaling, antigen presentation,antiviral responses, and production of cytokines or che-mokines. Several of the pathways were involved in theintracellular signaling induced by cytokines (such as thosemediated by Janus kinase/signal transducers and activa-tors of transcription, HIF-1� and NF-�B), apoptosis, cellcycle regulation, cell metabolism (e.g., Krebs [citrate]cycle), or in endoplasmic reticulum stress. Based on theidentification of these pathways, we focused on novelpathways of particular relevance for insulitis/�-cell apo-ptosis, aiming to identify regulatory transcription factorsby use of siRNA strategy (see below).Differential inflammatory signature of IL-1� andTNF-�. Cytokines regulate expression of many genesinvolved in the inflammatory response, such as “chemo-kines/cytokines/adhesion molecules” and “IFN-� signal-

384 622 846

IL TNF

6h

1000 1531 398

IL TNF

24h

FIG. 1. Effects of cytokine exposure on gene expression in FACS-purified rat �-cells. Ven diagram showing the number of �-cell geneswith the expression modified by cytokines after exposure to IL-1� �IFN-� (IL) or TNF-� � IFN-� (TNF) for 6 and 24 h. The diagram showsgenes modified by IL-1� � IFN-� alone (left part of the figure), TNF-� �IFN-� alone (right) or both (center). Results of three independentarray experiments were analyzed. mRNA expression was considered asmodified by cytokines when P < 0.02 and fold change >1.5 comparedwith control condition.

F. ORTIS AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 59, FEBRUARY 2010 359

Page 3: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

TA

BLE

1Se

lect

edge

nes

mod

ulat

edby

cyto

kine

trea

tmen

tde

tect

edby

arra

yan

alys

is

Pro

beG

enB

ank

Gen

ena

me/

func

tion

algr

oup

Sym

bol

6h

24h

IL�

IFN

TN

F�

IFN

IL�

IFN

TN

F�

IFN

Arg

inin

em

etab

olis

man

dN

Ofo

rmat

ion

1368

266_

atN

M_0

1713

4ar

g1A

rg1

0.65

�0.

060.

76�

0.16

0.15

�0.

030.

28�

0.18

1370

964_

atB

F28

3456

ASS

Ass

5.93

�1.

300.

75�

2.55

14.2

1�

4.66

1.73

�1.

3513

8766

7_at

L125

62iN

OS2

Nos

237

5.0

�59

.78

109.

3�

214.

511

3.8

�16

.03

57.8

0�

21.8

9G

luco

sem

etab

olis

m

1386

916_

atN

M_0

1732

1A

coni

tase

1(K

rebs

)A

co1

0.23

�0.

120.

38�

0.04

0.41

�0.

040.

56�

0.11

1367

589_

atN

M_0

2439

8A

coni

tase

2,m

itoc

hond

rial

(Kre

bs)

Aco

20.

45�

0.08

0.53

�0.

070.

61�

0.03

0.57

�0.

0613

7529

5_at

AI0

0965

7C

itra

tesy

ntha

se(K

rebs

)C

s0.

73�

0.18

0.60

�0.

041.

42�

0.15

1.31

�0.

0513

6767

0_at

NM

_017

005

Fum

aras

e/fu

mar

ate

hydr

atas

e1

(Kre

bs)

Fh1

0.34

�0.

030.

58�

0.04

0.64

�0.

030.

77�

0.04

1370

865_

atB

I277

627

Isoc

itra

tede

hydr

ogen

ase

3(N

AD

),�

(Kre

bs)

Idh3

g0.

61�

0.03

0.61

�0.

090.

56�

0.05

0.66

�0.

0213

8816

0_a_

atA

I171

793

Isoc

itra

tede

hydr

ogen

ase

3(N

AD

�)

�(K

rebs

)Id

h3B

0.67

�0.

230.

74�

0.10

1.30

�0.

181.

39�

0.05

1372

790_

atB

G67

1530

Mal

ate

dehy

drog

enas

e1,

NA

D(s

olub

le)

(Kre

bs)

Mdh

10.

47�

0.06

0.53

�0.

090.

20�

0.02

0.29

�0.

0213

7279

0_at

BG

6715

30M

alat

ede

hydr

ogen

ase

1,N

AD

(sol

uble

)(K

rebs

)M

dh1

0.47

�0.

060.

53�

0.09

0.20

�0.

020.

29�

0.02

1367

653_

a_at

NM

_033

235

Mal

ate

dehy

drog

enas

e1,

NA

D(s

olub

le)

(Kre

bs)

Mdh

10.

67�

0.12

0.66

�0.

030.

52�

0.02

0.52

�0.

0513

6765

3_a_

atN

M_0

3323

5M

alat

ede

hydr

ogen

ase

1,N

AD

(sol

uble

)(K

rebs

)M

dh1

0.67

�0.

120.

66�

0.03

0.52

�0.

020.

52�

0.05

1369

927_

atN

M_0

3115

1M

alat

ede

hydr

ogen

ase

2,N

AD

(mit

ocho

ndri

al)

(Kre

bs)

Mdh

20.

53�

0.16

0.48

�0.

050.

86�

0.03

0.79

�0.

09

1390

020_

atB

I277

513

�-K

etog

luta

rate

dehy

drog

enas

e(K

rebs

)�

kdgh

0.69

�0.

140.

75�

0.09

0.44

�0.

010.

55�

0.06

1380

813_

atA

A89

1239

Succ

inat

ede

hydr

ogen

ase

com

plex

(Kre

bs)

Sdhb

_pre

dict

ed0.

97�

0.23

1.00

�0.

080.

66�

0.10

0.89

�0.

0913

7212

3_at

AI1

7232

0Su

ccin

ate

dehy

drog

enas

eco

mpl

ex(K

rebs

)Sd

hb_p

redi

cted

0.84

�0.

250.

59�

0.11

1.43

�0.

051.

32�

0.07

1373

017_

atA

I237

518

Succ

inyl

-CoA

synt

heta

se,

�-s

ubun

it(K

rebs

)Su

clg2

0.84

�0.

240.

84�

0.06

0.65

�0.

080.

89�

0.02

1367

617_

atN

M_0

1249

5A

ldol

ase

A(g

lyco

lysi

s)A

ldoa

0.77

�0.

080.

66�

0.01

1.35

�0.

131.

27�

0.04

1387

312_

a_at

NM

_012

565

Glu

coki

nase

(gly

coly

sis)

Gck

0.20

�0.

070.

32�

0.14

0.22

�0.

050.

24�

0.10

1383

519_

atB

I294

137

Hex

okin

ase

2(g

lyco

lysi

s)H

k24.

99�

2.62

17.2

2�

5.44

16.7

7�

8.13

20.4

3�

10.5

313

6757

5_at

NM

_012

554

Eno

lase

1,al

pha

(gly

coly

sis)

Eno

10.

70�

0.07

0.62

�0.

041.

71�

0.08

1.62

�0.

1913

8831

8_at

BI2

7976

0P

hosp

hogl

ycer

ate

kina

se1

(gly

coly

sis)

Pgk

10.

81�

0.10

0.70

�0.

102.

08�

0.16

1.61

�0.

2013

8686

4_at

NM

_053

290

Pho

spho

glyc

erat

em

utas

e1

(gly

coly

sis)

Pga

m1

0.71

�0.

160.

70�

0.07

1.53

�0.

101.

52�

0.06

1378

382_

atA

I230

014

Pho

spho

glyc

erat

em

utas

efa

mily

mem

ber

5(g

lyco

lysi

s)P

gam

51.

05�

0.05

0.91

�0.

051.

64�

0.04

1.61

�0.

10

1391

577_

atB

I293

450

Pho

spho

glyc

erat

em

utas

efa

mily

mem

ber

5(g

lyco

lysi

s)P

gam

51.

12�

0.07

0.90

�0.

051.

56�

0.04

1.52

�0.

24

1367

864_

atN

M_0

3171

5P

hosp

hofr

ucto

kina

se,

mus

cle

(gly

coly

sis)

Pfk

m0.

46�

0.11

0.43

�0.

080.

21�

0.04

0.23

�0.

1213

7218

2_at

BM

3897

69P

hosp

hofr

ucto

kina

se,

plat

elet

(gly

coly

sis)

Pfk

p4.

98�

0.72

4.95

�0.

347.

76�

0.71

6.09

�0.

1313

8726

3_at

NM

_012

624

Pyr

uvat

eki

nase

,liv

eran

dre

dbl

ood

cell

(gly

coly

sis)

Pkl

r0.

51�

0.16

0.32

�0.

220.

15�

0.01

0.27

�0.

06

1368

651_

atM

1768

5P

yruv

ate

kina

se,

liver

and

red

bloo

dce

ll(g

lyco

lysi

s)P

klr

0.51

�0.

060.

52�

0.07

0.20

�0.

020.

22�

0.15

1370

200_

atB

I284

411

Glu

tam

ate

dehy

drog

enas

e1

Glu

d10.

32�

0.06

0.24

�0.

080.

26�

0.01

0.31

�0.

0913

8787

8_at

AW

9166

44G

luta

mat

ede

hydr

ogen

ase

1G

lud1

0.38

�0.

060.

26�

0.02

0.45

�0.

040.

43�

0.03

1370

870_

atM

3059

6M

alic

enzy

me

1M

e10.

51�

0.18

0.57

�0.

090.

54�

0.02

0.67

�0.

0813

7006

7_at

NM

_012

600

Mal

icen

zym

e1

Me1

0.56

�0.

130.

63�

0.09

0.56

�0.

030.

84�

0.07

1386

917_

atN

M_0

1274

4P

yruv

ate

carb

oxyl

ase

Pc

0.38

�0.

150.

75�

0.10

0.47

�0.

090.

61�

0.10

1371

388_

atB

M38

9223

Pyr

uvat

ede

hydr

ogen

ase

(lip

oam

ide)

�P

dhb

0.58

�0.

100.

52�

0.08

0.74

�0.

050.

73�

0.07

1372

229_

atA

I179

119

Pyr

uvat

ede

hydr

ogen

ase

kina

se,

isoe

nzym

e3

(map

ped)

Pdk

30.

21�

0.01

0.54

�0.

120.

09�

0.03

0.49

�0.

04

GENE NETWORKS AND �-CELL APOPTOSIS

360 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org

Page 4: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

TA

BLE

1C

onti

nued

Pro

beG

enB

ank

Gen

ena

me/

func

tion

algr

oup

Sym

bol

6h

24h

IL�

IFN

TN

F�

IFN

IL�

IFN

TN

F�

IFN

1370

848_

atB

I284

218

Solu

teca

rrie

rfa

mily

2(f

acili

tate

dgl

ucos

etr

ansp

orte

r),

mem

ber

1Sl

c2a1

4.25

�0.

372.

22�

0.28

12.8

0�

5.29

9.07

�2.

86

1387

228_

atN

M_0

1287

9So

lute

carr

ier

fam

ily2

(fac

ilita

ted

gluc

ose

tran

spor

ter)

,m

embe

r2

Slc2

a20.

37�

0.03

0.38

�0.

040.

27�

0.04

0.37

�0.

08

Lipi

dm

etab

olis

m

1367

763_

atD

1392

1A

cety

l-coe

nzym

eA

acet

yltr

ansf

eras

e1

Aca

t10.

47�

0.00

0.59

�0.

060.

30�

0.05

0.23

�0.

1213

8341

6_at

AA

8993

04A

cety

l-coe

nzym

eA

acet

yltr

ansf

eras

e1

Aca

t10.

32�

0.08

0.39

�0.

120.

36�

0.05

0.34

�0.

0513

7093

9_at

D90

109

Acy

l-CoA

synt

heta

selo

ng-c

hain

fam

ilym

embe

r1

Acs

l10.

78�

0.22

1.45

�0.

110.

61�

0.09

1.22

�0.

0513

6817

7_at

NM

_057

107

Acy

l-CoA

synt

heta

selo

ng-c

hain

fam

ilym

embe

r3

Acs

l30.

78�

0.16

0.51

�0.

101.

92�

0.39

1.34

�0.

3313

8692

6_at

NM

_053

607

Acy

l-CoA

synt

heta

selo

ng-c

hain

fam

ilym

embe

r5

Acs

l51.

67�

0.11

1.41

�0.

152.

35�

0.13

2.17

�0.

