Type 1 diabetes

From Wikipedia, the free encyclopedia

This is an old revision of this page, as edited by ClueBot NG (talk | contribs) at 13:27, 31 October 2014 (Reverting possible vandalism by 50.250.32.193 to version by Doc James. False positive? Report it. Thanks, ClueBot NG. (2012562) (Bot)). The present address (URL) is a permanent link to this revision, which may differ significantly from the current revision.

Type 1 diabetes
SpecialtyDiabetology Edit this on Wikidata

Diabetes mellitus type 1 (also known as type 1 diabetes, or T1DM; formerly insulin-dependent diabetes or juvenile diabetes) is a form of diabetes mellitus that results from the autoimmune destruction of the insulin-producing beta cells in the pancreas.[2] The subsequent lack of insulin leads to increased blood and urine glucose. The classical symptoms are polyuria (frequent urination), polydipsia (increased thirst), polyphagia (increased hunger) and weight loss.[3]

The cause of diabetes mellitus type 1 is unknown.[4] Type 1 diabetes can be distinguished from type 2 by autoantibody testing. The C-peptide assay, which measures endogenous insulin production, can also be used.

Lack of care can be lethal and administration of insulin is essential for survival. Insulin therapy must be continued indefinitely and does not usually impair normal daily activities. People are usually trained to manage their diabetes independently; however, for some this can be challenging. Untreated, diabetes can cause many complications.[4] Acute complications include diabetic ketoacidosis and nonketotic hyperosmolar coma. Serious long-term complications include heart disease, stroke, kidney failure, foot ulcers and damage to the eyes.[4] Furthermore, complications may arise from low blood sugar caused by excessive treatment.

Diabetes mellitus type 1 accounts for 5% of cases of diabetes.[5] Globally, the number of people with DM type 1 is unknown,[6] although it is estimated that about 80,000 children develop the disease each year.[6] Within the United States the number of affected persons is estimated at one to three million.[6][7] The development of new cases vary by country and region; the lowest rates appears to be in Japan and China with approximately 1 person per 100,000 per year; the highest rates are found in Scandinavia where it is closer to 35 new cases per 100,000 per year.[8] The United States and northern Europe fall somewhere in between with 8-17 new cases per 100,000 per year.[8]

Signs and symptoms

Overview of the most significant symptoms of diabetes

The classical symptoms of type 1 diabetes include: polyuria (frequent urination), polydipsia (increased thirst), xerostomia (dry mouth), polyphagia (increased hunger), fatigue, and weight loss.[3]

Many type 1 diabetics are diagnosed when they are present with diabetic ketoacidosis. The symptoms of diabetic ketoacidosis include xeroderma (dry skin), rapid deep breathing, drowsiness, abdominal pain, and vomiting.[9]

Depression

Depression and depressive symptoms are generally more common in people living with type 1 diabetes. One review article suggested that the prevalence rate of depression is more than three times higher in diabetics than non-diabetics; an average prevalence of 12% was found (range of 5.8–43.4% in studies reviewed).[10] Women with type 1 diabetes are more likely to be depressed than men with the disease,[10] and an increased incidence of depression has been associated with youth with type 1 diabetes.[11] According to the Canadian Diabetes Association, 15% of people living with diabetes have major depression.[12] Psychological distress is reported in the parents of youth with type 1 diabetes.[13] Recent evidence has suggested that reduced pre-frontal cortical thickness is associated with depression in people with type 1 diabetes. These neurological changes may be caused by long-term reduced glycemic control and may increase risk of depression.[14]

Social cognition and self-care

Results from recent research suggest that people with type 1 diabetes may neglect precise self-care due to social fear related to fear of hypoglycemia.[15] Type 1 diabetics may also neglect physical activity due to reduced perceived positive effects as well as increased perceived negative aspects of that activity.[16]

Cause

The cause of type 1 diabetes is unknown.[4] A number of explanatory theories have been put forward, and the cause may be one or more of the following: genetic susceptibility, a diabetogenic trigger, and/or exposure to an antigen.[17]

Genetics

Type 1 diabetes is a disease that involves many genes. Depending on locus or combination of loci, they can be dominant, recessive, or somewhere in between. The strongest gene, IDDM1, is located in the MHC Class II region on chromosome 6, at staining region 6p21. Certain variants of this gene increase the risk for decreased histocompatibility characteristic of type 1. Such variants include DRB1 0401, DRB1 0402, DRB1 0405, DQA 0301, DQB1 0302 and DQB1 0201, which are common in North Americans of European ancestry and in Europeans.[18] Some variants also appear to be protective.[18]

The risk of a child developing type 1 diabetes is about 10% if the father has it, about 10% if a sibling has it, about 4% if the mother has type 1 diabetes and was aged 25 or younger when the child was born, and about 1% if the mother was over 25 years old when the child was born.[19]

Environmental

Environmental factors can influence expression of type 1. For identical twins, when one twin had type 1 diabetes, the other twin only had it 30%–50% of the time. Despite having exactly the same genome, one twin had the disease, whereas the other did not; this suggests environmental factors, in addition to genetic factors, can influence the disease's prevalence.[20] Other indications of environmental influence include the presence of a 10-fold difference in occurrence among Caucasians living in different areas of Europe, and a tendency to acquire the incidence of the disease of the destination country for people who migrate.[17]

Virus

One theory proposes that type 1 diabetes is a virus-triggered autoimmune response in which the immune system attacks virus-infected cells along with the beta cells in the pancreas.[21] The Coxsackie virus family or rubella is implicated, although the evidence is inconclusive. In type 1, pancreatic beta cells in the islets of Langerhans are destroyed, decreasing endogenous insulin production. This distinguishes type 1's origin from type 2. The type of diabetes a patient has is determined only by the cause—fundamentally by whether the patient is insulin resistant (type 2) or insulin deficient without insulin resistance (type 1).

