<<

Page 1 of 58

Insulitis and βCell Mass in the Natural History of

Short title: Insulitis and Type 1 Diabetes

Martha CampbellThompson1, Ann Fu1, John S. Kaddis2, Clive Wasserfall1, Desmond A.

Schatz3, Alberto Pugliese4, and Mark A. Atkinson1,3

From the 1Department of , Immunology, & Laboratory Medicine, College of Medicine,

University of Florida, Gainesville, FL; 2City of Hope, Los Angeles, CA, 3Department of

Pediatrics, University of Florida, Gainesville, FL; 4Diabetes Research Institute and Departments

of Medicine, Microbiology and Immunology, University of Miami Miller School of Medicine,

Miami, FL.

Correspondence to: Martha CampbellThompson, Pathology, Immunology and Laboratory

Medicine, 1395 Center Drive, University of Florida, Gainesville, FL 326100275; Phone: (352)

2736129; Fax: (352) 2737753; Email: [email protected]

Word count: Abstract (229); Manuscript (4979); Tables (2); Online Supplementary Tables (5);

Figures (6); Online Supplementary Figures (6).

Page 1 of 40

Diabetes Publish Ahead of Print, published online November 18, 2015 Diabetes Page 2 of 58

Descriptions of insulitis in human islets throughout the natural history of type 1 diabetes

are limited. We determined insulitis frequency (proportion of islets displaying insulitis to

total islets, %), infiltrating leukocyte subtypes, and βcell and αcell mass in pancreata recovered from organ donors with type 1 diabetes (n=80), as well as donors without diabetes, both with (AAb+; n=18) and without (AAb; n=61) islet . Insulitis was observed in 4/4 (100%) donors with type 1 diabetes duration ≤1 year and two AAb+ donors (2/18; 11%). Insulitis frequency showed a significant but limited inverse correlation with diabetes duration (r=0.58; P=0.01) but not with age at onset. Residual βcells were observed in all type 1 diabetees donors with insulitis, while βcell area and mass were significantly higher in type 1 diabetes donors with insulitis compared to those without insulitis. Insulitis affected 33% of + islets compared to 2% of insulin islets in donors with type 1 diabetes. A significant correlation was observed between insulitis frequency and CD45+, CD3+, CD4+, CD8+, and CD20+ cell numbers within the insulitis (r= 0.53

0.73, p=0.0040.04) but not CD68+ or CD11c+ cells. The presence of βcells as well as insulitis several years after diagnosis in children and young adults suggest that the chronicity of islet autoimmunity extends well into the post–diagnosis period. This information should aid considerations of therapeutic strategies seeking type 1 diabetes prevention and reversal.

Page 2 of 40

Page 3 of 58 Diabetes

Abbreviations:

AAb/AAb+ negative/positive donors without diabetes

DKA Diabetic ketoacidosis

GADA Glutamic acid decarboxylase65 autoantibodies

IA2A associated protein2 autoantibodies

IAA Insulin autoantibodies

IHC Immunohistochemistry

INS/Ins+/Ins Insulin/insulin positive/insulin negative

GCG Glucagon

nPOD Network for Pancreatic Organ donors with Diabetes

T1D Type 1 diabetes

ZnT8A Zinc transporter 8 autoantibodies

Key words: βcell, αcell, autoantibody, autoimmunity, , insulin, glucagon, DKA, HLA

Page 3 of 40

Diabetes Page 4 of 58

Type 1 diabetes is a chronic autoimmune disorder resulting from poorly understood

combinations of immunologic, genetic, and environmental factors that drive immune responses

against multiple βcell antigens, resulting in the irreversible loss of functional pancreatic βcells

(1). These destructive processes are thought to begin months to years before the clinical

symptoms of type 1 diabetes occur. Ongoing autoimmunity and βcell destruction are

asymptomatic during this prediabetic period but can be identified serologically by the presence

of autoantibodies against one or more of several βcell autoantigens including glutamic acid

decarboxylase65 (GADA), insulinomaassociated protein2 (IA2A), insulin (IAA), and zinc

transporter 8 (ZnT8A) (2). The number, rather than titer, of these so called “islet autoantibodies”

can be used to determine risk for type 1 diabetes development (reviewed in (3)).

While the initial description for of pancreatic islet cells (i.e., insulitis) in

type 1 diabetes occurred over a century ago, a limited number of studies have characterized this

lesion in patients with the disease or during the preclinical phase (4). Certain exceptions exist,

yet a metaanalysis of the literature would suggest that insulitis is present in young donors (<14

years) within 1 year of type 1 diabetes diagnosis as well as in donors with multiple islet

autoantibodies but having no diabetes (59). Difficulties in studying human islets/βcells in vivo can be ascribed to several factors including their relative scarcity within the pancreas (12%), anatomical inaccessibility, declining patient autopsy rates, and inherent risks associated with pancreatic biopsy (reviewed in (10)). This inability to perform pathological evaluations is unfortunate as it holds the potential to help explain, in part, multiple facets of disease heterogeneity including age variation at diagnosis and disease progression including rate of C peptide decline after onset or with experimental therapy (11; 12). In an attempt to overcome these limitations, organized efforts have been developed to obtain high quality pancreas

Page 4 of 40

Page 5 of 58 Diabetes

biospecimens from organ donors to study mechanisms of βcell loss in type 1 diabetes (e.g.,

PanFinn, Belgian Diabetes Registry, JDRF Network for Pancreatic Organ donors with Diabetes

(nPOD)) (7; 13; 14).

In this current study, our major objective was to screen pancreata from nPOD donors

with and without type 1 diabetes, as well as donors with and without islet autoantibodies but no

diabetes, for islets with insulitis followed by leukocyte subtyping of infiltrating cells. Insulitis

frequency and leukocyte subtypes, in islets still expressing insulin as well as insulinnegative

islets, were correlated with donor clinical attributes (age at onset or demise, diabetes duration,

autoantibody numbers, HLA, diabetic ketoacidosis (DKA)). The βcell and αcell area and mass

were also determined for each donor group and tested for correlations to insulitis frequency and

diabetes duration.

Page 5 of 40

Diabetes Page 6 of 58

RESEARCH DESIGN AND METHODS

Study Design

Pancreata were recovered from organ donors during a seven year period through the JDRF nPOD program according to procedures previously described (1416). This report provides results from

donors with: 1) no history of diabetes or other and negative for islet cell

autoantibodies (AAb, n=61); 2) no history of diabetes or other pancreatic disease but positive

for islet autoantibodies (AAb+, n=18); or 3) type 1 diabetes (n=80). The lower age limit in this

study was 4 years as the youngest donor with type 1 diabetes was 4.4 years old. Clinical and

demographic data are summarized (Suppl. Table 1) and include proportions of donors by

numbers of islet autoantibodies and those with DKA in relation to cause of death. Diabetes

durations were known for 79/80 donors with type 1 diabetes; one donor with unknown diabetes

duration was a 34 yearold male Caucasian donor with clinical history of insulindependent

diabetes, undetectable Cpeptide levels, and no insulin+ islets by histopathology (nPOD case

6032). Ethical permission was obtained from the Institutional Review Board at the University of

Florida, and informed consent was obtained from a legal representative of each donor.

Laboratory Assessments

Islet autoantibody assays, serum Cpeptide levels, and high resolution HLA genotyping were performed as previously described (1416). Islet autoantibodies were measured by radioimmunoassay against all four major type 1 diabetesassociated autoantigens (GADA, IA2

A, ZnT8A, IAA) in all but one donor who was diagnosed with type 1 diabetes at 10.7 years old and had diabetes for 6 years (nPOD 6062). Of note, after institution of insulin therapy for type 1 diabetes, IAA could not be distinguished from antibodies induced by insulin injections, so IAA

Page 6 of 40

Page 7 of 58 Diabetes

were excluded from autoantibody counts for the donors with type 1 diabetes subgroup in Table 1

and Suppl. Tables 1 and 3 (19).

Insulitis Screening and Insulitic Islet Subtyping for Insulitis Frequencies

Pancreata were processed to formalin fixed paraffin blocks for each pancreas region (head, body,

tail) as previously described (20). For each donor, serial sections (average 2 blocks/region) were

stained by hematoxylin and eosin and two double immunohistochemistry (IHC) stains (Ki67 and

insulin, CD3 and glucagon) (Suppl. Table 2) (21). When insulitic islets were found in a given

donor, additional blocks were screened (as detailed below). Stained sections were scanned at 20x

magnification using an Aperio CS scanner (Leica/Aperio, Vista, CA) and all images stored in an

online pathology database (eSLIDE, Leica/Aperio, Vista, CA).

Screening for insulitic islets was performed on CD3+ glucagon stained sections. An islet

was defined as ≥10 αcells. Insulitis was defined as an islet with ≥6 CD3+ cells immediately

adjacent to or within the islet with ≥3 islets per pancreas section according to recent criteria (4).

Islets with insulitis were marked in an image layer using ImageScope software (Leica/Aperio,

Vista, CA). The two IHC serial images were aligned using the synchronization tool, and insulin+

islets were also marked on image layer. All islets/section from donors with insulitis were

subsequently subtyped as follows: 1) insulin+ CD3; 2) insulin+ CD3+ 3) insulin CD3+; and 4)

insulin CD3 (see Table 1 for numbers of islets analyzed). Then, all islets were counted by

subtype. The process was reversed for AAb+ donors (i.e., islet subtypes were counted using the

Ki67insulin image after markup for CD3+ insulitic islets and Ins islets using the CD3glucagon

image). The number of pancreas sections subtyped for insulitis ranged from 216/donor (8.1±4.1

Page 7 of 40

Diabetes Page 8 of 58

sections/donor, n=162 sections) (Table 1). The lowest number of available sections was due to partial pancreas recovery (tail only, 6198).

Insulitis frequency (%) was calculated as total insulitic islets (sum of insulin+ CD3+ and insulin CD3+ islets) divided by total number of islets (sum of 4 subtypes). Frequency of insulin+ insulitic islets to total insulin+ islets was determined by the ratio of (insulin+ CD3+ islets)/(sum of insulin+ CD3 and insulin+ CD3+ islets) with similar calculations for frequency of insulin insulitic islets (insulin CD3+/(sum of insulin CD3+ and insulin CD3)).

Insulitis Leukocyte Phenotyping

Paraffin sections from blocks having the maximum insulitis frequency for each donor were stained and positive leukocytes/insulitic islet (≥6 CD3+ cells) counted using multi immunofluorescence. Serial sections (4m) were dewaxed and rehydrated with Tris buffer. Heat induced antigen retrieval was performed using Triology (Cell Marque, Rocklin, CA) at 95⁰ for

20 minutes followed by rinsing in water for 20 minutes. The staining series was designed to phenotype leukocytes [total leukocytes (CD45), T (CD3) and B (CD20) , T subsets (CD4, CD8), and monocytes (dendritic cells (CD11c) and macrophages

(CD68)) (21)] in conjunction with subtyping islets for insulin immunopositivity. The staining series was as follows: 1) CD45+glucagon+insulin; 2) CD20+CD3+glucagon; 3)

CD8+CD4+glucagon; and 4) CD11c+CD68+insulin. Chromogranin A staining was also used to delineate endocrine cells. Antigens are listed in order of primary antibody incubation and corresponding secondary antibody and conjugated fluorochrome (AF488AF555AF647) (Suppl.

