Diabetes

Title page

“Function of isolated pancreatic islets from patients at onset of type 1 diabetes; Insulin secretion can be restored after some days in a non-diabetogenic environment in vitro. Results from the DiViD study”

Short running title: “In vitro studies of islets from type 1 diabetes patients”

List of authors Lars Krogvold,1,2,* Oskar Skog,3 Görel Sundström,4 Bjørn Edwin,2,5 Trond Buanes,2,6 Kristian F Hanssen,2,7 Johnny Ludvigsson,8 Manfred Grabherr,4 Olle Korsgren,3 and Knut Dahl-Jørgensen1,2

1Paediatric Department, Oslo University Hospital 2Faculty of Medicine, University of Oslo, Norway 3Department of Immunology, Genetics and Pathology, Uppsala University, Sweden 4Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden 5The Intervention Centre and Department of Surgery, Oslo University Hospital 6Division of Cancer, Surgery and Transplantation, Oslo University Hospital 7Department of Endocrinology, Oslo University Hospital 8Division of Paediatrics, Department of Clinical Experimental Medicine, Linköping University, Sweden

*Corresponding author: Lars Krogvold, Paediatric Department, Oslo University Hospital HF, P. O. Box 4950 Nydalen, N-0424 Oslo, Norway

Phone: +4797522277. Fax number: +4723015790. E-mail: [email protected]

LK, OS and GS contributed equally to the manuscript and are listed in alphabetical order. KDJ and OK contributed equally to the manuscript. KDJ is Principal Investigator of DiViD-study.

Word count, manuscript: 2024

Number of tables: 0 (1 online supplemental table) Number of figures: 3 (1 online supplemental figure)

1

Diabetes Publish Ahead of Print, published online February 12, 2015 Diabetes Page 2 of 24

Abstract:

The understanding of the etiology of type 1 diabetes (T1D) remains limited. One objective of the Diabetes Virus Detectionstudy (DiViD) was to collect pancreatic tissue from living subjects shortly after the diagnosis of T1D. Here we report the insulin secretion ability by in vitro glucose perifusion and explore the expression of insulin pathway in isolated islets of Langerhans from these patients. Whole genome RNA sequencing was performed on islets from six DiViD patients and two organ donors that died at the onset of T1D and compared with findings in three non diabetic organ donors. All human transcripts involved in the insulin pathway were present in the islets at onset of T1D. Glucoseinduced insulin secretion was present in some patients at the onset of T1D, and a perfectly normalized biphasic insulin release was obtained after some days in a nondiabetogenic environment in vitro. This indicates that the potential for endogenous insulin production is good, which could be taken advantage of if the disease process was reversed at diagnosis.

2

Page 3 of 24 Diabetes

Our understanding of the etiology of type 1 diabetes remains limited (1). Animal

models show only partial similarities with the human disease (2), and there has been

lack of wellpreserved human tissue samples (3). The functionality of islets of

Langerhans at onset of type 1 diabetes in humans remains poorly characterized.

Previous in vitro studies (4;5) have shown function of islet cells obtained several

months after diagnosis but so far there has been lack of in vitro access to isolated

islets obtained from subjects at onset of type 1 diabetes required to obtain indepth

understanding. Studies demonstrating the expression profiles for the human

pancreas and purified islets in type 1 diabetes have been published, providing

interesting data supporting that type 1 diabetes is caused by a chronic inflammatory

process with participation of innate immunity (6;7). The beta cells express and release

cytokines and chemokines, providing a link between the and the immune

system in early type 1 diabetes (8). However, these studies are based on few diseased

cases, mainly including tissue from subjects with longstanding type 1 diabetes (6;7).

Insulin secretion from islets obtained from nondiabetic human subjects exhibits a

typical biphasic pattern when stimulated with glucose (9). At diagnosis most patients

with type 1 diabetes have significant, but insufficient insulin secretion, even though

4050% of the βcells may be present (10) suggesting beta cell dysfunction (11;12).

Endoplasmic reticulum (ER) stress causes beta cell dysfunction, suggestively by

entrapment of the Wolfram syndrome 1 (WFS1), inhibiting the synthesis of

cAMP and thereby the secretion of insulin (13).

3

Diabetes Page 4 of 24

Here we report the ability of insulin secretion and explore the expression of insulin pathway genes in isolated islets of Langerhans obtained from subjects with recent onset type 1 diabetes and nondiabetic controls.

Materials and Methods

Patients and pancreas donors

Six patients (case 16), age 2435 years who gave their written informed consent were recruited to the DiViDstudy (14). In addition, the pancreases from two organ donors

(case 78) who died at the onset of type 1 diabetes and from three organ donors

(controls 13) without pancreatic disease were included in the study. Both donors with type 1 diabetes died of brain edema and total brain infarction described previously

(15). Clinical data regarding cases and controls are shown in eTable 1. The

Government’s Regional Ethics Committee in Norway and the Regional Ethics

Committee in Uppsala approved the study. For details regarding the methods, see supplementary material.