This vulnerability is not shared by everyone, for not everyone infected by the suspected virus develops type 1 diabetes. This has suggested presence of a genetic vulnerability[22] and there is indeed an observed inherited tendency to develop type 1. It has been traced to particular HLA genotypes, though the connection between them and the triggering of an autoimmune reaction is still poorly understood.

Chemicals and drugs

Some chemicals and drugs selectively destroy pancreatic cells. Pyrinuron (Vacor, N-3-pyridylmethyl-N'-p-nitrophenyl urea), a rodenticide introduced in the United States in 1976, selectively destroys pancreatic beta cells, resulting in type 1 diabetes after accidental or intentional ingestion. Vacor was withdrawn from the U.S. market in 1979, but is still used in some countries. Zanosar is the trade name for streptozotocin, an antibiotic and antineoplastic agent used in chemotherapy for pancreatic cancer; it kills beta cells, resulting in loss of insulin production. Other pancreatic problems, including trauma, pancreatitis or tumors (either malignant or benign), can also lead to loss of insulin production.

Pathophysiology

The pathophysiology in diabetes type 1 is a destruction of beta cells in the pancreas, regardless of which risk factors or causative entities have been present.

Individual risk factors can have separate pathophysiological processes to, in turn, cause this beta cell destruction. Still, a process that appears to be common to most risk factors is an autoimmune response towards beta cells, involving an expansion of autoreactive CD4+ T helper cells and CD8+ T cells, autoantibody-producing B cells and activation of the innate immune system.[18][23]

After starting treatment with insulin a persons own insulin levels may temporarily improve.[24] This is believed to be due to altered immunity and is known as the "honeymoon phase".[24]

Diagnosis

WHO diabetes diagnostic criteria[25][26]  edit
Condition 2-hour glucose Fasting glucose HbA1c
Unit mmol/L mg/dL mmol/L mg/dL mmol/mol DCCT %
Normal < 7.8 < 140 < 6.1 < 110 < 42 < 6.0
Impaired fasting glycaemia < 7.8 < 140 6.1–7.0 110–125 42–46 6.0–6.4
Impaired glucose tolerance ≥ 7.8 ≥ 140 < 7.0 < 126 42–46 6.0–6.4
Diabetes mellitus ≥ 11.1 ≥ 200 ≥ 7.0 ≥ 126 ≥ 48 ≥ 6.5

Diabetes mellitus is characterized by recurrent or persistent hyperglycemia, and is diagnosed by demonstrating any one of the following:[27]

  • Fasting plasma glucose level at or above 7.0 mmol/L (126 mg/dL).
  • Plasma glucose at or above 11.1 mmol/L (200 mg/dL) two hours after a 75 g oral glucose load as in a glucose tolerance test.
  • Symptoms of hyperglycemia and casual plasma glucose at or above 11.1 mmol/L (200 mg/dL).
  • Glycated hemoglobin (hemoglobin A1C) at or above 6.5. (This criterion was recommended by the American Diabetes Association in 2010, although it has yet to be adopted by the WHO.)[28]

About a quarter of people with new type 1 diabetes have developed some degree of diabetic ketoacidosis (a type of metabolic acidosis which is caused by high concentrations of ketone bodies, formed by the breakdown of fatty acids and the deamination of amino acids) by the time the diabetes is recognized. The diagnosis of other types of diabetes is usually made in other ways. These include ordinary health screening, detection of hyperglycemia during other medical investigations, and secondary symptoms such as vision changes or unexplainable fatigue. Diabetes is often detected when a person suffers a problem that may be caused by diabetes, such as a heart attack, stroke, neuropathy, poor wound healing or a foot ulcer, certain eye problems, certain fungal infections, or delivering a baby with macrosomia or hypoglycemia.

A positive result, in the absence of unequivocal hyperglycemia, should be confirmed by a repeat of any of the above-listed methods on a different day. Most physicians prefer to measure a fasting glucose level because of the ease of measurement and the considerable time commitment of formal glucose tolerance testing, which takes two hours to complete and offers no prognostic advantage over the fasting test.[29] According to the current definition, two fasting glucose measurements above 126 mg/dL (7.0 mmol/L) is considered diagnostic for diabetes mellitus.

Patients with fasting glucose levels from 100 to 125 mg/dL (5.6 to 6.9 mmol/L) are considered to have impaired fasting glucose. Patients with plasma glucose at or above 140 mg/dL (7.8 mmol/L), but not over 200 mg/dL (11.1 mmol/L), two hours after a 75 g oral glucose load are considered to have impaired glucose tolerance. Of these two pre-diabetic states, the latter in particular is a major risk factor for progression to full-blown diabetes mellitus and cardiovascular disease.[30]

Autoantibodies

The appearance of diabetes-related autoantibodies has been shown to be able to predict the appearance of diabetes type 1 before any hyperglycemia arises, the main ones being islet cell autoantibodies, insulin autoantibodies, autoantibodies targeting the 65-kDa isoform of glutamic acid decarboxylase (GAD), autoantibodies targeting the phosphatase-related IA-2 molecule, and zinc transporter autoantibodies (ZnT8).[17] By definition, the diagnosis of diabetes type 1 can be made first at the appearance of clinical symptoms and/or signs, but the emergence of autoantibodies may itself be termed "latent autoimmune diabetes". Not everyone with autoantibodies progresses to diabetes type 1, but the risk increases with the number of antibody types, with three to four antibody types giving a risk of progressing to diabetes type 1 of 60%–100%.[17] The time interval from emergence of autoantibodies to frank diabetes type 1 can be a few months in infants and young children, but in some people it may take years – in some cases more than 10 years.[17] Islet cell autoantibodies are detected by conventional immunofluorescence, while the rest are measured with specific radiobinding assays.[17]