Table 2). After blocking, sections were sequentially incubated with the primary antibody followed by the appropriate secondary antibody. For antiCD4, a Cy3 Tyramide signal

Page 8 of 40

Page 9 of 58 Diabetes

amplification kit (PerkinElmer, Waltham, MA) was used according to the manufacturer’s

instructions. All sections were mounted with ProLong Gold Antifade mounting media

containing DAPI (Life Technologies, Grand Island, NY). Positive controls included human

spleen, tonsil, and donor intrapancreatic lymph nodes, and negative controls included omission

of the primary antibody.

Numbers of leukocytes/insulitic islet were determined using multichannel image

acquisition software on a Zeiss Axiophot microscope (AxioVision, Carl Zeiss Inc., Thornwood,

NY). Fluorescent channels were viewed in combination with DAPI to count positive

leukocytes/islet.

βcell and αcell Area and Mass

Insulin and glucagon immunopositive areas were determined using the IHC sections to estimate

βcell and αcell areas, respectively, in relation to total tissue area using a single Aperio

colocalization algorithm (22). An average of 6 sections was used per donor (2 sections/head,

body, and tail regions). The βcell and αcell areas were expressed as a ratio (%) to the total

sectional area, including acinar and interstitial regions, to permit use of pancreas weights.

Average βcell and αcell area/pancreas was calculated from regional area averages. The βcell

or αcell mass (mg) was calculated by multiplication of the respective average area and pancreas

weight (g).

Statistical Analysis

Number and percent values were reported for all categorical factors, mean (± SD, n=number of

donors unless noted in figure legend) for normally distributed continuous variables, and median

Page 9 of 40

Diabetes Page 10 of 58

(minimum, maximum) for continuous variables with a skewed distribution. Donor characteristics

were analyzed using Satterthwaite corrected ttest if continuous and parametric, the Kruskal

Wallis test if continuous and nonparametric, Pearsons’s chi square if categorical values in all

cells exceeded 5, or by Fisher’s exact test if categorical values in one or more cells was <5. P

values were not calculated if values from 2 or more cells were equal to 0. Donor characteristics

were correlated to insulitis frequency and leukocyte subtypes/islet to other subtypes by Spearman

correlation and plotted with linear regression lines. Data were analyzed with exclusion of the

AAb+ donors with insulitis due to small sample size (n=2).

To obtain a subset of type 1 diabetes donors and compare clinical characteristics in the presence or absence of insulitis, selection of controls for all 18 type 1 diabetes insulitis cases was

attempted using a 1:1 ratio to identify the single best match. A matching method was used to

minimize bias lesioneffect estimates from key covariates (i.e., age at diabetes onset, sex,

ethnicity, and BMI). A propensity score, given the selected covariates, was calculated for all type

1 diabetes donors (23). The logit of the propensity score was used to match donors without

insulitis to those with insulitis. A randomly sorted nearestavailableneighbor matching method

without replacement was used with an optimal caliper width equal to 0.20 SD of the logit value

(0.15), as has been shown to be optimal (24), along with a SAS macro (25; 26). All reported P values are twotailed and significant if <0.05. All analyses were performed using GraphPad

Prism version 5.00 for Windows (GraphPad Software, San Diego, California CA, USA) or SAS software version 9.3 (SAS Institute, Cary, NC).

Page 10 of 40

Page 11 of 58 Diabetes

RESULTS

Insulitis Screening in Donors With Type 1 Diabetes and AAb+ Donors With No Diabetes

A total of 159 pancreata were reviewed for insulitis (no diabetes (n=61 AAb; n=18 AAb+);

n=80 type 1 diabetes) (Suppl. Table 1). Insulitis was observed in 18 type 1 diabetes donors

(18/80; 23%) and 2 AAb+ donors (2/18; 11%). Variability in the lobular distribution of insulin+

islets and insulitic islets was high in donors with type 1 diabetes (Fig. 1A-D). In AAb+ donors

with insulitis, insulitis was primarily observed in insulin+ islets (Suppl. Fig. 1). Numbers of

CD3+ cells/islet varied in a given donor irrespective of islet insulin immunopositivity or size,

and CD3+ cells were observed diffusely infiltrating islets, in various sized aggregates, or both

(Suppl. Fig. 2).

All 18 donors with type 1 diabetes and insulitis had insulin+ islets detected in at least one

section while only 5/62 (8%) donors with type 1 diabetes and no insulitis had insulin+ islets.

Insulitis was found in 509/1525 (33%) insulin+ islets and in 379/17,718 (2%) insulin islets in

donors with type 1 diabetes (Table 1). In AAb+ donors with insulitis, the opposite was found;

insulitis affected 167/4297 (4%) of insulin+ islets and 8/30 (27%) of those that were insulin.

Insulitis was found in type 1 diabetes donors ranging in age from 326.2 years (13.3±6.5) at

disease onset and with disease durations of 012 years (4.6±3.4) (Table 1). The highest insulitis

frequency (10.6%) was observed in a 13yearold who presented with DKA at disease onset

(nPOD 6228). The lowest insulitis frequency (0.3%) was observed in a 12yearold with type 1

diabetes for 9 years (nPOD 6264). The two AAb+ donors with insulitis had multiple

autoantibodies and were 22 and 23 years old (Table 1). The AAb+ donors had insulitis

frequencies (1.4%, 6.4%) within the range observed for donors with type 1 diabetes; few insulin

islets, with or without insulitis, were observed in these donors.

Page 11 of 40

Diabetes Page 12 of 58

The percent of islets with insulitis (insulitis frequency) was tested for a correlation to

diabetes duration. Insulitis frequency showed a low, yet significant, inverse relationship to

diabetes duration while age at onset or demise did not (Fig. 3).

Correlation of Insulitis Prevalence With Diabetes Duration

As past studies reporting insulitis primarily examined young donors with type 1 diabetes within a

year of onset, the prevalence of donors with insulitis were analyzed in this study using similar

criteria (6). All four donors with recent onset type 1 diabetes (duration ≤1 year) had insulitis; 3/4

were <15 years old at disease onset (Table 2). In donors with disease durations >1 year, insulitis

was found in 7/51 (14%) donors who were <15 years of age at disease onset and in 7/24 (29%)

donors who were ≥15 years at disease onset. Using 12 years diabetes duration as the cut off, the prevalence of insulitis was 45% (18/40) of donors with type 1 diabetes.

To further define other potential clinical variables as they relate to insulitis, 15 type 1

diabetes donors with insulitis were matched for age at onset, ethnicity, sex, and BMI with 15

type 1 diabetes donors without insulitis (Suppl. Table 3; In 3 cases, no matches were found and

thus excluded). Diabetes duration was significantly shorter in type 1 diabetes donors with

insulitis vs. those without insulitis (5 versus 14.5 years, respectively). Correspondingly, donors

with insulitis were significantly younger at demise compared to matched donors without insulitis

(20 versus 27.6 years, respectively). Interestingly, islet autoantibody numbers were not different between the matched type 1 diabetes donors. Hospitalization durations were not significantly

different in this matched subset and ranged from a mean of 3.23.7 days across all donor groups

(Suppl. Tables 1 and 3). Correlation of DKA during the hospitalization showed a significantly

higher proportion of donors with (7/15, 47%) versus without (1/15, 7%) insulitis (p=0.04).

Page 12 of 40

Page 13 of 58 Diabetes

Infiltrating Leukocytes Have Similar Proportions of B and T Cells and T Cell Subsets and

Numbers were Proportional to Insulitis Frequency

Infiltrating leukocytes in insulitic islets were examined by multiimmunofluorescence on serial

sections (Fig. 4). Mean numbers of CD45+ cells/islet in each donor were significantly correlated

to mean numbers of both T (CD3+) and B (CD20+) cells/islet (Fig. 5). Absolute numbers of

CD3+ and CD20+ cells varied between islets in a given donor depending on the nature of the

infiltrate (e.g., diffusely associated with islet, aggregate, mixture). Numbers of both CD3+ and

CD20+ cells appeared independent of islet insulin immunopositivity and diabetes duration

(Suppl. Fig. 23). Numbers of CD3+ cells were significantly correlated with CD8+ and CD4+

cells/islet (Fig. 5B). Leukocyte numbers/islet were also plotted for both donor groups with

insulitis, and high variability was observed particularly for CD45+, CD3+ and CD20+ (Fig. 5C).

This variability was also seen in the AAb+ donors without diabetes (Fig. 5C). Five donors had

insulitis aggregates with >50 CD3+ cells/islet; four were donors with type 1 diabetes who were

all less than 12 years of age but had widely varying diabetes durations (nPOD 6209, 6243, 6052,

6070) and one was an AAb+ donor without diabetes (nPOD 6267).

We also tested whether numbers of leukocytes/islet were correlated with insulitis

frequency and found significant correlations with CD45+, CD3+, CD20+, CD8+, and CD4+

cells/islet but not for CD68+ and CD11c+ cells/islet (Suppl. Fig. 4, Suppl. Table 4). Because of

the variability between islets in numbers of leukocytes/insulitic islet, we also tested whether a

ratio of markers (CD3+ cells / sum of CD3+ and CD20+ cells) would normalize interislet

variability but found no significant correlation to insulitis frequency (Suppl. Fig. 4).

Page 13 of 40

Diabetes Page 14 of 58

βcell and αcell Mass and Insulitis

Pancreas weights were variable in donors without diabetes: 71.1±24.2g (n=49) in AAb donors,

70.7±28.0g (n=15) in AAb+ donors without insulitis, and 60.7±17.9 in two AAb+ donors with insulitis (Fig. 6A). As anticipated, both donor groups without diabetes (AAb and AAb+) showed increasing pancreas weights after age 4 up to ~30 years (Suppl. Fig. 5A-B). Donors with type 1 diabetes had significantly lower pancreas weights (39.51±17.9g no insulitis, n=60;

33.8±12.9g with insulitis, n=16) compared to donors without diabetes (Fig. 6A). Pancreas weights were decreased at all ages in donors with type 1 diabetes compared to donors without diabetes (Suppl. Fig. 5C).

