| 
  • If you are citizen of an European Union member nation, you may not use this service unless you are at least 16 years old.

  • You already know Dokkio is an AI-powered assistant to organize & manage your digital files & messages. Very soon, Dokkio will support Outlook as well as One Drive. Check it out today!

View
 

Pathogenesis of Type 1A Diabetes

Page history last edited by Dhemy Padilla 14 years, 11 months ago

PATHOGENESIS OF TYPE 1A DIABETES

 

George Eisenbarth, MD 

 

 

 


 

Introduction

Type 1A diabetes mellitus is defined as immune mediated diabetes mellitus(1-5). It can become manifest with hyperglycemia presenting in the first days of life or in adults over the age of 60. Current estimates indicate that immune mediated diabetes represents approximately 5 to 10% of the diabetes developing in adults and that approximately as many individuals develop this form of diabetes as adults as do children(6-8). In the United States the great majority (>90%) of Caucasian children developing diabetes have type 1A diabetes while approximately 50% of African American and Hispanic American children developing diabetes lack the autoantibody and immunogenetic markers of typical type 1A diabetes(9-11). Most of these latter children appear to have variants of type 2 diabetes with a small number having specific characteristic genetic syndromes (e.g. MODY: Maturity Onset Diabetes of Youth) with identified mutations of genes such as glucokinase and HNF (Hepatic Nuclear Factors)(12). When an individual presents with type 1A diabetes it indicates that they and their relatives have an increased risk of having or developing a series of autoimmune disorders(11). Celiac disease, hypothyroidism, hyperthyroidism, Addison’s disease and pernicious anemia are some of the most prominent associated diseases. For example approximately 1/20 patients with type 1 diabetes have celiac disease(13;14). Most of these patients are asymptomatic and the disorder is only discovered if anti-transglutaminase autoantibodies are measured and individuals with positive antibodies biopsied. In that the therapy for celiac disease, namely gluten avoidance, is highly effective, at the Barbara Davis Center we routinely screen all type 1 diabetic patients. We also screen for thyroid disease and for Addison’s disease (21-hydroxylase autoantibodies)(15).

 

Studies of the pathogenesis of type 1 diabetes have blossomed during the past two decades and there are now complete books devoted to this subject (e.g. Immunology of Type 1 Diabetes available at www.barbaradaviscenter.org with appended “Teaching” slides). In terms of summarizing the pathogenesis of this disorder it is convenient to divide the disease into a series of stages (Figure 1) beginning with genetic susceptibility and ending (from an immunologic standpoint) with complete islet beta cell destruction(15). This is only a general description of the disease process. It is likely for instance that genetic determinants influence many of the stages of disease progression and are important determinants of individuals who express anti-islet autoantibodies but do not progress to diabetes (e.g. a major subset of those with anti-islet autoantibodies and the protective HLA allele DQB1*0602 allele)(16).

 

Figure 1. Hypothetical stages and loss of beta cells in an individual progressing to type 1A diabetes. From Eisenbarth, NEJM, 1986 (17)

 

 

 

STAGE I. GENETIC SUSCEPTIBILITY

An expert committee of the American Diabetes Association has divided type 1 diabetes into type 1A (immune mediated) and 1B (not immune mediated but with profound loss of insulin secretion)(18). The great majority of patients with insulin dependent diabetes have type 1A and firm examples of type 1B are either lacking or controversial. One recent example is a description from Japan of patients who extremely rapidly developed type 1 diabetes such that they had marked hyperglycemia, but at diabetes presentation their HbA1c was near normal, suggesting very recent onset of hyperglycemia(19). These patients lacked anti-islet autoantibodies, a subset had elevated serum pancreatic enzymes, but many had HLA alleles associated with type 1A diabetes. On biopsy beta cells were destroyed, there were lymphocytes in the acinar pancreas, but no typical insulitis (invasion of islets by lymphocytes). Whether this is truly type 1B or an extremely rapid variant of type 1A is currently debated.

 

 

 

Figure 2. Genetic heterogeneity of immune mediated type 1 diabetes with monogenic, polygenic, and oligogenic forms. Modified from teaching slides www.barbaradaviscenter.org

 

 

Type 1A diabetes is itself heterogeneous, with several forms of immune mediated diabetes with known genetic causes as parts of autoimmune syndromes (Thus likely to be classified as other Specific Forms of Diabetes). In particular patients with mutations of the AIRE gene (Autoimmune Regulator)(20)and the human gene homologous to the mutated gene causing Scurfy in mice develop immune mediated diabetes(21)(figure 2: monogenic disorders and figure 3). Mutations of the AIRE gene result in Autoimmune Polyendocrine Syndrome Type I(22). Mutations of the Scurfy homologue lead to overwhelming neonatal autoimmunity (XPID syndrome: X-linked polyendocrinopathy, immune dysfunction and diarrhea; also termed IPEX: Immune dysregulation, Polyendocrinoptahy, Enteropathy, X-linked)(21). A mutation of the FoxP3 genes, an essential transcription factor for CD4+CD25+ regulatory T cells is the cause of the IPEX syndrome(23;24). These children can develop type 1 diabetes in the first days of life and illustrate the importance of T cell regulation. Most other forms of type 1A diabetes are either oligogenic or polygenic in etiology and polymorphisms of genes within the major histocompatibility complex (HLA genes) play a major role in determining disease susceptibility(25;26). Such heterogeneity is also apparent in three spontaneous animal models of type 1A diabetes, the BB rat (Biobreeding), the NOD (Non-obese diabetic) and the Tokushima rat (Figure 4). For all three strains polymorphisms of genes homologous to HLA DR and DQ of man are essential for disease(27). In addition more than 15 other loci play an important role in diabetes susceptibility of the NOD mouse, but each locus contributes relatively little (polygenic inheritance) by itself.

 

 

Figure 4. Three rodent strains that develop immune mediated diabetes. Modified from teaching slides www.barbaradaviscenter.org

 

 

For the BB rat and Tokushima rat there are major loci outside of the MHC contributing to disease (Oligogenic inheritance)(28;29). It is not likely that human type 1 diabetes is less heterogeneous than these few animal models.

 

Identical twins of patients with type 1A diabetes have an overall risk of developing type 1 diabetes of fifty percent. Consistent with heterogeneity, that risk varies dramatically with the diabetic twin’s age of diabetes onset. If the identical twin develops diabetes prior to age 5, the risk for the other twin exceeds 50%. In contrast if the twin develops diabetes after age 25, the risk is less than 10%(27). The risk of developing diabetes is approximately 1/20 for a sibling or offspring of a patient with type 1A diabetes. The U.S. population risk for type 1A diabetes is approximately 1/300 and the country with the highest incidence in the world (Finland) has a risk of approximately 1/100. As will be discussed subsequently genetic polymorphisms greatly influence disease risk. A sibling of a patient with type 1 diabetes who has the highest risk genotype DR3/4 DQB1*0302 has an almost ½ risk of developing anti-islet autoantibodies and progressing to diabetes (DAISY study (30) and unpublished Rewers et al).

