fbpx
Wikipedia

Type 1 diabetes

Type 1 diabetes (T1D), formerly known as juvenile diabetes, is an autoimmune disease that originates when cells that make insulin (beta cells) are destroyed by the immune system.[4] Insulin is a hormone required for the cells to use blood sugar for energy and it helps regulate glucose levels in the bloodstream.[2] Before treatment this results in high blood sugar levels in the body.[1] The common symptoms of this elevated blood sugar are frequent urination, increased thirst, increased hunger, weight loss, and other serious complications.[4][10] Additional symptoms may include blurry vision, tiredness, and slow wound healing.[2] Symptoms typically develop over a short period of time, often a matter of weeks if not months.[1]

Type 1 Diabetes
Other namesDiabetes mellitus type 1, insulin-dependent diabetes,[1] juvenile diabetes[2]
A blue circle, the symbol for diabetes.[3]
Pronunciation
SpecialtyEndocrinology
SymptomsFrequent urination, increased thirst, increased hunger, weight loss,[4] blurred vision, fatigue and weakness, Bed-wetting in children who previously didn't wet the bed during the night[5]
ComplicationsDiabetic ketoacidosis, nonketotic hyperosmolar coma, poor healing, cardiovascular disease, damage to the eyes[2][4][6]
Usual onsetRelatively short period of time[1]
DurationLong term[4]
CausesBody does not produce enough insulin[4]
Risk factorsFamily history, celiac disease[6][7]
Diagnostic methodBlood sugar, A1C[6][8]
PreventionUnknown[4]
TreatmentInsulin, diabetic diet, exercise[1][2]
Frequency~7.5% of diabetes cases[9]

The cause of type 1 diabetes is unknown,[4] but it is believed to involve a combination of genetic and environmental factors.[1] The underlying mechanism involves an autoimmune destruction of the insulin-producing beta cells in the pancreas.[2] Diabetes is diagnosed by testing the level of sugar or glycated hemoglobin (HbA1C) in the blood.[6][8] Type 1 diabetes can typically be distinguished from type 2 by testing for the presence of autoantibodies[6] and/or declining levels/absence of C-peptide.

There is no known way to prevent type 1 diabetes.[4] Treatment with insulin is required for survival.[1] Insulin therapy is usually given by injection just under the skin but can also be delivered by an insulin pump.[11] A diabetic diet and exercise are important parts of management.[2] If left untreated, diabetes can cause many complications.[4] Complications of relatively rapid onset include diabetic ketoacidosis and nonketotic hyperosmolar coma.[6] Long-term complications include heart disease, stroke, kidney failure, foot ulcers and damage to the eyes.[4] Furthermore, since insulin lowers blood sugar levels, complications may arise from low blood sugar if more insulin is taken than necessary.[6]

Type 1 diabetes makes up an estimated 5–10% of all diabetes cases.[9] The number of people affected globally is unknown, although it is estimated that about 80,000 children develop the disease each year.[6] Within the United States the number of people affected is estimated at one to three million.[6][12] Rates of disease vary widely, with approximately one new case per 100,000 per year in East Asia and Latin America and around 30 new cases per 100,000 per year in Scandinavia and Kuwait.[13][14] It typically begins in children and young adults.[1]

Signs and symptoms edit

 
Overview of the most significant symptoms of diabetes

Type 1 diabetes begins suddenly, typically in childhood or adolescence.[15] The major sign of type 1 diabetes is very high blood sugar, which typically manifests in children as a few days to weeks of polyuria (increased urination), polydipsia (increased thirst), and weight loss.[16][17] Children may also experience increased appetite, blurred vision, bedwetting, recurrent skin infections, candidiasis of the perineum, irritability, and performance issues at school.[16][17] Adults with type 1 diabetes tend to have more varied symptoms that come on over months rather than days to weeks.[18][17]

Prolonged lack of insulin can also result in diabetic ketoacidosis, characterized by persistent fatigue, dry or flushed skin, abdominal pain, nausea or vomiting, confusion, trouble breathing, and a fruity breath odor.[18][19] Blood and urine tests reveal unusually high glucose and ketones in the blood and urine.[20] Untreated ketoacidosis can rapidly progress to loss of consciousness, coma, and death.[20] The percentage of children whose type 1 diabetes begins with an episode of diabetic ketoacidosis varies widely by geography, as low as 15% in parts of Europe and North America, and as high as 80% in the developing world.[20]

Cause edit

Type 1 diabetes is caused by the destruction of β-cells—the only cells in the body that produce insulin—and the consequent progressive insulin deficiency. Without insulin, the body is unable to respond effectively to increases in blood sugar. Due to this, people with diabetes have persistent hyperglycemia.[21] In 70–90% of cases, β-cells are destroyed by one's own immune system, for reasons that are not entirely clear.[21] The best-studied components of this autoimmune response are β-cell-targeted antibodies that begin to develop in the months or years before symptoms arise.[21] Typically someone will first develop antibodies against insulin or the protein GAD65, followed eventually by antibodies against the proteins IA-2, IA-2β, and/or ZNT8. People with more of these antibodies, and who develop them earlier in life, are at higher risk for developing symptomatic type 1 diabetes.[22] The trigger for the development of these antibodies remains unclear.[23] A number of explanatory theories have been put forward, and the cause may involve genetic susceptibility, a diabetogenic trigger, and/or exposure to an antigen.[24] The remaining 10–30% of type 1 diabetics have β-cell destruction but no sign of autoimmunity; this is called idiopathic type 1 diabetes and its cause is unknown.[21]

Environmental edit

Various environmental risks have been studied in an attempt to understand what triggers β-cell autoimmunity. Many aspects of environment and life history are associated with slight increases in type 1 diabetes risk, however the connection between each risk and diabetes often remains unclear.[citation needed] Type 1 diabetes risk is slightly higher for children whose mothers are obese or older than 35, or for children born by caesarean section.[25] Similarly, a child's weight gain in the first year of life, total weight, and BMI are associated with slightly increased type 1 diabetes risk.[25] Some dietary habits have also been associated with type 1 diabetes risk, namely consumption of cow's milk and dietary sugar intake.[25] Animal studies and some large human studies have found small associations between type 1 diabetes risk and intake of gluten or dietary fiber; however, other large human studies have found no such association.[25] Many potential environmental triggers have been investigated in large human studies and found to be unassociated with type 1 diabetes risk including duration of breastfeeding, time of introduction of cow milk into the diet, vitamin D consumption, blood levels of active vitamin D, and maternal intake of omega-3 fatty acids.[25][26]

A longstanding hypothesis for an environmental trigger is that some viral infection early in life contributes to type 1 diabetes development. Much of this work has focused on enteroviruses, with some studies finding slight associations with type 1 diabetes, and others finding none.[27] Large human studies have searched for, but not yet found an association between type 1 diabetes and various other viral infections, including infections of the mother during pregnancy.[27] Conversely, some have postulated that reduced exposure to pathogens in the developed world increases the risk of autoimmune diseases, often called the hygiene hypothesis. Various studies of hygiene-related factors—including household crowding, daycare attendance, population density, childhood vaccinations, antihelminth medication, and antibiotic usage during early life or pregnancy—show no association with type 1 diabetes.[28]

Genetics edit

Type 1 diabetes is partially caused by genetics, and family members of type 1 diabetics have a higher risk of developing the disease themselves. In the general population, the risk of developing type 1 diabetes is around 1 in 250. For someone whose parent has type 1 diabetes, the risk rises to 1–9%. If a sibling has type 1 diabetes, the risk is 6–7%. If someone's identical twin has type 1 diabetes, they have a 30–70% risk of developing it themselves.[29]

About half of the disease's heritability is due to variations in three HLA class II genes involved in antigen presentation: HLA-DRB1, HLA-DQA1, and HLA-DQB1.[29] The variation patterns associated with increased risk of type 1 diabetes are called HLA-DR3 and HLA-DR4-HLA-DQ8, and are common in people of European descent. A pattern associated with reduced risk of type 1 diabetes is called HLA-DR15-HLA-DQ6.[29] Large genome-wide association studies have identified dozens of other genes associated with type 1 diabetes risk, mostly genes involved in the immune system.[29]

Chemicals and drugs edit

Some medicines can reduce insulin production or damage β cells, resulting in disease that resembles type 1 diabetes. The antiviral drug didanosine triggers pancreas inflammation in 5 to 10% of those who take it, sometimes causing lasting β-cell damage.[30] Similarly, up to 5% of those who take the anti-protozoal drug pentamidine experience β-cell destruction and diabetes.[30] Several other drugs cause diabetes by reversibly reducing insulin secretion, namely statins (which may also damage β cells), the post-transplant immunosuppressants cyclosporin A and tacrolimus, the leukemia drug L-asparaginase, and the antibiotic gatifloxicin.[30][31] Pyrinuron (Vacor), a rodenticide introduced in the United States in 1976, selectively destroys pancreatic beta cells, resulting in type 1 diabetes after accidental poisoning.[32] Pyrinuron was withdrawn from the U.S. market in 1979.[33] Cancer Immunotherapy Drugs have also destroyed pancreatic beta cells, such as Opdivo among others.[34]

Diagnosis edit

Diabetes is typically diagnosed by a blood test showing unusually high blood sugar. The World Health Organization defines diabetes as blood sugar levels at or above 7.0 mmol/L (126 mg/dL) after fasting for at least eight hours, or a glucose level at or above 11.1 mmol/L (200 mg/dL) two hours after an oral glucose tolerance test.[35] The American Diabetes Association additionally recommends a diagnosis of diabetes for anyone with symptoms of hyperglycemia and blood sugar at any time at or above 11.1 mmol/L, or glycated hemoglobin (hemoglobin A1C) levels at or above 48 mmol/mol.[36]

Once a diagnosis of diabetes is established, type 1 diabetes is distinguished from other types by a blood test for the presence of autoantibodies that target various components of the beta cell.[37] The most commonly available tests detect antibodies against glutamic acid decarboxylase, the beta cell cytoplasm, or insulin, each of which are targeted by antibodies in around 80% of type 1 diabetics.[37] Some healthcare providers also have access to tests for antibodies targeting the beta cell proteins IA-2 and ZnT8; these antibodies are present in around 58% and 80% of type 1 diabetics respectively.[37] Some also test for C-peptide, a byproduct of insulin synthesis. Very low C-peptide levels are suggestive of type 1 diabetes.[37]