1513

6785

4_at

NM

_016

987

AT

Pci

trat

ely

ase

Acl

y0.

61�

0.06

0.66

�0.

020.

61�

0.01

0.71

�0.

0613

9871

6_at

BG

6708

22C

arni

tine

palm

itoy

ltra

nsfe

rase

1a,

liver

Cpt

1a0.

96�

0.41

0.79

�0.

230.

47�

0.04

0.43

�0.

1213

8288

2_x_

atA

A96

3228

Car

niti

nepa

lmit

oylt

rans

fera

se1a

,liv

erC

pt1a

0.78

�0.

040.

78�

0.06

0.51

�0.

070.

44�

0.15

1392

166_

atB

E09

9838

Car

niti

nepa

lmit

oylt

rans

fera

se1a

,liv

erC

pt1a

0.77

�0.

190.

83�

0.13

0.52

�0.

030.

48�

0.15

1397

700_

x_at

BG

6708

22C

arni

tine

palm

itoy

ltra

nsfe

rase

1a,

liver

Cpt

1a0.

83�

0.47

0.91

�0.

180.

55�

0.05

0.40

�0.

1313

8692

7_at

NM

_012

930

Car

niti

nepa

lmit

oylt

rans

fera

se2

Cpt

20.

29�

0.08

0.31

�0.

030.

17�

0.04

0.21

�0.

0913

6774

0_at

M14

400

Cea

tine

kina

se,

brai

nC

kb0.

19�

0.07

0.15

�0.

060.

12�

0.03

0.09

�0.

0913

9056

6_a_

atB

I301

453

Cre

atin

eki

nase

,m

itoc

hond

rial

1,ub

iqui

tous

Ckm

t16.

60�

0.94

5.46

�1.

916.

07�

1.56

3.82

�0.

7613

9153

4_at

BG

6667

35E

long

atio

nof

very

-long

-cha

infa

tty

acid

s(F

EN

1/E

lo2,

SUR

4/E

lo3,

yeas

t)-li

ke2

(pre

dict

ed)

Elo

vl2_

pred

icte

d0.

38�

0.28

0.36

�0.

270.

34�

0.04

0.39

�0.

09

1388

108_

atB

E11

6152

ELO

VL

fam

ilym

embe

r6,

elon

gati

onof

long

-cha

infa

tty

acid

s(y

east

)E

lovl

60.

43�

0.11

0.38

�0.

080.

36�

0.02

0.35

�0.

06

1367

857_

atN

M_0

5344

5F

atty

acid

desa

tura

se1

Fad

s10.

21�

0.07

0.25

�0.

070.

28�

0.03

0.27

�0.

1413

6770

7_at

NM

_017

332

Fat

tyac

idsy

ntha

seF

asn

0.33

�0.

090.

12�

0.08

0.50

�0.

020.

50�

0.14

1371

979_

atA

I170

663

Ster

olre

gula

tory

elem

ent–

bind

ing

fact

or2

(pre

dict

ed)

Sreb

f2_p

redi

cted

0.87

�0.

160.

87�

0.04

0.45

�0.

060.

43�

0.08

1389

611_

atA

A84

9857

VLD

Lre

cept

orV

ldlr

0.75

�0.

130.

34�

0.12

2.39

�0.

202.

28�

0.07

1387

455_

a_at

NM

_013

155

VLD

Llip

opro

tein

rece

ptor

Vld

lr0.

63�

0.15

0.50

�0.

092.

79�

0.25

2.35

�0.

09C

hem

okin

es/c

ytok

ines

/adh

esio

nm

olec

ules

1367

973_

atN

M_0

3153

0C

hem

okin

e(C

-Cm

otif

)lig

and

2C

cl2

962.

7�

378.

598

7.1

�71

3.1

511.

9�

362.

312

4.1

�16

3.7

1369

814_

atA

F05

3312

Che

mok

ine

(C-C

mot

if)

ligan

d20

/C

cl20

168.

9�

15.5

862

.2�

119.

213

9.7

�15

.95

14.8

6�

5.29

1369

983_

atN

M_0

3111

6C

hem

okin

e(C

-Cm

otif

)lig

and

5C

cl5

0.60

�1.

5577

.43

�32

.63

4.42

�2.

9418

2.1

�12

1.9

1379

935_

atB

F41

9899

Che

mok

ine

(C-C

mot

if)

ligan

d7

Ccl

713

1.3

�45

.08

128.

0�

74.8

559

.30

�6.

367.

41�

2.48

1387

316_

atN

M_0

3084

5C

hem

okin

e(C

-X-C

mot

if)

ligan

d1

(GR

O-a

lpha

)C

xcl1

392.

4�

101.

836

.4�

155.

418

9.8

�84

.53

6.64

�3.

3713

7206

4_at

BI2

9638

5Si

mila

rto

chem

okin

e(C

-X-C

mot

if)

ligan

d16

Cxc

l16

15.7

1�

2.78

19.8

7�

3.52

17.0

6�

1.37

24.6

6�

2.03

1368

760_

atN

M_0

3153

0C

hem

okin

e(C

-X-C

mot

if)

ligan

d2/

Cxc

l228

2.1

�61

.17

15.9

1�

203.

947

.3�

21.7

21.

54�

0.40

1373

544_

atA

I170

387

Che

mok

ine

(C-X

-Cm

otif

)lig

and

9C

xcl9

238.

2�

232.

758

8.0

�33

9.3

199.

8�

88.6

288.

0�

161.

213

8245

4_at

AI0

4422

2C

hem

okin

e(C

-X-C

mot

if)

ligan

d9

Cxc

l929

0.5

�17

2.6

913.

8�

269.

619

7.2

�21

6.5

334.

7�

398.

913

8720

2_at

NM

_012

967

Inte

rcel

lula

rad

hesi

onm

olec

ule

1Ic

am1

192.

9�

29.9

354

1.5

�11

0.6

93.2

�29

.84

152.

8�

32.9

213

6837

5_a_

atA

F01

5718

IL-1

5Il

1510

.36

�0.

4532

.08

�1.

366.

84�

1.37

20.4

2�

7.31

F. ORTIS AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 59, FEBRUARY 2010 361

Page 5: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

TA

BLE

1C

onti

nued

Pro

beG

enB

ank

Gen

ena

me/

func

tion

algr

oup

Sym

bol

6h

24h

IL�

IFN

TN

F�

IFN

IL�

IFN

TN

F�

IFN

1368

474_

atN

M_0

1288

9V

ascu

lar

cell

adhe

sion

mol

ecul

e1

Vca

m1

3.13

�0.

9534

.83

�5.

742.

50�

0.86

28.3

7�

12.4

5IF

N-�

sign

alin

g

1369

956_

atN

M_0

5378

3IF

N-�

rece

ptor

1If

ngr

1.47

�0.

341.

46�

0.28

1.81

�0.

261.

74�

0.15

1368

073_

atN

M_0

1259

1IR

F-1

Irf1

66.3

9�

15.0

910

5.83

�19

.34

19.8

5�

3.56

34.7

6�

2.17

1371

560_

atA

A89

3384

IRF

-3Ir

f30.

81�

0.09

0.87

�0.

230.

70�

0.05

0.58

�0.

0613

8356

4_at

BF

4110

36IR

F-

7Ir

f790

.31

�27

.87

427.

28�

111.

8822

.64

�11

.73

104.

71�

53.3

413

7209

7_at

BF

2842

62IR

F-8

Irf8

81.2

6�

2.01

57.6

3�

16.5

912

.28

�2.

733.

57�

2.74

1375

796_

atB

I300

770

IFN

-�–i

nduc

edG

TP

ase

Igtp

32.1

8�

11.5

845

.65

�10

.40

7.96

�4.

7611

.24

�9.

0413

7399

2_at

AI4

0844

0Si

mila

rto

IFN

-indu

cibl

eG

TP

ase

MG

C10

8823

140.

5�

42.7

748

2.2

�13

1.4

144.

4�

18.5

437

0.8

�95

.71

1377

950_

atA

A95

5213

Sim

ilar

toIF

N-in

duci

ble

GT

Pas

eR

GD

1309

362

63.4

�13

.823

5.0

�73

.529

.1�

11.5

142.

2�

43.5

1368

835_

atA

W43

4718

STA

T1

Stat

114

.50

�1.

7519

.51

�1.

895.

50�

0.69

8.67

�1.

4213

7275

7_at

BM

3868

75ST

AT

1St

at1

7.38

�0.

858.

27�

0.61

4.74

�0.

497.

28�

1.02

1387

354_

atN

M_0

3261

2ST

AT

1St

at1

28.1

3�

9.16

23.8

0�

8.68

9.90

�1.

9113

.39

�4.

0913

8957

1_at

BG

6663

68ST

AT

2St

at2

17.9

8�

5.43

25.5

0�

2.68

32.8

3�

16.2

349

.37

�12

.41

1370

224_

atB

E11

3920

STA

T3

Stat

32.

66�

0.43

2.86

�0.

252.

92�

0.66

1.91

�0.

7313

7178

1_at

BI2

8586

3ST

AT

3St

at3

2.02

�0.

152.

96�

0.34

1.62

�0.

221.

66�

0.28

1387

876_

atA

I177

626

STA

T5B

Stat

5b1.

33�

0.39

1.23

�0.

113.

24�

1.02

1.41

�0.

3113

8347

8_at

BG

6715

04Ja

nus

kina

se1

Jak1

1.06

�0.

160.

98�

0.09

2.71

�0.

112.

74�

0.45

1384

060_

atB

G66

3208

Janu

ski

nase

1Ja

k11.

31�

0.04

0.97

�0.

233.

71�

0.38

3.38

�0.

4513

6885

6_at

NM

_031

514

Janu

ski

nase

2Ja

k29.

74�

5.58

17.8

8�

3.18

4.23

�1.

485.

79�

1.98

1380

110_

atA

I229

643

Janu

ski

nase

2Ja

k212

.28

�1.

1614

.90

�2.

817.

27�

0.75

9.56

�1.

5413

6825

1_at

NM

_012

855

Janu

ski

nase

3Ja

k31.

46�

0.12

3.24

�0.

850.

99�

0.24

1.53

�0.

2013

7666

6_at

AI1

7086

4Su

ppre

ssor

ofcy

toki

nesi

gnal

ing

6(p

redi

cted

)So

cs6_

pred

icte

d0.

90�

0.17

1.28

�0.

251.

51�

0.08

1.37

�0.

0913

9148

4_at

BF

2847

86Su

ppre

ssor

ofcy

toki

nesi

gnal

ing

7(p

redi

cted

)So

cs7_

pred

icte

d1.

32�

0.06

1.53

�0.

131.

25�

0.03

1.41

�0.

18N

F-�

Bre

gula

tion

1383

474_

atB

I274

988

IL-1

rece

ptor

–ass

ocia

ted

kina

se2

Irak

23.

55�

0.66

3.57

�1.

513.

01�

0.20

1.34

�0.

2313

7096

8_at

AA

8588

01N

F�

light

-cha

inge

neen

hanc

erin

B-c

ells

1,p1

05N

fkb1

19.0

6�

3.30

17.7

6�

0.97

7.67

�1.

597.

48�

1.72

1389

538_

atA

W67

2589

NF

�lig

ht-c

hain

gene

enha

ncer

inB

-cel

lsin

hibi

tor,

�N

fkbi

a55

.16

�26

.23

82.8

4�

18.9

325

.93

�8.

4220

.09

�5.

05

1367

943_

atN

M_0

3086

7N

F�

light

-cha

inge

neen

hanc

erin

B-c

ells

inhi

bito

r,�

Nfk

bib

4.40

�1.

975.

78�

0.95

7.26

�2.

315.

67�

1.34

1375

989_

a_at

AI1

7036

2N

F�

light

poly

pept

ide

gene

enha

ncer

inB

-cel

ls2,

p49/

p100

Nfk

b212

.32

�2.

8418

.22

�5.

0210

.63

�6.

5511

.33

�5.

34

1376

835_

atB

I293

600

NF

�lig

htpo

lype

ptid

ege

neen

hanc

erin

B-c

ells

inhi

bito

r,ε

Nfk

bie

11.2

1�

3.38

41.2

7�

4.76

2.04

�1.

407.