Polymorphisms of genes within the major histocompatibility complex contribute to disease of both man and rodent models.

 

 

 

Figure 5. Schematic of the human major histocompatibility complex approximately 4 million base pairs in length. Redondo MJ, et al. Recent Prog Horm Res, 56: 69-89, 2001. Copyright The Endocrine Society.

 

 

The histocompatibility complex is divided into three regions, Class II, class III and class I. The most important determinants of type 1 diabetes are the HLA DQ and DR alleles. These molecules on the surface of antigen presenting cells (e.g. macrophages) bind and present short peptides that are recognized by T cell receptors of T lymphocytes(25;31;32). They are termed immune response genes in that the specific amino acid sequence of these molecules determine which peptides will be bound and to a large extent determine which peptides an individual will respond to. Each different amino acid sequence is given a number. For the DQ molecules both its alpha and beta chain gene are polymorphic and thus to specify a DQ molecule one must specify both chains. For DR molecules only the DRB chain is polymorphic and thus only this chain is specified. Each number after the star indicates a specific amino acid sequence of the HLA allele (Figure 6) and the letters and first number the gene (e.g. DRB1*0401, DR B chain gene number 1, allele 0401).

 

 

Figure 6. Hierarchy of HLA terminology with specific alleles, groups of alleles of different genes on the same chromosome, and finally genotype. Modified from teaching slides www.barbaradaviscenter.org

 

 

The alleles of different HLA genes (e.g. DRB1 and DQB1) are non-randomly associated with each other, such that with DRB1*0401 one usually finds one of three DQ alleles (e.g. DQB1*0301, DQB1*0302, DQB1*0303) rather than any one of more than forty different DQB molecules. Such non-random association of alleles of different genes on the same chromosome is termed linkage dysequilibrium.

 

 

Figure 7. Hierarchy of diabetes risk with examples of haplotypes that lead to diabetes susceptibility, are neutral, or protective. Modified from teaching slides www.barbaradaviscenter.org
There is a tremendous spectrum of diabetes risk associated with different DR and DQ genotypes(33-35). For Caucasians with type 1A diabetes the most common diabetes associated haplotypes are DR3 and DR4 associated. More than 90% of patients with type 1A diabetes have one or both of these alleles versus approximately 40% of the general U.S. population. With the finer sequence information that is now available DR4 haplotypes are subdivided based on specific variants of DRB1 and DQB1. The highest risk DR4 haplotypes have DRB1*0401, DRB1*0402, DRB1*0405, while DRB1*0403 is moderately protective. The highest risk DR4 haplotypes have DQB1*0302, with DQB1*0301 and DQB1*0303 of lower risk. Thus both DR and DQ alleles contribute to diabetes risk. DR3 haplotypes are almost always conserved with DRB1*03 combined with DQA1*0501, DQB1*0201(36). The highest risk genotype have both DR4/DR3 DQB1*0302/DQB1*0201. This genotype occurs in 2.4% of newborns in Denver Colorado and between 30 and 50% of children developing type 1A diabetes. Approximately 50% of children developing type 1A diabetes early (e.g. less than age 5) are DR3/4 heterozygotes versus 30% of young adults presenting with type 1A diabetes.
There are three HLA molecules that appear to provide dominant protection. The most common is DQB1*0602 that occurs in approximately 20% of U.S. individuals(37-39). Protection is not absolute, but less than 1% of children with type 1A diabetes have this molecule. DQA1*0201 with DQB1*0303 and DRB1*1401 also provide dramatic protection, rarely being found in patients with type 1 diabetes and rarely transmitted from a parent with the alleles to their diabetic offspring(34;35). It is noteworthy that both DR and DQ alleles can protect. The specific mechanism underlying both susceptibility and protection are not fully understood. One attractive hypothesis is that protective alleles when expressed within the thymus lead to deletion of T cells with receptors that recognize a critical islet peptide(40). With deletion of such T cells risk of diabetes would be reduced. In addition it is likely that high-risk HLA alleles present specific peptides of target islet molecules to T lymphocytes(26).

 

 

Figure 8. Summary of subset of confirmed loci from whole genome screens associated with type 1A diabetes (from Teaching Slides www.barbaradaviscenter.org) www.barbaradaviscenter.org. Modified from Todd et al. Robust Association of four new chromosomes regions from genome-wide analyses of type 1 diabetes, Nature Genetics, June 6, 2007.

 

 

Multiple genetic loci contributing to diabetes risk have been implicated (Figure 8). Polymorphisms of the insulin gene are well established as contributing to risk. A repeat sequence upstream (5’) of the insulin gene termed a Variable nucleotide tandem repeat or VNTR, is divided into three general repeat sizes with the longest set of repeats associated with protection from diabetes(41-43). This set of alleles is also associated with greater thymic production of insulin messenger RNA(44), leading to the hypothesis that greater thymic message and presumably greater proinsulin production dampens anti-insulin autoimmunity(44-46). Recently a functional polymorphism of the LYP gene (Lymphocyte Specific Phosphatase; PTPN22- Protein Tyrosine Phosphatase) has been associated wih type 1 diabetes, rheumatoid arthritis, and lupus erythematosus (47-49). The R620W missense mutation (trypophan replacing arginine) disrupts the binding of the phosphatase to the molecule Csk and this blocks its ability to down-regulate T cell receptor signaling. With an odds ratio of between 1.7 and 2.0 of the “autoimmunity” allele which is relatively common (5-10% allele frequency) there is a large multiplicative genetic effect, which is much greater than CTLA-4 polymorphisms associated with diabetes risk(50). Combining known diabetogenic polymorphisms of LYP, the insulin gene, alleles of DP, DQ, and DR class II immune response genes, as well all of the new loci account for approximately 48% of the familial aggregation of type 1A diabetes, with DR and DQ loci accounting for 41% of this 48%(Todd JA, Walker NM, Cooper JD, Smyth DJ, Downes K, Plagnol V et al. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet 2007; 39(7):857-864).  A recent study suggests that for a major subset of individuals with the highest risk HLA genotype (DR3/4-DQ2/DQ8 heterozygotes) who share both HLA haplotypes with a diabetic sibling, risk of activating anti-islet autoimmunity is as high as 80%.  This strongly suggests that other polymorphisms within or linked to the HLA region remain to be defined(Aly TA, Ide A, Jahromi MM, Barker JM, Fernando MS, Babu SR et al. Extreme Genetic Risk for Type 1A Diabetes. Proc Natl Acad Sci USA 2006; 103(38):14074-14079).

