Open access

Development of Improved Animal Models for the Study of Diabetes

Written By

Emilia Ciobotaru

Submitted: 02 May 2012 Published: 23 January 2013

DOI: 10.5772/49985

From the Edited Volume

Diabetes Mellitus - Insights and Perspectives

Edited by Oluwafemi O. Oguntibeju

Chapter metrics overview

2,344 Chapter Downloads

View Full Metrics

1. Introduction

Medical research based on animal model is rightly considered a “necessary evil”, being a “modus vivendi” in all research activities for more than 2,000 years. It is admitted that the major breakthroughs in medicine such as blood circulation, respiration physiology, the hormonal system used for research purpose different species of animals. In the last 150 years animals used in medical experiments brought huge benefits to humanity by providing crucial responses to the most intriguing questions about prevention and treatment of some devastating diseases. Furthermore, diseases as cancer, AIDS, malaria, tuberculosis, influenza, Alzheimer’s disease and diabetes mellitus were approached by creating specific animal models with respect to pathogenesis, genetic insights and treatment. Despite to all these achievements, over the years a lot of people or organizations were and still are reluctant to animal research because this brings intolerable suffer and pain. All of those mentioned emphasized that animal models are not the only scientific methods to achieve important and reliable results. Consecutively, it was constantly sustained that animal research should be abandoned at once and further efforts should be invested in creating alternative methods. For preventing barbarity against animals which was rightly condemned in the past, new concepts were necessary to be enforced. Thus, “animal rights” (animals are granted to live a life free from abuse and exploitation which also includes prevention of use an animal for scientific research) and “animal welfare” (for the animals used in research this implies assessment of breeding, transport, housing, nutrition, disease prevention and treatment, handling and, where necessary, euthanasia) were two of the most invoked [1].

Laboratory animal welfare was first defined in The Principles of Humane Experimental Technique written by William Russell and Rex Burch. The essence of this work refers to the three Rs (3Rs):

  • refinement: decrease in the incidence of the severity of inhumane procedures applied to those animals used for research purpose;

  • reduction: reduction in the number of animals used to obtain information of given amount and precision;

  • replacement: the substitution of conscious living animals with insentient materials.

Nevertheless, the 3Rs were the subject of dispute between animal research supporters and those who are against animal experimentation. Animal welfare was consistently improved by implementing of the 3Rs, but some important issues were created in some area of medical research. For instance, validation of the alternative methods which replaces the animals, reliable results based on statistical analysis when a smaller number of animals are used or refinement of the methods for induce less pain and suffering (e.g. administration of analgesics after surgical procedures) were the most debated in the last forty years. The scientific world is still preoccupied by further implementing of the 3Rs [2, 3]. In USA, National Institute of Health stopped financing almost all new projects which use chimpanzees as the closest human’s related animal model [4]. This species become nonessential due to alternative research tools and methods, this being one of the last benefits of Russell’s and Burch’s 3Rs.

Advertisement

2. Experimentally induced hyperglycemia

Hyperglycemia is one of the most important signs of diabetes mellitus, both surgical removal of the pancreas and administration of β-cell toxins being equally used. The first method has been used for the first time in a canine model designed by Oskar Minkowski and Josef von Mering. Partial or total surgical removal of the pancreas was followed by the most “popular” clinical sign of diabetes: glucosuria, body weight loss despite voracious appetite and intake of nourishing food, polyuria, polydipsia and ketonuria [5, 6]. This experiment was followed by another historical breakthrough accomplished by Frederick Banting and Charles Best. These two scientists performed a ligation of pancreas ducts to induce atrophy of exocrine acinar component and thereby to obtain a less contaminated extract of pancreatic islets. This extract succeeded to determine a substantial prolongation of life in dogs with pancreatectomy and also to save the life of a diabetic boy [7].

It is well known that the beginnings of the research in diabetes aimed as animal model the dogs and the rabbits. Later, the scientists preferred to conduct experiments in smaller animals, these being easier to manipulate and involve smaller expenses. Thus, rats and mice were subjected for pancreatectomy. This surgical procedure is challenging because of the particular anatomy of the pancreas and pancreatic ducts in this species. The rat pancreas is spread on a large anatomic area, being divided in three parts (biliary, duodenal and gastrosplenic portions). The duct system is quite polymorphic and represented by numerous independent pancreatic ducts which drain secretion from each corresponding part. The results of pancreatectomy in rat were not always followed by the rapid onset of the diabetes and do not reflect entirely the diabetes in humans, these being speculated by those who consider that larger species are more appropriate for diabetes study [8, 9].

Toxins as streptozotocin [10], alloxan [11], vacor [12], dithizone [13], and 8-hydroxyquinolone [14] were used as non surgical methods. Each toxins aim to induce various destruction of β-cells and produce diabetes and subsequent complications.

Both surgical removal of the pancreas and toxin induced diabetes are valuable methods used for studyingthe consequence of hyperglycemia and the onset of diabetes complications such as diabetic microangiopathy and macroangiopathy, retinopathy, neuropathy, and cardiomiopathy. Cardiomiopathy, as a complication of streptozotocin induced diabetes was revealed by gravimetric assessments and morphometry. Diabetic rats present hypertrophy of left ventricle, revealed by increased values of ventricular ratio, comparing with control group. Same groups exhibited significant increasing of heart weight/body weight ratio and liver weight/body weight ratio, comparing with control group [15]. Considering that cardiac hypertrophy is the result of potential interstitial fibrosis, thickening of arteriolar media, endothelial cells and basement membrane changes, morphometry of arteriolar media of heart arterioles and cellular density of media were assessed. Arteriolar media/diameter of arteriolar lumen was significantly bigger in rats with streptozotocin induced diabetes, this being the result of fibrosis in arteriolar media [16].

Islet cell transplantation and its consequence is one of the current research targets, being conducted on either surgical removal of the pancreas and toxin induced diabetes. Successful transplantation was achieved for the first time in 1966 in patients with diabetic nephropathy subjected for simultaneous pancreas and kidney transplantation [17]. Despite to consistent benefits of this therapeutic management, the lack of donors, the acquired chronic immunosupression, postoperative complications and graft rejection have to be considered. Thus, islet transplantation era began with two experiments in rodents previously rendered diabetic by the methods described above [17-19]. The methods of transplantation became more refined correlated with and requested by all the shortcomings resulted by immunosupression and graft rejection. Therefore, pancreatic islets graft may be transplanted as alginate or alginate-polylysine immunoisolated microcapsules [20-22], which are implanted in various anatomic sites (subcutaneously, into the splenic parenchyma, under renal capsule, into the peritoneum, into the portal vein for further colonization in the liver) [17-19, 23]. Unfortunately, the lifetime of transplanted islets is shortened by the deleterious immune reaction of the host. Without microcapsule protection and immunosuppressive treatment, islets transplanted into the liver are immediately surrounded by thrombi placed into the vessels of the surrounding tissue. Allogeneic islets from liver and spleen present lymphocytic infiltrations in 2 days after transplantation and are destroyed rapidly by the host [24].

Diabetic rodents are frequently used in research concerning pharmaceutical compounds aimed to lower the level of glycemia in diabetic persons. New formulas are previously tested on diabetic rodents in order to estimate efficacy, and potential toxic effect on the patients.

Advertisement

3. Experimentally induced glycosuria

Phlorizin is an organic compound, member of chalcone class, extracted for the first time from the bark of the apple tree. The compound was also isolated from roots bark, shoots, leaves and fruits, proving that phlorizin is usually ingested by humans. It was observed that ingestion of more than 1g of phlorizin is followed by glycosuria. Knowing that diabetes mellitus expresses urinary symptoms such as glucosuria and polyuria, an important correlation has been made between these symptoms and the effect of phlorizin. Chronic administration in dog was followed by glucosuria, polyuria and weight loss, creating this way an obvious resemblance between human spontaneous diabetes mellitus and phlorizin effect [25]. Diabetic rats treated with phlorizin express values of glycemia almost equal with normal parameter. This model was used to clarify the implication of hyperglycemia in the progression of islet lesions. The results proved that chronic hyperglycemia might have no effect of islets histopathological changes [26].

Advertisement

4. Chemically induced insulin dependent diabetes mellitus – animal models

Considering that insulin dependent diabetes mellitus (IDDM) features the immune-mediated destruction of β-cells and subsequent insulinopaenia, animal models which reproduce damage of pancreatic islets have been created. For this purpose, streptozotocin and alloxan induced diabetes mellitus were considered the handiest manners to create this condition, although naturally, β-cells become dysfunctional after a long period without evident clinical signs. Streptozotocin and alloxan are diabetogenic chemicals, both being framed in the group of glucose analogues. The onset of β-cells destruction is induced via different mechanisms. Alloxan was the first used as a toxic agent against β-cells, its ability being to generate both reactive oxygen species (ROS) and inhibition of glucose mediated insulin secretion through glucokinase blockage. During the destructive process, β-cells express reversible transformation of cytoplasmic organelles (cytoplasmic vacuolization, dilation of rough endoplasmic reticulum, reduced Golgi apparatus, scattered insulin content secretory granules and swollen mitochondria) finalizing with irreversible damaging of DNA (TUNEL positive staining of β-cells nuclei) [11]. Streptozotocin has antibiotic and chemotherapeutic properties, being isolated from Streptomyces achromogenes. The main action of streptozotocin is focused on β-cells DNA via alkylation process. Finally, DNA methylation results into the fragmentation and ultimately generates cell death [11, 14, 27]. Streptozotocin diabetes mellitus can be induced via a single large dose or multiple low doses administration. It is possible that the first option to induce diabetes because of direct toxic effect of streptozotocin, while, low doses repeatedly administrated may exert blockage of insulin secretion [14].