89�

1.86

1378

032_

atA

I176

265

NF

�lig

htpo

lype

ptid

ege

neen

hanc

erin

B-c

ells

inhi

bito

r,

(pre

dict

ed)

Nfk

biz_

pred

icte

d12

.91

�2.

159.

86�

1.36

13.4

8�

2.26

10.7

1�

1.32

Oth

ers

tran

scri

ptio

nfa

ctor

s

1379

368_

atA

I237

606

B-c

ell

leuk

emia

/lym

phom

a6

(pre

dict

ed)

Bcl

6_pr

edic

ted

1.90

�0.

902.

23�

0.86

9.63

�1.

5110

.86

�2.

2213

8559

2_at

BI2

8938

6B

cl6

inte

ract

ing

core

pres

sor

(pre

dict

ed)

Bco

r_pr

edic

ted

1.34

�0.

041.

53�

0.11

2.48

�0.

521.

76�

0.07

1391

632_

atA

A96

4568

CC

AA

T/e

nhan

cer

bind

ing

prot

ein

(C/E

BP

),

Ceb

pd0.

74�

0.05

0.60

�0.

032.

21�

0.63

1.74

�0.

0913

8734

3_at

NM

_013

154

CC

AA

T/e

nhan

cer

bind

ing

prot

ein

(C/E

BP

),

Ceb

pd2.

34�

0.36

1.11

�0.

1010

.82

�3.

475.

42�

0.99

1375

043_

atB

F41

5939

FB

Jm

urin

eos

teos

arco

ma

vira

lon

coge

neho

mol

ogF

os0.

33�

0.08

0.70

�0.

180.

33�

0.07

0.29

�0.

28

GENE NETWORKS AND �-CELL APOPTOSIS

362 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org

Page 6: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

TA

BLE

1C

onti

nued

Pro

beG

enB

ank

Gen

ena

me/

func

tion

algr

oup

Sym

bol

6h

24h

IL�

IFN

TN

F�

IFN

IL�

IFN

TN

F�

IFN

1388

761_

atA

I180

339

His

tone

deac

etyl

ase

1(p

redi

cted

)H

dac1

_pre

dict

ed0.

59�

0.04

0.72

�0.

090.

34�

0.05

0.56

�0.

0213

9682

0_at

AW

5301

95H

isto

nede

acet

ylas

e1

(pre

dict

ed)

Hda

c1_p

redi

cted

0.97

�0.

160.

99�

0.24

0.54

�0.

020.

56�

0.03

1370

908_

atA

A89

2297

His

tone

deac

etyl

ase

2H

dac2

0.61

�0.

040.

97�

0.16

0.73

�0.

050.

82�

0.02

1387

076_

atN

M_0

2435

9H

IF-1

,�

-sub

unit

Hif

1a2.

02�

0.17

1.58

�0.

312.

07�

0.20

2.16

�0.

1913

6968

1_at

NM

_017

339

ISL1

tran

scri

ptio

nfa

ctor

,LI

M/h

omeo

dom

ain

1Is

l10.

53�

0.08

0.14

�0.

050.

31�

0.23

0.29

�0.

0613

9313

8_at

BE

3293

77Ju

nD

prot

o-on

coge

neJu

nd1.

31�

0.10

1.43

�0.

052.

25�

0.23

1.83

�0.

2313

6951

6_at

NM

_022

852

Pan

crea

tic

and

duod

enal

hom

eobo

xge

ne1

Pdx

11.

01�

0.23

0.21

�0.

030.

49�

0.06

0.25

�0.

2113

6924

2_at

NM

_013

001

Pai

red

box

gene

6P

ax6

0.50

�0.

010.

52�

0.08

0.35

�0.

040.

53�

0.05

1374

404_

atB

I288

619

Pro

to-o

ncog

ene

c-ju

nJu

n2.

17�

1.36

2.44

�1.

094.

95�

0.94

2.76

�0.

3613

6978

8_s_

atN

M_0

2183

5P

roto

-onc

ogen

ec-

jun

Jun

4.71

�1.

356.

30�

1.77

7.39

�0.

136.

65�

1.20

1389

528_

s_at

BI2

8861

9P

roto

-onc

ogen

ec-

jun

Jun

4.00

�0.

933.

05�

0.68

9.12

�1.

527.

04�

2.01

Hor

mon

es

1387

235_

atN

M_0

2165

5C

hrom

ogra

nin

AC

hga

0.64

�0.

160.

65�

0.01

0.30

�0.

020.

27�

0.05

1368

034_

atN

M_0

1252

6C

hrom

ogra

nin

BC

hgb

0.79

�0.

130.

66�

0.02

0.30

�0.

020.

35�

0.04

1369

888_

atN

M_0

1270

7G

luca

gon

Gcg

0.71

�0.

040.

77�

0.06

0.10

�0.

010.

16�

0.06

1387

815_

atN

M_0

1912

9In

sulin

1In

s10.

94�

0.09

1.04

�0.

120.

75�

0.06

0.79

�0.

0213

7007

7_at

NM

_019

130

Insu

lin2

Ins2

0.86

�0.

070.

99�

0.09

0.68

�0.

030.

75�

0.01

1387

660_

atM

2539

0Is

let

amyl

oid

poly

pept

ide

Iapp

0.86

�0.

100.

85�

0.02

0.48

�0.

030.

60�

0.06

1368

559_

atN

M_0

1709

1P

ropr

otei

nco

nver

tase

subt

ilisi

n/ke

xin

type

1P

csk1

0.48

�0.

180.

48�

0.07

0.18

�0.

030.

27�

0.05

1387

247_

atM

8374

5P

ropr

otei

nco

nver

tase

subt

ilisi

n/ke

xin

type

1P

csk1

0.41

�0.

140.

56�

0.06

0.17

�0.

020.

23�

0.14

1387

155_

atN

M_0

1274

6P

ropr

otei

nco

nver

tase

subt

ilisi

n/ke

xin

type

2P

csk2

0.78

�0.

080.

76�

0.08

0.45

�0.

020.

58�

0.04

1397

662_

atB

F39

5791

Pro

prot

ein

conv

erta

sesu

btili

sin/

kexi

nty

pe2

Pcs

k20.

92�

0.17

1.05

�0.

080.

26�

0.04

0.55

�0.

1413

6777

8_at

NM

_019

331

Pro

prot

ein

conv

erta

sesu

btili

sin/

kexi

nty

pe3

Pcs

k30.

80�

0.06

0.51

�0.

010.

67�

0.05

0.57

�0.

0313

6776

2_at

NM

_012

659

Som

atos

tati

nSs

t0.

48�

0.06

0.48

�0.

040.

08�

0.01

0.08

�0.

08H

orm

one

rece

ptor

s

1369

787_

atN

M_0

1268

8C

hole

cyst

okin

inA

rece

ptor

Cck

ar0.

23�

0.04

0.25

�0.

040.

06�

0.01

0.11

�0.

1213

6848

1_at

NM

_012

714

Gas

tric

inhi

bito

rypo

lype

ptid

ere

cept

orG

ipr

0.55

�0.

050.

64�

0.16

0.14

�0.

020.

12�

0.08

1369

699_

atN

M_0

1272

8G

LP-1

rece

ptor

Glp

1r0.

47�

0.34

0.96

�0.

260.

32�

0.04

0.39

�0.

0813

6892

4_at

NM

_017

094

Gro

wth

horm

one

rece

ptor

Ghr

0.41

�0.

070.

49�

0.17

0.31

�0.

060.

44�

0.07

1370

384_

a_at

M57

668

Pro

lact

inre

cept

orP

rlr

0.36

�0.

120.

38�

0.18

0.17

�0.

060.

23�

0.20

1370

789_

a_at

L480

60P

rola

ctin

rece

ptor

Prl

r0.

15�

0.08

0.38

�0.

140.

21�

0.01

0.21

�0.

0513

7694

4_at

AI4

0716

3P

rola

ctin

rece

ptor

Prl

r0.

41�

0.03

0.49

�0.

030.

17�

0.04

0.35

�0.

1113

9261

2_at

AW

1429

62P

rola

ctin

rece

ptor

Prl

r0.

37�

0.11

0.50

�0.

120.

13�

0.03

0.14

�0.

1013

8717

7_at

NM

_017

238

Vas

oact

ive

inte

stin

alpe

ptid

ere

cept

or2

Vip

r20.

35�

0.07

0.36

�0.

060.

13�

0.02

0.11

�0.

03F

ree

radi

cal

scav

ange

r/D

NA

dam

age

1367

995_

atN

M_0

1252

0C

atal

ase

Cat

0.79

�0.

090.

72�

0.09

1.96

�0.

142.

12�

0.29

1367

774_

atN

M_0

3150

9G

luta

thio

neS

-tra

nsfe

rase

A3

Gst

a30.

58�

0.19

0.45

�0.

166.

34�

1.77

1.78

�0.

1313

8983

2_at

BE

1134

59G

luta

thio

neS

-tra

nsfe

rase

,�

1G

sto1

0.62

�0.

120.

66�

0.04

1.54

�0.

151.

59�

0.05

1387

023_

atN

M_0

3115

4G

luta

thio

neS

-tra

nsfe

rase

,�

type

3G

stm

30.

38�

0.06

0.38

�0.

040.

14�

0.01

0.12

�0.

0513

8812

2_at

X02

904

Glu

tath

ione

S-t

rans

fera

se,

2

Gst

p20.

83�

0.32

0.52

�0.

144.

61�

1.84

6.30

�2.

1013

7201

6_at

BI2

8797

8G

row

thar

rest

and

DN

Ada

mag

e–in

duci

ble

45�

Gad

d45b

44.5

2�

6.02

21.0

1�

34.6

413

1.03

�53

.38

38.8

1�

10.3

113

8879

2_at

AI5

9942

3G

row

thar

rest

and

DN

Ada

mag

e–in

duci

ble

45�

Gad

d45g

3.85

�1.

062.

43�

0.88

3.53

�0.

481.

89�

0.54

1388

267_

a_at

M24

327

Met

allo

thio

nein

1aM

t1a

2.41

�1.

743.

61�

1.23

26.3

6�

4.49

30.5

2�

4.33

1371

237_

a_at

AF

4113

18M

etal

loth

ione

in1a

Mt1

a3.

02�

1.19

4.03

�1.

0736

.70

�11

.37

40.6

4�

9.73

1374

911_

atA

W25

1534

Oxi

dati

vest

ress

resp

onsi

vege

neR

GD

1303

142

1.01

�1.

071.

00�

0.34

8.43

�2.

516.

51�

0.66

F. ORTIS AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 59, FEBRUARY 2010 363

Page 7: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

TA

BLE

1C

onti

nued

Pro

beG

enB

ank

Gen

ena

me/

func

tion

algr

oup

Sym

bol

6h

24h

IL�

IFN

TN

F�

IFN

IL�

IFN

TN

F�

IFN

1372

941_

atB

I273

897

p53

and

DN

Ada

mag

ere

gula

ted

1P

drg1

2.01

�0.

501.

30�

0.03

2.61

�0.

182.

07�

0.07

1380

071_

atB

I285

978

Pol

y(A

DP

-rib

ose)

poly

mer

ase

fam

ily,

mem

ber

12(p

redi

cted

)P

arp1

2_pr

edic

ted

7.04

�1.

258.

87�

0.94

2.89

�0.

134.

96�

0.65

1383

251_

atA

W52

4533

Pol

y(A

DP

-rib

ose)

poly

mer

ase

fam

ily,

mem

ber

2(p

redi

cted

)P

arp2

_pre

dict

ed0.

99�

0.18

0.77

�0.

201.

22�

0.09

1.40

�0.

13

1376

144_

atA

A81

9679

Pol

y(A

DP

-rib

ose)

poly

mer

ase

fam

ily,

mem

ber

9(p

redi

cted

)P

arp9

_pre

dict

ed27

.68

�9.

8248

.25

�13

.19

15.1

7�

1.94

22.4

0�

2.13

1370

173_

atB

G67

1549

Supe

roxi

dedi

smut

ase

2,m

itoc

hond

rial

Sod2

6.74

�0.

417.

02�

0.19

7.51

�1.

275.

53�

0.35

1370

172_

atA

A89

2254

Supe

roxi

dedi

smut

ase

2,m

itoc

hond

rial

Sod2

8.69

�0.

509.

23�

0.82

13.0

7�

0.94

9.18

�0.