Management edit

The mainstay of type 1 diabetes treatment is the regular injection of insulin to manage hyperglycemia.[38] Injections of insulin via subcutaneous injection using either a syringe or an insulin pump are necessary multiple times per day, adjusting dosages to account for food intake, blood glucose levels and physical activity.[38] The goal of treatment is to maintain blood sugar in a normal range—80–130 mg/dL before a meal; <180 mg/dL after—as often as possible.[39] To achieve this, people with diabetes often monitor their blood glucose levels at home. Around 83% of type 1 diabetics monitor their blood glucose by capillary blood testing: pricking the finger to draw a drop of blood, and determining blood glucose with a glucose meter.[40] The American Diabetes Association recommends testing blood glucose around 6–10 times per day: before each meal, before exercise, at bedtime, occasionally after a meal, and any time someone feels the symptoms of hypoglycemia.[40] Around 17% of people with type 1 diabetes use a continuous glucose monitor, a device with a sensor under the skin that constantly measures glucose levels and communicates those levels to an external device.[40] Continuous glucose monitoring is associated with better blood sugar control than capillary blood testing alone; however, continuous glucose monitoring tends to be substantially more expensive.[40] Healthcare providers can also monitor someone's hemoglobin A1C levels which reflect the average blood sugar over the last three months.[41] The American Diabetes Association recommends a goal of keeping hemoglobin A1C levels under 7% for most adults and 7.5% for children.[41][42]

The goal of insulin therapy is to mimic normal pancreatic insulin secretion: low levels of insulin constantly present to support basic metabolism, plus the two-phase secretion of additional insulin in response to high blood sugar, then an extended phase of continued insulin secretion.[43] This is accomplished by combining different insulin preparations that act with differing speeds and durations. The standard of care for type 1 diabetes is a bolus of rapid-acting insulin 10–15 minutes before each meal or snacks, and as-needed to correct hyperglycemia.[43] In addition, constant low levels of insulin are achieved with one or two daily doses of long-acting insulin, or by steady infusion of low insulin levels by an insulin pump.[43] The exact dose of insulin appropriate for each injection depends on the content of the meal/snack, and the individual person's sensitivity to insulin, and is therefore typically calculated by the individual with diabetes or a family member by hand or assistive device (calculator, chart, mobile app, etc.).[43] People unable to manage these intensive insulin regimens are sometimes prescribed alternate plans relying on mixtures of rapid- or short-acting and intermediate-acting insulin, which are administered at fixed times along with meals of pre-planned times and carbohydrate composition.[43] The National Institute of Health and Care Excellence now recommends closed-loop insulin systems as an option for all women with type 1 diabetes who are pregnant or planning pregnancy.[44][45][46]

A non-insulin medication approved by the U.S. Food and Drug Administration for treating type 1 diabetes is the amylin analog pramlintide, which replaces the beta-cell hormone amylin. Addition of pramlintide to mealtime insulin injections reduces the boost in blood sugar after a meal, improving blood sugar control.[47] Occasionally, metformin, GLP-1 receptor agonists, Dipeptidyl peptidase-4 inhibitors, or SGLT2 inhibitor are prescribed off-label to people with type 1 diabetes, although fewer than 5% of type 1 diabetics use these drugs.[38]

Lifestyle edit

Besides insulin, the major way type 1 diabetics control their blood sugar is by learning how various foods impact their blood sugar levels. This is primarily done by tracking their intake of carbohydrates, the type of food with the greatest impact on blood sugar.[48] In general, people with type 1 diabetes are advised to follow an individualized eating plan rather than a pre-decided one.[49] There are camps for children to teach them how and when to use or monitor their insulin without parental help.[50] As psychological stress may have a negative effect on diabetes, a number of measures have been recommended including: exercising, taking up a new hobby, or joining a charity, among others.[51]

Regular exercise is important for maintaining general health, though the effect of exercise on blood sugar can be challenging to predict.[52] Exogenous insulin can drive down blood sugar, leaving those with diabetes at risk of hypoglycemia during and immediately after exercise, then again seven to eleven hours after exercise (called the "lag effect").[52] Conversely, high-intensity exercise can result in a shortage of insulin, and consequent hyperglycemia.[52] The risk of hypoglycemia can be managed by beginning exercise when blood sugar is relatively high (above 100 mg/dL), ingesting carbohydrates during or shortly after exercise, and reducing the amount of injected insulin within two hours of the planned exercise.[52] Similarly, the risk of exercise-induced hyperglycemia can be managed by avoiding exercise when insulin levels are very low, when blood sugar is extremely high (above 350 mg/dL), or when one feels unwell.[52]

Transplant edit

In some cases, people can receive transplants of the pancreas or isolated islet cells to restore insulin production and alleviate diabetic symptoms. Transplantation of the whole pancreas is rare, due in part to the few available donor organs, and to the need for lifelong immunosuppressive therapy to prevent transplant rejection.[53][54] The American Diabetes Association recommends pancreas transplant only in people who also require a kidney transplant, or who struggle to perform regular insulin therapy and experience repeated severe side effects of poor blood sugar control.[54] Most pancreas transplants are done simultaneously with a kidney transplant, with both organs from the same donor.[55] The transplanted pancreas continues to function for at least five years in around three quarters of recipients, allowing them to stop taking insulin.[56]

Transplantations of islets alone have become increasingly common.[57] Pancreatic islets are isolated from a donor pancreas, then injected into the recipient's portal vein from which they implant onto the recipient's liver.[58] In nearly half of recipients, the islet transplant continues to work well enough that they still do not need exogenous insulin five years after transplantation.[59] If a transplant fails, recipients can receive subsequent injections of islets from additional donors into the portal vein.[58] Like with whole pancreas transplantation, islet transplantation requires lifelong immunosuppression and depends on the limited supply of donor organs; it is therefore similarly limited to people with severe poorly controlled diabetes and those who have had or are scheduled for a kidney transplant.[57][60]

Donislecel (Lantidra) allogeneic (donor) pancreatic islet cellular therapy was approved for medical use in the United States in June 2023.[61]

Pathogenesis edit

Type 1 diabetes is a result of the destruction of pancreatic beta cells, although what triggers that destruction remains unclear.[62] People with type 1 diabetes tend to have more CD8+ T-cells and B-cells that specifically target islet antigens than those without type 1 diabetes, suggesting a role for the adaptive immune system in beta cell destruction.[62][63] Type 1 diabetics also tend to have reduced regulatory T cell function, which may exacerbate autoimmunity.[62] Destruction of beta cells results in inflammation of the islet of Langerhans, called insulitis. These inflamed islets tend to contain CD8+ T-cells and – to a lesser extent – CD4+ T cells.[62] Abnormalities in the pancreas or the beta cells themselves may also contribute to beta-cell destruction. The pancreases of people with type 1 diabetes tend to be smaller, lighter, and have abnormal blood vessels, nerve innervations, and extracellular matrix organization.[64] In addition, beta cells from people with type 1 diabetes sometimes overexpress HLA class I molecules (responsible for signaling to the immune system) and have increased endoplasmic reticulum stress and issues with synthesizing and folding new proteins, any of which could contribute to their demise.[64]

The mechanism by which the beta cells actually die likely involves both necroptosis and apoptosis induced or exacerbated by CD8+ T-cells and macrophages.[65] Necroptosis can be triggered by activated T cells – which secrete toxic granzymes and perforin – or indirectly as a result of reduced blood flow or the generation of reactive oxygen species.[65] As some beta cells die, they may release cellular components that amplify the immune response, exacerbating inflammation and cell death.[65] Pancreases from people with type 1 diabetes also have signs of beta cell apoptosis, linked to activation of the janus kinase and TYK2 pathways.[65]

Partial ablation of beta-cell function is enough to cause diabetes; at diagnosis, people with type 1 diabetes often still have detectable beta-cell function. Once insulin therapy is started, many people experience a resurgence in beta-cell function, and can go some time with little-to-no insulin treatment – called the "honeymoon phase".[64] This eventually fades as beta-cells continue to be destroyed, and insulin treatment is required again.[64] Beta-cell destruction is not always complete, as 30–80% of type 1 diabetics produce small amounts of insulin years or decades after diagnosis.[64]

Alpha cell dysfunction edit

Onset of autoimmune diabetes is accompanied by impaired ability to regulate the hormone glucagon,[66] which acts in antagonism with insulin to regulate blood sugar and metabolism. Progressive beta cell destruction leads to dysfunction in the neighboring alpha cells which secrete glucagon, exacerbating excursions away from euglycemia in both directions; overproduction of glucagon after meals causes sharper hyperglycemia, and failure to stimulate glucagon upon hypoglycemia prevents a glucagon-mediated rescue of glucose levels.[67]

Hyperglucagonemia edit

Onset of type 1 diabetes is followed by an increase in glucagon secretion after meals. Increases have been measured up to 37% during the first year of diagnosis, while c-peptide levels (indicative of islet-derived insulin), decline by up to 45%.[68] Insulin production will continue to fall as the immune system destroys beta cells, and islet-derived insulin will continue to be replaced by therapeutic exogenous insulin. Simultaneously, there is measurable alpha cell hypertrophy and hyperplasia in the early stage of the disease, leading to expanded alpha cell mass. This, together with failing beta cell insulin secretion, begins to account for rising glucagon levels that contribute to hyperglycemia.[67] Some researchers believe glucagon dysregulation to be the primary cause of early stage hyperglycemia.[69] Leading hypotheses for the cause of postprandial hyperglucagonemia suggest that exogenous insulin therapy is inadequate to replace the lost intraislet signalling to alpha cells previously mediated by beta cell-derived pulsatile insulin secretion.[70][71] Under this working hypothesis intensive insulin therapy has attempted to mimic natural insulin secretion profiles in exogenous insulin infusion therapies.[72] In young people with type 1 diabetes, unexplained deaths could be due to nighttime hypoglycemia triggering abnormal heart rhythms or cardiac autonomic neuropathy, damage to nerves that control the function of the heart.

Hypoglycemic glucagon impairment edit

Glucagon secretion is normally increased upon falling glucose levels, but normal glucagon response to hypoglycemia is blunted in type 1 diabetics.[73][74] Beta cell glucose sensing and subsequent suppression of administered insulin secretion is absent, leading to islet hyperinsulinemia which inhibits glucagon release.[73][75]

Autonomic inputs to alpha cells are much more important for glucagon stimulation in the moderate to severe ranges of hypoglycemia, yet the autonomic response is blunted in a number of ways. Recurrent hypoglycemia leads to metabolic adjustments in the glucose sensing areas of the brain, shifting the threshold for counter regulatory activation of the sympathetic nervous system to lower glucose concentration.[75] This is known as hypoglycemic unawareness. Subsequent hypoglycemia is met with impairment in sending of counter regulatory signals to the islets and adrenal cortex. This accounts for the lack of glucagon stimulation and epinephrine release that would normally stimulate and enhance glucose release and production from the liver, rescuing the diabetic from severe hypoglycemia, coma, and death. Numerous hypotheses have been produced in the search for a cellular mechanism of hypoglycemic unawareness, and a consensus has yet to be reached.[76] The major hypotheses are summarized in the following table:[77][75][76]

Mechanisms of hypoglycemic unawareness
Glycogen supercompensation Increased glycogen stores in astrocytes might contribute supplementary glycosyl units for metabolism, counteracting the central nervous system perception of hypoglycemia.
Enhanced glucose metabolism Altered glucose transport and enhanced metabolic efficiency upon recurring hypoglycemia relieves oxidative stress that would activate sympathetic response.
Alternative fuel hypothesis Decreased reliance on glucose, supplementation of lactate from astrocytes, or ketones meet metabolic demands and reduce stress to brain.
Brain neuronal communication Hypothalamic inhibitory GABA normally decreases during hypoglycemia, disinhibiting signals for sympathetic tone. Recurrent episodes of hypoglycemia result in increased basal GABA which fails to decrease normally during subsequent hypoglycemia. Inhibitory tone remains and sympathetic tone is not increased.