Other diabetogenic compounds were used in experimental models such as dithizone [28]. Administration of this chelator in rabbit has a particular effect expressed as initial hyperglycemia after 2 hours, followed by normoglycemia in 8 hours and finalized by permanent hyperglycemia due to degranulation of β-cells [29].

Advertisement

5. Spontaneous IDDM based on animal models

The non-obese diabetic (NOD) mouse (table 1) is a spontaneous IDDM animal model. This was spontaneously obtained in one of two sublines derived from CTS mice (Immune Deficiency of Cataract Shionogi). The diabetic line was established after six generations of breeding [30]. About 20% of males and 80% of females develop type 1 diabetes mellitus around 30 weeks of age in particular environment (the incidence of diabetes is higher in colonies maintained in relatively germ-free conditions). The lesions of Langerhans islets are expressed as insulitis, the onset of insulinopaenia being recorded in 12-week-old females. Polyuria, polydipsia, hyperglycemia, glucosuria and hypercholesterolemia are the main clinical signs [30, 31]. Daily administration of insulin improves consistently the body weight and life span, although the mice can survive for weeks without insulin supplement. It is noteworthy that the low level of insulin in NOD mice is correlated with increase secretion of glucagon in treatead and non-treated with insulin individuals. Thus, is concluded that insulin deficiency and glucagon hypersecretion might have an important role in the development and clinical progress of diabetes in NOD mice [32]. Pinealectomy in newborn mice is followed by a more rapid onset of diabetes in female and supplementary melatonin administration protects the animals. The results are somehow intriguing, knowing that melatonin induces increase of insulin autoantibodies [33]. NOD mice are prone to develop autoimmune inflammations, especially those with anti-diabetogenic MHC haplotype and programmed death cell deficiency (sialadenitis of submandibular gland, thyroiditis, gastritis, vasculitis of renal arteries, neuritis) [34-36]. The most important studies which have been run on NOD mice targeted gene implication, MHC genes class II having an important role. Also, knowing that NOD mouse develop cell immune mediated diabetes, many of the experiments aim to picture the immunological status which is responsible for the onset of diabetes [37, 38]. It is important to bear in mind that diabetes in NOD mice is not only the result of cell mediated immunity but also of humoral factors as GAD and IgM [39].

Akita mouse (Ins2Akita) was obtained from a spontaneous point mutation in a female of C57BL/6 line. This mutation disrupts normal synthesis of insulin via incapacity to produce and secrete mature insulin. Clinical signs of diabetes are clearly expressed in male, comparing with female. Heterozygous mutant mice present hyperglycemia, hypoinsulinemia, polydipsia and polyuria. The mice are lean and do not present insulitis. Pancreatic islets exhibit decreased density of β-cells and decreased density of secretory granules in the existing β-cells, increase amount of endoplasmic reticulum and swollen mitochondria [40]. Progressive diabetic retinopathy begins around 12 weeks of age after the onset of hyperglycemia and is consistent with increased vascular permeability, morphological abnormalities of astrocytes and microglia, apoptosis and thinning of inner layer of the retina [41]. Heterozygous Ins2Akita are suited for allogeneic and xenogeneic islet transplantation, because it provides a biological status free of unwanted toxic effect of streptozotocin and alloxan and without β-cell autoimmunity [42].

BB (bio breeding) rat also known as BBDP (bio-breeding diabetes prone)rat is an inbred laboratory rodent which spontaneously develops IDDM. The animals between 2 and 4 months of age develop spontaneous hyperglycemia, different degrees of mononuclear infiltration of the pancreatic islets or total loss of β-cells, insulinopaenia and ketogenesis [43-45]. The overt diabetes can be reversed in 36% of diabetic rats when BB/Worchester (BB/W) are treated with rabbit anitiserum to rat lymphocytes. These results highlight that diabetes mellitus in BB rats is a cell-mediated autoimmune disease [46]. Destruction of β-cellsis performed by a cohort of immune cells such as T and B-lymphocytes, macrophages and natural killer cells [38, 47]. The BB/Worchester diabetic rats may develop lymphocytic thyroiditis in individuals between 8 and 10 months of age [48]. The onset of diabetes in BB rats is attributable to many genes, the most important being those which trigger the age of the onset of diabetes, diabetes susceptibility, severity of islet infiltration with inflammatory cells and islet atrophy [49].

RodentsMouseNon-obese diabetic (NOD) mouse
Akita mouse
RatBB/BBDP
Long Evans Tokushima Lean (LETL)
Komeda Diabetes Prone (KDP)
RabbitNew Zealand White Rabbit
HamsterChinese hamster (Cricetulus griseus)
DogKeeshond dog

Table 1.

Animal models for insulin dependent diabetes mellitus

As an overview of either differences or resemblances between NOD mouse, BB rats and human IDDM data are presented in table 2 [50].

CharacteristicsHumanNOD miceBB rats
Genetic predisposition (MHC class II)yesyesyes
Genetic controlpolygenicpolygenicpolygenic
Haemopoietic stem cell transferyesyes [50]yes [50]
Lymphocytic insulitis (with T-lymphocytes)yesyesyes
Lymphocytic infiltrates in other organssometimesyesyes
Humoral reactivity to β-cellsyesyes [39]no
Diabetic ketoacidosis (without treatment)yesmildyes
Detection of retroviral antigens expressed in beta cellsnoyes [51]no
Sex predispositionnoyesno

Table 2.

Comparative overview in human, NOD mouse and BB IDDM

Long Evans Tokushima Lean (LETL). An outbred colony of Long-Evans rats developed spontaneously remarkable signs attributable to diabetes (polyuria, polyphagia, and polydipsia). This line has been maintained since 1983 in Tokushima Research Institute (Otsuka Pharmaceutical, Yokushima, Japan) and generated another line (Long Evans Tokushima Lean - LETL). LETL rats present no sex predilection concerning the onset of the disease or severity, sudden onset of the diabetes expressed as hyperglycemia, polyuria, polydipsia and weight loss, lymphocytic insulitis at 120-220 days of age followed by the destruction of β-cells, normal levels of T-lymphocytes, lymphocytic infiltration of salivary and lacrimal glands [52, 53].

Komeda Diabetes Prone(KDP)rat is a substrain of LETL, all the individuals presenting moderate to mild insulitis around 220 days of age. The onset of diabetes is 70% at 120 days and 82% within 220 days. This strain present a major IDDM susceptibility gene named Iddm/kdp1 placed on chromosome 11. Homozigous alleles at this locus are strongly linked with the capacity to develop moderate or severe insulitis [54-56]

New Zealand White Rabbit developed spontaneous diabetes mellitus for the first time in a female in 1969. By inbreeding this female and her offspring, a diabetic line was obtained. The overt diabetes was diagnosed in 19% of animals aged between 1 and 3 years. The diabetic animals present fasting hyperglycemia, hypoinsulinemia and absent ketoacidosis [57]. The lesions of β-cells are expressed as cytoplasmic hypergranulation, this being different comparing with previous animal models featured by insulitis, islet atrophy, degranulation of β-cells. It was postulated that the lesion is the consequence of a secretion defect. In addition, diabetic rabbits present mineral deposits in kidney, particularly in basement membrane of the tubules and Bowman capsules and into the lining cells of proximal convoluted tubules [58, 59].

Certain lines of Keeshond dog may develop inherited IDDM expressed as overt diabetes around 2-6 months of age. The dogs have low level of insulin as a consequence of β-cells aplasia. In addition, glucagon secretion is also depressed. The dogs can survive 2-4 months without insulin supplement. Concurrent lesions such as cataracts, skin infections and poor bodily growth are observed. The incidence of diabetes is higher in females. The fertility in diabetic individuals is very low, non-diabetic dogs being used to obtain diabetic offspring. An autosomal recessive disorder is consistent with the onset of diabetes. Keeshond dogs are suitable for studying long term complications of diabetes [60, 61].

Chinese hamster(Cricetulus griseus) has become the subject of research in diabetes mellitus as an animal model since 1959 [62]. The incidence of diabetes in Chinese hamster sublines is more than 85%. At the time of birth, the pups are prediabetic. The overt diabetes range from mild to severe and it is characterized by polyphagia, hyperglycemia, severe polyuria, glucosuria and elevated gluconeogenesis. β-cells present degranulation and hydropic degeneration [63-65]. Other morphologic changes occur in kidneys (glomerulosclerosis), brain (vascular lesions expressed as duplication and thickening of the basement membrane, degeneration in either dendrites or axons, focal demyelination and synaptic degeneration) [66], exocrine pancreas (pancreatic adenoma and adenocarcinoma) [67], teeth (periodontal disease) [68], and macroangiopathy of the thoracic aorta [69]. Genetic defects are responsible for the onset of diabetes, four autosomal recessive genes being involved [70]. Chinese hamster with IDDM have an impaired humoral antibody response similar to that developed in human diabetes, which makes it suitable for research concerning the consequence of diabetes mellitus induced by impaired immune response [71], as well as for diabetic nephropathy [72].

