NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets...

64

Transcript of NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets...

Page 1: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of
Page 2: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of
Page 3: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

N

B: P

leas

e do

not

read

the

lyric

s whi

lst l

iste

ning

to th

e re

cord

ings

.

Page 4: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

Front cover photo: Isolated human islets as viewed in a light microscope. © Monika Hodik, Uppsala, 2012. Back cover photo by Mikael Carlsson, Bankeryd, © 2011.

Page 5: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

List of Papers

This thesis is based on the following papers, which are referred to in the text by their Roman numerals.

I Moëll, A.*, Skog, O.*, Åhlin, E., Korsgren, O., Frisk, G.

(2009) Antiviral effect of nicotinamide on enterovirus-infected human islets in vitro: effect on virus replication and chemokine secretion. J. Med. Virol. 81(6):1082–1087. *Contributed equally

II Skog, O., Korsgren, O., Frisk, G. (2011) Modulation of innate immunity in human pancreatic islets infected with enterovirus in vitro. J. Med. Virol. 83(4):658-664,

III Skog, O.*, Hodik, M.*, Lukinius, A., Korsgren, O., Frisk, G. Ongoing pathogenic processes in the pancreas at onset of type 1 diabetes in humans. Manuscript. *Contributed equally

IV Skog, O., Hodik, M., Korsgren, O., Frisk, G. Enterovirus-induced disintegration of human pancreatic islets, but not viral replication per se, reduces the beta cell function selectively. Manuscript.

Reprints of paper I and II were made with permission from the publisher (Wiley-Liss, Inc.).

Page 6: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of
Page 7: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

Contents

Introduction ................................................................................................... 11 Autoimmune Disease ................................................................................ 11 

Autoimmune Disease and Virus Infection ........................................... 11 The Pancreas and the Islets of Langerhans ............................................... 12 Normal Regulation of Glucose Homeostasis ............................................ 12 Type 1 Diabetes ........................................................................................ 13 

Beta Cell Loss in Type 1 Diabetes ....................................................... 13 Autoimmunity in Type 1 Diabetes ....................................................... 14 Enterovirus in Type 1 Diabetes ............................................................ 16 

Innate Immunity ....................................................................................... 18 Innate Immunity to Virus ..................................................................... 18 

Enterovirus ................................................................................................ 19 Taxonomy, Structure, and Life Cycle .................................................. 19 Effects on Hosts and Host Cells ........................................................... 20 Interfering with Antiviral Innate Immunity ......................................... 21 Enterovirus Persistence ........................................................................ 21 Enterovirus in Myocarditis – Similarities with Type 1 Diabetes ......... 22 

Possible Mechanisms of Enterovirus-Induced Beta Cell Destruction ...... 23 Aims .............................................................................................................. 25 Considerations on Research Design and Methods ........................................ 27 

Virus Strains (Paper I-II and IV) .............................................................. 27 Use of Primary Human Islets (Paper I-IV) ............................................... 28 Pancreata from Recent Onset T1D Organ Donors (Paper III) .................. 28 Detection of Enterovirus in Pancreatic Tissue (Paper III) ........................ 29 

Results and Discussion .................................................................................. 30 Paper I - Antiviral Effect of Nicotinamide on Enterovirus-Infected Human Islets in vitro: Effect on Virus Replication and Chemokine Secretion ................................................................................................... 30 Paper II – Modulation of Innate Immunity in Human Pancreatic Islets Infected with Enterovirus in vitro ............................................................. 31 Paper III – Ongoing Pathogenic Processes in the Pancreas at Onset of Type 1 Diabetes in Humans ...................................................................... 32 Paper IV – Enterovirus-Induced Disintegration of Human Pancreatic Islets, but not Viral Replication per se, Reduces the Beta Cell Function Selectively ................................................................................................ 34 

Page 8: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

Conclusions ................................................................................................... 37 Paper I ....................................................................................................... 37 Paper II ..................................................................................................... 37 Paper III .................................................................................................... 37 Paper IV .................................................................................................... 38 

General Discussion ........................................................................................ 39 

Future Perspectives ....................................................................................... 41 What Causes Type 1 Diabetes? ................................................................ 41 Can it be Prevented? ................................................................................. 42 

Populärvetenskaplig sammanfattning på svenska ......................................... 44 

Acknowledgements ....................................................................................... 46 

References ..................................................................................................... 50 

Page 9: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

Abbreviations

ADP Adenosine diphosphate ATP Adenosine triphosphate CAR Coxsackievirus and Adenovirus receptor CD Cluster of differentiation CNS Central nervous system COPII Coat protein complex II CPE Cytopathic effect CTL Cytotoxic T lymphocyte CVB Coxsackievirus B DAF Decay accelerating factor DNA Deoxyribonucleic acid dsRNA Double-stranded RNA eIF Eukaryotic initiation factor ELISA Enzyme-linked immunosorbent assay EM Electron microscopy ER Endoplasmatic reticulum EV Enterovirus GAD65 Glutamic acid decarboxylase 65 kDa GADA GAD65 antibody GLUT Glucose transporter GSIS Glucose-stimulated insulin secretion HLA Human leukocyte antigen HSP Heat-shock protein IA2 Tyrosine phosphatase-like insulinoma antigen 2 IA2A IA2 antibody ICAM-1 Intercellular adhesion molecule 1 IFN Interferon Ig Immunoglobulin IHC Immunohistochemistry IL Interleukin IP-10 IFN-inducible protein 10 IPS-1 IFN promoter stimulator 1 IRES Internal ribosome entry site IRF3 Interferon regulatory factor 3 ISH In-situ hybridization JDRF Juvenile Diabetes Research Foundation

Page 10: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

MafA Mammalian homologue of Avian MCP-1 Monocyte chemotactic protein-1 MDA5 Melanoma differentiation-associated protein 5 MHC Major histocompatibility complex mRNA Messenger RNA NFkB Nuclear factor kappa B NK Natural killer NOD Non-obese diabetic nPOD Network for Pancreatic Organ donors with Diabetes ORF Open reading frame PAMP Pathogen-associated molecular pattern PARP-1 ADP-ribose polymerase 1 PCR Polymerase chain reaction PDX1 Pancreatic and duodenal homeobox 1 Poly(I:C) Polyinosinic:polycytidylic acid PP Pancreatic polypeptide PRR Pattern recognition receptor PVR Poliovirus receptor RANTES Regulated upon activation, normal T cell expressed, and

secreted RIG-I Retinoic acid gene I RLH RIG-I-like helicase RNA Ribonucleic acid RT-PCR Reverse transcription PCR ssRNA Single-stranded RNA T1D Type 1 diabetes TLR Toll-like receptor UK United Kingdom UTR Un-translated region VI Virus isolation VP Viral protein VPg Viral protein genome-linked ZnT8 Zinc transporter 8

Page 11: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

11

Introduction

Autoimmune Disease Oxford English Dictionary defines the term autoimmunity as “the occurrence or development of an immune response against a normal constituent of the body; the condition resulting from this” [1]. Considering that autoreactive antibodies and T cells circulate naturally, constituting a significant propor-tion of all antibodies, autoimmunity is not limited to autoimmune disease [2]. Autoantibodies and autoreactive T cells can be triggered by traumatic injury or virus infection [3, 4], and even if a direct pathogenic effect has been demonstrated in some diseases [5], they may in other cases be an epi-phenomenon to the underlying pathogenic event. For an autoimmune disease to appear, the regulatory mechanisms that control reactions against self-antigens must be dysfunctional.

Despite the fact that autoimmune diseases affect ~5% of the western world population very little is known about the genetic and environmental factors triggering these events [5]. The criteria for autoimmune disease were defined in 1957 by Witebsky’s postulate [6, 7]:

1. A defined circulating antibody- or cell-mediated immunity to au-toantigen(s).

2. Identification of the specific autoantigen(s). 3. The ability to produce the disease in an experimental animal by

passive transfer of the antibody or self-reactive cells. 4. The ability to induce an analogous autoimmune response in an

experimental animal by immunization with the self-antigen. 5. The ability to generate a similar disease in experimental animals

following immunization with the self-antigen.

Autoimmune Disease and Virus Infection There is a strong association between viruses and autoimmune disease. Most diseases with a proposed autoimmune etiology have been associated to one or more viruses in at least one study (reviewed in [8, 9]). Onset of most auto-immune diseases is accompanied by virus-like ‘mild febrile illness’ and sev-eral different viruses have been detected in patients at onset. Three main mechanisms are proposed in which a virus could induce or trigger autoim-mune tissue destruction; molecular mimicry, bystander activation, and direct

Page 12: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

12

infection [9]. The concept of molecular mimicry is based on sequence ho-mologies between viral and cellular epitopes, which could lead to cross-reaction between the immune response evoked by the viral infection and endogenous cellular proteins. In bystander activation a virus infection of cells in the target organ induces an inflammatory milieu that in turn may be toxic to uninfected neighboring cells. The third possibility is that a direct infection of the target cells attracts an immune response. These three differ-ent mechanisms will be discussed in more detail regarding their possible contribution to the etiology of type 1 diabetes in a later section.

The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of many different cell types. The dominating part (~80%) of the gland is exocrine, with acinar cells producing digestive enzymes that are secreted to the duode-num via the pancreatic duct where they digest lipids and proteins. Inter-spersed in the exocrine gland is an endocrine gland made up of around 1 000 000 islets of Langerhans representing only 1-2% of the total pancreas mass [10]. These islets were first described in 1869 by Paul Langerhans as islands of clear cells that stained differently from the rest of the pancreatic tissue [11]. It is now known that the islets of Langerhans contain several thousand endocrine cells of five different types that produce hormones involved in metabolism regulation. Most abundant are the insulin-producing beta cells and the glucagon-producing alpha cells, making up around 60% and 30% of the islet mass respectively. Present, but less abundant are the somatostatin-producing delta cells, pancreatic polypeptide (PP)-producing cells, and epsi-lon cells producing ghrelin [12]. Also, the islets are highly vascularized and endothelial cells make up a significant part of the islet volume [13]. In hu-mans, the relative proportion of the different endocrine cell types varies be-tween islets in different anatomical parts of the pancreas. For instance, in the uncinate process in the posterior head of the pancreas PP-producing cells are the dominating cell type [14].

Normal Regulation of Glucose Homeostasis Twenty-five years after the discovery of the islets of Langerhans, Sir Edward Albert Sharpey-Schäfer suggested them to play a role in controlling blood glucose by secreting a substance that he later termed insuline [15]. Insulin is a hormone produced by the beta cells in the islets of Langerhans and essen-tial for intracellular uptake of glucose in most tissues. Insulin is encoded by the pre-proinsulin gene on the short arm of chromosome 11 [16], by activa-tion of several transcription factors, including PDX1 and MAFA [17]. Pre-

Page 13: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

13

proinsulin is processed in the endoplasmatic reticulum (ER) into proinsulin, consisting of the A and B chains of insulin joined by the C peptide. The C peptide is subsequently cleaved off from the proinsulin and packed together with insulin in secretory granules eventually ready to be exocytosed [12]. The insulin secretory granules also contain high concentration of zinc, which crystallizes the insulin within the granule [18].

In healthy individuals insulin is exocytosed from the beta cells in re-sponse to glucose. The process leading to insulin release is complex, starting with intracellular uptake of glucose through glucose transporters (GLUTs) 1 and 3 on the beta cell surface. Glucose is metabolized in the beta cell, lead-ing to ATP production and closure of ATP-dependent K+ channels, altered electrical activity, influx of Ca2+ through voltage-gated calcium channels, and exocytosis of insulin granules. The beta cell response to a sudden in-crease in extracellular glucose is biphasic, consisting of an initial rapid phase where insulin granules that are already docked to the beta cell membrane are released, followed by a sustained second phase where granules migrate from the cytosol to the plasma membrane and insulin is released [12]. In mouse and rat islets, beta cell sensing of high glucose also leads to up-regulated transcription of the pre-proinsulin gene and production of new insulin gran-ules, but this was recently shown not to be the case in human islets [17], unless the exposure is prolonged [19].

Type 1 Diabetes Type 1 diabetes (T1D) is characterized by lost insulin production and secre-tion due to destruction of the beta cells in the pancreas. The incidence ranges from <1/100000 per year in Venezuela and China [20] to 64/100000 per year in Finland [21]. The increase in incidence during the past decades has been dramatic, especially in young children [22-25]. Despite the high numbers of affected patients, and that the cause of the disease (loss of insulin) has been known for more than a century, there is no preventive or curative therapy, but patients with type 1 diabetes will require lifelong injections of recombi-nant insulin.

Beta Cell Loss in Type 1 Diabetes The availability of human pancreatic samples from prediabetic subjects is scarce and non-invasive imaging methods for monitoring beta cell mass are not yet available. Thus, the ideas of pancreatic events leading to develop-ment of type 1 diabetes are mainly derived from animal models and specula-tions based on immunological markers in the circulation. The perhaps most widespread view is that the total beta cell mass decreases gradually in a line-ar manner over several years and that symptoms (hyperglycemia) arise when

Page 14: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

14

only around 10% of the beta cells remain [26] (Fig. 1A). A refined model (Fig. 1B) was recently proposed by von Herrath et al, suggesting that T1D is a relapsing-remitting disease [27, 28], similar to what is seen in multiple sclerosis, with repeated autoimmune events to an increased number of anti-gens, followed by a remitting phase with regulatory T cells and beta cell regeneration.

The idea that the beta cell mass is decreasing for several years before on-set of T1D is mainly based on the appearance of autoantibodies against islet antigens several years before the onset of T1D in many subjects, and a few reports of decreased glucose clearance in pre-diabetic subjects [29, 30]. However, the correlation of such metabolic tests with the actual beta cell mass is only limited [31]. In fact, an immunohistochemical study of pancreas from 62 organ donors with autoantibodies against islet antigens showed no evidence of decreased beta cell mass, even in cases with at least three differ-ent autoantibodies [32]. Also, a meta-analysis of histopathological studies has suggested that the reduction in beta cell mass required to initiate clinical symptoms of T1D is highly variable and decreases with age; at 20 years of age a 40% reduction in beta cell mass was sufficient to induce hyperglyce-mia and T1D [33, 34]. An alternative view would be that T1D is an acute and rapid disease. The autoantibodies, often appearing years before T1D onset, do not necessarily reflect what is going on in the pancreas and could possibly reflect limited beta cell destruction in a small part of the pancreas (Fig. 1C, arrows). The strong correlation of the appearance of autoantibodies with a later development of T1D may be explained by an increased suscepti-bility to the agent(s) inducing this beta cell damage.