63E

ndop

lasm

icre

ticu

lum

stre

ss/a

popt

osis

rela

ted

1369

268_

atN

M_0

1291

2A

TF

3A

tf3

15.7

3�

3.69

23.2

7�

4.00

53.8

9�

24.8

726

.59

�27

.09

1367

624_

atN

M_0

2440

3A

TF

4A

tf4

1.33

�0.

081.

07�

0.05

2.32

�0.

272.

16�

0.12

1368

066_

atN

M_0

5381

2B

CL2

-ant

agon

ist/

kille

r1

Bak

16.

45�

3.37

5.77

�0.

933.

83�

0.76

6.81

�2.

7913

6912

2_at

AF

2359

93B

cl2-

asso

ciat

edX

prot

ein

Bax

1.17

�0.

061.

24�

0.06

2.34

�0.

222.

32�

0.22

1377

759_

atB

G66

6928

BH

3-in

tera

ctin

gdo

mai

nde

ath

agon

ist

Bid

6.29

�1.

663.

48�

1.36

9.39

�1.

304.

30�

0.71

1370

283_

atM

1405

0B

ipH

spa5

0.89

�0.

100.

91�

0.05

1.46

�0.

031.

35�

0.06

1370

283_

atM

1405

0B

ipH

spa5

0.89

�0.

100.

91�

0.05

1.46

�0.

031.

35�

0.06

1381

173_

atB

G37

5010

Cas

pase

4,ap

opto

sis-

rela

ted

cyst

eine

pept

idas

eC

asp4

17.9

7�

7.54

15.8

9�

5.15

5.70

�0.

8917

.31

�4.

9813

8781

8_at

NM

_053

736

Cas

pase

4,ap

opto

sis-

rela

ted

cyst

eine

pept

idas

eC

asp4

18.8

9�

7.24

26.1

3�

13.1

141

.24

�27

.83

65.6

1�

19.2

913

8917

0_at

BF

2837

54C

aspa

se7

Cas

p70.

40�

0.12

0.31

�0.

180.

57�

0.09

0.72

�0.

0813

6752

9_at

BE

1139

89D

erlin

1R

GD

1311

835

0.81

�0.

060.

78�

0.08

1.52

�0.

201.

44�

0.08

1374

581_

atB

M38

4392

Der

lin1

RG

D13

1183

50.

91�

0.08

0.80

�0.

091.

73�

0.18

1.43

�0.

1313

8961

5_at

BI2

8480

1D

erlin

1R

GD

1311

835

0.58

�0.

190.

55�

0.02

2.34

�0.

252.

07�

0.17

1369

590_

a_at

NM

_024

134

Cho

pD

dit3

1.66

�0.

271.

19�

0.33

7.86

�1.

117.

10�

0.91

1383

011_

atA

I501

182

Euk

aryo

tic

tran

slat

ion

init

iati

onfa

ctor

2AE

if2a

1.62

�0.

091.

62�

0.25

1.86

�0.

201.

70�

0.05

1388

898_

atA

I236

601

Hea

tsh

ock

105

kDa/

110

kDa

prot

ein

1H

sph1

0.71

�0.

080.

74�

0.15

2.01

�0.

661.

50�

0.20

1385

620_

atB

F52

5282

Hea

tsh

ock

105

kDa/

110

kDa

prot

ein

1H

sph1

0.54

�0.

070.

31�

0.15

3.56

�0.

851.

35�

0.20

1388

721_

atB

G38

0282

Hea

tsh

ock

22kD

apr

otei

n8

Hsp

b835

.81

�13

.54

12.6

7�

5.18

9.02

�2.

865.

92�

2.36

1368

247_

atN

M_0

3197

1H

eat

shoc

k70

kDpr

otei

n1A

Hsp

a1a

1.21

�0.

161.

25�

0.23

5.57

�1.

021.

52�

1.14

1370

912_

atB

I278

231

Hea

tsh

ock

70kD

prot

ein

1B(m

appe

d)H

spa1

b1.

19�

0.26

1.17

�0.

356.

15�

1.09

2.03

�1.

4813

8885

1_at

BI2

8228

1H

eat

shoc

k70

kDa

prot

ein

9A(p

redi

cted

)H

spa9

a_pr

edic

ted

0.80

�0.

070.

69�

0.03

1.97

�0.

101.

91�

0.12

1386

894_

atN

M_0

2222

9H

eat

shoc

kpr

otei

n1

(cha

pero

nin)

Hsp

d10.

52�

0.01

0.49

�0.

011.

37�

0.11

1.24

�0.

0513

7270

1_at

AI2

3759

7H

eat

shoc

kpr

otei

n1,

�H

spca

0.76

�0.

120.

76�

0.18

2.79

�0.

201.

63�

0.26

1388

850_

atB

G67

1521

Hea

tsh

ock

prot

ein

1,�

Hsp

ca0.

67�

0.06

0.79

�0.

093.

06�

0.68

1.66

�0.

2713

9824

0_at

NM

_024

351

Hea

tsh

ock

prot

ein

8H

spa8

0.66

�0.

060.

68�

0.08

1.51

�0.

101.

21�

0.05

1368

195_

atN

M_1

3441

9H

spb-

asso

ciat

edpr

otei

n1

Hsp

bap1

0.81

�0.

771.

00�

0.25

3.89

�0.

302.

22�

0.34

1370

174_

atB

I284

349

Mye

loid

diff

eren

tiat

ion

prim

ary

resp

onse

gene

116

Myd

116

6.11

�0.

816.

55�

4.61

21.4

6�

2.39

9.67

�6.

56

1382

615_

atB

I284

366

Sec6

1�

1su

buni

t(S

.ce

revi

siae

)Se

c61a

10.

76�

0.18

0.97

�0.

090.

75�

0.29

0.54

�0.

0413

7565

9_at

BG

3815

29Se

c61,

�-s

ubun

it2

(S.

cere

visi

ae)

(pre

dict

ed)

Sec6

1a2_

pred

icte

d0.

77�

0.06

0.84

�0.

030.

71�

0.04

0.81

�0.

0113

7253

3_at

AI1

7579

0Si

mila

rto

mK

IAA

0212

prot

ein

(pre

dict

ed)

edem

1.35

�0.

122.

06�

0.14

1.44

�0.

051.

51�

0.16

1370

695_

s_at

AB

0209

67T

ribb

les

hom

olog

3(D

roso

phila

)T

rib3

1.88

�0.

760.

88�

0.13

8.15

�3.

617.

74�

0.85

1370

694_

atA

B02

0967

Tri

bble

sho

mol

og3

(Dro

soph

ila)

Tri

b31.

94�

1.04

1.16

�0.

239.

76�

1.28

7.81

�0.

4713

8632

1_s_

atH

3128

7T

ribb

les

hom

olog

3(D

roso

phila

)T

rib3

2.12

�0.

611.

30�

0.16

14.6

6�

2.32

10.4

7�

1.31

1369

065_

a_at

NM

_017

290

Serc

a2A

tp2a

20.

40�

0.08

0.45

�0.

070.

21�

0.02

0.25

�0.

07

GENE NETWORKS AND �-CELL APOPTOSIS

364 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org

Page 8: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

TA

BLE

1C

onti

nued

Pro

beG

enB

ank

Gen

ena

me/

func

tion

algr

oup

Sym

bol

6h

24h

IL�

IFN

TN

F�

IFN

IL�

IFN

TN

F�

IFN

1370

426_

a_at

AI1

7549

2Se

rca2

Atp

2a2

0.51

�0.

060.

52�

0.03

0.63

�0.

060.

65�

0.03

Cel

lcy

cle

1390

343_

atA

A99

8893

Cyc

linC

Ccn

c0.

61�

0.15

0.59

�0.

030.

48�

0.01

0.68

�0.

0413

8910

1_at

BE

1203

40C

yclin

CC

cnc

0.90

�0.

140.

81�

0.21

1.33

�0.

211.

62�

0.04

1371

643_

atA

W14

3798

Cyc

linD

1C

cnd1

0.48

�0.

120.

69�

0.14

0.43

�0.

120.

55�

0.17

1369

935_

atN

M_0

1276

6C

yclin

D3

Ccn

d30.

38�

0.11

0.52

�0.

020.

62�

0.12

0.62

�0.

1013

7195

3_at

AI4

0830

9C

yclin

G2

(pre

dict

ed)

Ccn

g2_p

redi

cted

4.59

�0.

724.

51�

1.85

2.45

�0.

332.

54�

0.37

1388

370_

atA

A94

5706

Cyc

linI

(pre

dict

ed)

Ccn

i_pr

edic

ted

0.93

�0.

061.

05�

0.06

0.74

�0.

020.

88�

0.04

1368

050_

atN

M_0

5366

2C

yclin

L1C

cnl1

1.61

�0.

351.

96�

0.55

2.76

�0.

471.

80�

0.76

1390

815_

atB

F28

2870

Cyc

linM

1(p

redi

cted

)C

nnm

1_pr

edic

ted

1.08

�0.

110.

76�

0.17

0.49

�0.

040.

38�

0.05

1391

270_

atB

E11

2177

Cyc

linM

3(p

redi

cted

)C

nnm

3_pr

edic

ted

0.37

�0.

050.

33�

0.05

0.19

�0.

040.

37�

0.07

1384

214_

a_at

AI0

4545

9C

yclin

T2

(pre

dict

ed)

Ccn

t2_p

redi

cted

0.50

�0.

230.

75�

0.30

0.27

�0.

060.

42�

0.09

1390

470_

atB

E10

7044

Cyc

linT

2(p

redi

cted

)C

cnt2

_pre

dict

ed1.

73�

0.29

1.21

�0.

120.

53�

0.09

0.71

�0.

16Sp

licin

gm

achi

nery

Seri

ne-r

ich

and

seri

ne-r

ich–

rela

ted

prot

ein

1376

252_

atA

I145

784

Splic

ing

fact

or,

argi

nine

/ser

ine-

rich

3(S

Rp2

0)(p

redi

cted

)Sf

rs3_

pred

icte

d1.

03�

0.08

0.76

�0.

270.

39�

0.02

0.50

�0.

12

1379

010_

atA

A95

6727

Splic

ing

fact

or,

argi

nine

/ser

ine-

rich

3(S

Rp2

0)(p

redi

cted

)Sf

rs3_

pred

icte

d2.

11�

0.43

1.18

�0.

283.

98�

0.87

2.05

�0.

67

1376

594_

atA

W52

4517

Sim

ilar

tosp

licin

gfa

ctor

,ar

gini

ne/s

erin

e-ri

ch1

(ASF

/SF

2)V

ezf1

_pre

dict

ed1.

73�

0.60

1.45

�0.

233.

16�

0.36

3.44

�0.

51

1383

537_

atB

F52

2715

Sim

ilar

tosp

licin

gfa

ctor

,ar

gini

ne/s

erin

e-ri

ch1

(ASF

/SF

2)V

ezf1

_pre

dict

ed1.

84�

0.46

1.79

�0.

301.

67�

0.29

1.80

�0.

11

1371

838_

atA

I411

155

Sim

ilar

tosp

licin

gfa

ctor

,ar

gini

ne/s

erin

e-ri

ch2

Sfrs

21.

11�

0.07

1.13

�0.

121.

43�

0.04

1.33

�0.

0413

7183

9_at

AA

8193

69Si

mila

rto

splic

ing

fact

or,

argi

nine

/ser

ine-

rich

2Sf

rs2

0.63

�0.

130.

66�

0.07

1.59

�0.

101.

33�

0.17

1368

992_

a_at

AI1

0400

5Sp

licin

gfa

ctor

,ar

gini

ne/s

erin

e-ri

ch5

Sfrs

50.

56�

0.09

0.77

�0.

170.

53�

0.03

0.57

�0.

0613

7199

9_at

BI3

0364

1Sp

licin

gfa

ctor

argi

nine

/ser

ine-

rich

6(S

RP

55-2

)(i

sofo

rm2)

Sfrs

60.

37�

0.13

0.71

�0.

060.

19�

0.04

0.20

�0.

09

1381

623_

atB

F39

1476

Ssim

ilar

toSf

rs4

prot

ein

(pre

dict

ed)

Sfrs

4_pr

edic

ted

1.23

�0.

371.

59�

0.14

2.08

�0.