Advertisement

6. Animal models of non-insulin dependent diabetes mellitus (NIDDM)

NIDDM is generated by the failure of β-cells to adapt to a more challenging conditions created by insulin resistance, this being induced by over-nutrition and lack of physical exercises. Mechanisms as oxidative stress, islet amyloidosis, glucotoxicity and lipotoxicity were associated with inappropriate secretory behavior of β-cells. Autoimmune attack and islet inflammation considered previously as a hallmark for IDDM, is now associated with NIDDM. This concept is sustained by the fact that all mentioned mechanisms may initiate inflammation or are initiated by the inflammation. One of the reasons is that human pancreatic islets release IL-1β as response to glucotoxicity. The inflamation is somehow blocked in the initial stages for allowing β-cell regeneration. The more necrosis and apoptosis become obvious, the more infiltration with inflammatory cells (e.g. macrophages) are seen in pancreatic islets [73, 74].

Creation of animal models of NIDDM needs to meet the heterogeneous background which features human condition. Roughly, the animals have to express insulin resistance, impaired insulin secretion in the condition of fasting or post-challenge hyperglycemia. On the other hand the existent animal models present as dominant at least one characteristic: some animals are insulin resistant, other express mainly glucose intolerance as a part of obesity, others express NIDDM because of a particular sensitivity to dietary components. The animal models used for research in NIDDM present an important diversity, although mice and rats are constantly preferred (table 2).

RodentsMouseObeseob/ob mouse
db/db mouse
KK mouse
NZO mouse
NONcNZO10 mouse
NSY mouse
TH mouse
TSOD mouse
M16 mouse
CBA/ca mouse
Gene mutation
Diet inducedC57/BL 6J mouseDiet-gene interaction
RatObeseZDF rat
Wistar fatty rat
OLETF rat
SHR/NIH-cp
Gene mutation
Non-obeseGK rat
Torii rat
Gene mutation
Diet inducedCohen diabetic rat
Israeli sand rat
Nile rat
Diet-gene interaction
Pig [75]-Yucatan minipig
Göttingen minipigs
Sinclair minipigs
Yorkshire and Yorkshire crossed strains
Chinese Guizhou minipig
Ossabaw minipigs
Familial hypercholesterolemic pigs
Low-birth-weight pigs
Cardiovascular complications
Cat [76]-Shorthaired malesIslet amyloidosis
Monkey [77]-Non human primatesIslet amyloidosis

Table 3.

Animal models for NIDDM

Ob/Ob mouse was created in Jackson Laboratories in 1949 and resulted from mutation on both obese (ob) genes [78]. The main characteristic of this mutant is the uncontrolled appetite which results rapidly in the onset of obesity and NIDDM around 11 weeks of age. Polyphagia in ob/ob mouse is generated by ob genes mutations which also encode leptine. This hormone is synthesized by adipose tissue and has an important role in appetite downregulation and regulation of body weight. Leptin is absent in obese mice, the treatment with this monomer lowers consistently the food intake and body weight and also improve up to normal the plasma levels of glucose and insulin [79]. Persistent mild hyperglycemia is linked with 60% enlargement of pancreatic islets and subsequent hyperinsulinemia comparing with lean mice. Interestingly, β-cell from obese mice secretes insulin at a lower threshold of glucose that lean mice [80]. High level of plasma insulin may result from metabolic alteration of β-cells that leads to insulin overproduction or is the consequence of the heterogeneity in glucose sensitivity of these cells. Increased concentration of glucose is followed by recruitment of new β-cells with increased glucose sensitivity [81]. Infertility is a current feature in obese mouse, this being supported by fatty degeneration of the ovaries, follicular atresia, damaged mitochondria and apoptosis of the ovocytes [82]. Many studies have been run in ob/ob mice such as amelioration of insulin resistance [83], hypoglycemic effects of some polysaccharids [84, 85] and complication of NIDDM as diabetic cardiomiopathy [86] and peripheral neuropathy [87].

Db/db mouse is a diabetic mutant mouse created in Dunn Nutritional Laboratory, Cambridge, United Kingdom in 1966. Particularly, this mutant expresses a mutation on db gene which encodes the leptin receptor [88]. Thus, leptin signaling in the hypothalamus is absent leading to persistent high levels of both insulin and leptin. The mouse becomes obese around 4-6 weeks of age and develops progressively high levels of plasma insulin and glucose. All characteristic clinical signs are recorded: polyuria, polydipsia, polyphagia, proteinuria, and glucosuria. One of the most intriguing aspects is that the mice of some strains maintain hyperinsulinemia despite severe depletion of β-cells. This can be attributed to stem cells differentiation from pancreatic ducts. Body weight and insulin levels begin to decrease in association with β-cell degeneration when the mouse reaches 5-6 months of age. The cause of death remains unclear, although the mice present ketonuria, hematuria and gastrointestinal hemorrhages in terminal stage [89]. Db/db mouse has a long history in comparative research to human diabetes. Thus, human dietary habits were reproduced in db/db mouse. High lipid and cholesterol reach diet induce dyslipidemia and create similarities with the patients with type 2 diabetes mellitus [90]. Furthermore, diabetic nephropathy in db/db mice is consistent with some features encountered in human diabetic nephropathy such as renal hypertrophy, glomeruli enlargement, albuminuria, and mesangial matrix expansion [91].

KK mouse history began in 1957, this line being derived from numerous strains of Japanese native mice. Later, after many inbreeding procedures, Nakamura obtained KK mouse strain, which was a polygenic model, spontaneously diabetic and named after the region where the strain was founded (Kasukabe in Saitama prefecture) [92]. The KK mice become obese once with the onset of adulthood and develop insulin resistance, subsequent hyperinsulinemia and β-cell hyperplasia. Particularly, KK mice present a chemical diabetic stage preceded by prediabetes stage accompanied by renal, neurological and retinal complications [93]. The severity of diabetes is strongly correlated with environmental factors such as diet, food intake and social isolation of the animals, the chemical diabetic state being replaced with overt diabetes [94, 95]. Diabetes and obesity in KK mouse has a moderate expression. Introduction of Ay allele creates a new line (KK- Ay) which present enhanced pathophysiological characteristics especially for glucose intolerance [96].

New Zealand obese (NZO)mouse is a polygenic animal model which is prone to express obesity, insulin resistance, glucose intolerance and also autoimmunity featured by perturbation of splenic lymphocyte function and IgM antibodies to insulin receptor. These characteristics are concomitant with poor breeding performance due to ovarian degeneration [97] and diabetic nephropathy expressed as glomerular proliferation, mesangial deposits, mild thickening of basement membrane, glomerular eosinophilic nodules and glomerulosclerosis [98-100]. There are research which concluded that the obesity develops independently to dietary content, the onset of diabetes being recorded earlier in mice fed with carbohydrates and fat reach diet [101]. Other studies emphasized that obesity in NZO individuals is the results of hyperphagia and low energy expenditure due to insufficient physical activity [102].

NONcNZO10 mouse is a recombinant congenic new strain of NIDDM developed by introgressing 5 genomic intervals containg NZO/H1Lt (NZO) diabetogenic quantitative trait loci onto non-obese non-diabetic (NON/Lt or NON) genetic background [103]. Particularly, these mice do not express polyphagia, morbid obesity, poor fertility and variable frequency of hyperglycemia as their parental NZO males do. NONcNZO10 males are normophagic, moderately obese and exhibit normal fertility. NONcNZO10 males become hyperglycemic in 12-20 weeks of age and present atrophy of pancreatic islets and hepatic lipidosis. The resemblance between NONcNZO10 mice and human obesity/diabetes syndrome in higher than ob/ob and db/db mice because of lack of hyperphagia, normal levels of leptin and leptin signaling, normal thermoregulation and lack of hypercorticism [104].

Nagoya-Shibata-Yasuda (NSY) mouse is a spontaneous model of NIDDM, having the same ancestor with NOD mouse (Jcl ICR line). Surprisingly, three major loci contributing to susceptibility to NIDDM in the NSY mouse presented overlapping with the region where susceptibility genes for IDDM have been mapped in NOD mouse. It was postulated that some responsible genes for the onset of diabetes come from the same ancestor genes which express IDDM phenotype in NOD mice and NIDDM in NSY mice [105]. Age and sex related onset of diabetes is the most prominent characteristic for NSY mice. Males develop diabetes at 48 weeks of age as mild obesity and mild hyperinsulinemia. The impaired insulin secretion via glucose challenge is observed after 24 weeks of age. There were no morphological changes in pancreatic islets in NSY mice at any age, these findings suggesting that defective secretion of β-cells may be one of the causes in NIDDM in the NSY mouse. Fasting hyperinsulinemia may contribute to the pathogenesis of diabetes in NSY mouse, insulin sensitivity being under genetic control of Nidd2nsy and Nidd3nsy genes. Genetic analysis of NSY identified a specific gene mutation of Tcf2 responsible for encoding hepatocyte nuclear factor 1β (HNF-1β) and implicated in MODY pathogenesis [106, 107]. Spontaneous amyloidosis was reported in old individuals, deposits being remarked mainly in kidneys, but also in the different segments of digestive system, lung, heart and adrenal glands [108]. NSY mice as well as ob/ob mice prove recently to be the source of creating of new animal models for simultaneous development and research of Allzheimer’s disease and NIDDM [109].