Autoimmunity in Type 1 Diabetes Inflammatory infiltrations of islet tissue in patients that died soon after the onset of T1D were observed already in the early 20th century [35, 36], and autoimmunity was put forward as an important contributor to beta cell de-struction in the landmark paper by Gepts in 1965 [37]. The concept of auto-immunity in T1D development gained additional support in the 1970s by the finding of circulating autoantibodies against islet antigens in patients with T1D [38, 39] and by the discovery of a genetic association between T1D and certain HLA genotypes [40-42]. Autoantibodies against several islet antigens have been discovered (Table 1.) but also against antigens present only in the exocrine pancreas [43-45]. It is not known whether autoantibodies contribute to the pathogenesis of beta cell destruction or only is a consequence of it. Nonetheless, circulating autoantibodies have a strong predictive value for T1D development, especially when several autoantibodies are present [46, 47]. However, a case of development of T1D in a patient with severe inherit-ed B cell (antibody) deficiency has been reported [48], indicating that auto-antibodies are not a prerequisite for the development of the disease. In addi-

Page 15: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

15

tion to autoantibodies, autoreactive T cells directed against islet antigens have been found in the circulation of diabetic and prediabetic subjects [49-51].

Most knowledge of the autoimmune process in T1D is derived from find-ings in peripheral blood and murine models. Thus, the knowledge of what is actually going on in the pancreas is weak. In the most widely studied mouse model of autoimmune diabetes, the non-obese diabetic (NOD) mouse, the insulitic lesion is dense with extreme numbers of T cells and B cells engulf-ing the islets [52, 53]. The presence of immune cells in human pancreata from patients that died at onset of T1D has most extensively been studied in a collection of samples obtained from hospitals in the UK between 1960 and 1985 by Foulis et al. [54-57], and in a few isolated case reports [58-65]. In contrast to studies in the NOD mouse, in human T1D the insulitis is discrete and less frequent. The presence of insulitis in humans is more common in young children and decreases with time after onset [61]. Most studies de-scribe an infiltrate dominated by cytotoxic CD8+ T cells and macrophages [56, 62, 65], but in some cases significant numbers of NK cells [63], CD4+ T cells, and B cells have also been reported [56, 65]. The distribution of immune cells throughout the pancreas is heterogeneous and three different categories of islets have been described; islets with insulin without insulitis, islets with insulin and with insulitis, and islets without insulin and without insulitis [54]. A consensus for the definition of insulitis has yet to be defined. Based on the lowest number of immune cells that was rarely exceeded in control islets [56], Richardson et al. [66] suggested a threshold of five mon-onuclear cells per islet section for insulitis, but whether this is enough to play a pathogenic role is not known. In pancreata collected in the recently established Network for Pancreatic Organ donors with Diabetes (nPOD), Coppieters et al. [67] have demonstrated the presence of T cells directed against islet antigens in the islets of recently diagnosed T1D donors, but also in this study the presence of immune cells was scarce. Table 1. Autoantibodies against islet antigens

Antigen Prevalence at T1D onset Discovery year References GAD65 70-80% 1982 [68-70] IA2 35-75%* 1990 [71-73] Insulin 20-90%* 1963 [74-76] ZnT8 60-80% 2007 [46, 77]

*Higher in young children; GAD65: glutamic acid decarboxylase 65; IA2: tyrosine phosphatase-like insulinoma antigen 2; ZnT8: Zinc transporter 8

Page 16: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

16

Figure 1. Different theories on the time course of beta cell loss in T1D. In the classical view (A) the beta cells are lost in a linear fashion after the appearance of auto-antibodies. A relapsing-remitting autoimmune beta cell destruction (B) was proposed by von Herrath et al. [27]. It is also possible that the majority of beta cells are destroyed rapidly in the acute phase of the disease (C).

Enterovirus in Type 1 Diabetes Although several genetic T1D susceptibility loci have been identified, there is only a less than 40% concordance in monozygotic twins [78, 79], which suggests environmental factors to be important contributors to the disease. The first study to suggest a linkage between enterovirus (EV) infections and T1D was published more than four decades ago [80]. Since then, numerous epidemiological studies and case reports have been presented that support this linkage and a number of recent publications have provided substantial evidence for EV being a direct causative agent of T1D [63, 81-85]. There is

Page 17: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

17

clear evidence that EVs can infect human pancreatic islets [86-88]. EV has been detected in human islets in vivo by virus isolation [89], in situ hybridi-zation [90] and immunostaining [63, 91, 92]. Richardson et al recently demonstrated immunopositivity for EV capsid protein 1 (EV-P1) in the islets in 44 out of 72 recent-onset type 1 diabetic patients, and very rarely in islets from control donors [91]. Also, inoculation of mice with EV isolated from human pancreas induced beta cell destruction and hyperglycemia [89]. The exact mechanisms of beta cell destruction in EV-induced T1D are however not known. A most interesting recent study by Duenas-Cubas et al. showed that that a significant fraction of first-degree relatives with islet autoantibod-ies had EV genome in their circulation [93]. Also, immunopositivity to EV capsid protein has been demonstrated in the islets from an autoantibody posi-tive organ donor [94]. These two studies together may suggest that EV infec-tion of the islets is one of the events that can trigger the induction of these autoantibodies.

The ability of EV to replicate in cultured pancreatic islets and also to cause beta cell death has been shown to vary between different strains of a serotype [86]. Most EV strains cause direct beta cell damage when human islets are infected in culture [86], but productive non-lytic EV infection of cultured islets has also been described [95]. Lytic as well as non-lytic EV infection of pancreatic islets in vitro results in the induction of a number of pro-inflammatory cytokines and chemokines, including IL-1β, IL-6, IL-8, MCP-1, and IP-10 [96, 97]. These could either cause beta cell destruction directly or by promoting insulitis. Interferon inducible peptide-10 (IP-10) and monocyte chemotactic protein-1 (MCP-1) have chemoattractant effects on specific immune cells and have been implicated in T1D development [98]. It is also possible that the initial beta cell destruction caused by a direct EV infection in vivo leads to the release of intracellular proteins that were previously invisible to the humoral immune system. These proteins would be recognized as foreign and could explain the appearance of auto-antibodies associated with autoimmune disease (reviewed in [99]).

Recognition and induction of pro-inflammatory substances by members of the picornavirus family have been shown to be mediated by the intracellu-lar helicase MDA5 [100]. Most interestingly, recent genetic studies have found four rare variants of MDA5 that confer protection against T1D [101, 102], suggesting a role for MDA5 in T1D development, and also providing a possible link between T1D and EV infection. It is tempting to speculate that certain variants of MDA5 either make the cells more susceptible to EV in-fection or alter the cytokine pattern induced by EV leading to a more dia-betogenic immune reaction.

Page 18: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

18

Innate Immunity The first line of host defense against invading pathogens is the innate im-mune system. It is non-specific by nature in the sense that it recognizes and responds to pathogens in a generic fashion. The most basal function of innate immunity is to provide a “barrier” against infection. This barrier can be di-rectly mechanical, like the epithelia of the skin and other surface areas of the body prohibiting foreign objects to enter, or more sophisticated recognizing the existence of an invading foreign object, eliminate it or prohibit its spread. In this way, the innate immune system may function to limit the infection, but it also plays an important role in alerting and shaping the later, antigen-specific, adaptive immune response. In the perspective of auto-immune dis-ease, local induction of innate immunity has been proposed to induce a so-called “fertile field” for pre-existing and latent adaptive auto-immunity [103].

Innate Immunity to Virus Viral particles, infecting their target cell, or being phagocytosed by poly-morphonuclear leucocytes, are recognized by several receptors of the innate immune system. These receptors are so-called pattern recognition receptors (PRRs) that are activated through the recognition of pathogen-associated molecular patterns (PAMPs) of, or produced by, the invading virus [104]. Two classes of PRRs are important for production of type I interferon (IFN) and pro-inflammatory cytokines upon viral infection; Toll-like receptors (TLRs) and RIG-I like helicases (RLHs) (Fig. 2). TLRs, involved in virus detection, are transmembrane proteins present on the plasma membrane (TLR2 and TLR4) or on cytoplasmic vesicles (TLR3, TLR7, and TLR9). These are highly expressed in plasmacytoid dendritic cells (pDCs) special-ized to produce extremely high levels of type I IFN in response to virus in-fection. RLHs consist of two pro-inflammatory members designated retin-oic-acid gene I (RIG-I) [105] and melanoma differentiation-associated pro-tein 5 (MDA-5) [100]. These are intracellular helicases detecting viral nucle-ic acid in the cytoplasm of any infected cell. Viral PAMPs detected by RLHs may be dsRNA or ssRNA with 5’-triposphate ends. Long dsRNA is normal-ly not found in uninfected cells, and most cellular mRNA is capped and nev-er 5’-triphosphorylated. The relative importance of different components of the innate immune system for virus recognition is not fully clarified yet and varies among different viruses and cell types [100]. However, some studies have indicated that RIG-I is essential for detection of 5’-triphosphorylated ssRNA [106], whereas dsRNA is recognized mainly by MDA-5 [100].

Recognition of virus by TLRs or RLHs leads to transcription of type I in-terferon (IFN) and pro-inflammatory cytokines/chemokines (Fig. 2). Type I IFNs induce the transcription of more than 300 IFN-stimulated genes, induc-

Page 19: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

19

ing an antiviral state in surrounding cells, and limiting viral spread [107]. The induced pattern of cytokines and chemokines is important in attracting, activating, and shaping the later adaptive immune response [108].  

Figure 2. Two classes of pattern-recognition receptors (PRRs) are important for production of type I interferon and proinflammatory cytokines upon viral infection. The illustration is modified from Takeuchi and Akira, J Immunol. Rev., 2009 [104].

Enterovirus Taxonomy, Structure, and Life Cycle Enterovirus (EV) is a genus in the family Picornaviridae and grouped into 10 EV species; Human EVs A-D, Simian EV A, Bovine EV, Porcine EV B, and Human rhinoviruses A-C [109]. Each of these 10 species can be further subdivided into different serotypes, which by definition are viruses that are neutralized by treatment with a type-specific anti-serum. Also, the extremely high mutation rate of picornaviruses results in the existence of several viral strains, with different characteristics, within one single serotype, so-called quasispecies [110].

All members of the EV genus are small (~30 nm in diameter), icosahedral particles, containing a single-stranded RNA genome of positive sense. The

Page 20: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

20

genome of ~7500 nucleotides consists of one single open reading frame (ORF), flanked by 5’ and 3’ un-translated regions (UTRs). The 3’UTR con-tains a poly(A)tail, but unlike most mammalian mRNA transcripts, the 5’UTR of EV is not capped. Instead, translation of the EV RNA is initiated by ribosomal recognition of the internal ribosome entry site (IRES) located in the 5’UTR, and primed by VPg, allowing cap-independent translation. Upon entry to the host cell through the specific binding of EV structural proteins to cellular surface receptors (DAF, CAR, PVR, and ICAM-1 being some of the most well-studied), the EV genome is uncoated and can be translated directly in the ribosomes. The EV ORF encodes one single poly-protein that is subsequently cleaved by virus-encoded protease in a series of events, into four structural (VP1-VP4) and 7 nonstructural (2A-2C and 3A-3D) proteins. Replication of the EV genome takes place at membranes of virus-induced vesicles formed from the endoplasmatic reticulum and Golgi, and is carried out by the viral RNA-dependent RNA polymerase through a minus stranded RNA intermediate (the EV life cycle is reviewed in [111]).

Effects on Hosts and Host Cells Enteroviruses, originally named because of their ability so survive the gastric low pH and establish infection in the intestinal mucosa, are very common and mostly cause mild or subclinical infection. However, EVs can some-times spread from their primary site of infection in the gastrointestinal mu-cosa or upper respiratory tract, to secondary replication sites and cause more severe disease; i.e. meningitis or encephalitis in the brain, paralysis in the CNS, and myocarditis/pericarditis in some cases leading to chronic myocar-ditis or dilated cardiomyopathy of the heart [112]. EV has also been detected in other organs, such as the pancreatic islets of Langerhans [63, 90-92, 94], in which a clear causal relationship to disease remains to be proven.

EV has developed efficient ways to promote viral replication. Since its translation is cap-independent, EV proteases cause shut-off of cellular trans-lation by cleavage of components of the eIF4F complex, involved in cap-dependent translation [113]. It also reduces protein transport in the host cell through the inhibition of ER-to-Golgi traffic by the viral non-structural pro-teins 2B and 3A [114, 115]. Numerous other virus-cell interactions that facil-itate increased viral replication and reduce the host defense have been de-scribed, and are reviewed in [111]. Ultrastructurally, the most prominent EV-induced change is the accumulation of membranous vesicles in the cyto-plasm [116]. This is achieved by use of cellular COPII [117] and integration of viral proteins 2B and 2BC into the membrane of the host Golgi and ER complex, producing virus-induced vesicles [118]. In the light-microscope the EV-induced cytopathic effect (CPE) is characterized by rounding of the cells, making the infected cells refractory to light.

Page 21: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

21

Interfering with Antiviral Innate Immunity The importance of innate immunity in controlling virus infection is empha-sized by the fact that viruses have evolved mechanisms to interfere with almost all aspects of activation of the innate immune system. During co-evolution of viruses and their host cells, there has been a constant battle be-tween immune mechanisms and viral countermeasures, and it is not always obvious if a particular mechanism is in the interest of the host trying to pro-tect itself or the virus trying to evade host protection in order to replicate and spread. Numerous mechanisms for EV interference with host cell innate immune responses have been described. Given the large number of different EV serotypes, and almost unlimited number of strains and quasi-species, it is for obvious reasons not possible to characterize what exact mechanisms each specific virus utilizes. Also, it should be noted that the same virus might have different effects in different cell types. However, many mechanisms have been observed in several systems and can be expected to be more or less consistent.

Sensing of EV in the cytoplasm of the host cell is interfered by cleavage of the intracellular helicases RIG-I and MDA5 during EV replication, the former directly by the viral protease 3C [119] and the latter by cellular caspases activated during EV infection [120]. Downstream of these RLHs, transcription of type 1 IFN is attenuated by viral proteases 2A and 3C through degradation of IPS-1, which abolishes activation of IRF3, a tran-scription factor for type 1 IFN [121, 122]. Also, the inhibition of protein secretion by EV 2B and 3A [114, 115], and shut-off of cap-dependent trans-lation [113], discussed in a previous section, reduces the secretion of IFN and pro-inflammatory cytokines and chemokines.

