<<

          

               

 !" #

        $$ %&%'&()&    $! *+'%','*+)&'-   .  ... '%((*%               ! "# $ !%!&' #   '   ' (#  #)* '  +,"#  -   . #,

 ,%,"# / )   +0   ' #  ' ,1   ,         &&2,&% , ,345 26727&&87697!,

"# /  )  +         # -# /         #-  , :      /  /     #     #  #       / #     0 #      ,3# - #      #  ;0, -#<    - #  , (     '        /  #      '  # -   '       '     #;0,1  '  /  '' # #' ',3  '      /  ' & %6       #             #, -' ' /               ,"# ##   ''  # '' #  '          ,*#   '     #          '     '  / ' 86#,"#''  5=7          '   # -  # '5=4 #      # / '',"#      /              '  /   #     '# ,"# / #'       ' #  ' ,1    #  '#>?   -##'   # /   ,  /           ' ' ##     % -#   '          ,= '   # /   #      ' % , - /  #    #   '  '# ,

  /    #         '  / 

   !        ! "#$!  !%&#"$'(  !

@ %

34459&79%9 34526727&&87697!     7%%&)# AA ,/,A B C    7%%&+ List of Papers

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

I Blixt, M., Niklasson, B., Sandler, S. (2007) Characterization of -cell function of pancreatic islets isolated from bank de- veloping glucose intolerance/diabetes: an model show- ing features of both type 1 and type 2 diabetes mellitus, and a possible role of the Ljungan virus. General and Comparative Endocrinology, 154(1-3):41–47

II Blixt, M., Niklasson, B., Sandler, S. (2009) Suppression of bank pancreatic islet function by proinflammatory cytokines. Molecular and Cellular Endocrinology, 305(1-2):1–5

III Blixt, M., Niklasson, B., Sandler, S. (2010) Pancreatic islets of bank vole show signs of dysfunction after prolonged exposure to high glucose in vitro. Submitted

IV Blixt, M., Niklasson, B., Sandler, S. (2010) Morphologic inves- tigation of the endocrine pancreas in diabetic bank voles indi- cates a type 2 diabetes profile. Manuscript

Reprints were made with permission from the respective publishers.

Contents

Introduction ...... 9 Historical aspects of diabetes mellitus ...... 9 Background ...... 11 Animal models used in diabetes research ...... 11 The bank vole ...... 12 The Ljungan virus ...... 13 Proinflammatory cytokines ...... 14 Effects of high glucose ...... 15 Morphological changes of pancreatic islets in diabetes ...... 16 Aim ...... 18 Materials and methods ...... 19 Animal and sample preparations (I – IV) ...... 19 Islet isolation and culture condition (I – III) ...... 20 Islet (pro)insulin biosynthesis rate (I – III) ...... 20 Islet glucose oxidation rate (I – III) ...... 21 Medium insulin and proinsulin accumulation, islet insulin release and insulin content (I– III) ...... 21 Medium nitrite accumulation (II) ...... 22 RNA isolation, cDNA synthesis and Real Time PCR (II, III) ...... 22 DNA quantification (III) ...... 23 Islet cell viability (II) ...... 23 Paraffin removal and antigen retrieval (IV) ...... 23 Haematoxylin staining (I, IV) ...... 24 Fluorescence staining (IV) ...... 24 Morphological evaluation (IV) ...... 24 Statistical analysis (I –IV) ...... 25 Results and discussion ...... 26 Glucose intolerance in bank voles (I) ...... 26 NO independent reduction in insulin biosynthesis (II) ...... 27 Prolonged exposure to high glucose (III) ...... 28 Islet morphology and function (IV) ...... 30 Ljungan virus (I–IV) ...... 31

Conclusions ...... 32 Acknowledgements ...... 33 References ...... 35

Abbreviations

AMV Avian myelobalstosis virus ANOVA Analysis of variance BB Bio-breeding CD-1 Inbred mouse strain derived from ICR mouse cDNA Complementary deoxyribonucleic acid Ct Cycle threshold CY2 Fluorescent dye fluorescein isothiocyanate Db/db–model Monogenic mouse model of obesity –leptin resistant DNAse Enzyme degrading deoxyribonucleic acid dNTP Deoxyribonucleotide triphosphate ELISA Enzyme–linked immunosorbent assay Fa/fa–model Monogenic rat model of obesity –leptin resistant GAD65 Glutamic acid carboxylase isoform 65 GK Goto Kakizaki inbred rat strain HBSS Hanks’ balanced salt solution IA–2 Islet antigen–2 IFN– Interferon gamma IL–1 Interleukin one beta iNOS Inducible nitric oxide synthase IPGGT Intraperitoneal glucose tolerance test Jc1–ICR Inbred mouse strain from Jackson laboratories KK Inbred mouse stain from Kasukabe in Saintama, Japan KRBH Krebs’ Ringer bicarbonate HEPES buffer LADA Latent autoimmune diabetes in adults LV Ljungan virus NOD Non obese diabetic inbred mouse strain NYS Nagoya–Shibata–Yasuda inbred mouse strain Ob/ob–model Monogenic mouse model of obesity –leptin deficient PDX–1 Pancreatic–duodenal homeobox – 1 RNAse Enzyme degrading ribonucleic acid RPMI Roswell Park Memorial Institute RT–PCR Reverse transcriptase polymerase chain reaction S.E.M. Standard error of the mean TBS Tris buffered saline TCA Trichloroacetic acid TNF– Tumor necrosis factor alfa UV Ultraviolet

Introduction

Diabetes mellitus is a group of metabolic disorders where the control of blood glucose homeostasis has been lost due to reduced ability to use carbo- hydrates as energy source. This is a consequence of a failing insulin hor- monal signaling system. The exact etiology of diabetes mellitus is still un- clear however, several theories have been presented arguing that genetic predisposition and environmental factors e.g. viral infections, dietary pro- teins, toxins as well as lifestyle may influence disease development [1-7]. The prevalence of diabetes mellitus is increasing rapidly. At present approx- imately 280 million adult individuals suffer from the disease. It is estimated that within 20 years this number will rise to over 400 million [8]. Diabetes mellitus can be divided into several variants of which type 1 and type 2 are the major diagnostic groups [9]. Type 1 diabetes mellitus is a chronic au- toimmune disease that affects the –cells in the pancreatic islet of Langer- hans, which results in an insufficient endogenous insulin production [10]. Type 2 diabetes mellitus evolves from relative insulin deficiency that is a result of an increasing desensitization of insulin sensitivity in the peripheral tissue and an impaired –cell function [11]. Ultimately both these conditions leads to increased blood glucose concentration and a failing –cell mass. The diabetic patients’ incapacity to metabolize sufficient carbohydrates leads to increasing use of other fuels, preferably fat and protein. This shift in meta- bolism will lead to loss of normal physiological conditions and eventually result in presentation of the classical symptoms of diabetes; fatigue, polydip- sia and polyuria depending on the graveness of the disease.

Historical aspects of diabetes mellitus The first documented observation of diabetes is from the ancient Egypt pre- served in the Ebers’ papyrus dated to 1552 BC [12]. In this papyrus there are descriptions of many diseases, symptoms and treatments including a phrase regarding a remedy that has been translated in two ways “"A medicine to drive away the passing of too much urine," and "To eliminate urine which is too plentiful.". These sentences indicate that diabetes may have been present at this point of time in history. Ebers was not the author but the man who became the owner of the papyrus in 1862 AD. The first known user of the word diabetes, which is Greek, “to pass through”, was Demetrios that lived in the late 2nd century BC in Apameia, today a part of Turkey [13]. It was

9

not until the late 18th century that William Cullen used the word mellitus that is “honey” in Latin [14]. One century later the German histologist Paul Langerhans published a description of distinct groups of cells seen through- out the pancreatic gland [15]. These groups of cells were later referred to as islets of Langerhans. In 1921 Frederick Banting at the University of Toronto and his student assistant Charles Best successfully isolated insulin (initially called "iletin) from dog pancreases, using facilities and resources provided by John Mac- Leod. The findings of the experiments were published in early 1922. The same year in Toronto, Leonard Thompson, a 14 year old boy was the first human treated with insulin. The initial attempt made on January 11 was not satisfactory and James Collip who joined the team in December 1921 made further efforts to increase the purity of insulin. The second injection was made on January 23 and it was a success [16]. In 1923 Frederick Banting and John MacLeod were both awarded the Nobel Prize in medicine. They decided to share their prize sum with Charles Best and James Collip. In 1958 the Nobel Prize in chemistry was awarded to the British molecu- lar biologist Frederick Sanger for his work on the structure of proteins. Fre- derick Sanger was the first to describe the amino acid sequence of insulin [17]. In 1977 the American medical physician Rosalyn Sussman Yalow was awarded the Nobel Prize in medicine for the development of the radioimmu- noassays of peptide hormones like insulin[18]. This assay has become a very important tool in biological and medical research. A major breakthrough in the research of islet biology was the development of islet isolation tech- niques. In 1964 the Swedish islet researcher Claes Hellerström at Uppsala University developed the micro dissection procedure that allowed the isola- tion of the endocrine islets from the exocrine pancreatic tissue [19]. This technique later became an enzyme based procedure and it is today frequently used worldwide in islet research [20, 21]. In 1967 there was a major break- through in insulin research when Donald Steiner discovered the insulin pre- cursor - proinsulin and the insulin biosyntheses pathway [22].

10

Background

Animal models used in diabetes research Perhaps the most famous experimental animal in the research history of di- abetes mellitus was the diabetic dog Marjorie. The dog was pancreatecto- mised and thus developed diabetes. Pancreatectomised dogs seldom survived very long and usually died within a week or two as a consequence of ele- vated blood glucose. Marjorie was the first exception from this scenario as she stayed alive for an extended time period given injections with pancreatic extracts by Banting and Best [23]. Today we still use surgical removal to induce type 1 diabetes in . A less invasive action is to selectively destroy the endocrine pancreas or islet –cells by toxins of which streptozo- tocin [24] and alloxan [25] are two common examples. The use of streptozo- tocin in one large single dose given to will result in the destruction of the –cells and insulin dependence, while multiple low doses may pro- voke diabetes and lead to a scenario more similar to the development of type 1 diabetes with activated immune cells as a key player [26]. Animal models that spontaneously develop type 1 diabetes have been bred in the laboratory. The two most commonly used in research are the non obese diabetic (NOD) mouse model and the BB rat (Bio Breeding Laborato- ries, Ottawa). Both these species develop autoimmune mediated type 1 di- abetes with infiltrating T–cells, B–cells, macrophages and natural killer cells mounting to insulitis, a state of inflammation leading to the destruction of the islets [27, 28]. Also, a variety of islet auto–antibodies have been reported in both the NOD mouse and the BB rat [29, 30]. Islet autoantibodies are tools of the immune system that falsely identify islet constituents as harmful agents leading to disease development. The NOD mouse model originates from the Jc1–ICR mouse strain. Diabetes presents at 12 – 30 weeks of age and there is gender difference with 90% of the females but only 10% of the males developing diabetes [31]. The BB rat is bred from the Wistar rat strain and the animals develop diabetes between 9 – 21 weeks of age [32]. Animal models of type 2 diabetes are a heterogeneous group characte- rized by insulin resistance and impaired insulin secretion. Obesity has a strong relationship with type 2 diabetes mellitus in humans. Animal models of monogenic obesity have therefore been used in research to gain insights into the human disease. An increasing functional demand on the –cell in

11

some models, ob/ob mouse and fa/fa rats is compensated by a vast insulin production leading to hyperinsulinemia, while in the db/db mouse the ani- mals develop hyperglycemia when their –cells are unable to meet the re- quired levels of insulin production [33]. The ob/ob, fa/fa and db/db mouse models are all a result of different changes in the leptin signaling system [34- 36]. Leptin is a peptide hormone involved in the regulation of appetite. The GK (Goto Kakizaki) rat bred from the Wistar rat, NSY (Nagoya– Shibata–Yasuda) mouse derived from the Jc1–ICR mouse and KK mouse are all examples of animal models of polygenic type 2 diabetes [37-39]. The GK rat is suitable to study the diabetes complications; retinopathy, neuropa- thy and nephropathy. The NSY mice spontaneously develop diabetes over time with impaired insulin secretion and mild insulin resistance in adult ani- mals. The KK mouse is another model that with increasing age gradually develops insulin resistance with compensatory hyperinsulinemia and islet cell hyperplasia along with mild obesity. In contrast to the monogenic obesi- ty type 2 diabetes models, obesity is not a major feature of these animals phenotype. In the type 2 diabetes models there is a pronounced gender dif- ference favoring male animals, that to a high extent develop hyperglycemia while in females often less than a third is affected [39]. The Israeli sand rat (Psammomys obesus) is a good model when studying the effects of diet and exercise in the development of type 2 diabetes [40]. Sand rats kept in captivity and given laboratory chow, become obese, insulin resistant and hyperglycemic [41]. The glucose intolerant animals also devel- op hyperlipidemia and atherosclerosis when given a high cholesterol diet [42]. This model has an impaired insulin biosynthesis that results in in- creased circulating proinsulin also, a feature seen in human type 2 diabetes [43].