TallyHo (TH) mouse is a relatively new NIDDM animal model, reported for the first time in 2001. The mice present obesity, hyperinsulinemia, hyperlipidemia and male-limited hyperglycemia, insulin resistance and glucose intolerance. It has been postulated that female diabetes resistance is the consequence of estrogens which enhance hepatic insulin sensitivity [110]. The genome wide scan proves polygenic involvement and also additional gene-gene interactions to express hyperglycemic phenotype [111]. Comparing with ob/ob and db/db mice, which present severe obesity attributable to leptin synthesis and leptin receptor deficiencies respectively, TH has normal levels of this hormone and also intact leptin signaling. Carbohydrates and fat reach diet enhance the levels of leptin and also the other specific features of NIDDM [112]. The treatment with leptin results in decreased glucose-stimulated insulin secretion, which demonstrate that letin plays an important role in initiation of glucose intolerance in TH mice [113]. Both males and females develop early moderate hyperplasia and hypertrophy of pancreatic islets, but only the males continue these lesions with β-cell degranulation, discrete vacuolization, different degrees of islet atrophy and fibrosis [114]. Vascular dysfunction occurs in TH mice, expressed mainly in aorta, carotid arteries and cerebral arterioles as a consequence of PGH2/TxA2 receptor activation and cytochrome p450 products and oxidative stress and elevated activity of Rho kinase, respectively [115, 116].

Tsumura Suzuki obese diabetic (TSOD) mouse resulted from inbreeding procedure of ddY strain. The diabetic line includes only moderate obese males with polyphagia, polydipsia, glucosuria, hyperglycemia, hyperinsulinemia, and hyperlipidemia. Pancreatic islets exhibit hypertrophy and hyperplasia, without any signs of insulitis or islet fibrosis [117]. Diabetic nephropathy is consistent with thickened basement membrane of the glomeruli and increased mesangial area. Peripheral neuropathy involves both sensitive and motor nerves and expresses high resemblance with human counterpart. The most prominent lesions of the nerve are decreased density of nervous fibers due to endoneurial fibrosis, degeneration of myelin sheath, intralamellar edema and remyelination, total destruction of lamellar structure associated with macrophage invasion around and into the myelin sheath [118]. Insulin resistance in TSOD mouse is probably induced, at least partially, by a decreased GLUT 4 translocation by insulin in skeletal muscles and adipose tissue [119].

M16 mouse is a new obese animal model created in Institute of Cancer Research, London, UK. Both male and female express early onset of a moderate obesity due to hyperphagia and have high levels of insulin, leptin and cholesterol. The diabetic phenotype of M16 permits research of obesity/diabetes syndrome with early onset as it recorded in human population as a current tendency [120].

CBA/Ca mouse diabetes is recorded only in 10-20% of males. The incidence can be enhanced by inbreeding. Hyperphagia, obesity, hyperglycemia, glucose intolerance, hyperinsulinemia, hypertriglyceridemia occur around 12-16 week of age. Pancreatic islets are hypertrophied, with increased insulin content that persist up to 48 weeks of age. The islet degeneration as a prove of β-cell exhaustion does not appear in this mouse [121, 122].

Zucker diabetic fatty rats (ZDF rats) resulted from inbred as well as outbred lines of rats, which maintain hyperglycemia and glucose intolerance featuring NIDDM. The scientist Lois M. Zucker and Theodore F. Zucher created in 1961 a line of rats which express a gene responsible for the onset of obesity (fatty gene/fa). Usually these rats are not hyperglycemic and present leptine-receptor deficiency, although both male and female express some parameters attributable to insulin resistance. The original colony began spontaneously to present hyperglycemia and glucose intolerance in some bucks and does. These individuals were the founders of the Zucker diabetic fatty rats. ZDF rats develop hyperglycemia with concomitant β-cell death. Compensatory proliferation is maintained as long as plasma glucose levels remain moderate [123-125]. Subsequent exhaustion of β-cells is followed by an increased rate of apoptosis [126]. Lipotoxicity is also considered as a potential cause of β-cell population reduction. Thus, elevated lipogenesis prior to, or in association with hyperglycemia results in excessive accumulation of fatty acid into the β-cell cytoplasm [127]. ZDF rats are frequently used in comparative studies with non-diabetic fatty rats and lean ZDF.

Besides the ZDF rats, other strains have been created, all receiving fa-gene from Zucker rats. Wistar fatty rats (fa/fa) resulted from mating of Zucker with Wistar-Kyoto individuals. The rats from this line are obese, and present hyperlypidemia, hyperinsulinemia and insulin resistance. Wistar fatty rats are prone to develop hypertrophy of pancreatic islets and degranulation of β-cells. The symptoms of diabetes have been observed only in males [125].

Otsuka Long-Evans Tokushima fatty (OLETF) rat resulted from an outbred colony of Long-Evans rats which spontaneously develop polyuria, polydipsia and mild obesity. The onset of hyperglycemia occurs in male and relatively late comparing with other lines (after 18 weeks of age). Particularly, OLETF rats present a specific diabetogenic gene associated with X-chromosome Implication of testosterone is considered to have an important influence in the onset of diabetes in male. This feature is sustained also by the administration of estrogen in castrated males which suppress or delay diabetes. The lesions of pancreatic islets begin with discrete lymphocytic infiltration, followed by the second stage expressed as islet hyperplasia with or without fibrosis in or around the islets and final stage represented by islet atrophy [128, 129]. OLETF are prone to develop diabetic nephropathy, some features of this complication comparable with human diabetes being recorded (diffuse glomerulosclerosis, thickening of basement membrane, PAS-positive deposits in the mesangium or capillaries). Mesangial lesions might express some nodular aspect similar but not identical with specific Kiemmelstiel-Wilson lesions [130].

Spontaneously Hypertensive rat/National Institute of Health-cp (SHR/NIH-cp) was created in Bethesda Maryland USA and associates obesity, NIDDM and hypertension. This rat presents a homozygous genotype for corpulent gene (cp/cp). The males are early hyperphagic and become obese and express hyperglycemia, impaired glucose tolerance, hyperinsulinemia, insulin resistance, high plasma levels of cholesterol and triglycerides, hyperleptinemia and mild essential hypertension [131].

JCR/LA-cp rat (James C. Russel/LA-cp rat) was reported in 1984 as a homozygous genotype for cp gene which develops hyperphagia, obesity, insulin resistance, hyperinsulinemia, glucose intolerance, hyperlipidemia and leptin receptor deficiency. Obese males also manifest cardiovascular lesions such as atherosclerosis and myocardial lesions. Hyperinsulinemia is caused by β-cell hyperplasia followed by islet hypertrophy and fibrosis [132]. Pharmacological researches use this animal model to determine the effectiveness of anti-obesity compounds and also to evaluate long-term benefit to prevent atherosclerosis [133-135]

Goto Kikazaki rat (GK) is one of the polygenic non-obese models of NIDDM which exhibit high resemblances with human condition, especially on hormonal, metabolic and vascular disorders. The line was founded in Japan (Tohoku University in 1975) based on selective repeated inbreeding of non-diabetic Wistar-Kyoto rats with minor glucose intolerance. Diabetes became overt and stable after 30 generations. Despite minor differences between subcolonies of GK, common characteristics were noticed such as decreased β-cell mass, moderate and stable hyperglycemia in adults, hepatic and peripheric insulin resistance and polyuria. Defective function and morphology of pancreatic islets was recorded since embryonic and fetal period featured by reduction of β-cell mass and insulin levels [136-138]. The complications of diabetes in GK rats refer to nephropathy (significant enhancement of kidney weight, glomerular volume, basement membrane thickness, mesangial fraction and total mesangial volume) [139], peripheral neuropathy [140, 141], diabetic osteopathy (trabecular osteopaenia) [142] and diabetic retinopathy (reduction of retinal blood flow, pericytes ghosts, acellular capillaries, increased production of vascular endothelial growth factor) [143, 144].

Spontaneously Diabetic Torii (SDT) rat has been developed from Sprague-Dawley rats in 1997 in Research Laboratories of Torii Pharmaceutical, Ohnodai, Chiba, Japan. This rat is particularly characterized by non-obese, sex related onset of NIDDM with insulin hyposecretion and severe diabetic retinopathy. The males develop glucosuria around 20 weeks of age. All males are diabetic by 40 weeks, while only 33% of female rats present diabetes even by 65 weeks of age. Glucose intolerance is noticed in 16-week-old individuals and continues with the onset of hyperglycemia, hypoinsulinemia, long-term survival without insulin treatment and hypertriglyceridemia. Fibrosis of pancreatic islets and ocular lesions such as hypermature cataract, hemorrhages in anterior chamber, tractional retinal detachment and subsequent retinal fibrovascular proliferation are the most important histopathological findings in SDT rats [145, 146]. The attempt to clarify the genetic basis of diabetes in SDT rats succeeded to identify seven quantitative trait loci which affect the levels of plasma glucose and one for body weight. One of them (Dmsdt1) have particular involvement in islet inflammation and fibrosis. It was suspected that this gene might also have implication in retinal lesions [147].