βcell area was also variable in donors without diabetes: 1.18±0.71% for AAb donors

(n=57) and 0.67±0.38% for AAb+ donors without insulitis (n=16) (Fig. 6B). The two AAb+ donors with insulitis had βcell area of 2.26±0.4%. As expected, significant reductions in βcell area was observed for donors with type 1 diabetes compared to nondiabetic donors (Fig. 6B). β cell area was also significantly different between the two donor groups with type 1 diabetes

(0.02±0.05% no insulitis, n=54; 0.15±0.17% with insulitis, n=18) (Fig. 6B).

βcell mass was 797±523mg for AAb donors (n=45) and 448±268mg for AAb+ donors without insulitis (n=15) (Fig. 6C). βcell mass was 1407± 649mg in the two AAb+ donors with insulitis. βcell mass was decreased in donors with type 1 diabetes regardless of insulitis status compared to those without type 1 diabetes; however, βcell mass was significantly higher in type

1 diabetes donors with insulitis (47.9±53.4mg, n=17; P<0.05) compared to those without insulitis (5.5±10.1mg, n=53) (Fig. 6C). βcell mass from five donors without diabetes (3 AAb,

2AAb+) overlapped with the highest values for βcell mass in donors with T1D and insulitis due to low pancreas weight (3) or low βcell area (2). Estimated βcell mass was further tested for

Page 14 of 40

Page 15 of 58 Diabetes

correlation with insulitis frequency, but no association was found (Suppl. Fig. 6A, P>0.05).

Similarly, we compared βcell mass to type 1 diabetes duration and age at onset to determine if

insulin mass was greater in donors with shorter diabetes durations or younger donors; again, no

significant associations were found (Suppl. Fig. 6B-C, P>0.05).

In marked contrast to differences in βcell area and mass, αcell area and mass were not

significantly different between groups (Fig. 6D-E). The αcell area was 0.73±0.45% in AAb

donors (n=57), 0.62±0.44% in AAb+ donors without insulitis (n=16), 1.37±0.04% in AAb+

donors with insulitis (n=2), 1.04±0.84% in donors with type 1 diabetes and no insulitis (n=57),

and 1.37±0.62% in donors with type 1 diabetes and insulitis (n=17). The αcell mass was

495±320mg in AAb donors, 402±428mg in AAb+ donors without insulitis, 827±224mg in

AAb+ donors with insulitis, 396±377mg in donors with type 1 diabetes and no insulitis, and

513±375mg in donors with type 1 diabetes and insulitis.

Influence of HLA on Insulitis

Donors with type 1 diabetes had a higher frequency of DRB1*03:01DQB1*02:01 or DRB1*04

DQB1*03:02 genotypes (DR3/DR4) compared to AAb or AAb+ donors (35.9% vs. 0%,

P<0.0001 and P=0.0012, respectively). HLA DR3/DR4 frequencies were not different when

comparing the matched donors within the type 1 diabetes group, with and without insulitis

(Suppl. Table 3). The DRB1*1501DQB1*0602 haplotype was very rare and considered

protective for type 1 diabetes. The overall frequency of this haplotype in AAb donors (25%)

was similar to the general U.S. Caucasian population (22%) (27). This haplotype was found in

four AAb+ donors with single autoantibodies and in none of those with multiple autoantibodies.

Page 15 of 40

Diabetes Page 16 of 58

In the type 1 diabetes donors, only one had the DRB1*1501DQB1*0602 haplotype, and quite interestingly, this donor also had insulitis (nPOD 6195).

Page 16 of 40

Page 17 of 58 Diabetes

DISCUSSION

The concept of insulitis has been the subject of previous reports; however, this study is the first

to report insulitis frequency, leukocyte subtypes, pancreatic weights, and βcell and αcell mass

in organ donors encompassing the entire natural history of type 1 diabetes [i.e., preclinical phase

(no diabetes, islet autoantibody positive) as well as acute and chronic phases]. Beyond this, the

current study is large, utilized standard operating proceduredriven protocols, and utilized only

high quality tissues. As a result, we believe the results noted herein provide much in the way of

new information as well as validating, or refuting, past observations.

Insulitis was defined by the presence of ≥6 CD3+ cells/islet and 3 insulitic islets/section

(4). Overall, insulitis was identified in 18/80 (23%) donors with type 1 diabetes and 2/18 (11%)

AAb+ donors without diabetes. Notably, numbers of donors with insulitis (insulitis prevalence)

was higher for older donors and for longer disease durations in both young and older patients

than reported using metaanalysis of existing studies (6). Historically, insulitis prevalence has

been reported in 51% (41/81) of patients with disease duration ≤1 year compared to only 3%

(4/132) in patients with durations >1 year (6). As shown in Table 2, we observed an insulitis

prevalence of 100% (4/4) in donors with type 1 diabetes durations ≤1 year and 19% (14/75) in

donors with durations >1 year. We also observed a high proportion of donors with insulitis for

diabetes durations up to 12 years (18/40 (45%)). At the same time, our overall insulitis

prevalence of 23% (18/79) in donors with type 1 diabetes compares well with the overall insulitis

prevalence rate of 21% (45/213) from former studies (6). Heterogeneity in the distribution of

insulitic islets could partly explain the lower detection rates from former studies using autopsy

collections or biopsies as fewer blocks and pancreas regions could be tested per patient (5; 7).

Page 17 of 40

Diabetes Page 18 of 58

Studies by Oram et al. and several others support the persistence of βcells in patients with long standing type 1 diabetes (11; 2830). Our data also show the persistence of βcells in donors with type 1 diabetes, with and without insulitis, up to 12 years post onset. While other investigators have also reported insulin+ islets without insulitis in patients with type 1 diabetes, few have emphasized that insulin islets are also involved in the insulitic process (reviewed in

(31)). The proportions of insulin+ and insulin insulitic islets in our study (33% insulin+, 2% insulin) were similar to those reported by Foulis and others (2835% insulin+, 15% insulin) (5;

32). Persistent insulitis after loss of βcells could imply the presence of degranulated and/or dedifferentiated βcells, undetected by these methods, or alternatively, spread of autoimmunity or

“innocent bystander” effects on other endocrine cells. Prospective studies demonstrated that appearance of multiple islet autoantibodies is usually sequential rather than simultaneous, and that their number (≥2) is a strong predictor of progression to type 1 diabetes (1). Herein, insulitis was observed in 2/5 (40%) donors with multiple autoantibodies, a finding similar to In’t Veld et al. where insulitis was detected in 2/7 (29%) donors with multiple autoantibodies (7). Of note, the AAb+ donors with insulitis in our study were younger (21 and 22 years compared to 46 and

52.5 years) and the entire pancreas was available for study compared to the former study of biopsy material. Interestingly, all four donors showed similar insulitis frequencies (1.46.4%; 3

9%) and normal βcell mass, and all four donors had the autoantibody combination of GADA+ and IA2A+.

Another novel aspect of our study is in showing similarities between the preclinical and clinical phases of type 1 diabetes in the leukocyte subtypes in insulitic islets and independent of the presence of residual βcells. Both phases were characterized by diffuse and/or focal aggregates and varying numbers of leukocytes/islet within and between donors. Several studies

Page 18 of 40

Page 19 of 58 Diabetes

show a predominance of CD8+ T lymphocytes and macrophages in insulitic islets (7; 30; 3238).

The defining study was considered that by Willcox et al. using 29 patients from the Foulis

collection, and reexamination of 21 patients from the Foulis collection with two nPOD donors

(6052, 6113) was recently reported by Arif et al. (32; 39). In the former study, insulitic islets

were ranked by both insulin percentage and immune cell numbers (n= 279 insulitic islets), while

only insulin+ islets were evaluated in the latter study. All leukocytes were significantly reduced

in insulin islets (32). In marked contrast, our study found that CD45+, CD3+, CD20+, and T cell

subsets increased linearly with numbers of insulitic islets (insulitis frequency) irrespective of

islet βcells. Potential differences between studies may result from differences in age or disease

duration or technical differences in methods.

An intriguing hypothesis by Skog et al. proposed that ascending infection through the

pancreatic ducts could result in insulitis due to greater susceptibility of βcells compared to other

endocrine cells (40). Though insulitic islets were frequently observed near pancreatic ducts in

this study, no consistent pattern was observed for the presence of periductal infiltrates and

insulitic islets. The heterogeneous leukocyte composition of insulitis might support this

hypothesis if the offending agent (virus or bacterium) or their toxic byproducts had minimal

proclivity for ductular epithelial cells.

In the present study, donors with type 1 diabetes had small pancreata regardless of age or

disease duration. Pancreas weights were not different between AAb or AAb+ donor groups

without diabetes and are similar to the agerelated changes reported in normal pancreas volume

by Saisho et al. (41). Our current findings contrast with our previous report using a subset (n=8)

of the 18 AAb+ donors in this study (42). In our first study, all donors were >18 years and

excluded if pancreatitis was noted in the medical chart or by histopathology. Age or pancreatitis

Page 19 of 40

Diabetes Page 20 of 58

exclusion criteria were not utilized in the current study. As it is difficult to predict when organ

donors with multiple AAb+ could progress to type 1 diabetes, pancreas size loss in the natural

history of type 1 diabetes might be best studied in longitudinal clinical trials of AAb+ subjects by noninvasive radiography (43; 44). Beyond this, the notion of pancreatitis in type 1 diabetes is

of evolving interest, given histopathological findings of multifocal CD3+ infiltrates in acinar

regions of donors with type 1 diabetes. Indeed, RodriguezCalvo et al. reported an increased

number of exocrine CD8+ T cells in nPOD donors with type 1 and (45).

Reports indicate marked variability in βcell area or mass in normal adults with adult

levels reached by 5 years (17; 18; 46; 47). Indeed, some donors without diabetes (both AAb+ and

AAb) exhibit βcell mass within the range observed for type 1 diabetes donors. Reasons for this

heterogeneity are unknown but could result from host genetic and environmental factors

including in utero and neonatal periods when βcell mass is increasing. Rising rates of obesity

could also account for some variability. Our βcell area and mass data are in agreement with past

literature also reporting high variability in both measures of islet βcell content. For instance, our

mean control βcell area (1.2%) was similar to that reported in children and young adults by

Meier et. al. [1.7% (5.715 years of age), 1.3% (18.521.5 years of age)] (18) as well as In’t

Veld’s report of AAb+ donors without diabetes (mean relative βcell area of 1.2% (0.51–2.61)

(7). In the report by In’t Veld et al., comparison of βcell areas and mass in AAb+ and AAb

donors did not show a significant difference (7). Two other groups have also reported normal β

cell mass in patients with autoantibodies (48; 49). Interestingly, βcell mass in five donors

without diabetes was in the upper range found for donors with T1D and insulitis and was related

to young donor age, low pancreas weights, and/or low βcell area.