The bank vole The number of rodents in the northern hemisphere has been shown to fluc- tuate from year to year. The fluctuations are synchronous and regular with density peaks every third to fourth year [44]. The cause of these population variations has not been explained although several theories including “sui- cide marches” and infectious agents have been presented over the years, none being widely accepted. One strain of these rodents, the bank vole (Myodes glareolus previously known as Clethrionomys glareolus) is a small with a body length of 7 – 13.5 cm, tail length of 3.5 – 6.5 cm and a body weight of approximately 12 – 35 g [45]. These animals do not hibernate i.e. are active throughout the year. During summertime they become nocturnal, however, for the rest of the year they are active both day and night. The main diet of the bank vole is vegetarian, including fruit, soft seeds, leaves, fungi, roots, grass, buds and

12

moss. They are also known to eat invertebrate food as snails, worms and insects occasionally. The bank vole has its breeding period from April to October and the gestation period is approximately 21 days long. The bank vole has an expected life span of up to 18 months [46]. In the north and the middle part of Sweden the bank vole population has synchronous cyclic density peaks. The population densities in the adminis- trative provinces of Västerbotten and Örebro have been documented since 1973 [47, 48]. In 1998 it was shown that during the period 1973 – 1989 there was a correlation between the bank vole population density and the inci- dence of human type 1 diabetes mellitus [48]. In a subsequent study of bank voles kept in captivity it was shown that animals develop polydipsia and polyuria, symptoms that suggest that the animals have a disturbed glucose metabolism and there was also gender dif- ference, where male bank voles to a larger extent were affected compared to the females [49]. Indeed, those animals that presented symptoms of glucose intolerance also displayed elevated levels of autoantibodies to glutamic acid carboxylase (GAD65), tyrosine phosphatase–like protein IA–2 and insulin [50]. These findings may suggest that the diabetic condition in these bank voles resembles the type 1 form of the disease [51-53]. Subsequently, it was shown that wild bank voles develop type 1 diabetes mellitus at density peaks [50].

The Ljungan virus The bank voles have been found to host a novel picornavirus named the Ljungan virus (LV) related to both human parechovirus 1 and cardiovirus genus [54, 55]. The first LV was isolated from bank voles resident in the Ljungan valley along the Ljungan River in the Medelpad County, Sweden. Picornavirus particles have been found in the pancreas of the bank voles [54]. Also, it was recently reported that the BB rat model for type 1 diabetes is a carrier of the LV [56]. LV exposure often results in a chronic persistent or long lasting infection. Inoculation of CD–1 mice with the LV during gestation results in the devel- opment of glucose intolerance in the offspring [57]. Furthermore, environ- mental stress has proven to be an important factor playing a vital role for the development of diabetes in this mouse strain. Diabetic CD–1 mice have been tested for the presence of LV with a diagnostic method based on RT–PCR [58]. Although, these mice tested negative with this method there are several examples of animals later proven infectious. The false negative result in this case is probably a consequence of very low viral RNA copy number in per- sistently infected animals. The breeding colony of bank voles at the Astrid Fagreus Laboratory, Ka- rolinska Institute, Stockholm was originally established in Sweden from animals trapped in the provinces of Västerbotten, Småland and Skåne in the

13

1980:s [50]. The breeding colony is considered LV infected. Attempts to determine with accuracy if an individual diabetic bank vole is LV infected or not, have so far not been successful.

Proinflammatory cytokines Cytokines are signal and effector peptides released mainly by the cells of the immune system to induce various responses in target cells. The cytokine signaling has shown to play an important regulatory role during the devel- opment of type 1 diabetes [59]. Besides modulatory effects of the inflamma- tory responses they are also believed to be important for the impairment of the –cell function during the disease progression [60, 61]. Also, in type 2 diabetes the impaired –cell function and the changes seen in peripheral tissues may be related to the effects of cytokine signaling [62-65]. Proinflammatory cytokines are a subgroup of cytokines that favors in- flammation. The peptides are mainly released from activated macrophages and T–lymphocytes and contribute to immune cell activation and recruitment to the site of inflammation. The proinflammatory cytokines IL–1, TNF– and IFN– have been studied extensively in islet research and have been proposed to exert key roles for the detrimental effects seen on the –cell in type 1 diabetes [60, 66]. IL–1 alone or in combination with TNF– and/or IFN– has been suggested to mediate some of these effects through activa- tion of the inducible nitric oxide synthase (iNOS) [67]. Activated iNOS pro- duce nitrogen oxide free radicals (NO) that may perturb islet function and even be toxic to the –cell and ultimately lead to cell death [68-70]. Nitric oxide synthase (NOS) exists in several isoforms all catalyzing NO biosyn- thesis via a reaction involving conversion of L-arginine to L-citruline [71]. The main production of NO in the pancreatic islet takes place in activated intra–islet macrophages but other sources of NO production have also been reported like the –cell itself and endothelial cells [72-75]. NO is a very central effector molecule thus inhibition of NOS can prevent some of the effects seen when islets are exposed to proinflammatory cytokines [69, 76- 79]. There is a high diversity between species in the response to these cyto- kines. In mouse islet cells the iNOS activation can be mediated by IL–1 alone or in combination with TNF– and IFN– [80, 81]. Mouse islets ex- posed to IFN– alone show an impairment of insulin secretion though some conflicting studies exist [80, 82]. In rat islets IL–1 alone or TNF– alone can activate iNOS though when used in combination there is a massive pro- duction of NO [69, 70]. However, in human islets a combination of IL–1, IFN– or IL–1, TNF– and IFN– is needed to activate iNOS [83]. The synergistic effect of IFN– in cytokine mediated –cell damage may depend on enhancement of the IL–1 induced gene expression in the target cells [84-

14

86]. In human islets, IFN– alone induced minor effects like suppressed in- sulin secretion [83, 87, 88]. One explanation for the difference seen between species in regulation of iNOS expression in pancreatic islets has been suggested to be a result of variation in the requirement of transcription factors [89].

Effects of high glucose Prolonged periods of hyperglycemia have proven harmful. The secondary complications often seen in kidneys, eyes, nerves and blood vessels in di- abetic patients have been suggested to be a result of extended time periods with hyperglycemia [90]. However, not only these organs are affected. High glucose concentration also has negative effects on the insulin producing cells. An increased glucose load in vivo on islet –cells has been proposed to participate in disease progression in type 1 and type 2 diabetes [91, 92]. In type 2 diabetes the formation of noxious free oxygen radicals damages the - cell and affects the islet function [93]. However, in type 1 diabetes the de- struction of the –cell mass is primarily mediated by immune cells [94, 95]. An increased functional demand on isolated islets in vitro has shown that hyperglycemic–like conditions can alter islet function and be detrimental for the –cell, but depending on the species of origin this effect varies [93-97]. Mouse islets cultured at 28 mM glucose for 3 days are not affected by an increased functional demand. However, after 1 week in culture the islets display decreased insulin content and enhanced glucose stimulated insulin release. These islets also display increased accumulation of insulin in their culture medium. Islets cultured at 5.6 mM glucose also had an increased (pro)insulin biosynthesis rate in short–term incubation at 1.7 mM glucose. With small variations between strains murine –cells are affected after long term high glucose culture in vitro. However, it appears that they can adapt to this harsh environment and avoid glucose induced cell death [94, 98]. Rat islets cultured at high glucose for 2 days have reduced insulin content [99]. In some studies a reduced glucose stimulated insulin release could be seen [100]. However, in other studies no obvious effect was observed [97]. Islets isolated from the Israeli sand rat and cultured at high glucose have markedly decreased insulin content after 3 days. Further culture of these islets resulted in an increase in the proportion of proinsulin in the islets [101]. Moreover, in a study where islet cells from normal Psammomys ob- esus were explanted in vitro and cultured as monolayer it was found that an elevated glucose concentration (33.3 mM) for 10 days caused a pronounced reduction in glucose induced insulin secretion and insulin content. There was also increased proinsulin–related peptide in the culture medium [96]. Islets from animals kept on a high energy diet display glucose-induced apoptosis and reduced -cell proliferation [102].

15

Human islets exposed to a prolonged culture period at high glucose have reduced insulin content, increased basal insulin secretion and reduced re- sponse upon glucose stimulation. These islets also have a reduced (pro)insulin biosynthesis rate and total protein biosynthesis rate. However, the cell viability was not changed although the glucose oxidation rate was reduced [93].

Morphological changes of pancreatic islets in diabetes The clinical manifestation of diabetes mellitus is probably preceded by a prolonged period of cell mass destruction [103, 104]. The morphologic hallmark of type 1 diabetes has been considered the presence of inflamma- tion lesions in the pancreatic islets [105, 106]. In these type 1 diabetic pa- tients the partial or total islet destruction is supposed to be mediated by im- mune cells and proinflammatory cytokines [60, 107-109], while in type 2 diabetic patients the question of cell mass deterioration is more controver- sial. Previous studies have reported of unchanged cell mass in type 2 di- abetic patients [110, 111], whereas more recent studies have shown that the cell mass is affected even in these subjects [112, 113]. The reduction of the cell mass in type 2 diabetic patients has been suggested to be a result of lipotoxicity as well as glucotoxicity [114, 115]. The Israeli sand rat develops morphological changes in the pancreatic is- lets in parallel with the development of hyperglycemia, when given high energy diet [116]. Islets from severely diabetic sand rats have vacuolization in their cells and deposits of lipids [116-118]. The severely altered islet structure seen in these animals is a result of prolonged disease progression and is apparent only at the end–stage of the disease [116]. Islets in glucose intolerant sand rats early during disease progression have degranulated – cells, which can be seen as weak staining for insulin [119][122]. arvalis is another wild rodent that develops glucose intolerance. In accordance with the sand rat, early during disease progression, the pan- creatic islet cells display degranulation and the animals develop hypergly- cemia and hyperinsulinemia. In severely diabetic animals the pancreatic islet –cells display vacuoles described as glycogen deposits and the animals become hypoinsulinemic [120]. Another animal model showing deposits of glycogen in the islets when cultured at high glucose conditions in vitro is the Wistar rat [121]. In these islets the glycogen appears to be localized to the – cell while the other islet cells seem unaffected [122]. Hydropic degeneration is a degenerative change of the cell structure that resembles cloudy swelling. In rabbit islets hydropic degeneration has been demonstrated in the –cell and proven to be glycogen deposits [123]. Mor- phologic changes resembling hydropic degeneration can be induced in both

16

rabbit –cells and in rat –cells using various chemical substances [124- 126].