Cohen diabetic rat (CD) is a particular experimental model for study in NIDDM, which make it distinctive comparing with the other models presented. Diet-induced diabetes correlated with a genetic sensitivity is truly considered the most prominent feature of this rat, although it is still unclear which of the dietary components are responsible for the onset of the diabetes. It was observed that CD rats become overtly diabetic when their diet has a high-sucrose low copper content. In addition with diet profile, a sex predilection can be observed: male record a lower growth rate and more severe glucose intolerance than female. CD rats fed with diabetogenic diet do not express obesity or hyperlipidemia [148]. Other studies concluded that high-casein low copper diet is responsible for the onset of the diabetes. This result is based on genetic analysis, a gene (Ica1) being associated with diabetes and bovine casein. Routine histopathological investigation reveals intact pancreas islets and replacement of exocrine acini with adipose tissue. Degeneration of exocrine pancreas remains intact when diabetogenic diet is replaced with a regular one [149]. CD rats develop early hyperinsulinemia and insulin resistance, followed by the exhaustion of β-cells and hypoinsulinemia. The most common complications are diabetic retinopathy and nephropathy [150, 151].

The Israeli sand rat (Psammomys obesus) is a terrestrial mammal, being mostly found in the desert area of North Africa and Middle East. The sand rat is another experimental model for diet induced NIDDM. The high resemblance with human condition derives from distribution of adipose tissue into the subcutaneous and visceral compartments. This animal readily becomes obese when the diet from the natural habitat is replaced with usual laboratory rat chow. It was suspected but not proved yet that some components from the natural diet might have hypoglycemic effect. Thus, the juice from Atriplex halimus (saltbush which has low energy, high water and electrolyte content and represents the basis of the food intake), as well as water extract and dialysate induce a significant decrease of glucose in diabetic sand rat [152]. The development of obesity is accompanied by hyperglycemia, hyperinsulinemia, decreased insulin sensitivity in adipose tissue and liver, and glucosuria, [153, 154]. Comparing with normoglycemic individuals, pancreatic β-cell volume begin to decrease in the obese and diabetic sand rats, as well as GLUT 2 glucose transporter on the cellular membrane and glucokinase in the cytoplasm of β-cells [155, 156]. Progressive loss of β-cells due to cell death is accompanied by hypoinsulinemia and persistent hyperglycemia, generating an irreversible diabetic state in sand rat. Proinflammatory cytokines such as IL-1β are not involved in producing deleterious effect on β-cells [157]. However, initiation of inflammation in sand rat NIDDM seems to be induced by other pathogenic pathways. For instance, a gene named Tanis (the Hebrew word for fasting) and expressed as hepatic receptor for serum amyloid A (SAA) is regulated by glucose and become dysfunctional when diabetes occur. Knowing that SAA and other acute-phase protein received special attention because their implication in cardiovascular disease, Tanis gene may provide answers for questions about the link between diabetes, inflammation and cardiovascular disease [158].

Nile rat (Arvicanthis niloticus) is a recently reported diet-induced model which expresses the features of both Metabolic Syndrome and NIDDM. Nile rats fed with current lab diet present characteristic signs as excessive abdominal adipose tissue, hyperglycemia, hyperinsulinemia, impaired peripheral insulin sensitivity, dyslipidemia (high level of cholesterol and triglycerides), microalbuminuria, and hypertension. Sex predilection was observed in males, which present segregation in two groups: early-onset diabetes and late-onset diabetes. Dietary modulation (high-fat diet) induce the early onset, as well as more accumulation of body fat [159].

Advertisement

7. Transgenic and knockout models used for research in diabetes mellitus

The specific techniques of molecular biology had a valuable contribution for the study of diabetes mellitus. As it was mentioned before, diabetes mellitus involves a considerable heterogeneity given by the multifactorial genetic and environmental conditions. Thus, interpretation of the results in a particular experiment is challenged by this complicated background. For this particular reason, the scientists have felt the need to create transgenic animal model which provide good conditions for studying the effect or implication of a specific gene and corresponding product according to physiological and environmental conditions. The most important outcomes of the transgenic animals are knowledge about gene regulation and development, pathogenesis of diabetes and new approaches in the therapy of this disease.

Transgenic animals, particularly mice, result from two basic techniques of genetic engineering. The first aims to transfer a gene (a new genetic material presented as a foreign DNA construct containing a regulatory region and a coding region for a protein), into the pronucleus of a fertilized ovocyte. After the gene inoculation, the modified ovocytes are transferred in the uterus of a foster mother for further development. After the birth, the pups are genetically scanned to verify whether the new genetic material was incorporated into the host genome. The animals which manifest the transgene are bred and the pups are also analyzed for the same DNA construct. Positive offspring of the second generation are further bred to establish a transgenic line for studying a particular transgenic phenotype. This revolutionary technique has both advantages and disadvantages. The major advantage is that the method enables to obtain transgenic animals with minimal cost and in a short time. The disadvantages are generated by the hazardous integration of the DNA construct in the genome of the host. The locus of integration, as well as the number of copies is unpredictable. Transgene phenotype expression is limited to use for studying a specific protein or RNA. Therefore, this protein will be overexpressed in the transgenic animal. If the target of experimentation is to reduce the expression of a protein, a RNA antisense transgene is used. It is noteworthy that this technique is also disadvantageous because of unpredictable complications and misinterpretation of the results [160].

The second method used for obtaining genetically engineered mice is focused on deleting a specific endogenous gene or gene fragment (knockout) and replacing with an exogenous DNA which present homologous sequences with the endogenous DNA fragment (homologous recombination). The engineered DNA fragment (a vector which is designed to produce a disruption in the target gene) is inoculated in an embryonic stem cell culture. The positive targeted cells are inoculated in a mouse embryo, which will be finally transferred into the uterus of a foster mother. If the experiment is successful, this embryonic stem cells will participate to generate germ cells and finally organs, all having the new recombinant gene [161].

Double transgenic mice can be obtained by maiting. Thus, the offspring of transgenic mice expressing the hemagglutinin of influenza virus under the insulin promoter and transgenic mice expressing T-lymphocytes with receptor for immunodominant epitope of the same virus present typical features for IDDM. The mice are hyperglycemic, hypoinsulinemic, present lymphocytic insulitis, glucosuria and poor bodily growth, features which are consistent with IDDM. The mortality is up to 90% at 3 months of age [162]. This line (TCR-HA Ins-HA) has consistent improvement glucose levels when treated with potato buds lectin [163, 164].

Advertisement

8. Conclusions

The experimentation in diabetes mellitus has known a long history, as well as a continuous and diverse development. Banting and Best as discoverers of insulin and Minkowski as the scientist who create the first experimental model of diabetes mellitus are truly recognized as the pioneers of the research in this area. Although the diversity of animal models created in the last fifty years is somehow overwhelming, their classification and usefulness follows the pathogenesis, corresponding lesions and subsequent complications recorded in human diabetes mellitus. The scientific literature describes many animal models of IDDM, NIDDM and secondary diabetes, although mice and rats are constantly used regardless the purpose of the research. It is easily noticed that the most famous research centers and laboratories developed their own experimental models and also provided genetic material for the creation of other colonies. Considering that hyperglycemia and glucosuria are two of the most important clinical signs of diabetes, some basic substances which induce these signs are described. Thus, Streptozotocin, Alloxan, Vacor, 8-hydroxyquinolone, Dithizone are usually used in experimentation which reproduce hyperglycemia, while phlorizin is recognized as a vegetal component which is responsible for glucosuria. The animal models of spontaneous diabetes mellitus are consistently represented by rodents, although other species as dog, cat, pig and primates are recommended. The research in NIDDM is sustained by experimental models divided in three major categories: obese, non-obese and diet-induced models. Molecular biology techniques have an important contribution in creation of transgenic animals for research the depth of the pathogenesis of diabetes mellitus.