Prevention

Type 1 diabetes is not currently preventable.[31] Some researchers believe it might be prevented at the latent autoimmune stage, before it starts destroying beta cells.[18]

Immunosuppressive drugs

Cyclosporine A, an immunosuppressive agent, has apparently halted destruction of beta cells (on the basis of reduced insulin usage), but its nephrotoxicity and other side effects make it highly inappropriate for long-term use.[18]

Anti-CD3 antibodies, including teplizumab and otelixizumab, had suggested evidence of preserving insulin production (as evidenced by sustained C-peptide production) in newly diagnosed type 1 diabetes patients.[18] A probable mechanism of this effect was believed to be preservation of regulatory T cells that suppress activation of the immune system and thereby maintain immune system homeostasis and tolerance to self-antigens.[18] The duration of the effect is still unknown, however.[18] In 2011, Phase III studies with otelixizumab and teplizumab both failed to show clinical efficacy, potentially due to an insufficient dosing schedule.[32][33]

An anti-CD20 antibody, rituximab, inhibits B cells and has been shown to provoke C-peptide responses three months after diagnosis of type 1 diabetes, but long-term effects of this have not been reported.[18]

Diet

Some research has suggested breastfeeding decreases the risk in later life;[34][35] various other nutritional risk factors are being studied, but no firm evidence has been found.[36] Giving children 2000 IU of Vitamin D during their first year of life is associated with reduced risk of type 1 diabetes, though the causal relationship is obscure.[37]

Children with antibodies to beta cell proteins (i.e. at early stages of an immune reaction to them) but no overt diabetes, and treated with vitamin B3 the niacinamide version, had less than half the diabetes onset incidence in a seven-year time span than did the general population, and an even lower incidence relative to those with antibodies as above, but who received no niacinamide.[38]

Management

Insulin

There are four main types of insulin, rapid acting insulin, short acting insulin, intermediate acting insulin, and long acting insulin. The rapid acting insulin is used as a bolus dosage. The action onsets in 15 minutes with peak actions in 30 to 90 minutes. Short acting insulin action onsets within 30 minutes with the peak action around 2 to 4 hours. Intermediate acting insulin action onsets within 1 to 2 hours with peak action of 4 to 10 hours. Long acting insulin is usually given around bedtime. The action onset is roughly 1 to 2 hours with a sustained action of 24 hours.

Injections of insulin—either via subcutaneous injection or insulin pump— is necessary for those living with type 1 diabetes. It can't be treated with diet and exercise alone. [citation needed] In addition to insulin therapy dietary management is important. This includes keeping track of the carbohydrate content of food, and careful monitoring of blood glucose levels using glucose meters. Today, the most common insulins are biosynthetic products produced using genetic recombination techniques; formerly, cattle or pig insulins were used, and even sometimes insulin from fish.[39] Major global suppliers include Eli Lilly and Company, Novo Nordisk, and Sanofi-Aventis. A more recent trend, from several suppliers, is insulin analogs which are slightly modified insulins with different onset or duration of action times.

Untreated type 1 diabetes commonly leads to coma, often from diabetic ketoacidosis, which is fatal if untreated. Diabetic ketoacidosis can cause cerebral edema (accumulation of liquid in the brain). This complication is life-threatening. Children are at an increased risk for cerebral edema, making ketoacidosis the most common cause of death in pediatric diabetes.[40]

Treatment of diabetes focuses on lowering blood sugar or glucose (BG) to the near normal range, approximately 80–140 mg/dl (4.4–7.8 mmol/L).[41] The ultimate goal of normalizing BG is to avoid long-term complications that affect the nervous system (e.g. peripheral neuropathy leading to pain and/or loss of feeling in the extremities), and the cardiovascular system (e.g. heart attacks, vision loss). This level of control over a prolonged period of time can be varied by a target HbA1c level of less than 7.5%.[6]

People with type 1 diabetes always need to use insulin, but treatment can lead to low BG (hypoglycemia), i.e. BG less than 70 mg/dl (3.9 mmol/l). Hypoglycemia is a very common occurrence in people with diabetes, usually the result of a mismatch in the balance among insulin, food and physical activity, although the nonphysiological method of delivery[clarification needed] also plays a role. Continuous glucose monitors can alert patients to the presence of dangerously high or low blood sugar levels, but technical issues have limited the effect these devices have had on clinical practice .[citation needed]

Pancreas transplantation

In more extreme cases, a pancreas transplant can restore proper glucose regulation. However, the surgery and accompanying immunosuppression required may be more dangerous than continued insulin replacement therapy, so is generally only used with or some time after a kidney transplant. One reason for this is that introducing a new kidney requires taking immunosuppressive drugs such as cyclosporine. Nevertheless this allows the introduction of a new, functioning pancreas to a patient with diabetes without any additional immunosuppressive therapy. However, pancreas transplants alone can be wise in patients with extremely labile type 1 diabetes mellitus.[42]

Islet cell transplantation

Experimental replacement of beta cells (by transplant or from stem cells) is being investigated in several research programs. Islet cell transplantation is less invasive than a pancreas transplant, which is currently the most commonly used approach in humans.

In one variant of this procedure, islet cells are injected into the patient's liver, where they take up residence and begin to produce insulin. The liver is expected to be the most reasonable choice because it is more accessible than the pancreas, and islet cells seem to produce insulin well in that environment. The patient's body, however, will treat the new cells just as it would any other introduction of foreign tissue, unless a method is developed to produce them from the patient's own stem cells or an identical twin is available who can donate stem cells. The immune system will attack the cells as it would a bacterial infection or a skin graft. Thus, patients now also need to undergo treatment involving immunosuppressants, which reduce immune system activity.