Results

Islet function

The mean glucosestimulated insulin secretion (GSIS) was reduced in islets from type

1 diabetes subjects (Fig. 1A). GSIS was lowest when islets were examined day 1 after isolation but seemed to increase after 3 and 6 days of in vitro culture (Fig. 1A). Islets from individual subjects with type 1 diabetes had varying levels of GSIS (Fig. 1B).

When examined day 1 after isolation, islets from all subjects except case 6 had a very low or undetectable GSIS (Fig. 1B). After 3 and 6 days of culture, islets from two of the subjects (Case 1 and 2) secreted slightly increased amount of insulin upon glucose

4

Page 5 of 24 Diabetes

stimulation, but did not display biphasic insulin secretion, whereas islets from case 4

that displayed a poor GSIS one day after isolation, recovered to a normal biphasic

secretion after 3 and 6 days of culture. Case 6 displayed a closetonormal GSIS

already one day after isolation, and the insulin secretion levels increased further after

3 and 6 days of culture (Fig. 1B). For the rest of the cases (3, 5, 7 and 8) the insulin

secretion remained low or undetectable after culturing. Islets from nondiabetic

subjects responded with a biphasic insulin release already on day one and were not

further stimulated.

Whole transcriptome sequencing

We generated a total of 362 million reads and mapped those to the .

Both cases and controls generated approximately the same amount of mapable reads.

The full data set (reads) is openly available on doi: 10.17044/BILS/g000002.

When comparing expression similarities, using RPKM values, across all genes, islets

from 5 of the 6 live subjects (Case 15, but not case 6) are grouped together and

separate from the brain dead donors (case 7 and 8 and control 13), regardless of

whether the islets are from type 1 diabetes or nondiabetic subjects (Fig. 2A),

reflecting the major impact of brain death on the results obtained from the whole

transcriptome analysis. Similarly, genes that are part of the complement system

pathway also reflect the differences between islets from live and brain dead subjects

(Fig. 2B). By contrast, the insulin secretion pathway groups islet samples by type 1

diabetes or not, notably resulting in longer branch lengths compared to the

complement system pathway (Fig. 2C).

5

Diabetes Page 6 of 24

In all cases, except case 6, the genes that (according to KEGG database) is involved in the secretion of insulin, the insulin pathway, were less expressed when compared to the controls (Fig. 3). That includes all the genes in the insulin pathway, both the insulin gene INS itself, and the genes involved in production and release of insulin. In case 6, all the genes in the pathway were upregulated compared to the nondiabetic controls. INS is about tenfold lower expressed in all other diabetic samples, together with lowered expression of two upstream regulators, the pancreatic and duodenal homeobox 1 (PDX1), and MAFA, a transcription factor that binds RIPE3b, a conserved enhancer element that regulates pancreatic beta cellspecific expression of

INS (16). Additional genes that are consistently lower in expression include (a) the adenylate cyclase activating polypeptide (PACAP) and its PACAPR; (b)

FFAR1, a member of the GPR40 family of G proteincoupled receptors and may be involved in the metabolic regulation of insulin secretion; (c) Gprotein controlled integral and inwardrectifier type (KCNJ11); (d)

ATPbinding cassette transporter of the MRP subfamily (ABCC8), which is involved in multidrug resistance but also functions as a modulator of ATPsensitive potassium channels and insulin release; (e) calciumactivated nonselective that mediates transport of monovalent cations across membranes (TRPM4); and (f) three major glucose transporters in the mammalian bloodbrain barrier, which are found primarily in the cell membrane and on the cell surface, where they also function as receptors for human Tcell leukemia virus I and II (GLUT1/2/3). By contrast, the muscarinic cholinergic receptor CHRM3, and the Gprotein coupled receptor CCKAR, which binds nonsulfated members of the cholecystokinin family of peptide hormones and acts as a major physiologic mediator of pancreatic enzyme secretion, are unchanged in expression, or slightly more highly expressed.

6

Page 7 of 24 Diabetes

Discussion

The results show that the expression of key regulatory elements, the insulin pathway

and the GSIS were reduced in islets isolated from 7 of the 8 subjects with type 1

diabetes compared to islets from nondiabetic organ donors. However, in 1 of the 8

cases, the insulin pathway expression and the GSIS were remarkably preserved

already at the day of isolation. The observed GSIS improvement after three and six

days of culture in 4 of the 8 cases suggests that when the islets are removed from the

‘diabetic milieu’, remaining beta cells can recover. Soluble factors released within the

vicinity of the islets could induce a functional impairment of the beta cells, which

could be reversed after islet isolation and in vitro culture. However, a screening of 41

cytokines and chemokines in isolated islets from the six type 1 diabetes patients

showed no difference from that in isolated islets from brain dead organ donors (data

not shown).