These mechanisms all reduce the ability of the infected cell to alert sur-rounding cells and induce an antiviral state preventing viral spread. In addi-tion to this, EVs have evolved mechanisms to antagonize the effect of IFN-stimulated genes, and some EVs can infect and replicate in cells pretreated with type I IFN [123]. In conclusion, one can say that the result of this inter-action between the host’s innate immune system and the virus’ ability to avoid clearance may determine the outcome of infection by a particular EV strain in a particular host/individual. For instance, the tropism and patho-genicity of poliovirus and CVB3 have been shown to be controlled partly by the type I IFN response [124, 125].

Enterovirus Persistence Most EVs rapidly lyse infected cells in vitro and in vivo, but EV persistence has been shown or suggested to play a role in a number of different diseases [126-128], including T1D [66, 129, 130]. Persistence of EV in the host has been poorly understood, but shown to occur in both naturally infected hu-

Page 22: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

22

mans and in experimental systems by the evolution of attenuated phenotypes that evade immune surveillance [131-134].

Some strains of EV have been reported to replicate without cell lysis in certain cell types but not in others [81, 95, 134], indicating that the course of EV infection is dependent on both viral and host cell properties. The high mutation rate during EV replication leads to a constant generation of multi-ple quasi-species of which different variants may be favored in different cell types. Replication of the EV genome is facilitated partly by host cell proteins that are not available in the cytoplasm during cell cycle arrest in a non-replicating state (G0) [135, 136]. EV infection of primary, rarely replicating, terminally differentiated cells has been shown to generate EV genomes with deletions of up to 49 nucleotides in the 5’ terminus. These terminally deleted viruses replicated slowly but did not induce any visible cytopathic effect [131, 132]. Persistence of such terminally deleted EV genomes was detected for up to five months in mice cardiomyocytes [131], and in the heart of a human with myocarditis [133]. Also, EVs with terminal deletions have been shown to encapsidate the negative strand [131] and EV persistence has been suggested to occur in the form of stable enteroviral dsRNA [137].

In the case of a possible link between T1D and EV, viral persistence is of great interest since a persistent infection with a virus in vivo may lead to inflammation, which can be difficult to distinguish from an autoimmune attack. An obvious case of this scenario is chronic hepatitis, in which persis-tent Hepatitis C infection in the liver causes an immune-mediated destruc-tion of the liver cells, also involving an autoimmune response against endog-enous liver antigens [138]. Also, the presence of a very low amount of EV RNA, due to the slow replication of EV with terminal deletions, may explain difficulties in detection of virus in pancreatic samples.

Enterovirus in Myocarditis – Similarities with Type 1 Diabetes Other EV-associated diseases that involve autoimmunity are chronic myo-carditis and dilated cardiomyopathy. EV infection of the heart is present in a significant proportion of cases of these diseases [139]. In animal models of EV-induced myocarditis the virus has been shown to persist in the myocar-dium, causing impaired function of the infected cardiomyocytes [126, 131, 140, 141]. Chronic myocarditis is in many ways similar to T1D. In addition to its association with EV, it is also characterized by the presence of inflam-matory infiltrates (predominated by CD8+ T cells) in the heart tissue and circulating autoantibodies to endogenous antigens [142, 143]. Flu-like symp-toms with fever and gastrointestinal or respiratory symptoms are commonly reported in close association to disease onset [142]. As in T1D, a slight male predominance has been reported in the incidence of myocarditis and dilated cardiomyopathy [144]. In contrast to T1D, the involvement of EV in the pathogenesis of myocarditis is proven and generally accepted. However, as

Page 23: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

23

with T1D, most EV infections do not cause heart disease and a better under-standing of the host and viral genetic determinants that lead to severe disease is needed [145]. In heart tissue from patients with myocarditis or dilated myocardiopathy, EV can frequently be detected both by IHC for EV P1 and by PCR for EV genome. However, the detection of EV protein is more fre-quent than EV genome, suggesting that in tissue samples this method is more sensitive [146]. This is in line with T1D, where immunopositivity to EV P1 is frequently found in the islets, but detection of EV genome has been re-ported less frequently.

Figure 3. Possible mechanisms of EV-induced beta cell destruction. Figure from [147].

Possible Mechanisms of Enterovirus-Induced Beta Cell Destruction Three main pathways by which EV possibly could induce beta cell destruc-tion have been proposed (Fig. 3); direct cytolysis of infected beta cells, local virus-induced inflammation, and molecular mimicry.

A direct cytolytic effect is supported by the findings that EV can infect human beta cells in vitro [86-88] and has been discovered in the islets at onset of T1D [64, 90]. In addition to causing T1D by direct virus-mediated beta cell destruction, this has been suggested to lead to release of intracellu-

Page 24: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

24

lar beta cell antigens that were previously hidden from the immune system and induce autoimmunity [148]. Indeed, EV has been detected in the blood of first-degree relatives with autoantibodies [93] and in the islets of an auto-antibody positive organ donor [94].

Infection of beta cells, or other cells in close association to the islets, in-duces an inflammatory milieu [97, 149] that can be directly toxic to the islets [150-152] or attract immune cells to the site of infection [92, 153]. It has also been suggested that activation of innate immunity in the islets by an EV infection could provide a so-called fertile field for self-directed T cells al-ready present in the circulation [103].

Molecular mimicry is a hypothetical mechanism in which the immune re-sponse evoked by a virus cross-reacts with endogenous cells due to epitope similarities. The infection can thus be in any cell type in the body and indi-rectly cause autoimmune beta cell destruction. This theory is mainly based on a six amino acids long sequence homology between the EV protein 2C and the islet autoantigen GAD65 [154].

Page 25: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

25

Aims

The studies included in this thesis aimed at elucidating mechanisms of EV-induced innate immune responses and islet destruction, both in vitro and in vivo. We also had the opportunity to study ongoing pathogenic mechanisms at onset of T1D and in pancreata from donors with autoantibodies against islets antigens. The rationale behind studies of in vitro infected human islets is based on the idea that a direct EV infection of islets in vivo initiates or triggers T1D [66, 155]. However, since most knowledge of viral effects on their host cells is based on studies in animals or in immortalized cell lines and since innate immune signaling varies greatly between different primary cell types [156], these studies are important also from a basic research per-spective. Specific aims of the four papers were:

• To investigate the anti-inflammatory and islet-protective effects of nico-tinamide (Paper I) and the female sex hormone 17β-estradiol (Paper II) in EV-infected human islets.

• To study if nicotinamide has an antiviral effect on EV replication Paper I).

• To study induction of innate immunity by EV and synthetic dsRNA in human islets (Paper II).

• To study the ability of pre-induction of innate immunity by dsRNA to induce an antiviral state in human islets, reducing EV replication (Paper II).

• To characterize the morphology, mode of cell death, and induction of innate immunity, at onset of T1D in the pancreata of two organ donors (Paper III).

• To study pancreatic immune infiltration and islet insulin content in organ donors at onset of T1D, several years post diagnosis, and in islet autoan-tibody-positive subjects (Paper III).

Page 26: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

26

• To search for the presence of viral proteins and/or viral nucleic acid in

the pancreas at onset of T1D (Paper III).

• To test if, and at what stage of virus-induced islet destruction, EV infec-tion affects the glucose-stimulated insulin secretion in human islets (Pa-per IV).

• To test if dsRNA alone, without the presence of infectious virus, affects beta cell function (Paper IV).

• To investigate if EV infection of islets results in altered expression of beta cell specific genes (Paper IV).

Page 27: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

27

Considerations on Research Design and Methods

Here some considerations on a few important methods used in this thesis are brought forward. For more detailed information regarding all the methods used, please read the material and methods section in the respective papers (I-IV).

Virus Strains (Paper I-II and IV) Since there are more than 100 different serotypes of human EV [109, 110], and numerous strains of each serotype, all with different properties, the studies in this thesis had to be limited to a few particulary interesting viruses from a T1D perspective. Even though it is not known whether some genotypes or strains are more ‘diabetogenic’ than others, the EVs most prominently linked to T1D are of the Coxsackievirus B type [28].

Four strains of CVB were used; VD2921 (Paper I and IV) and V89-4557 (Paper IV), isolated from patients suffering from aseptic meningitis [86, 95, 157, 158], and Adr (Paper I-II and IV) and E2-Yoon (Paper IV), isolated at onset of T1D. Adr was isolated from stool [87], whereas E2-Yoon in the 1970s was the first virus to be isolated from the pancreas at onset of T1D [89]. VD2921 and E2-Yoon have been genotyped as CVB4 whereas Adr and V89-2921 are of the CVB5 genotype.

Adr and E2-Yoon were chosen because of their temporal relationship with T1D onset in two clinical cases. V89-4557 is interesting since it, in contrast to most other CVB can infect cells lacking both CAR and DAF [157] and has been shown to replicate fast in islets causing pronounced islet disintegration [87]. VD2921 has been shown to establish persistent non-lytic infection in human islets in vitro [86, 95], and persistent infection in vivo has been put forward as one of the main suspects for causing/triggering T1D [9, 155].

Page 28: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

28

Use of Primary Human Islets (Paper I-IV) The pancreases were obtained from organ donors within the Nordic Network after informed consent from the donor or close relatives. Islets of Langer-hans were isolated in Uppsala, Sweden, using a protocol approved by the Local Ethics Committee, as described previously [159]. Islet preparations were available for research only because the total islet volume was too low for clinical transplantation or a suitable recipient was not available.

Despite the good quality of the islets used in these studies, one must take into account that the islets are affected by the isolation procedure and culture and may not function exactly as they do in vivo [160]. Another challenge when working with primary human cells is that the genetic background of each donor may generate divergent results that are difficult to interpret. This, however, is also a benefit of primary cells since each inbred animal model and each cell line is to look at as an individual donor and studies on these actually represents single case reports [161].

Pancreata from Recent Onset T1D Organ Donors (Paper III) In recent years, initiatives to collect and distribute tissue from human diabet-ic pancreata have gained increased interest. The Network for Pancreatic Or-gan donors with Diabetes (nPOD) funded by the JDRF (www.jdrfnpod.org) collects and procures pancreata from centers in the US and around the world. To a large extent inspired by this program, similar initiatives have been initi-ated in Europe. Tissue from pancreatic organ donors in the Nordic countries is collected through the Nordic Network for Islet Transplantation, in Uppsa-la, Sweden. Some of these donors have been diagnosed with diabetes and all are screened for autoantibodies against islet antigens.

In paper III we had the unique opportunity to study ongoing pathogenic processes in the pancreas at onset of T1D in humans. Two patients, with no previous history of T1D, presented with acute severe hyperglycemia and diabetic ketoacidosis. Despite treatment at intensive care units at hospitals in Finland and Sweden respectively, both developed cerebral edema and brain death. They were selected as multi-organ donors, and their pancreata were removed while heart beating and transported to Uppsala in proper preserva-tion solution minimizing post-mortal changes. Islets were isolated from the tail of both pancreases, and the remaining parts (body and head) were used for biopsies. Also, pancreata from organ donors with antibodies against GAD65 and/or IA2 were available, allowing the possibility to examine the pancreatic tissue before clinical presentation of T1D. Since the material was from diseased organ donors, one obvious limitation is that there is no possi-bility to follow up the patients for development of diabetes.

Page 29: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

29

Most previous case reports describing the pancreas at onset of T1D are from autopsy samples, suffering to a large extent from tissue degeneration and post mortem changes [54]. The pancreata used in our study are well preserved for clinical transplantation, minimizing artifacts caused post mor-tem. However, the time and treatment in intensive care units before organ procurement may influence, for instance, the presence of inflammatory markers.

Detection of Enterovirus in Pancreatic Tissue (Paper III) Detection of EV in pancreatic tissue has proven challenging. Five main de-tection methods have been applied; Immunohistochemistry (IHC), in-situ hybridization (ISH), reverse transcription polymerase chain reaction (RT-PCR), electron microscopy (EM), and virus isolation (VI). Each of these methods has its limitations, and there has been a striking problem with in-consistent results generated by the different methods.

The most widely used method for detection of EV in pancreatic tissue has been IHC with a monoclonal antibody against the EV-P1 (clone 5D8/1; Dako, Glostrup, Denmark). This method is also applied in this thesis (Paper III). This antibody seems to be the most reliable commercial antibody avail-able, detecting most, but not all, serotypes of EV [162]. It has been suggest-ed to cross-react with islet cell antigens (IA-2) [163] and HSP60 [164] under certain conditions, but by careful optimization it does not generate positive staining in pancreas tissue from the majority of non-diabetic organ donors [91]. However, cross-reactivity to the altered protein expression in diabetic islets has not been ruled out.

The limitations of IHC does definitely not rule out that the results ob-tained by this method indeed represent true virus positivity, but stresses the importance of confirming positive findings by several methods. In this the-sis, additional EV staining with a blend of antibodies against CVB1-6 was applied in order to confirm the staining with the 5D8/1 clone if the present virus was of the CVB type. Since the material in our study was fresh, and preserved for organ transplantation, we also had the opportunity to screen for EV with EM, VI, and RT-PCR, methods that are difficult or impossible to apply on formalin fixed tissues [130]. Even if these methods also have their limitations regarding sensitivity and specificity, confirmative results with multiple methods dramatically reduce the risk for false positives as well as negatives. In addition, detection of viral footprints, such as markers of innate immunity activation, may argue for the presence of virus even if the virus itself cannot be detected.

 

 

Page 30: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

30

Results and Discussion

Paper I - Antiviral Effect of Nicotinamide on Enterovirus-Infected Human Islets in vitro: Effect on Virus Replication and Chemokine Secretion EV infection of pancreatic islets might promote insulitis by inducing local expression of pro-inflammatory cytokines and chemokines, leading to leuko-cyte recruitment. In paper I, we demonstrate the induction of Interferon-inducible peptide 10/CXCL10 (IP-10) and monocyte chemotactic protein 1/CCL2 (MCP-1), by a non-lytic CVB4 strain (VD2921) as well as by a lytic CVB5 strain (Adr). These two chemokines are involved in the pathogenesis of various autoimmune diseases such as multiple sclerosis and rheumatoid arthritis [87]. In addition they have been shown to induce beta cell death [165], and to be elevated in serum [166, 167], and in pancreatic islets [92] from T1D patients at onset. We used ELISA to measure accumulation of IP-10 and MCP-1 in the culture media of islets infected with VD2921 or Adr, and compared it to media from mock-infected islets. IP-10 was secreted ex-clusively from EV-infected islets whereas mock-infected islets secreted ba-sal levels of MCP-1, which was significantly increased by EV infection. Expression of several pro-inflammatory genes, including mcp-1, has been demonstrated in isolated human islets [168, 169] and is probably a result of various stress factors, including the process of brain death and organ pro-curement.