17

Aim

Paper I In this paper we aimed to characterize the function of pancreatic islets iso- lated from glucose intolerant/diabetic and glucose tolerant/normal bank voles, in order to investigate if the bank vole could become a future model for the study of human diabetes.

Paper II Herein we studied the function of pancreatic islets isolated from glucose tolerant/normal male bank voles after prolonged exposure to proinflammato- ry cytokines in vitro.

Paper III The aim of this study was to investigate functional alterations of islets iso- lated from glucose tolerant bank voles after prolonged exposure to various glucose concentrations in vitro.

Paper IV The focus of this study was to characterize the islet morphology of glucose intolerant/diabetic animals and further investigate if a relationship could be seen between structural changes in the islets and the observed animal pheno- type.

18

Materials and methods

Animal and sample preparations (I – IV) Bank voles (Myodes glareolus) were housed at Astrid Fagreus Laboratory, Karolinska Institute, Stockholm. The breeding colony was originally estab- lished from animals trapped in Sweden in the provinces of Västerbotten, Småland and Skåne [50]. Animals had free access to water and standard laboratory chew (LABFOR R3, Lactamin, Kimstad, Sweden) with an energy content of 3.01 kcal/g and were occasionally given pieces of vegetables. The experimental procedures were approved by the animals’ ethical committee in Stockholm (N248/03, N276/06) and in accordance with international guide- lines (NIH publications no.85–23, revised 1985). Animals were tested with an intraperitoneal glucose tolerance test (IPGTT). Blood glucose determinations were performed with an automated glucose meter (Paper I, Precision PCX; Abbott Inc., Stockholm, Sweden; Paper II, III, IV, Accu-Chek Aviva; Roche Diagnostics, Bromma, Sweden) on whole blood. Animals were injected with 2 g glucose/kg body weight and samples were taken from the retro–orbital sinus immediately prior to injec- tion (0 min), at 60 and 120 min after glucose injection. A bank vole with 120 min blood glucose 11.1 mM was classified as glucose intolerant/diabetic. The animals were killed by cervical dislocation and exsanguinations via the carotid arteries. The animals were anaesthetized with Isoflurane (Abbott Inc.) prior to testing and killing. After killing the animals the pancreas was removed and kept on ice in Hanks’ balanced salt solution (HBSS; SBL Vac- cine, Stockholm, Sweden) supplemented with 50 U/ml benzylpenicillin and 0.05 mg/ml streptomycin (Roche) or placed in 4 % formalin for 48 hrs. In the latter case the fixed pancreas was washed and stored in 70 % ethanol until embedded in paraffin. The tissue was sectioned (5 μm) and attached to POLYLYSINETM glass slides (Menzel–Gläser; Braunschweig, Germany). Serum was prepared from the blood samples, frozen and stored at -70°C. The serum insulin level was later measured using high range rat insulin ELI- SA kit or rat insulin ELISA kit (Mercodia AB, Uppsala, Sweden) according to the instructions of the manufacturer.

19

Islet isolation and culture condition (I – III) The pancreas was inflated with HBSS, cut in pieces and digested at 37 C with collagenase A (Roche) dissolved in HBSS to a final concentration of 3.125 mg/ml. Islets of Langerhans were handpicked using a braking pipette under a stereomicroscope and transferred to sterile non-attachment culture dishes containing culture medium RPMI 1640 with 11.1 mM glucose sup- plemented with 10 % fetal calf serum (FCS), 2 mM glutamine (Sigma– Aldrich Sweden AB, Stockholm, Sweden) and antibiotics (see above). (I) Islets from glucose tolerant/normal and glucose intolerant/diabetic female and male bank voles 4 to 44 weeks of age were either studied immediately after isolation or cultured at 37 °C, in humidified air + 5 % CO2 (AGA, Stockholm, Sweden) for 7 days. Culture medium was exchanged every second day. (II) Islets from glucose tolerant/normal male bank voles 1520 weeks of age were precultured in 5 days and then transferred to new culture dishes con- taining culture medium as above, with or without addition of IL–1 (25 U/ml; human; PeproTech EC Ltd., London, UK) alone or in combination with TNF– (1000 U/ml; human; PeproTech) + IFN– (1000 U/ml; murine; PeproTech) and cultured for additional 48 hrs before tested. Bank vole spe- cific cytokines are not commercially available. Also we added 2 mM amino- guanidine to randomly selected dishes with or without all three cytokines during the 48 hrs culture period. (III) Glucose tolerant/normal female and male bank voles 4–42 week of age were used to study the functional changes upon prolonged exposure to high glu- cose concentration. Islets from two bank voles of the same gender and simi- lar body weight were pooled and distributed into three new culture dishes with 50 islets of equal size in each and further cultured for 5 days in culture media supplemented with glucose to a final concentration of 5.6, 11.1 or 28 mM before examined. The culture media was exchanged on day 1 and 3. In separate experiments islets were cultured in medium containing 11.1 mM D– glucose + 16.9 mM L–glucose to explore possible osmotic effects of a high glucose concentration.

Islet (pro)insulin biosynthesis rate (I – III) The (pro)insulin biosynthesis analysis was performed using 10 islets in dup- licate that were incubated for 2 hrs at 37°C in humidified air + 5 % CO2 in

20

1.7 mM or 16.7 mM glucose, Krebs-Ringer bicarbonate buffer [127] with 10 mM HEPES (Merck Eurolab, Stockholm, Sweden), hereafter designated as KRBH, supplemented with 3H-labeled leucine to a concentration of 0.4 μM (120 Ci/mmol; American radiolabeled Chemicals, St. Louis, MO, USA) and 2 mg/ml bovine serum albumin (BSA, ICN Biochemicals, Irvine, USA). After the incubation the islets were washed with non–radioactive leucine (10 mM; Sigma–Aldrich) dissolved in HBSS and the islets were dispersed in 200 μl H2O through sonication. A volume of 10 μl of the homogenate was mixed with 50 mM glycine (Merck Eurolab), 6 mM NaOH, 0.1 % Triton X–100 (Sigma–Aldrich) and 2.5 mg/ml BSA pH 8.8 and 5 mg Sepharose A (Amer- sham Pharmacia Biotech AB, Uppsala, Sweden). (Pro)insulin was precipi- tated using a polyclonal anti–serum raised in guinea pig against bovine insu- lin (Chemicon Inc., Hampshire, UK) and normal guinea pig serum was used to assess unspecific bindings. Another 10 μl homogenate was dissolved in 25 mM glycine, 3 mM NaOH and 1.25 mg/ml 0.305 M TCA (Merck) to calcu- late total protein synthesis rates. The pellet was dissolved in 0.15 M NaOH. Radioactivity was measured in a liquid scintillation counter after adding 4 ml of Ultima Gold scintillation fluid (PerkinElmer Life Sciences, Boston, USA).

Islet glucose oxidation rate (I – III) The islet glucose oxidation rate was studied using duplicate or triplicate vials containing 10 islets in KRBH, supplemented with radioactive labeled D-[U- 14C]glucose (16 Ci/mmol; Amersham Pharmacia) and non-radioactive glu- cose to a final concentration of 1.7 mM or 16.7 mM. Islets were incubated at 37°C (O2/CO2; 95/5 %; AGA) under slow-shaking for 90 min. After the incubation antimycin A (Sigma–Aldrich) was added to a final concentration of 0.017 mM and 250 μl 1 M hyamine 10–X (PerkinElmer) in the outer vials reincubation at 37°C in 2 hrs. Radioactivity was measured in a liquid scintil- lation counter after adding 5 ml of Ultima Gold scintillation fluid (Perki- nElmer) [128].

Medium insulin and proinsulin accumulation, islet insulin release and insulin content (I– III) Media from the last 48 h of culture was collected for determination of me- dium insulin and proinsulin accumulation. Islets in triplicate of 10 were in- cubated at 1.7 mM glucose in KRBH supplemented with 2 mg/ml BSA at 37°C (O2/CO2; 95/5 %; AGA) for 1 h. Subsequently the buffer was replaced with KRBH supplemented with 16.7 mM glucose and incubated in an addi- tional h. Islet insulin content was measured on islets pooled from the tripli- cates above after disruption in H2O and extraction overnight at 4°C in 6870

21

% ethanol (Solveco Chemicals AB, Stockholm, Sweden) supplemented with 0.13 mM HCl (Merck). The insulin concentrations in the culture media, the incubation buffers and the insulin extractions were measured with either a rat insulin high range ELISA kit or a rat insulin ELISA kit (Mercodia). The proinsulin concentration in the culture media was measured with a rat Proin- sulin ELISA kit (Mercodia) according to the instructions of the manufactur- er.

Medium nitrite accumulation (II) Samples from the culture medium were mixed with 0.05 % N–(1–naphtyl)– ethylenediamine dihydrochloride (Sigma–Aldrich) and 0.5 % sulfanilamide dissolved in phosphoric acid (Merck). Samples were incubated at 60°C for 2 min followed by incubation at room temperature in 20 min. Nitrite levels were determined through spectrophotometeric absorbance measurement at 546 nm. Culture media collected from wells without islets was used as con- trol of the original nitrite level in the media.

RNA isolation, cDNA synthesis and Real Time PCR (II, III) Total RNA was extracted from 50 islets using RNeasy Micro kit (Qiagen, Hilden, Germany) complemented with RNase–free DNAse (Qiagen) to en- sure DNA free samples and eluted in RNAse free water. The RNA amount and purity was tested using the Nanodrop ND–1000 system (NanoDrop Technologies, Delaware, USA). Synthesis of cDNA was performed with Reverse Transcription System (Promega, Madison, USA) using 1:2 volume of total RNA and 0.025 g/μl Oligo(dT)15 primer per cDNA synthesis reac- tion mixed with reaction buffer (5 mM MgCl2, 10 mM Tris-HCl (pH 9.0), 50 mM KCl, 0.1% Triton® X–100, 1 mM each dNTP, 0.5 U recombinant RNa- sin® ribonuclease inhibitor, 15 U AMV (Avian Myeloblastosis Virus) re- verse transcriptase. The reactions were incubated at 42 C for 60 min fol- lowed by 99 C in 5 min and subsequently stored at -20 C. The LightCycler Instrument (Roche) combined with sequence indepen- dent detection with SYBR Green I was used to amplify and analyze generat- ed cDNA. The sequence of the primers used were: insulin (mouse) Forward 5´-CCATCAGCAAGCAGGTTAT-3´ Reverse 5´-GGGTGTGTAGAAGAAGCCA-3´, PDX–1 (mouse) Forward 5´-GGTGCCAGAGTTCAGCGCTA -3´ Reverse 5´-TTGTTTTCCTCGGGTTCCGC-3´ –actin (mouse) Forward 5´-CCACCGATCCACACAGAGTACTTG-3´ Reverse 5´-GCTCTGGCTCCTAGCACC-3´ (TIB MOLBIOL Syntheselabor GmbH, Berlin, Germany) PCR amplifica- tion of 0.1 μg of cDNA sample was performed with 0.2 μM of each insulin

22

or PDX–1 primer and 0.5 μM of each –actin primer in SYBR Green JumpStart ready mixture (FastStart Taq DNA polymerase, dNTP mix and SYBR Green I dye; Roche) and 3.5 mM MgCl2 (Roche). For each reaction, the polymerase was activated by preincubation in 95 C 30 sec. Amplifica- tion was as follows: 5 sec at 94 C, 10 sec at 47 C and 15 sec at 72 C in 40 cycles. Cycle threshold (Ct) values were obtained for individual samples with the second derivative maximum method [129, 130]. The relative ex- pression was calculated from the formula 2-(Ct), were Ct is the difference between the insulin Ct value or PDX–1 Ct value and the –actin Ct value. RNase-free water was used as a negative control to assure contamination free reagents.