References

  1. 1. The ethics of research involving animals,2005Nuffield Council on Bioethics: http://www.nuffieldbioethics.org/animal-research.
  2. 2. FlecknellP.Replacementreduction.refinementA. L. T. E. X.20027378
  3. 3. Sechzer, J.A., Historical issues concerning animal experimentation in the United States.Soc Sci Med F, 19811317
  4. 4. Nolen, R.S. NIH suspends new chimp research grants.Agency adopts strict conditions set out in IOM report. JAVMA News, 2012
  5. 5. JörgensV.OskarMinkowski.(1858-1931An outstanding master of diabetes research. Hormones, 2006310311
  6. 6. MinkowskiO.Perspectivesin.diabetesHistorical development of the theory of pancreatic diabetes (introduction and translation by Rachmiel Levine). Diabetes, 198916
  7. 7. SimoniR. D.HillR. L.VaughanM.Thediscovery.ofinsulin.thework.ofFrederick.BantingCharlesBest.J Biol Chem, 20023132
  8. 8. ScowR. O.Total”“.pancreatectomyin.therat.operationeffects.postoperativecare.Endocrinology, 1957359367
  9. 9. Kara, M.E., The anatomical study on the rat pancreas and its ducts with emphasis on the surgical approach.Ann Anat, 2005105112
  10. 10. Junod, A., et al., Diabetogenic action of streptozotocin: relationship of dose to metabolic response. J Clin Invest, 196921292139
  11. 11. Lenzen, S., The mechanisms of alloxan- and streptozotocin-induced diabetes.Diabetologia, 2008216226
  12. 12. EspostiM. D.NgoA.MyersM. A.Inhibitionof.mitochondrialcomplex. I.mayaccount.forI. D. D. M.inducedby.intoxicationwith.therodenticide.VacorDiabetes, 199615311534
  13. 13. Lazaris, J.A. and Z.E. Bavelsky, Dithisone diabetes in rabbits and its prevention by sulfhydryl and imidazole containing compounds.Endocrinol Exp, 1984157167
  14. 14. Rees, D.A. and J.C. Alcolado, Animal models od diabetes mellitus.Diabetic Medicine, 2005359370
  15. 15. Ciobotaru, E., et al., Gravimetric and morphometric assessments in Wistar rats with experimental diabetes mellitus type 1 and cardiac failure.Acta Veterinaria Beograd, 2008583592
  16. 16. Ciobotaru, E., et al., Morphological chages of myocardial arterioles in rats with experimentally induced diabetes mellitus and cardiac failure (unpublished), 2012.
  17. 17. JamiolkowskiR. M.et al.Islettransplantation.intype. I.diabetesmellitus.Yale J Biol Med, 20123743
  18. 18. Reckard, C.R. and C.F. Barker,Transplantation of isolated pancreatic islets across strong and weak histocompatibility barriers. Transplant Proc, 1973761763
  19. 19. Ballinger, W.F. and P.E. Lacy, Transplantation of intact pancreatic islets in rats.Surgery, 1972175186
  20. 20. KulsengB.EspevikT.Skjak-BraekG.Treatmentof.diabetesmellitus.withtransplantation.ofimmunoprotected.pancreaticislet.tissueTidsskr Nor Laegeforen, 199942194223
  21. 21. Kulseng, B., et al., Transplantation of alginate microcapsules: generation of antibodies against alginates and encapsulated porcine islet-like cell clusters.Transplantation, 1999978984
  22. 22. Rabanel, J.M., et al., Progress technology in microencapsulation methods for cell therapy.Biotechnol Prog, 2009946963
  23. 23. FiedorP. S.OluwoleS. F.HardyM. A.Localizationof.endocrinepancreatic.isletsWorld J Surg, 199610161022discussion 1022-3.
  24. 24. Franklin, W.A., J.A. Schulak, and C.R. Reckard, The fate of transplanted pancreatic islets in the rat.Am J Pathol, 19798595
  25. 25. EhrenkranzJ. R. L.et al.Phlorizina.reviewDiabetes.Diabetes-Metabolism Research and Reviews, 20053138
  26. 26. Janssen, S.W., et al., Phlorizin treatment prevents the decrease in plasma insulin levels but not the progressive histopathological changes in the pancreatic islets during aging of Zucker diabetic fatty rats. J Endocrinol Invest, 2003508515
  27. 27. Bono, V.H., Jr., Review of mechanism of action studies of the nitrosoureas.Cancer Treat Rep, 1976699702
  28. 28. Meiramov, G.G. and N.I. Trukhanov, [The ultrastructure of pancreatic beta cells in dithizone diabetes and its prevention by sodium diethyldithiocarbamate].Probl Endokrinol (Mosk), 19759295
  29. 29. Goldberg, E.D., V.A. Eshchenko, and V.D. Bovt, The diabetogenic and acidotropic effects of chelators.Exp Pathol, 19915964
  30. 30. Makino, S., et al., Breeding of a non-obese, diabetic strain of mice.Jikken Dobutsu, 1980113
  31. 31. KikutaniH.MakinoS.Themurine.autoimmunediabetes.modelN. O. D.relatedstrains.Adv Immunol, 1992285322
  32. 32. Ohneda, A., et al., Insulin and glucagon in spontaneously diabetic non-obese mice.Diabetologia, 1984460463
  33. 33. ContiA.MaestroniG. J.Roleof.thepineal.glandmelatoninin.thedevelopment.ofautoimmune.diabetesin.non-obesediabetic.miceJ. Maestroni, Role of the pineal gland and melatonin in the development of autoimmune diabetes in non-obese diabetic mice. J Pineal Res, 1996164172
  34. 34. Jiang, F., et al., Identification of QTLs that modify peripheral neuropathy in NOD.H2b-Pdcd1-/- mice.Int Immunol, 2009499509
  35. 35. Cihakova, D., et al., Animal models for autoimmune myocarditis and autoimmune thyroiditis.Methods Mol Med, 2004175193
  36. 36. RoseN. R.BonitaR.BurekC. L.Iodinean.environmentaltrigger.ofthyroiditis.Autoimmun Rev, 200297103
  37. 37. Bonifacio, E., et al., International Workshop on Lessons From Animal Models for Human Type 1 Diabetes: identification of insulin but not glutamic acid decarboxylase or IA-2 as specific autoantigens of humoral autoimmunity in nonobese diabetic mice.Diabetes, 200124512458
  38. 38. Yoon, J.W. and H.S. Jun, Cellular and molecular pathogenic mechanisms of insulin-dependent diabetes mellitus.Ann N Y Acad Sci, 2001200211
  39. 39. Shieh, D.C., et al., Insulin-dependent diabetes in the NOD mouse model.I. Detection and characterization of autoantibody bound to the surface of pancreatic beta cells prior to development of the insulitis lesion in prediabetic NOD mice. Autoimmunity, 1993123135
  40. 40. Izumi, T., et al., Dominant negative pathogenesis by mutant proinsulin in the Akita diabetic mouse.Diabetes, 2003409416
  41. 41. Barber, A.J., et al., The Ins2Akita mouse as a model of early retinal complications in diabetes.Invest Ophthalmol Vis Sci, 200522102218
  42. 42. Mathews, C.E., S.H. Langley, and E.H. Leiter, New mouse model to study islet transplantation in insulin-dependent diabetes mellitus.Transplantation, 200213331336
  43. 43. MarlissE. B.NakhoodaA. F.PoussierP.Clinicalforms.naturalhistory.ofthe.diabeticsyndrome.insulinglucagonsecretion.inthe. B. B.ratMetabolism, 1983Suppl 1): 1117
  44. 44. Rossini, A.A., et al., Spontaneous diabetes in the gnotobiotic BB/W rat.Diabetes, 197910311032
  45. 45. Lam-TseW. K.LernmarkA.DrexhageH. A.Animalmodels.ofendocrine/organ-specific.autoimmunediseases.dotheyreally.helpus.tounderstand.humanautoimmunity?.SpringerSemin.Immunopathol2002297321
  46. 46. Like, A.A., et al., Spontaneous diabetes mellitus: reversal and prevention in the BB/W rat with antiserum to rat lymphocytes.Science, 197914211423
  47. 47. Bone, A.J., et al., Insulitis and mechanisms of disease resistance: studies in an animal model of insulin dependent diabetes mellitus. J Mol Med (Berl), 19995761
  48. 48. Sternthal, E., et al., Lymphocytic thyroiditis and diabetes in the BB/W rat.A new model of autoimmune endocrinopathy. Diabetes, 198110581061
  49. 49. Wallis, R.H., et al., Type 1 diabetes in the BB rat: a polygenic disease.Diabetes, 200910071017
  50. 50. LeiterE. H.vonM.HerrathAnimal.modelshave.littleto.teachus.abouttype. .diabetes.In opposition to this proposal. Diabetologia, 200416571660
  51. 51. Tsumura, H., et al., Detection of endogenous retrovirus antigens in NOD mouse pancreatic beta-cells.Lab Anim, 19988694
  52. 52. Komeda, K., et al., Establishment of two substrains, diabetes-prone and non-diabetic, from Long-Evans Tokushima Lean (LETL) rats.Endocr J, 1998737744
  53. 53. Kawano, K., et al., New inbred strain of Long-Evans Tokushima lean rats with IDDM without lymphopenia.Diabetes, 199113751381
  54. 54. YokoiN.et al.non-MA.locusH. C.essentialfor.autoimmunetype. I.diabetesin.theKomeda.Diabetes-Pronerat.J Clin Invest, 199720152021
  55. 55. Yokoi, N., et al., Establishment and characterization of the Komeda diabetes-prone rat as a segregating inbred strain.Exp Anim, 2003295301
  56. 56. Yokoi, N., Identification of a major gene responsible for type 1 diabetes in the Komeda diabetes-prone rat.Exp Anim, 2005111115
  57. 57. Conaway, H.H., et al., Spontaneous diabetes mellitus in the New Zealand white rabbit: physiologic characteristics.Metabolism, 19815056