Nicotinamide is used routinely during the isolation procedure of human islets [159] because of its demonstrated protective [170] and anti-inflammatory effects on islets in vitro [170, 171]. Based on these findings, we were interested in evaluating the effect of nicotinamide on EV-infected islets. Isolated human islets were cultured with or without addition of nico-tinamide to the culture medium for 24 h prior to infection with VD2921 or Adr. The secretion of IP-10 upon infection was blocked almost completely (Adr) or reduced dramatically (VD2921) when the islets had been pre-cultured with nicotinamide. The increased MCP-1 secretion induced by EV infection was also reduced by nicotinamide, to a level comparable to that in mock-infected islets. We also studied the effect of nicotinamide on virus replication and virus-induced islet destruction/CPE. Most interestingly, nico-tinamide reduced the replication of VD2921 as well as of Adr significantly.

Page 31: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

31

Also, the islet destruction induced by Adr infection was reduced by nicotin-amide. The timing of nicotinamide exposure was not important, since its effects on chemokine secretion, virus replication, and islet destruction, were similar when nicotinamide was added to the islets simultaneously, or 24 h post infection.

The mechanism of action of nicotinamide is not demonstrated in Paper I, but the substance is known to interact with poly ADP-ribose polymerase 1 (PARP-1) [172]. PARP-1 is involved in many cellular processes, including regulation of transcription of inflammatory mediators, and DNA repair. In-hibition of PARP-1 could lead to defect NFκB activation and reduced ex-pression of inflammatory mediators [173]. Although nicotinamide has been tested in a large scale human trial (ENDIT), in which it failed to prevent T1D in patients at risk of developing the disease [174], the findings de-scribed in Paper I could have implications for the treatment/prevention of virus- and/or immune-mediated disease. Also, this study highlights a possi-ble mechanism of virus-induced T1D through the induction of MCP-1 and IP-10 in pancreatic islets.

Paper II – Modulation of Innate Immunity in Human Pancreatic Islets Infected with Enterovirus in vitro There is a male predominance in the incidence of T1D after puberty [175], and female sex hormones have been suggested to have a protective role. In Paper II we demonstrate that the female sex hormone 17β-estradiol, like nicotinamide in Paper I, reduces EV-induced secretion of IP-10 by human islets. In contrast to nicotinamide, 17β-estradiol did not affect viral replica-tion. One of the most interesting findings in this paper is that although IP-10 was induced in islets by synthetic dsRNA (poly[I:C]) to the same extent as by EV infection, this secretion was not reduced by 17β-estradiol, indicating that in primary human islets, poly(I:C) and EV induce innate immunity via separate mechanisms. The paper further demonstrates for the first time that EV infection induces the expression of IFNβ, MDA5, and TLR3 mRNA in human islets, and that the virus continues to replicate even after the induc-tion of these antiviral genes. However, when the islets are pre-exposed to poly(I:C) for 24 h before EV-infection, the mean viral replication is reduced significantly (n=9), indicating that poly(I:C) induces an antiviral state in the islets that cannot be counteracted by the virus. The variations between indi-vidual islet donors were large, and poly(I:C) either blocked (5/9 donors), reduced (3/9 donors), or did not affect (1/9 donors) EV replication. Like EV infection, poly(I:C) exposure of islets led to induction of the innate immuni-ty genes IFNβ, MDA5, and TLR3. The induction levels varied greatly be-tween the nine tested donors, but did not correlate with the level of inhibition

Page 32: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

32

of viral replication. The data presented in this paper provide insight on the innate immune responses induced by EV in human islets, and show that this can be modulated by 17β-estradiol. It further demonstrates an important difference between virus- and poly(I:C)-induced signaling.

Paper III – Ongoing Pathogenic Processes in the Pancreas at Onset of Type 1 Diabetes in Humans In Paper III we had the rare opportunity to study pancreatic tissue and iso-lated islets from two organ donors that died at onset of T1D and 15 organ donors with circulating diabetes-associated autoantibodies (GADA and/or IA2A). Isolated islets were used for functional analyses, gene and protein expression studies, whereas pancreatic sections were used for IHC staining and ultrastructural analysis.

No abnormalities were found in pancreas from autoantibody-positive do-nors. The insulin-positive area was not significantly different from control pancreata and no proof of insulitis or EV infection was found. It should be noted that only two of the fifteen studied donors were positive for both IA2A and GADA (the rest were only positive for GADA), but even in these we found no signs of ongoing pathogenesis. However, only a limited part of the pancreases were available for IHC studies and it is fully possible that limited beta cell destruction was ongoing in a different anatomical area.

IHC staining of the pancreatic tissue from the donors with recent onset T1D revealed that insulin was reduced but still present in many or most is-lets. However, insulin secretion in response to a dynamic glucose challenge was extremely low or missing. Lost beta cell function despite the presence of a large number of intact insulin-containing cells at onset of T1D has been described previously [63], and could indicate that a functional beta cell block, in addition to cell destruction, is involved in T1D pathogenesis. A finding that was striking, considering what has been reported in animal mod-els and in a few cases of human T1D, was the lack of accumulation of in-flammatory cells in or around the islets. Staining for cytotoxic CD8+ T lym-phocytes (CTLs) and macrophages (CD68+) showed an increased presence of these cells in the pancreatic sections from the two T1D donors compared to controls, indicating an increased immunologic activity, but the pattern of distribution was more or less even throughout the pancreas. T helper lym-phocytes (CD4+) or B cells (CD20+) were not present. In one of the two T1D donors, clusters of CTLs that grouped together could be found, a few times in an islet, but more often localized to the exocrine pancreas. Most case re-ports describing islet infiltration do not report the frequency of immune cells not associated to islets, but only count the number of islet-infiltrating mono-nuclear cells. The definition of insulitis is also very vague, with some case

Page 33: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

33

reports defining islets as insulitis-positive when containing “two or more infiltrating mononuclear cells” [176]. Our results clearly show that to fully understand the relevance of such a finding, the number of islet-infiltrating immune cells must be compared to the number in the exocrine pancreas.

Most islets in one, and a few islets in the other, of the two T1D pancreas-es had a dramatic appearance with intra-islet bleedings (Fig 4A) and large swollen cells (Fig 4C). Swollen islet cells and bleedings in human diabetes were described already in the early 20th century by A. Weichselbaum (Fig 4B and 4D) [177, 178], and in the classic work from 1965 by W. Gepts [37]. The phenomenon has been termed hydropic degeneration and has also been described in islets of rabbits treated with cortisone [179], and in bank-voles infected with Ljungan virus [180].

Figure 4. The characteristic islet appearance in one of the two described cases of recent onset T1D (A and C) were described more than 100 years ago by A. Weich-selbaum (B and D) [177].

Consistent with a viral infection, staining of the pancreas from the two T1D donors revealed prevalence of cells intensely immunopositive for EV-P1 in multiple islets. No EV-P1 positive cells could be detected in any of the test-ed normal controls. In one of the two T1D donors, the suspected presence of

Page 34: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

34

EV could be strengthened by staining with a blend of antibodies against Coxsackievirus B. However, despite several attempts to isolate virus from pancreata from the T1D donors, and multiple PCRs against a wide variety of picornavirus and other viruses, we failed to confirm the positive results from the IHC analyses. Isolation of virus is not a sensitive method, and failure to isolate virus can easily be explained by low concentration of virus in the islets, or lack of permissiveness of the inoculated cells to the specific virus. Even though the used PCRs were very sensitive at detecting virus in culture, their sensitivity may decrease dramatically when trying to detect virus in tissue due to the presence of inhibitors. Another explanation of the failure to detect virus with PCR despite the positive EV stainings may be that these infections were caused by a new picornavirus that is not detected with the primers used here. Also, one obvious explanation could be that no virus is present and that the antibody against EV cross-reacts with an endogenous protein that is expressed in diabetic but not in healthy islets. However, since the EV-P1 staining in one of the T1D cases co-localized with staining with an antibody-blend against CVB, absence of virus is less likely. We also de-tected some signs of an activated innate immune response in the islets from the T1D donors, consistent with a viral infection; expression levels of the chemokines IP-10, MCP-1, and RANTES were up-regulated to some extent in at least one of the T1D donors.

Ultrastructural analysis of islets from the two T1D cases revealed a high degree of decomposed endocrine cytoplasm, mostly in beta cells. An inter-esting finding in affected cells was the presence of insulin granules contain-ing more than one insulin crystal giving the impression of fused granules. The islet septa and perivascular compartments were frequently filled with fibrogenous material. Surprisingly few nuclei showed signs of apoptosis, and the frequent formation of vacuoles and crinophagic bodies suggested a wide-spread ongoing autophagy in these cells. Absence of apoptosis was support-ed by negative IHC staining for cleaved caspase-3. These findings coincide with a functional beta cell block, in which autophagy is increased to clear old insulin granules and maintain the intracellular storage level [181]. Final-ly, the heavy cytoplasmic vacuolization that was found is consistent with a viral remodeling of the cells.

Paper IV – Enterovirus-Induced Disintegration of Human Pancreatic Islets, but not Viral Replication per se, Reduces the Beta Cell Function Selectively Based on the finding in Paper III and in the work by Dotta et al. [63], that islets from cases of recent onset T1D have lost their ability of glucose-stimulated insulin secretion (GSIS) despite a large fraction of cells being

Page 35: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

35

immunopositive for insulin, we in Paper IV wanted to examine whether such a functional beta cell block could be caused by EV infection. EV had been shown previously to reduce insulin secretion by cell lysis [63, 88, 95], but it was not known whether viral replication itself could cause a functional block in the beta cells.

When isolated human islets were infected in this study, the degree and rapidness of virus-induced islet disintegration varied between the different strains of CVB, but also between different donors (Paper IV, Table 1). Pre-vious studies of virus-induced effects on beta cell function had looked at one or more predetermined time point(s)[86-88]. In this study instead, the degree of islet disintegration, not the specific time point or strain of virus, was com-pared with the islet GSIS, revealing that the GSIS is greatly affected when the islets are markedly disintegrated but not when the virus has replicated without observable islet destruction. This may not be surprising, but suggests that a persistent and/or non-lytic infection do not cause T1D by itself. How-ever, in the presence of immune cells and a possible pre-existing latent auto-immunity in vivo, such an infection could be a triggering event.

We also found that the expression of insulin but not glucagon mRNA was lower in islets infected with E2 or VD2921, than in mock-infected control islets. An obvious interpretation of this finding is that these viruses destroy beta cell function selectively. This supports the previous studies arguing for a selective tropism of many EV for beta cells [63, 90, 91]. In islets infected with V89-4557, both glucagon and insulin transcription was downregulated. This virus has been shown previously to, in contrast to most other CVB, infect cells lacking both CAR and DAF [157], giving it a broader tropism possibly also infecting alpha cells.

An important finding in Paper III and previously by Dotta et al [63] was that, in cases of recent onset T1D, a significant number of islet cells stain positive for insulin with IHC despite a completely lost GSIS. In line with this, in paper IV, islets with a very low GSIS and greatly reduced insulin mRNA levels still stained positive for insulin. This is likely due to insulin remaining in beta cells that are about to die, but could also be explained by a functional block. EV infection leads to shut-off of cap dependent translation, and even though insulin can partly be translated via a cap-independent mechanism, most of the insulin synthesis is blocked by eIF4A cleavage [182]. Thus, when beta cells are infected it is likely that if infected, but not disintegrated, islets would be challenged with high glucose for a longer dura-tion than in this present study, the insulin stores would be emptied and the beta cells made unresponsive to high glucose.

A surprising finding in Paper IV was that synthetic double-stranded RNA (Poly[I:C]) potentiated the GSIS in human islets. This is directly contradicto-ry to several reports from poly(I:C) exposure of murine islets, where it has been shown to induce apoptosis and reduce the GSIS [183-187]. The data presented in this paper gives no clues to the mechanism of this potentiating

Page 36: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

36

effect in human islets, but one can speculate that the molecular pathways induced by poly(I:C) in human islets in some way interacts with the path-ways involved in insulin secretion and/or glucose sensing.

In summary, the data in paper IV support that EV can destroy beta cell function selectively, but also show that viral replication alone does not nega-tively affect insulin secretion. Rather, the presence of dsRNA during viral replication or persistence may potentiate the GSIS, contradictory to results in murine models.  

  

Page 37: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

37

Conclusions

Paper I 1. Nicotinamide reduces EV replication and EV-induced islet destruc-

tion/CPE in human islets. 2. Nicotinamide inhibits or reduces EV-induced secretion of IP-10 and

MCP-1 from human islets.

Paper II 1. 17β-estradiol reduces EV- but not poly(I:C)-induced secretion of IP-10

from human islets. 2. EV replication in human islets induces the expression of IFNβ, MDA5,

and TLR3 mRNA. 3. Poly(I:C) exposure of human islets induces an antiviral state which can-

not be counteracted by the virus.

Paper III Two cases of recent onset T1D revealed:

1. Lost beta cell function despite the presence of a relatively large number of intact insulin-containing cells, suggesting a functional beta cell block.

2. Intra-islet bleedings and large swollen cells. 3. Infiltrating immune cells, dominated by CTLs and macrophages, evenly

distributed in the pancreas without obvious accumulation in or around the islets.

4. Multiple islets immunopositive for EV-P1 which in one case was con-firmed with an antibody blend against Coxsackievirus B.

5. Signs of an activated innate immune response in the islets; up-regulation of TLR3, and several chemokines

6. Decomposed endocrine cytoplasm, fused insulin granulae, cytoplasmic vacuolization, dilated ER, few apoptotic cells, and signs of extensive au-tophagy.

Page 38: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

38

Paper IV 1. Infection of human islets with the CVB strains E2-Yoon or Adr, isolated

from T1D patients, reduces the expression of beta but not alpha cell spe-cific genes.

2. EV-induced islet disintegration, affects the ability of the beta cells to secrete insulin in response to high glucose. EV replication without ob-servable induction of islet disintegration has no effect on the glucose-stimulated insulin secretion in vitro.

3. Synthetic dsRNA (polyI:C) has a potentiating effect on glucose-stimulated insulin secretion in human islets.

4. Human islets infected with EV are positively stained with IHC for insu-lin despite extensive islet disintegration and lost ability to secrete insulin in response to high glucose.