In addition, autoimmune diabetes is characterized by a loss of islet specific sympathetic innervation.[78] This loss constitutes an 80–90% reduction of islet sympathetic nerve endings, happens early in the progression of the disease, and is persistent though the life of the patient.[79] It is linked to the autoimmune aspect of type 1 diabetics and fails to occur in type 2 diabetics. Early in the autoimmune event, the axon pruning is activated in islet sympathetic nerves. Increased BDNF and ROS that result from insulitis and beta cell death stimulate the p75 neurotrophin receptor (p75NTR), which acts to prune off axons. Axons are normally protected from pruning by activation of tropomyosin receptor kinase A (Trk A) receptors by NGF, which in islets is primarily produced by beta cells. Progressive autoimmune beta cell destruction, therefore, causes both the activation of pruning factors and the loss of protective factors to the islet sympathetic nerves. This unique form of neuropathy is a hallmark of type 1 diabetes, and plays a part in the loss of glucagon rescue of severe hypoglycemia.[78]

Complications edit

The most pressing complication of type 1 diabetes are the always present risks of poor blood sugar control: severe hypoglycemia and diabetic ketoacidosis. Hypoglycemia – typically blood sugar below 70 mg/dL – triggers the release of epinephrine, and can cause people to feel shaky, anxious, or irritable.[80] People with hypoglycemia may also experience hunger, nausea, sweats, chills, dizziness, and a fast heartbeat.[80] Some feel lightheaded, sleepy, or weak.[80] Severe hypoglycemia can develop rapidly, causing confusion, coordination problems, loss of consciousness, and seizure.[80][81] On average, people with type 1 diabetes experience a hypoglycemia event that requires assistance of another 16–20 times in 100 person-years, and an event leading to unconsciousness or seizure 2–8 times per 100 person-years.[81] The American Diabetes Association recommends treating hypoglycemia by the "15-15 rule": eat 15 grams of carbohydrates, then wait 15 minutes before checking blood sugar; repeat until blood sugar is at least 70 mg/dL.[80] Severe hypoglycemia that impairs someone's ability to eat is typically treated with injectable glucagon, which triggers glucose release from the liver into the bloodstream.[80] People with repeated bouts of hypoglycemia can develop hypoglycemia unawareness, where the blood sugar threshold at which they experience symptoms of hypoglycemia decreases, increasing their risk of severe hypoglycemic events.[82] Rates of severe hypoglycemia have generally declined due to the advent of rapid-acting and long-acting insulin products in the 1990s and early 2000s;[43] however, acute hypoglycemic still causes 4–10% of type 1 diabetes-related deaths.[81]

The other persistent risk is diabetic ketoacidosis – a state where lack of insulin results in cells burning fat rather than sugar, producing toxic ketones as a byproduct.[19] Ketoacidosis symptoms can develop rapidly, with frequent urination, excessive thirst, nausea, vomiting, and severe abdominal pain all common.[83] More severe ketoacidosis can result in labored breathing, and loss of consciousness due to cerebral edema.[83] People with type 1 diabetes experience diabetic ketoacidosis 1–5 times per 100 person-years, the majority of which result in hospitalization.[84] 13–19% of type 1 diabetes-related deaths are caused by ketoacidosis,[81] making ketoacidosis the leading cause of death in people with type 1 diabetes less than 58 years old.[84]

Long-term complications edit

In addition to the acute complications of diabetes, long-term hyperglycemia results in damage to the small blood vessels throughout the body. This damage tends to manifest particularly in the eyes, nerves, and kidneys causing diabetic retinopathy, diabetic neuropathy, and diabetic nephropathy respectively.[82] In the eyes, prolonged high blood sugar causes the blood vessels in the retina to become fragile.[85]

People with type 1 diabetes also have increased risk of cardiovascular disease, which is estimated to shorten the life of the average type 1 diabetic by 8–13 years.[86] Cardiovascular disease[87] as well as neuropathy[88] may have an autoimmune basis, as well. Women with type 1 DM have a 40% higher risk of death as compared to men with type 1 DM.[89]

About 12 percent of people with type 1 diabetes have clinical depression.[90] About 6 percent of people with type 1 diabetes also have celiac disease, but in most cases there are no digestive symptoms[7][91] or are mistakenly attributed to poor control of diabetes, gastroparesis or diabetic neuropathy.[91] In most cases, celiac disease is diagnosed after onset of type 1 diabetes. The association of celiac disease with type 1 diabetes increases the risk of complications, such as retinopathy and mortality. This association can be explained by shared genetic factors, and inflammation or nutritional deficiencies caused by untreated celiac disease, even if type 1 diabetes is diagnosed first.[7]

Urinary tract infection edit

People with diabetes show an increased rate of urinary tract infection.[92] The reason is bladder dysfunction is more common in people with diabetes than people without diabetes due to diabetes nephropathy. When present, nephropathy can cause a decrease in bladder sensation, which in turn, can cause increased residual urine, a risk factor for urinary tract infections.[93]

Sexual dysfunction edit

Sexual dysfunction in people with diabetes is often a result of physical factors such as nerve damage and poor circulation, and psychological factors such as stress and/or depression caused by the demands of the disease.[94] The most common sexual issues in males with diabetes are problems with erections and ejaculation: "With diabetes, blood vessels supplying the penis's erectile tissue can get hard and narrow, preventing the adequate blood supply needed for a firm erection. The nerve damage caused by poor blood glucose control can also cause ejaculate to go into the bladder instead of through the penis during ejaculation, called retrograde ejaculation. When this happens, semen leaves the body in the urine." Another cause of erectile dysfunction is reactive oxygen species created as a result of the disease. Antioxidants can be used to help combat this.[95] Sexual problems are common in women who have diabetes,[94] including reduced sensation in the genitals, dryness, difficulty/inability to orgasm, pain during sex, and decreased libido. Diabetes sometimes decreases estrogen levels in females, which can affect vaginal lubrication. Less is known about the correlation between diabetes and sexual dysfunction in females than in males.[94]

Oral contraceptive pills can cause blood sugar imbalances in women who have diabetes. Dosage changes can help address that, at the risk of side effects and complications.[94]

Women with type 1 diabetes show a higher than normal rate of polycystic ovarian syndrome (PCOS).[96] The reason may be that the ovaries are exposed to high insulin concentrations since women with type 1 diabetes can have frequent hyperglycemia.[97]

Autoimmune disorders edit

People with type 1 diabetes are at an increased risk for developing several autoimmune disorders, particularly thyroid problems – around 20% of people with type 1 diabetes have hypothyroidism or hyperthyroidism, typically caused by Hashimoto thyroiditis or Graves' disease respectiveley.[98][81] Celiac disease affects 2–8% of people with type 1 diabetes, and is more common in those who were younger at diabetes diagnosis, and in white people.[98] Type 1 diabetics are also at increased risk of rheumatoid arthritis, lupus, autoimmune gastritis, pernicious anemia, vitiligo, and Addison's disease.[81] Conversely, complex autoimmune syndromes caused by mutations in the immunity-related genes AIRE (causing autoimmune polyglandular syndrome), FoxP3 (causing IPEX syndrome), or STAT3 include type 1 diabetes in their effects.[99]

Prevention edit

There is no way to prevent type 1 diabetes;[100] however, the development of diabetes symptoms can be delayed in some people who are at high risk of developing the disease. In 2022 the FDA approved an intravenous injection of teplizumab to delay the progression of type 1 diabetes in those older than eight who have already developed diabetes-related autoantibodies and problems with blood sugar control. In that population, the anti-CD3 monoclonal antibody teplizumab can delay the development of type 1 diabetes symptoms by around two years.[101]

In addition to anti-CD3 antibodies, several other immunosuppressive agents have been trialled with the aim of preventing beta cell destruction. Large trials of cyclosporine treatment suggested that cyclosporine could improve insulin secretion in those recently diagnosed with type 1 diabetes; however, people who stopped taking cyclosporine rapidly stopped making insulin, and cyclosporine's kidney toxicity and increased risk of cancer prevented people from using it long-term.[102] Several other immunosuppressive agents – prednisone, azathioprine, anti-thymocyte globulin, mycophenolate, and antibodies against CD20 and IL2 receptor α – have been the subject of research, but none have provided lasting protection from development of type 1 diabetes.[102] There have also been clinical trials attempting to induce immune tolerance by vaccination with insulin, GAD65, and various short peptides targeted by immune cells during type 1 diabetes; none have yet delayed or prevented development of disease.[103]

Several trials have attempted dietary interventions with the hope of reducing the autoimmunity that leads to type 1 diabetes. Trials that withheld cow's milk or gave infants formula free of bovine insulin decreased the development of β-cell-targeted antibodies, but did not prevent the development of type 1 diabetes.[104] Similarly, trials that gave high-risk individuals injected insulin, oral insulin, or nicotinamide did not prevent diabetes development.[104]

Epidemiology edit

Type 1 diabetes makes up an estimated 10–15% of all diabetes cases[22] or 11–22 million cases worldwide.[105] Symptoms can begin at any age, but onset is most common in children, with diagnoses slightly more common in 5 to 7 year olds, and much more common around the age of puberty.[106][15] In contrast to most autoimmune diseases, type 1 diabetes is slightly more common in males than in females.[106]

In 2006, type 1 diabetes affected 440,000 children under 14 years of age and was the primary cause of diabetes in those less than 15 years of age.[107][22]

Rates vary widely by country and region. Incidence is highest in Scandinavia, at 30–60 new cases per 100,000 children per year, intermediate in the U.S. and Southern Europe at 10–20 cases per 100,000 per year, and lowest in China, much of Asia, and South America at 1–3 cases per 100,000 per year.[26]

In the United States, type 1 and 2 diabetes affected about 208,000 youths under the age of 20 in 2015. Over 18,000 youths are diagnosed with Type 1 diabetes every year. Every year about 234,051 Americans die due to diabetes (type I or II) or diabetes-related complications, with 69,071 having it as the primary cause of death.[108]

In Australia, about one million people have been diagnosed with diabetes and of this figure 130,000 people have been diagnosed with type 1 diabetes. Australia ranks 6th-highest in the world with children under 14 years of age. Between 2000 and 2013, 31,895 new cases were established, with 2,323 in 2013, a rate of 10–13 cases per 100,00 people each year. Aboriginals and Torres Strait Islander people are less affected.[109][110]

Since the 1950s, the incidence of type 1 diabetes has been gradually increasing across the world by an average 3–4% per year.[26] The increase is more pronounced in countries that began with a lower incidence of type 1 diabetes.[26] A single 2023 study suggested a relationship between COVID-19 infection and the incidence of type 1 diabetes in children;[111] confirmatory studies have not appeared to date.