It is known that prolonged exposure of human islets to high glucose impairs beta cell

function (17) and since even short episodes of hyperglycemia dramatically affect

glucose tolerance (18) it is possible that glucotoxicity could play a role in the reduced

GSIS observed in islets from these patients. In 1940 Jackson described that intensive

insulin therapy at diagnosis of T1D lead to a long partial remission of the disease (19).

Later these findings were confirmed using Cpeptide determinations (20). However,

in an experimental transplantation model human islets were exposed 46 weeks in

vivo to hyperglycemia followed by normoglycemia for 2 weeks (21). The results

showed that even if the hyperglycemia induced impairment in glucose metabolism,

depletion of insulin mRNA, decreased (pro)insulin biosynthesis, increased glycogen

7

Diabetes Page 8 of 24

accumulation and depletion of insulin content was reversed by the 2 week period of normoglycemia, the deleterious effects of the diabetic state on human islet insulin release remained.

Endoplasmatic reticulum (ER) stress has been proposed as an important contributor to beta cell dysfunction in type 1 diabetes (22). In this study, the of ER stress markers (BiP, CHOP, ATF4, and XBP1) was high in islets from all subjects with type 1 diabetes, but did not differ from the islets from the brain dead non diabetic controls. The process of brain death has been shown to induce ER stress (23) and the finding of a similar level of expression of these genes in islets isolated from subjects with recent onset type 1 diabetes imply that the ER stress in these islets is a feature of type 1 diabetes. The expression of WFS1 was lower in islets from subjects with T1D (both braindead and live subjects) than in islets from the control donors, with the exception of case 6 from whom the islets with almost preserved GSIS were isolated. It may be speculated that ER stress and low expression of WFS1 contribute to the reduced beta cell function in T1D.

The level of transcription of the genes in the insulin pathway was reduced in most of the diabetic cases, compared to the nondiabetic controls. However, all genes in the insulin pathway were expressed, also in islets from the subjects with no or very low

GSIS. This is in line with earlier studies, describing insulin synthesis and storage remaining in islets even many years after type 1 diabetes onset (6;24), and show that the destruction of the βcells is a slow ongoing process (24). A limitation in all studies of transcriptome profiling of an unfractioned tissue is that any changes observed can be due to differences in either modulation of gene expression or from changes in its

8

Page 9 of 24 Diabetes

cell composition. The current analysis was conducted on handpicked islets, thereby

avoiding to a large extent the problem with analyzing pancreatic biopsies in which the

islets constitute only about 1%. However, the insulin producing beta cells constitute

only about 60% of the total cell number in human islets. A reduction in the number of

insulin producing cells in the islets isolated from subjects with recent onset type 1

diabetes could be in agreement with the observation of a reduction in the insulin

pathway.

The most obvious limitation of the present study is the small number of cases

examined. The number of recruited patients was limited by the unexpectedly high

frequency of complications arising from the biopsy procedure (14). Therefore we

considered it unethical to continue to enrol patients. A further important consideration

is the degree of matching between the tissues obtained from the patients with T1D and

those used as controls. This study did not include pancreatic biopsies from healthy

individuals and we therefore chose to employ tissue harvested by alternative methods.

Nondiabetic organ donors were chosen as their clinical characteristics are well

defined, all having normal HbA1c and being negative for 4 auto antibodies. The islets

from the controls were not cultured and stimulated with glucose on day 3 and 6, but it

has previously been shown that culturing of normal beta cells does not improve their

insulin secretion ability (25). Due to the limited number of islets available from each

of the cases, we chose to stimulate with glucose only, knowing that additional

secretagogues would potentially provide more complete information.

In summary, our findings illustrate the importance of βcell dysfunction, and not only

loss of number of insulin producing cells, at onset of type 1 diabetes. The restoration

9

Diabetes Page 10 of 24

of specific function of the isolated islets removed from the diabetogenic milieu should encourage further characterization of the underlying mechanism(s) of this functional impairment. Hopefully, this would allow initiation of clinical intervention trials specifically aiming to restore beta cell function alone or combined with drugs targeting the injurious processes ongoing within the pancreas at onset of type 1 diabetes.

Acknowledgements: The project was founded by SouthEastern Norway Regional

Health Authority (Grant to KDJ), The Novo Nordisk Foundation (Grant to KDJ and

OK), through the PEVNET Study Group founded by the European Union's Seventh

Framework Programme [FP7/20072013] under grant agreement n°261441 PEVNET

(http://www.uta.fi/med/pevnet/publications.html), Barndiabetesfonden (Swedish

Child Diabetes Foundation), the Swedish Medical Research Council (65X1221915

6, K201554X12219194), Diabetes Wellness Sweden, the Swedish Diabetes

Foundation and the Family Ernfors Fund. OK’s position is in part supported by the

National Institutes of Health (U01AI06519206). Human pancreatic islets were obtained from the Nordic Network for Clinical Islet Transplantation supported by the

Swedish national strategic research initiative EXODIAB (Excellence of Diabetes

Research in Sweden) and the Juvenile Diabetes Research Foundation. The authors would like to acknowledge support of Uppsala Genome Center and UPPMAX for providing assistance in massive parallel sequencing and computational infrastructure.