Page 39: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

39

General Discussion

The data presented in this thesis focus on enteroviral effects on human pan-creatic islets. Most knowledge on viral effects on host cells relies on studies of immortalized cell lines or animal models. The islets, and more specifically the beta cells, represent a fundamentally different and less well studied cellu-lar host from a viral perspective. First, the beta cells can be considered ter-minally differentiated, meaning that some nuclear factors shown to partici-pate in EV replication may not be present in the cytoplasm [135, 136], which may lead to selection of different quasi-species than when the virus repli-cates in a transformed cell line. Also, it has been shown that the innate im-mune signaling varies heavily between different cell lines and between dif-ferent primary human cells [156]. Given the suspected involvement of beta cell infection in T1D pathogenesis, increasing our knowledge of EV effects on primary human pancreatic islets is obviously of great importance.

Since the most important property of any virus is the ability to replicate and spread efficiently between hosts, it is not likely that infecting the islets of Langerhans, “trapped” in the pancreas far from the surrounding environ-ment, is in the interest of the virus. Rather, the beta cell tropism of EV is more likely a “side-effect” of adaptation to replication in other tissues. Un-derstanding how such “accidental” infection, by EV and other viruses, af-fects the infected cells may have implications for our understanding of many different diseases of so far unknown etiology.

In this thesis it is shown that EV that replicates in human islets activates innate immunity genes and induces secretion of the chemokines MCP-1 and IP-10. An important difference in activation of innate immunity by replicat-ing EV and synthetic dsRNA is suggested, since the chemokine secretion induced by EV infection but not by dsRNA is reduced by addition of the female sex hormone 17beta-estradiol to the culture medium. We also, for the first time, demonstrate a direct antiviral effect of nicotinamide, and even though this substance failed to prevent T1D in a large scale study [174], this finding could have implications for the treatment/prevention of virus- and/or immune-mediated disease.

The rare access to well preserved human pancreata from two organ do-nors with recent onset T1D and several donors with T1D-related autoanti-bodies described in Paper III gave us a great opportunity to study ongoing pathogenic processes at and before the onset of T1D. Despite this, we could neither confirm nor reject the hypothesis that EV is involved in T1D devel-

Page 40: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

40

opment. Several signs, such as the ultrastructural remodeling of the beta cell cytoplasm, activation of innate immunity, and immunopositivity to EV cap-sid protein 1, are supportive of an ongoing virus infection at onset of T1D, but direct evidence is still lacking. Detection of virus with other methods will be essential to provide evidence for EV infection of the islets at onset of T1D, but it is also possible that the virus is present in an earlier stage of dis-ease development, or that T1D is caused by another mechanism.

In conclusion I would like to state that these studies of EV in isolated human islets in vitro support the idea that these viruses can cause T1D in vivo, but future studies will have to show if and how frequently this happens.

Page 41: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

41

Future Perspectives

In this section I will present my personal view of where the field stands to-day and where I think the next years of research should focus.

What Causes Type 1 Diabetes? From the low concordance rate in monozygotic twins, the uneven geographic distribution, the seasonal pattern of onset, and the dramatic increase in inci-dence of T1D, we can draw the conclusion that environmental factors play an important role in the development of the disease.

The basic hypothesis behind this thesis is that EV is involved in the etio-logic mechanisms behind T1D. Although it is almost 50 years old, this hy-pothesis still can be neither confirmed nor rejected. The examination of pan-creata from recent onset T1D patients in this (Paper III) and earlier studies provides no conclusive evidence of virus infection. Most studies are based on IHC with an antibody only (see page 29), and in cases where viral ge-nome has been detected the method used has in most cases been ISH, which does not allow confirmation of specificity by sequencing. The paper by Dot-ta et al [63], isolating an EV from the islets from a case of recent onset of T1D has been questioned since, when sequenced, the virus showed a 99.6% nucleotide identity with JVB, an EV strain isolated during the 1970s and in many labs used as a reference strain. It is quite clear however, that ongoing (detection of EV in blood) and/or recent (EV-IgM in serum) EV infection is temporally associated with the onset of T1D [188-190]. The common pres-ence of EV viremia at T1D onset, together with the susceptibility of human islets to most EV infections in vitro, to me suggests that EV could also commonly be present in the islets at onset of the disease. This is supported by simultaneous detection of EV in cardiac myocytes and in islet cells in children that died of acute EV myocarditis [191] but has yet to be proven.

Even if it can be established that EV (or any other virus) is present in the islets at or before the onset of T1D, in all or most cases, its pathogenic role still needs to be elucidated. The interplay between viral and host genetic variants must be examined. Final proof of EV as a direct cause of T1D is probably not established until vaccination, and/or antiviral therapy, in high-risk individuals have proven to prevent the disease in some or all subjects.

Page 42: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

42

The relative lack of pancreatic tissue and islets from humans with and without T1D has lead many researchers to focus on animal models of the disease, especially the NOD mouse. This has to a large extent taken the fo-cus of the research community from what has actually been described in human pancreata already in the early half of the 20th century. In contrast to studies in the NOD mouse, in human T1D the insulitis is discrete and less frequent. The establishment of more and more centers for islet isolation and transplantation in the last decade has given the scientific community access to human islet tissue, which has proven to be a valuable resource. Also, initi-atives to build up biobanks of human pancreatic tissue has led the field to now shift its focus back to human T1D. In the NOD mice there is no doubt that beta cells are destroyed by an aggressive autoimmune attack directed against the beta cells. However, in humans there is little or no evidence that an autoimmune attack is the major cause of the disease. Immune cells are sometimes present at onset and most likely contribute to the beta cell de-struction, but it may not be the initial or main destructive factor. This is sup-ported by the fact that all attempts to prevent the disease by regulating the immune response have been unsuccessful. Even one-year treatment of newly onset T1D patients with full dose cyclosporine, a very strong inhibitor of T cell function, failed to improve the metabolic control compared to the place-bo group [192].

It is quiet possible that an unknown pathogen or toxin causes the initial beta cell destruction, leading to a subsequent activation of the immune re-sponse, the latter being interpreted as autoimmunity. Also, it is highly possi-ble that T1D is not one or two diseases, but rather a state occurring as a re-sult of lack of insulin that may have several different etiologies. The key to success of future research to identify these mechanisms is most likely to keep research broad and continue to test new hypotheses, and perhaps even more importantly, to reject any hypothesis that is proven false.

Can it be Prevented? Most therapeutic approach to prevent T1D has aimed at regulating the im-mune response, either in general or specifically against islet antigens [28]. But the question needs to be asked; what can we expect from immunomodu-lating therapy if the major and initial cause of beta cell destruction is not the insulitis? So far, no clinical trial has made it past phase III [28], and even heavy immune suppression has failed to effectively prevent T1D [192]. Common for many tested compounds are that they show promising results in NOD mice and in small early studies, with few patients and only limited follow-up time, but fail to efficiently prevent T1D in large clinical studies. Perhaps this can be explained by a scenario where immune mediated de-struction enhances and contributes to the islet destruction, but if the underly-

Page 43: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

43

ing cause is something else the islets will be destroyed anyway, even if the progression may be slightly delayed. Novel tools for earlier prediction and identification of patients at risk for developing T1D may increase the effica-cy of treatment, but identification of one or a few causative factor(s), of viral or other origin, may be necessary to completely prevent T1D.

If EV can be identified as a direct cause of beta cell destruction in T1D, it is possible that vaccination could more or less eradicate T1D. However, vaccination conferring protection against all available EV serotypes is likely not achievable, and identification of one or a few “diabetogenic” serotypes would be necessary. An approach that possibly can have effect against a broader spectrum of EV is antiviral drugs. Three capsid-binding antiviral drugs are in clinical trials for treatment of EV infection today [193], and one of these has been shown to prevent replication of EV in human islets in vitro [194]. Unfortunately, this approach struggles with the rapid emergence of drug resistance [193, 194] and may have only limited effect on persistent EV infection. Development of new anti-EV drugs targeting non-structural pro-teins may be needed. Another strategy could be preventing EV spread from its primary replication site to sites where it may cause severe disease, i.e. the islets of Langerhans. Understanding innate immunity to EV in the islets may aid in development of new drugs. For instance, tissue tropism and patho-genicity of poliovirus can be controlled by the type I IFN response [124]. Also, if the virus is already present in the islets at start of treatment, control-ling the activation of innate immunity (e.g. secretion of chemokines and up-regulation of MHC class I) may be the best way to prevent immune mediated destruction of the beta cells.

Page 44: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

44

Populärvetenskaplig sammanfattning på svenska

Diabetes typ 1 är en sjukdom som beror på att betacellerna i de Langerhan-ska öarna i bukspottskörteln inte längre producerar insulin, ett hormon som behövs för att kroppen ska kunna tillgodogöra sig socker som vi får i oss genom födan. När inte detta fungerar blir blodsockerkoncentrationen hög och utan behandling leder det till döden. Sedan 1920-talet har dock dia-betiker kunnat behandlas med konstgjort insulin.

Trots att man i över 100 år har känt till att sjukdomen beror på att betacel-lerna förstörs vet man än idag inte vad som orsakar den här förstörelsen. Man tror att den delvis beror på att immunförsvaret av någon anledning at-tackerar och förstör betacellerna (s.k. autoimmunitet) men vad som orsakar detta är inte känt. En av de starkaste teorierna är att ett virus bidrar till sjuk-domsförloppet. Redan på 1960-talet kopplades s.k. enterovirus ihop med diabetes typ 1 då man upptäckte att patienter som nyss insjuknat i sjukdomen hade en typ av antikroppar mot enterovirus, som tyder på att de hade en pågående infektion. Enterovirus är en virusgrupp där också Polioviruset ingår, men orsakar normalt endast förkylning eller mycket vaga symptom. Det kan dock i vissa fall även orsaka allvarlig sjukdom såsom hjärtmuskelin-flammation och kanske alltså diabetes. Virus till skillnad från bakterier behöver hjälp av en värdcell för att kunna föröka sig och finns därför alltid inne i cellen. När en cell är infekterad drar den till sig uppmärksamhet från immunförsvaret som kommer och förstör cellen för att på så sätt göra sig av med viruset. En sådan reaktion kan vara mycket svår att skilja från en auto-immun attack.

I den här avhandlingen ingår tre arbeten i vilka vi har renat fram Langerhan-ska öar från bukspottskörteln från avlidna donatorer och infekterat dessa med Enterovirus i cellodlingsskålar för att studera olika effekter som en sådan virusinfektion har på cellerna. I ett fjärde arbete hade vi en unik möjlighet att studera bukspottskörtlar från två organdonatorer som avled precis vid insjuknandet i diabetes typ 1, något som är väldigt ovanligt. När vi infekterade de Langerhanska öarna på vårt laboratorium upptäckte vi att dessa då utsöndrar flera substanser som lockar till sig celler från immun-försvaret, så kallade kemokiner. Vi kunde även minska eller förhindra utsöndringen av dessa substanser genom att tillsätta vitamin B3 (nicotina-

Page 45: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

45

mid) eller kvinnligt könshormon (estradiol). Dessa upptäckter kan bidra till vår förståelse för hur Enterovirus kan orsaka att vårt immunförsvar at-tackerar betacellerna och möjligen hur detta i så fall kan förhindras kliniskt. Vi såg även att viruset i sig förstörde betacellernas förmåga att producera och utsöndra insulin. I arbetet där vi beskriver två patienter som dog vid insjuknandet i diabetes typ 1 försökte vi hitta virus eller spår av virusinfek-tion i öarna, med flera olika metoder. Tyvärr gav detta inte något fullständigt säkert svar. Flera saker tydde på att Enterovirus var närvarande i öarna, bland annat färgades de positivt med en metod för att märka in Enterovirus med hjälp av en konstgjord antikropp, men vi kunde inte påvisa vi-rusförekomst med andra metoder. Ett annat intressant fynd i dessa två buk-spottskörtlar var att inflammationen var mindre än man tidigare trott. Detta har nu även visats i andra studier, och det är tydligt att immunförsvaret inte lika aktivt bidrar till förstörelsen av betaceller som det gör i flera frekvent använda musmodeller av sjukdomen. Efter att har studerat dessa två pank-reasar är det svårt att tro att diabetes typ 1 är en autoimmun sjukdom, ial-lafall i det akuta stadiet. Snarare kan man tänka sig att något, t ex en vi-rusinfektion, inleder förstörelsen men att immunförsvaret bidrar i ett senare skede.

Page 46: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

46

Acknowledgements

The work presented in this thesis was carried out at the Faculty of Medicine at Uppsala University, partly at the Department of Women’s and Children’s Health and at the Department of Immunology, Genetics and Pathology. This could not have been done without an enormous support from a lot of people. Especially I would like to express my gratitude to: My main supervisor, Gun Frisk, for always being open and supportive of my ideas, and for spamming my inbox with suggestions, results, comments, abstracts, and funny links . You always have time for a discussion, and arguing with you has been very stimulating. My co-supervisor and the Islet Group head honcho, Olle Korsgren, for your optimism and for always having a good argument in hand. I truly admire your scientific thinking and your almost pathological obsession of thinking out of the box. You are a great source of inspiration. My fellow PhD student and co-author, Monika Hodik, for your calmness, your diligence and witty personality. I think we have complemented each other well and cooperation with you has always been easy. Thank you also for taking the great cover photo on this thesis with your iPhone! My other co-authors, Annika Moëll and Erik Åhlin, for your great work on Paper I in this thesis, and Annika for fun company in the Keystone snow. Agneta Lukinius, for invaluable expertise on electronmicroscopy and ultra-structure. Other past and present members of the “virus and diabetes” group; Therese Rosenling, Mahesh Anagandula, Maria Hindersson, Asma Elshebani, and Anna-Karin Berg. Also, the students that I have had the pleasure to co-supervise; Victor Mill and Anna Wiberg. Anna who also, together with Rebecka Lindén, made being trapped in Vancouver a good thing. I almost whished Eyjafjallajökull would keep erupting a little longer.