History edit

The connection between diabetes and pancreatic damage was first described by the German pathologist Martin Schmidt, who in a 1902 paper noted inflammation around the pancreatic islet of a child who had died of diabetes.[112] The connection between this inflammation and diabetes onset was further developed through the 1920s by Shields Warren, and the term "insulitis" was coined by Hanns von Meyenburg in 1940 to describe the phenomenon.[112]

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.[113] 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.[114] The name juvenile diabetes was used earlier as it often first is diagnosed in childhood.

Society and culture edit

Type 1 and 2 diabetes 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.[115] In the United States $245 billion every year is attributed to diabetes. Individuals diagnosed with diabetes have 2.3 times the health care costs as individuals who do not have diabetes. One in ten health care dollars are spent on individuals with type 1 and 2 diabetes.[108]

Research edit

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 Institutes of Health, and in the UK via the National Institute for Health and Care Research or the Medical Research Council. The Juvenile Diabetes Research Foundation (JDRF), founded by parents of children with type 1 diabetes, is the world's largest provider of charity-based funding for type 1 diabetes research.[citation needed] Other charities include the American Diabetes Association, Diabetes UK, Diabetes Research and Wellness Foundation,[116] Diabetes Australia, and the Canadian Diabetes Association.

Organ replacement edit

Pluripotent stem cells can be used to generate beta cells but previously these cells did not function as well as normal beta cells.[117] In 2014 more mature beta cells were produced which released insulin in response to blood sugar when transplanted into mice.[118][119] Before these techniques can be used in humans, more evidence of safety and effectiveness is needed.[117]

There has also been substantial effort to develop a fully automated insulin delivery system or "artificial pancreas" that could sense glucose levels and inject appropriate insulin without conscious input from the user.[120] Current "hybrid closed-loop systems" use a continuous glucose monitor to sense blood sugar levels, and a subcutaneous insulin pump to deliver insulin; however, due to the delay between insulin injection and its action, current systems require the user to initiate insulin before taking meals.[121] Several improvements to these systems are currently undergoing clinical trials in humans, including a dual-hormone system that injects glucagon in addition to insulin, and an implantable device that injects insulin intraperitoneally where it can be absorbed more quickly.[122]

Disease models edit

Various animal models of disease are used to understand the pathogenesis and etiology of type 1 diabetes. Currently available models of T1D can be divided into spontaneously autoimmune, chemically induced, virus induced and genetically induced.[123]

The nonobese diabetic (NOD) mouse is the most widely studied model of type 1 diabetes.[123] It is an inbred strain that spontaneously develops type 1 diabetes in 30–100% of female mice depending on housing conditions.[124] Diabetes in NOD mice is caused by several genes, primarily MHC genes involved in antigen presentation.[124] Like diabetic humans, NOD mice develop islet autoantibodies and inflammation in the islet, followed by reduced insulin production and hyperglycemia.[124][125] Some features of human diabetes are exaggerated in NOD mice, namely the mice have more severe islet inflammation than humans, and have a much more pronounced sex bias, with females developing diabetes far more frequently than males.[124] In NOD mice the onset of insulitis occurs at 3–4 weeks of age. The islets of Langerhans are infiltrated by CD4+, CD8+ T lymphocytes, NK cells, B lymphocytes, dendritic cells, macrophages and neutrophils, similar to the disease process in humans.[126] In addition to sex, breeding conditions, gut microbiome composition or diet also influence the onset of T1D.[127]

The BioBreeding Diabetes-Prone (BB) rat is another widely used spontaneous experimental model for T1D. The onset of diabetes occurs, in up to 90% of individuals (regardless of sex) at 8–16 weeks of age.[126] During insulitis, the pancreatic islets are infiltrated by T lymphocytes, B lymphocytes, macrophages, and NK cells, with the difference from the human course of insulitis being that CD4 + T lymphocytes are markedly reduced and CD8 + T lymphocytes are almost absent. The aforementioned lymphopenia is the major drawback of this model. The disease is characterized by hyperglycemia, hypoinsulinemia, weight loss, ketonuria, and the need for insulin therapy for survival.[126] BB Rats are used to study the genetic aspects of T1D and are also used for interventional studies and diabetic nephropathy studies.[128]

LEW-1AR1 / -iddm rats are derived from congenital Lewis rats and represent a rarer spontaneous model for T1D. These rats develop diabetes at about 8–9 weeks of age with no sex differences unlike NOD mice.[129] In LEW mice, diabetes presents with hyperglycemia, glycosuria, ketonuria, and polyuria.[130][126] The advantage of the model is the progression of the prediabetic phase, which is very similar to human disease, with infiltration of islet by immune cells about a week before hyperglycemia is observed. This model is suitable for intervention studies or for the search for predictive biomarkers. It is also possible to observe individual phases of pancreatic infiltration by immune cells. The advantage of congenic LEW mice is also the good viability after the manifestation of T1D (compared to NOD mice and BB rats).[131]

Chemically induced edit

The chemical compounds aloxan and streptozotocin (STZ) are commonly used to induce diabetes and destroy β-cells in mouse/rat animal models.[126] In both cases, it is a cytotoxic analog of glucose that passes GLUT2 transport and accumulates in β-cells, causing their destruction. The chemically induced destruction of β-cells leads to decreased insulin production, hyperglycemia and weight loss in the experimental animal.[132] The animal models prepared in this way are suitable for research into blood sugar-lowering drugs and therapies (e.g. for testing new insulin preparations). They are also the most commonly used genetically induced T1D model is the so-called AKITA mouse (originally C57BL/6NSIc mouse). The development of diabetes in AKITA mice is caused by a spontaneous point mutation in the Ins2 gene, which is responsible for the correct composition of insulin in the endoplasmic reticulum. Decreased insulin production is then associated with hyperglycemia, polydipsia and polyuria. If severe diabetes develops within 3–4 weeks, AKITA mice survive no longer than 12 weeks without treatment intervention. The description of the etiology of the disease shows that, unlike spontaneous models, the early stages of the disease are not accompanied by insulitis.[133] AKITA mice are used to test drugs targeting endoplasmic reticulum stress reduction, to test islet transplants, and to study diabetes-related complications such as nephropathy, sympathetic autonomic neuropathy, and vascular disease.[126][134] for testing transplantation therapies. Their advantage is mainly the low cost, the disadvantage is the cytotoxicity of the chemical compounds.[135]

Genetically induced edit

Virally induced edit

Viral infections play a role in the development of a number of autoimmune diseases, including human type 1 diabetes. However, the mechanisms by which viruses are involved in the induction of type 1 DM are not fully understood. Virus-induced models are used to study the etiology and pathogenesis of the disease, in particular the mechanisms by which environmental factors contribute to or protect against the occurrence of type 1 DM.[136] Among the most commonly used are Coxsackie virus, lymphocytic choriomeningitis virus, encephalomyocarditis virus, and Kilham rat virus. Examples of virus-induced animals include NOD mice infected with coxsackie B4 that developed type 1 DM within two weeks.[137]