 

Multiple additional loci (Figure 8) have been implicated with estimates that approximately 50% of the familial aggregation of type 1 diabetes is attributable to the HLA region, perhaps 10% to the insulin locus, with all other loci contributing much less, though in aggregate their contribution is important. In the Cox analysis (figure 8) of approximately 700 sibling pairs the only significant LOD score was for a locus on chromosome 16q that was not given an iddm designation with earlier genome screens. Several areas implicated in the past had suggestive scores, but there is overlap with the families from which the original evidence was generated. It is likely that contributing loci may differ between populations contributing to the difficulty of replicating putative loci in different studies. There also may be marked heterogeneity, and for instance IDDM17 was implicated in the study of a single Bedouin Arab family where the locus segregates as a major gene(51). Heterogeneity may also explain the very weak signals for these non-MHC loci. In general such genome screens have failed to identify new genes and likely produce more false positives than relevant loci. Emphasis to association studies with candidate genes such as the LYP polymorphism, and with a detailed haplotype map may foster further genetic analysis(52).

 

 

STAGE II: TRIGGERING

Anti-islet autoimmunity (e.g. insulin autoantibodies), insulitis and immune mediated diabetes can be triggered in a number of animal models by a large number of immunologic and genetic manipulations(53-55). Perhaps the most relevant of these manipulations is the administration of poly-IC (poly inosinic cytodylic acid) to normal rat strains that have the diabetes susceptible major histocompatibility RT1-U haplotype(56). Administration of poly-IC in some of these normal strains leads to insulitis while in others it leads to overt diabetes with islet beta cell destruction. Poly-IC interacts with Toll 3 receptors of the innate immune system leading to a cascade of intracellular and cytokine mediated events. Infection of Diabetes Resistant BB rats, a strain of rats that does not develop diabetes and lacks the lymphopenia gene of the spontaneously diabetic BB rat strain, leads to diabetes probably by a pathogenic mechanism similar to the effects of poly-IC(57). Poly-IC is a mimic of viral double stranded RNA and thus it is easy to envision that many common RNA viral infections may induce diabetes in genetically susceptible patients. Normal mouse strains such as Balb/c mice rapidly develop insulin autoantibodies if challenged with an insulin peptide (B chain peptide, amino acids 9 to 23)(53). If the peptide is administered with poly-IC, insulitis is induced and in genetically susceptible mice diabetes can be induced. These and many other studies in animal models indicate that normal animals harbor autoreactive B and T lymphocytes that can be expanded and activated, with resultant diabetes. Though these strains are “normal” they have variants of MHC molecules that determine disease susceptibility, by influencing T cell responses to relevant peptides. The class II MHC molecules (equivalent of DR and DQ of man) appear to be most important and these molecules probably influence disease either by the peptides they bind and present to T cells within islets and draining lymph nodes and by their influence on the thymic T cell repertoire(58).

 

In man environmental factors that trigger anti-islet autoimmunity are largely unknown. Congenital rubella infection is associated with a risk of type 1 diabetes that exceeds 1/5(59;60). It is however only congenital infection that increases the risk of diabetes, and leads to a series of autoimmune disorders (e.g. thyroid autoimmunity). It is thus likely that the congenital infection damages the developing immune system leading to relatively broad disease susceptibility(61).

 

Figure 9 lists a number of additional environmental factors that may impact on the development of type 1A diabetes. The enteroviruses are probably the most extensively studied(62;63). Investigators have associated for instance antibodies to Coxsackie viruses and Coxsackie viral RNA with type 1A diabetes(64). An important study from Finland has provided

 

 

 

Figure 9. Environmental factors that may increase or decrease diabetes risk. Congenital rubella infection is bolded as it is the only clearly defined factor.

 

 

 

evidence that enterovirus infection may be associated with the activation of anti-islet autoimmunity as measured by the appearance of anti-islet autoantibodies. Such studies have been difficult to replicate and the DAISY study from Denver Colorado that also follows newborns from birth for evidence of anti-islet autoimmunity has not found an association with the enteroviral infection(63).

 

 

Figure 10. Secular trend in the incidence of type 1A diabetes in Finland subdivided by the age of appearance of diabetes. Modified from teaching slides www.barbaradaviscenter.org

 

 

Dietary factors that may contribute to diabetes are being extensively studied. One hypothesis is that bovine milk ingestion, particularly in the first months of life is associated with development of diabetes(65). Again there is conflicting data with reports that bovine milk ingestion increases the development of anti-islet autoantibodies and studies from Denver, Munich, and Melbourne, that it does not(66). Recently two studies (Germany and Denver) have implicated early (<3 months) introduction of cereals as a risk factor for type 1 diabetes(67;68). In the Denver study late introduction (>7 months) also inceased risk. At present it is my view that the data does not allow any firm recommendation in terms of changing infant diet, except to emphasize the current standard recommendation for introduction of cereals between 3 and 7 months..

 

Environmental factors might not only increase the development of diabetes but may also provide protection. There is a very wide range in the risk of type 1 diabetes ranging from an annual incidence of less than 1/100,000 in China to approximately 50/100,000 in Finland(69). Much of this difference may relate to genetic factors but there is strong evidence that environmental factors are influencing diabetes risk. The strongest evidence comes from a marked secular trend in terms of increasing diabetes incidence in multiple populations(70). As shown in figure 10, the incidence has increased dramatically particularly for children developing diabetes prior to age 5, increasing more than 3 fold over the past three decades. Such a rapid change in disease incidence cannot be due to changes in gene pool. Something that increases risk has been added or more likely an environmental factor that decreases risk, has been removed from the population. With increasing public health, a “hygiene” hypothesis has been advanced, particularly directed at asthma and type 1 diabetes(71). It is hypothesized that as the environment becomes “cleaner” the normal development of the immune system is disrupted (e.g. regulatory T cell development is subnormal) resulting in increases of both presumed Th2 (asthma) and Th1 (Type 1 diabetes) mediated diseases. For instance, one review discusses decreasing pinworm infection as a potential factor.

 

 

STAGE III AUTOIMMUNITY

The assays for anti-islet autoantibodies have improved remarkably over the past three decades(72). A series of anti-islet autoantibody workshops where sera is sent blinded to multiple laboratories throughout the world has stimulated assay improvements and standardization(62). Such workshops have evaluated not only anti-islet autoantibodies of man but also of the NOD mouse model of type 1 diabetes. In the mouse model the only specific autoantibody detected reacted with insulin(73) and similar to studies in man fluid phase radioassays provided better sensitivity and specificity compared to ELISA assays.

The autoantibodies that are primarily measured in man react with insulin, glutamic acid decarboxylase 65, and ICA512 (IA-2) (Figure 11). GAD67 autoantibodies are primarily a subset of antibodies that cross-react with GAD65, and similarly IA-2beta antibodies are predominantly a subset of ICA512 autoantibodies.