Page 20 of 40

Page 21 of 58 Diabetes

In terms of potential limitations, it must be emphasized that these data were obtained

from pancreata of multiorgan donors with brain death who were subjected to intensive care

procedures. Interpretations of data must be considered, as posed by In’t Veld et al., in light of

medications received during critical care and duration of intensive care (reviewed in (50)). The

majority of donors in this study had short hospitalization durations, yet further studies are needed

to both validate and expand these findings. The computerassisted image analysis algorithm used

in this study has inherent limitations with single minimum and maximum intensity thresholds

applied to all sections. Alternatively, its unbiased approach utilizing the entire section cross

sectional areas could add greater consistency compared to manual pointcounting methods.

When studying organ donors, there is the possibility that the referring/initial diagnosis provided

to nPOD may be of questionable nature. To address this, full medical histories are carefully

reviewed by Board Certified Endocrinologists and Pathologists. Even when histories are limited,

we carefully consider age at diagnosis, BMI, time to insulin dependence, history of insulin

dependence, treatment with oral agents, cpeptide, HLA, presence of DKA, ketosis, and more.

In sum, the proportion of insulitic islets (insulitis frequency) in donors with type 1

diabetes was inversely associated with disease duration and not age at onset, number of

autoantibodies, or HLA genotype. Numbers of donors with insulitis (insulitis prevalence) was

higher in older patients and for longer disease durations in both young and older patients than

previously reported. In donors with type 1 diabetes, proportions of insulin+ islets with insulitis

was higher than in insulin islets, yet insulin islets were also affected by insulitis, particularly in

donors with multiple AAb+. Insulitic lesions were composed of a mixture of B and T

lymphocytes whose numbers increased with insulitis frequency. The βcell mass was

significantly higher in donors with type 1 diabetes and insulitis compared to donors without

Page 21 of 40

Diabetes Page 22 of 58

insulitis while βcell mass was similar between AAb+ donors and AAb donors. These data

underscore the chronic and heterogeneous nature of insulitis in type 1 diabetes. The presence of

insulitis as well as insulin+ islets several years after diagnosis, and the limited correlation between insulitis frequency and disease duration suggest that the chronicity of islet

autoimmunity extends well into the post–diagnosis period. Together with growing evidence for

the chronicity of autoimmune responses and for the persistence of Cpeptide secretion in patients, even decades after onset, our findings support the existence of a potential long window of therapeutic opportunity (51; 52).

Page 22 of 40

Page 23 of 58 Diabetes

Acknowledgments. The authors acknowledge Dr. Alvin Powers (Vanderbilt University,

Memphis, TN) and Dr. Jake Kushner (Baylor College of Medicine, Houston, TX) for nPOD

donor referrals and Robert Pietras for image analysis software design. The authors thank Dr.

Amanda Posgai (University of Florida) for editorial assistance. All nPOD staff members are

thanked for donor recovery, data collection, and helpful discussions. Organ Procurement

Organizations partnering with nPOD are listed at http://www.jdrfnpod.org/forpartners/npod

partners. Immunohistochemistry slides used in this study are available for viewing through the

nPOD Online Pathology database (21). This study used data from the Organ Procurement and

Transplantation Network (OPTN) supplied by the United Network for Organ Sharing as the

contractor for the OPTN. The interpretation and reporting of these data are the responsibility of

the author(s) and in no way should be seen as an official policy of or interpretation by the OPTN

or the U.S. Government.

Funding. This work was supported by the JDRF (252013268, 1720123, and 252012516

M.CT., M.A.A., A.P., J.S.K.) and NIDDK (UC4 DK 104155 01, 1DP3DK10112001 M.CT.,

M.A.A., D.A.S.).

Duality of interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. M.C.T. designed the experiments, performed pathology reviews, image

analyses, and statistical calculations, and wrote the manuscript. A.F. performed experiments and

edited the manuscript. C.W. designed the autoantibody studies, performed experiments, and

edited the manuscript. J.S.K. and A.P. researched data, performed statistical calculations and

contributed to discussion and editing of the manuscript. D.A.S and M.A.A. contributed to study

design and discussion and edited the manuscript. M.C.T. is the guarantor of this work and, as

Page 23 of 40

Diabetes Page 24 of 58

such, had full access to all the data in the study and takes responsibility for the integrity of the

data and the accuracy of the data analysis.

Prior Presentation. Preliminary immunotyping results were presented at the Immunology of

Diabetes Society 12th International Congress, Vancouver Island, Canada, June 1519, 2012 and

13th International Congress, Lorne, Australia, December 711, 2013. Pancreas weights were

reported on a subset of these donors (53). Of the 159 nPOD cases presented herein, 43 were previously examined by Coppieters et al (Suppl. Table 5).

Page 24 of 40

Page 25 of 58 Diabetes

References

1. Atkinson MA, Eisenbarth GS, Michels AW: Type 1 diabetes. Lancet 2014;383:6982

2. Winter WE, Schatz DA: Autoimmune markers in diabetes. Clin Chem 2011;57:168175

3. Brorsson CA, Onengut S, Chen WM, Wenzlau J, Yu L, Baker P, Williams AJ, Bingley PJ,

Hutton JC, Eisenbarth GS, Concannon P, Rich SS, Pociot F, Consortium TDG: Novel

Association Between ImmuneMediated Susceptibility Loci and Persistent Autoantibody

Positivity in Type 1 Diabetes. Diabetes 2015;64:30173027

4. CampbellThompson ML, Atkinson MA, Butler AE, Chapman NM, Frisk G, Gianani R,

Giepmans BN, von Herrath MG, Hyöty H, Kay TW, Korsgren O, Morgan NG, Powers AC,

Pugliese A, Richardson SJ, Rowe PA, Tracy S, In't Veld PA: The diagnosis of insulitis in human

type 1 diabetes. Diabetologia 2013;56:25412543

5. Foulis AK, Liddle CN, Farquharson MA, Richmond JA, Weir RS: The histopathology of the

pancreas in type 1 (insulindependent) diabetes mellitus: a 25year review of deaths in patients

under 20 years of age in the United Kingdom. Diabetologia 1986;29:267274

6. In't Veld P: Insulitis in human type 1 diabetes: The quest for an elusive lesion. Islets

2011;3:131138

7. In't Veld P, Lievens D, De Grijse J, Ling Z, Van der Auwera B, PipeleersMarichal M, Gorus

F, Pipeleers D: Screening for insulitis in adult autoantibodypositive organ donors. Diabetes

2007;56:24002404

8. Gianani R, Putnam A, Still T, Yu L, Miao D, Gill RG, Beilke J, Supon P, Valentine A, Iveson

A, Dunn S, Eisenbarth GS, Hutton J, Gottlieb P, Wiseman A: Initial results of screening of

nondiabetic organ donors for expression of islet autoantibodies. J Clin Endocrinol Metab

2006;91:18551861

Page 25 of 40

Diabetes Page 26 of 58

9. Wiberg A, Granstam A, Ingvast S, Härkönen T, Knip M, Korsgren O, Skog O:

Characterization of Human Organ Donors Testing Positive for Type 1 DiabetesAssociated

Autoantibodies. Clin Exp Immunol 2015;

10. Krogvold L, Edwin B, Buanes T, Ludvigsson J, Korsgren O, Hyöty H, Frisk G, Hanssen KF,

DahlJørgensen 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;57:841843

11. Oram RA, Jones AG, Besser RE, Knight BA, Shields BM, Brown RJ, Hattersley AT,

McDonald TJ: The majority of patients with longduration type 1 diabetes are insulin

microsecretors and have functioning beta cells. Diabetologia 2013;57:187191

12. Greenbaum CJ, Beam CA, Boulware D, Gitelman SE, Gottlieb PA, Herold KC, Lachin JM,

McGee P, Palmer JP, Pescovitz MD, KrauseSteinrauf H, Skyler JS, Sosenko JM, Group TDTS:

Fall in Cpeptide during first 2 years from diagnosis: evidence of at least two distinct phases

from composite Type 1 Diabetes TrialNet data. Diabetes 2012;61:20662073

13. Tauriainen S, Salmela K, Rantala I, Knip M, Hyöty H: Collecting highquality pancreatic

tissue for experimental study from organ donors with signs of βcell autoimmunity. Diabetes

Metab Res Rev 2010;26:585592

14. CampbellThompson M, Wasserfall C, Kaddis J, AlbaneseO'Neill A, Staeva T, Nierras C,

Moraski J, Rowe P, Gianani R, Eisenbarth G, Crawford J, Schatz D, Pugliese A, Atkinson M:

Network for Pancreatic Organ Donors with Diabetes (nPOD): developing a tissue biobank for

type 1 diabetes. Diabetes Metab Res Rev 2012;28:608617

15. Pugliese A, Vendrame F, Reijonen H, Atkinson MA, CampbellThompson M, Burke GW:

New insight on human type 1 diabetes biology: nPOD and nPODtransplantation. Curr Diab Rep

2014;14:530

Page 26 of 40

Page 27 of 58 Diabetes

16. Pugliese A, Yang M, Kusmarteva I, Heiple T, Vendrame F, Wasserfall C, Rowe P, Moraski

JM, Ball S, Jebson L, Schatz DA, Gianani R, Burke GW, Nierras C, Staeva T, Kaddis JS,

CampbellThompson M, Atkinson MA: The Juvenile Diabetes Research Foundation Network for

Pancreatic Organ Donors with Diabetes (nPOD) Program: goals, operational model and

emerging findings. Pediatr Diabetes 2014;15:19

17. Gregg BE, Moore PC, Demozay D, Hall BA, Li M, Husain A, Wright AJ, Atkinson MA,

Rhodes CJ: Formation of a Human βCell Population within Is Set Early in

Life. J Clin Endocrinol Metab 2012;97:31973206

18. Meier J, Butler A, Saisho Y, Monchamp T, Galasso R, Bhushan A, Rizza R, Butler P: Beta

cell replication is the primary mechanism subserving the postnatal expansion of betacell mass in

humans. Diabetes 2008;57:15841594

19. Fineberg SE, Kawabata TT, FincoKent D, Fountaine RJ, Finch GL, Krasner AS:

Immunological responses to exogenous insulin. Endocr Rev 2007;28:625652

20. CampbellThompson ML, Montgomery EL, Foss RM, Kolheffer KM, Phipps G, Schneider

L, Atkinson MA: Collection protocol for human pancreas. J Vis Exp 2012:e4039

21. CampbellThompson ML, Heiple T, Montgomery E, Zhang L, Schneider L: Staining

protocols for human pancreatic islets. J Vis Exp 2012;63:4068

22. Butler AE, CampbellThompson M, Gurlo T, Dawson DW, Atkinson M, Butler PC: Marked

Expansion of Exocrine and Endocrine Pancreas with Incretin Therapy in Humans with increased

Exocrine Pancreas Dysplasia and the potential for Glucagonproducing Neuroendocrine Tumors.