221.

89�

0.17

1370

188_

atA

W25

2670

Splic

ing

fact

or,

argi

nine

/ser

ine-

rich

10(t

rans

form

er2

hom

olog

,D

roso

phila

)Sf

rs10

1.02

�0.

161.

04�

0.06

2.18

�0.

291.

41�

0.13

1371

425_

atB

F39

6399

Seri

ne/a

rgin

ine

repe

titi

vem

atri

x1

(pre

dict

ed)

Srrm

1_pr

edic

ted

1.29

�0.

211.

20�

0.13

1.40

�0.

091.

37�

0.06

1383

410_

atB

I290

777

Sign

alre

cogn

itio

npa

rtic

le54

Srp5

40.

91�

0.09

0.88

�0.

061.

50�

0.13

1.21

�0.

0313

7159

6_at

AI0

0897

1R

ibon

ucle

icac

idbi

ndin

gpr

otei

nS1

Rnp

s11.

01�

0.34

0.90

�0.

121.

84�

0.08

1.54

�0.

06H

eter

ogen

eous

nucl

ear

ribo

nucl

eopr

otei

nfa

mily

1398

883_

atB

I296

284

Het

erog

eneo

usnu

clea

rri

bonu

cleo

prot

ein

A2/

B1

(pre

dict

ed)

Hnr

pa2b

1_pr

edic

ted

0.77

�0.

100.

85�

0.05

0.45

�0.

040.

53�

0.06

1371

505_

atB

G38

1750

Het

erog

eneo

usnu

clea

rri

bonu

cleo

prot

ein

CH

nrpc

1.15

�0.

031.

15�

0.06

2.25

�0.

282.

05�

0.18

1367

931_

a_at

X60

790

Pol

ypyr

imid

ine

trac

tbi

ndin

gpr

otei

n1

Ptb

p10.

67�

0.12

0.60

�0.

020.

83�

0.14

0.73

�0.

1013

7091

9_at

AI1

0346

7H

eter

ogen

eous

nucl

ear

ribo

nucl

eopr

otei

nM

Hnr

pm0.

85�

0.09

0.81

�0.

030.

79�

0.07

0.78

�0.

07O

ther

splic

ing

fact

ors

1389

975_

atB

E11

6949

ELA

V(e

mbr

yoni

cle

thal

,ab

norm

alvi

sion

,D

roso

phila

)-lik

e4

(Hu

anti

gen

D)

HuD

0.63

�0.

120.

59�

0.11

0.32

�0.

020.

33�

0.06

F. ORTIS AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 59, FEBRUARY 2010 365

Page 9: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

ing” (Table 1 and supplementary Fig. 3). For some of thesegenes and pathways, there was a different regulation byIL-1� and TNF-�, with TNF-� � IFN-� preferentiallyinducing IL-15, chemokine (CXC-motif) ligand (CXCL) 9(or Mig), CXCL10 (or IP-10), CCL5 (or RANTES), IRF-1,IRF-7, and signal transducer and activator of transcrip-tion-1 (STAT1)-�, while IL-1� preferentially upregulatedCCL2 (or MCP-1) and CXCL1 (or Gro�) (Table 1). Thesedifferences were to a large extent confirmed by real-timeRT-PCR (Fig. 2A). TNF-�–induced higher CCL5 and IRF-7expression was also confirmed at the protein level (sup-plementary Fig. 4A–C). TNF-� � IFN-� leads also to higherexpression of IFN-�, a downstream gene of IRF-7, thanIL-1� � IFN-� (supplementary Fig. 4E). TNF-�–inducedIRF-7 expression upregulates expression of IRF-1 andproinflammatory chemokines in other tissues (20,21). Useof a specific siRNA against IRF-7 induced a 90% knockdown of IRF-7, which partially prevented TNF-� � IFN-�–induced, but not IL-1� � IFN-�–induced, IRF-1, CCL5(confirmed at protein level) (supplementary Fig. 4C),IL-15, and CXCL10 expression (Fig. 2B). The role of IRF-7is apparently specific for genes preferentially induced byTNF-� � IFN-�, since CXCL1 expression, which is higherafter IL-1� � IFN-� exposure (Fig. 2), was not significantlydecreased by IRF-7 knock down (Fig. 2B). These observa-tions were confirmed by the use of a second siRNA againstIRF-7 (data not shown).Differential modulation of the citrulline-NO cycle byIL-1� and TNF-�. IL-1� � IFN-� treatment in �-cells ledto higher expression of inducible NO synthase (iNOS)(Table 1) and NO accumulation in the medium (supple-mentary Fig. 1B) than TNF-� � IFN-�. iNOS utilizesarginine as the substrate for NO formation, generatingcitrulline as a by-product. Citrulline can be used to regen-erate arginine by the citrulline-NO cycle (Fig. 3A) (22),which is regulated by argininosuccinate synthetase (ASS)expression (22). The array analysis indicated that ASS isstrongly induced by IL-1� � IFN-� but not by TNF-� �IFN-� (Table 1). In addition, IL-1� � IFN-� inhibited theexpression of arginase-1 (arg1) more efficiently thanTNF-� � IFN-� (Table 1), preserving arginine for NOformation (Fig. 3A). In line with the mRNA data, IL-1� �IFN-�, but not TNF-� � IFN-�, induced NO formation inthe absence of arginine but presence of citrulline (Fig. 3B).Cytokines decrease the expression of genes involvedin maintenance of a differentiated �-cell phenotype.We next examined the expression of a group of 14 genes(Fig. 4) previously shown to be of particular relevance forthe induction and maintenance of the differentiated phe-notype in �-cells (23). These genes are either directlyrelated to �-cell–differentiated functions (Fig. 4A) or func-tion as master regulatory transcription factors (Fig. 4B).They were all inhibited by IL-1� � IFN-� or TNF-� �IFN-�, a finding confirmed by real-time RT-PCR for se-lected genes (Fig. 4C). For many of these genes, inhibitionwas already present after 6 h of cytokine exposure (Fig. 4),suggesting an early and specific effect.Cytokines decrease the expression of genes encodingenzymes of the Krebs cycle. Exposure of �-cells to IL-1� �IFN-� or TNF-� � IFN-� decreased to a similar extentexpression of genes encoding enzymes of the Krebs cycle(Table 1, glucose metabolism). This was confirmed byreal-time RT-PCR for seven of eight genes present in theKrebs cycle, with a more important inhibitory effect at 24 h(Fig. 5A). The promoter region of these genes was ana-lyzed by an in silico approach, and a binding site for (ATF4T

AB

LE1

Con

tinu

ed

Pro

beG

enB

ank

Gen

ena

me/

func

tion

algr

oup

Sym

bol

6h

24h

IL�

IFN

TN

F�

IFN

IL�

IFN

TN

F�

IFN

1394

546_

atA

I556

229

ELA

V(e

mbr

yoni

cle

thal

,ab

norm

alvi

sion

,D

roso

phila

)-lik

e4

(Hu

anti

gen

D)

HuD

0.82

�0.

370.

59�

0.03

0.10

�0.

010.

21�

0.09

1395

083_

atA

A92

6313

Neu

ro-o

ncol

ogic

alve

ntra

lan

tige

n1

Nov

a10.

34�

0.18

0.68

�0.

180.

22�

0.03

0.37

�0.

1113

8847

6_at

AI1

0139

1T

ial1

cyto

toxi

cgr

anul

e–as

soci

ated

RN

Abi

ndin

gpr

otei

n–lik

e1

(map

ped)

Tia

l11.

15�

0.21

1.03

�0.

211.

39�

0.02

1.32

�0.

05

1374

463_

atA

I172

068

Qua

king

hom

olog

,K

Hdo

mai

nR

NA

bind

ing

(mou

se)

Qki

1.33

�0.

041.

07�

0.22

1.87

�0.

081.

57�

0.16

1370

899_

atA

I599

699

Splic

ing

fact

orpr

olin

e/gl

utam

ine

rich

(pol

ypyr

imid

ine

trac

tbi

ndin

gpr

otei

nas

soci

ated

)Sf

pq0.

42�

0.05

0.48

�0.

020.

40�

0.08

0.30

�0.

13

1386

896_

atA

F39

3783

KH

dom

ain

cont

aini

ng,

RN

Abi

ndin

g,si

gnal

tran

sduc

tion

–ass

ocia

ted

1K

hdrb

s10.

88�

0.12

0.82

�0.

042.

15�

0.19

1.66

�0.

17

1398

773_

atN

M_1

3040

5K

Hdo

mai

nco

ntai

ning

,R

NA

bind

ing,

sign

altr

ansd

ucti

on–a

ssoc

iate

d1

Khd

rbs1

0.96

�0.

180.

96�

0.04

1.31

�0.

061.

39�

0.05

1372

496_

atB

G37

1538

Rib

onuc

leop

rote

inP

TB

-bin

ding

1(p

rote

inra

ver-

1)R

aver

1h0.

55�

0.24

0.68

�0.

073.

26�

0.38

1.85

�0.

29

1371

367_

atB

E10

7459

TA

RD

NA

-bin

ding

prot

ein

43(T

DP

-43)

Tar

dbp

0.93

�0.

070.

83�

0.10

0.40

�0.

030.

51�

0.05

Dat

aar

em

eans

�SE

ofth

ree

inde

pend

ent

expe

rim

ents

and

are

expr

esse

das

fold

chan

geve

rsus

cont

rolc

ells

,stu

died

atth

esa

me

tim

epo

ints

.Gen

eex

pres

sion

was

cons

ider

edm

odifi

edby

cyto

kine

sw

hen

P�

0.02

(pai

red

tte

st)

and

expr

essi

onle

vel

�1.

5-fo

ldhi

gher

orlo

wer

asco

mpa

red

wit

hco

ntro

lco

ndit

ions

.P

rim

ary

rat

�-c

ells

wer

ele

ftun

trea

ted

orex

pose

dto

IL1�

�IF

N-�

(IL

�IF

N)

orT

NF

-��

IFN

-�(T

NF

�IF

N)

for

6an

d24

h.

GENE NETWORKS AND �-CELL APOPTOSIS

366 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org

Page 10: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

identified as overrepresentative in this set of genes. ATF4is induced by cytokines (Table 1 and Fig. 5B) and has animportant role in the unfolded protein response (UPR) in�-cells (24,25). Against this background, we analyzed therole of ATF4 knock down on cytokine-induced changes inKrebs cycle–regulating genes. Since both cytokine combi-

nations have similar effects on this group of genes, weused only IL-1� � IFN�. siRNA targeting ATF4 inhibitedcytokine-induced ATF4 expression by �80% (Fig. 5B).Expression of ATF3, an ATF4-regulated gene, was signifi-cantly decreased confirming functional consequences ofATF4 knock down (Fig. 5E). Inhibition of ATF4 expression

*

control IL+IFN TNF+IFN

CC

L2/G

APD

H

control IL+IFN TNF+IFN

CXC

L1/G

APD

H

control IL+IFN TNF+IFN

IL-1

5/G

APD

H

control IL+IFN TNF+IFN

CXC

L10/

GA

PDH

control IL+IFN TNF+IFN

CXC

L9/G

APD

H

control IL+IFN TNF+IFN

CC

L5/G

APD

Hcontrol IL+IFN TNF+IFN

IRF1

/GA

PDH

*

**

* *

* *

control IL+IFN TNF+IFN

IRF7

/GA

PDH

control IL+IFN TNF+IFN

STAT

1/G

APD

H

A

B

control IL+IFN TNF+IFN

IRF7

/GA

PDH

control IL+IFN TNF+IFN

IRF1

/GA

PDH

control IL+IFN TNF+IFN

CC

L5/G

APD

H

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2control IL+IFN TNF+IFN

IL-1

5/G

APD

H

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

CXC

L10/

GA

PDH

0.0

0.4

0.8

1.2

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

CXC

L1/G

APD

H

§ §

§ § §

§§

FIG. 2. TNF-� and IL-1� differentially modulate expression of �-cell genes involved in the inflammatory response. Gene expression was analyzedby real-time RT-PCR. A: FACS-purified rat �-cells were exposed or not (control) to IL-1� � IFN-� (IL�IFN) or to TNF-� � IFN-� (TNF�IFN)for 6 h (�) or 24 h (f). B: FACS-purified rat �-cells were transfected with siRNA control (�) or siRNA against IRF-7 (f) and exposed or not(control) to IL-1� � IFN-� (IL�IFN) or to TNF-� � IFN-� (TNF�IFN) for 24 h. Results are means � SE of three to six independent experiments.*P < 0.05 vs. IL�IFN at the same time point; §P < 0.05 vs. siControl at the same time point and treatment.