 

 

 

Figure 11. Five characterized islet autoantigens with specific/sensitive autoantibody assays. Modified from teaching slides www.barbaradaviscenter.org

 

 

 

There are a number of important caveats in the utilization of anti-islet autoantibody assays. The field developed from the initial observation that patient’s sera “stained” islets of cut sections of human pancreas, the cytoplasmic islet cell antibody (ICA) assay(74). This assay, given its utilization of human pancreas from cadaveric donation and subjective reading of slides, has proven the most difficult to standardize(62). The assay predominantly detects antibodies reacting with GAD65 and ICA512, but does not detect anti-insulin autoantibodies. Given the difficulty in standardization, reliability over time, and major overlap with defined autoantibody assays, a number of investigators no longer utilize this assay. For research purposes and potentially in older adults with what has been termed LADA (latent autoimmune diabetes of adults) the ICA assay may have utility in that there is evidence of one or more additional autoantibodies detected with this assay and not with GAD65, ICA512 and insulin autoantibody determination.

 

Insulin autoantibodies are usually the first autoantibody to appear in children followed from birth for the development of type 1A diabetes(75;76). These autoantibodies can appear in the first six months of life. Once insulin autoantibodies appear in such young children there is a high risk of development of additional anti-islet autoantibodies and progression to diabetes. More than 90% of children developing type 1A diabetes prior to age 5 have insulin autoantibodies while less than 50% of children developing diabetes after age 12 have such autoantibodies(77). Therapy with human insulin induces insulin antibodies that cannot at present be distinguished from insulin autoantibodies. Thus if an individual has been treated with insulin for more than several weeks, positive insulin autoantibodies are not interpretable. For all autoantibodies measured in the first 9 months of life, the antibodies may be transplacental in origin, a particular problem if a mother has type 1 diabetes and is treated with insulin.

 

 

 

Figure 12. Progression to diabetes of first degree relatives of patients with type 1 diabetes subdivided by the number of autoantibodies expressed (of GAD65, ICA512, and insulin). Adapted from Verge et al, Diabetes 1996 (78)

 

 

A single autoantibody, even when present on multiple occasions is associated with only a modest risk of progression to diabetes of approximately 20%(78;79). Once two or more anti-islet autoantibodies are present (of GAD65, ICA512, or insulin) progression to diabetes is very high, exceeding 75% with a decade of follow up. In addition once multiple autoantibodies are present it is very unusual for an individual to lose all expression of autoantibodies prior to the development of overt diabetes. Following the development of diabetes, ICA512 and more slowly GAD65 (over decades) autoantibodies wane. Following islet or pancreatic transplantation expression of GAD65 and ICA512 autoantibodies can be induced in patients with long-standing diabetes(80).

 

The most specific of the autoantibodies react with the molecule ICA512, but ICA512 autoantibodies are usually detected following the appearance of insulin and/or GAD65 autoantibodies(75). Even with ICA512 autoantibodies however there are apparent “false” positives in terms of diabetes risk. We evaluated approximately 10 individuals with either transient ICA512 autoantibodies or normal controls with ICA512 autoantibodies. None of these individuals expressed an additional anti-islet autoantibody. In contrast to patients with or developing type 1 diabetes, the ICA512 autoantibodies of 9/10 of these normal individuals did not recognize multiple ICA512 epitopes and did not react with the dominant ICA512 autoantigenic domain(81). This indicates that even with a highly specific radioassay if one screens tens of thousands of sera, one can find sera that presumably by chance cross-react with some epitope of the ICA512 molecule. It is much less likely to find an individual with antibodies that by chance react with two different islet autoantigens using fluid phase radioassays set with specificity at the 99th percentile of controls.

 

Not all individuals with two or more autoantibodies are destined to progress to type 1 diabetes. For instance the diabetes risk is unknown for individuals with expression of two or more anti-islet autoantibodies with the protective HLA molecule DQA1*0102, DQB1*0602. Figure 13 is a life table of progression to diabetes of first degree relatives with high titer cytoplasmic autoantibodies with a subset of relatives having the protective HLA allele DQB1*0602. Two of the DQB1*0602 relatives expressed multiple “biochemical” anti-islet autoantibodies (one with two autoantibodies and the other three) and neither of these individuals has yet progressed to diabetes. In that approximately 1% of patients with type 1 diabetes have DQB1*0602 it is possible that these individuals will eventually progress to diabetes.

 

 

 

Figure 13. Lack of progression to diabetes of cytoplasmic ICA positive first-degree relatives that have the protective HLA allele DQB1*0602. Modified from teaching slides www.barbaradaviscenter.org

 

 

 

STAGE IV. LOSS OF INSULIN SECRETION

There are three general hypotheses in terms of progression to type 1A diabetes (Figure 14). In that at present beta cell mass is not readily measured over time in man it is not possible to absolutely define progression of beta cell loss. There is however no doubt that measurable anti-islet autoimmunity precedes the development of diabetes in terms of anti-islet autoantibodies in man, and autoantibodies and T cell invasion in animal models. In the NOD mouse there is evidence of some beta cell destruction and beta cell regeneration prior to the onset of diabetes (82). There is also evidence for a change in the immune system close to the time of onset of diabetes (e.g. Th2 to Th1) (83-86). This change is associated with more rapid disease progression, ability to transfer diabetes by T cells, and a time window during which a specific immunotherapy (monoclonal anti-CD3 antibodies) is effective(87). In man the best evidence for progressive loss of beta cell function comes from studies of insulin and C-peptide secretion(88). C-peptide the connecting peptide of proinsulin is secreted in equimolar amount to insulin, but C-peptide is not present in insulin preparations utilized to treat diabetes. Thus C-peptide has become an important indicator of remaining beta cell function. Following the onset of diabetes it has long been appreciated that C-peptide secretion progressively declines, until for most patients with type 1 diabetes C-peptide becomes non-detectable, associated with true insulin dependence. In a similar manner first phase insulin secretion following a bolus of glucose on intravenous glucose tolerance testing is progressively lost for relatives followed to the development of type 1 diabetes(89). Such metabolic abnormalities may result in part from functional inhibition of beta cell secretion, but pathologic studies indicate that beta cell mass is normal for identical twins of patients that have not activated anti-islet autoimmunity, and for new onset patients that bulk of beta cells are destroyed(90). Within the pancreas of a patient with type 1 diabetes there is heterogeneity of islet lesions, with most islets lacking all beta cells and with no lymphocytic infiltrates (pseudoatrophic islets), few normal islets with no infiltrates, and few islets with remaining beta cells and having infiltrates. This is perhaps analogous to the progressive development of vitiligo in patients, with patches of skin with all melanocytes destroyed, whereas other skin is normal.