Diabetes 2013;62(7):2595604

23. D'Agostino RB: Propensity score methods for bias reduction in the comparison of a treatment

to a nonrandomized control group. Stat Med 1998;17:22652281

Page 27 of 40

Diabetes Page 28 of 58

24. Austin PC: Optimal caliper widths for propensityscore matching when estimating

differences in means and differences in proportions in observational studies. Pharm Stat

2011;10:150161

25. CocaPerraillon M: Local and global optimal propensity score matching. Proceedings of the

SAS Global Forum, SAS Institute, Inc., 2007

26. CocaPerraillon M: Matching with propensity scores to reduce bias in observational studies.

Philadelphia, PA, Proceedings of the Northeast SAS Users Group Conference, 2006

27. GonzalezGalarza FF, Christmas S, Middleton D, Jones AR: Allele frequency net: a database

and online repository for immune gene frequencies in worldwide populations. Nucleic Acids Res

2011;39:D913919

28. Keenan HA, Sun JK, Levine J, Doria A, Aiello LP, Eisenbarth G, BonnerWeir S, King GL:

Residual insulin production and pancreatic ßcell turnover after 50 years of diabetes: Joslin

Medalist Study. Diabetes 2010;59:28462853

29. Liu EH, Digon BJ, Hirshberg B, Chang R, Wood BJ, Neeman Z, Kam A, Wesley RA, Polly

SM, Hofmann RM, Rother KI, Harlan DM: Pancreatic function persists in many patients with chronic type 1 diabetes, but is not dramatically improved by prolonged

immunosuppression and euglycaemia from a beta cell allograft. Diabetologia 2009;52:13691380

30. Coppieters KT, Dotta F, Amirian N, Campbell PD, Kay TW, Atkinson MA, Roep BO, von

Herrath MG: Demonstration of isletautoreactive CD8 T cells in insulitic lesions from recent

onset and longterm type 1 diabetes patients. J Exp Med 2012;209:5160

31. Gepts W: Pathologic anatomy of the pancreas in juvenile diabetes mellitus. Diabetes

1965;14:619633

Page 28 of 40

Page 29 of 58 Diabetes

32. Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG: Analysis of islet inflammation

in human type 1 diabetes. Clin Exp Immunol 2009;155:173181

33. Hanafusa T, Imagawa A: Insulitis in human type 1 diabetes. Ann N Y Acad Sci

2008;1150:297299

34. Itoh N, Hanafusa T, Miyazaki A, Miyagawa J, Yamagata K, Yamamoto K, Waguri M,

Imagawa A, Tamura S, Inada M: Mononuclear cell infiltration and its relation to the expression

of major histocompatibility complex antigens and adhesion molecules in pancreas biopsy

specimens from newly diagnosed insulindependent diabetes mellitus patients. J Clin Invest

1993;92:23132322

35. Dotta F, Censini S, van Halteren A, Marselli L, Masini M, Dionisi S, Mosca F, Boggi U,

Muda A, Prato S, Elliott J, Covacci A, Rappuoli R, Roep B, Marchetti P: Coxsackie B4 virus

infection of beta cells and natural killer cell insulitis in recentonset type 1 diabetic patients. Proc

Natl Acad Sci U S A 2007;104:51155120

36. Bottazzo GF, Dean BM, McNally JM, MacKay EH, Swift PG, Gamble DR: In situ

characterization of autoimmune phenomena and expression of HLA molecules in the pancreas in

diabetic insulitis. N Engl J Med 1985;313:353360

37. Hänninen A, Jalkanen S, Salmi M, Toikkanen S, Nikolakaros G, Simell O: Macrophages, T

cell receptor usage, and endothelial cell activation in the pancreas at the onset of insulin

dependent diabetes mellitus. J Clin Invest 1992;90:19011910

38. Somoza N, Vargas F, RouraMir C, VivesPi M, FernándezFigueras MT, Ariza A, Gomis R,

Bragado R, Martí M, Jaraquemada D: Pancreas in recent onset insulindependent diabetes

mellitus. Changes in HLA, adhesion molecules and autoantigens, restricted T cell receptor V

beta usage, and cytokine profile. J Immunol 1994;153:13601377

Page 29 of 40

Diabetes Page 30 of 58

39. Arif S, Leete P, Nguyen V, Marks K, Nor NM, Estorninho M, KronenbergVersteeg D,

Bingley PJ, Todd JA, Guy C, Dunger DB, Powrie J, Willcox A, Foulis AK, Richardson SJ, de

Rinaldis E, Morgan NG, Lorenc A, Peakman M: Blood and islet phenotypes indicate

immunological heterogeneity in type 1 diabetes. Diabetes 2014;63:38353845

40. Skog O, Korsgren S, Melhus A, Korsgren O: Revisiting the notion of type 1 diabetes being a

Tcellmediated autoimmune disease. Curr Opin Endocrinol Diabetes Obes 2013;20:118123

41. Saisho Y, Butler AE, Meier JJ, Monchamp T, AllenAuerbach M, Rizza RA, Butler PC:

Pancreas volumes in humans from birth to age one hundred taking into account sex, obesity, and presence of type2 diabetes. Clin Anat 2007;20:933942

42. CampbellThompson M, Wasserfall C, Montgomery EL, Atkinson MA, Kaddis JS: Pancreas organ weight in individuals with diseaseassociated autoantibodies at risk for type 1 diabetes.

JAMA 2012;308:23372339

43. Gaglia JL, Guimaraes AR, Harisinghani M, Turvey SE, Jackson R, Benoist C, Mathis D,

Weissleder R: Noninvasive imaging of pancreatic islet inflammation in type 1A diabetes patients. J Clin Invest 2011;121:442445

44. Williams AJ, Thrower SL, Sequeiros IM, Ward A, Bickerton AS, Triay JM, Callaway MP,

Dayan CM: Pancreatic Volume Is Reduced in Adult Patients with Recently Diagnosed Type 1

Diabetes. J Clin Endocrinol Metab 2012;97(11):E210913

45. RodriguezCalvo T, Ekwall O, Amirian N, ZapardielGonzalo J, von Herrath MG: Increased

Immune Cell Infiltration of the Exocrine Pancreas: A Possible Contribution to the Pathogenesis of Type 1 Diabetes. Diabetes 2014;63(11):38803890

46. Saisho Y, Butler AE, Manesso E, Elashoff D, Rizza RA, Butler PC: βcell mass and turnover in humans: effects of obesity and aging. Diabetes Care 2013;36:111117

Page 30 of 40

Page 31 of 58 Diabetes

47. Rahier J, Guiot Y, Goebbels RM, Sempoux C, Henquin JC: Pancreatic betacell mass in

European subjects with type 2 diabetes. Diabetes Obes Metab 2008;10 Suppl 4:3242

48. Wagner R, McNally JM, Bonifacio E, Genovese S, Foulis A, McGill M, Christie MR,

Betterle C, Bosi E, Bottazzo GF: Lack of immunohistological changes in the islets of

nondiabetic, autoimmune, polyendocrine patients with betaselective GADspecific islet cell

antibodies. Diabetes 1994;43:851856

49. Oikarinen M, Tauriainen S, Honkanen T, Vuori K, Karhunen P, VasamaNolvi C, Oikarinen

S, Verbeke C, Blair GE, Rantala I, Ilonen J, Simell O, Knip M, Hyöty H: Analysis of pancreas

tissue in a child positive for islet cell antibodies. Diabetologia 2008;51:17961802

50. Atkinson MA: Losing a grip on the notion of βcell specificity for immune responses in type

1 diabetes: can we handle the truth? Diabetes 2014;63:35723574

51. WilmotRoussel H, Lévy DJ, Carette C, CaillatZucman S, Boitard C, Timsit J, Dubois

Laforgue D: Factors associated with the presence of glutamic acid decarboxylase and islet

antigen2 autoantibodies in patients with longstanding type 1 diabetes. Diabetes Metab

2013;39:244249

52. Oram RA, McDonald TJ, Shields BM, Hudson MM, Shepherd MH, Hammersley S, Pearson

ER, Hattersley AT, Team obotU: Most People With LongDuration Type 1 Diabetes in a Large

PopulationBased Study Are Insulin Microsecretors. Diabetes Care 2014;38:323328

53. CampbellThompson M, Kaddis J, Wasserfall C, Haller M, Pugliese A, Schatz D, Shuster J,

Atkinson M: The influence of type 1 diabetes on pancreatic weight. Diabetologia 2015;(in press)

Page 31 of 40

Diabetes Page 32 of 58

Table 1. Insulitis frequency in donors. Multiple slide sections were reviewed from each donor pancreas and islets were counted by insulin immunopositivity (Ins+, Ins) and insulitis status (CD3 (negative), CD3+ (positive)). the total number of insulitic islets divided by total islet numbers was used to calculate insulitis frequency (%) for each donor. Donors were listed in order of increasing insulitis frequency (%).Demographic information for ages at demise and diabetes onset, diabetes duration, and autoantibody types were also shown.

Insulitis Case No. Ins+ Ins+ Ins Ins Total Age Diabetes Age Frequency § || ID Sections CD3 CD3+ CD3+ CD3 Islets* Onset Duration Demise Autoantibody Results (%) † (yrs) (yrs) (yrs) 6264 6 7 3 0 1017 1027 0.3% 3.00 9.00 12.00 Negative 6212 12 51 13 10 1601 1675 1.4% 15.00 5.00 20.00 6268 5 0 1 13 808 822 1.7% 9.00 3.00 12.00 6265 6 8 6 5 508 527 2.1% 3.00 8.00 11.00 GADA+ 6039 4 0 2 5 324 331 2.1% 16.70 12.00 28.70 GADA+ IA2A+ ZnT8A+ 6046 5 92 2 6 268 368 2.2% 10.80 8.00 18.80 IA2A+ ZnT8A+ 6113 14 21 28 9 1640 1698 2.2% 11.52 1.58 13.10 6224 8 30 24 12 1486 1552 2.3% 19.50 1.50 21.00 Negative 6088 7 73 7 6 433 519 2.5% 26.20 5.00 31.20 GADA+ IA2A+ ZnT8A+

T1D 6245 6 131 7 28 1021 1187 2.9% 15.00 7.00 22.00 GADA+ IA2A+ 6247 6 77 20 25 1068 1190 3.8% 23.40 0.60 24.00 6195 16 0 2 53 1385 1440 3.8% 14.20 5.00 19.20 GADA+ IA2A+ ZnT8A+ 6070 4 56 15 8 333 412 5.6% 15.60 7.00 22.60 IA2A+ 6198 2 16 9 5 181 211 6.6% 19.00 3.00 22.00 GADA+ IA2A+ ZnT8A+ 6052 9 10 27 41 921 999 6.8% 11.00 1.00 12.00 IA2A+ 6209 11 32 42 54 1247 1375 7.0% 4.75 0.25 5.00 IA2A+ ZnT8A+ 6243 8 162 55 32 859 1108 7.9% 8.00 5.00 13.00 6228 15 250 246 74 2447 3017 10.6% 13.00 0.00 13.00 GADA+ IA2A+ ZnT8A+ Total 144 1,016 509 386 17,547 19,458 6197 5 1981 28 0 4 2013 1.4% 22.00 GADA+ IA2A+ AAb+¶ 6267 13 2149 139 8 18 2314 6.4% 23.00 GADA+ IA2A+

Total 18 4,130 167 8 22 4,327

Page 32 of 40

Page 33 of 58 Diabetes

* Total Islets calculated as sum of 4 islet subtypes. †Insulitis frequency (%) calculated as sum insulitic islets (Ins+ CD3+ and Ins CD3+)/Total Islets. ‡ Calculated as age at demise minus diabetes duration. § All donors with insulitis had autoantibody testing by RIA. RIA data values were converted to NIDDK units and defined as positive if one or more of the following applied: GADA if >20, IA2A if >5, or ZnT8A if >0.020. || IAA were not listed for donors with T1D as they could not be distinguished as autoantibodies versus antibodies to parenteral insulin as described in Methods. An empty field indicates that only insulin autoantibodies were detected by RIA. ¶ AAb+ donors without diabetes.