References edit

  1. ^ a b c d e f g h "Causes of Diabetes". NIDDK. August 2014. from the original on 10 August 2016. Retrieved 31 July 2016.
  2. ^ a b c d e f g "Types of Diabetes". NIDDK. February 2014. from the original on 16 August 2016. Retrieved 31 July 2016.
  3. ^ . International Diabetes Federation. 17 March 2006. Archived from the original on 5 August 2007.
  4. ^ a b c d e f g h i j k "Diabetes Fact sheet N°312". WHO. November 2016. from the original on 26 August 2013. Retrieved 29 May 2017.
  5. ^ "Diabetes mellitus Type 1". Autoimmune Registry Inc. Retrieved 15 June 2022.
  6. ^ a b c d e f g h i Chiang JL, Kirkman MS, Laffel LM, Peters AL (July 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. PMC 5865481. PMID 24935775.
  7. ^ a b c Elfström P, Sundström J, Ludvigsson JF (November 2014). "Systematic review with meta-analysis: associations between coeliac disease and type 1 diabetes". Alimentary Pharmacology & Therapeutics. 40 (10): 1123–1132. doi:10.1111/apt.12973. PMID 25270960. S2CID 25468009.
  8. ^ a b "Diagnosis of Diabetes and Prediabetes". NIDDK. May 2015. from the original on 16 August 2016. Retrieved 31 July 2016.
  9. ^ a b Daneman D (March 2006). "Type 1 diabetes". Lancet. 367 (9513): 847–858. doi:10.1016/S0140-6736(06)68341-4. PMID 16530579. S2CID 21485081.
  10. ^ Torpy JM, Lynm C, Glass RM (September 2007). "JAMA patient page. Type 1 diabetes". JAMA. 298 (12): 1472. doi:10.1001/jama.298.12.1472. PMID 17895465.
  11. ^ "Alternative Devices for Taking Insulin". NIDDK. July 2016. from the original on 16 August 2016. Retrieved 31 July 2016.
  12. ^ "Fast Facts Data and Statistics about Diabetes". American Diabetes Association. from the original on 16 December 2015. Retrieved 25 July 2014.
  13. ^ Global report on diabetes (PDF). World Health Organization. 2016. pp. 26–27. ISBN 978-92-4-156525-7. (PDF) from the original on 7 October 2016. Retrieved 31 July 2016.
  14. ^ Skyler J (2012). Atlas of diabetes (4th ed.). New York: Springer. pp. 67–68. ISBN 978-1-4614-1028-7. from the original on 8 September 2017.
  15. ^ a b Atkinson et al. 2020, Table 36.1.
  16. ^ a b Wolsdorf & Garvey 2016, "Type 1 Diabetes".
  17. ^ a b c Atkinson et al. 2020, "Clinical presentation".
  18. ^ a b DiMeglio, Evans-Molina & Oram 2018, p. 2449.
  19. ^ a b "DKA (Ketoacidosis) & Ketones". American Diabetes Association. Retrieved 28 July 2021.
  20. ^ a b c Delli & Lernmark 2016, "Signs and symptoms".
  21. ^ a b c d Katsarou et al. 2017, p. 1.
  22. ^ a b c Katsarou et al. 2017, "Epidemiology".
  23. ^ Katsarou et al. 2017, "Introduction".
  24. ^ Knip M, Veijola R, Virtanen SM, Hyöty H, Vaarala O, Akerblom HK (December 2005). "Environmental triggers and determinants of type 1 diabetes". Diabetes. 54 (Suppl 2): S125–S136. doi:10.2337/diabetes.54.suppl_2.S125. PMID 16306330.
  25. ^ a b c d e Norris, Johnson & Stene 2020, "Environmental factors".
  26. ^ a b c d Norris, Johnson & Stene 2020, "Trends in epidemiology".
  27. ^ a b Norris, Johnson & Stene 2020, "Infections".
  28. ^ Norris, Johnson & Stene 2020, "The hygiene hypothesis and proxies of microbial exposures".
  29. ^ a b c d DiMeglio, Evans-Molina & Oram 2018, p. 2450.
  30. ^ a b c Repaske 2016, "Additional medications that decrease insulin release".
  31. ^ Repaske 2016, "A common medication that decreases insulin release".
  32. ^ Thayer KA, Heindel JJ, Bucher JR, Gallo MA (June 2012). "Role of environmental chemicals in diabetes and obesity: a National Toxicology Program workshop review". Environmental Health Perspectives (Review). 120 (6): 779–789. doi:10.1289/ehp.1104597. PMC 3385443. PMID 22296744.
  33. ^ "Pyriminil". U.S. National Library of Medicine. from the original on 4 July 2013.
  34. ^ Lucier, Jessica; Tabatabai, Laila (8 May 2020). "SAT-677 A Case of Opdivo Induced Type 1 Diabetes". Journal of the Endocrine Society. 4 (Suppl 1): SAT–677. doi:10.1210/jendso/bvaa046.469. PMC 7208640.
  35. ^ Definition and Diagnosis of Diabetes Mellitus and Intermediate Hyperglycemia (PDF). Geneva: World Health Organization. 2006. p. 1. ISBN 978-92-4-159493-6. Retrieved 28 July 2021.
  36. ^ American Diabetes Association (January 2021). "2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2021". Diabetes Care. American Diabetes Association. 44 (Suppl 1): S15–S33. doi:10.2337/dc21-S002. PMID 33298413.
  37. ^ a b c d Butler & Misselbrook 2020, "What is the next investigation?".
  38. ^ a b c DiMeglio, Evans-Molina & Oram 2018, p. 2453.
  39. ^ Katsarou et al. 2017, p. 11.
  40. ^ a b c d Smith & Harris 2018, "Self monitoring".
  41. ^ a b American Diabetes Association (6) 2021, "Glycemic assessment".
  42. ^ DiMeglio, Evans-Molina & Oram 2018, "Management of clinical disease".
  43. ^ a b c d e f Atkinson et al. 2020, "Insulin Therapy".
  44. ^ "Overview | Diabetes in pregnancy: management from preconception to the postnatal period | Guidance | NICE". www.nice.org.uk. 25 February 2015. Retrieved 31 January 2024.
  45. ^ Lee, Tara T.M.; Collett, Corinne; Bergford, Simon; Hartnell, Sara; Scott, Eleanor M.; Lindsay, Robert S.; Hunt, Katharine F.; McCance, David R.; Barnard-Kelly, Katharine; Rankin, David; Lawton, Julia; Reynolds, Rebecca M.; Flanagan, Emma; Hammond, Matthew; Shepstone, Lee (26 October 2023). "Automated Insulin Delivery in Women with Pregnancy Complicated by Type 1 Diabetes". New England Journal of Medicine. 389 (17): 1566–1578. doi:10.1056/NEJMoa2303911. ISSN 0028-4793.
  46. ^ "Closed-loop insulin systems are effective for pregnant women with type 1 diabetes". NIHR Evidence. 16 January 2024.
  47. ^ Atkinson et al. 2020, "Use of Adjunctive Drugs in T1DM".
  48. ^ Atkinson et al. 2020, "Nutrition Therapy".
  49. ^ Seckold R, Fisher E, de Bock M, King BR, Smart CE (March 2019). "The ups and downs of low-carbohydrate diets in the management of Type 1 diabetes: a review of clinical outcomes". Diabetic Medicine (Review). 36 (3): 326–334. doi:10.1111/dme.13845. PMID 30362180. S2CID 53102654. Low‐carbohydrate diets are of interest for improving glycaemic outcomes in the management of Type 1 diabetes. There is limited evidence to support their routine use in the management of Type 1 diabetes.
  50. ^ Ly TT (2015). "Technology and type 1 diabetes: Closed-loop therapies". Current Pediatrics Reports. 3 (2): 170–176. doi:10.1007/s40124-015-0083-y. S2CID 68302123.
  51. ^ "Stress". diabetes.org. American Diabetes Association. from the original on 12 November 2014. Retrieved 11 November 2014.
  52. ^ a b c d e Atkinson et al. 2020, "Physical Activity and Exercise".
  53. ^ Atkinson et al. 2020, "Pancreas and Islet Cell Transplantation".
  54. ^ a b Robertson RP, Davis C, Larsen J, Stratta R, Sutherland DE (April 2006). "Pancreas and islet transplantation in type 1 diabetes". Diabetes Care. 29 (4): 935. doi:10.2337/diacare.29.04.06.dc06-9908. PMID 16567844.
  55. ^ Dean et al. 2017, "Simultaneous pancreas-kidney transplant".
  56. ^ Dean et al. 2017, "Outcomes of pancreas transplantation".
  57. ^ a b Shapiro, Pokrywczynska & Ricordi 2017, "Main".
  58. ^ a b Rickels & Robertson 2019, "Islet allotransplantation for the treatment of type 1 diabetes".
  59. ^ Rickels & Robertson 2019, "Long-term outcomes and comparison with pancreas transplantation".
  60. ^ Shapiro, Pokrywczynska & Ricordi 2017, "Indications for islet transplantation".
  61. ^ "FDA Approves First Cellular Therapy to Treat Patients with Type 1 Diabetes". U.S. Food and Drug Administration (FDA) (Press release). 28 June 2023. Retrieved 28 June 2023.   This article incorporates text from this source, which is in the public domain.
  62. ^ a b c d DiMeglio, Evans-Molina & Oram 2018, "The immune phenotype of type 1 diabetes".
  63. ^ DiMeglio, Evans-Molina & Oram 2018, "Diagnosis".
  64. ^ a b c d e DiMeglio, Evans-Molina & Oram 2018, "The β-cell phenotype of type 1 diabetes".
  65. ^ a b c d Atkinson et al. 2020, "Mechanisms of Beta-Cell Death in T1DM".
  66. ^ Farhy LS, McCall AL (July 2015). "Glucagon – the new 'insulin' in the pathophysiology of diabetes". Current Opinion in Clinical Nutrition and Metabolic Care. 18 (4): 407–414. doi:10.1097/mco.0000000000000192. PMID 26049639. S2CID 19872862.
  67. ^ a b Yosten GL (February 2018). "Alpha cell dysfunction in type 1 diabetes". Peptides. 100: 54–60. doi:10.1016/j.peptides.2017.12.001. PMID 29412832. S2CID 46878644.
  68. ^ Brown RJ, Sinaii N, Rother KI (July 2008). "Too much glucagon, too little insulin: time course of pancreatic islet dysfunction in new-onset type 1 diabetes". Diabetes Care. 31 (7): 1403–1404. doi:10.2337/dc08-0575. PMC 2453684. PMID 18594062.
  69. ^ Unger RH, Cherrington AD (January 2012). "Glucagonocentric restructuring of diabetes: a pathophysiologic and therapeutic makeover". The Journal of Clinical Investigation. 122 (1): 4–12. doi:10.1172/JCI60016. PMC 3248306. PMID 22214853.
  70. ^ Meier JJ, Kjems LL, Veldhuis JD, Lefèbvre P, Butler PC (April 2006). "Postprandial suppression of glucagon secretion depends on intact pulsatile insulin secretion: further evidence for the intraislet insulin hypothesis". Diabetes. 55 (4): 1051–1056. doi:10.2337/diabetes.55.04.06.db05-1449. PMID 16567528.
  71. ^ Cooperberg BA, Cryer PE (December 2009). "Beta-cell-mediated signaling predominates over direct alpha-cell signaling in the regulation of glucagon secretion in humans". Diabetes Care. 32 (12): 2275–2280. doi:10.2337/dc09-0798. PMC 2782990. PMID 19729529.
  72. ^ Paolisso G, Sgambato S, Torella R, Varricchio M, Scheen A, D'Onofrio F, Lefèbvre PJ (June 1988). "Pulsatile insulin delivery is more efficient than continuous infusion in modulating islet cell function in normal subjects and patients with type 1 diabetes". The Journal of Clinical Endocrinology and Metabolism. 66 (6): 1220–1226. doi:10.1210/jcem-66-6-1220. PMID 3286673.
  73. ^ a b Banarer S, McGregor VP, Cryer PE (April 2002). "Intraislet hyperinsulinemia prevents the glucagon response to hypoglycemia despite an intact autonomic response". Diabetes. 51 (4): 958–965. doi:10.2337/diabetes.51.4.958. PMID 11916913.
  74. ^ Raju B, Cryer PE (March 2005). "Loss of the decrement in intraislet insulin plausibly explains loss of the glucagon response to hypoglycemia in insulin-deficient diabetes: documentation of the intraislet insulin hypothesis in humans". Diabetes. 54 (3): 757–764. doi:10.2337/diabetes.54.3.757. PMID 15734853.
  75. ^ a b c Tesfaye N, Seaquist ER (November 2010). "Neuroendocrine responses to hypoglycemia". Annals of the New York Academy of Sciences. 1212 (1): 12–28. Bibcode:2010NYASA1212...12T. doi:10.1111/j.1749-6632.2010.05820.x. PMC 2991551. PMID 21039590.
  76. ^ a b Reno CM, Litvin M, Clark AL, Fisher SJ (March 2013). "Defective counterregulation and hypoglycemia unawareness in diabetes: mechanisms and emerging treatments". Endocrinology and Metabolism Clinics of North America. 42 (1): 15–38. doi:10.1016/j.ecl.2012.11.005. PMC 3568263. PMID 23391237.
  77. ^ Martín-Timón I, Del Cañizo-Gómez FJ (July 2015). "Mechanisms of hypoglycemia unawareness and implications in diabetic patients". World Journal of Diabetes. 6 (7): 912–926. doi:10.4239/wjd.v6.i7.912. PMC 4499525. PMID 26185599.