DNA quantification (III) DNA content was measured with fluorometric assay using picoGreen (Mole- cular Probes, Eugene, CA, USA) labeling system according to the instruc- tions of the manufacturer. The DNA content was measured in water homo- genates in conjunction to determinations of the islet insulin content, total protein and (pro)insulin biosynthesis rates.

Islet cell viability (II) Islet viability was determined using Hoechst 33342 and propidium iodide (Sigma–Aldrich) staining [131]. In each test 10 islets were incubated with 5 μg/ml bisBenzimide (Sigma–Aldrich) and 20 μg/ml propidium iodide for 20 min. The islets were washed in PBS and the emitted fluorescence was ob- served at 461 nm and 615 nm respectively after excitation with UV light. The nucleus morphology and the number of blue and red fluorescent cells were counted in a microscope (Leica Microsystems GmbH, Wetzlar, Ger- many) by a blinded observer.

Paraffin removal and antigen retrieval (IV) All incubations were performed in room temperature and the water was deionized prior to use unless otherwise stated. The slides were washed twice for 10 min in xylene to ensure paraffin removal. The sections were rehy- drated using an ethanol dilution series finishing in water. The antigen re- trieval was performed using the pressure cooker 2100 retriever system (Pres- tige Medical, California,USA) and the BORG–decloakerTM RTU buffer (Biocare Medical, California, USA) according to the description of the man- ufacturer. After boiling the retriever buffer was removed and the sections were washed twice with heated Hot Rinse buffer (Biocare Medical). Subse- quently, the buffer was exchanged and the slides washed twice for 5 min with water and twice for 5 min in Tris–buffered saline pH 7.6 with TWEEN

23

20 (TBS, Biocare Medical). TBS was hereafter used as wash buffer and sol- vent for antibody preparations.

Haematoxylin staining (I, IV) The tissue was counterstained using Mayer’s haematoxylin solution (Histo- lab, Göteborg, Sweden). The excess color was removed by washing in non- deionized water in 10 min. The sections were dehydrated using an ethanol dilution series finishing in ethanol. The slides were mounted with cover glasses (Menzel–Gläser) using the Microm CTM6 system (Microm Interna- tional GmbH, Walldorf, Germany). The sections were examined in bright field microscope (Leica Microsystems GmbH) using 40x and 400x magnifi- cations.

Fluorescence staining (IV) Unspecific binding of the secondary antibody was blocked during 1 h incu- bation in a humidified chamber using normal donkey serum (1:20; Jackson ImmunoResearch Laboratories, West Grove, Pennsylvania, USA). The pri- mary antibodies were monoclonal antiserum raised in chicken against human insulin (1:1000; Immunsystem, Uppsala, Sweden) and human glucagon (1:100; a kind gift from Professor Anders Larsson, Uppsala University Hos- pital, Sweden) as well as monoclonal antiserum raised in sheep against so- matostatin (1:75; Guildhay, Guildford, UK) and human pancreatic polypep- tide (1:1; SeroTech, Oxford, UK). Excess blocking buffer was removed prior to application of the primary antibody thereafter the slides were reincubated in the humidified chamber at 4 C over night. The secondary antibodies were raised in donkey against chicken or sheep and conjugated with cyanine dye– Cy2 (1:200; IgG, Jackson ImmunoResearch Laboratories). Unbound primary antibody was removed by washing thrice for 5 min. The secondary antibody was applied to the sections and the slides were reincubated in the humidified chamber for 1 h and subsequently washed thrice for 5 min. The slides were mounted with cover glasses (Menzel–Gläser) using 87% glycerol (Calbio- chem, California, USA). The islets were photographed using a fluorescence microscope with filter cube L5 (emission filter 527/30 nm) at 200x magnifi- cation (Leica Microsystems GmbH). The specificity of the antibodies was tested using different dilutions of the antibodies as well as by omitting the primary antibodies.

Morphological evaluation (IV) Glucose tolerant/normal and glucose intolerant/diabetic male and female bank voles 928 weeks of age were used to study the morphology of the

24

endocrine pancreas. The total islet area as percent of the total pancreatic area was measured in 2 sections at least 300 μm apart stained with haematoxylin. The insulin, glucagon and somatostatin positive areas and total islet area were studied on slides stained using immunohistochemistry and fluorescent dyes. For each animal we measured the islet positive area of insulin, gluca- gon, somatostatin and pancreatic polypeptide in 10 islets from 2 sections and expressed the results as percent of the total islet area.

Statistical analysis (I –IV) Mean values were calculated from duplicate or triplicate measurements and then considered as one separate observation (n). In paper I, II and IV each observation (n) represents a single donor. In paper III each observation (n) represents pooled islets from two donors matched by gender and body weight. Values are expressed as means  S.E.M., and groups of data were compared using Student’s unpaired or paired t-test, one or two way ANOVA using Bonferroni correction, Fisher’s exact test or Mann-Whitney rank sum test. We also tested if an independent variable could be used to predict a dependent variable using linear regression and Galton log-normal regression analysis [132]. Observed differences were considered statistically significant when P 0.05. Statistical analysis were performed using SigmaStat0? (SPSS Inc., Chicago, IL, USA).

25

Results and discussion

Glucose intolerance in bank voles (I) Bank voles caught in nature and subsequently bred in captivity develop a disturbed glucose homeostasis. The bank vole breeding colony at the Astrid Fagreus Laboratory has been shown to have a glucose intolerance/diabetes incidence of approximately 20 % when maintained on a standard laboratory diet. The animals developed glucose intolerance early in life and the majori- ty of all diagnosed bank voles were between 5 to 20 weeks of age. In this colony it appears as if more males than females developed glucose intoler- ance/diabetes early during this period. We used an intraperitoneal glucose tolerance test (IPGTT) as a diagnostic method to determine the glucose tolerance status of the animals in the colo- ny. Based on the IPGTT test results two groups of glucose intolerant/diabetic animals could be distinguished, those with blood glucose close to the cutoff value of 11.1 mM glucose (~70%) and those with severely elevated blood glucose (> 16.7 mM). The glucose intolerant/diabetic animals were also suffering from hyperinsulinemia, and although without obvious signs of obesity, they displayed a trend towards slightly higher body weight than glucose tolerant bank voles. We also observed that male bank voles classi- fied as glucose intolerant/diabetic alone were hyperglycemic. The bank voles suffering from glucose intolerance/diabetes have pre- viously been reported to display features of human type 1 diabetes. The mas- sive destruction of the islet –cells and the presence of islet autoantibodies in these animals support the notion of a new possible animal model of type 1 diabetes [133]. However, the symptoms described above are more similar to type 2 diabetes–like condition. Perhaps the condition observed in the glucose intolerant/diabetic bank voles are a mixture of the two variants. In humans there are a somewhat similar diabetic condition known as latent autoimmune diabetes in the adults (LADA) in which patients with type 2 diabetes pheno- type exhibit islet autoantibodies [134]. The diabetic bank vole shares fea- tures with this condition. Pancreatic islets isolated from glucose intolerant/diabetic male bank voles displayed an increased basal insulin secretion acute after isolation compared to islets from glucose tolerant/normal male bank voles. After one week in culture the islets also increased their response to glucose and the basal insu- lin secretion was now even more elevated compared to the acute performed

26

experiments. After culture the glucose stimulated insulin release was ele- vated in islets isolated from glucose intolerant/diabetic males compared to the basal insulin secretion of such islets. Furthermore, the insulin content of islets from glucose intolerant/diabetic males was elevated after culture. Islets isolated from glucose intolerant/diabetic female bank voles did not have these features. However, islets isolated from glucose tolerant/normal female bank voles had increased insulin content acute after isolation compared to islets from normal males. These findings may indicate a gender difference in that the -cells of the glucose intolerant/diabetic males were more affected by an increased func- tional demand in vivo than the females. Indeed, an increased functional load on the –cells may well exist during development of both type 1 diabetes [94] and type 2 diabetes in rodents [135]. Furthermore the altered function observed acutely in islets isolated from glucose intolerant/diabetic male bank voles could be reversed by culture and did not represent a permanent dys- function. It has previously been shown that islets isolated from prediabetic NOD mice had an impaired glucose stimulated insulin secretion, but the – cell function could then subsequently be restored after culture [95]. This does not exclude that bank voles developing diabetes have –cells that dege- nerate and islets that gradually disappear from the pancreas.

NO independent reduction in insulin biosynthesis (II) The proinflammatory cytokines, IL–1, TNF– and IFN– together reduced the relative contribution of labeled (pro)insulin to the total protein biosyn- thesis rate in bank vole islets after 48 hrs exposure. This was mainly a result of a decreased (pro)insulin biosynthesis rate. Further, these islets also had markedly reduced insulin gene expression and a tendency towards reduced PDX–1 gene transcription. The proinflammatory cytokines also led to re- duced islet insulin content and increased basal insulin secretion in the bank vole after 48 hrs exposure. IL–1 alone had the same effect on insulin con- tent after only 24 hrs exposure, however, the basal insulin secretion was not affected at this time point compared to non–exposed control islets. The proinflammatory cytokines had little effect on the glucose stimulated insulin release and no effect on the insulin accumulation in the medium. These find- ings suggests that the reduction in islet insulin content was not attributed to an enhanced exocytosis and not related to an altered glucose metabolism, but rather was due to a decline in the formation of insulin. This pattern is partly similar and partly different from what has been seen in similar experiments using islet preparations from other species. In rat pancreatic islets, IL–1 induced strong suppressive effects on all functions described above and it is likely that impaired glucose metabolism is a key factor [70, 88, 97]. Con- cerning mouse pancreatic islets, exposed to IL–1, the insulin content and

27

(pro)insulin biosynthesis rates were depressed, medium insulin accumulation and glucose oxidation rates essentially unaffected, but glucose stimulated insulin release reduced by about 50 % [136]. Furthermore, human islets cul- tured with IL–1 alone for a prolonged period can be shown to have a stimu- lated –cell function [137]. When a combination of cytokines was added a marked increase in medium insulin accumulation was present as well as a decrease in islet insulin content, a decline in relative glucose induced insulin release (16.7/1.7), but no change in glucose oxidation rate [83]. NO is often assessed by nitrite medium accumulation in cell culture expe- riments. Bank vole islets exhibited a modest increase in medium nitrite after 48 hours cytokine exposure. When aminoguanidine, a preferential iNOS inhibitor was added to these islets [138] the medium nitrite level was re- duced, but the reduction in islet insulin content was not affected suggesting that the decline in islet insulin content was not NO–dependent. It is likely that an iNOS and NO–independent pathway is responsible for the actions we observed in proinflammatory cytokine exposed bank vole islets. iNOS and NO–independent pathways have been suggested previously to be based on experiments with human islets [83], rat–derived –cell preparations [139, 140] and pancreatic islets from mice with an inactivated iNOS gene [141]. In experiments performed with the latter type of islets it has been shown that mRNA expression for the transcription factor PDX–1 was also reduced by cytokines [142]. Indeed, this was what we saw in bank vole islets after expo- sure to IL–1 + TNF– + IFN– though the effect was more pronounced on the insulin gene expression where a 50 % reduction could be seen.