Page 33 of 40

Diabetes Page 34 of 58

Table 2. Table of insulitis prevalence in nPOD donors by age of onset and diabetes duration.

Diabetes Duration ≤ 1 year Diabetes Duration > 1 year Totals*

Age at Number Total Proportion Number Total Proportion Number Total Proportion

Onset Donors Number with Donors Number with Donors Number with

(Years) Insulitis+ Donors Insulitis% Insulitis+ Donors Insulitis% Insulitis+ Donors Insulitis%

1.414.2 3 3 100% 7 51 14% 10 54 19%

1536 1 1 100% 7 24 29% 8 25 32%

Total 4 4 100% 14 75 19% 18 79 23%

Age groups and diabetes durations adapted from In’t Veld (6).

*One donor with T1D had unknown duration of disease (Methods).

Page 34 of 40

Page 35 of 58 Diabetes

Figure Legends

Figure 1. Lobular variability in insulin+ islets and insulitis. Islets were imaged from a 13year

old patient with T1D for 5 years (6243). Serial paraffin sections were stained for Ki67+INS (A,

C, E) and CD3+GCG (B, D,F) and islets were subtyped as described in Methods. Five insulin+

islets (A) are seen in a lobule adjacent to a lobule with two insulin islets in the patient with T1D

(A, B, blue arrows). Three insulin+ islets had insulitis (B, black arrows) and both insulin islets

did not have insulitis. One of the insulin+ islet with insulitis is shown at higher magnification

((C, D) as well as an insulin insulitis islet (E, F). Few islet cells were Ki67+ (A, C) indicating

no effect of insulitis on proliferating cell numbers. Scale bars: A, B 500m; C-F 50µm.

Figure 2 Heterogeneity of islet βcells and numbers of CD3+ cells in insulitic islets in young

donors at diabetes onset and after 5 years duration. Representative islets were imaged for a 13

yearold at diabetes onset (6228; A-F) and another 13yearold with T1D for 5 years (6243; G-L).

Serial paraffin sections were stained using dual immunohistochemistry (Ki67+Insulin,

CD3+Glucagon) as described in Methods. Decreasing proportions of βcells are shown in

different islets from both donors (A-C, G-I) as well as heterogeneity in numbers of CD3+

cells/islet (D-F, J-L). Heterogeneity in CD3+ counts between two insulin islets of similar sizes

is also shown for one donor (K, L). Scale bars: A-F, H, K: 50m; G, I, J, L: 100µm.

Figure 3 Insulitis frequency in relation to diabetes duration and donor ages at demise or disease

onset. The insulitis frequency (%, total insulitic islets/total islets) is shown on the Y axis in

comparison to years for diabetes duration (A), age at T1D onset (B), and age at demise (C). Data

were displayed (pink) for the two AAb+ donors with insulitis for age at demise but were

excluded from statistical analyses due to small sample size. Insulitis frequency (%) had a low but

Page 35 of 40

Diabetes Page 36 of 58

significant correlation to diabetes duration but not to donor age at demise or disease onset. Linear

regression line with Spearmen r and p values are shown.

Figure 4. Insulitis leukocyte subtyping by multiple immunofluorescence. Serial sections of an islet with insulitic aggregate were imaged from a 5yearold donor with T1D for 4 months

(6209). Numbers of leukocytes/islet were counted for total leukocytes (CD45, A), B and T

lymphocytes (CD20, CD3, B), T lymphocyte subsets (CD8, CD4, C), dendritic cells and

macrophages (CD11c, CD68, D) as described in Methods. Islet endocrine cells were identified

using glucagon (GCG) and insulin (INS) stains. The merged images (A-D) include DAPIstained

DNA and the individual fluorescence channels are shown below the merged images. Scale bars:

50µm

Figure 5 Leukocyte numbers increase in parallel with CD45+ and CD3+ numbers but are heterogeneous between islets. Numbers of CD45+, CD3+, CD20+, CD8+ and CD4+ cells were counted for each insulitic islet and averaged per donor (n=13 donors with T1D, 210 islets/donor). Data were also displayed (pink) for each leukocyte marker for the two AAb+ donors with insulitis for comparison purposes but were excluded from statistical analyses due to small sample size. Correlations between numbers of leukocytes/islet were performed by

Spearman analysis with linear regression lines shown. Numbers of CD45+ cells/islet were significantly correlated with numbers of CD3+ and CD20+cells/islet (A). Numbers of CD3+ cells/islet were also significantly correlated with numbers of CD8+ and CD4+ cells/islets (B).

Numbers of leukocyte subtypes/islet were also analyzed to examine variability in cell numbers between all islets for donors with T1D and two AAb+ donors with insulitis with means depicted

Page 36 of 40

Page 37 of 58 Diabetes

by boxandwhisker plots with Tukey error bars (nonparametric) (C). Numbers in parentheses

following each marker indicate the total numbers of individual islets analyzed for each leukocyte

marker (CD45) or set of markers. Variability in overall numbers of leukocytes/islet was

observed. Numbers of leukocytes /islet had similar trends in AAb+ donors without diabetes.

Figure 6 Pancreas weights and βcell area and mass are decreased in donors with T1D. Group

means were depicted by boxandwhisker plots with Tukey error bars (nonparametric). Data

were displayed (pink) for the two AAb+ donors with insulitis and were excluded from statistical

analyses due to small sample size (n=2). Pancreas weights were significantly lower in donors

with T1D compared to AAb and AAb+ donors without diabetes and no difference was detected

between donors with T1D based on insulitis status (A). The βcell area was significantly lower in

both donor groups with T1D compared to nondiabetic AAb and AAb+ donors (B). T1D donors

with insulitis had a significantly higher βcell area compared to T1D donors without insulitis (B).

The βcell mass was similar between AAb and AAb+ donors and was significantly different

from donor groups with T1D (C). T1D donors with insulitis had significantly higher insulin+ β

cell mass compared to T1D donors without insulitis (C). The αcell area (D) and mass (E) were

not significantly different between groups. *Significantly different from AAb and AAb+ donors

(P<0.001), **Significantly different from AAb and AAb+ donors (P<0.001) and T1D donors

without insulitis (P<0.05).

Supplementary Figure 1. Insulitic islets in AAb+ donors without diabetes. Islets were imaged

from a 22yearold donor (6197; A-B) and a 23yearold donor (6267; C-D). Cells in the insulitic

islets were immunotyped using multiple immunofluorescence as described in Methods. Islet

endocrine cells were identified using glucagon (GCG) and insulin (INS) stains. Heterogeneous

Page 37 of 40

Diabetes Page 38 of 58

distribution of Ins+ and Ins islets was observed between lobules (A). An insulitic islet is shown with diffuse infiltration by CD45+ cells (B). A large insulitic aggregate is shown composed of similar numbers of CD20+ and CD3+ lymphocytes (C). Another large islet with fewer CD20+ and CD3+ cells demonstrates the wide variability between leukocyte numbers/islet within a given donor (D). Scale bars: 200µm A, 50 µm B, D, 100µm C.

Supplementary Figure 2. B and T lymphocytes in insulitic islets from donors with long standing T1D. Focal aggregates in islets were imaged from a 12yearold donor with T1D for 3 years (6268) (A-D) and a 22.6yearold patient with T1D for 7 years (6070) (E-I). Sections were stained for CD20 (B, F), CD3 (C, G), CD8 and CD4 (I), glucagon (GCG, D), insulin (INS, H), or chromogranin A (CGA, I) as described in Methods. Both CD20+ and CD3+ lymphocytes were found in insulitic aggregates though in varying numbers. DAPIstained DNA to delineate nuclei is shown in merged images (A, E, I). Scale bars: 50µm.

Supplementary Figure 3 Leukocytes in insulitic islets from insulin+ and insulin islets. Two islets in a pancreas section were imaged from a 13yearold donor with T1D for 5 years (6243)

(insulin+ islet (A-D); insulin islet (E-H)). Serial sections were stained as described in Methods with endocrine cells identified using glucagon (GCG) and insulin (INS) stains. Aggregates in both islets were comprised of both CD20+ and CD3+ cells (C, G) as well as CD8+ and CD4+ T cells (D, H). Dendritic cells (CD11c+) and CD68+ macrophages were observed adjacent to mononuclear aggregates and endocrine cells (B, F). A large number of macrophages (CD68) are also seen in the adjacent exocrine region in panel B. Scale bars: A-D 50m; E-H 100m.

Supplementary Figure 4. Heterogeneity in leukocyte markers in insulitic islets and correlations with insulitis frequency. Mean numbers of leukocyte subtypes / insulitic islets from each donor

Page 38 of 40

Page 39 of 58 Diabetes

with T1D were plotted in comparison to insulitis frequency (%) (A-G). Data were displayed

(pink) for the two AAb+ donors with insulitis and were excluded from statistical analyses due to

small sample size. The ratio of CD3/(CD3+CD20) was also calculated and plotted by insulitis

frequency (H). Numbers of CD45+, CD3+, CD20+, CD8+, and CD4+ cells/islet had variable

significant correlations with insulitis frequency but numbers of CD68+ and CD11c+ cells/islet or

the ratio of CD3+/(CD3+CD20) did not. Linear regression lines with Spearmen r and p values

are shown.

Supplementary Figure 5. Pancreas weight and βcell area and mass by donor age and group.

Data from donors with no diabetes (AAb (A, D, G), AAb+ (B, E, H)) or T1D (C, F, I) were

depicted with data for donors with insulitis indicated in color (AAb+, pink; T1D, blue) within

each respective group. An xaxis of 080 years was used in all plots; although, some older donors

did not have pancreas weight data due to receipt of partial pancreas sample (n=3) or no recording

(n=13). Smaller Y axis scales were used for donors with T1D in panels for βcell area (F) and

mass (I) to allow better visualization of the significant differences, as shown in Fig. 6, for these

two variables by insulitis status within this group. Pancreas weights were plotted with regression

lines from bestfit second order polynomial equations (A-C). Variations in βcell area (%) were

seen by age in donors without diabetes (AAb, AAb+) (D-E) and in donors with T1D (F). The β

cell mass (mg) was calculated from βcell area (%) multiplied by pancreas weight (gram) (G-I)

and similar variations were observed as for βcell areas.