Work performed at Uppsala Genome Center has been funded by RFI/VR “SNISS”

Swedish National Infrastructure for large Scale Sequencing and Science for Life

Laboratory, Uppsala.

10

Page 11 of 24 Diabetes

No potential conflict of interest relevant to this article was reported.

LK was responsible for clinical coordination and recruitment of patient, data

collection, analysis and interpretation, and drafted the manuscript. OS performed the

GSIS analyses, and analysed, interpreted and drafted the manuscript. GS performed

the whole transcriptome sequencing analysis and interpretation and drafted the

manuscript. BE and TB performed the surgery and participated in writing of the

article. KFH, JL, MG and OK contributed to study design, data analysis,

interpretation and writing of the manuscript. KDJ was the principal investigator of the

study, had the initial idea of the DiViD study, and participated in study design,

funding, regulatory issues, international collaboration, data collection, analysis and

interpretation, and in writing of the manuscript. LK and KDJ are the guarantors of this

work and, as such, had full access to all the data in the study and take responsibility

for the integrity of the data and the accuracy of the data analysis.

The authors thank specialist nurse Trine Roald, who has provided invaluable efforts in

coordination of the study, Sofie Ingvast for excellent technical assistance, nurses and

doctors at the local hospitals, providing contact with the patients, and finally the

patients who participated in this study.

11

Diabetes Page 12 of 24

Figure legends

FIG. 1. Glucose-stimulated insulin secretion by isolated islets. Twenty handpicked islets were perifused with low glucose (1.67 mmol/L) for 42 min, high glucose (20 mmol/L) for 48 min, and then low glucose again as indicated. Fractions were collected at 6 min intervals and the secreted insulin was measured by ELISA. A:

Mean insulin secretion from islets isolated from six type 1 diabetes patients (case 16) and cultured for 1 (open circles), 3 (black squares) or 6 days (open squares), and from islets isolated from 15 organ donors on day one without pancreatic disease (open triangles). B: Individual insulin secretion from islets from two organ donors with type

1 diabetes (case 78) and from six live patients with type 1 diabetes (case 16), cultured for 1 (open circles), 3 (black squares), or 6 days (open squares).

Observational data.

FIG. 2. Neighbor-joining trees based on expression similarity. Shown are groupings for all genes (a), the complement system pathway (b), and the insulin secretion pathway (c). With the exception of sample case 6, live and brain dead donors cluster together in (a) and (b), while the insulin secretion pathway (c) splits diabetic from nondiabetic samples.

FIG. 3. Insulin secretion pathway for all diabetic samples except case 6 (A) and for case 6 (B). We normalized expression by three brain dead controls and show lower expression in blue and higher expression in red. With the exception of case 6, the majority of genes in the pathway are more lowly expressed in the diabetic samples. Similar figures for the rest of the cases separately are shown in eFigure 1.