Prolonged exposure to high glucose (III) Isolated bank vole islets displayed a markedly elevated basal insulin secre- tion and reduced insulin content compared to control islets (11.1 mM glu- cose) after prolonged exposure to high glucose conditions in vitro (28 mM glucose). However, only a minor effect was seen on glucose stimulated insu- lin release compared to controls. The reduced insulin content in islets cul- tured at high glucose suggests that the islets were not able to adjust to the increased functional demand. Bank vole islets maintained at low glucose condition (5.6 mM glucose) showed a reduced basal insulin secretion and glucose stimulated insulin release while the islet insulin content was essen- tially unaffected compared to control islets (11.1 mM glucose). The stimula- tion index, the fraction between the insulin release upon a glucose challenge and the basal insulin secretion, reveal that the islets maintained at high glu- cose conditions were less responsive compared to controls suggesting that their functional ability was exceeded. In contrast to the bank vole islets maintained at low glucose conditions displayed increased stimulation index compared to controls. The latter finding suggests that the control islets are

28

not cultured at an optimal glucose concentration. The increased response seen at 5.6 mM glucose suggests that this glucose concentration, using the RPMI 1640 culture medium, may be the most favorable culture conditions for bank vole islets. It has previously been shown that human islets cultured in RPMI 1640 have an optimal glucose concentration lower than the stan- dard 11.1 mM glucose [143]. In similarity to our experiments in bank vole human islets also display a similar response in stimulation index and in insu- lin content with increasing glucose concentration in the culture medium [143]. The selection of culture media appears to be of importance, at least when studying isolated rat islets in vitro. Normal rat islets cultured at high glucose has in some studies shown to express a decrease in glucose stimu- lated insulin secretion so–called desensitization [100]. However, in other studies high glucose culture has been shown to have no obvious harmful effect on the islets [97]. Islets isolated from female bank voles and maintained at low glucose condition had reduced (pro)insulin biosynthesis and reduced insulin gene expression compared to control islets. In islets isolated from male donors no difference was detected in the (pro)insulin biosynthesis rate or the insulin gene expression in response to the different glucose concentrations used in the culture media. These results may suggest that female islets are more suited, than male islets, to adapt their (pro)insulin biosynthesis rates and insulin gene expression to the increasing functional demand. The insulin accumulation in medium was not changed after islet culture in high glucose compared to the control group. However, the proinsulin frac- tion in this medium, expressed as percent of total medium insulin accumula- tion, was increased. The insulin and proinsulin concentration in medium with a low glucose concentration collected after islet culture displayed a decline compared to control islets. The increased proinsulin amount in the culture medium with high glucose concentration may suggest that the islet insulin production capacity was exceeded. The Israeli sand rat shares some of these features. Diabetic sand rats kept under non–fasting conditions have markedly increased proinsulin levels in their serum [144]. Also, pancreatic extracts from these animals revealed that they had depleted insulin stores. These findings indicate that the functional demand on the –cell in vivo has surpassed the capacity to produce insulin. In a study where islet cells from normal Psammomys obesus were explanted in vitro and cultured as mono- layers it was found that an elevated glucose concentration (33.3 mM) for 10 days caused a pronounced reduction in glucose induced insulin secretion and depletion of islet insulin stores as well as increased proportion of proinsulin related peptides [96].

29

Islet morphology and function (IV) Bank vole pancreatic islets develop a markedly changed islet structure con- taining balloon-like cells and severe reduction of the number of endocrine cells. The changed structure of these islets resembles so–called hydropic degeneration [123]. Hydropic degeneration was observed to be present only in islets from bank voles classified as glucose intolerant/diabetic following IPGTT. The severely changed islet structure seen in islets with hydropic degenera- tion did not affect the relative islet area of the total pancreas area. However, the loss of endocrine cells resulted in a reduced insulin positive area com- pared to total islet area in these islets. In pancreas from female donors, islets with hydropic degeneration also had reduced glucagon and somatostatin positive areas. This difference was not apparent in islets from male donors. Microtus arvalis is another vole that as a response to feeding on a low fi- ber diet develops similar islet structural changes as presently could be found in the severely affected bank voles [120]. The vacuoles observed in these animals were described as enlarged cells with glycogen deposits. Another animal model showing deposits of glycogen in the islets when cultured at high glucose conditions in vitro is the Wistar rat [121]. This may suggest that the vacuoles seen in islets from severely affected bank voles could re- flect glycogen deposition. Similar to the severely affected bank voles the glucose intolerant sand rat develops markedly changed islet structure [118]. In the sand rat this altered islet structure is a result of prolonged disease progression [116]. This altered islet structure has been presented as a result of extensive cell death that leads to the formation of vacuoles in the cells [102]. Based on this change seen in severely diabetic sand rats it is reasonable to assume that the altered islet structure seen in severely affected bank voles may be due to increased cell death. However, previous studies have shown that islets exposed to pro- longed period of elevated glucose levels in vitro does not reveal any change in islet viability nor could increased cell death be observed in islets exposed to proinflammatory cytokines despite a suppressed function. The observed extensive morphological islet changes seen in diabetic bank voles has made us suspect that a subpopulation of islets has been studied throughout this project namely those that we were able to isolate upon the collagenase isolation procedure. It may well be that the islets with the most deteriorated structure, could not be retrieved upon islet isolation. We found that the islet yield during isolation from a glucose tolerant/normal animal is approximately 90 islets per pancreas, while in severely diabetic animals it usually decreases to 60 islets or less. The serum insulin level was elevated in female bank voles with islets showing hydropic degeneration. However, this increase was not as clear in male bank voles. But when both female and male serum insulin were plotted

30

the bank vole serum insulin could be predicted from the non–fasting blood glucose, using the Galton log–normal regression [132] (P<0.018). It is possi- ble that with increasing functional demand the bank vole –cell an initial period of hyperinsulinemia will occur but eventually it will lead to loss of insulin production and hypoinsulinemia. Although the islet structure was severely changed and the number of en- docrine cells reduced morphologic screening of pancreatic sections from bank voles of various age have not revealed any clear signs of insulitis, which argues against a classical human type 1 diabetes pathogenesis. In hu- man type 2 diabetic patients, the loss of cell mass does not affect the frac- tional islet area which is consistent with our finding in the bank vole [112, 113]. Bank vole has been considered as a diabetes model showing features of both type 1 and type 2 diabetes. However, our present findings suggest that the bank voles bred in the laboratory for a prolonged period is more likely a diabetic animal model showing mainly features of type 2 diabetes. In this study of glucose intolerant/diabetic animals we have observed elevated se- rum insulin levels, increased body weight, reduced cell mass, markedly changed islet structure and the absence of insulitis, all signs of type 2 di- abetes mellitus. These observations are made on a 30 years old breeding colony and we cannot exclude that bank voles caught in nature instead rather develop a type 1 form of the disease. This argues for a decisive role for inte- ractions between environmental factors in the diabetes development in bank voles.

Ljungan virus (I–IV) The role of the LV in the bank vole pathology is still unclear. Whether or not the LV infection has contributed to the development of glucose intoler- ance/diabetes in the bank vole still remains to be revealed. Therefore, we cannot exclude that the LV may be the agent responsible for the develop- ment of diabetes in bank voles. Furthermore, such infection may have influ- enced our findings when cytokines were added to the isolated pancreatic islets or when the islets were exposed to an increased functional demand.

31

Conclusions

Paper I We report that bank voles to a significant extent develop glucose intoler- ance/diabetes in the laboratory, a condition which was accompanied by hyperinsulinemia. When islet functions were studied in vitro, signs of an increased functional demand on the -cells were seen, and that there might be a gender difference in that the -cells of males are more affected than the islets of the females. Paper II The pancreatic islets isolated from glucose tolerant bank voles are affected by prolonged exposure to proinflammatory cytokines. This effect seems to be essentially NO-independent, and a key cellular event appears to be a de- creased level of insulin mRNA leading to a decline in insulin translation that result in a depletion of insulin stores in the -cells and a lowered insulin response to a glucose challenge. Paper III We found that islets isolated from female and male bank voles display signs of dysfunction after high glucose culture in vitro, although this did not cause direct toxicity and/or cell death. A gender difference was observed suggest- ing that the islets of the females may more readily adapt to the elevated glu- cose concentration than islets of male bank voles. Paper IV The bank vole has been considered as a diabetes model showing features of both type 1 and type 2 diabetes. In this study we report that islets in severely affected animals have markedly changed structure, reduced cell mass and hyperinsulinemia, all of which are characteristics of a type 2 diabetes. These findings suggest that bank voles bred in the laboratory may develop similar features to type 2 diabetes. However, it cannot be excluded that bank voles caught in nature instead rather develop a type 1 form of the disease. This argues for a decisive role for interactions between environmental factors in the diabetes development in bank voles.

32

Acknowledgements

This work was carried out at the department of Medical Cell Biology, Upp- sala University, Uppsala, Sweden and the Astrid Fagreus Laboratory, Karo- linska Institute, Stockholm. I would like to express my sincere thankfulness to all people who guided, supported and inspired me on the path of research.

Jag vill dessutom framföra mitt vördnadsfulla och hjärtliga tack till:

Min huvudhandledare Stellan Sandler för att han delat med sig av sin enor- ma kunskap inom diabetes och för hans oändliga tålamod. Du har introduce- rat mig, stöttat mig och guidat mig på forskningens oändliga vidder.

Mina handledare Bo Niklasson för din brinnande entusiasm som för Ljung- anvirus projektet framåt och Carina Carlsson för att du delat med dig av din expertis inom molekylär biologi, och er båda för inspirerande vetenskapliga diskussioner och värdefull hjälp.

Nuvarande och tidigare prefekter för institutionen Erik Gylfe och Arne An- dersson för er professionella hängivenhet och ert engagemang för institutio- nens välbefinnande och skapandet av en god forskningsatmosfär.

Professorer och seniora forskare för ert kunnande, ert bidrag till en veten- skapligt stimulerande utvecklingsmiljö och alla konstruktiva kommentarer under ”Brainpubs”.

Ing-Britt Hallgren för ovärderlig hjälp och idogt kämpande med öplockning och öfunktionstest, Astrid Nordin och Eva Törnelius för er expertis då labo- ratoriefrågorna blivit för svåra, Ing-Marie Mörsare, Lisbeth Sagulin, Göran Ståhl, Birgitta Bodin för hjälp med både det ena och det andra, Agneta Sand- ler Bäfwe, Marianne Ljungkvist and Lina Thorvaldson för ovärderlig hjälp med många utmanande administrativa problem. Stort tack till er alla för liv- liga diskussioner, livsexpertis och mycket gott sällskap.

Alla tidigare och nuvarande studenter och doktorander, på institutionen för gott kamratskap och roliga upptåg.

33

Tidigare och nuvarande rumskamrater för språklig rådgivning, förslag på intressanta experiment, alternativ tolkning av resultat, kulturell fortbildning, och anvisningar om var fokus för verksamheten bör ligga – må Champagnen flöda… ”and this will give You world domination!” I’m sure .

Det stora antalet sorkar som offrat sig för vetenskapen.

Alla vänner, för att ni fyller mitt liv med glädje och äventyr.

Min svärfamilj för att ni välkomnat mig in i eran gemenskap och för att ni stöttar oss då det behövs.

Mina föräldrar för att ni hjälpt mig till att bli den jag är idag och för att ni är underbara farföräldrar för Oscar och Isabella. Maria, min allra bästa lillasyster och Torbjörn, min allra bästa storebror .

Min egna Skatt – Eva, Oscar och Isabella Tack för allt stöd och omtanke som gjort denna avhandling möjlig.

34

References

1. Bogardus, C., Lillioja, S., Mott, D., Reaven, G.R., Kashiwagi, A. and Foley, J.E., Relationship between obesity and maximal insulin- stimulated glucose uptake in vivo and in vitro in man. International Journal of Obesity, 1985. 9 Suppl 1: p. 149.

2. Dahlquist, G., Non-genetic risk determinants of type 1 diabetes. Di- abetes and Metabolism, 1994. 20(3): p. 251-257.

3. Hyöty, H. and Taylor, K.W., The role of viruses in human diabetes. Diabetologia, 2002. 45(10): p. 1353-1361.

4. Knip, M. and Åkerblom, H.K., Environmental factors in the pathoge- nesis of type 1 diabetes mellitus. Experimental and Clinical Endocri- nology & Diabetes 1999. 107 Suppl 3: p. S93-100.

5. Ludvigsson, J., Why diabetes incidence increases--a unifying theory. Annals of the New York Academy of Sciences, 2006. 1079: p. 374- 82.