Page 47: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

47

My toughest Swedish competitors, Pär Larsson and Katharina Lind, in the Malin Flodström Tullberg lab, for great company at conferences and meet-ings, and for making the competition more friendly than fierce. If you are nice at the “questions from the audience” maybe you will see that missing SNP finally… Other collaborators and colleagues, Alkwin Wanders, Niklas Nyström, Luis Sarmiento, Corrado Cilio, Sisko Taurianinen, Sami Oikarinen, Heikki Hyöty, Michael Lindberg, and many others. You are great scientist and have contributed to many fun meetings and to stimulating my scientific thinking. The Uppsala Graduate School of Biomedical Research, headed by Cath-arina Svensson, for giving me the best introduction to scientific research one can get, and my fellow UGSBR students for numerous fun pub nights. Björn Carlsson who, in addition to being a great supervisor on my third UGSBR project, introduced me to the Rudbeck world with warnings over orcs and saber-toothed tigers. All past and present members of the islet isolation team. You are too many to mention, but with all of you, endless days of islet isolation have been bearable and even quite fun. I am sorry that I tended to attract multiple pan-creases on the days when I was on-call. Extra credit should be given to; An-na Svensson, who for some strange reason had to deal with me being late a few Sunday mornings, Andrew Friberg for always recommending interest-ing articles and for proof-reading this thesis (unfortunately, all remaining mistakes are my own!), Arian Sadeghi for giving me someone to blame when things went wrong (sorry) and for being a great person, and Fredrik Carlsson for introducing me to the wonderful world of IPAs. Peetra Magnusson and her group members Johan Brännström, Sofia Nordling, and Moa Fransson for great discussions, both scientific and non-scientific. A special appreciation to Johan for boosting my self-esteem by always letting me win in Wordfeud and Rumble (I know you cannot be that bad for real). Everyone else at IGP, and especially Clinical Immunology (both academic and hospital people). You all contribute to making this an excellent place to be! Special thanks to all the technical staff who helped me and others in the lab. Elisabeth Wijkström and the rest of the Q-test people. Anna Karlsson and Susanne Lindblom for never saying no when I came running with blood and spleen samples asking for help, often late in the evenings. Mar-gareta “Bumsan” Engkvist, for helping everyone with everything! I cannot

Page 48: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

48

imagine how this lab will work without you. Inga Hansson, for your ex-tremely hard work with all the stainings and for your positive personality. Berith Nilsson for your pleasant and helpful manner. Past and present ad-ministrative staff at IGP and Klinimm, especially Mona Persson, Ann-Sofie “Anso” Lindberg, and Anna-Maria Andersson. Anso for being my per-sonal administrator, even after leaving Klinimm . Also Christina Anders-son deserves a special mention for helping me getting all the paperwork ready for my dissertation in extremely short notice. All the great people at the Department of Women’s and Children’s Health for providing a good working environment and for great cooperation on solving crossword puzzles. Särskilt tack till klipporna Barbro Westerberg och Karin Wennqvist för era raka sätt och för all hjälp inom alla tänkbara områden. Elisabet Söderberg för din envishet och sköna humor; vä-nskapligt småtjafsande med dig var en självklar höjdpunkt under tiden på KBH. Torsten Tuvemo för att han alltid trott på vår forskning och gett oss sitt fulla stöd. Alla andra vid lunchbordet; Orvar, Lemm, Janne, Helene, Ingrid, Tony, Kjell m. fl. My friends from the Biomedical Program; Knocke (Stor-Fjun), Pontus (Lill-Fjun), Jacob, Danne, Helena, Helén, Ulle, Åsa, Marlene, Sissi, An-ders and Jenny. Special thanks to Knocke for numerous IPAs at Monks, letting me be the best man on your wedding, and for being a great friend; Pontus, for Lederhosen, Maß und Tim Toupet; And of course, Ulle, my personal stylist and smakråd in all areas. I don’t know what I would be with-out you! Teammates and friends in IBS Nabla. Tolerans, kamratskap och ambition, glöm aldrig det! Mina fantastiska uppsalavänner, Torbjörn, Piss-Danne, Lärkan, Poooo, Megap (Ja, jag vet att du blir tackad på två ställen, men Pär Larsson och bajskars-Megap är väl egentligen två olika personer?), Delfinskötarn and many more. Tobias “Torbjörn” Hellström förtjänar här ett särskilt tack för att han ALDRIG säger nej till en liten after work och för att han slog mig i pingis så jag var tvungen att klippa av mig håret. Niklas & Malin Johansson at Agave Tequila Bar for providing a place of refuge and making the best Margaritas in the world! (Alla ni som läser det här borde besöka dem åtminstone en gång och pröva både deras drinkar och chiligryta. Drottninggatan 8. OBS! Jag får inte betalt för att skriva det här.) Jönköpingsvänner, vi ses inte så ofta men de gånger vi gör det brukar det vara årets höjdpunkt. Simon & Jennie ni är de bästa och jag ser fram emot

Page 49: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

49

nästa gång man får komma ut och hälsa på i Konungsö. Fredrik, min stän-dige semesterkompis, det är faktiskt en rätt ful bil du har köpt (sorry), men det ska bli kul att cruisa runt med dig i den i sommar. Viktor, snart kommer det ett vykort i din brevlåda och då hoppas jag att du och din bättre hälft kommer på grillfest! Malte, jag är fortfarande rörd av den fina visaknar-dig.se-hälsningen! Grattis till dig och Maria för den lille Ing…förlåt Ivar! Staffan, tack för folkabussturer, festivalsällskap och fantastiska duetter med skönsång! Gabbe, nu tycker jag det är dags att du flyttar lite längre upp i landet så man får se din resliga kroppshydda lite oftare. Uppsala rekommen-deras, men Stockholm kan duga! Amelie, du är min favoritwordfeudmot-ståndare! Synd att man träffar dig alltför sällan “irl”, men Bruce i sommar blir underbart! CJ, min gamla stensamlarvän och gäddhåvare. Snart har du nog kommit ifatt mig i antal fångade gäddor. Övriga fantastiska (f.d.) jön-nemänniskor som förtjänar ett omnämnande är Elin och såklart Anders & Anna. Sist och viktigast vill jag såklart tacka min släkt och familj. Mamma Ann-Mari & Lennart, Pappa Mikael & Viktoria, mina syskon Viktor, Rebec-ca, Anton och Edvin. Mormor Ruth och Morfar Agnar samt all övrig släkt. Tack för att ni alltid har stöttat mig med allt. Även om vi bara ses någon gång om året så vet ni att ni betyder allt!

Oskar, 17:e April, 2012

Page 50: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

50

References

1. Oxford English Dictionary Online. 2011. 2. Dighiero, G., et al., Natural autoantibodies constitute a

substantial part of normal circulating immunoglobulins. Ann N Y Acad Sci, 1986. 475: p. 135-45.

3. Nussinovitch, U. and Y. Shoenfeld, The Clinical and Diagnostic Significance of Anti-myosin Autoantibodies in Cardiac Disease. Clin Rev Allergy Immunol, 2011.

4. Racanelli, V., et al., Autoantibodies to intracellular antigens: generation and pathogenetic role. Autoimmun Rev, 2011. 10(8): p. 503-8.

5. Lleo, A., et al., Definition of human autoimmunity--autoantibodies versus autoimmune disease. Autoimmun Rev, 2010. 9(5): p. A259-66.

6. Ferreira, C.A., Autoimmune diseases: beyond clinical and/or immune parameters to pathogenic process. Pathol Biol (Paris), 2006. 54(3): p. 119-21.

7. Witebsky, E., et al., Chronic thyroiditis and autoimmunization. J Am Med Assoc, 1957. 164(13): p. 1439-47.

8. Selin, L.K., et al., Heterologous immunity: immunopathology, autoimmunity and protection during viral infections. Autoimmunity, 2011. 44(4): p. 328-47.

9. Fujinami, R.S., et al., Molecular mimicry, bystander activation, or viral persistence: infections and autoimmune disease. Clin Microbiol Rev, 2006. 19(1): p. 80-94.

10. Rahier, J., R.M. Goebbels, and J.C. Henquin, Cellular composition of the human diabetic pancreas. Diabetologia, 1983. 24(5): p. 366-71.

11. Langerhans, P., Beitrage zur mikroskopischen Anatomie der Bauchspeicheldruse. 1869, Friedrich Wilhelm Universität.

12. Jones, P.P., SJ., Islet Function and Insulin Secretion, in Textbook of Diabetes, 4th edition, chapter 6. 2010. p. 87-103.

13. Ballian, N. and F.C. Brunicardi, Islet vasculature as a regulator of endocrine pancreas function. World J Surg, 2007. 31(4): p. 705-14.

14. Suckale, J. and M. Solimena, Pancreas islets in metabolic signaling--focus on the beta-cell. Front Biosci, 2008. 13: p. 7156-71.

15. Rosenfeld, L., Insulin: discovery and controversy. Clin Chem, 2002. 48(12): p. 2270-88.

Page 51: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

51

16. Bell, G.I., et al., Sequence of the human insulin gene. Nature, 1980. 284(5751): p. 26-32.

17. Dai, C., et al., Islet-enriched gene expression and glucose-induced insulin secretion in human and mouse islets. Diabetologia, 2011.

18. Dunn, M.F., Zinc-ligand interactions modulate assembly and stability of the insulin hexamer -- a review. Biometals, 2005. 18(4): p. 295-303.

19. Muller, D., et al., Identification of insulin signaling elements in human beta-cells: autocrine regulation of insulin gene expression. Diabetes, 2006. 55(10): p. 2835-42.

20. DIAMOND, p.g., Incidence and trends of childhood Type 1 diabetes worldwide 1990-1999. Diabet Med, 2006. 23(8): p. 857-66.

21. Harjutsalo, V., L. Sjoberg, and J. Tuomilehto, Time trends in the incidence of type 1 diabetes in Finnish children: a cohort study. Lancet, 2008. 371(9626): p. 1777-82.

22. Variation and trends in incidence of childhood diabetes in Europe. EURODIAB ACE Study Group. Lancet, 2000. 355(9207): p. 873-6.

23. Knip, M., Descriptive epidemiology of type 1 diabetes-is it still in? Diabetologia, 2010.

24. Hermann, R., et al., Temporal changes in the frequencies of HLA genotypes in patients with Type 1 diabetes--indication of an increased environmental pressure? Diabetologia, 2003. 46(3): p. 420-5.

25. Patterson, C.C., et al., Incidence trends for childhood type 1 diabetes in Europe during 1989-2003 and predicted new cases 2005-20: a multicentre prospective registration study. Lancet, 2009. 373(9680): p. 2027-33.

26. Eisenbarth, G.S., Type I diabetes mellitus. A chronic autoimmune disease. N Engl J Med, 1986. 314(21): p. 1360-8.

27. von Herrath, M., S. Sanda, and K. Herold, Type 1 diabetes as a relapsing-remitting disease? Nat Rev Immunol, 2007. 7(12): p. 988-94.

28. van Belle, T.L., K.T. Coppieters, and M.G. von Herrath, Type 1 diabetes: etiology, immunology, and therapeutic strategies. Physiol Rev, 2011. 91(1): p. 79-118.

29. Srikanta, S., et al., Pre-type I diabetes. Linear loss of beta cell response to intravenous glucose. Diabetes, 1984. 33(8): p. 717-20.

30. Lo, S.S., et al., Altered islet beta-cell function before the onset of type 1 (insulin-dependent) diabetes mellitus. Diabetologia, 1992. 35(3): p. 277-82.

31. Robertson, R.P., Estimation of beta-cell mass by metabolic tests: necessary, but how sufficient? Diabetes, 2007. 56(10): p. 2420-4.

Page 52: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

52

32. In't Veld, P., et al., Screening for insulitis in adult autoantibody-positive organ donors. Diabetes, 2007. 56(9): p. 2400-4.

33. Klinke, D.J., 2nd, Extent of beta cell destruction is important but insufficient to predict the onset of type 1 diabetes mellitus. PLoS ONE, 2008. 3(1): p. e1374.

34. Klinke, D.J., 2nd, Age-corrected beta cell mass following onset of type 1 diabetes mellitus correlates with plasma C-peptide in humans. PLoS ONE, 2011. 6(11): p. e26873.

35. Kraus, E.J., Die Pathologischen-Anatomischen Veränderungen des Pankreas beim Diabetes Mellitus. Handbuch der speziellen Pathologischen Anatomie und Histologie. 1922, Berlin: Julius Springer Verlag.

36. Stansfield, O.H. and S. Warren, Inflammation Involving the Islands of Langerhans in Diabetes. New England Journal of Medicine, 1928. 198(13): p. 686-687.

37. Gepts, W., Pathologic anatomy of the pancreas in juvenile diabetes mellitus. Diabetes, 1965. 14(10): p. 619-33.

38. Bottazzo, G.F., A. Florin-Christensen, and D. Doniach, Islet-cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies. Lancet, 1974. 2(7892): p. 1279-83.

39. MacCuish, A.C., et al., Antibodies to pancreatic islet cells in insulin-dependent diabetics with coexistent autoimmune disease. Lancet, 1974. 2(7896): p. 1529-31.

40. Nerup, J., et al., HL-A antigens and diabetes mellitus. Lancet, 1974. 2(7885): p. 864-6.

41. Owerbach, D., et al., HLA-D region beta-chain DNA endonuclease fragments differ between HLA-DR identical healthy and insulin-dependent diabetic individuals. Nature, 1983. 303(5920): p. 815-7.

42. Platz, P., et al., HLA-D and -DR antigens in genetic analysis of insulin dependent diabetes mellitus. Diabetologia, 1981. 21(2): p. 108-15.

43. Kobayashi, T., et al., Pancreatic cytokeratin: an antigen of pancreatic exocrine cell autoantibodies in type 1 (insulin-dependent) diabetes mellitus. Diabetologia, 1990. 33(6): p. 363-70.

44. Hardt, P.D., et al., Distinct autoantibodies against exocrine pancreatic antigens in European patients with type 1 diabetes mellitus and non-alcoholic chronic pancreatitis. JOP, 2008. 9(6): p. 683-9.

45. Panicot, L., et al., Circulating antibodies against an exocrine pancreatic enzyme in type 1 diabetes. Diabetes, 1999. 48(12): p. 2316-23.

46. Andersson, C., et al., The three ZNT8 autoantibody variants together improve the diagnostic sensitivity of childhood and

Page 53: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

53

adolescent type 1 diabetes. Autoimmunity, 2011. 44(5): p. 394-405.

47. Zhang, L. and G.S. Eisenbarth, Prediction and prevention of Type 1 diabetes mellitus. J Diabetes, 2011. 3(1): p. 48-57.

48. Martin, S., et al., Development of type 1 diabetes despite severe hereditary B-lymphocyte deficiency. N Engl J Med, 2001. 345(14): p. 1036-40.