F. ORTIS AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 59, FEBRUARY 2010 367

Page 11: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

partially prevented the inhibitory effects of cytokines onthe expression of the two Krebs cycles genes analyzed,namely malate dehydrogenase and �-ketoglutarate dehy-drogenase (Fig. 5C). ATF4 knock down was confirmed atthe functional level by Western blot for ATF3, a down-stream gene of ATF4. Cytokine-induced expression ofATF3 was prevented by ATF4 knock down, at a similarlevel of the inhibition observed when a siATF3 was used(Fig. 5E).Cytokines decrease the expression of incretin andhormone receptors at least in part via activation ofHIF-1�. IL-1� � IFN� and TNF-� � IFN� inhibited theexpression of key hormone receptors in rat �-cells (Table1). This was confirmed by real-time RT-PCR for thereceptors of glucagon-like peptide (GLP)-1, prolactin(PRL), growth hormone (GH), and cholecystokinin A(CCKA) (Fig. 6A). Cytokine-induced inhibition was in therange of 50–90% and more marked after 24 h (Fig. 6A).Analysis of the promoter region of these genes found acommon binding site for the transcription factor HIF-1(26,27). Cytokines upregulated transcriptional activity(Fig. 6B) and mRNA expression (Table 1 and Fig. 6C) ofHIF-1�, the regulatory HIF-1 subunit (26). This could inpart be explained by cytokine activation of AKT (supple-mentray Fig. 5C). HIF-1� knock down inhibited by 75%cytokine-induced HIF-1� expression (Fig. 6D) and by 60%HIF transcriptional activity (supplementary Fig. 5A and B).Knock down of HIF-1� partially prevented cytokine-in-duced apoptosis in �-cells (Fig. 6D) and inhibition of tworeceptors analyzed, GLP-1 receptor (R) and PRLR (Fig.6E). This partial effect of HIF-1� knock down in GLP-1Rand PRLR expression suggest that other transcriptionfactors may be involved in this process, as supported bythe in silico identification of other relevant candidatetranscription factors (supplementary Table 8).

Cytokines regulate the splicing machinery and alter-native splicing in primary �-cells. A large number ofcytokine-modified genes are involved in alternative splic-ing (Table 1, splicing machinery). To determine whetherthis triggers modifications in the splice variants present in�-cells, 24-h cytokine-treated samples from the three�-cell–independent preparation/experiments used in theinitial array analysis (Fig. 1 and Table 1) were pooled aspreviously described (7,9) and analyzed for the presenceof splice variants using the rat exon-array from Affymetrix.Cytokine treatment led to important changes in alternativesplicing, with IL-1� � IFN� potentially modulating differ-ential splicing of 2,651 genes (21% of the total number ofthe expressed genes) (supplementary Table 9, Fig. 7A).From these, only 396 were also modified at the expressionlevel. These findings suggest that �50% of IL-1� � IFN-�–modulated genes undergo alternative splicing. For TNF-� �IFN-�, there was induction of alternative splicing in 2,206genes (19%), with only 207 of these being also modified atexpression level (supplementary Table 10, Fig. 7A). Thespliced genes were classified according with their putativemolecular function as shown in supplementary Tables 11and 12. Alternative splicing was confirmed for three genesanalyzed by RT-PCR (Fig. 7), namely iNOS and ASS, whichparticipate in the citrulline-NO cycle (Fig. 3A) and theNF-�B subunit p100/p52 (NF-�B2). iNOS was not detectedin control cells, but it was induced by cytokines, and therewas a difference in the size of the amplified region after 6and 24 h of cytokine treatment (Fig. 7C). At 6 h, there wasamplification of two bands of 1,237 and 1,137 bp, thesecond one corresponding to iNOS lacking exon 8 or 9 (bysequencing analysis of the PCR product we confirmed thatexon 8 is missing (data not shown), while at 24 h themajority of the amplified bands contained exon 8 (Fig. 7C).This confirms that posttranscriptional processing of iNOSis differentially modified by cytokines at different timepoints. Using the same approach, we observed that cyto-kines decreased utilization of exon 1 from ASS while itincreased utilization of exon 22 from NF-�B2 (Fig. 7C).

DISCUSSION

We have presently used state-of-the-art array analysis offluorescence-activated cell sorter–purified �-cells to unveilthe global pattern of genes modified by the inflammatorycytokines IL-1� � IFN-� and TNF-� � IL-1�. The use ofprimary and pure cell preparations (�90% �-cells) is ofspecial relevance, since it enabled us to obtain a broadpicture of �-cell responses to proapoptotic inflammatorymediators without the confounding signals generated byother endocrine and nonendocrine islet cells. We cannot,however, discard that interactions between �-cells andothers cells in the islets, and with infiltrating mononuclearcells during insulitis, will lead to changes in �-cell geneexpression that are not detected by the present model. Thearray data were evaluated by both nonbiased pathwayanalysis (IPA) and investigator-based analysis. Selectedpathways were chosen for additional studies, with specialemphasis on the role of novel transcription factors.Prompted by the observation of cytokine-induced changesin a large number of genes involved in alternative splicing,an exon-array analysis was performed to evaluate thepresence of splicing variants in �-cells. The following aremain novel observations of the study. 1) Nearly 8,000genes were detected as present in �-cell, with 96% confir-mation of selected cytokine-modified genes by real-time

Ass

Citrulline

Arginine

Citrulline+NO

iNOS

Urea+ornithine

arg1

A

B

*

0

2

4

6

8

10

12

control IL+IFN TNF+IFN

pM n

itrite

/105

cel

ls

FIG. 3. Differential usage of the NO synthesis pathway by IL-1� andTNF-�. A: Schematic view of the NO synthesis pathway. Ellipticalshapes represent enzymes. B: Synthesis of NO by rat primary �-cellscultured in arginine-citrulline–free medium (o) or in medium contain-ing 1 mmol/l citrulline (�) and exposed to IL-1� � IFN-� (IL�IFN) orTNF-� � IFN-� (TNF�IFN) for 48 h. Results are mean of five indepen-dent experiments. *P < 0.05 vs. arginine-citrulline–free medium.

GENE NETWORKS AND �-CELL APOPTOSIS

368 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org

Page 12: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

RT-PCR. This more than doubles the known �-cell ex-pressed genes. 2) There are temporal, qualitative, andquantitative differences in the genes induced by TNF-� andIL-1� regarding inflammation and NO production. This isprobably secondary to the differential expression andusage of transcription factors such as NF-�B and IRF-7. 3)Key gene networks related to �-cell–differentiated pheno-type and the Krebs cycle are similarly inhibited by TNF-� �IFN-� and IL-1� � IFN-�. 4) Cytokines induce majorchanges in alternative splicing of genes, indicating a novellevel of functional regulation in �-cells.

IL-1� � IFN-� induces a higher expression of iNOS andASS and a more marked inhibition of arg1 as comparedwith TNF-� � IFN-�, leading to higher NO productionfrom either arginine or citrulline (supplementary Fig. 1Band Fig. 3). This enables continuous NO production ininflammation sites where arginine is usually depleted. NOformation induced by proinflammatory cytokines contrib-utes for �-cell death in some rodent models of diabetes (1).

Furthermore, 46% of cytokine-modulated genes are NOdependent in INS-1E cells (8), suggesting that differencesin NO production may explain why IL-1� � IFN-� modu-lates a higher number of genes compared with TNF-� �IFN-� at 24 h (Fig. 1).

Exposure of �-cells to proinflammatory cytokines dur-ing insulitis induce release of chemokines and cytokines,which may contribute to recruit and activate immune cellsand thus amplify local inflammation and the autoimmuneassault (2,10). The present data suggest differential rolesfor IL-1� and TNF-� in this “dialogue” between the �-cellsand the immune system. Thus, while TNF-� � IFN-�induces higher expression of IL-15, CCL5, CXCL9, andCXCL10, IL-1� � IFN-� preferentially induces CCL2 andCXCL1. These inflammatory mediators contribute for in-sulitis and destruction of �-cells by the immune system(1,2,10), and the present observations suggest that thebalance between TNF-� and IL-1� expression during insu-litis can lead to different outcomes. These differences may

1.2 1.2

0.8

0.0

0.4* * * * * *

Reg

ulat

ory

tran

scrip

tion

fact

ors

(fold

var

iatio

n vs

con

trol

)G

enes

rela

ted

to β

-cel

l fun

ctio

n(fo

ld v

aria

tion

vs c

ontr

ol)

0.0

0.2

0.4

0.6

0.8

1.0

1.2

Ins1 Ins2 Glut2 Gck PSCK1 PSCK3

##

##

#

##

0.0

0.4

0.8

1.2

1.6

Pdx1 Neurod1 Nkx2.2 Nkx6.1 MafA Pax6 Isl1 foxo1

#

# *

*

*

*

###

**

**

A

B

C

0.0

0.4

0.8

control IL+IFN TNF+IFN

isl1

/GA

PDH

control IL+IFN TNF+IFN

Maf

A/G

APD

H

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

Pdx1

/GA

PDH

#

§§

§§

§

§

§

§

§

FIG. 4. Cytokines decrease the expression of genes involved in the maintenance of a differentiated �-cell phenotype. Expression of genes relatedto �-cell function (A) or regulatory transcription factors (B) were analyzed by microarray (n � 3) in FACS-purified rat �-cells exposed to IL-1� �IFN-� for 6 h (�) or 24 h (f) or to TNF-� � IFN-� for 6 h (o) or 24 h (vertical striped bars). Results are shown as fold change compared withcontrol (no cytokine added), considered as one (line). C: confirmation by real-time RT-PCR of cytokine effects on the expression of PDX-1, MafA,and Isl1; �, 6 h; f, 24 h. Results are means � SE of three to four independent experiments. *P < 0.05; #P < 0.01; §P < 0.001 vs. control.

F. ORTIS AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 59, FEBRUARY 2010 369

Page 13: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

reflect differential usage of two key transcription factors,namely NF-�B and IRF-7. Thus, higher and earlier activa-tion of NF-�B by IL-1� � IFN-�, as presently shown inprimary �-cells, probably explains the higher expressionof NF-�B target genes such as CCL2 (28). On the otherhand, TNF-� preferentially triggers IRF-7 and IRF-1 acti-vation (present data). In other cell types, TNF-�–inducedIFN-� expression synergistically activates the IRF-7/1–STAT-1 pathway, leading to sustained expression of cyto-kines and chemokines (21). TNF-� � IFN-� leads to higherinduction of IFN-� expression in �-cells than IL-1� �IFN-�, which may explain the differences in the expressionof chemokines/cytokines induced by IL-1� or TNF-�. Therole of STAT-1 in this process was previously shown inislets from STAT-1 knockout mice (29), and we presentlyshow that IRF-7 knock down partially prevents TNF-� �IFN-�–induced expression of IRF-1, IL-15, CCL5, andCXCL1.