Recent studies have shown islet cell transplants have progressed to the point where 58% of the patients in one study were insulin-independent one year after transplantation.[43] Scientists in New Zealand with Living Cell Technologies are currently in human trials with Diabecell, placing pig islets within a protective capsule derived of seaweed which enables insulin to flow out and nutrients to flow in, while protecting the islets from immune system attack via white blood cells.

Complications

Complications of poorly managed type 1 diabetes mellitus may include cardiovascular disease, diabetic neuropathy, and diabetic retinopathy, among others. However, cardiovascular disease[44] as well as neuropathy[45] may have an autoimmune basis, as well.

Driving

Studies conducted in the United States[46] and Europe[47] showed that drivers with type 1 diabetes had twice as many collisions as their nondiabetic spouses, demonstrating the increased risk of driving collisions in the type 1 diabetes population. Diabetes can compromise driving safety in several ways. First, long-term complications of diabetes can interfere with the safe operation of a vehicle. For example, diabetic retinopathy (loss of peripheral vision or visual acuity), or peripheral neuropathy (loss of feeling in the feet) can impair a driver's ability to read street signs, control the speed of the vehicle, apply appropriate pressure to the brakes, etc.

Second, hypoglycemia can affect a person's thinking processes, coordination, and state of consciousness.[48][49] This disruption in brain functioning, neuroglycopenia, can impair driving ability.[48][50] A study involving people with type 1 diabetes found that individuals reporting two or more hypoglycemia-related driving mishaps differ physiologically and behaviorally from their counterparts who report no such mishaps.[51] For example, during hypoglycemia, drivers who had two or more mishaps reported fewer warning symptoms, their driving was more impaired, and their body released less epinephrine (a hormone that helps raise BG). Additionally, individuals with a history of hypoglycemia-related driving mishaps appear to use sugar at a faster rate[52] and are relatively slower at processing information.[53] These findings indicate that although anyone with type 1 diabetes may be at some risk of experiencing disruptive hypoglycemia while driving, there is a subgroup of type 1 drivers who are more vulnerable to such events.

Given the above research findings, drivers with type 1 diabetes and a history of driving mishaps are recommended to never drive when their BG is less than 80 mg/dl. Instead, these drivers are advised to treat hypoglycemia and delay driving until their BG is above 90 mg/dl.[51] Such drivers should also learn as much as possible about what causes their hypoglycemia, and use this information to avoid future hypoglycemia while driving.

Studies funded by the National Institutes of Health (NIH) have demonstrated that face-to-face training programs designed to help individuals with type 1 diabetes better anticipate, detect, and prevent extreme BG can reduce the occurrence of future hypoglycemia-related driving mishaps.[54][55][56] An internet-version of this training has also been shown to have significant beneficial results.[57] Additional NIH funded research to develop internet interventions specifically to help improve driving safety in drivers with type 1 diabetes is currently underway.[58]

Epidemiology

Type 1 diabetes causes an estimated 5–10% of all diabetes cases[59] or 11–22 million worldwide.[31] In 2006 it affected 440,000 children under 14 years of age and was the primary cause of diabetes in those less than 10 years of age.[60] The incidence of type 1 diabetes has been increasing by about 3% per year.[60]

Rates vary widely by country. In Finland, the incidence is a high of 57 per 100,000 per year, in Japan and China a low of 1 to 3 per 100,000 per year, and in Northern Europe and the U.S., an intermediate of 8 to 17 per 100,000 per year.[8][61]

History

Type 1 diabetes was described as an autoimmune disease in the 1970s, based on observations that autoantibodies against islets were discovered in diabetics with other autoimmune deficiencies.[62] It was also shown in the 1980s that immunosuppressive therapies could slow disease progression, further supporting the idea that type 1 diabetes is an autoimmune disorder.[63]

Society and culture

The disease was estimated to cause $10.5 billion in annual medical costs ($875 per month per diabetic) and an additional $4.4 billion in indirect costs ($366 per month per person with diabetes) in the U.S.[64]

Research

Funding for research into type 1 diabetes originates from government, industry (e.g., pharmaceutical companies), and charitable organizations. Government funding in the United States is distributed via the National Institute of Health, and in the UK via the National Institute for Health Research or the Medical Research Council. The Juvenile Diabetes Research Foundation, originally founded by parents of children with type 1 diabetes, is the world's largest provider of charity based funding for type 1 diabetes research. Other charities include the American Diabetes Association, Diabetes UK, Diabetes Research and Wellness Foundation,[65] Diabetes Australia, the Canadian Diabetes Association.

Stem cells

Stem cells are being studied in mice.[66] An early trial has also been carried out in humans.[67][68]

Vaccine

Injections with a vaccine containing GAD65, an autoantigen involved in type 1 diabetes, has in clinical trials delayed the destruction of beta cells when treated within six months of diagnosis.[18] Patients treated with the substance showed higher levels of regulatory cytokines, thought to protect the beta cells.[69] Phase III trials are under way in the USA[70] and in Europe.[71][72][73] Two prevention studies, where the vaccine is given to persons who have not yet developed diabetes are underway.[74][75][76]

Labile diabetes

Insulin-dependent diabetes characterized by dramatic and recurrent swings in glucose levels, often occurring for no apparent reason, is sometimes known as brittle diabetes, unstable diabetes or labile diabetes, although some experts say the "brittle diabetes" concept "has no biologic basis and should not be used".[77] The results of such swings can be irregular and unpredictable hyperglycemias, frequently involving ketosis, and sometimes serious hypoglycemias. Brittle diabetes occurs no more frequently than in 1% to 2% of diabetics.[78] An insulin pump may be recommended for brittle diabetes to reduce the number of hypoglycemic episodes and better control the morning rise of blood sugar due to the dawn phenomenon.[79] In a small study, 10 of 20 brittle diabetic patients aged 18–23 years who could be traced had died within 22 years, and the remainder, though suffering high rates of complications, were no longer brittle.[80] These results were similar to those of an earlier study by the same authors which found a 19% mortality in 26 patients after 10.5 years.[81]