  58. 58. Roth, S.I., et al., Spontaneous diabetes mellitus in the New Zealand white rabbit: preliminary morphologic characterization.Lab Invest, 1980571579
  59. 59. Roth, S.I. and H.H. Conaway, Animal model of human disease.Spontaneous diabetes mellitus in the New Zealand white rabbit. Am J Pathol, 1982359363
  60. 60. Kramer, J.W., et al., Inherited, early onset, insulin-requiring diabetes mellitus of Keeshond dogs.Diabetes, 1980558565
  61. 61. Kramer, J.W., Animal model of human disease: Inherited early-onset, insulin-requiring diabetes mellitus in keeshond dogs.Am J Pathol, 1981194196
  62. 62. MeierH.YerganianG. A.Spontaneoushereditary.diabetesmellitus.inChinese.hamster.Cricetulusgriseus.Pathological findings. Proc Soc Exp Biol Med, 1959810815
  63. 63. Gerritsen, G.C., The Chinese hamster as a model for the study of diabetes mellitus.Diabetes, 1982Suppl 1 Pt 2): 1423
  64. 64. GreenM. N.YerganianG.GagnonH. J.Predictionof.spontaneoushereditary.diabetesmellitus.inChinese.hamstersby.meansof.elevatedalpha.serumlevels.Nature, 1963396
  65. 65. Gerritsen, G.C. and W.E. Dulin, Characterization of diabetes in the Chinese hamster.Diabetologia, 19677484
  66. 66. Luse, S.A., The ultrastructure of the brain in the diabetic Chinese hamster with special reference to synaptic abnormalities.Electroencephalogr Clin Neurophysiol, 1970410
  67. 67. PoelW. E.YerganianG.Adenocarcinomaof.thepancreas.indiabetes-prone.Chinesehamsters.Am J Med, 1961861863
  68. 68. CohenM. M.ShklarG.YerganianG.Periodontalpathology.ina.strainof.Chinesehamster.Cricetulusgriseus.withhereditary.diabetesmellitus.Am J Med, 1961864867
  69. 69. McCombs, H.L., et al., Morphologic changes in the aorta of the diabetic Chinese hamster.Diabetologia, 1974Suppl: 601606
  70. 70. Gerritsen, G.C., et al., Epidemiology of Chinese hamsters and preliminary evidence for genetic heterogeneity of diabetes.Diabetologia, 1974Suppl: 581588
  71. 71. Fletcher-McGruder, B.L. and G.C. Gerritsen, Deficient humoral antibody response of the spontaneously diabetic Chinese hamster.Proc Soc Exp Biol Med, 19847478
  72. 72. Diani, A.R., et al., Systematic evaluation of microangiopathy in diabetic Chinese hamsters.I. Morphometric analysis of minimal glomerular basement membrane thickness in 11- to 15- and 19- to 23-month-old Chinese hamsters. Microvasc Res, 1986306316
  73. 73. Donath, M.Y., et al., Islet inflammation impairs the pancreatic beta-cell in type 2 diabetes.Physiology (Bethesda), 2009325331
  74. 74. Donath, M.Y., et al., Islet inflammation in type 2 diabetes: from metabolic stress to therapy.Diabetes Care, 2008Suppl 2: S161S164
  75. 75. Bellinger, D.A., E.P. Merricks, and T.C. Nichols, Swine models of type 2 diabetes mellitus: insulin resistance, glucose tolerance, and cardiovascular complications.ILAR J, 2006243258
  76. 76. HoenigM.et al.felineA.modelof.experimentallyinduced.isletamyloidosis.Am J Pathol, 200021432150
  77. 77. Wagner, J.E., et al., Old world nonhuman primate models of type 2 diabetes mellitus.ILAR J, 2006259271
  78. 78. Ingalls, A.M., M.M. Dickie, and G.D. Snell, Obese, a new mutation in the house mouse.J Hered, 1950317318
  79. 79. McGarry, J.D., Appetite control: Does leptin lighten the problem of obesity? Curr Biol,199513421344
  80. 80. TassavaT. M.OkudaT.RomsosD. R.Insulinsecretion.fromob/ob.mousepancreatic.isletseffects.ofneurotransmitters.Am J Physiol, 1992Pt 1): E338E343
  81. 81. Chen, N.G., T.M. Tassava, and D.R. Romsos, Threshold for glucose-stimulated insulin secretion in pancreatic islets of genetically obese (ob/ob) mice is abnormally low.J Nutr, 199315671574
  82. 82. Serke, H., et al., Leptin-deficient (ob/ob) mouse ovaries show fatty degeneration, enhanced apoptosis and decreased expression of steroidogenic acute regulatory enzyme.Int J Obes (Lond), 2011
  83. 83. Takeshita, S., et al., Amelioration of insulin resistance in diabetic ob/ob mice by a new type of orally active insulin-mimetic vanadyl complex: bis(1-oxy-2-pyridinethiolato)oxovanadium(IV) with VO(S(2)O(2)) coordination mode.J Inorg Biochem, 2001179186
  84. 84. Xu, J., et al., Hypoglycemic effects of MDG-1, a polysaccharide derived from Ophiopogon japonicas, in the ob/ob mouse model of type 2 diabetes mellitus.Int J Biol Macromol, 2011657662
  85. 85. Kim, S.W., et al., Proteomic analysis in ob/ob mice before and after hypoglycemic polysaccharide treatments.J Microbiol Biotechnol, 200911091121
  86. 86. Mazumder, P.K., et al., Impaired cardiac efficiency and increased fatty acid oxidation in insulin-resistant ob/ob mouse hearts.Diabetes, 200423662374
  87. 87. Drel, V.R., et al., The leptin-deficient (ob/ob) mouse: a new animal model of peripheral neuropathy of type 2 diabetes and obesity.Diabetes, 200633353343
  88. 88. Chen, H., et al., Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice.Cell, 1996491495
  89. 89. Hummel, K.P., M.M. Dickie, and D.L. Coleman, Diabetes, a new mutation in the mouse.Science, 196611271128
  90. 90. Kobayashi, K., et al., The db/db mouse, a model for diabetic dyslipidemia: molecular characterization and effects of Western diet feeding.Metabolism, 20002231
  91. 91. SharmaK.Mc CueP.DunnS. R.Diabetickidney.diseasein.thedb/db.mouseAm J Physiol Renal Physiol, 2003F1138F1144
  92. 92. TaketomiS.K. K.MiceK. K. A. y.inAnimal.modelsof.diabetesSecond.EditionE.Shafrir, Editor 2007CRC Press Taylor & Francis Group: USA. 335348
  93. 93. Reddi, A.S. and R.A. Camerini-Davalos, Hereditary diabetes in the KK mouse: an overview.Adv Exp Med Biol, 1988715
  94. 94. NonogakiK.NozueK.OkaY.Socialisolation.affectsthe.developmentof.obesitytype.diabetesin.miceEndocrinology, 200746584666
  95. 95. IkedaH.mouseK. K.Diabetes Res Clin Pract, 1994Suppl: S313S316
  96. 96. Suto, J., et al., Genetic analysis of non-insulin-dependent diabetes mellitus in KK and KK-Ay mice.Eur J Endocrinol, 1998654661
  97. 97. Radavelli-Bagatini, S., et al., The New Zealand obese mouse model of obesity insulin resistance and poor breeding performance: evaluation of ovarian structure and function.J Endocrinol, 2011307315
  98. 98. Melez, K.A., et al., Diabetes is associated with autoimmunity in the New Zealand obese (NZO) mouse.Diabetes, 1980835840
  99. 99. BreyerM. D.et al.Mousemodels.ofdiabetic.nephropathyJ Am Soc Nephrol, 20052745
  100. 100. Melez, K.A., et al., Immune abnormalities in the diabetic New Zealand obese (NZO) mouse: insulin treatment partially suppresses splenic hyperactivity measured by flow cytometric analysis.Clin Immunol Immunopathol, 1985110119
  101. 101. Mirhashemi, F., et al., Diet dependence of diabetes in the New Zealand Obese (NZO) mouse: total fat, but not fat quality or sucrose accelerates and aggravates diabetes.Exp Clin Endocrinol Diabetes, 2011167171
  102. 102. Jurgens, H.S., et al., Hyperphagia, lower body temperature, and reduced running wheel activity precede development of morbid obesity in New Zealand obese mice.Physiol Genomics, 2006234241
  103. 103. HaskellB. D.et al.Thediabetes-prone. N. Z. O.HlLt.strainI. Immunophenotypic comparison to the related NZB/BlNJ and NZW/LacJ strains. Lab Invest, 2002833842
  104. 104. Pan, H.J., et al., Pharmacogenetic analysis of rosiglitazone-induced hepatosteatosis in new mouse models of type 2 diabetes.Diabetes, 200518541862
  105. 105. IkegamiH.FujisawaT.OgiharaT.Mousemodels.oftype. .type.diabetesderived.fromthe.sameclosed.colonygenetic.susceptibilityshared.betweentwo.typesof.diabetesI. L. A.ILAR J, 2004268277
  106. 106. Ueda, H., et al., Genetic analysis of late-onset type 2 diabetes in a mouse model of human complex trait.Diabetes, 199911681174
  107. 107. Ueda, H., et al., The NSY mouse: a new animal model of spontaneous NIDDM with moderate obesity.Diabetologia, 1995503508
  108. 108. Shimizu, K., et al., Spontaneous amyloidosis in senile NSY mice.Acta Pathol Jpn, 1993215221
  109. 109. HanW.LiC.Linkingtype. .diabetesAlzheimer’sdisease.Proc Natl Acad Sci U S A, 201065576558
  110. 110. Buck, D.W., 2nd and T.A. Mustoe, Reply: TallyHo Diabetic Phenotype Limited to Male Mice: Female Mice Provide Obese, Nondiabetic Mouse Model.Plast Reconstr Surg, 2012727e728e
  111. 111. Kim, J.H., et al., Genetic analysis of a new mouse model for non-insulin-dependent diabetes.Genomics, 2001273286
  112. 112. Kim, J.H., et al., Type 2 diabetes mouse model TallyHo carries an obesity gene on chromosome 6 that exaggerates dietary obesity.Physiol Genomics, 2005171181