6. Pociot, F., Rønningen, K.S., Bergholdt, R., Lorenzen, T., Johannesen, J., Ye, K., Dinarello, C.A. and Nerup, J., Genetic susceptibility markers in Danish patients with type 1 (insulin-dependent) diabetes- -evidence for polygenicity in man. Danish Study Group of Diabetes in Childhood. Autoimmunity, 1994. 19(3): p. 169-178.

7. Virtanen, S.M. and Knip, M., Nutritional risk predictors of  cell au- toimmunity and type 1 diabetes at a young age. The American Jour- nal of Clinical Nutrition, 2003. 78(6): p. 1053-1067.

8. International Diabetes Federation, www.eatlas.idf.org/Prevalence/All_diabetes. Diabetes Atlas, 2010.

9. Report of the expert committee on the diagnosis and classification of diabetes mellitus, Classification of diabetes mellitus and other cate- gories of glucose regulation Diabetes Care, 2003. 26 p. 5-20.

10. Eisenbarth, G.S., Connelly, J. and Soeldner, J.S., The "natural" history of type 1 diabetes. Diabetes/Metabolism Reviews, 1987. 3(4): p. 873-891.

35

11. Turner, R.C., Matthews, D.R., Clark, A., O'Rahilly, S., Rudenski, A.S. and Levy, J., Pathogenesis of NIDDM--a disease of deficient insulin secretion. Baillière's Clinical Endocrinology and Metabolism, 1988. 2(2): p. 327-342.

12. Papaspyrus, N.S., The history of diabetes mellitus. 1964, Stuttgart: Georg Thieme Verlag

13. Ahlström, H., Strand, P.O., Ahlström, G. and Nilsson, G., Läkarlexi- kon. 1983, Malmö: Bra Böckers förlag

14. Cullen, W., Synopsis nosologiae methodicae. 1769, Edingburgh.

15. Lanerhans, P., Beitrage zur mikroskopischen Anatomie de bauschspei- cherdrusse. 1869: Berlin.

16. Banting, F.G., Best, C.H., Campbell, W.R. and Fletcher, A.A., Pan- creatic extracts in the treatment of diabetes mellitus. Canadian Med- ical Association Journal, 1922. 12: p. 141-146.

17. Shampo, M.A. and Kyle, R.A., Frederick Sanger--winner of 2 Nobel prizes. Mayo Clinic Proceedings, 2002. 77(3): p. 212.

18. Meites, J., The 1977 Nobel Prize in Physiology or Medicine. Science, 1977. 198(4317): p. 594-596.

19. Hellerström, C., A Method for the Microdissection of Intact Pancreatic Islets of . Acta Endocrinologica, 1964. 45: p. 122-32.

20. Lacy, P.E. and Kostianovsky, M., Method for the isolation of intact islets of Langerhans from the rat pancreas. Diabetes, 1967. 16(1): p. 35-9.

21. Moskalewski, S., Isolation and Culture of the Islets of Langerhans of the Guinea Pig. General and Comparative Endocrinology, 1965. 44: p. 342-53.

22. Steiner, D.F. and Oyer, P.E., The Biosynthesis of Insulin and a Proba- ble Precursor of Insulin by a Human Islet Cell Adenoma. Proceed- ings of the National Academy of Sciences of the United States of America, 1967. 57(2): p. 473-480.

23. Bliss, M., The discovery of insulin. 3rd pbk. ed. 2000, Toronto: Uni- versity of Toronto Press. 304 p., [16] p. of plates.

36

24. Junod, A., Lambert, A.E., Stauffacher, W. and Renold, A.E., Diabeto- genic action of streptozotocin: relationship of dose to metabolic re- sponse. Journal of Clinical Investigation, 1969. 48(11): p. 2129-39.

25. Lenzen, S. and Panten, U., Alloxan: history and mechanism of action. Diabetologia, 1988. 31(6): p. 337-42.

26. Holstad, M. and Sandler, S., A transcriptional inhibitor of TNF- pre- vents diabetes induced by multiple low-dose streptozotocin injec- tions in mice. Journal of Autoimmunity, 2001. 16(4): p. 441-7.

27. Atkinson, M.A. and Leiter, E.H., The NOD mouse model of type 1 diabetes: as good as it gets? Nature Medicine, 1999. 5(6): p. 601- 604.

28. Yoon, J.W., Jun, H.S. and Santamaria, P., Cellular and molecular mechanisms for the initiation and progression of  cell destruction resulting from the collaboration between macrophages and T cells. Autoimmunity, 1998. 27(2): p. 109-22.

29. Dean, B.M., Bone, A.J., Varey, A.M., Walker, R., Baird, J.D. and Cooke, A., Insulin autoantibodies, islet cell surface antibodies and the development of spontaneous diabetes in the BB/Edinburgh rat. Clinical and Experimental Immunology, 1987. 69(2): p. 308-13.

30. Yoon, J.W., Yoon, C.S., Lim, H.W., Huang, Q.Q., Kang, Y., Pyun, K.H., Hirasawa, K., Sherwin, R.S. and Jun, H.S., Control of au- toimmune diabetes in NOD mice by GAD expression or suppression in  cells. Science, 1999. 284(5417): p. 1183-7.

31. Makino, S., Kunimoto, K., Muraoka, Y., Mizushima, Y., Katagiri, K. and Tochino, Y., Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu, 1980. 29(1): p. 1-13.

32. Nakhooda, A.F., Like, A.A., Chappel, C.I., Murray, F.T. and Marliss, E.B., The spontaneously diabetic Wistar rat. Metabolic and morpho- logic studies. Diabetes, 1977. 26(2): p. 100-12.

33. Chagnon, Y.C. and Bouchard, C., Genetics of obesity: advances from rodent studies. Trends in Genetics 1996. 12(11): p. 441-4.

34. Chen, H., Charlat, O., Tartaglia, L.A., Woolf, E.A., Weng, X., Ellis, S.J., Lakey, N.D., Culpepper, J., Moore, K.J., Breitbart, R.E., Duyk, G.M., Tepper, R.I. and Morgenstern, J.P., Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell, 1996. 84(3): p. 491-5.

37

35. Considine, R.V., Sinha, M.K., Heiman, M.L., Kriauciunas, A., Ste- phens, T.W., Nyce, M.R., Ohannesian, J.P., Marco, C.C., McKee, L.J., Bauer, T.L. and et al., Serum immunoreactive-leptin concentra- tions in normal-weight and obese humans. The New England Jour- nal of Medicine, 1996. 334(5): p. 292-5.

36. Truett, G.E., Bahary, N., Friedman, J.M. and Leibel, R.L., Rat obesity gene fatty (fa) maps to chromosome 5: evidence for homology with the mouse gene diabetes (db). Proceedings of the National Academy of Sciences of the United States of America, 1991. 88(17): p. 7806- 9.

37. Goto, Y., Kakizaki, M. and Masaki, N., Production of spontaneous diabetic rats by repetition of selective breeding. The Tohoku Journal of Experimental Medicine, 1976. 119(1): p. 85-90.

38. Nakamura, M. and Yamada, K., Studies on a diabetic (KK) strain of the mouse. Diabetologia, 1967. 3(2): p. 212-21.

39. Ueda, H., Ikegami, H., Yamato, E., Fu, J., Fukuda, M., Shen, G., Ka- waguchi, Y., Takekawa, K., Fujioka, Y., Fujisawa, T. and et al., The NSY mouse: a new animal model of spontaneous NIDDM with mod- erate obesity. Diabetologia, 1995. 38(5): p. 503-8.

40. Heled, Y., Shapiro, Y., Shani, Y., Moran, D.S., Langzam, L., Braiman, L., Sampson, S.R. and Meyerovitch, J., Physical exercise prevents the development of type 2 diabetes mellitus in Psammomys obesus. American Journal of Physiology. Endocrinology and Metabolism, 2002. 282(2): p. E370-5.

41. Ziv, E., Shafrir, E., Kalman, R., Galer, S. and Bar-On, H., Changing pattern of prevalence of insulin resistance in Psammomys obesus, a model of nutritionally induced type 2 diabetes. Metabolism, 1999. 48(12): p. 1549-54.

42. Marquie, G., Hadjiisky, P., Arnaud, O. and Duhault, J., Development of macroangiopathy in sand rats (Psammomys obesus), an animal model of non-insulin-dependent diabetes mellitus: effect of glicla- zide. The American Journal of Medicine, 1991. 90(6A): p. 55S-61S.

43. Cerasi, E., Kaiser, N. and Gross, D.J., From sand rats to diabetic pa- tients: is non-insulin-dependent diabetes mellitus a disease of the  cell? Diabetes & Metabolism, 1997. 23 Suppl 2: p. 47-51.

44. Myers, J.H. and Krebs, C.J., Population Cycles in Rodents. Scientific American, 1974. 230(6): p. 38-46.

38

45. Burnie, D. and Wilson, D.E., Animal: The Definitive Visual Guide to the World's Wildlife. 2001, London Dorling Kinersley Publishing.

46. Macdonnald, D.W., The New Encyclopedia of Mammals. 2001, Ox- ford Oxford University Press.

47. Hornfeldt, B., Synchronous Population Fluctuations in Voles, Small Game, Owls, and Tularemia in Northern Sweden. Oecologia, 1978. 32(2): p. 141-152.

48. Niklasson, B., Hörnfeldt, B. and Lundman, B., Could myocarditis, insulin-dependent diabetes mellitus, and Guillain-Barre syndrome be caused by one or more infectious agents carried by rodents? Emerging Infectious Diseases, 1998. 4(2): p. 187-193.

49. Schønecker, B., Heller, K.E. and Freimanis, T., Development of ste- reotypies and polydipsia in wild caught bank voles (Clethrionomys glareolus) and their laboratory-bred offspring. Is polydipsia a symp- tom of diabetes mellitus? Applied Animal Behaviour Science, 2000. 68(4): p. 349-357.

50. Niklasson, B., Hörnfeldt, B., Nyholm, E., Niedrig, M., Donoso- Mantke, O., Gelderblom, H.R. and Lernmark, A., Type 1 diabetes in Swedish bank voles (Clethrionomys glareolus): signs of disease in both colonized and wild cyclic populations at peak density. Annals of the New York Academy of Sciences, 2003. 1005: p. 170-175.

51. Baekkeskov, S., Aanstoot, H.J., Christgau, S., Reetz, A., Solimena, M., Cascalho, M., Folli, F., Richter-Olesen, H., De Camilli, P. and Camilli, P.D., Identification of the 64K autoantigen in insulin- dependent diabetes as the GABA-synthesizing enzyme glutamic acid decarboxylase. Nature, 1990. 347(6289): p. 151-156.

52. Palmer, J.P., Asplin, C.M., Clemons, P., Lyen, K., Tatpati, O., Raghu, P.K. and Paquette, T.L., Insulin antibodies in insulin-dependent di- abetics before insulin treatment. Science, 1983. 222(4630): p. 1337- 1339.

53. Payton, M.A., Hawkes, C.J. and Christie, M.R., Relationship of the 37,000- and 40,000-M(r) tryptic fragments of islet antigens in insu- lin-dependent diabetes to the protein tyrosine phosphatase-like mo- lecule IA-2 (ICA512). Journal of Clinical Investigation, 1995. 96(3): p. 1506-1511.

54. Niklasson, B., Kinnunen, L., Hörnfeldt, B., Hörling, J., Benemar, C., Hedlund, K.O., Matskova, L., Hyypia, T. and Winberg, G., A new

39

picornavirus isolated from bank voles (Clethrionomys glareolus). Virology, 1999. 255(1): p. 86-93.

55. Wesslen, L., Påhlson, C., Friman, G., Fohlman, J., Lindquist, O. and Johansson, C., Myocarditis caused by Chlamydia pneumoniae (TWAR) and sudden unexpected death in a Swedish elite orienteer. Lancet, 1992. 340(8816): p. 427-428.