 

 

 

Figure 14. Three hypotheses of progression to type 1A diabetes. Modified from teaching slides www.barbaradaviscenter.org

 

 

 

STAGE V. OVERT DIABETES

The development of type 1 diabetes is usually perceived as an abrupt event, and some individuals may rapidly manifest severe hyperglycemia. With the ability to now follow individuals to the development of type 1A diabetes it is apparent that anti-islet autoantibodies can precede hyperglycemia by years and there is usually some deterioration in glucose tolerance more than one year prior to diabetes onset (particularly with intravenous glucose tolerance testing)(91). The majority of individuals identified to be diabetic following autoantibody testing are found to have a diabetic 2-hour glucose on oral glucose tolerance testing (>200mg%) rather than fasting hyperglycemia. The acute presentation with severe hyperglycemia and ketoacidosis is life threatening and it is estimated that approximately 1/200 children die at the onset of type 1 diabetes(92;93). Such children typically have a medical history where the first health care providers have failed to make the diagnosis of diabetes, the child then presents again later and dies with cerebral edema. The classic symptoms of polyuria, polydipsia, and weight loss are usually present but the initial diagnosis is still missed. The alternative diagnosis of nausea and vomiting due to viral illness is the most common mistaken diagnosis and with the ready availability of glucose determination from a finger or heel stick, there should be a low threshold in emergency rooms and physicians offices for ruling out diabetes. I am also aware of at least one child with hyperglycemia thought to be illness induced who died several weeks later without any therapy. Though transient hyperglycemia can occur, such children obviously need close follow up. We usually arrange glucose monitoring for children thought to have transient hyperglycemia, and measure anti-islet autoantibodies(94). Of those with anti-islet autoantibodies and transient hyperglycemia, almost all progress to type 1 diabetes within several months.

 

A recent report by Barker and coworkers illustrates the potential importance of screening for anti-islet autoantibodies and monitoring high risk children. Less than 5% of children in such intensive follow-up who developed diabetes were hospitalized with ketoacidosis compared to 40% of children not being screened. Figure illustrates the blood glucose for the two groups at the time of diagnosis with markedly elevated levels in the non-screened children, including values greater than 1,000 mg% associated with severe metabolic decompensation(95).

 

 

 

Figure 15. Glucose at diagnosis in children screened for anti-islet autoantibodies and metabolically monitored, versus children from the general population presenting with diabetes(95). Modified from teaching slides www.barbaradaviscenter.org of Type 1 Diabetes: Molecular, Cellular and Clinical Immunology.

 

 

 

SUMMARY

At the onset of type 1 diabetes almost all individuals have residual insulin secretion and there is convincing evidence that residual insulin secretion as measured by C-peptide secretion is of clinical benefit (less hypoglycemia, less microvascular complications and much easier diabetes management). With many immunologic therapies it is possible to prevent diabetes in animal models and as reviewed, the disorder is predictably in man. At present there is no proven therapy to either prevent progression to type 1 diabetes or to halt beta cell loss after presentation with diabetes. One arm of the DPT-1 prevention trial, where low doses of subcutaneous insulin were administered, did not delay progression to diabetes(96). The DPT-1 trial of oral insulin indicated that overall, oral insulin did not delay progression to diabetes(C).  A subset analysis of antibody positive relatives entering the trial with elevated levels of insulin autoantibodies however, suggests a delay in progression to diabetes of approximately 4 years.  A repeat trial of oral insulin for diabetes prevention is underway by Trialnet.( Effects of Oral Insulin in Relatives of Patients With Type 1 Diabetes: The Diabetes Prevention Trial-Type 1. Diab care 2005; 28(5):1068-1076.)  A study of anti-CD3 monoclonal antibody modified to reduce cytokine release has shown promise in patients with new onset diabetes(97). The National Institutes of Health has created a cooperative trial network to develop therapies to prevent beta cell loss (in prediabetics and new onset patients), termed TrialNet. Relatives of type 1 patients throughout North America can be screened for diabetes risk and trials are underway to preserve beta cell function in new onset patients. A contact number for TrialNet is 1-800-HALT-DM1. In addition the Immune Tolerance Network is evaluating therapies in a series of disorders designed to restore “tolerance”, including type 1A diabetes. They are seeking innovative therapies with a web site for applications (immunetolerance.org).

 

Acknowledgments: Our research is supported by Grants DK-32083, DK-32493, AI39213, DK55969, DK62718, AI50864, AI95380, DK50970, AI46374, MO1RR00069 (Clinical Research Centers Program), Diabetes Endocrine Research Center (P30 DK57516), Autoimmunity Center of Excellence, U19 AI46374 and Autoimmunity Prevention Center from the National Institute of Health, Clinical Research Centers at Children’s Hospital (RR00069) and University Hospital (M01 RR00051), the Juvenile Diabetes Foundation, the American Diabetes Association, and a grant from the Children's Diabetes Foundation at Denver

 

 

(1) Devendra D, Liu E, Eisenbarth GS. Type 1 diabetes: recent developments. Brit Med J 2004; 328(7442):750-754.

(2) Pugliese A. Genetics of type 1 diabetes. Endocrinol Metab Clin North Am 2004; 33(1):1-16, vii.

(3) Tree TIM, Peakman M. Autoreactive T cells in human type 1 diabetes. Endocrinology and Metabolism Clinics of North America 2004; 33(1):113-+.

(4) Daaboul J, Schatz D. Overview of prevention and intervention trials for type 1 diabetes. Rev Endocr Metab Disord 2003; 4(4):317-323.

(5) Wong FS, Wen L. The study of HLA class II and autoimmune diabetes. Curr Mol Med 2003; 3(1):1-15.

(6) Lorenzen T, Pociot F, Hougaard P, Nerup J. Long-term risk of IDDM in first-degree relatives of patients with IDDM. diabetol 1994; 37:321-327.

(7) Janzon L, Bergentz SE, Ericsson BF, Lindell SE. The arm-ankle pressure gradient in relation to cardiovascular risk factors in intermittent claudication. Circulation 1981; 63(6):1339-1341.

(8) Zimmet P, Turner R, McCarty D, Rowley M, Mackay I. Crucial points at diagnosis - Type 2 diabetes or slow type 1 diabetes. Diab care 1999; 22:B59-B64.

(9) Pinhas-Hamiel O, Dolan LM, Daniels SR, Standiford D, Khoury PR, Zeitler P. Increased incidence of non-insulin dependent diabetes mellitus among adolescents. J Pediatr 128, 608-615. 1996.

(10) Pinhas-Hamiel O, Zeitler P. Type 2 diabetes in adolescents, no longer rare. Pediatr Rev 1998; 19(12):434-435.

(11) Winter WE, Maclaren NK, Riley WJ, Clarke DW, Kappy MS, Spillar RP. Maturity-onset diabetes of youth in Black Americans. N Engl J Med 1987; 316(6):285-291.