49. Panina-Bordignon, P., et al., Cytotoxic T cells specific for glutamic acid decarboxylase in autoimmune diabetes. J Exp Med, 1995. 181(5): p. 1923-7.

50. Roep, B.O., et al., T-cell reactivity to beta-cell membrane antigens associated with beta-cell destruction in IDDM. Diabetes, 1995. 44(3): p. 278-83.

51. Petersen, L.D., et al., Increased numbers of in vivo activated T cells in patients with recent onset insulin-dependent diabetes mellitus. J Autoimmun, 1996. 9(6): p. 731-7.

52. Kendall, P.L., et al., Tertiary lymphoid structures in the pancreas promote selection of B lymphocytes in autoimmune diabetes. J Immunol, 2007. 178(9): p. 5643-51.

53. Wang, B., et al., The role of CD8+ T cells in the initiation of insulin-dependent diabetes mellitus. Eur J Immunol, 1996. 26(8): p. 1762-9.

54. Foulis, A.K., et al., The histopathology of the pancreas in type 1 (insulin-dependent) diabetes mellitus: a 25-year review of deaths in patients under 20 years of age in the United Kingdom. Diabetologia, 1986. 29(5): p. 267-74.

55. Foulis, A.K., M. McGill, and M.A. Farquharson, Insulitis in type 1 (insulin-dependent) diabetes mellitus in man--macrophages, lymphocytes, and interferon-gamma containing cells. J Pathol, 1991. 165(2): p. 97-103.

56. Willcox, A., et al., Analysis of islet inflammation in human type 1 diabetes. Clin Exp Immunol, 2009. 155(2): p. 173-81.

57. Foulis, A.K., et al., Massive synchronous B-cell necrosis causing type 1 (insulin-dependent) diabetes--a unique histopathological case report. Diabetologia, 1988. 31(1): p. 46-50.

58. Hanafusa, T., et al., Examination of islets in the pancreas biopsy specimens from newly diagnosed type 1 (insulin-dependent) diabetic patients. Diabetologia, 1990. 33(2): p. 105-11.

59. Kimura, N., et al., Vanished islets with pancreatitis in acute-onset insulin-dependent diabetes mellitus in an adult. Arch Pathol Lab Med, 1994. 118(1): p. 84-8.

60. Hanninen, A., et al., 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(5): p. 1901-10.

Page 54: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

54

61. Lernmark, A., et al., Heterogeneity of islet pathology in two infants with recent onset diabetes mellitus. Virchows Arch, 1995. 425(6): p. 631-40.

62. Somoza, N., et al., Pancreas in recent onset insulin-dependent diabetes mellitus. Changes in HLA, adhesion molecules and autoantigens, restricted T cell receptor V beta usage, and cytokine profile. J Immunol, 1994. 153(3): p. 1360-77.

63. Dotta, F., et al., Coxsackie B4 virus infection of beta cells and natural killer cell insulitis in recent-onset type 1 diabetic patients. Proc Natl Acad Sci U S A, 2007. 104(12): p. 5115-20.

64. Shibasaki, S., et al., Expression of toll-like receptors in the pancreas of recent-onset fulminant type 1 diabetes. Endocr J, 2010. 57(3): p. 211-9.

65. Itoh, N., et al., Mononuclear cell infiltration and its relation to the expression of major histocompatibility complex antigens and adhesion molecules in pancreas biopsy specimens from newly diagnosed insulin-dependent diabetes mellitus patients. J Clin Invest, 1993. 92(5): p. 2313-22.

66. Richardson, S.J., et al., Immunopathology of the human pancreas in type-I diabetes. Semin Immunopathol, 2011. 33(1): p. 9-21.

67. Coppieters, K.T., et al., Demonstration of islet-autoreactive CD8 T cells in insulitic lesions from recent onset and long-term type 1 diabetes patients. J Exp Med, 2012. 209(1): p. 51-60.

68. Baekkeskov, S., et al., Antibodies to a 64,000 Mr human islet cell antigen precede the clinical onset of insulin-dependent diabetes. J Clin Invest, 1987. 79(3): p. 926-34.

69. Baekkeskov, S., et al., Autoantibodies in newly diagnosed diabetic children immunoprecipitate human pancreatic islet cell proteins. Nature, 1982. 298(5870): p. 167-9.

70. Baekkeskov, S., et al., Identification of the 64K autoantigen in insulin-dependent diabetes as the GABA-synthesizing enzyme glutamic acid decarboxylase. Nature, 1990. 347(6289): p. 151-6.

71. Christie, M.R., et al., Distinct antibody specificities to a 64-kD islet cell antigen in type 1 diabetes as revealed by trypsin treatment. J Exp Med, 1990. 172(3): p. 789-94.

72. Bonifacio, E., et al., Identification of protein tyrosine phosphatase-like IA2 (islet cell antigen 512) as the insulin-dependent diabetes-related 37/40K autoantigen and a target of islet-cell antibodies. J Immunol, 1995. 155(11): p. 5419-26.

73. Gorus, F.K., et al., IA-2-autoantibodies complement GAD65-autoantibodies in new-onset IDDM patients and help predict impending diabetes in their siblings. The Belgian Diabetes Registry. Diabetologia, 1997. 40(1): p. 95-9.

Page 55: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

55

74. Pav, J., Z. Jezkova, and F. Skrha, Insulin antibodies. Lancet, 1963. 2(7301): p. 221-2.

75. Palmer, J.P., et al., Insulin antibodies in insulin-dependent diabetics before insulin treatment. Science, 1983. 222(4630): p. 1337-9.

76. Landin-Olsson, M., et al., Predictive value of islet cell and insulin autoantibodies for type 1 (insulin-dependent) diabetes mellitus in a population-based study of newly-diagnosed diabetic and matched control children. Diabetologia, 1992. 35(11): p. 1068-73.

77. Wenzlau, J.M., et al., The cation efflux transporter ZnT8 (Slc30A8) is a major autoantigen in human type 1 diabetes. Proc Natl Acad Sci U S A, 2007. 104(43): p. 17040-5.

78. Kumar, D., et al., North-American twins with IDDM. Genetic, etiological, and clinical significance of disease concordance according to age, zygosity, and the interval after diagnosis in first twin. Diabetes, 1993. 42(9): p. 1351-63.

79. Lo, S.S., M.S. Sutton, and R.D. Leslie, Information on type 1 diabetes mellitus and QT interval from identical twins. Am J Cardiol, 1993. 72(3): p. 305-9.

80. Gamble, D.R., et al., Viral antibodies in diabetes mellitus. Br Med J, 1969. 3(5671): p. 627-30.

81. Chehadeh, W., et al., Persistent infection of human pancreatic islets by coxsackievirus B is associated with alpha interferon synthesis in beta cells. J Virol, 2000. 74(21): p. 10153-64.

82. Salminen, K.K., et al., Isolation of enterovirus strains from children with preclinical Type 1 diabetes. Diabet Med, 2004. 21(2): p. 156-64.

83. Salminen, K., et al., Enterovirus infections are associated with the induction of beta-cell autoimmunity in a prospective birth cohort study. J Med Virol, 2003. 69(1): p. 91-8.

84. Hindersson, M., et al., Simultaneous type 1 diabetes onset in mother and son coincident with an enteroviral infection. J Clin Virol, 2005. 33(2): p. 158-67.

85. Yin, H., et al., Enterovirus RNA is found in peripheral blood mononuclear cells in a majority of type 1 diabetic children at onset. Diabetes, 2002. 51(6): p. 1964-71.

86. Frisk, G. and H. Diderholm, Tissue culture of isolated human pancreatic islets infected with different strains of coxsackievirus B4: assessment of virus replication and effects on islet morphology and insulin release. Int J Exp Diabetes Res, 2000. 1(3): p. 165-75.

87. Elshebani, A., et al., Effects on isolated human pancreatic islet cells after infection with strains of enterovirus isolated at clinical presentation of type 1 diabetes. Virus Res, 2007. 124(1-2): p. 193-203.

Page 56: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

56

88. Roivainen, M., et al., Mechanisms of coxsackievirus-induced damage to human pancreatic beta-cells. J Clin Endocrinol Metab, 2000. 85(1): p. 432-40.

89. Yoon, J.W., et al., Isolation of a virus from the pancreas of a child with diabetic ketoacidosis. N Engl J Med, 1979. 300(21): p. 1173-9.

90. Ylipaasto, P., et al., Enterovirus infection in human pancreatic islet cells, islet tropism in vivo and receptor involvement in cultured islet beta cells. Diabetologia, 2004. 47(2): p. 225-39.

91. Richardson, S.J., et al., The prevalence of enteroviral capsid protein vp1 immunostaining in pancreatic islets in human type 1 diabetes. Diabetologia, 2009. 52(6): p. 1143-51.

92. Tanaka, S., et al., Enterovirus infection, CXC chemokine ligand 10 (CXCL10), and CXCR3 circuit: a mechanism of accelerated beta-cell failure in fulminant type 1 diabetes. Diabetes, 2009. 58(10): p. 2285-91.

93. Cubas-Duenas, I., et al., First-degree relatives of persons with type 1 diabetes: insulin resistance and enterovirus infection are associated with different patterns of islet cell autoimmunity. Acta Diabetol, 2011.

94. Oikarinen, M., et al., Analysis of pancreas tissue in a child positive for islet cell antibodies. Diabetologia, 2008. 51(10): p. 1796-802.

95. Yin, H., et al., Complete nucleotide sequence of a Coxsackievirus B-4 strain capable of establishing persistent infection in human pancreatic islet cells: effects on insulin release, proinsulin synthesis, and cell morphology. J Med Virol, 2002. 68(4): p. 544-57.

96. Berg, A.K., O. Korsgren, and G. Frisk, Induction of the chemokine interferon-gamma-inducible protein-10 in human pancreatic islets during enterovirus infection. Diabetologia, 2006. 49(11): p. 2697-703.

97. Olsson, A., et al., Inflammatory gene expression in Coxsackievirus B-4-infected human islets of Langerhans. Biochem Biophys Res Commun, 2005. 330(2): p. 571-6.

98. Godessart, N. and S.L. Kunkel, Chemokines in autoimmune disease. Curr Opin Immunol, 2001. 13(6): p. 670-5.

99. van der Werf, N., et al., Viral infections as potential triggers of type 1 diabetes. Diabetes Metab Res Rev, 2007. 23(3): p. 169-83.

100. Kato, H., et al., Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature, 2006. 441(7089): p. 101-5.

101. Nejentsev, S., et al., Rare Variants of IFIH1, a Gene Implicated in Antiviral Responses, Protect Against Type 1 Diabetes. Science, 2009. 324(5925): p. 387-389.

Page 57: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

57

102. Jermendy, A., et al., The interferon-induced helicase IFIH1 Ala946Thr polymorphism is associated with type 1 diabetes in both the high-incidence Finnish and the medium-incidence Hungarian populations. Diabetologia, 2009.

103. von Herrath, M., Can We Learn From Viruses How to Prevent Type 1 Diabetes? Diabetes, 2009. 58(1): p. 2-11.

104. Takeuchi, O. and S. Akira, Innate immunity to virus infection. Immunol Rev, 2009. 227(1): p. 75-86.

105. Yoneyama, M., et al., The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol, 2004. 5(7): p. 730-7.

106. Pichlmair, A., et al., RIG-I-mediated antiviral responses to single-stranded RNA bearing 5'-phosphates. Science, 2006. 314(5801): p. 997-1001.

107. de Veer, M.J., et al., Functional classification of interferon-stimulated genes identified using microarrays. J Leukoc Biol, 2001. 69(6): p. 912-20.

108. Durand, V., et al., Shaping of adaptive immune responses to soluble proteins by TLR agonists: A role for IFN-alpha/beta. Immunology and Cell Biology, 2004. 82(6): p. 596-602.

109. Picornavirus Home Page. 2012 [cited 2012 Mars 27]; Available from: http://www.picornaviridae.com/.

110. Domingo, E., et al., Quasispecies structure and persistence of RNA viruses. Emerg Infect Dis, 1998. 4(4): p. 521-7.

111. Lin, J.Y., et al., Viral and host proteins involved in picornavirus life cycle. J Biomed Sci, 2009. 16: p. 103.

112. Khetsuriani, N., et al., Enterovirus surveillance--United States, 1970-2005. MMWR Surveill Summ, 2006. 55(8): p. 1-20.

113. Gradi, A., et al., Proteolysis of human eukaryotic translation initiation factor eIF4GII, but not eIF4GI, coincides with the shutoff of host protein synthesis after poliovirus infection. Proc Natl Acad Sci U S A, 1998. 95(19): p. 11089-94.

114. Doedens, J.R. and K. Kirkegaard, Inhibition of cellular protein secretion by poliovirus proteins 2B and 3A. Embo J, 1995. 14(5): p. 894-907.

115. Choe, S.S., D.A. Dodd, and K. Kirkegaard, Inhibition of cellular protein secretion by picornaviral 3A proteins. Virology, 2005. 337(1): p. 18-29.

116. van Kuppeveld, F.J., et al., Coxsackievirus protein 2B modifies endoplasmic reticulum membrane and plasma membrane permeability and facilitates virus release. Embo J, 1997. 16(12): p. 3519-32.

117. Rust, R.C., et al., Cellular COPII proteins are involved in production of the vesicles that form the poliovirus replication complex. J Virol, 2001. 75(20): p. 9808-18.

118. de Jong, A.S., et al., Determinants for membrane association and permeabilization of the coxsackievirus 2B protein and the

Page 58: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

58

identification of the Golgi complex as the target organelle. J Biol Chem, 2003. 278(2): p. 1012-21.

119. Barral, P.M., et al., RIG-I is cleaved during picornavirus infection. Virology, 2009. 391(2): p. 171-6.

120. Barral, P.M., et al., MDA-5 is cleaved in poliovirus-infected cells. J Virol, 2007. 81(8): p. 3677-84.

121. Drahos, J. and V.R. Racaniello, Cleavage of IPS-1 in cells infected with human rhinovirus. J Virol, 2009. 83(22): p. 11581-7.

122. Kotla, S., et al., Attenuation of the type I interferon response in cells infected with human rhinovirus. Virology, 2008. 374(2): p. 399-410.

123. Morrison, J.M. and V.R. Racaniello, Proteinase 2Apro is essential for enterovirus replication in type I interferon-treated cells. J Virol, 2009. 83(9): p. 4412-22.

124. Ida-Hosonuma, M., et al., The alpha/beta interferon response controls tissue tropism and pathogenicity of poliovirus. J Virol, 2005. 79(7): p. 4460-9.