56. Niklasson, B., Hultman, T., Kallies, R., Niedrig, M., Nilsson, R., Berggren, P.O., Juntti-Berggren, L., Efendic, S., Lernmark, A. and Klitz, W., The BioBreeding rat diabetes model is infected with Ljun- gan virus. Diabetologia, 2007. 50(7): p. 1559-1560.

57. Niklasson, B., Samsioe, A., Blixt, M., Sandler, S., Sjöholm, A., La- gerquist, E., Lernmark, A. and Klitz, W., Prenatal viral exposure followed by adult stress produces glucose intolerance in a mouse model. Diabetologia, 2006. 49(9): p. 2192-2199.

58. Donoso Mantke, O., Kallies, R., Niklasson, B., Nitsche, A. and Nie- drig, M., A new quantitative real-time reverse transcriptase PCR as- say and melting curve analysis for detection and genotyping of Ljungan virus strains. Journal of Virological Methods, 2007. 141(1): p. 71-7.

59. Karlsson Faresjö, M.G., Ernerudh, J. and Ludvigsson, J., Cytokine profile in children during the first 3 months after the diagnosis of type 1 diabetes. Scandinavian Journal of Immunology, 2004. 59(5): p. 517-26.

60. Rabinovitch, A. and Suarez-Pinzon, W.L., Cytokines and their roles in pancreatic islet -cell destruction and insulin-dependent diabetes mellitus. Biochemical Pharmacology, 1998. 55(8): p. 1139-1149.

61. Sandler, S., Eizirik, D.L., Svensson, C., Strandell, E., Welsh, M. and Welsh, N., Biochemical and molecular actions of interleukin-1 on pancreatic -cells. Autoimmunity, 1991. 10(3): p. 241-253.

62. Boni-Schnetzler, M., Ehses, J.A., Faulenbach, M. and Donath, M.Y., Insulitis in type 2 diabetes. Diabetes, Obesity & Metabolism, 2008. 10 Suppl 4: p. 201-4.

63. Donath, M.Y., Størling, J., Maedler, K. and Mandrup-Poulsen, T., Inflammatory mediators and islet -cell failure: a link between type 1 and type 2 diabetes. Journal of Molecular Medicine, 2003. 81(8): p. 455-470.

40

64. Kolb, H. and Mandrup-Poulsen, T., An immune origin of type 2 di- abetes? Diabetologia, 2005. 48(6): p. 1038-1050.

65. Larsen, C.M., Faulenbach, M., Vaag, A., Volund, A., Ehses, J.A., Seifert, B., Mandrup-Poulsen, T. and Donath, M.Y., Interleukin-1- receptor antagonist in type 2 diabetes mellitus. The New England Journal of Medicine, 2007. 356(15): p. 1517-1526.

66. Xie, Q. and Nathan, C., The high-output nitric oxide pathway: role and regulation. Journal of Leukocyte Biology, 1994. 56(5): p. 576- 582.

67. Mandrup-Poulsen, T., The role of interleukin-1 in the pathogenesis of IDDM. Diabetologia, 1996. 39(9): p. 1005-1029.

68. Eizirik, D.L., Cagliero, E., Bjorklund, A. and Welsh, N., Interleukin- 1 induces the expression of an isoform of nitric oxide synthase in insulin-producing cells, which is similar to that observed in acti- vated macrophages. Federation of European Biochemical Societies Letters, 1992. 308(3): p. 249-252.

69. Southern, C., Schulster, D. and Green, I.C., Inhibition of insulin secre- tion by interleukin-1  and tumour necrosis factor- via an L- arginine-dependent nitric oxide generating mechanism. Federation of European Biochemical Societies Letters, 1990. 276(1-2): p. 42- 44.

70. Welsh, N., Eizirik, D.L., Bendtzen, K. and Sandler, S., Interleukin-1 -induced nitric oxide production in isolated rat pancreatic islets requires gene transcription and may lead to inhibition of the Krebs cycle enzyme aconitase. Endocrinology, 1991. 129(6): p. 3167-3173.

71. Griffith, O.W. and Stuehr, D.J., Nitric oxide synthases: properties and catalytic mechanism. Annual Review of Physiology, 1995. 57: p. 707-736.

72. Kleemann, R., Rothe, H., Kolb-Bachofen, V., Xie, Q.W., Nathan, C., Martin, S. and Kolb, H., Transcription and translation of inducible nitric oxide synthase in the pancreas of prediabetic BB rats. Federa- tion of European Biochemical Societies Letters, 1993. 328(1-2): p. 9-12.

73. Kolb, H. and Kolb-Bachofen, V., Nitric oxide: a pathogenetic factor in autoimmunity. Immunology Today, 1992. 13(5): p. 157-160.

74. Rabinovitch, A., Suarez-Pinzon, W.L., Sorensen, O. and Bleackley, R.C., Inducible nitric oxide synthase (iNOS) in pancreatic islets of

41

nonobese diabetic mice: identification of. Endocrinology, 1996. 137(5): p. 2093-2099.

75. Rothe, H., Faust, A., Schade, U., Kleemann, R., Bosse, G., Hibino, T., Martin, S. and Kolb, H., Cyclophosphamide treatment of female non-obese diabetic mice causes enhanced expression of inducible ni- tric oxide synthase and interferon-gamma, but not of interleukin-4. Diabetologia, 1994. 37(11): p. 1154-1158.

76. Bergmann, L., Kroncke, K.D., Suschek, C., Kolb, H. and Kolb- Bachofern, V., Cytotoxic action of IL-1 against pancreatic islets is mediated via nitric oxide formation and is inhibited by NG- monomethyl-L-arginine. Federation of European Biochemical Socie- ties Letters, 1992. 299(1): p. 103-106.

77. Corbett, J.A., Mikhael, A., Shimizu, J., Frederick, K., Misko, T.P., McDaniel, M.L., Kanagawa, O. and Unanue, E.R., Nitric oxide pro- duction in islets from nonobese diabetic mice: aminoguanidine- sensitive and -resistant stages in the immunological diabetic process. Proceedings of the National Academy of Sciences of the United States of America, 1993. 90(19): p. 8992-5.

78. Holstad, M. and Sandler, S., Aminoguanidine, an inhibitor of nitric oxide formation, fails to protect against insulitis and hyperglycemia induced by multiple low dose streptozotocin injections in mice. Au- toimmunity, 1993. 15(4): p. 311-314.

79. Sternesjö, J., Welsh, N. and Sandler, S., S-methyl-L-thiocitrulline counteracts interleukin 1  induced suppression of pancreatic islet function in vitro, but does not protect against multiple low-dose streptozotocin-induced diabetes in vivo. Cytokine, 1997. 9(5): p. 352-359.

80. Cetkovic-Cvrlje, M. and Eizirik, D.L., TNF- and IFN-y potentiate the deleterious effects of IL-1 on mouse pancreatic islets mainly via generation of nitric oxide. Cytokine, 1994. 6(4): p. 399-406.

81. Welsh, N. and Sandler, S., Interleukin-1 induces nitric oxide produc- tion and inhibits the activity of aconitase without decreasing glucose oxidation rates in isolated mouse pancreatic islets. Biochemical and Biophysical Research Communications, 1992. 182(1): p. 333-340.

82. Campbell, I.L., Iscaro, A. and Harrison, L.C., IFN- and tumor necro- sis factor-. Cytotoxicity to murine islets of Langerhans. The Jour- nal of Immunology, 1988. 141(7): p. 2325-2329.

42

83. Eizirik, D.L., Sandler, S., Welsh, N., Cetkovic-Cvrlje, M., Nieman, A., Geller, D.A., Pipeleers, D.G., Bendtzen, K. and Hellerström, C., Cy- tokines suppress human islet function irrespective of their effects on nitric oxide generation. The Journal of Clinical Investigation, 1994. 93(5): p. 1968-1974.

84. Flodström, M. and Eizirik, D.L., Interferon--induced interferon regu- latory factor-1 (IRF-1) expression in rodent and human islet cells precedes nitric oxide production. Endocrinology, 1997. 138(7): p. 2747-2753.

85. Heitmeier, M.R., Scarim, A.L. and Corbett, J.A., Prolonged STAT1 activation is associated with interferon- priming for interleukin-1- induced inducible nitric-oxide synthase expression by islets of Lan- gerhans. The Journal of Biological Chemistry, 1999. 274(41): p. 29266-29273.

86. Pavlovic, D., Chen, M.C., Bouwens, L., Eizirik, D.L. and Pipeleers, D., Contribution of ductal cells to cytokine responses by human pancreatic islets. Diabetes, 1999. 48(1): p. 29-33.

87. Kawahara, D.J. and Kenney, J.S., Species differences in human and rat islet sensitivity to human cytokines. Monoclonal anti-interleukin- 1 (IL-1) influences on direct and indirect IL-1-mediated islet effects. Cytokine, 1991. 3(2): p. 117-124.

88. Sandler, S., Andersson, A. and Hellerström, C., Inhibitory effects of interleukin 1 on insulin secretion, insulin biosynthesis, and oxidative metabolism of isolated rat pancreatic islets. Endocrinology, 1987. 121(4): p. 1424-1431.

89. Darville, M.I. and Eizirik, D.L., Regulation by cytokines of the induci- ble nitric oxide synthase promoter in insulin-producing cells. Diabe- tologia, 1998. 41(9): p. 1101-1108.

90. Stolar, M., Glycemic control and complications in type 2 diabetes mellitus. The American Journal of Medicine, 2010. 123(3 Suppl): p. S3-11.

91. Rossetti, L., Giaccari, A. and DeFronzo, R.A., Glucose toxicity. Di- abetes Care, 1990. 13(6): p. 610-30.

92. Weir, G.C., Leahy, J.L. and Bonner-Weir, S., Experimental reduction of -cell mass: implications for the pathogenesis of diabetes. Di- abetes/Metabolism Reviews, 1986. 2(1-2): p. 125-161.

43

93. Eizirik, D.L., Korbutt, G.S. and Hellerström, C., Prolonged exposure of human pancreatic islets to high glucose concentrations in vitro impairs the -cell function. The Journal of Clinical Investigation, 1992. 90(4): p. 1263-8.

94. Eizirik, D.L., Strandell, E. and Sandler, S., Culture of mouse pancrea- tic islets in different glucose concentrations modifies cell sensitivi- ty to streptozotocin. Diabetologia, 1988. 31(3): p. 168-174.

95. Eizirik, D.L., Strandell, E. and Sandler, S., Prolonged exposure of pancreatic islets isolated from "pre-diabetic" non-obese diabetic mice to a high glucose concentration does not impair Beta-cell func- tion. Diabetologia, 1991. 34(1): p. 6-11.

96. Gross, D.J., Leibowitz, G., Cerasi, E. and Kaiser, N., Increased sus- ceptibility of islets from diabetes-prone Psammomys obesus to the deleterious effects of chronic glucose exposure. Endocrinology, 1996. 137(12): p. 5610-5615.

97. Sandler, S., Bendtzen, K., Borg, L.A., Eizirik, D.L., Strandell, E. and Welsh, N., Studies on the mechanisms causing inhibition of insulin secretion in rat pancreatic islets exposed to human interleukin-1  indicate a perturbation in the mitochondrial function. Endocrinolo- gy, 1989. 124(3): p. 1492-1501.

98. Svensson, C., Sandler, S. and Hellerström, C., Lack of long-term -cell glucotoxicity in vitro in pancreatic islets isolated from two mouse strains (C57BL/6J; C57BL/KsJ) with different sensitivities of the - cells to hyperglycaemia in vivo. The Journal of Endocrinology, 1993. 136(2): p. 289-296.

99. Tsuboi, T., Ravier, M.A., Parton, L.E. and Rutter, G.A., Sustained exposure to high glucose concentrations modifies glucose signaling and the mechanics of secretory vesicle fusion in primary rat pan- creatic -cells. Diabetes, 2006. 55(4): p. 1057-65.