12

Page 13 of 24 Diabetes

REFERENCES

1. Atkinson,MA, Eisenbarth,GS, Michels,AW: Type 1 diabetes. Lancet

2014;383:69-82

2. In't Veld,P: Insulitis in human type 1 diabetes: a comparison between

patients and animal models. Semin Immunopathol 2014;36:569-579

3. Coppieters,KT, Roep,BO, von Herrath,MG: Beta cells under attack: toward a

better understanding of type 1 diabetes immunopathology. Semin

Immunopathol 2011;33:1-7

4. Marchetti,P, Dotta,F, Ling,Z, Lupi,R, Del,GS, Santangelo,C, Realacci,M,

Marselli,L, Di,MU, Navalesi,R: Function of pancreatic islets isolated from a

type 1 diabetic patient. Diabetes Care 2000;23:701-703

5. Lupi,R, Marselli,L, Dionisi,S, Del,GS, Boggi,U, Del,CM, Lencioni,C, Bugliani,M,

Mosca,F, Di,MU, Del,PS, Dotta,F, Marchetti,P: Improved insulin secretory

function and reduced chemotactic properties after tissue culture of islets

from type 1 diabetic patients. Diabetes Metab Res Rev 2004;20:246-251

6. Planas,R, Carrillo,J, Sanchez,A, Ruiz de Villa,MC, Nunez,F, Verdaguer,J,

James,RFL, Pujol-Borrell,R, Vives-Pi,M: Gene expression profiles for the

human pancreas and purified islets in Type 1 diabetes: new findings at

clinical onset and in long-standing diabetes. Clin Exp Immunol

2010;159:23-44

13

Diabetes Page 14 of 24

7. Nica,AC, Ongen,H, Irminger,JC, Bosco,D, Berney,T, Antonarakis,SE,

Halban,PA, Dermitzakis,ET: Cell-type, allelic, and genetic signatures in the

human pancreatic beta cell transcriptome. Genome Res 2013;23:1554-1562

8. Eizirik,DL, Sammeth,M, Bouckenooghe,T, Bottu,G, Sisino,G, Igoillo-Esteve,M,

Ortis,F, Santin,I, Colli,ML, Barthson,J, Bouwens,L, Hughes,L, Gregory,L,

Lunter,G, Marselli,L, Marchetti,P, McCarthy,MI, Cnop,M: The human

pancreatic islet transcriptome: expression of candidate genes for type 1

diabetes and the impact of pro-inflammatory cytokines. PLoS Genet

2012;8:e1002552

9. Cerasi,E, Luft,R: The plasma insulin response to glucose infusion in healthy

subjects and in diabetes mellitus. Acta Endocrinol (Copenh) 1967;55:278-

304

10. Akirav,E, Kushner,JA, Herold,KC: Beta-cell mass and type 1 diabetes: going,

going, gone? Diabetes 2008;57:2883-2888

11. Drucker,D, Zinman,B: Pathophysiology of beta cell failure after prolonged

remission of insulin-dependent diabetes mellitus (IDDM). Diabetes Care

1984;7:83-87

12. Dupre,J, Jenner,MR, Mahon,JL, Purdon,C, Rodger,NW, Stiller,CR: Endocrine-

metabolic function in remission-phase IDDM during administration of

cyclosporine. Diabetes 1991;40:598-604

14

Page 15 of 24 Diabetes

13. Fonseca,SG, Urano,F, Weir,GC, Gromada,J, Burcin,M: Wolfram syndrome 1

and adenylyl cyclase 8 interact at the plasma membrane to regulate insulin

production and secretion. Nat Cell Biol 2012;14:1105-1112

14. Krogvold,L, Edwin,B, Buanes,T, Ludvigsson,J, Korsgren,O, Hyoty,H, Frisk,G,

Hanssen,KF, Dahl-Jorgensen,K: Pancreatic biopsy by minimal tail resection

in live adult patients at the onset of type 1 diabetes: experiences from the

DiViD study. Diabetologia 2014;57:841-843

15. Korsgren,S, Molin,Y, Salmela,K, Lundgren,T, Melhus,A, Korsgren,O: On the

etiology of type 1 diabetes: a new animal model signifying a decisive role

for bacteria eliciting an adverse innate immunity response. Am J Pathol

2012;181:1735-1748

16. Olbrot,M, Rud,J, Moss,LG, Sharma,A: Identification of beta-cell-specific

insulin gene transcription factor RIPE3b1 as mammalian MafA. Proc Natl

Acad Sci U S A 2002;99:6737-6742

17. Eizirik,DL, Korbutt,GS, Hellerstrom,C: Prolonged exposure of human

pancreatic islets to high glucose concentrations in vitro impairs the beta-

cell function. J Clin Invest 1992;90:1263-1268

18. Brunzell,JD, Robertson,RP, Lerner,RL, Hazzard,WR, Ensinck,JW, Bierman,EL,

Porte,D, Jr.: Relationships between fasting plasma glucose levels and insulin

secretion during intravenous glucose tolerance tests. J Clin Endocrinol

Metab 1976;42:222-229

15

Diabetes Page 16 of 24

19. Jackson RL, Boyd JD, Smith TE: Stabilization of the diabetic child. Am J Dis

Child 1940;59:332-341

20. Ludvigsson,J, Heding,LG, Larsson,Y, Leander,E: C-peptide in juvenile

diabetics beyond the postinitial remission period. Relation to clinical

manifestations at onset of diabetes, remission and diabetic control. Acta

Paediatr Scand 1977;66:177-184

21. Eizirik,DL, Jansson,L, Flodstrom,M, Hellerstrom,C, Andersson,A:

Mechanisms of defective glucose-induced insulin release in human

pancreatic islets transplanted to diabetic nude mice. J Clin Endocrinol

Metab 1997;82:2660-2663

22. Tersey,SA, Nishiki,Y, Templin,AT, Cabrera,SM, Stull,ND, Colvin,SC, Evans-

Molina,C, Rickus,JL, Maier,B, Mirmira,RG: Islet beta-cell endoplasmic

reticulum stress precedes the onset of type 1 diabetes in the nonobese

diabetic mouse model. Diabetes 2012;61:818-827

23. Contreras JL, Jenkins S Smyth C Young C Eckhoff DE. Role of Endoplasmatic

Reticulum (ER)-Stress on Brain-Death (BD)-Induced Human Beta-Cell

Death. In 65th Scientific Sessions of the American Diabetes Association. San

Diego, California, US. 2005.