  113. 113. Sung, Y.Y., et al., Glucose intolerance in young TallyHo mice is induced by leptin-mediated inhibition of insulin secretion.Biochem Biophys Res Commun, 200517791787
  114. 114. Kim,J.H., et al., Phenotypic characterization of polygenic type 2 diabetes in TALLYHO/JngJ mice. J Endocrinol, 2006437446
  115. 115. Didion, S.P., C.M. Lynch, and F.M. Faraci, Cerebral vascular dysfunction in TallyHo mice: a new model of Type II diabetes.Am J Physiol Heart Circ Physiol, 2007H1579H1583
  116. 116. Cheng, Z.J., et al., Vascular dysfunction in type 2 diabetic TallyHo mice: role for an increase in the contribution of PGH2/TxA2 receptor activation and cytochrome p450 products.Can J Physiol Pharmacol, 2007404412
  117. 117. SuzukiW.et al.newA.mousemodel.ofspontaneous.diabetesderived.fromdd. Y.strainExp Anim, 1999181189
  118. 118. Iizuka, S., et al., Diabetic complications in a new animal model (TSOD mouse) of spontaneous NIDDM with obesity.Exp Anim, 20057183
  119. 119. Miura, T., et al., Impairment of insulin-stimulated GLUT4 translocation in skeletal muscle and adipose tissue in the Tsumura Suzuki obese diabetic mouse: a new genetic animal model of type 2 diabetes.Eur J Endocrinol, 2001785790
  120. 120. AllanM. F.EisenE. J.PompD.TheM.mousean.outbredanimal.modelof.earlyonset.polygenicobesity.diabesityObes Res, 200413971407
  121. 121. Connelly, D.M. and P.V. Taberner, Characterization of the spontaneous diabetes obesity syndrome in mature male CBA/Ca mice.Pharmacol Biochem Behav, 1989255259
  122. 122. Figueroa, C.D. and P.V. Taberner, Pancreatic islet hypertrophy in spontaneous maturity onset obese-diabetic CBA/Ca mice.Int J Biochem, 199412991303
  123. 123. Finegood, D.T., et al., Beta-cell mass dynamics in Zucker diabetic fatty rats.Rosiglitazone prevents the rise in net cell death. Diabetes, 200110211029
  124. 124. Unger, R.H., How obesity causes diabetes in Zucker diabetic fatty rats.Trends Endocrinol Metab, 1997276282
  125. 125. Wind, D.S. Type 2 diabetes, obesity, and bumblefoot: a possible correlation? 2003 [cited 2012 24.04.].
  126. 126. Pick, A., et al., Role of apoptosis in failure of beta-cell mass compensation for insulin resistance and beta-cell defects in the male Zucker diabetic fatty rat.Diabetes, 1998358364
  127. 127. Finegood, D.T. and B.G. Topp, beta-cell deterioration- prospects for reversal or prevention.Diabetes Obes Metab, 2001Suppl 1: 2027
  128. 128. KawanoK.et al.Spontaneouslydiabetic.rat. O. L. E. T. F.asmodela.ofN. I. D. D. M.inhumans.inLessons.fromanimal.diabetesV. I. E.Shafrir, Editor 1996Birkhausser Boston: USA. 225236
  129. 129. Kawano, K., et al., OLETF (Otsuka Long-Evans Tokushima Fatty) rat: a new NIDDM rat strain.Diabetes Res Clin Pract, 1994Suppl: S317S320
  130. 130. Kawano, K., et al., Examination of the pathogenesis of diabetic nephropathy in OLETF rats.J Vet Med Sci, 199912191228
  131. 131. Velasque, M.T., S.J. Bhathena, and C.T. Hansen, Leptin and its relation to obesity and insulin in the SHR/N-corpulent rat, a model of type II diabetes mellitus.Int J Exp Diabetes Res, 2001217223
  132. 132. Russell, J.C., et al., Insulin resistance and impaired glucose tolerance in the atherosclerosis-prone LA/N corpulent rat.Arteriosclerosis, 1987620626
  133. 133. Brindley, D.N., et al., Sustained decreases in weight and serum insulin, glucose, triacylglycerol and cholesterol in JCR:LA-corpulent rats treated with D-fenfluramine. Br J Pharmacol, 1992679685
  134. 134. Brindley, D.N. and J.C. Russell, Metabolic abnormalities linked to obesity: effects of dexfenfluramine in the corpulent rat.Metabolism, 1995Suppl 2): 2327
  135. 135. Russell, J.C., et al., Cardioprotective effect of probucol in the atherosclerosis-prone JCR:LA-cp rat.Eur J Pharmacol, 1998203210
  136. 136. Gauguier, D., et al., Chromosomal mapping of genetic loci associated with non-insulin dependent diabetes in the GK rat.Nat Genet, 19963843
  137. 137. Galli, J., et al., Pathophysiological and genetic characterization of the major diabetes locus in GK rats.Diabetes, 199924632470
  138. 138. Zhou, H., et al., Network screening of Goto-Kakizaki rat liver microarray data during diabetic progression.BMC Syst Biol, 2011Suppl 1): S1S16
  139. 139. Schrijvers, B.F., et al., Long-term renal changes in the Goto-Kakizaki rat, a model of lean type 2 diabetes.Nephrol Dial Transplant, 200410921097
  140. 140. Wada, R., et al., Effects of long-term treatment with alpha-glucosidase inhibitor on the peripheral nerve function and structure in Goto-Kakizaki rats: a genetic model for type 2 diabetes.Diabetes Metab Res Rev, 1999332337
  141. 141. Murakawa, Y., et al., Impaired glucose tolerance and insulinopenia in the GK-rat causes peripheral neuropathy.Diabetes Metab Res Rev, 2002473483
  142. 142. Ahmad, T., et al., Skeletal changes in type-2 diabetic Goto-Kakizaki rats.J Endocrinol, 2003111116
  143. 143. Sone, H., et al., Ocular vascular endothelial growth factor levels in diabetic rats are elevated before observable retinal proliferative changes.Diabetologia, 1997726730
  144. 144. Yatoh, S., et al., Antioxidants and an inhibitor of advanced glycation ameliorate death of retinal microvascular cells in diabetic retinopathy.Diabetes Metab Res Rev, 20063845
  145. 145. ShinoharaM.et al.newA.spontaneouslydiabetic.non-obeseTorii.ratstrain.withsevere.ocularcomplications.Int J Exp Diabetes Res, 200089100
  146. 146. Sasase, T., Pathophysiological characteristics of diabetic ocular complications in spontaneously diabetic torii rat.J Ophthalmol, 2010615641
  147. 147. YokoiN.FuseM.SeinoS.Geneticsof.thespontaneously.diabeticTorii.ratThe Open Diabetes Journal, 20112125
  148. 148. Weksler-Zangen, S., et al., The newly inbred cohen diabetic rat: a nonobese normolipidemic genetic model of diet-induced type 2 diabetes expressing sex differences.Diabetes, 200125212529
  149. 149. Yagil, C., et al., Metabolic and genomic dissection of diabetes in the Cohen rat.Physiol Genomics, 2007181192
  150. 150. Hammes, H.P., et al., Islet transplantation inhibits diabetic retinopathy in the sucrose-fed diabetic Cohen rat.Invest Ophthalmol Vis Sci, 199320922096
  151. 151. Yagil, C., et al., Nonproteinuric diabetes-associated nephropathy in the Cohen rat model of type 2 diabetes.Diabetes, 200514871496
  152. 152. AharonsonZ.ShaniJ.SulmanF. G.Hypoglycaemiceffect.ofthe.saltbush. .Atriplexhalimus)--a.feedingsource.ofthe.sandrat. .PsammomysObesus.Diabetologia, 1969379383
  153. 153. FrenkelG.KraicerP. F.ShaniJ.Diabetesin.thesand-rat.diabetogenesisresponses.tomannoheptulose.atriplexash.Diabetologia, 1972313318
  154. 154. De FronzoR.MikiE.SteinkeJ.Diabeticsyndrome.insand.ratDiabetologia, 1967140142
  155. 155. Donev, S., et al., Immunohistochemical investigations of the endocrine pancreas in normoglycemic sand rats (Psammomys obesus).Acta Diabetol Lat, 1989309313
  156. 156. Jorns, A., et al., Gradual loss of pancreatic beta-cell insulin, glucokinase and GLUT2 glucose transporter immunoreactivities during the time course of nutritionally induced type-2 diabetes in Psammomys obesus (sand rat).Virchows Arch, 20026369
  157. 157. Jorns, A., et al., Beta cell death in hyperglycaemic Psammomys obesus is not cytokine-mediated.Diabetologia, 200627042712
  158. 158. Walder, K., et al., Tanis: a link between type 2 diabetes and inflammation? Diabetes, 200218591866
  159. 159. Chaabo, F., et al.,Nutritional correlates and dynamics of diabetes in the Nile rat (Arvicanthis niloticus): a novel model for diet-induced type 2 diabetes and the metabolic syndrome. Nutr Metab (Lond), 201029
  160. 160. Livingston,J.N., Genetically engineered mice in drug development. J Intern Med, 1999627635
  161. 161. BronsonS. K.SmithiesO.Alteringmice.byhomologous.recombinationusing.embryonicstem.cellsJ Biol Chem, 19942715527158
  162. 162. Radu, D.L., et al., Double transgenic mice with type 1 diabetes mellitus develop somatic, metabolic and vascular disorders.J Cell Mol Med, 2004349358
  163. 163. Ciobotaru, E., et al., Histological aspects in TCR-HA Ins-HA double transgenic mice treated with potato buds lectin, in 24th Meeting of European Society of VeterinaryPathology2006Edinburgh. 135
  164. 164. Pop, A., et al., Potato buds lectin reduces hyperglicemia in TCR-HA, INS-HA duble transgenic mice.Buletin USAMV Cluj Napoca, 2005259261

Written By

Emilia Ciobotaru

Submitted: 02 May 2012 Published: 23 January 2013