Because labile diabetes is defined as "episodes of hypoglycemia or hyperglycemia that, whatever their cause, constantly disrupt a patient's life", it can have many causes, some of which include:[82]

  • errors in diabetes management, which can include too much insulin being given, in ratio, to carbohydrate being consumed.
  • interactions with other medical conditions.
  • psychological problems.
  • biological factors that interfere with how insulin is processed within the body.
  • hypoglycemia and hyperglycemia due to strenuous exercise, however hypoglycemia is more frequent.
  • Insulin exposed to higher temperatures reduces effectiveness of the insulin hormone in the body.
  • Spontaneous production of insulin in the body due to activity in the beta cells during the period shortly after diagnosis of type 1 diabetes.

Exercise related hyperglycemia is caused when hormones (such as adrenaline and cortisol) are released during moderate to strenuous exercise. This happens when the muscles signal the liver to release glucose into the bloodstream by converting stored glycogen into glucose. The cause of exercise related hypoglycemia, on the other hand, occurs when the muscle group being exercised uses up glucose faster than it can be replenished by the body.

One of these biological factors is the production of insulin autoantibodies. High antibody titers can cause episodes of hyperglycemia by neutralizing the insulin, cause clinical insulin resistance requiring doses of over 200 IU/day. However, antibodies may also fail to buffer the release of the injected insulin into the bloodstream after subcutaneous injection, resulting in episodes of hypoglycemia. In some cases, changing the type of insulin administered can resolve this problem.[82] There have been a number of reports that insulin autoantibodies can act as a "sink" for insulin and affect the time to peak, half-life, distribution space, and metabolic clearance, though in most patients these effects are small.[83]