(12) Sussel L, Eisenbarth GS. From bedside to bench and back: The tale of MODY diabetes. Trends in Endocrinology & Metabolism 2001; 12:277-278.

(13) Bao F, Yu L, Babu S, Wang T, Hoffenberg EJ, Rewers M et al. One third of HLA DQ2 homozygous patients with type 1 diabetes express celiac disease associated transglutaminase autoantibodies. J Autoimmunity 1999; 13:143-148.

(14) Hoffenberg EJ, Bao F, Eisenbarth GS, Uhlhorn C, Haas J, Sokol RJ et al. Transglutaminase antibodies in children with a genetic risk for celiac disease. J Pediatr 2000; 137(3):356-360.

(15) Yu L, Brewer KW, Gates S, Wang T, Babu S, Gottlieb PA et al. DRB1*04 and DQ alleles: expression of 21-hydroxylase autoantibodies and risk of progression to Addison's disease. J Clin Endocrinol Metab 1999; 84(1):328-335.

(16) Atkinson MA, Eisenbarth GS. Type 1 Diabetes: New Perspectives on Disease Pathogenesis and Treatment. Lancet 2001; 358(9277):221-229.

(17) Eisenbarth GS. Type I diabetes mellitus. A chronic autoimmune disease. N Engl J Med 1986; 314:1360-1368.

(18) American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diab care 2004; 27 Suppl 1:S5-S10.:S5-S10.

(19) Imagawa A, Hanafusa T, Miyagawa J, Matsuzawa Y. A novel subtype of type 1 diabetes mellitus characterized by a rapid onset and an absence of diabetes-related antibodies. Osaka IDDM Study Group. New Engl J Med 2000; 342:301-307.

(20) Robles DT, Eisenbarth GS, Ikegami H, Pugliese A, Dotta F, She J-X et al. Endocrinology and Metabolism Clinics of North America. Philadelphia: W.B. Saunders Company, 2002.

(21) Patel DD. Escape from tolerance in the human X-linked autoimmunity-allergic disregulation syndrome and the Scurfy mouse. J Clin Invest 2001; 107(2):155-157.

(22) Bjorses P, Aaltonen J, Horelli-Kuitunen N, Yaspo ML, Peltonen L. Gene defect behind APECED: a new clue to autoimmunity. Hum Mol Genet 1998; 7(10):1547-1553.

(23) Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003; 4(4):330-336.

(24) Owen CJ, Jennings CE, Imrie H, Lachaux A, Bridges NA, Cheetham TD et al. Mutational analysis of the FOXP3 gene and evidence for genetic heterogeneity in the immunodysregulation, polyendocrinopathy, enteropathy syndrome. J Clin Endocrinol Metab 2003; 88(12):6034-6039.

(25) Klein J, Sato A. The HLA system. First of two parts. N Engl J Med 2000; 343(10):702-709.

(26) Lee KH, Wucherpfennig KW, Wiley DC. Structure of a human insulin peptide/HLA-DQ8 complex and susceptibility to type 1 diabetes. Nature Immunology 2001; 2:501-507.

(27) Mordes JP, Greiner DL, Rossini AA. Animal models of autoimmune diabetes mellitus. In: LeRoith D, Taylor SI, Olefsky JM, editors. Diabetes mellitus: a fundamental and clinical text. Philadelphia: Lippincott-Raven, 1996: 349-360.

(28) Martin AM, Maxson MN, Leif J, Mordes JP, Greiner DL, Blankenhorn EP. Diabetes-prone and diabetes-resistant BB rats share a common major diabetes susceptibility locus, iddm4: additional evidence for a "universal autoimmunity locus" on rat chromosome 4. diab 1999; 48(11):2138-2144.

(29) Yu B, Gauthier L, Hausmann DH, Wucherpfennig KW. Binding of conserved islet peptides by human and murine MHC class II molecules associated with susceptibility to type I diabetes [In Process Citation]. Eur J Immunol 2000; 30(9):2497-2506.

(30) Rewers M, Bugawan TL, Norris JM, Blair A, Beaty B, Hoffman M et al. Newborn screening for HLA markers associated with IDDM: diabetes autoimmunity study in the young (DAISY). diabetol 1996; 39(7):807-812.

(31) Kwon OJ, Brautbar C, Weintrob N, Sprecher E, Saphirman C, Bloch K et al. Immunogenetics of HLA class II in Israeli Ashkenazi Jewish, Israeli non-Ashkenazi Jewish, and in Israeli Arab IDDM patients. Hum Immunol 2001; 62(1):85-91.

(32) Undlien DE, Lie BA, Thorsby E. HLA complex genes in type 1 diabetes and other autoimmune diseases. Which genes are involved? Trends Genet 2001; 17(2):93-100.

(33) Lie BA, Ronningen KS, Akselsen HE, Thorsby E, Undlien DE. Application and interpretation of transmission/disequilibrium tests: transmission of HLA-DQ haplotypes to unaffected siblings in 526 families with type 1 diabetes. Am J Hum Genet 2000; 66(2):740-743.

(34) Redondo MJ, Kawasaki E, Mulgrew CL, Noble JA, Erlich HA, Freed BM et al. DR and DQ associated protection from type 1 diabetes: comparison of DRB1*1401 and DQA1*0102-DQB1*0602. J Clin Endocrinol Metab 2000; 85(10):3793-3797.

(35) Kawasaki E, Noble J, Erlich H, Mulgrew CL, Fain PR, Eisenbarth GS. Transmission of DQ haplotypes to patients with type 1 diabetes. diab 1998; 47(12):1971-1973.

(36) Stewart CA, Horton R, Allcock RJ, Ashurst JL, Atrazhev AM, Coggill P et al. Complete MHC haplotype sequencing for common disease gene mapping. Genome Res 2004; 14(6):1176-1187.

(37) Baisch JM, Weeks T, Giles R, Hoover M, Stastny P, Capra JD. Analysis of HLA-DQ genotypes and susceptibility in insulin- dependent diabetes mellitus. N Engl J Med 1990; 322(26):1836-1841.

(38) Pugliese A, Gianani R, Moromisato R, Awdeh ZL, Alper CA, Erlich HA et al. HLA-DQB1*0602 is associated with dominant protection from diabetes even among islet cell antibody-positive first-degree relatives of patients with IDDM. diab 1995; 44(6):608-613.

(39) Pugliese A, Kawasaki E, Zeller M, Yu L, Babu S, Solimena M et al. Sequence analysis of the diabetes-protective human leukocyte antigen-DQB1*0602 allele in unaffected, islet cell antibody-positive first degree relatives and in rare patients with type 1 diabetes. J Clin Endocrinol Metab 1999; 84(5):1722-1728.