100. Bolaffi, J.L., Bruno, L., Heldt, A. and Grodsky, G.M., Characteristics of desensitization of insulin secretion in fully in vitro systems. Endo- crinology, 1988. 122(5): p. 1801-1809.

101. Leibowitz, G., Yuli, M., Donath, M.Y., Nesher, R., Melloul, D., Cera- si, E., Gross, D.J. and Kaiser, N., -cell glucotoxicity in the Psam- momys obesus model of type 2 diabetes. Diabetes, 2001. 50 Suppl 1: p. S113-7.

44

102. Donath, M.Y., Gross, D.J., Cerasi, E. and Kaiser, N., Hyperglycemia- induced -cell apoptosis in pancreatic islets of Psammomys obesus during development of diabetes. Diabetes, 1999. 48(4): p. 738-44.

103. Eisenbarth, G.S., Type 1 diabetes mellitus. A chronic autoimmune disease. The New England Journal of Medicine, 1986. 314(21): p. 1360-1368.

104. Maclean, N. and Ogilvie, R.F., Quantitative estimation of the pancrea- tic islet tissue in diabetic subjects. Diabetes, 1955. 4(5): p. 367-76.

105. Foulis, A.K., Liddle, C.N., Farquharson, M.A., Richmond, J.A. and Weir, R.S., 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.

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

107. Bach, J.F., Autoimmunity and type I diabetes. Trends in Endocrinology and Metabolism, 1997. 8(2): p. 71-4.

108. Eizirik, D.L., Colli, M.L. and Ortis, F., The role of inflammation in insulitis and -cell loss in type 1 diabetes. Nature Reviews. Endo- crinology, 2009. 5(4): p. 219-26.

109. Itoh, N., Hanafusa, T., Miyazaki, A., Miyagawa, J., Yamagata, K., Yamamoto, K., Waguri, M., Imagawa, A., Tamura, S., Inada, M. and et al., Mononuclear cell infiltration and its relation to the ex- pression of major histocompatibility complex antigens and adhesion molecules in pancreas biopsy specimens from newly diagnosed insu- lin-dependent diabetes mellitus patients. The Journal of Clinical In- vestigation, 1993. 92(5): p. 2313-22.

110. Guiot, Y., Sempoux, C., Moulin, P. and Rahier, J., No decrease of the -cell mass in type 2 diabetic patients. Diabetes, 2001. 50 Suppl 1: p. S188.

111. Sempoux, C., Guiot, Y., Dubois, D., Moulin, P. and Rahier, J., Human type 2 diabetes: morphological evidence for abnormal -cell func- tion. Diabetes, 2001. 50 Suppl 1: p. S172-7.

112. Butler, A.E., Janson, J., Bonner-Weir, S., Ritzel, R., Rizza, R.A. and Butler, P.C., -cell deficit and increased -cell apoptosis in humans with type 2 diabetes. Diabetes, 2003. 52(1): p. 102-10.

45

113. Yoon, K.H., Ko, S.H., Cho, J.H., Lee, J.M., Ahn, Y.B., Song, K.H., Yoo, S.J., Kang, M.I., Cha, B.Y., Lee, K.W., Son, H.Y., Kang, S.K., Kim, H.S., Lee, I.K. and Bonner-Weir, S., Selective -cell loss and -cell expansion in patients with type 2 diabetes mellitus in Korea. The Journal of Clinical Endocrinology and Metabolism, 2003. 88(5): p. 2300-8.

114. Donath, M.Y. and Halban, P.A., Decreased beta-cell mass in diabetes: significance, mechanisms and therapeutic implications. Diabetolo- gia, 2004. 47(3): p. 581-9.

115. Robertson, R.P., Harmon, J., Tran, P.O. and Poitout, V., -cell glucose toxicity, lipotoxicity, and chronic oxidative stress in type 2 diabetes. Diabetes, 2004. 53 Suppl 1: p. S119-24.

116. Kaiser, N., Yuli, M., Uckaya, G., Oprescu, A.I., Berthault, M.F., Kar- gar, C., Donath, M.Y., Cerasi, E. and Ktorza, A., Dynamic changes in -cell mass and pancreatic insulin during the evolution of nutri- tion-dependent diabetes in psammomys obesus: impact of glycemic control. Diabetes, 2005. 54(1): p. 138-45.

117. Kanety, H., Moshe, S., Shafrir, E., Lunenfeld, B. and Karasik, A., Hyperinsulinemia induces a reversible impairment in insulin recep- tor function leading to diabetes in the sand rat model of non-insulin- dependent diabetes mellitus. Proceedings of the National Academy of Sciences of the United States of America, 1994. 91(5): p. 1853-7.

118. Petkov, P., Marquie, G., Donev, S., Dahmani, Y. and Duhault, J., Morphology of the endocrine pancreas in diabetic sand rat (Psam- momys obesus). Cellular and Molecular Biology, 1985. 31(2): p. 61- 74.

119. Bendayan, M., Malide, D., Ziv, E., Levy, E., Ben-Sasson, R., Kalman, R., Bar-On, H., Chretien, M. and Seidah, N., Immunocytochemical investigation of insulin secretion by pancreatic -cells in control and diabetic Psammomys obesus. The Journal of Histochemistry and Cytochemistry 1995. 43(8): p. 771-84.

120. Sasaki, M., Arai, T., Usui, T. and Oki, Y., Immunohistochemical, ul- trastructural, and hormonal studies on the endocrine pancreas of voles (Microtus arvalis) with monosodium aspartate-induced di- abetes. Veterinary Pathology, 1991. 28(6): p. 497-505.

121. Doherty, M. and Malaisse, W.J., Glycogen accumulation in rat pan- creatic islets: in vitro experiments. Endocrine, 2001. 14(3): p. 303-9.

46

122. Graf, R. and Klessen, C., Glycogen in pancreatic islets of steroid di- abetic rats. Carbohydrate histochemical detection and localization using an immunocytochemical technique. Histochemistry, 1981. 73(2): p. 225-32.

123. Toreson, W.E., Glycogen infiltration (so-called hydropic degenera- tion) in the pancreas in human and experimental diabetes mellitus. The American Journal of Pathology, 1951. 27(2): p. 327-347.

124. Davis, J.C., Hydropic degeneration of the  cells of the pancreatic islets produced by synthalin A. The Journal of Pathology and Bacte- riology, 1952. 64(3): p. 575-84.

125. Helmchen, U., Schmidt, W.E., Siegel, E.G. and Creutzfeldt, W., Mor- phological and functional changes of pancreatic B cells in cyclospo- rin A-treated rats. Diabetologia, 1984. 27(3): p. 416-8.

126. Molello, J.A., Barnard, S.D. and Thompson, D.J., Pancreatic cell vacuolation in rats after oral administration of hexamethylmela- mine. Toxicology and Applied Pharmacology, 1984. 72(2): p. 255- 61.

127. Krebs, H.A. and Henseleit, K., Untersuchungen über die Harnstoffbil- dung im Tierkörper. Hoppe-Seyler's Zeitschrift für Physiologische Chemie, 1932. 210 p. 33-66.

128. Andersson, A. and Sandler, S., Viability tests of cryopreserved endo- crine pancreatic cells. Cryobiology, 1983. 20(2): p. 161-168.

129. Gibson, U.E., Heid, C.A. and Williams, P.M., A novel method for real time quantitative RT-PCR. Genome Research, 1996. 6(10): p. 995- 1001.

130. Heid, C.A., Stevens, J., Livak, K.J. and Williams, P.M., Real time quantitative PCR. Genome Research, 1996. 6(10): p. 986-994.

131. Saldeen, J., Cytokines induce both necrosis and apoptosis via a com- mon Bcl-2-inhibitable pathway in rat insulin-producing cells. Endo- crinology, 2000. 141(6): p. 2003-2010.

132. Limpert, E., Stahel, W.A. and Abbt, M., Log-normal distributions across the sciences: Keys and clues. Bioscience, 2001. 51(5): p. 341-352.

133. Niklasson, B., Heller, K.E., Schønecker, B., Bildsøe, M., Daniels, T., Hampe, C.S., Widlund, P., Simonson, W.T., Schaefer, J.B., Rut- ledge, E., Bekris, L., Lindberg, A.M., Johansson, S., Ortqvist, E.,

47

Persson, B. and Lernmark, A., Development of type 1 diabetes in wild bank voles associated with islet autoantibodies and the novel ljungan virus. International Journal of Experimental Diabesity Re- search, 2003. 4(1): p. 35-44.

134. Naik, R.G. and Palmer, J.P., Latent autoimmune diabetes in adults (LADA). Reviews in Endocrine Metabolic Disorders, 2003. 4(3): p. 233-241.

135. Leahy, J.L., Cooper, H.E., Deal, D.A. and Weir, G.C., Chronic hyper- glycemia is associated with impaired glucose influence on insulin secretion. A study in normal rats using chronic in vivo glucose infu- sions. Journal of Clinical Investigation, 1986. 77(3): p. 908-915.

136. Eizirik, D.L., Welsh, M., Strandell, E., Welsh, N. and Sandler, S., Interleukin-1 depletes insulin messenger ribonucleic acid and in- creases the heat shock protein hsp70 in mouse pancreatic islets without impairing the glucose metabolism. Endocrinology, 1990. 127(5): p. 2290-2297.

137. Eizirik, D.L., Welsh, N. and Hellerström, C., Predominance of stimu- latory effects of interleukin-1 on isolated human pancreatic islets. The Journal of Clinical Endocrinology and Metabolism, 1993. 76(2): p. 399-403.

138. Holstad, M., Jansson, L. and Sandler, S., Inhibition of nitric oxide formation by aminoguanidine: an attempt to prevent insulin- dependent diabetes mellitus. General Pharmacology, 1997. 29(5): p. 697-700.

139. Dunger, A., Cunningham, J.M., Delaney, C.A., Lowe, J.E., Green, M.H., Bone, A.J. and Green, I.C., Tumor necrosis factor- and in- terferon- inhibit insulin secretion and cause DNA damage in un- weaned-rat islets. Extent of nitric oxide involvement. Diabetes, 1996. 45(2): p. 183-189.

140. Suarez-Pinzon, W.L., Strynadka, K., Schulz, R. and Rabinovitch, A., Mechanisms of cytokine-induced destruction of rat insulinoma cells: the role of nitric oxide. Endocrinology, 1994. 134(3): p. 1006-1010.

141. Andersson, A.K., Flodström, M. and Sandler, S., Cytokine-induced inhibition of insulin release from mouse pancreatic -cells deficient in inducible nitric oxide synthase. Biochemical and Biophysical Re- search Communications, 2001. 281(2): p. 396-403.

142. Andersson, A.K., Börjesson, A., Sandgren, J. and Sandler, S., Cyto- kines affect PDX-1 expression, insulin and proinsulin secretion from

48

iNOS deficient murine islets. Molecular and Cellular Endocrinology, 2005. 240(1-2): p. 50-57.

143. Eizirik, D.L., Tracey, D.E., Bendtzen, K. and Sandler, S., Role of re- ceptor binding and gene transcription for both the stimulatory and inhibitory effects of interleukin-1 in pancreatic -cells. Autoimmuni- ty, 1992. 12(2): p. 127-133.

144. Gadot, M., Leibowitz, G., Shafrir, E., Cerasi, E., Gross, D.J. and Kais- er, N., Hyperproinsulinemia and insulin deficiency in the diabetic Psammomys obesus. Endocrinology, 1994. 135(2): p. 610-616.

49 / 0   01              2 .3  4 / / 

 /     4 / / 5011  0  5       1 167 / 1 / 1    1  8 $7  / 57        3 9 1   $  011  3   4 /  / 6:  ;  5())57 1  <9 1  $  011  3   4 / / =6

        3 .1/  66     .  ... '%((*%