Supplementary Figure 6. βcell mass from donors with T1D and insulitis in comparison to

insulitis frequency, diabetes duration and age at onset. The βcell mass in donors with T1D and

insulitis was not significantly correlated with insulitis frequency (A), diabetes duration (B) or age

Page 39 of 40

Diabetes Page 40 of 58

at onset (C) due to heterogeneity in residual βcell distribution independent of age. Linear regression lines with Spearmen r and p values are shown.

Page 40 of 40

Page 41 of 58 Diabetes

A Ki67 INS B CD3 GCG

E,F C,D

C Ki67 INS D CD3 GCG E Ki67 INS F CD3 GCG

Figure 1. Figure2 .

CD3 GCG Ki67 INS CD3 GCG Ki67 INS G G A D J

K H E B B Diabetes L I C F F Page 42of58 Page 43 of 58 Diabetes

A B C

)

)

)

% %

1 5 1 5 %

( 1 5

(

(

r= 0 .1 1 p = n s r= -0 .0 2 p = n s y

y r= -0 .5 8 p = 0 .0 1

y

c

c

c

n n

n A A b +

e e 1 0 1 0 e

u 1 0

u

u

q

q

q

e

e

e

r

r

r

F

F

F

s s 5 5

s 5

i

i

i

t

t

t

i

i

i

l

l

l

u

u

u

s

s

s

n

n

n I I 0 0 I 0 0 5 1 0 1 5 0 1 0 2 0 3 0 0 1 0 2 0 3 0 4 0 D ia b e te s D u ra tio n (Y e a rs ) A g e a t O n s e t (Y e a rs ) A g e a t D e m is e (Y e a rs )

Figure 3. Diabetes Page 44 of 58

A B C D Merge Merge

CD45 CD20 CD8 CD11c

GCG CD3 CD4 CD68

INS GCG GCG INS Endocrine Endocrine

Figure 4 Page 45 of 58 Diabetes

A

t e

l 6 0

s

i /

s C D 3 + r= 0 .9 5 p < 0 .0 0 1

l l

e C D 2 0 + r= 0 .9 6 p < 0 .0 0 1 c C

+ 4 0 A A b + C D 3 + 0

2 A A b + C D 2 0 + D

1 4 0

C

r 1 2 0 o

2 0 1 0 0 +

3 8 0

D 7 0

t

C e

0 l 6 0

s I

0 5 0 1 0 0 1 5 0 / s

l 5 0

C D 4 5 + c e lls /is le t l

e c

B 4 0

] +

5 0 D 3 0

C [

2 0

t

e l

s 4 0 i

/ 1 0

s

l l

e 0 c 3 0 + C D 8 + r= 0 .8 7 p < 0 .0 0 1 ) ) ) 4 0 ) c ) ) ) 4 0 ) c

8 7 1 3 2 5 0 1 6 0 7 0 D 2 4 1 1 2 2 D 2 1 1 D C D 4 + r= 0 .9 0 p < 0 .0 0 1 1 ( 1 C D ( 1 ( ( ( C D ( 1 ( ( D D C 3 C 8 5 3 8 C 8 A A b + C D 4 + 5 8 6 C 4 + 6 C r D D D b + 4 D D D D o 2 0 C C C A b + D A A b + C D 8 + C C C A C b + + + A C + A + A

4 b b b A A b A D A A A A C 1 0 A A

0 0 2 0 4 0 6 0 8 0

C D 3 + c e lls /is le t Figure 5

Diabetes Page 46 of 58

A B C

) 1 5 0

g 4 2 5 0 0

(

)

t )

* * g

h %

m 2 0 0 0

g

( (

i 3

1 0 0

e

a

s e

s 1 5 0 0

W

r a

* ** a

s 2

l

m

a

l

l 1 0 0 0

e e

5 0 l

r

c

e

- c

1 c

- 

n 5 0 0 a

 * ** P 0 0 0 - b + is D is - + s s - + s s b t 1 t b i D i b i D i A li li b it 1 it b it 1 it A T A A l T l A A l T l A A u u A u u A u u s s A s s A s s in in n n n n I I I I + D b + D + D 1 b 1 b 1 A T A A A T T D A E A

4 2 5 0 0

)

g )

m 2 0 0 0

%

( (

3

s

a s

e 1 5 0 0

r

a a

2

m

l

l

l 1 0 0 0

l

e

e

c -

1 c -

 5 0 0  0 0 - - + s s + is D is b ti D i b b t t b i 1 it A li 1 i A A l T l A T l A u u A A u u A s s s s n n in in i i + D + D b 1 b 1 A T A T A A

Figure 6. Page 47 of 58 Diabetes

A B Insulin+ INS

Insulin- GCG CD45 GCG

C D GCG GCG

CD3

CD20

Suppl. Figure 1 Diabetes Page 48 of 58

A B CD20 C CD3 D GCG

E H INS I CD8 CD4 CGA

F CD20 G CD3

Suppl. Figure 2. Page 49 of 58 Diabetes

E CD45 GCG INS F CD11c CD68 INS

A CD45 GCG INS B CD11c CD68 INS

C CD20 CD3 GCG D CD8 CD4 GCG G CD20 CD3 GCG H CD8 CD4 GCG

Suppl. Figure 3. Diabetes Page 50 of 58 A B

1 5 0 8 0

t r= 0 .7 3 p = 0 .0 0 4

r= 0 .5 3 p = 0 .0 3 t

e

e

l l

s A A b +

s i

A A b + I 6 0

/

/ s

1 0 0 s

l

l

l

l

e e

c 4 0

C

+ +

5 5 0

3 4

D 2 0

D

C C 0 0 0 5 1 0 1 5 0 5 1 0 1 5 In s u litis F re q u e n c y (% ) C D In s u litis F re q u e n c y (% )

5 0 6 0

t r= 0 .5 6 p = 0 .0 4 r= 0 .6 2 p = 0 .0 2

t

e

e l A A b + l

s 4 0 s

i A A b +

i

/ /

s 4 0

s

l

l l

3 0 l

e

e

c

c

+ 2 0

+ 0

8 2 0

2 D

D 1 0

C C 0 0 0 5 1 0 1 5 0 5 1 0 1 5 F E In s u litis F re q u e n c y (% ) In s u litis F re q u e n c y (% )

2 5 r= 0 .6 2 p = 0 .0 2 2 5

t r= 0 .3 3 p = n s

t

e

l e

l 2 0 A A b + 2 0

s A A b +

i

s

/

i

/

s

l

s l

l 1 5 1 5

l

e

e

c

c

1 0 + 1 0

+

8

4

6 D

5 D 5

C C 0 0 0 5 1 0 1 5 0 5 1 0 1 5

H

s l

G In s u litis F re q u e n c y (% ) l In s u litis F re q u e n c y (% )

e

c

)

2 0 + 0 1 .5

t r= 0 .5 3 p = n s r= -0 .4 6 p = n s

2

e l

A A b + D s

C A A b +

i

/ 1 5

+

s l

3 1 .0

l

D

e

c

C

1 0 (

l

+

a

c t

1 0 .5

o 1

5 T

/

D

s

l

l

C

e c

0 0 .0 +

0 5 1 0 1 5 3 0 5 1 0 1 5

D C In s u litis F re q u e n c y (% ) In s u litis F re q u e n c y (% ) Suppl. Figure 4. Page 51 of 58 Diabetes

A B C

A A b +

1 5 0 A A b - 1 5 0 1 5 0

) )

) A A b + In s u litis T 1D

g

g

g

(

(

(

T 1 D In s u litis

t

t

t

h

h

h

g

g g

i 1 0 0

i 1 0 0

i 1 0 0

e

e

e

W

W

W

s

s

s

a

a

a e e 5 0

5 0 e 5 0

r

r

r

c

c

c

n

n

n

a

a

a

P

P P 0 0 0 0 2 0 4 0 6 0 8 0 0 2 0 4 0 6 0 8 0 0 2 0 4 0 6 0 8 0 A g e (y r) A g e (y r) A g e (y r) D E F

3 .5 A A b - 3 .5 0 .6 A A b + T 1D

3 .0 3 .0

) )

) A A b + In s u litis T 1 D In s u litis

%

% %

2 .5 ( 2 .5

( (

0 .4

a

a a

e e

e 2 .0 2 .0

r

r r

a

a a

l l

l 1 .5 1 .5

l

l l

e e

e 0 .2

c c

c 1 .0 1 .0

-

- -

  0 .5 0 .5

0 .0 0 .0 0 .0 0 2 0 4 0 6 0 8 0 0 2 0 4 0 6 0 8 0 0 2 0 4 0 6 0 8 0 A g e (y r) A g e (y r) A g e (y r)

G H I

2 5 0 0 2 5 0 0 2 0 0

A A b - A A b + T 1D

)

) )

g A A b + In s u litis g 2 0 0 0 2 0 0 0 g T 1 D In s u litis

m 1 5 0

m

m

(

(

(

s s

1 5 0 0 1 5 0 0 s

s

s

s

a a

a 1 0 0

m

m

m

1 0 0 0

1 0 0 0

l

l

l

l

l

l

e

e

e

c c

c 5 0

- -

5 0 0 5 0 0 -

 

0 0 0 0 2 0 4 0 6 0 8 0 0 2 0 4 0 6 0 8 0 0 2 0 4 0 6 0 8 0 A g e (y r) A g e (y r) A g e (y r)

Suppl. Figure 5. Diabetes Page 52 of 58

A B C

)

s

)

r

)

a %

1 5 1 5 s 3 0

( e r= -0 .2 7 p = n s r

r= 0 .2 1 p = n s r= -0 .0 0 p = n s

y

a

y

(

e

c

y

n

n

(

o

e i

1 0 1 0 t 2 0

t

u

e

a

q

s

r

e

n

u

r

F

D

O

t

s 5 5 1 0

i

s

a

t

i

e

l

t

e

u

e

g

s

b

A

n a

I 0 0 0 i

0 5 0 1 0 0 1 5 0 2 0 0 D 0 5 0 1 0 0 1 5 0 2 0 0 0 5 0 1 0 0 1 5 0 2 0 0

 -c e ll m a s s (m g )  -c e ll m a s s (m g )  -c e ll m a s s (m g )