125. Wessely, R., et al., Cardioselective infection with coxsackievirus B3 requires intact type I interferon signaling: implications for mortality and early viral replication. Circulation, 2001. 103(5): p. 756-61.

126. Chapman, N.M. and K.S. Kim, Persistent coxsackievirus infection: enterovirus persistence in chronic myocarditis and dilated cardiomyopathy. Curr Top Microbiol Immunol, 2008. 323: p. 275-92.

127. Julien, J., et al., Postpolio syndrome: poliovirus persistence is involved in the pathogenesis. J Neurol, 1999. 246(6): p. 472-6.

128. Chia, J.K. and A.Y. Chia, Chronic fatigue syndrome is associated with chronic enterovirus infection of the stomach. J Clin Pathol, 2008. 61(1): p. 43-8.

129. Oikarinen, M., et al., Type 1 diabetes is associated with enterovirus infection in gut mucosa. Diabetes, 2012. 61(3): p. 687-91.

130. Tauriainen, S., et al., Enteroviruses in the pathogenesis of type 1 diabetes. Semin Immunopathol, 2011.

131. Kim, K.S., et al., 5'-Terminal deletions occur in coxsackievirus B3 during replication in murine hearts and cardiac myocyte cultures and correlate with encapsidation of negative-strand viral RNA. J Virol, 2005. 79(11): p. 7024-41.

132. Kim, K.S., N.M. Chapman, and S. Tracy, Replication of coxsackievirus B3 in primary cell cultures generates novel viral genome deletions. J Virol, 2008. 82(4): p. 2033-7.

133. Chapman, N.M., et al., 5' terminal deletions in the genome of a coxsackievirus B2 strain occurred naturally in human heart. Virology, 2008. 375(2): p. 480-91.

Page 59: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

59

134. Conaldi, P.G., et al., Persistent infection of human vascular endothelial cells by group B coxsackieviruses. J Infect Dis, 1997. 175(3): p. 693-6.

135. Feuer, R., et al., Cell cycle status affects coxsackievirus replication, persistence, and reactivation in vitro. J Virol, 2002. 76(9): p. 4430-40.

136. Brunner, J.E., et al., Functional interaction of heterogeneous nuclear ribonucleoprotein C with poliovirus RNA synthesis initiation complexes. J Virol, 2005. 79(6): p. 3254-66.

137. Tam, P.E. and R.P. Messner, Molecular mechanisms of coxsackievirus persistence in chronic inflammatory myopathy: viral RNA persists through formation of a double-stranded complex without associated genomic mutations or evolution. J Virol, 1999. 73(12): p. 10113-21.

138. Koskinas, J., et al., Cellular and humoral immune reactions against autoantigens and hepatitis C viral antigens in chronic hepatitis C. Gastroenterology, 1994. 107(5): p. 1436-42.

139. Baboonian, C. and T. Treasure, Meta-analysis of the association of enteroviruses with human heart disease. Heart, 1997. 78(6): p. 539-43.

140. Nishtala, K., et al., Virus-induced dilated cardiomyopathy is characterized by increased levels of fibrotic extracellular matrix proteins and reduced amounts of energy-producing enzymes. Proteomics, 2011. 11(22): p. 4310-20.

141. Gluck, B., et al., Persistent expression of cytokines in the chronic stage of CVB3-induced myocarditis in NMRI mice. J Mol Cell Cardiol, 2001. 33(9): p. 1615-26.

142. Cooper, L.T., Jr., Myocarditis. N Engl J Med, 2009. 360(15): p. 1526-38.

143. Caforio, A.L., et al., Circulating cardiac autoantibodies in dilated cardiomyopathy and myocarditis: pathogenetic and clinical significance. Eur J Heart Fail, 2002. 4(4): p. 411-7.

144. Herskowitz, A., et al., Demographic features and prevalence of idiopathic myocarditis in patients undergoing endomyocardial biopsy. Am J Cardiol, 1993. 71(11): p. 982-6.

145. Gauntt, C.J. and M.A. Pallansch, Coxsackievirus B3 clinical isolates and murine myocarditis. Virus Res, 1996. 41(1): p. 89-99.

146. Li, Y., et al., Enteroviral capsid protein VP1 is present in myocardial tissues from some patients with myocarditis or dilated cardiomyopathy. Circulation, 2000. 101(3): p. 231-4.

147. Roivainen, M., Enteroviruses: new findings on the role of enteroviruses in type 1 diabetes. Int J Biochem Cell Biol, 2006. 38(5-6): p. 721-5.

148. Jun, H.S. and J.W. Yoon, A new look at viruses in type 1 diabetes. Diabetes Metab Res Rev, 2003. 19(1): p. 8-31.

Page 60: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

60

149. Ylipaasto, P., et al., Global profiling of coxsackievirus- and cytokine-induced gene expression in human pancreatic islets. Diabetologia, 2005. 48(8): p. 1510-22.

150. Contreras, J.L., et al., 17beta-Estradiol protects isolated human pancreatic islets against proinflammatory cytokine-induced cell death: molecular mechanisms and islet functionality. Transplantation, 2002. 74(9): p. 1252-9.

151. Eizirik, D.L., et al., Cytokines suppress human islet function irrespective of their effects on nitric oxide generation. J Clin Invest, 1994. 93(5): p. 1968-74.

152. Pavlovic, D., et al., Contribution of ductal cells to cytokine responses by human pancreatic islets. Diabetes, 1999. 48(1): p. 29-33.

153. Horwitz, M.S., et al., Diabetes induced by Coxsackie virus: initiation by bystander damage and not molecular mimicry. Nat Med, 1998. 4(7): p. 781-5.

154. Vreugdenhil, G.R., et al., Molecular mimicry in diabetes mellitus: the homologous domain in coxsackie B virus protein 2C and islet autoantigen GAD65 is highly conserved in the coxsackie B-like enteroviruses and binds to the diabetes associated HLA-DR3 molecule. Diabetologia, 1998. 41(1): p. 40-6.

155. Hyoty, H., Enterovirus infections and type 1 diabetes. Ann Med, 2002. 34(3): p. 138-47.

156. Lundberg, A.M., et al., Key differences in TLR3/poly I:C signaling and cytokine induction by human primary cells: a phenomenon absent from murine cell systems. Blood, 2007. 110(9): p. 3245-52.

157. Frisk, G., T. Elfstrom, and H. Diderholm, The replication of certain Coxsackie B virus strains in CHO cells. J Virol Methods, 2001. 98(2): p. 161-5.

158. Frisk, G. and T. Tuvemo, Enterovirus infections with beta-cell tropic strains are frequent in siblings of children diagnosed with type 1 diabetes children and in association with elevated levels of GAD65 antibodies. J Med Virol, 2004. 73(3): p. 450-9.

159. Goto, M., et al., Refinement of the automated method for human islet isolation and presentation of a closed system for in vitro islet culture. Transplantation, 2004. 78(9): p. 1367-75.

160. Negi, S., et al., Analysis of beta-cell gene expression reveals inflammatory signaling and evidence of dedifferentiation following human islet isolation and culture. PLoS ONE, 2012. 7(1): p. e30415.

161. Roep, B.O. and M. Atkinson, Animal models have little to teach us about type 1 diabetes: 1. In support of this proposal. Diabetologia, 2004. 47(10): p. 1650-6.

Page 61: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

61

162. Oikarinen, M., et al., Evaluation of immunohistochemistry and in situ hybridization methods for the detection of enteroviruses using infected cell culture samples. J Clin Virol, 2010. 47(3): p. 224-8.

163. Harkonen, T., et al., Enterovirus infection can induce immune responses that cross-react with beta-cell autoantigen tyrosine phosphatase IA-2/IAR. J Med Virol, 2002. 66(3): p. 340-50.

164. Harkonen, T., et al., Picornavirus proteins share antigenic determinants with heat shock proteins 60/65. J Med Virol, 2000. 62(3): p. 383-91.

165. Schulthess, F.T., et al., CXCL10 impairs beta cell function and viability in diabetes through TLR4 signaling. Cell Metab, 2009. 9(2): p. 125-39.

166. Shimada, A., et al., Elevated serum IP-10 levels observed in type 1 diabetes. Diabetes Care, 2001. 24(3): p. 510-5.

167. Nicoletti, F., et al., Serum concentrations of the interferon-gamma-inducible chemokine IP-10/CXCL10 are augmented in both newly diagnosed Type I diabetes mellitus patients and subjects at risk of developing the disease. Diabetologia, 2002. 45(8): p. 1107-10.

168. Piemonti, L., et al., Human pancreatic islets produce and secrete MCP-1/CCL2: relevance in human islet transplantation. Diabetes, 2002. 51(1): p. 55-65.

169. Johansson, U., et al., Inflammatory mediators expressed in human islets of Langerhans: implications for islet transplantation. Biochem Biophys Res Commun, 2003. 308(3): p. 474-9.

170. Ichii, H., et al., Improved human islet isolation using nicotinamide. Am J Transplant, 2006. 6(9): p. 2060-8.

171. Moberg, L., et al., Nicotinamide inhibits tissue factor expression in isolated human pancreatic islets: implications for clinical islet transplantation. Transplantation, 2003. 76(9): p. 1285-8.

172. Virag, L., et al., Nitric oxide-peroxynitrite-poly(ADP-ribose) polymerase pathway in the skin. Exp Dermatol, 2002. 11(3): p. 189-202.

173. Oliver, F.J., et al., Resistance to endotoxic shock as a consequence of defective NF-kappaB activation in poly (ADP-ribose) polymerase-1 deficient mice. Embo J, 1999. 18(16): p. 4446-54.

174. Gale, E.A., et al., European Nicotinamide Diabetes Intervention Trial (ENDIT): a randomised controlled trial of intervention before the onset of type 1 diabetes. Lancet, 2004. 363(9413): p. 925-31.

175. Kyvik, K.O., et al., The epidemiology of Type 1 diabetes mellitus is not the same in young adults as in children. Diabetologia, 2004. 47(3): p. 377-84.

Page 62: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

62

176. Uno, S., et al., Macrophages and dendritic cells infiltrating islets with or without beta cells produce tumour necrosis factor-alpha in patients with recent-onset type 1 diabetes. Diabetologia, 2007. 50(3): p. 596-601.

177. Weichselbaum, A. and E. Stangl, Zur Kenntniss der feineren Veränderungen des Pankreas bei Diabetes mellitus. Wiener Klinische Wochenschrift, 1901. 41: p. 968-972.

178. Weichselbaum, A., Über die Veränderungen des Pankreas bei Diabetes mellitus. Sitzungsberichte der kaiserlichen Akademie der Wissenschaften, 1910. 119: p. 73-281.

179. Toreson, W.E., Glycogen infiltration (so-called hydropic degeneration) in the pancreas in human and experimental diabetes mellitus. Am J Pathol, 1951. 27(2): p. 327-47.

180. Blixt, M., B. Niklasson, and S. Sandler, Characterization of [beta]-cell function of pancreatic islets isolated from bank voles developing glucose intolerance/diabetes: An animal model showing features of both type 1 and type 2 diabetes mellitus, and a possible role of the Ljungan virus. General and Comparative Endocrinology, 2007. 154(1-3): p. 41-47.

181. Marsh, B.J., et al., Regulated autophagy controls hormone content in secretory-deficient pancreatic endocrine beta-cells. Mol Endocrinol, 2007. 21(9): p. 2255-69.

182. Fred, R.G., et al., The human insulin mRNA is partly translated via a cap- and eIF4A-independent mechanism. Biochem Biophys Res Commun, 2011. 412(4): p. 693-8.

183. Heitmeier, M.R., A.L. Scarim, and J.A. Corbett, Double-stranded RNA inhibits beta-cell function and induces islet damage by stimulating beta-cell production of nitric oxide. J Biol Chem, 1999. 274(18): p. 12531-6.

184. Rhodes, C.J. and K.W. Taylor, Effect of interferon and double-stranded RNA on B-cell function in mouse islets of Langerhans. Biochem J, 1985. 228(1): p. 87-94.

185. Colli, M.L., et al., Exposure to the viral by-product dsRNA or Coxsackievirus B5 triggers pancreatic beta cell apoptosis via a Bim / Mcl-1 imbalance. PLoS Pathog, 2011. 7(9): p. e1002267.

186. Dogusan, Z., et al., Double-stranded RNA induces pancreatic beta-cell apoptosis by activation of the toll-like receptor 3 and interferon regulatory factor 3 pathways. Diabetes, 2008. 57(5): p. 1236-45.

187. Scarim, A.L., et al., Mechanisms of beta-cell death in response to double-stranded (ds) RNA and interferon-gamma: dsRNA-dependent protein kinase apoptosis and nitric oxide-dependent necrosis. Am J Pathol, 2001. 159(1): p. 273-83.

188. Yeung, W.C., W.D. Rawlinson, and M.E. Craig, Enterovirus infection and type 1 diabetes mellitus: systematic review and

Page 63: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of

63

meta-analysis of observational molecular studies. BMJ, 2011. 342: p. d35.

189. Frisk, G., et al., High frequency of Coxsackie-B-virus-specific IgM in children developing type I diabetes during a period of high diabetes morbidity. J Med Virol, 1985. 17(3): p. 219-27.

190. Helfand, R.F., et al., Serologic evidence of an association between enteroviruses and the onset of type 1 diabetes mellitus. Pittsburgh Diabetes Research Group. J Infect Dis, 1995. 172(5): p. 1206-11.

191. Foulis, A.K., et al., A search for the presence of the enteroviral capsid protein VP1 in pancreases of patients with type 1 (insulin-dependent) diabetes and pancreases and hearts of infants who died of coxsackieviral myocarditis. Diabetologia, 1990. 33(5): p. 290-8.

192. Cyclosporin-induced remission of IDDM after early intervention. Association of 1 yr of cyclosporin treatment with enhanced insulin secretion. The Canadian-European Randomized Control Trial Group. Diabetes, 1988. 37(11): p. 1574-82.

193. Thibaut, H.J., A.M. De Palma, and J. Neyts, Combating enterovirus replication: state-of-the-art on antiviral research. Biochem Pharmacol, 2012. 83(2): p. 185-92.

194. Berg, A.K., et al., Antiviral treatment of Coxsackie B virus infection in human pancreatic islets. Antiviral Res, 2007. 74(1): p. 65-71.

Page 64: NB: Please do not read the lyrics whilst listening to the ...515158/...The Pancreas and the Islets of Langerhans The pancreas is a combined exo- and endocrine gland, consisting of