Loss of differentiated �-cell functions is another impor-tant consequence of exposure to cytokines (30). We pres-ently describe three gene networks whose inhibition maycontribute to this outcome, namely key transcription fac-tors for the maintenance of �-cell phenotype, mRNAsencoding receptors for growth factors and incretins, andmRNAs encoding enzymes of the Kreb’s cycle. Zhou et al.(23) reported that inducing expression of the transcriptionfactors neurogenin 3 (Ngn3), pancreatic and duodenalhomeobox-1 (Pdx-1), and mammalian homologue of avianMafA/L-Maf (MafA) reprograms pancreatic mouse exo-crine cells into cells that closely resemble �-cells. Repro-gramming of pancreatic exocrine cells to �-cells shouldbenefit patients with type 1 diabetes, an autoimmunedisease characterized by local inflammation (2,10). Insulinepitopes are targets of the immune assault in type 1diabetes (31), and new insulin-producing cells will berecognized and attacked by the immune system (32). The

gene

/GA

PDH

1.2

0.0

0.4

0.8

control IL+IFN TNF+IFN

*

aconitase 2

* *

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

fumarase 1.2

*

0.0

0.4

0.8

control IL+IFN TNF+IFN

*

α-ketoglutarate dehy

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

*

isocitrate dehy 3b

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

**

**

malate dehy

* *

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

succinate dehy

**

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

succinyl-CoA synthase

gene

/GA

PDH

A

B

gene

/GA

PDH

0.0

0.2

0.4

0.6

0.8

1.0

control IL+IFN

ATF4

0.0

0.2

0.4

0.6

0.8

1.0

control IL+IFN

malate dehy

0.0

0.2

0.4

0.6

0.8

1.0

control IL+IFN

α-ketoglutarate dehy

1.2

0.0

0.4

0.8

control IL+IFN TNF+IFN

Citrate synthase

C

++ + +cyt

ATF3

tubulin

control siNEG siATF3 siATF4

E

§ §

FIG. 5. Cytokines inhibit expression of genes encoding Krebs cycleenzymes, which is partially dependent of ATF4 activation. A: Confirma-tion by real-time RT-PCR of microarray analysis data in rat purified�-cells untreated (control) or exposed to a combination of IL-1� � IFN-�(IL�IFN) or TNF-� � IFN-� (TNF�IFN) for 6 h (�) or 24 h (f). Resultsare means � SE of four to five independent experiments. *P > 0.05 vs.control. B and C: �-Cells were transfected with siControl (�) or siATF4(f) and then left untreated or treated with IL-1� � IFN-� (IL�IFN) for24 h. B: Confirmation of ATF4 knockdown (KD) by real-time RT-PCR. C:Effects of ATF4 KD on the expression of malate dehydrogenase and�-ketoglutarase dehydrogenase. Results are mean of six independentexperiments. §P < 0.05 cytokines � siATF4 vs. cytokines � siControl.

Dehy � dehydrogenase. E: Western blot for ATF3 protein in cells transfected with siATF4 or siATF3. The figure is representative of fourindependent experiments.

GENE NETWORKS AND �-CELL APOPTOSIS

370 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org

Page 14: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

present data suggest that immune mediators of insulitis,such as cytokines, will push back newly developed �-cellsinto a dedifferentiated state, preceding actual �-cell death.

The hormones GLP-1, CCKA, PRL, and GH are involvedin mitotic and functional activation of rodent �-cells(33,34). Due to these characteristics, GLP-1 analogs arebeing presently tested as an adjuvant therapy in early type1 diabetes (35). Of concern, cytokines induce an early andprofound inhibition of mRNAs encoding for the receptors

of GLP-1, CCKA, PRL, and GH, which may prevent therestorative effects of these hormones. These mRNAs areinhibited in parallel, suggesting the role for a commoninhibitory transcription factor downstream of cytokines.In silico analysis and siRNA experiments suggest thatHIF-1� is at least in part involved in this inhibitory effect ofcytokines. HIF-1 is a key regulator of adaptive cellularresponses to hypoxia, and it is active when its regulatorysubunit HIF-1� is stabilized during hypoxia (26). HIF-1�

0

10

20

30

control IL+IFN

% a

popt

osis

0.0

0.2

0.4

0.6

0.8

1.0

control IL+IFN

PRLR

/GA

PDH

0.0

0.2

0.4

0.6

0.8

1.0

control IL+IFN

GLP

1R/G

APD

H

A

B C D

E

0.0

0.4

0.8

1.2

luci

fera

se a

ctiv

ity ** *

0.0

0.2

0.4

0.6

0.8

1.0

control IL+IFN

HIF

1α/G

APD

H

*

* *

growth hormone R

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

* *

cholecystokinin A R

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

* *

glucagon like peptide 1 R

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

* *

prolactin R

0.0

0.4

0.8

1.2

control IL+IFN TNF+IFN

* *

gene

/GA

PD

Hge

ne/G

APD

H

§

FIG. 6. Cytokines decrease expression of key hormone receptor genes partially via HIF-1� induction. Rat purified �-cells were left untreated(control) or exposed to a combination of IL-1� � IFN-� (IL�IFN) or TNF-� � IFN-� (TNF�IFN). A: Real-time RT-PCR to confirm microarrayanalysis of hormone receptors (R) expression after cytokine exposure for 6 h (�) or 24 h (f). Results are means � SE of three to fourexperiments. *P < 0.05 vs. control. B: Luciferase reporter assay of HIF-1� activation by cytokines. Cells were cotransfected with an HREluciferase reporter gene and the internal control pRL-CMV, then left untreated (o) or exposed to IL�IFN (�) or the positive control Cobaltchloride (CoCl2, f) for 12 h. Results are normalized for Renilla luciferase activity and are means � SE of five experiments. *P < 0.05 vs. untreatedcells. C–E: HIF-1� knockdown by siRNA. Cells were transfected with siControl (�) or siHIF-1� (f) and then left untreated or treated withIL�IFN for 24 h. Results are means � SE of four to six experiments. C: HIF-1� knockdown analyzed by real-time RT-PCR. *P < 0.05 vs. siControlunder the same treatment and §P < 0.05 vs. control (not cytokine treated). D: Viability of cells after HIF-1� knockdown and 48 h cytokineexposure. *P < 0.05 vs. siControl � cytokines. E: Expression of PRLR and GLP-1R after HIF-1� knockdown and 24-h cytokine exposure, measuredby real-time RT-PCR. *P < 0.05 vs. siControl � cytokines.

F. ORTIS AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 59, FEBRUARY 2010 371

Page 15: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

stabilization/activation has important roles in other cellu-lar responses, such as glucose metabolism, cell growth/apoptosis, and the inflammatory response (26,36,37). We

now show that cytokines induce both HIF-1� mRNAexpression and transcriptional activity in �-cells and thatHIF-1� knock down partially prevents cytokine-induced

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16

1 2 3 4 5 6 7 8 9 10 11 12 1314 15 16 17 18 19 20 21 22 23 24 25 26 27

54321

ATG TGA

6 7 10X

Ass

iNOS

GAPDH

Exon 1 +

Exon 1

Exon 8 +

Exon 8 –

–Exon 22 +

Exon 22

ATG TAG

5432

1237bp

1137bp

iNOS

453bp

364bpAss

1 2 3 4 5 6 7 8 9 10 11 12 13 14 5 16 17 18 19 20 21 22

ATG TGA

370bp

140bp21

NF-κB2

NF-κB2

A

B

C IL TNF C IL TNF

6h 24h

1999 207 169

AS Exp

TNF+IFN

2255 396 308

AS Exp

IL+IFN

C

FIG. 7. Cytokines induce alternative splicing in rat pancreatic �-cells. A: Ven diagram representing the number of �-cell genes that undergoalternative splicing (alternative splicing) and/or expression (Exp) changes after 24 h of cytokine treatment compared with control condition, asidentified by exon array analysis (GeneChip Rat exon 1.0 ST Array). B: Schematic diagram of inducible iNOS, ASS, and NF�B subunit p100/p52exon structures and of the PCR primers presently used to identify spliced forms. Start (ATG) and stop (TGA or TAG) codons are indicated inthe figure. The arrows show the positions of the PCR primers, while the lines below indicate the size of the amplified region in the presence orabsence of the respective exon analyzed. C: RT-PCR of rat primary �-cells exposed to control condition (C), IL-1� � IFN-� (IL), or TNF-� � IFN-�(TNF) for 6 or 24 h to amplify the regions of iNOS, ASS, and NF-�B indicated in B. GAPDH was amplified in parallel to control for the amountof cDNA loaded in each reaction. The figure is representative of three to five experiments.

GENE NETWORKS AND �-CELL APOPTOSIS

372 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org

Page 16: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

inhibition of key hormone receptors and �-cell apoptosis.This suggests a novel role for HIF-1� in �-cells, as one ofthe mediators of cytokine-induced �-cell dysfunction anddeath. Of note, prolonged �-cell exposure to high glucosetriggers HIF-1� expression (38), while constitutive HIF-1�expression in �-cells impairs glucose-stimulated insulinrelease (39).

We have previously shown that cytokine-induced NOformation in �-cells inhibits mitochondrial glucose oxida-tion via functional impairment of the enzyme aconitase(40). We presently show that cytokines also inhibit expres-sion of several mRNAs encoding enzymes of the Krebscycle. This is mediated, at least in part, via ATF4 activa-tion, as suggested by both in silico analysis and siRNA. Thetranscription factor ATF4 is part of the UPR response incytokine-treated �-cells (24,25). Endoplasmic reticulumstress may contribute to HIF-1� activation (41), potentiallylinking three cytokine-induced effects in �-cells, namelyendoplasmic reticulum stress, HIF-1� activation, and inhi-bition of the Krebs cycle. Additional experiments are nowrequired to further investigate this possibility and to clarifyhow gene networks regulating mitochondria and endoplas-mic reticulum function may provide the signaling for �-cellapoptosis.

Cytokines modulate expression of several genes relatedto the alternative splicing machinery (present data), whichis in line with recent proteomic data (42). Alternativesplicing is an important determinant of cellular function.More than 85% of the human genes may undergo alterna-tive splicing (14,43), and many of these spliced forms aretissue specific, contributing for the generation of pro-teomic diversity (43). The complex interactions requiredfor correct splicing can be disturbed by changes in theexpression of splicing factors and cellular energy stores(15,44). By exon-array analysis, we presently observedthat cytokines modulate the expression of splicing vari-ants in �-cells, with potentially 20% of the detected genesshowing alternative splicing. This findings must be inter-preted with caution, since they represent a pool of threeexperiments that precludes adequate statistical analysis.In addition, this methodology can lead to false positivedetection (45). Here, for at least three of the modifiedgenes (iNOS ASS, and NF-�B2) there was independentconfirmation by RT-PCR. Cytokine-induced iNOS splicingvariants may provide another level of regulation of iNOSactivity in a tissue-specific way (46). Many of the presentlyidentified genes are modified only at the splicing level,without changes in expression. This indicates a new levelof complexity in the effects of cytokines (and potentially ofother modulators of �-cell function and survival) that mustbe taken into account in future studies. The functionalimpact of these diverse splice variants in �-cells remain tobe investigated, but data available from other tissuesindicate that it is huge, increasing the number of moleculespecies that are involved in normal regulation of cell ordisease susceptibility (14–16,43,47,48). Interestingly, splic-ing may also have a role in the augmentation of autoimu-nity in type 1 diabetes (49).

In conclusion, the present study doubles the number ofknown genes modified by cytokines in primary rat �-cellsand suggests temporal, qualitative, and quantitative differ-ences between the effects of TNF-� � IFN-� and IL-1� �IFN-�. Cytokines decrease the expression of genes relatedto �-cell function and growth/regeneration, indicating thatimmune mediators of insulitis can push back newlyformed �-cells into a dedifferentiated state. Interestingly,

cytokines modify alternative splicing in �-cells, indicatinga new level of complexity in the �-cell responses toimmune-mediated damage.

ACKNOWLEDGMENTS

This work has been supported by grants from the Euro-pean Union (Projects Savebeta and Naimit in the Frame-work Programs 6 and 7 of the European Community); theFNRS (Fonds National de la Recherche Scientifique) andARC (Actions de Recherche Concertee de la CommunauteFrancaise), Belgium; the Belgium Program on Interuniver-sity Poles of Attraction initiated by the Belgian state (IUAPP5/17 and P6/40); and the Expert Center Grant 2008.40.001from the Dutch Diabetes Research Foundation. M.L.C. isthe recipient of a scholarship from CAPES (BrazilianCoordination for the Improvement of Higher EducationPersonnel). F.M. is the recipient of a Postdoctoral Fellow-ship from FNRS, Belgium. No potential conflicts of interestrelevant to this article were reported.

We thank the personnel from Laboratory of Experimen-tal Medicine–Universite Libre de Bruxelles, M.A. Neef, G.Vandenbroeck, M. Urbain, J. Schoonheydt, R. Leeman, S.Mertens, R. Makhnas, and A.E. Musaya for excellenttechnical support.