  78. ^ a b Mundinger TO, Taborsky GJ (October 2016). "Early sympathetic islet neuropathy in autoimmune diabetes: lessons learned and opportunities for investigation". Diabetologia. 59 (10): 2058–2067. doi:10.1007/s00125-016-4026-0. PMC 6214182. PMID 27342407.
  79. ^ Mundinger TO, Mei Q, Foulis AK, Fligner CL, Hull RL, Taborsky GJ (August 2016). "Human Type 1 Diabetes Is Characterized by an Early, Marked, Sustained, and Islet-Selective Loss of Sympathetic Nerves". Diabetes. 65 (8): 2322–2330. doi:10.2337/db16-0284. PMC 4955989. PMID 27207540.
  80. ^ a b c d e f "Hypoglycemia (Low blood sugar)". American Diabetes Association. Retrieved 20 March 2022.
  81. ^ a b c d e f DiMeglio, Evans-Molina & Oram 2018, p. 2455.
  82. ^ a b DiMeglio, Evans-Molina & Oram 2018, "Complications of type 1 diabetes".
  83. ^ a b Cashen & Petersen 2019, "Diagnosis, screening and prevention".
  84. ^ a b Cashen & Petersen 2019, "Epidemiology".
  85. ^ Brownlee et al. 2020, "Pathophysiology of diabetic retinopathy".
  86. ^ DiMeglio, Evans-Molina & Oram 2018, p. 2456.
  87. ^ 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): 774–779. doi:10.2337/diabetes.55.03.06.db05-1417. PMID 16505242.
  88. ^ Granberg V, Ejskjaer N, Peakman M, Sundkvist G (August 2005). "Autoantibodies to autonomic nerves associated with cardiac and peripheral autonomic neuropathy". Diabetes Care. 28 (8): 1959–1964. doi:10.2337/diacare.28.8.1959. PMID 16043739.
  89. ^ Huxley RR, Peters SA, Mishra GD, Woodward M (March 2015). "Risk of all-cause mortality and vascular events in women versus men with type 1 diabetes: a systematic review and meta-analysis". The Lancet. Diabetes & Endocrinology. 3 (3): 198–206. doi:10.1016/S2213-8587(14)70248-7. PMID 25660575.
  90. ^ Roy T, Lloyd CE (October 2012). "Epidemiology of depression and diabetes: a systematic review". Journal of Affective Disorders. 142 (Suppl): S8–21. doi:10.1016/S0165-0327(12)70004-6. PMID 23062861.
  91. ^ a b See JA, Kaukinen K, Makharia GK, Gibson PR, Murray JA (October 2015). "Practical insights into gluten-free diets". Nature Reviews. Gastroenterology & Hepatology (Review). 12 (10): 580–591. doi:10.1038/nrgastro.2015.156. PMID 26392070. S2CID 20270743. Coeliac disease in T1DM is asymptomatic ...Clinical manifestations of coeliac disease, such as abdominal pain, gas, bloating, diarrhoea and weight loss can be present in patients with T1DM, but are often attributed to poor control of diabetes, gastroparesis or diabetic neuropathy
  92. ^ Chen HS, Su LT, Lin SZ, Sung FC, Ko MC, Li CY (January 2012). "Increased risk of urinary tract calculi among patients with diabetes mellitus – a population-based cohort study". Urology. 79 (1): 86–92. doi:10.1016/j.urology.2011.07.1431. PMID 22119251.
  93. ^ James R, Hijaz A (October 2014). "Lower urinary tract symptoms in women with diabetes mellitus: a current review". Current Urology Reports. 15 (10): 440. doi:10.1007/s11934-014-0440-3. PMID 25118849. S2CID 30653959.
  94. ^ a b c d "Sexual Dysfunction in Women". Diabetes.co.uk. Diabetes Digital Media Ltd. from the original on 9 November 2014. Retrieved 28 November 2014.
  95. ^ Goswami SK, Vishwanath M, Gangadarappa SK, Razdan R, Inamdar MN (August 2014). "Efficacy of ellagic acid and sildenafil in diabetes-induced sexual dysfunction". Pharmacognosy Magazine. 10 (Suppl 3): S581–S587. doi:10.4103/0973-1296.139790. PMC 4189276. PMID 25298678. ProQuest 1610759650.
  96. ^ Escobar-Morreale HF, Roldán B, Barrio R, Alonso M, Sancho J, de la Calle H, García-Robles R (November 2000). "High prevalence of the polycystic ovary syndrome and hirsutism in women with type 1 diabetes mellitus". The Journal of Clinical Endocrinology and Metabolism. 85 (11): 4182–4187. doi:10.1210/jcem.85.11.6931. PMID 11095451.
  97. ^ Codner E, Escobar-Morreale HF (April 2007). "Clinical review: Hyperandrogenism and polycystic ovary syndrome in women with type 1 diabetes mellitus". The Journal of Clinical Endocrinology and Metabolism. 92 (4): 1209–1216. doi:10.1210/jc.2006-2641. PMID 17284617.
  98. ^ a b Atkinson et al. 2020, "Other Complications".
  99. ^ Redondo, Steck & Pugliese 2018, "Evidence for the contribution of genetics to type I diabetes".
  100. ^ "What is Type 1 Diabetes?". Centers for Disease Control and Prevention. 11 March 2022. Retrieved 15 February 2023.
  101. ^ "FDA Approves Tzield". Drugs.com. November 2022. Retrieved 15 February 2023.
  102. ^ a b Atkinson et al. 2020, "Immunosuppresion".
  103. ^ von Scholten et al. 2021, "Antigen vaccination".
  104. ^ a b Dayan et al. 2019, "Previous prevention trials".
  105. ^ "Diabetes". World Health Organization. from the original on 26 January 2011. Retrieved 24 January 2011.
  106. ^ a b Atkinson et al. 2020, "Diagnosis".
  107. ^ Aanstoot HJ, Anderson BJ, Daneman D, Danne T, Donaghue K, Kaufman F, et al. (October 2007). "The global burden of youth diabetes: perspectives and potential". Pediatric Diabetes. 8. 8 (s8): 1–44. doi:10.1111/j.1399-5448.2007.00326.x. PMID 17767619. S2CID 222102948.
  108. ^ a b (PDF). American Diabetes Association. Archived from the original (PDF) on 29 April 2015.
  109. ^ Australian Institute of Health and Welfare (2015). "Incidence of type 1 diabetes in Australia 2000–2013". from the original on 7 October 2016. Retrieved 19 October 2016.
  110. ^ Shaw J (2012). "diabetes: the silent pandemic and its impact on Australia" (PDF). (PDF) from the original on 7 October 2016. Retrieved 19 October 2016.
  111. ^ Weiss A, Donnachie E, Beyerlein A, Ziegler AG, Bonifacio E (22 May 2023). "Type 1 Diabetes Incidence and Risk in Children With a Diagnosis of COVID-19" (PDF). JAMA (Research Letter). 329 (23): 2089–2091. doi:10.1001/jama.2023.8674. PMC 10203966. PMID 37213115. S2CID 258831851.
  112. ^ a b Atkinson et al. 2020, "Introduction".
  113. ^ Bottazzo GF, Florin-Christensen A, Doniach D (November 1974). "Islet-cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies". Lancet. 2 (7892): 1279–1283. doi:10.1016/s0140-6736(74)90140-8. PMID 4139522.
  114. ^ 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): 243–256. doi:10.1038/nri3422. PMC 4172461. PMID 23524461.
  115. ^ Johnson L (18 November 2008). "Study: Cost of diabetes $218B". USA Today. Associated Press. from the original on 1 July 2012.
  116. ^ "About DRWF". Diabetes Research & Wellness Foundation. from the original on 11 May 2013.
  117. ^ a b Minami K, Seino S (March 2013). "Current status of regeneration of pancreatic β-cells". Journal of Diabetes Investigation. 4 (2): 131–141. doi:10.1111/jdi.12062. PMC 4019265. PMID 24843642.
  118. ^ Pagliuca FW, Millman JR, Gürtler M, Segel M, Van Dervort A, Ryu JH, et al. (October 2014). "Generation of functional human pancreatic β cells in vitro". Cell. 159 (2): 428–439. doi:10.1016/j.cell.2014.09.040. PMC 4617632. PMID 25303535.
  119. ^ Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, et al. (November 2014). "Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells". Nature Biotechnology. 32 (11): 1121–1133. doi:10.1038/nbt.3033. PMID 25211370. S2CID 205280579.
  120. ^ Boughton & Hovorka 2020, "Introduction".
  121. ^ Boughton & Hovorka 2020, "Regulatory Approval of Closed-Loop Systems".
  122. ^ Ramli R, Reddy M, Oliver N (July 2019). "Artificial Pancreas: Current Progress and Future Outlook in the Treatment of Type 1 Diabetes". Drugs. 79 (10): 1089–1101. doi:10.1007/s40265-019-01149-2. hdl:10044/1/71348. PMID 31190305. S2CID 186207231.
  123. ^ a b King AJ (2020). Animal models of diabetes: methods and protocols. New York, NY: Humana Press. ISBN 978-1-0716-0385-7. OCLC 1149391907.[page needed]
  124. ^ a b c d Atkinson et al. 2020, "Animal models".
  125. ^ "NOD/ShiLtJ". The Jackson Laborataory. Retrieved 18 January 2022.
  126. ^ a b c d e f Pandey S, Dvorakova MC (7 January 2020). "Future Perspective of Diabetic Animal Models". Endocrine, Metabolic & Immune Disorders Drug Targets. 20 (1): 25–38. doi:10.2174/1871530319666190626143832. PMC 7360914. PMID 31241444.
  127. ^ Chen D, Thayer TC, Wen L, Wong FS (2020). "Mouse Models of Autoimmune Diabetes: The Nonobese Diabetic (NOD) Mouse". In King AJ (ed.). Animal Models of Diabetes. Methods in Molecular Biology. Vol. 2128. New York, NY: Springer US. pp. 87–92. doi:10.1007/978-1-0716-0385-7_6. ISBN 978-1-0716-0384-0. PMC 8253669. PMID 32180187.
  128. ^ Lenzen S, Arndt T, Elsner M, Wedekind D, Jörns A (2020). "Rat Models of Human Type 1 Diabetes". In King AJ (ed.). Animal Models of Diabetes. Methods in Molecular Biology. Vol. 2128. New York, NY: Springer US. pp. 69–85. doi:10.1007/978-1-0716-0385-7_5. ISBN 978-1-0716-0384-0. PMID 32180186. S2CID 212741496.
  129. ^ Al-Awar A, Kupai K, Veszelka M, Szűcs G, Attieh Z, Murlasits Z, et al. (2016). "Experimental Diabetes Mellitus in Different Animal Models". Journal of Diabetes Research. 2016: 9051426. doi:10.1155/2016/9051426. PMC 4993915. PMID 27595114.
  130. ^ Lenzen S, Tiedge M, Elsner M, Lortz S, Weiss H, Jörns A, et al. (September 2001). "The LEW.1AR1/Ztm-iddm rat: a new model of spontaneous insulin-dependent diabetes mellitus". Diabetologia. 44 (9): 1189–1196. doi:10.1007/s001250100625. PMID 11596676.
  131. ^ Lenzen S (October 2017). "Animal models of human type 1 diabetes for evaluating combination therapies and successful translation to the patient with type 1 diabetes". Diabetes/Metabolism Research and Reviews. 33 (7): e2915. doi:10.1002/dmrr.2915. PMID 28692149. S2CID 34331597.
  132. ^ Radenković M, Stojanović M, Prostran M (March 2016). "Experimental diabetes induced by alloxan and streptozotocin: The current state of the art". Journal of Pharmacological and Toxicological Methods. 78: 13–31. doi:10.1016/j.vascn.2015.11.004. PMID 26596652.
  133. ^ Salpea P, Cosentino C, Igoillo-Esteve M (2020). "A Review of Mouse Models of Monogenic Diabetes and ER Stress Signaling". In King AJ (ed.). Animal Models of Diabetes. Methods in Molecular Biology. Vol. 2128. New York, NY: Springer US. pp. 55–67. doi:10.1007/978-1-0716-0385-7_4. ISBN 978-1-0716-0384-0. PMID 32180185. S2CID 212740474.
  134. ^ Chang JH, Gurley SB (2012). "Assessment of Diabetic Nephropathy in the Akita Mouse". In Hans-Georg J, Hadi AH, Schürmann A (eds.). Animal Models in Diabetes Research. Methods in Molecular Biology. Vol. 933. Totowa, NJ: Humana Press. pp. 17–29. doi:10.1007/978-1-62703-068-7_2. ISBN 978-1-62703-067-0. PMID 22893398.
  135. ^ King AJ, Estil Les E, Montanya E (2020). "Use of Streptozotocin in Rodent Models of Islet Transplantation". In King AJ (ed.). Animal Models of Diabetes. Methods in Molecular Biology. Vol. 2128. New York, NY: Springer US. pp. 135–147. doi:10.1007/978-1-0716-0385-7_10. ISBN 978-1-0716-0384-0. PMID 32180191. S2CID 212739708.
  136. ^ Christoffersson G, Flodström-Tullberg M (2020). "Mouse Models of Virus-Induced Type 1 Diabetes". In King AJ (ed.). Animal Models of Diabetes. Methods in Molecular Biology. Vol. 2128. New York, NY: Springer US. pp. 93–105. doi:10.1007/978-1-0716-0385-7_7. ISBN 978-1-0716-0384-0. PMID 32180188. S2CID 212739248.
  137. ^ King AJ (June 2012). "The use of animal models in diabetes research". British Journal of Pharmacology. 166 (3): 877–894. doi:10.1111/j.1476-5381.2012.01911.x. PMC 3417415. PMID 22352879.