Ref Type: Conference Proceeding

16

Page 17 of 24 Diabetes

24. Keenan,HA, Sun,JK, Levine,J, Doria,A, Aiello,LP, Eisenbarth,G, Bonner-Weir,S,

King,GL: Residual insulin production and pancreatic ss-cell turnover after

50 years of diabetes: Joslin Medalist Study. Diabetes 2010;59:2846-2853

25. Lee,RH, Carter,J, Szot,GL, Posselt,A, Stock,P: Human albumin preserves islet

mass and function better than whole serum during pretransplantation islet

culture. Transplant Proc 2008;40:384-386

17

Diabetes Page 18 of 24

A 800 1.67 20 1.67 mmol/L

) 600

L

/

l

o

m p

( 400

n

i

l

u

s n i 200

0 40 60 80 100 120 B Time (minutes)

800 Case 1 800 Case 2

600 600

400 400

200 200

0 0 40 60 80 100 120 40 60 80 100 120 800 Case 3 800 Case 4

600 600

400 400

)

L /

l 200 200

o

m

p (

0 0

40 60 80 100 120 40 60 80 100 120

n

i l

800 800 u

s Case 5 Case 6

n I 600 600

400 400

200 200

0 0 40 60 80 100 120 40 60 80 100 120

800 800 Case 7 Case 8

600 600

400 400

200 200

0 0 40 60 80 100 120 40 60 80 100 120

1.67 2 0 1 .67 mmol/L 1.67 2 0 1 .67 mmol/L

Time (minutes)

Page 19 ofa) 24 All genes Diabetes Case 1 live patients Case 2

Case 3

Case 4

Case 5

Case 8 brain-dead donors Case 6*

Control 2

Case 7

Control 1

0.09 Control 3

b) Complement system pathway

Case 7

Control 3

Control 1

Control 2

Case 6* brain-dead donors Case 8

Case 4

Case 5

Case 1 live patients Case 2

Case 3 0.07

c) Insulin secretion pathway

Case 1

Case 3

Case 5

Case 4

Case 2

Case 8 diabetic

Case 7

Control 2 non-diabetic

Case 6*

Control 1

0.09 Control 3 Diabetes Page 20 of 24 GIP GLP-1 PACAP A Fatty acid GIPR GLP1R KCNJ11 ABCC8 PACAPR Acetylecholine ATPase FFAR1 Gs TRPM4 depolarization CHRM3 CCK AC VDCC CCKAR Gq/G11 repolarization RAPGEF4 PKA 2+ RIMS2 Ca cAMP ATP PCLO K-CaK- PLC ER MT RYR2 Citric cycle ITPR33 CREB PDX1 DAG RAB3A Pyruvate Ca2+ CAMK nucleus IP3 DNA Glucose 6- Glucose phosphate CAMK INS PKC GCK

Glut1/2/3 Glucose

VAMP2 STX1A SNAP25 INS

GIP GLP-1 PACAP B Fatty acid GIPR KCNC11 ABCC8 PACAPR GLP1R Acetylecholine ATPase FFAR1 Gs TRPM4 depolarization CHRM3 CCK AC VDCC CCKAR Gq/G11 repolarization RAPGEF4 PKA 2+ RIMS2 Ca cAMP ATP PCLO K-CaK- PLC ER MT RYR Citric cycle ITPR33 CREB PDX1 DAG RAB3A Pyruvate Ca2+ CAMK nucleus IP3 DNA Glucose 6- Glucose phosphate CAMK INS PKC GCK Glut1/2/3 Glucose

VAMP2 STX1A SNAP25 INS Page 21 of 24 Diabetes

ONLINE SUPPLEMENTAL MATERIALS

Function of isolated pancreatic islets from patients at onset of type 1 diabetes; Insulin secretion can be restored after some days in a non-diabetogenic environment in vitro. Results from the DiViD study.

Table of contents Islet isolation ...... 2 Islet function ...... 2 Whole transcriptome sequencing and analyses ...... 2 eTable 1.Demographic data, cases and controls...... 3 Legend eFig 1...... 3 References ...... 3

Diabetes Page 22 of 24

Islet isolation The most distal part (0.5-1 cm) of laparoscopic pancreatic tail resections performed at Oslo University Hospital was immediately shipped by air courier in cold organ preservation solution (Viaspan®, UW) to Uppsala University for islet isolation. The islets were isolated by a method based on the procedure used for clinical islet isolation that has been described previously (1). Basically, the pancreatic duct was located under a surgical microscope, cannulated with a fine catheter, and collagenase (Liberase®, Roche) was injected continuously. After about 30 min, the pancreatic tissue was cut into 3–4 pieces, transferred to two 15 mL glass tubes, and digested at 37°C with continuous shaking for 30 min. The digested pancreata was washed twice with cold culture media (CMRL-1066, ICN Biomedicals, Costa Mesa, CA supplemented with 10 mM HEPES, 2 mM L-glutamine, 50 µg/mL gentamycin, 20 µg/mL ciproxfloxacin 10 mM Nicotinamide, and 10% heat-inactivated fetal calf serum) and immediately transferred to culture dishes. 300-700 islets from each patient were handpicked from the digested tissue under a microscope by skilled islet technicians with multiple years of experience. Islets from the brain-dead organ donors were isolated and cultured as described previously (1;2).