(40) Schmidt D, Verdaguer J, Averill N, Santamaria P. A mechanism for the major histocompatibility complex-linked resistance to autoimmunity. J Exp Med 1997; 186:1059-1075.

(41) Bell GI, Horita S, Karam JH. A polymorphic locus near the human insulin gene is associated with insulin-dependent diabetes mellitus. diab 1984; 33:176-183.

(42) Undlien DE, Bennett ST, Todd JA, Akselsen HE, Ikaheimo I, Reijonen H et al. Insulin gene region-encoded susceptibility to IDDM maps upstream of the insulin gene. diab 1995; 44:620-625.

(43) Barratt BJ, Payne F, Lowe CE, Hermann R, Healy BC, Harold D et al. Remapping the Insulin Gene/IDDM2 Locus in Type 1 Diabetes. diab 2004; 53(7):1884-1889.

(44) Pugliese A, Brown D, Garza D, Zeller M, Redondo MJ, Eisenbarth GS et al. Self-Antigen Presenting Cells Expressing Islet Cell Molecules in Human Thymus and Peripheral Lymphoid Organs: Phenotypic Characterization and Implications for Immunological Tolerance and Type 1 Diabetes. J Clin Invest 2001; 107(5):555-564.

(45) Pugliese A, Zeller M, Fernandez A, Zalcberg LJ, Bartlett RJ, Ricordi C et al. The insulin gene is transcribed in the human thymus and transcription levels correlate with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type I diabetes. Nat Genet 1997; 15:293-297.

(46) Bennet ST, Wilson AJ, Esposito L, Bouzekri J, Undlien D, Cucca F et al. Insulin VNTR allele-specific effect in type 1 diabetes depends on identity of untransmitted paternal allele. Nat Genet 1997; 17:350-352.

(47) Bottini N, Muscumeci L, Alonso A, Rahmouni S, Nika K, Rostamkhani M et al. A functional variant of lympoid tyrosine phosphatase is associated with type I diabetes. Nature Genetics 36, 337-338. 2004.

Ref Type: Journal (Full)

(48) Kyogoku C, Langefeld CD, Ortmann WA, Lee A, Selby S, Carlton VE et al. Genetic Association of the R620W Polymorphism of Protein Tyrosine Phosphatase PTPN22 with Human SLE. Am J Hum Genet 2004; 75(3):504-507.

(49) Begovich AB, Carlton VE, Honigberg LA, Schrodi SJ, Chokkalingam AP, Alexander HC et al. A Missense Single-Nucleotide Polymorphism in a Gene Encoding a Protein Tyrosine Phosphatase (PTPN22) Is Associated with Rheumatoid Arthritis. Am J Hum Genet 2004; 75(2).

(50) Ueda H, Howson JM, Esposito L, Heward J, Snook H, Chamberlain G et al. Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 2003; 423:506-511.

(51) Verge CF, Vardi P, Babu S, Bao F, Erlich HA, Bugawan T et al. Evidence for oligogenic inheritance of type 1A diabetes in a large Bedouin Arab family. J Clin Invest 1998; 102(8):1569-1575.

(52) Walsh EC, Mather KA, Schaffner SF, Farwell L, Daly MJ, Patterson N et al. An integrated haplotype map of the human major histocompatibility complex. Am J Hum Genet 2003; 73(3):580-590.

(53) Abiru N, Maniatis AK, Yu L, Miao D, Moriyama H, Wegmann D et al. Peptide and MHC specific breaking of humoral tolerance to native insulin with the B:9-23 peptide in diabetes prone and normal mice. diab 2001; 50:1274-1281.

(54) Heath VL, Hutchings P, Fowell DJ, Cooke A, Mason DW. Peptides derived from murine insulin are diabetogenic in both rats and mice, but the disease-inducing epitopes are different: evidence against a common environmental cross-reactivity in the pathogenicity of type 1 diabetes. diab 1999; 48(11):2157-2165.

(55) Wen L, Chen NY, Tang J, Sherwin R, Wong FS. The regulatory role of DR4 in a spontaneous diabetes DQ8 transgenic model. J Clin Invest 2001; 107(7):871-880.

(56) Ellerman KE, Like AA. Susceptibility to diabetes is widely distributed in normal class IIu haplotype rats. diabetol 2000; 43(7):890-898.

(57) Ellerman KE, Richards CA, Guberski DL, Shek WR, Like AA. Kilham rat virus triggers T-cell-dependent autoimmune diabetes in multiple strains of rat. diab 1996; 45:557-562.

(58) Wucherpfennig KW, Eisenbarth GS. Type 1 Diabetes. Nature Immunology 2001; 2(7):1-3.

(59) Ginsberg-Fellner F, Witt ME, Yagihashi S, Doberson MJ, Taub F, Fedun B et al. Congenital rubella syndrome as a model for type I (insulin- dependent) diabetes mellitus: increased prevalence of islet cell surface antibodies. diabetol 1984; 27 Suppl:87-89.

(60) Shaver KA, Boughman JA, Nance WE. Congenital rubella syndrome and diabetes: A review of epidemiologic, genetic, and immunologic factors. Am Ann Deaf 1985; 130:526-532.

(61) Rabinowe SL, George KL, Loughlin R, Soeldner JS, Eisenbarth GS. Congenital rubella. Monoclonal antibody-defined T cell abnormalities in young adults. Am J Med 1986; 81(5):779-782.

(62) Lonnrot M, Korpela K, Knip M, Ilonen J, Simell O, Korhonen S et al. Enterovirus infection as a risk factor for beta-cell autoimmunity in a prospectively observed birth cohort: the Finnish Diabetes Prediction and Prevention Study. diab 2000; 49(8):1314-1318.

(63) Graves PM, Rewers M. The role of enteroviral infections in the development of IDDM: limitations of current approaches. diab 1997; 46(2):161-168.

(64) Rewers M, Norris JM. Epidemiology of type I diabetes. In: Eisenbarth GS, Lafferty KJ, editors. Type I Diabetes: Molecular, Cellular, and Clinical Immunology. New York: Oxford University Press, 1996: 172-208.

(65) Martin JM, Trink B, Daneman D, Dosch HM, Robinson B. Milk proteins in the etiology of insulin-dependent diabetes mellitus (IDDM). Ann Med 1998; 23:447-452.

(66) Norris JM, Beaty B, Klingensmith G, Yu L, Hoffman M, Chase HP et al. Lack of association between early exposure to cow's milk protein and b-cell autoimmunity: Diabetes Autoimmunity Study in the Young (DAISY). JAMA 1996; 276:609-614.

(67) Norris JM, Barriga K, Klingensmith G, Hoffman M, Eisenbarth GS, Erlich H et al. Timing of cereal exposure in infancy and risk of islet autoimmunity. The Diabetes Autoimmunity Study in the Young (DAISY). JAMA 2003; 290:1713-1720.