Suppl. Figure 6. Page 53 of 58 Diabetes

Supplementary Table 1. Clinical characteristics of 159 organ donors from study. No Diabetes Autoantibody Positive (AAb+) T1D No Diabetes (AAb) Characteristic No Insulitis Insulitis No Insulitis Insulitis n Value* n Value* n Value* n Value* n Value* Age at Death (yr) 61 28.6 (±16.4) 16 38.8 (±17.8) 2 22.5 (±0.7) 62 30.6 (±11.9) 18 17.8 (±6.8) † Age at Diabetes Onset (yr) 61 11.4 (±8.2) 18 13.3 (±6.5) Diabetes Duration (yr) 61 19.1 (±11.5) 18 4.6 (±3.4) Ethnicity African American 5 8% 1 6% 1 50% 7 11% 1 6% White 49 82% 13 81% 1 50% 53 86% 16 88% Hispanic/Latino 6 10% 2 13% 0 0% 2 3% 1 6% Sex Female 23 38% 7 44% 1 50% 26 42% 9 50% Male 38 62% 9 56% 1 50% 36 58% 9 50% Death‡ or Admit Course§ DKA Related 0 0% 0 0% 0 0% 16 26% 8 44% Other 61 100% 15 100% 2 100% 46 74% 10 56% Whole Body Weight (kg) 61 73.7 (±21.9) 16 75.3 (±27.0) 2 76.5 (±17.7) 62 71.4 (±16.7) 18 59.5 (±19.8) BMI (kg/m2) 61 25.4 (±5.2) 16 25.8 (±6.5) 2 25.9 (±3.3) 62 25.0 (±4.1) 18 22.2 (±4.4) Cpeptide (ng/mL)|| 61 4.6 (0.4, 22.9) 14 3.9 (0.001, 26.2) 2 17.0 (16.6, 17.5) 61 0.001 (0.001, 0.140) 18 0.001 (0.001, 0.470) HbA1c (% ) 21 5.4 (±1.3) 4 5.8 (±0.8) 2 5.3 (±0.4) 19 9.8 (±2.3) 5 10.3 (±2.9) (mmol/mol) 35.1 (±14.4) 39.6 (±8.4) 33.9 (±3.9) 83.6 (±24.6) 89.1 (±32.0) Autoantibodies Detected¶ 0 61 100% 0 0% 0 0% 30 49% 7 39% 1 0 0% 13 89% 0 0% 23 38% 3 17% ≥2 0 0% 3 19% 2 100% 8 13% 8 44% Hospitalization Stay (days) 57 3.2 (0.3, 23.6) 13 3.2 (1.1, 6.8) 2 3.7 (3.0, 4.5) 60 3.3 (0.9, 11.3) 3.4 (0.9, 21.7) *% reported for all categorical variables; use of mean or median (minimum, maximum) based on evaluation of normal distribution † Calculated as age at death minus diabetes duration ‡ Cause or mechanism of death § Clinical records were examined for mention of DKA || A fill value of 0.001 was used when assay results <0.05 (lower limit of detection) ¶ All RIA data values were converted to NIDDK units and defined as positive if one or more of the following applied: GADA if >20, IA2A if >5, ZnT8A if >0.020, or insulin if >0.010. Insulin autoantibodies excluded from counts for donors with T1D as described in Methods. Three AAb+ donors with no insulitis had positive autoantibody results from the screening serum samples by ELISA and/or RIA in lieu of positive results from the serum sample obtained at pancreas recovery.

Diabetes Page 54 of 58

Supplementary Table 2. Primary and secondary antibodies used in immunolocalization assays.

Catalogue Antigen Species Clone Company Number CD11c Rabbit EP1347Y Abcam ab52632

CD3 Rabbit Polyclonal DAKO A0452 CD4 Mouse 4B12 DAKO M7310

CD8 Rabbit Sp16 Abcam ab27605 CD20cy Mouse L26 DAKO M0755

CD45 Mouse 2B11 + PD7/26 DAKO M0701 CD68 Mouse PGM1 DAKO M0876

Chromogranin A Rabbit DAKA3 DAKO M0869 Glucagon Mouse K79bB10 Abcam ab10988

Insulin Guinea Pig Polyclonal DAKO A0564 Ki67 Mouse MIB1 DAKO M7240

Goat AF488 conjugate Invitrogen A11029 Mouse IgG Rabbit IgG Goat AF555 conjugate Invitrogen A21429

Guinea pig IgG Goat AF647 conjugate Invitrogen A21450 Page 55 of 58 Diabetes

Supplementary Table 3. Clinical and analytical characteristics of matched donors with T1D with and without insulitis.

Name (%)*, Mean (± 1 SD)*, or Median (min, max)* P-value n No Insulitis n Insulitis Age at Demise (yrs) 15 27.6 (4.4, 61.0) 15 20 (12.0, 31.2) 0.005 Age at Diabetes Onset (yrs) †, ‡ 14 11.7 (±8.9) 15 14.5 (±6.0) 0.319 Diabetes Duration (yrs) 14 14.5 (3.0, 52.0) 15 5 (0.6, 12.0) <0.001 Ethnicity † 0.791 Caucasian 11 73% 13 86% African American 3 20% 1 7% Hispanic/Latino 1 7% 1 7% Sex † 0.264 Female 4 27% 8 53% Male 11 73% 7 47% Death§ or Admit Course|| 0.035 DKA related 1 7% 7 47% Other 14 93% 8 53% Height (cm) 15 172.7 (81.3, 193.0) 15 165.1 (124.5, 182.8) 0.134 Whole Body Weight (kg) 15 69.9 (±19.8) 15 65.7 (±14.1) 0.508 BMI (kg/m2)† 15 23.9 (±2.5) 15 23.9 (±2.3) 0.984 Cpeptide (ng/mL) ¶ 15 0.001 (0.001, 0.060) 15 0.001 (0.001, 0.470) 0.142 HbA1c (%) (mmol/mol) 5 11.5 (±1.5) 102.0 (±16.8) 4 9.6 (±2.8) 80.9 (±30.3) 0.223 Autoantibody Results# 0.403 0 8 53% 7 47% 1 4 27% 2 13% >2 3 20% 6 40% Hospitalization Stay (days) 14 3.2 (0.9, 11.3) 14 3.2 (0.9, 21.7) 0.801 Transport Duration (hrs) 13 16.8 (±4.6) 15 14.4 (±5.7) 0.235 Cytomegalovirus Present 0.999 No 8 62% 9 64% Yes 5 38% 5 36% History of Hypertension 0.999 No 8 62% 9 64% Yes 5 38% 5 36% Heavy Alcohol Use No 14 100% 13 100% History of Cigarette Use 0.482 Diabetes Page 56 of 58

No 12 93% 14 100% Yes 1 7% 0 0% Cocaine Use 0.222 No 13 100% 11 79% Yes 0 0% 3 21% Other Drug Use 0.648 No 10 71% 11 85% Yes 4 29% 2 15% HLA Haplotypes DRB1*04DQB1*03:02 alone 0.390 Absent 13 87% 10 67% Present 2 13% 5 33% DRB1*03:01DQB1*02:01 alone 0.999 Absent 10 67% 11 73% Present 5 33% 4 27% Both of Above 0.427 Absent 9 60% 12 80% Present 6 40% 3 20% DRB1*15:01DQB1*06:02 0.999 Absent 15 100% 14 93% Present 0 0% 1 7% Pancreas Weight (grams) 14 39.2 (11.1, 117.4) 14 33.8 (20.4, 59.8) 0.6625 Fractional Insulin Area (%) 14 0.00 (0.00, 0.14) 15 0.08 (0.00, 0.55) 0.0016 Insulin Mass (mg) 13 1.73 (0.11, 30.80) 14 26.77 (1.27, 174.18) 0.0028 * % reported for all categorical variables; use of mean or median based on evaluation of normal distribution. † Factors used for matching (see Table 1 for characteristics of all donors in study). ‡ Calculated as age at death minus diabetes duration. § Cause or mechanism of death. || Clinical records were examined for DKA in relation to cause or mechanism of death or during the terminal admission history. ¶ A fill value of 0.001 was used when assay results <0.05 (lower limit of detection). # All data values were converted to NIDDK units and defined as positive if one or more of the following applied: GADA if >20, IA2A if >5, or ZnT8A if >0.020. Insulin autoantibodies were excluded from counts for donors with T1D as described in Methods.

Page 57 of 58 Diabetes

Supplementary Table 4. Correlation analyses between numbers of leukocytes/islet and insulitis frequency, diabetes duration, and age of onset. Mean numbers of leukocytes/islet for each donor with T1D and insulitis were analyzed for correlations to other CD markers and insulitis frequency (%), diabetes duration, and age of onset. Spearman r values are shown with grey shading indicating no significant correlations. Numbers of CD3+, CD20+, CD8+ and CD4+ cells/islet showed significant correlations to each other, total leukocytes (CD45), and insulitis frequency (%). Mean numbers of CD11c+ or CD68 cells/islet were significantly correlated with each other and numbers of CD3+ cells/islet but not to other leukocytes or other donor variables. * P<0.05 ** P<0.01 *** P <0.001.

Insulitis Diabetes Age CD45 CD3 CD20 CD8 CD4 CD68 CD11c Frequency Duration onset

(%) (Years) (Years) CD45 1.00 0.95*** 0.96*** 0.90*** 0.85*** 0.58 0.51 0.67* 0.42 0.26 CD3 1.00 0.89*** 0.87*** 0.90*** 0.63* 0.68* 0.78*** 0.24 0.38

CD20 1.00 0.78*** 0.78*** 0.33 0.49 0.56* 0.38 0.07

CD8 1.00 0.85 0.22 0.54 0.62* 0.55 0.08

CD4 1.00 0.33 0.55 0.62* 0.42 0.15

CD68 1.00 0.75 0.33 0.32 0.47

CD11c 1.00 0.53 0.55 0.17

Insulitis Frequency (%) 1.00 0.51 0.05

Diabetes Page 58 of 58

Supplementary Table 5. 43 nPOD cases reported herein were previously examined by Coppieters et al. (N = 14 donors without diabetes, N = 4 AAb+ donors without diabetes, and N = 25 donors with T1D). nPOD ID Diabetes Status 6009 No diabetes 6012 No diabetes 6013 No diabetes 6017 No diabetes 6019 No diabetes 6020 No diabetes 6021 No diabetes 6024 No diabetes 6030 No diabetes 6034 No diabetes 6047 No diabetes 6075 No diabetes 6098 No diabetes 6099 No diabetes 6027 AAb+ 6044 AAb+ 6080 AAb+ 6101 AAb+ 6025 T1D 6026 T1D 6031 T1D 6032 T1D 6035 T1D 6039 T1D 6041 T1D 6045 T1D 6049 T1D 6051 T1D 6054 T1D 6061 T1D 6062 T1D 6063 T1D 6064 T1D 6067 T1D 6076 T1D 6077 T1D 6079 T1D 6083 T1D 6087 T1D 6088 T1D 6089 T1D 6113 T1D 6180 T1D