Islet function Glucose-stimulated insulin-secretion (GSIS) was assessed in a dynamic perfusion system, Suprafusion 1000 (BRANDEL, Gaithersburg, MD). Twenty handpicked islets cultured for 1, 3, or 6 days were perifused with low glucose (1.67 mM) for 42 min, high glucose (20 mM) for 48 min, and then low glucose again. Fractions were collected at 6 min intervals and the secreted insulin was measured by ELISA.

Whole transcriptome sequencing and analyses RNA was extracted with AllPrep DNA/RNA/Protein Mini Kit (Qiagen) from 50-100 islets per subject, immediately after handpicking from the digested pancreatic tail resections, or after storage of isolated islets from multi-organ donors (case 7 and 8, and controls 1-3) on day 1 after isolation at -80°C in RNAlater (Qiagen). The extracted RNA was of good quality (RIN values between 7.1 and 9.5) and sufficient quantity (>1 µg) for performing whole transcriptome sequencing. For details regarding the methods, see supplementary material. Total RNA samples were depleted from rRNA using the RiboMinus Eukaryote Kit for RNA and the RiboMinus Concentration Module. Total RNA was used to prepare a fragment library using the SOLiD Total RNA-seq Kit (Applied Biosystems). The libraries were sequenced using the AB SOLiD™ 5500xl-W system. The read length was 50 bp for all samples and directionality of RNA molecules was preserved in the sequencing. Reads were aligned to version GRCh37/hg19 of the human genome using version 1.1 of the Applied Biosystems whole transcriptome analysis tool (Uppsala genome Center). The neighbor-joining method, based on the normalized Euclidean distance of reads per kilobase of exon per million fragments mapped (RPKM) between samples was used to analyze differences in expression between all samples, using uniform gene expression to root the tree. Gene lists for the insulin secretion pathway and the complement system pathway were extracted from the KEGG database and relevant literature. RPKM values for all genes in the pathways were calculated (3) and each case was compared to the mean of the three controls in order to identify up- and down-regulated genes. Genes with RPKM values below 0.1 were considered non-expressed.

Page 23 of 24 Diabetes

eTable 1.Demographic data, cases and controls.

Time from HbA1c at Age Sex Insulin Anti Anti- T1D biopsy anti-ZnT8 Anti IA2 Category No (years (M/ (U/kg/d GAD insulin diagnosis (% (<0,12ai)* (<0,10ai)* ) F) ay) (<0,08ai)* (<0,08ai)* (weeks) mmol/mol) Case 1 25 F 4 6.7 (50) 0,5 1,76 0,7 0,28 0,16 DiViD- Case 2 24 M 3 10.3 (89) 0,35 0,79 <0,01 0,44 >3,00 cases, living newly Case 3 34 F 9 7.1 (54) 0,17 1,77 < 0,05 1,45 > 3,00 diagnosed Case 4 31 M 5 7.4 (57) 0,4 0,77 0,1 < 0,01 2,54 T1D Case 5 24 F 5 7.4 (57) 0,36 0,46 0,1 0,06 >3 patients Case 6 35 M 5 7.1 (54) 0,52 1,85 < 0,05 < 0,01 < 0,04

Organ- Case 7 40 M 0 nd - Neg. Neg. Neg. Neg. donors died at onset of T1D Case 8 29 M 0 10.4 (90) - Neg. Neg. Neg. Neg.

Organ- Control 1 22 M - 5.5 (37) - Neg. Neg. Neg. Neg. donors Control 2 20 M - 5.8 (40) - Neg. Neg. Neg. Neg. without T1D Control 3 25 M - 5.9 (41) - Neg. Neg. Neg. Neg.

*Arbitrary units according to Diabetes Antibody Standardization Program (DASP) (4)

Legend eFig 1

Insulin secretion pathway for all diabetic cases (1-8) shown separately. We normalized expression by three brain dead controls and show lower expression in blue and higher expression in red. With the exception of case 6, the majority of genes in the pathway are more lowly expressed in the diabetic samples.