References

  1. ^ "Diabetes Blue Circle Symbol". International Diabetes Federation. 17 March 2006.
  2. ^ "Type 1 Diabetes Mellitus". Retrieved 4 August 2008.
  3. ^ a b Cooke DW, Plotnick L (November 2008). "Type 1 diabetes mellitus in pediatrics". Pediatr Rev. 29 (11): 374–84, quiz 385. doi:10.1542/pir.29-11-374. PMID 18977856.
  4. ^ a b c d "Diabetes Fact sheet N°312". WHO. October 2013. Retrieved 25 March 2014.
  5. ^ "Type 1 Diabetes". American Diabetes Association. American Diabetes Association. Retrieved 26 October 2014.
  6. ^ a b c d Chiang, J. L.; Kirkman, M. S.; Laffel, L. M. B.; Peters, A. L. (16 June 2014). "Type 1 Diabetes Through the Life Span: A Position Statement of the American Diabetes Association". Diabetes Care. 37 (7): 2034–2054. doi:10.2337/dc14-1140.
  7. ^ "FAST FACTS Data and Statistics about Diabetes" (PDF). professional.diabetes.org/facts. American Diabetes Association. Retrieved 25 July 2014.
  8. ^ a b c Kasper, Dennis L; Braunwald, Eugene; Fauci, Anthony; et al. (2005). Harrison's Principles of Internal Medicine (16th ed.). New York: McGraw-Hill. ISBN 0-07-139140-1. {{cite book}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  9. ^ "webmd Symptoms Type I Diabetes".
  10. ^ a b Roy T, Lloyd CE (2012). "Epidemiology of depression and diabetes: a systematic review". J Affect Disord. 142 Suppl: S8–21. doi:10.1016/S0165-0327(12)70004-6. PMID 23062861.
  11. ^ Type 1 Diabetes: Poor blood sugar control linked to depression in youth with Diabetes. Mental Health Business Week (6 May 2006) 97.
  12. ^ [1][dead link]
  13. ^ Jaser SS, Linsky R, Grey M (2014). "Coping and psychological distress in mothers of adolescents with type 1 diabetes". Matern Child Health J. 18 (1): 101–8. doi:10.1007/s10995-013-1239-4. PMID 23420308.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  14. ^ Lyoo IK, Yoon S, Jacobson AM, Hwang J, Musen G, Kim JE, Simonson DC, Bae S, Bolo N, Kim DJ, Weinger K, Lee JH, Ryan CM, Renshaw PF (2012). "Prefrontal cortical deficits in type 1 diabetes mellitus: brain correlates of comorbid depression". Arch. Gen. Psychiatry. 69 (12): 1267–76. doi:10.1001/archgenpsychiatry.2012.543. PMID 23090665.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  15. ^ Di Battista, Ashley M., Hart, Trevor A., Greco, & Laurie, Gloizer. (2009). Type 1 Diabetes among adolescents: Reduced Diabetes self-care caused by social fear of hypoglycemia. The Diabetes Educator, 35, 465–475.
  16. ^ Plotnikoff RC, Karunamuni N, Brunet S (2009). "A comparison of physical activity-related social-cognitive factors between those with type 1 diabetes, type 2 diabetes and diabetes free adults". Psychol Health Med. 14 (5): 536–44. doi:10.1080/13548500903012863. PMID 19844832.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  17. ^ a b c d e f Knip M, Veijola R, Virtanen SM, Hyöty H, Vaarala O, Akerblom HK (2005). "Environmental Triggers and Determinants of Type 1 Diabetes". Diabetes. 54: S125–S136. doi:10.2337/diabetes.54.suppl_2.S125. PMID 16306330.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  18. ^ a b c d e f g h i j Bluestone JA, Herold K, Eisenbarth G (2010). "Genetics, pathogenesis and clinical interventions in type 1 diabetes". Nature. 464 (7293): 1293–1300. Bibcode:2010Natur.464.1293B. doi:10.1038/nature08933. PMID 20432533.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  19. ^ Genetics & Diabetes, Diabetes Information. Dr. Warram. Joslin Diabetes Center and Joslin Clinic
  20. ^ "OMIM Entry – %222100 – DIABETES MELLITUS, INSULIN-DEPENDENT; IDDM". Ncbi.nlm.nih.gov. Retrieved 29 November 2011.
  21. ^ Fairweather D, Rose NR (2002). "Type 1 diabetes: virus infection or autoimmune disease?". Nat. Immunol. 3 (4): 338–40. doi:10.1038/ni0402-338. PMID 11919574.
  22. ^ Donner H, Rau H, Walfish PG, Braun J, Siegmund T, Finke R, Herwig J, Usadel KH, Badenhoop K (1997). "CTLA4 alanine-17 confers genetic susceptibility to Graves' disease and to type 1 diabetes mellitus". J. Clin. Endocrinol. Metab. 82 (1): 143–6. doi:10.1210/jcem.82.1.3699. PMID 8989248.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  23. ^ Chatzigeorgiou A, Harokopos V, Mylona-Karagianni C, Tsouvalas E, Aidinis V, Kamper EF (September 2010). "The pattern of inflammatory/anti-inflammatory cytokines and chemokines in type 1 diabetic patients over time". Ann. Med. 42 (6): 426–38. doi:10.3109/07853890.2010.495951. PMID 20568978.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  24. ^ a b Aly H, Gottlieb P (August 2009). "The honeymoon phase: intersection of metabolism and immunology". Current opinion in endocrinology, diabetes, and obesity. 16 (4): 286–92. doi:10.1097/med.0b013e32832e0693. PMID 19506474.
  25. ^ Definition and diagnosis of diabetes mellitus and intermediate hyperglycemia: Report of a WHO/IDF consultation (PDF). Geneva: World Health Organization. 2006. p. 21. ISBN 978-92-4-159493-6.
  26. ^ Vijan S (March 2010). "In the clinic. Type 2 diabetes". Annals of Internal Medicine. 152 (5): ITC31-15, quiz ITC316. doi:10.7326/0003-4819-152-5-201003020-01003. PMID 20194231.
  27. ^ World Health Organisation Department of Noncommunicable Disease Surveillance (1999). "Definition, Diagnosis and Classification of Diabetes Mellitus and its Complications" (PDF).
  28. ^ ""Diabetes Care" January 2010". American Diabetes Association. Retrieved 29 January 2010.
  29. ^ Saydah SH, Miret M, Sung J, Varas C, Gause D, Brancati FL (August 2001). "Postchallenge hyperglycemia and mortality in a national sample of U.S. adults". Diabetes Care. 24 (8): 1397–402. doi:10.2337/diacare.24.8.1397. PMID 11473076.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  30. ^ Santaguida PL, Balion C, Hunt D, Morrison K, Gerstein H, Raina P, Booker L, Yazdi H. "Diagnosis, Prognosis, and Treatment of Impaired Glucose Tolerance and Impaired Fasting Glucose". Summary of Evidence Report/Technology Assessment, No. 128. Agency for Healthcare Research and Quality. Retrieved 20 July 2008.{{cite web}}: CS1 maint: multiple names: authors list (link)