Works cited edit

  • American Diabetes Association (January 2021). "6. Glycemic Targets: Standards of Medical Care in Diabetes-2021". Diabetes Care. 44 (Suppl 1): S73–S84. doi:10.2337/dc21-S006. PMID 33298417. S2CID 228087604.
  • Atkinson MA, Mcgill DE, Dassau E, Laffel L (2020). "Type 1 diabetes mellitus". Williams Textbook of Endocrinology. Elsevier. pp. 1403–1437.
  • Boughton CK, Hovorka R (August 2020). "The artificial pancreas". Curr Opin Organ Transplant. 25 (4): 336–342. doi:10.1097/MOT.0000000000000786. PMID 32618719. S2CID 220326946.
  • Brownlee M, Aiello LP, Sun JK, Cooper ME, Feldman EL, Plutzky J, Boulton AJ (2020). "Complications of Diabetes Mellitus". Williams Textbook of Endocrinology. Elsevier. pp. 1438–1524. ISBN 978-0-323-55596-8.
  • Butler AE, Misselbrook D (August 2020). "Distinguishing between type 1 and type 2 diabetes". The BMJ. 370: m2998. doi:10.1136/bmj.m2998. PMID 32784223. S2CID 221097632.
  • Cashen K, Petersen T (August 2019). "Diabetic Ketoacidosis". Pediatr Rev. 40 (8): 412–420. doi:10.1542/pir.2018-0231. PMID 31371634. S2CID 199381655.
  • Dayan CM, Korah M, Tatovic D, Bundy BN, Herold KC (October 2019). "Changing the landscape for type 1 diabetes: the first step to prevention". Lancet. 394 (10205): 1286–1296. doi:10.1016/S0140-6736(19)32127-0. PMID 31533907. S2CID 202575545.
  • Dean PG, Kukla A, Stegall MD, Kudva YC (April 2017). "Pancreas transplantation". The BMJ. 357: j1321. doi:10.1136/bmj.j1321. PMID 28373161. S2CID 11374615.
  • Delli AJ, Lernmark A (2016). "Type 1 (insulin-dependent) diabetes mellitus: etiology, pathogenesis, prediction, and prevention". In Jameson JL (ed.). Endocrinology: Adult and Pediatric (7 ed.). Saunders. pp. 672–690. ISBN 978-0-323-18907-1.
  • DiMeglio LA, Evans-Molina C, Oram RA (June 2018). "Type 1 diabetes". Lancet. 391 (10138): 2449–2462. doi:10.1016/S0140-6736(18)31320-5. PMC 6661119. PMID 29916386.
  • Katsarou A, Gudbjörnsdottir S, Rawshani A, Dabelea D, Bonifacio E, Anderson BJ, et al. (March 2017). "Type 1 diabetes mellitus". Nature Reviews. Disease Primers. 3: 17016. doi:10.1038/nrdp.2017.16. PMID 28358037. S2CID 23127616.
  • Norris JM, Johnson RK, Stene LC (March 2020). "Type 1 diabetes-early life origins and changing epidemiology". The Lancet. Diabetes & Endocrinology. 8 (3): 226–238. doi:10.1016/S2213-8587(19)30412-7. PMC 7332108. PMID 31999944.
  • Redondo MJ, Steck AK, Pugliese A (May 2018). "Genetics of type 1 diabetes". Pediatric Diabetes. 19 (3): 346–353. doi:10.1111/pedi.12597. PMC 5918237. PMID 29094512.
  • Repaske DR (September 2016). "Medication-induced diabetes mellitus". Pediatr Diabetes. 17 (6): 392–7. doi:10.1111/pedi.12406. PMID 27492964. S2CID 4684512.
  • Rickels MR, Robertson RP (April 2019). "Pancreatic Islet Transplantation in Humans: Recent Progress and Future Directions". Endocr Rev. 40 (2): 631–668. doi:10.1210/er.2018-00154. PMC 6424003. PMID 30541144.
  • Shapiro AM, Pokrywczynska M, Ricordi C (May 2017). "Clinical pancreatic islet transplantation". Nat Rev Endocrinol. 13 (5): 268–277. doi:10.1038/nrendo.2016.178. PMID 27834384. S2CID 28784928.
  • Smith A, Harris C (August 2018). "Type 1 diabetes: Management strategies". American Family Physician. 98 (3): 154–156. PMID 30215903. Retrieved 12 January 2022.
  • von Scholten BJ, Kreiner FF, Gough SC, von Herrath M (May 2021). "Current and future therapies for type 1 diabetes". Diabetologia. 64 (5): 1037–1048. doi:10.1007/s00125-021-05398-3. PMC 8012324. PMID 33595677.
  • Wolsdorf, Joseph I.; Garvey, Katharine C. (2016). "Management of Diabetes in Children". Endocrinology: Adult and Pediatric. pp. 854–882.e6. doi:10.1016/B978-0-323-18907-1.00049-4. ISBN 978-0-323-18907-1.

External links edit

  • National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) 25 April 2011 at the Wayback Machine – Diabetes in America Textbook (PDFs)
  • IDF Diabetes Atlas
  • Type 1 Diabetes 30 October 2009 at the Wayback Machine at the American Diabetes Association
  • ADA's Standards of Medical Care in Diabetes 2019