(68) Ziegler AG, Schmid S, Huber D, Hummel M, Bonifacio E. Early infant feeding and risk of developing type 1 diabetes-associated autoantibodies. JAMA 2003; 290(13):1721-1728.

(69) Yang Z, Wang K, Li T, Sun W, Li Y, Chang YF et al. Childhood diabetes in China. Enormous variation by place and ethnic group. Diab care 1998; 21(4):525-529.

(70) Onkamo P, Vaananen S, Karvonen M, Tuomilehto J. Worldwide increase in incidence of Type I diabetes--the analysis of the data on published incidence trends. diabetol 1999; 42(12):1395-1403.

(71) Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med 2002; 347(12):911-920.

(72) Kawasaki E, Eisenbarth G. High-Throughput Radioassays for Autoantibodies to Recombinant Autoantigens. Frontiers in Bioscience 2000; 5:181-190.

(73) Bonifacio E, Atkinson M, Eisenbarth GS, Serreze D, Kay TW, Lee-Chan E et al. International workshop on autoimmunity in animal models of autoimmune diabetes identifies insulin, but not GAD or IA-2 as specific antigens of humoral autoimmunity in the non-obese diabetic mouse. diab 2001; 50:2451-2458.

(74) Bottazzo GF, Florin-Christensen A, Doniach D. Islet-cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies. Lancet 1974; 2(7892):1279-1283.

(75) Bonifacio E, Scirpoli M, Kredel K, Fuchtenbusch M, Ziegler AG. Early autoantibody responses in prediabetes are IgG1 dominated and suggest antigen-specific regulation. J Immunol 1999; 163(1):525-532.

(76) Yu L, Robles DT, Abiru N, Kaur P, Rewers M, Kelemen K et al. Early expression of antiinsulin autoantibodies of humans and the NOD mouse: evidence for early determination of subsequent diabetes. Proc Natl Acad Sci USA 2000; 97(4):1701-1706.

(77) Vardi P, Ziegler AG, Matthews JH, Dib S, Keller RJ, Ricker AT et al. Concentration of insulin autoantibodies at onset of type I diabetes. Inverse log-linear correlation with age. Diab care 1988; 11(9):736-739.

(78) Verge CF, Gianani R, Kawasaki E, Yu L, Pietropaolo M, Jackson RA et al. Prediction of type I diabetes in first-degree relatives using a combination of insulin, GAD, and ICA512bdc/IA-2 autoantibodies. diab 1996; 45(7):926-933.

(79) Bingley PJ, Bonifacio E, Williams AJK, Genovese S, Bottazzo GF, Gale EAM. Prediction of IDDM in the general population: strategies based on combinations of autoantibody markers. diab 1997; 46:1701-1710.

(80) Bosi E, Braghi S, Maffi P, Scirpoli M, Bertuzzi F, Pozza G et al. Autoantibody response to islet transplantation in type 1 diabetes. diab 2001; 50(11):2464-2471.

(81) Clark CM, Jr. How should we respond to the worldwide diabetes epidemic? [editorial; comment]. Diab care 1998; 21(4):475-476.

(82) Sreenan S, Pick AJ, Levisetti M, Baldwin AC, Pugh W, Polonsky KS. Increased b-cell proliferation and reduced mass before diabetes onset in the nonobese diabetic mouse. diab 1999; 48:989-996.

(83) Gazda LS, Charlton B, Lafferty KJ. Diabetes results from a late change in the autoimmune response of NOD mice. J Autoimmun 1997; 10:261-270.

(84) Dilts SM, Lafferty KJ. Autoimmune diabetes: the involvement of benign and malignant autoimmunity. J Autoimmun 1999; 12(4):229-232.

(85) Andre I, Gonzalez A, Wang B, Katz J, Benoist C, Mathis D. Checkpoints in the progression of autoimmune disease: lessons from diabetes models. Proc Natl Acad Sci U S A 1996; 93(6):2260-2263.

(86) Shimada A, Charlton B, Taylor-Edwards C, Fathman CG. b-cell destruction may be a late consequence of the autoimmune process in nonobese diabetic mice. diab 1996; 45:1063-1067.

(87) Chatenoud L, Primo J, Bach JF. CD3 antibody-induced dominant self tolerance in overtly diabetic NOD mice. J Immunol 1997; 158(6):2947-2954.

(88) Rosenbauer J, Herzig P, von Kries R, Neu A, Giani G. Temporal, seasonal, and geographical incidence patterns of type I diabetes mellitus in children under 5 years of age in Germany. diabetol 1999; 42(9):1055-1059.

(89) Chase HP, Dolan LM, Krischer JP, White N, Wolfsdorf JI, Cuthbertson DD et al. First phase insulin release during the intravenous glucose tolerance test as a risk factor for type 1 diabetes. J Pediatr 2001; 138(2):244-249.

(90) Foulis AK, McGill M, Farquharson MA, Hilton DA. A search for evidence of viral infection in panreases of newly diagnosed patients with IDDM. diabetol 1997; 40:53-61.

(91) Srikanta S, Ganda OP, Rabizadeh A, Soeldner JS, Eisenbarth GS. First-degree relatives of patients with type I diabetes mellitus. Islet cell antibodies and abnormal insulin secretion. N Engl J Med 1985; 313(8):461-464.

(92) Laing SP, Swerdlow AJ, Slater SD, Botha JL, Burden AC, Waugh NR et al. The British Diabetic Association Cohort Study, II: cause-specific mortality in patients with insulin-treated diabetes mellitus. Diabet Med 1999; 16(6):466-471.

(93) Laing SP, Swerdlow AJ, Slater SD, Botha JL, Burden AC, Waugh NR et al. The British Diabetic Association Cohort Study, I: all-cause mortality in patients with insulin-treated diabetes mellitus. Diabet Med 1999; 16(6):459-465.

(94) Herskowitz RD, Wolfsdorf JI, Ricker AT, Vardi P, Dib S, Soeldner JS et al. Transient hyperglycemia in childhood: identification of a subgroup with imminent diabetes mellitus. Diabetes Res 1988; 9(4):161-167.

(95) Barker JM, Goehrig SH, Barriga K, Hoffman M, Slover R, Eisenbarth GS et al. Clinical characteristics of children diagnosed with type 1 diabetes through intensive screening and follow-up. Diab care 2004; 27(6):1399-1404.

(96) Effects of insulin in relatives of patients with type 1 diabetes mellitus. N Engl J Med 2002; 346(22):1685-1691.

(97) Herold KC, Hagopian W, Auger JA, Poumian-Ruiz E, Taylor L, Donaldson D et al. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med 2002; 346(22):1692-1698.

 

 

Comments (0)

You don't have permission to comment on this page.