  31. ^ a b "Diabetes". World Health Organization. Retrieved 24 January 2011.
  32. ^ "''Tolerx, Inc. and GlaxoSmithKline (GSK) Announce Phase 3 Defend-1 Study of Otelixizumab in Type 1 Diabetes Did Not Meet Its Primary Endpoint''". Biospace. Retrieved 29 November 2011.
  33. ^ "Macrogenics press release: ''MacroGenics and Lilly Announce Pivotal Clinical Trial of Teplizumab Did Not Meet Primary Efficacy Endpoint''". Macrogenics.com. 20 October 2010. Retrieved 29 November 2011.
  34. ^ Borch-Johnsen K, Joner G, Mandrup-Poulsen T, Christy M, Zachau-Christiansen B, Kastrup K, Nerup J (November 1984). "Relation between breast-feeding and incidence rates of insulin-dependent diabetes mellitus. A hypothesis". Lancet. 2 (8411): 1083–6. doi:10.1016/S0140-6736(84)91517-4. PMID 6150150.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  35. ^ Shehadeh N, Shamir R, Berant M, Etzioni A (2001). "Insulin in human milk and the prevention of type 1 diabetes". Pediatric Diabetes. 2 (4): 175–7. doi:10.1034/j.1399-5448.2001.20406.x. PMID 15016183.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  36. ^ Virtanen SM, Knip M (December 2003). "Nutritional risk predictors of beta cell autoimmunity and type 1 diabetes at a young age". The American Journal of Clinical Nutrition. 78 (6): 1053–67. PMID 14668264.
  37. ^ Hyppönen E, Läärä E, Reunanen A, Järvelin MR, Virtanen SM (November 2001). "Intake of vitamin D and risk of type 1 diabetes: a birth-cohort study". Lancet. 358 (9292): 1500–3. doi:10.1016/S0140-6736(01)06580-1. PMID 11705562.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  38. ^ Elliott RB, Pilcher CC, Fergusson DM, Stewart AW (1996). "A population based strategy to prevent insulin-dependent diabetes using nicotinamide". Journal of Pediatric Endocrinology & Metabolism. 9 (5): 501–9. doi:10.1515/JPEM.1996.9.5.501. PMID 8961125.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  39. ^ Wright JR (2002). "From ugly fish to conquer death: J J R Macleod's fish insulin research, 1922-24". Lancet. 359 (9313): 1238–42. doi:10.1016/S0140-6736(02)08222-3. PMID 11955558.
  40. ^ Rosenbloom AL, Hanas R (1996). "Diabetic ketoacidosis (DKA): treatment guidelines" (PDF). Clin Pediatr (Phila). 35 (5): 261–6. doi:10.1177/000992289603500506. PMID 8804545.
  41. ^ American Diabetes Association Clinical Guidelines, 2010.
  42. ^ Jennifer L. Larsen. "Pancreas Transplantation: Indications and Consequences". Edrv.endojournals.org. Retrieved 29 November 2011.
  43. ^ "Islet cell transplant: Experimental treatment for type 1 diabetes". Archived from the original on 10 April 2007. Retrieved 4 June 2007.
  44. ^ Devaraj S, Glaser N, Griffen S, Wang-Polagruto J, Miguelino E, Jialal I (March 2006). "Increased monocytic activity and biomarkers of inflammation in patients with type 1 diabetes". Diabetes. 55 (3): 774–9. doi:10.2337/diabetes.55.03.06.db05-1417. PMID 16505242.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  45. ^ Granberg V, Ejskjaer N, Peakman M, Sundkvist G (2005). "Autoantibodies to autonomic nerves associated with cardiac and peripheral autonomic neuropathy". Diabetes Care. 28 (8): 1959–64. doi:10.2337/diacare.28.8.1959. PMID 16043739.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  46. ^ Songer, TJ. Low blood sugar and motor vehicle crashes in persons with type 1 diabetes, Annu Proc Assoc Adv Automotive Med, 46:424–427 (2002)
  47. ^ Cox DJ, Penberthy JK, Zrebiec J, Weinger K, Aikens JE, Frier B, Stetson B, DeGroot M, Trief P, Schaechinger H, Hermanns N, Gonder-Frederick L, Clarke W (2003). "Diabetes and Driving Mishaps: Frequency and correlations from a multinational survey". Diabetes Care. 26 (8): 2329–2334. doi:10.2337/diacare.26.8.2329. PMID 12882857.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  48. ^ a b Cox DJ, Gonder-Frederick L, Clarke W (1993). "Driving decrements in type 1 diabetes during moderate hypoglycemia". [[Diabetes (journal)|]]. 42 (2): 239–243. doi:10.2337/diabetes.42.2.239. PMID 8425660.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  49. ^ Clarke WL, Cox DJ, Gonder-Frederick LA, Kovatchev B (1999). "Hypoglycemia and the Decision to Drive a Motor Vehicle by Persons With Diabetes". JAMA. 282 (8): 750–754. doi:10.1001/jama.282.8.750. PMID 10463710.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  50. ^ Cox DJ, Gonder-Frederick LA, Kovatchev BP, Julian DM, Clarke WL (2000). "Progressive hypoglycemia's impact on driving simulation performance". Diabetes Care. 23 (2): 163–170. doi:10.2337/diacare.23.2.163. PMID 10868825.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  51. ^ a b Cox DJ, Kovatchev BP, Anderson SM, Clarke WL, Gonder-Frederick LA (November 2010). "Type 1 diabetic drivers with and without a history of recurrent hypoglycemia-related driving mishaps: physiological and performance differences during euglycemia and the induction of hypoglycemia". Diabetes Care. 33 (11): 2430–5. doi:10.2337/dc09-2130. PMC 2963507. PMID 20699432.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  52. ^ Cox DJ, Gonder-Frederick LA, Kovatchev BP, Clarke WL (2002). "The metabolic demands of driving for drivers with type 1 diabetes mellitus". Diabetes/Metabolism Research and Review. 18 (5): 381–385. doi:10.1002/dmrr.306.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  53. ^ Campbell LK, Gonder-Frederick LA, Broshek DK, Kovatchev BP, Anderson S, Clarke WL & Cox DJ (2010). Neurocognitive differences between drivers with type 1 diabetes with and without a recent history of recurrent driving mishaps. International Journal of Diabetes in Developing Countries, 2(2), 73–77. NIHMS[211748]