References

1. Goto,M, Eich,TM, Felldin,M, Foss,A, Kallen,R, Salmela,K, Tibell,A, Tufveson,G, Fujimori,K, Engkvist,M, Korsgren,O: Refinement of the automated method for human islet isolation and presentation of a closed system for in vitro islet culture. Transplantation 2004;78:1367-1375 2. Korsgren,S, Molin,Y, Salmela,K, Lundgren,T, Melhus,A, Korsgren,O: On the etiology of type 1 diabetes: a new animal model signifying a decisive role for bacteria eliciting an adverse innate immunity response. Am J Pathol 2012;181:1735-1748 3. Mortazavi,A, Williams,BA, McCue,K, Schaeffer,L, Wold,B: Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods 2008;5:621-628 4. Bingley,PJ, Bonifacio,E, Mueller,PW: Diabetes Antibody Standardization Program: first assay proficiency evaluation. Diabetes 2003;52:1128-1136

Diabetes Page 24 of 24

pacap pacap

Fatty acid gipr glp-1r kir6.2 sur1 Fatty acid gipr glp-1r kir6.2 sur1 pacapr pacapr Acetylecholine Case 1 Acetylecholine Case 2 atpase atpase gpr40 gs nscc depolarization gpr40 gs nscc depolarization m3r m3r CCK ac CCK ac vdcc vdcc cckar cckar gq/g11 repolarization gq/g11 epac pka repolarization epac pka 2+ rim2 Ca2+ rim2 Ca cAMP ATP cAMP ATP piccolo k-caK- piccolo k-caK- plc plc ER MT ER MT ryr ip3r ryr ip3r3 creb pdx-1 Citric cycle 3 creb pdx-1 Citric cycle rab3 rab3 DAG Pyruvate DAG Pyruvate Ca2+ camk Ca2+ camk nucleus nucleus IP3 DNA Glucose 6- Glucose IP3 DNA Glucose 6- Glucose phosphate phosphate camk ins camk ins pkc gck pkc gck

glut1/2 Glucose glut1/2 Glucose

vamp2 vamp2 syntaxin syntaxin snap25 snap25 ins ins

pacap pacap

Fatty acid gipr glp-1r kir6.2 sur1 Fatty acid gipr glp-1r kir6.2 sur1 pacapr Case 3 pacapr Case 4 Acetylecholine atpase Acetylecholine atpase gpr40 gs nscc depolarization gpr40 gs nscc depolarization m3r m3r CCK ac CCK ac vdcc vdcc cckar cckar gq/g11 repolarization gq/g11 repolarization epac pka 2+ epac pka 2+ rim2 Ca rim2 Ca cAMP ATP cAMP ATP piccolo k-caK- piccolo k-caK- plc plc ER MT ER MT ryr ryr 3 Citric cycle ip3r3 Citric cycle ip3r creb pdx-1 creb pdx-1 DAG rab3 Pyruvate DAG rab3 Pyruvate Ca2+ camk Ca2+ camk nucleus nucleus IP3 DNA Glucose 6- Glucose IP3 DNA Glucose 6- Glucose phosphate phosphate camk ins camk ins pkc gck pkc gck

glut1/2 Glucose glut1/2 Glucose

vamp2 vamp2 syntaxin syntaxin snap25 snap25 ins ins

pacap pacap

Fatty acid gipr glp-1r kir6.2 sur1 Fatty acid gipr glp-1r kir6.2 sur1 pacapr pacapr Acetylecholine Case 5 Acetylecholine Case 6 atpase atpase gpr40 gs nscc depolarization gpr40 gs nscc depolarization m3r m3r CCK ac CCK ac vdcc vdcc cckar cckar gq/g11 epac pka repolarization gq/g11 epac pka repolarization Ca2+ Ca2+ rim2 rim2 ATP cAMP ATP cAMP piccolo k-caK- piccolo k-caK- plc plc MT ER MT ER ryr ip3r ryr Citric cycle ip3r3 creb pdx-1 Citric cycle 3 creb pdx-1 rab3 rab3 DAG Pyruvate DAG Pyruvate 2+ camk Ca2+ camk Ca nucleus nucleus IP IP3 DNA Glucose 6- Glucose 3 DNA Glucose 6- Glucose phosphate phosphate camk ins camk ins pkc gck pkc gck

glut1/2 Glucose glut1/2 Glucose

vamp2 vamp2 syntaxin syntaxin snap25 snap25 ins ins

pacap pacap

Fatty acid gipr glp-1r kir6.2 sur1 Fatty acid gipr glp-1r kir6.2 sur1 pacapr pacapr Acetylecholine Acetylecholine Case 8 atpase Case 7 atpase gpr40 gs nscc depolarization gpr40 gs nscc depolarization m3r m3r CCK ac CCK ac vdcc vdcc cckar cckar gq/g11 epac pka repolarization gq/g11 epac pka repolarization Ca2+ Ca2+ rim2 rim2 ATP cAMP ATP cAMP piccolo k-caK- piccolo k-caK- plc plc MT ER MT ER ryr ip3r ryr Citric cycle ip3r3 creb pdx-1 Citric cycle 3 creb pdx-1 rab3 DAG rab3 Pyruvate DAG Pyruvate Ca2+ camk Ca2+ camk nucleus nucleus IP3 DNA Glucose 6- Glucose IP3 DNA Glucose 6- Glucose phosphate phosphate camk ins camk ins pkc gck pkc gck

glut1/2 Glucose glut1/2 Glucose

vamp2 vamp2 syntaxin syntaxin snap25 snap25 ins ins