REFERENCES

1. Eizirik DL, Mandrup-Poulsen T. A choice of death-the signal-transductionof immune-mediated �-cell apoptosis. Diabetologia 2001;44:2115–2133

2. Kaminitz A, Stein J, Yaniv I, Askenasy N. The vicious cycle of apoptotic�-cell death in type 1 diabetes. Immunol Cell Biol 2007;85:582–589

3. Uno S, Imagawa A, Okita K, Sayama K, Moriwaki M, Iwahashi H, YamagataK, Tamura S, Matsuzawa Y, Hanafusa T, Miyagawa J, Shimomura I.Macrophages and dendritic cells infiltrating islets with or without �-cellsproduce tumour necrosis factor-� in patients with recent-onset type 1diabetes. Diabetologia 2007;50:596–601

4. Mastrandrea L, Yu J, Behrens T, Buchlis J, Albini C, Fourtner S, Quattrin T.Etanercept treatment in children with new-onset type 1 diabetes: pilotrandomized, placebo-controlled, double-blind study. Diabetes Care 2009;32:1244–1249

5. Pickersgill LM, Mandrup-Poulsen TR. The anti-interleukin-1 in type 1diabetes action trial-background and rationale. Diabetes Metab Res Rev2009;25:321–324

6. Eizirik DL, Moore F, Flamez D, Ortis F. Use of a systems biology approachto understand pancreatic �-cell death in type 1 diabetes. Biochem SocTrans 2008;36:321–327

7. Cardozo AK, Kruhoffer M, Leeman R, Orntoft T, Eizirik DL. Identificationof novel cytokine-induced genes in pancreatic �-cells by high-densityoligonucleotide arrays. Diabetes 2001;50:909–920

8. Kutlu B, Cardozo AK, Darville MI, Kruhoffer M, Magnusson N, Orntoft T,Eizirik DL. Discovery of gene networks regulating cytokine-induced dys-function and apoptosis in insulin-producing INS-1 cells. Diabetes 2003;52:2701–2719

9. Cardozo AK, Heimberg H, Heremans Y, Leeman R, Kutlu B, Kruhoffer M,Orntoft T, Eizirik DL. A comprehensive analysis of cytokine-induced andnuclear factor-�B-dependent genes in primary rat pancreatic �-cells. J BiolChem 2001;276:48879–48886

10. Eizirik DL, Colli ML, Ortis F. The role of inflammation in insulitis and �-cellloss in type 1 diabetes. Nat Rev Endocrinol 2009;5:219–226

11. Ortis F, Cardozo AK, Crispim D, Storling J, Mandrup-Poulsen T, Eizirik DL.Cytokine-induced pro-apoptotic gene expression in insulin-producing cellsis related to rapid, sustained and non-oscillatory NF-�B activation. MolEndocrinol 2006;20:1867–1879

12. Ortis F, Pirot P, Naamane N, Kreins AY, Rasschaert J, Moore F, Theatre E,Verhaeghe C, Magnusson N, Chariot A, Orntoft T, Eizirik DL. TNF-� andIL-1� induction of NF-�B and its downstream genes has a pro-apoptoticrole in pancreatic �-cells. Diabetologia 2008;51:1213–1225

13. Magnusson NE, Cardozo AK, Kruhoffer M, Eizirik DL, Orntoft TF, JensenJL. Construction and validation of the APOCHIP, a spotted oligo-microar-ray for the study of �-cell apoptosis. BMC Bioinformatics 2005;6:311

14. Pan Q, Shai O, Lee LJ, Frey BJ, Blencowe BJ. Deep surveying of alternativesplicing complexity in the human transcriptome by high-throughput se-quencing. Nat Genet 2008;40:1413–1415

F. ORTIS AND ASSOCIATES

diabetes.diabetesjournals.org DIABETES, VOL. 59, FEBRUARY 2010 373

Page 17: Cytokines Interleukin1 and Tumor Necrosis Factor Regulate Different Transcriptional and Alternative Splicing Networks in Primary Cells

15. Blencowe BJ. Alternative splicing: new insights from global analyses. Cell2006;126:37–47

16. Colobran R, Pujol-Borrell R, Armengol MP, Juan M. The chemokinenetwork: II. on how polymorphisms and alternative splicing increase thenumber of molecular species and configure intricate patterns of diseasesusceptibility. Clin Exp Immunol 2007;150:1–12

17. Thomas HE, McKenzie MD, Angstetra E, Campbell PD, Kay TW. �-Cellapoptosis in diabetes. Apoptosis 2009;14:1389–1404

18. Wu Z, Irizarry RA, Gentleman R, Martinez-Murillo F, Spencer F. A modelbased background adjustment for oligonucleotide expression array. J AmStat Assoc 2004;99:909–917

19. Moore F, Colli ML, Cnop M, Esteve MI, Cardozo AK, Cunha DA, Bugliani M,Marchetti P, Eizirik DL. PTPN2, a candidate gene for type 1 diabetes,modulates interferon-�–induced pancreatic �-cell apoptosis. Diabetes2009;58:1283–1291

20. Sgarbanti M, Marsili G, Remoli AL, Orsatti R, Battistini A. IRF-7: new rolein the regulation of genes involved in adaptive immunity. Ann N Y Acad Sci2007;1095:325–333

21. Yarilina A, Park-Min KH, Antoniv T, Hu X, Ivashkiv LB. TNF activates anIRF1-dependent autocrine loop leading to sustained expression of chemo-kines and STAT1-dependent type I interferon-response genes. Nat Immu-nol 2008;9:378–387

22. Flodstrom M, Niemann A, Bedoya FJ, Morris SM Jr, Eizirik DL. Expressionof the citrulline-nitric oxide cycle in rodent and human pancreatic �-cells:induction of argininosuccinate synthetase by cytokines. Endocrinology1995;136:3200–3206

23. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA: In vivo reprogram-ming of adult pancreatic exocrine cells to �-cells. Nature 2008;1476–4687

24. Eizirik DL, Cardozo AK, Cnop M. The role for endoplasmic reticulum stressin diabetes mellitus. Endocr Rev 2008;29:42–61

25. Cardozo AK, Ortis F, Storling J, Feng YM, Rasschaert J, Tonnesen M, VanEylen F, Mandrup-Poulsen T, Herchuelz A, Eizirik DL. Cytokines down-regulate the sarcoendoplasmic reticulum pump Ca2� ATPase 2b anddeplete endoplasmic reticulum Ca2�, leading to induction of endoplasmicreticulum stress in pancreatic �-cells. Diabetes 2005;54:452–461

26. Ke Q, Costa M. Hypoxia-inducible factor-1 (HIF-1). Mol Pharmacol 2006;70:1469–1480

27. O’Hagan KA, Cocchiglia S, Zhdanov AV, Tambuwala MM, Cummins EP,Monfared M, Agbor TA, Garvey JF, Papkovsky DB, Taylor CT, Allan BB.PGC-1� is coupled to HIF-1�-dependent gene expression by increasingmitochondrial oxygen consumption in skeletal muscle cells. Proc NatlAcad Sci U S A 2009;106:2188–2193

28. Kutlu B, Darville MI, Cardozo AK, Eizirik DL. Molecular regulation ofmonocyte chemoattractant protein-1 expression in pancreatic �-cells.Diabetes 2003;52:348–355

29. Gysemans CA, Ladriere L, Callewaert H, Rasschaert J, Flamez D, Levy DE,Matthys P, Eizirik DL, Mathieu C. Disruption of the �-interferon signalingpathway at the level of signal transducer and activator of transcription-1prevents immune destruction of �-cells. Diabetes 2005;54:2396–2403

30. Cnop M, Welsh N, Jonas JC, Jorns A, Lenzen S, Eizirik DL. Mechanisms ofpancreatic �-cell death in type 1 and type 2 diabetes: many differences, fewsimilarities. Diabetes 2005;54(Suppl. 2):S97–S107

31. Kent SC, Chen Y, Bregoli L, Clemmings SM, Kenyon NS, Ricordi C, HeringBJ, Hafler DA. Expanded T cells from pancreatic lymph nodes of type 1diabetic subjects recognize an insulin epitope. Nature 2005;435:224–228

32. Sibley RK, Sutherland DE, Goetz F, Michael AF. Recurrent diabetesmellitus in the pancreas iso- and allograft: a light and electron microscopic

and immunohistochemical analysis of four cases. Lab Invest 1985;53:132–144

33. Nielsen JH, Svensson C, Galsgaard ED, Moldrup A, Billestrup N. �-Cellproliferation and growth factors. J Mol Med 1999;77:62–66

34. Ahren B: Potentiators and inhibitors of insulin secretion. In Advances in

Molecular Cell Biology. Vol. 29. Bittar EE, Howel SL, Eds. Greenwich, CT,Jay Press, 1999, p. 191–202

35. Suarez-Pinzon WL, Power RF, Yan Y, Wasserfall C, Atkinson M, Rabino-vitch A. Combination therapy with glucagon-like peptide-1 and gastrinrestores normoglycemia in diabetic NOD mice. Diabetes 2008;57:3281–3288

36. Dehne N, Brune B. HIF-1 in the inflammatory microenvironment. Exp CellRes 2009;315:1791–1797

37. Piret JP, Mottet D, Raes M, Michiels C. Is HIF-1� a pro- or an anti-apoptoticprotein? Biochem Pharmacol 2002;64:889–892

38. Bensellam M, Jonas J-C. High glucose activates hypoxia inducible factors1 and 2 in cultured rat islets and INS-1E cell monolayers. In Proceedings of

the 44th Annual Meeting of the European Association for the Study of

Diabetes, Rome, Italy, 7–11 September 200839. Gribble FM. Intolerant of glucose and gasping for oxygen. J Clin Invest

2009;15:247–24940. Welsh N, Eizirik DL, Bendtzen K, Sandler S. Interleukin-1 �-induced nitric

oxide production in isolated rat pancreatic islets requires gene transcrip-tion and may lead to inhibition of the Krebs cycle enzyme aconitase.Endocrinology 1991;129:3167–3173

41. Werno C, Zhou J, Brune B. A23187, ionomycin and thapsigargin upregulatemRNA of HIF-1� via endoplasmic reticulum stress rather than a rise inintracellular calcium. J Cell Physiol 2008;215:708–714

42. D’Hertog W, Overbergh L, Lage K, Ferreira GB, Maris M, Gysemans C,Flamez D, Cardozo AK, Van den Bergh G, Schoofs L, Arckens L, Moreau Y,Hansen DA, Eizirik DL, Waelkens E, Mathieu C. Proteomics analysis ofcytokine-induced dysfunction and death in insulin-producing INS-1E cells:new insights into the pathways involved. Mol Cell Proteomics 2007;6:2180–2199

43. Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, KingsmoreSF, Schroth GP, Burge CB. Alternative isoform regulation in human tissuetranscriptomes. Nature 2008;456:470–476

44. Long JC, Caceres JF. The SR protein family of splicing factors: masterregulators of gene expression. Biochem J 2009;417:15–27

45. Gaidatzis D, Jacobeit K, Oakeley EJ, Stadler MB: Overestimation ofalternative splicing caused by variable probe characteristics in exonarrays. Nucleic Acid Res 2009;37:e107

46. Eissa NT, Strauss AJ, Haggerty CM, Choo EK, Chu SC, Moss J. Alternativesplicing of human inducible nitric-oxide synthase mRNA. tissue-specificregulation and induction by cytokines. J Biol Chem 1996;271:27184–27187

47. Gardina PJ, Clark TA, Shimada B, Staples MK, Yang Q, Veitch J, SchweitzerA, Awad T, Sugnet C, Dee S, Davies C, Williams A, Turpaz Y. Alternativesplicing and differential gene expression in colon cancer detected by awhole genome exon array. BMC Genomics 2006;7:325

48. Zhang W, Duan S, Bleibel WK, Wisel SA, Huang RS, Wu X, He L, Clark TA,Chen TX, Schweitzer AC, Blume JE, Dolan ME, Cox NJ. Identification ofcommon genetic variants that account for transcript isoform variationbetween human populations. Hum Genet 2009;125:81–93

49. Diez J, Park Y, Zeller M, Brown D, Garza D, Ricordi C, Hutton J, EisenbarthGS, Pugliese A. Differential splicing of the IA-2 mRNA in pancreas andlymphoid organs as a permissive genetic mechanism for autoimmunityagainst the IA-2 type 1 diabetes autoantigen. Diabetes 2001;50:895–900

GENE NETWORKS AND �-CELL APOPTOSIS

374 DIABETES, VOL. 59, FEBRUARY 2010 diabetes.diabetesjournals.org