  54. ^ Cox DJ, Gonder-Frederick L, Julian DM, Clarke W (1994). "Long-term follow-up evaluation of blood glucose awareness training". Diabetes Care. 17 (1): 1–5. doi:10.2337/diacare.17.1.1. PMID 8112183.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  55. ^ Cox DJ, Gonder-Frederick L, Polonsky W, Schlundt D, Kovatchev B, Clarke W (2001). "Blood Glucose Awareness Training (BGAT-II): Long term benefits". Diabetes Care. 24 (4): 637–642. doi:10.2337/diacare.24.4.637. PMID 11315822.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  56. ^ Broers S, le Cessie S, van Vliet KP, Spinhoven P, van der Ven NC, Radder JK (2002). "Blood glucose awareness training in Dutch type 1 diabetes patients". Diabet. Med. 19 (2): 157–161. doi:10.1046/j.1464-5491.2002.00682.x. PMID 11874433.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  57. ^ Cox D, Ritterband L, Magee J, Clarke W, Gonder-Frederick L (2008). "Blood Glucose Awareness Training Delivered Over The Internet". Diabetes Care. 31 (8): 1527–1528. doi:10.2337/dc07-1956. PMC 2494647. PMID 18477813.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  58. ^ http://www.DiabetesDriving.com Diabetes Driving.
  59. ^ Daneman D (11 March 2006). "Type 1 diabetes". Lancet. 367 (9513): 847–58. doi:10.1016/S0140-6736(06)68341-4. PMID 16530579.
  60. ^ a b Aanstoot HJ, Anderson BJ, Daneman D, Danne T, Donaghue K, Kaufman F, Réa RR, Uchigata Y (October 2007). "The global burden of youth diabetes: perspectives and potential". Pediatric diabetes. 8. Suppl 8 (s8): 1–44. doi:10.1111/j.1399-5448.2007.00326.x. PMID 17767619.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  61. ^ Soltesz G, Patterson CC, Dahlquist G (October 2007). "Worldwide childhood type 1 diabetes incidence—what can we learn from epidemiology?". Pediatric diabetes. 8. Suppl 6 (s6): 6–14. doi:10.1111/j.1399-5448.2007.00280.x. PMID 17727380.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  62. ^ Bottazzo, GF; Florin-Christensen, A; Doniach, D (30 November 1974). "Islet-cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies". Lancet. 2 (7892): 1279–83. doi:10.1016/s0140-6736(74)90140-8. PMID 4139522.
  63. ^ Herold, KC; Vignali, DA; Cooke, A; Bluestone, JA (April 2013). "Type 1 diabetes: translating mechanistic observations into effective clinical outcomes". Nature reviews. Immunology. 13 (4): 243–56. doi:10.1038/nri3422. PMID 23524461.
  64. ^ Johnson, Linda (18 November 2008). "Study: Cost of diabetes $218B". USA Today. Associated Press.
  65. ^ Diabetes Research and Wellness Foundation
  66. ^ Zhao Y, Lin B, Dingeldein M, Guo C, Hwang D, Holterman MJ (May 2010). "New type of human blood stem cell: a double-edged sword for the treatment of type 1 diabetes". Translational research : the journal of laboratory and clinical medicine. 155 (5): 211–6. doi:10.1016/j.trsl.2010.01.003. PMID 20403575.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  67. ^ Zhao Y, Jiang Z, Zhao T, Ye M, Hu C, Yin Z, Li H, Zhang Y, Diao Y, Li Y, Chen Y, Sun X, Fisk MB, Skidgel R, Holterman M, Prabhakar B, Mazzone T (10 January 2012). "Reversal of type 1 diabetes via islet β cell regeneration following immune modulation by cord blood-derived multipotent stem cells". BMC medicine. 10: 3. doi:10.1186/1741-7015-10-3. PMC 3322343. PMID 22233865.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unflagged free DOI (link)
  68. ^ Zhao Y (October 2012). "Stem cell educator therapy and induction of immune balance". Current Diabetes Reports. 12 (5): 517–23. doi:10.1007/s11892-012-0308-1. PMID 22833322.
  69. ^ "New England Journal of Medicine: GAD Treatment and Insulin Secretion in Recent-Onset type 1 Diabetes". Content.nejm.org. Retrieved 29 November 2011.
  70. ^ "Diamyd US Phase III Trial". Clinicaltrials.gov. Retrieved 29 November 2011.
  71. ^ "Diamyd European Phase III Trial". Clinicaltrials.gov. Retrieved 29 November 2011.
  72. ^ "Further Evidence for Lasting Immunological Efficacy of Diamyd Diabets Vaccine". Diamyd.com. 19 September 2007. Retrieved 29 November 2011.
  73. ^ "Diamyd Announces Completion of type 1 Diabetes Vaccine Trial with Long Term Efficcacy Demonstrated at 30 Months". Diamyd.com. 21 January 2008. Retrieved 29 November 2011.
  74. ^ MSNBC News: Pioneering Diamyd(r) Study to Prevent Childhood Diabetes Approved[dead link]
  75. ^ "Diamyd press release: Diamyd approved for groundbreking study in Norway". Diamyd.com. Retrieved 29 November 2011.
  76. ^ "Diabetes Prevention – Immune Tolerance (DIAPREV-IT)". Clinicaltrials.gov. Retrieved 29 November 2011.
  77. ^ "Diabetes Mellitus (DM): Diabetes Mellitus and Disorders of Carbohydrate Metabolism: Merck Manual Professional". Merck.com. Retrieved 30 July 2010.
  78. ^ Dorner M, Pinget M, Brogard JM (May 1977). "[Essential labile diabetes (author's transl)]". MMW Munch Med Wochenschr. 119 (19): 671–4. PMID 406527.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  79. ^ Higuchi, Chigusa; Tone, Atsuhito; Iseda, Izumi; Tsukamoto, Keiko; Katayama, Akihiro (2010). "A Pregnant Patient with Brittle Type 1 Diabetes Successfully Managed by CSII Therapy with Insulin Aspart". Journal of Diabetes & Metabolism. 01. doi:10.4172/2155-6156.1000104.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  80. ^ Cartwright A, Wallymahmed M, Macfarlane IA, Wallymahmed A, Williams G, Gill GV (2011). "The outcome of brittle type 1 diabetes--a 20 year study". QJM. 104 (7): 575–9. doi:10.1093/qjmed/hcr010. PMID 21285231.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  81. ^ Kent LA, Gill GV, Williams G (September 1994). "Mortality and outcome of patients with brittle diabetes and recurrent ketoacidosis". Lancet. 344 (8925): 778–81. doi:10.1016/S0140-6736(94)92340-X. PMID 7916072.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  82. ^ a b Davidson MB, Kumar D, Smith W (1991). "Successful treatment of unusual case of brittle diabetes with sulfated beef insulin". Diabetes Care. 14 (11): 1109–10. PMID 1797500.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  83. ^ Fineberg SE, Kawabata TT, Finco-Kent D, Fountaine RJ, Finch GL, Krasner AS (2007). "Immunological Responses to Exogenous Insulin". Endocrine Reviews. 28 (6): 625–52. doi:10.1210/er.2007-0002. PMID 17785428.{{cite journal}}: CS1 maint: multiple names: authors list (link)

External links

This template is no longer used; please see Template:Endocrine pathology for a suitable replacement