type, diabetes, formerly, known, juvenile, diabetes, autoimmune, disease, that, originates, when, cells, that, make, insulin, beta, cells, destroyed, immune, system, insulin, hormone, required, cells, blood, sugar, energy, helps, regulate, glucose, levels, blo. Type 1 diabetes T1D formerly known as juvenile diabetes is an autoimmune disease that originates when cells that make insulin beta cells are destroyed by the immune system 4 Insulin is a hormone required for the cells to use blood sugar for energy and it helps regulate glucose levels in the bloodstream 2 Before treatment this results in high blood sugar levels in the body 1 The common symptoms of this elevated blood sugar are frequent urination increased thirst increased hunger weight loss and other serious complications 4 10 Additional symptoms may include blurry vision tiredness and slow wound healing 2 Symptoms typically develop over a short period of time often a matter of weeks if not months 1 Type 1 DiabetesOther namesDiabetes mellitus type 1 insulin dependent diabetes 1 juvenile diabetes 2 A blue circle the symbol for diabetes 3 Pronunciation d aɪ e ˈ b iː t iː z SpecialtyEndocrinologySymptomsFrequent urination increased thirst increased hunger weight loss 4 blurred vision fatigue and weakness Bed wetting in children who previously didn t wet the bed during the night 5 ComplicationsDiabetic ketoacidosis nonketotic hyperosmolar coma poor healing cardiovascular disease damage to the eyes 2 4 6 Usual onsetRelatively short period of time 1 DurationLong term 4 CausesBody does not produce enough insulin 4 Risk factorsFamily history celiac disease 6 7 Diagnostic methodBlood sugar A1C 6 8 PreventionUnknown 4 TreatmentInsulin diabetic diet exercise 1 2 Frequency 7 5 of diabetes cases 9 The cause of type 1 diabetes is unknown 4 but it is believed to involve a combination of genetic and environmental factors 1 The underlying mechanism involves an autoimmune destruction of the insulin producing beta cells in the pancreas 2 Diabetes is diagnosed by testing the level of sugar or glycated hemoglobin HbA1C in the blood 6 8 Type 1 diabetes can typically be distinguished from type 2 by testing for the presence of autoantibodies 6 and or declining levels absence of C peptide There is no known way to prevent type 1 diabetes 4 Treatment with insulin is required for survival 1 Insulin therapy is usually given by injection just under the skin but can also be delivered by an insulin pump 11 A diabetic diet and exercise are important parts of management 2 If left untreated diabetes can cause many complications 4 Complications of relatively rapid onset include diabetic ketoacidosis and nonketotic hyperosmolar coma 6 Long term complications include heart disease stroke kidney failure foot ulcers and damage to the eyes 4 Furthermore since insulin lowers blood sugar levels complications may arise from low blood sugar if more insulin is taken than necessary 6 Type 1 diabetes makes up an estimated 5 10 of all diabetes cases 9 The number of people affected globally is unknown although it is estimated that about 80 000 children develop the disease each year 6 Within the United States the number of people affected is estimated at one to three million 6 12 Rates of disease vary widely with approximately one new case per 100 000 per year in East Asia and Latin America and around 30 new cases per 100 000 per year in Scandinavia and Kuwait 13 14 It typically begins in children and young adults 1 Contents 1 Signs and symptoms 2 Cause 2 1 Environmental 2 2 Genetics 2 3 Chemicals and drugs 3 Diagnosis 4 Management 4 1 Lifestyle 4 2 Transplant 5 Pathogenesis 5 1 Alpha cell dysfunction 5 1 1 Hyperglucagonemia 5 1 2 Hypoglycemic glucagon impairment 6 Complications 6 1 Long term complications 6 2 Urinary tract infection 6 3 Sexual dysfunction 6 4 Autoimmune disorders 7 Prevention 8 Epidemiology 9 History 10 Society and culture 11 Research 11 1 Organ replacement 11 2 Disease models 11 2 1 Chemically induced 11 2 2 Genetically induced 11 2 3 Virally induced 12 References 13 Works cited 14 External linksSigns and symptoms edit nbsp Overview of the most significant symptoms of diabetesType 1 diabetes begins suddenly typically in childhood or adolescence 15 The major sign of type 1 diabetes is very high blood sugar which typically manifests in children as a few days to weeks of polyuria increased urination polydipsia increased thirst and weight loss 16 17 Children may also experience increased appetite blurred vision bedwetting recurrent skin infections candidiasis of the perineum irritability and performance issues at school 16 17 Adults with type 1 diabetes tend to have more varied symptoms that come on over months rather than days to weeks 18 17 Prolonged lack of insulin can also result in diabetic ketoacidosis characterized by persistent fatigue dry or flushed skin abdominal pain nausea or vomiting confusion trouble breathing and a fruity breath odor 18 19 Blood and urine tests reveal unusually high glucose and ketones in the blood and urine 20 Untreated ketoacidosis can rapidly progress to loss of consciousness coma and death 20 The percentage of children whose type 1 diabetes begins with an episode of diabetic ketoacidosis varies widely by geography as low as 15 in parts of Europe and North America and as high as 80 in the developing world 20 Cause editType 1 diabetes is caused by the destruction of b cells the only cells in the body that produce insulin and the consequent progressive insulin deficiency Without insulin the body is unable to respond effectively to increases in blood sugar Due to this people with diabetes have persistent hyperglycemia 21 In 70 90 of cases b cells are destroyed by one s own immune system for reasons that are not entirely clear 21 The best studied components of this autoimmune response are b cell targeted antibodies that begin to develop in the months or years before symptoms arise 21 Typically someone will first develop antibodies against insulin or the protein GAD65 followed eventually by antibodies against the proteins IA 2 IA 2b and or ZNT8 People with more of these antibodies and who develop them earlier in life are at higher risk for developing symptomatic type 1 diabetes 22 The trigger for the development of these antibodies remains unclear 23 A number of explanatory theories have been put forward and the cause may involve genetic susceptibility a diabetogenic trigger and or exposure to an antigen 24 The remaining 10 30 of type 1 diabetics have b cell destruction but no sign of autoimmunity this is called idiopathic type 1 diabetes and its cause is unknown 21 Environmental edit Various environmental risks have been studied in an attempt to understand what triggers b cell autoimmunity Many aspects of environment and life history are associated with slight increases in type 1 diabetes risk however the connection between each risk and diabetes often remains unclear citation needed Type 1 diabetes risk is slightly higher for children whose mothers are obese or older than 35 or for children born by caesarean section 25 Similarly a child s weight gain in the first year of life total weight and BMI are associated with slightly increased type 1 diabetes risk 25 Some dietary habits have also been associated with type 1 diabetes risk namely consumption of cow s milk and dietary sugar intake 25 Animal studies and some large human studies have found small associations between type 1 diabetes risk and intake of gluten or dietary fiber however other large human studies have found no such association 25 Many potential environmental triggers have been investigated in large human studies and found to be unassociated with type 1 diabetes risk including duration of breastfeeding time of introduction of cow milk into the diet vitamin D consumption blood levels of active vitamin D and maternal intake of omega 3 fatty acids 25 26 A longstanding hypothesis for an environmental trigger is that some viral infection early in life contributes to type 1 diabetes development Much of this work has focused on enteroviruses with some studies finding slight associations with type 1 diabetes and others finding none 27 Large human studies have searched for but not yet found an association between type 1 diabetes and various other viral infections including infections of the mother during pregnancy 27 Conversely some have postulated that reduced exposure to pathogens in the developed world increases the risk of autoimmune diseases often called the hygiene hypothesis Various studies of hygiene related factors including household crowding daycare attendance population density childhood vaccinations antihelminth medication and antibiotic usage during early life or pregnancy show no association with type 1 diabetes 28 Genetics edit Type 1 diabetes is partially caused by genetics and family members of type 1 diabetics have a higher risk of developing the disease themselves In the general population the risk of developing type 1 diabetes is around 1 in 250 For someone whose parent has type 1 diabetes the risk rises to 1 9 If a sibling has type 1 diabetes the risk is 6 7 If someone s identical twin has type 1 diabetes they have a 30 70 risk of developing it themselves 29 About half of the disease s heritability is due to variations in three HLA class II genes involved in antigen presentation HLA DRB1 HLA DQA1 and HLA DQB1 29 The variation patterns associated with increased risk of type 1 diabetes are called HLA DR3 and HLA DR4 HLA DQ8 and are common in people of European descent A pattern associated with reduced risk of type 1 diabetes is called HLA DR15 HLA DQ6 29 Large genome wide association studies have identified dozens of other genes associated with type 1 diabetes risk mostly genes involved in the immune system 29 Chemicals and drugs edit Some medicines can reduce insulin production or damage b cells resulting in disease that resembles type 1 diabetes The antiviral drug didanosine triggers pancreas inflammation in 5 to 10 of those who take it sometimes causing lasting b cell damage 30 Similarly up to 5 of those who take the anti protozoal drug pentamidine experience b cell destruction and diabetes 30 Several other drugs cause diabetes by reversibly reducing insulin secretion namely statins which may also damage b cells the post transplant immunosuppressants cyclosporin A and tacrolimus the leukemia drug L asparaginase and the antibiotic gatifloxicin 30 31 Pyrinuron Vacor a rodenticide introduced in the United States in 1976 selectively destroys pancreatic beta cells resulting in type 1 diabetes after accidental poisoning 32 Pyrinuron was withdrawn from the U S market in 1979 33 Cancer Immunotherapy Drugs have also destroyed pancreatic beta cells such as Opdivo among others 34 Diagnosis editDiabetes is typically diagnosed by a blood test showing unusually high blood sugar The World Health Organization defines diabetes as blood sugar levels at or above 7 0 mmol L 126 mg dL after fasting for at least eight hours or a glucose level at or above 11 1 mmol L 200 mg dL two hours after an oral glucose tolerance test 35 The American Diabetes Association additionally recommends a diagnosis of diabetes for anyone with symptoms of hyperglycemia and blood sugar at any time at or above 11 1 mmol L or glycated hemoglobin hemoglobin A1C levels at or above 48 mmol mol 36 Once a diagnosis of diabetes is established type 1 diabetes is distinguished from other types by a blood test for the presence of autoantibodies that target various components of the beta cell 37 The most commonly available tests detect antibodies against glutamic acid decarboxylase the beta cell cytoplasm or insulin each of which are targeted by antibodies in around 80 of type 1 diabetics 37 Some healthcare providers also have access to tests for antibodies targeting the beta cell proteins IA 2 and ZnT8 these antibodies are present in around 58 and 80 of type 1 diabetics respectively 37 Some also test for C peptide a byproduct of insulin synthesis Very low C peptide levels are suggestive of type 1 diabetes 37 Management editFurther information Diabetes management The mainstay of type 1 diabetes treatment is the regular injection of insulin to manage hyperglycemia 38 Injections of insulin via subcutaneous injection using either a syringe or an insulin pump are necessary multiple times per day adjusting dosages to account for food intake blood glucose levels and physical activity 38 The goal of treatment is to maintain blood sugar in a normal range 80 130 mg dL before a meal lt 180 mg dL after as often as possible 39 To achieve this people with diabetes often monitor their blood glucose levels at home Around 83 of type 1 diabetics monitor their blood glucose by capillary blood testing pricking the finger to draw a drop of blood and determining blood glucose with a glucose meter 40 The American Diabetes Association recommends testing blood glucose around 6 10 times per day before each meal before exercise at bedtime occasionally after a meal and any time someone feels the symptoms of hypoglycemia 40 Around 17 of people with type 1 diabetes use a continuous glucose monitor a device with a sensor under the skin that constantly measures glucose levels and communicates those levels to an external device 40 Continuous glucose monitoring is associated with better blood sugar control than capillary blood testing alone however continuous glucose monitoring tends to be substantially more expensive 40 Healthcare providers can also monitor someone s hemoglobin A1C levels which reflect the average blood sugar over the last three months 41 The American Diabetes Association recommends a goal of keeping hemoglobin A1C levels under 7 for most adults and 7 5 for children 41 42 The goal of insulin therapy is to mimic normal pancreatic insulin secretion low levels of insulin constantly present to support basic metabolism plus the two phase secretion of additional insulin in response to high blood sugar then an extended phase of continued insulin secretion 43 This is accomplished by combining different insulin preparations that act with differing speeds and durations The standard of care for type 1 diabetes is a bolus of rapid acting insulin 10 15 minutes before each meal or snacks and as needed to correct hyperglycemia 43 In addition constant low levels of insulin are achieved with one or two daily doses of long acting insulin or by steady infusion of low insulin levels by an insulin pump 43 The exact dose of insulin appropriate for each injection depends on the content of the meal snack and the individual person s sensitivity to insulin and is therefore typically calculated by the individual with diabetes or a family member by hand or assistive device calculator chart mobile app etc 43 People unable to manage these intensive insulin regimens are sometimes prescribed alternate plans relying on mixtures of rapid or short acting and intermediate acting insulin which are administered at fixed times along with meals of pre planned times and carbohydrate composition 43 The National Institute of Health and Care Excellence now recommends closed loop insulin systems as an option for all women with type 1 diabetes who are pregnant or planning pregnancy 44 45 46 A non insulin medication approved by the U S Food and Drug Administration for treating type 1 diabetes is the amylin analog pramlintide which replaces the beta cell hormone amylin Addition of pramlintide to mealtime insulin injections reduces the boost in blood sugar after a meal improving blood sugar control 47 Occasionally metformin GLP 1 receptor agonists Dipeptidyl peptidase 4 inhibitors or SGLT2 inhibitor are prescribed off label to people with type 1 diabetes although fewer than 5 of type 1 diabetics use these drugs 38 Lifestyle edit Besides insulin the major way type 1 diabetics control their blood sugar is by learning how various foods impact their blood sugar levels This is primarily done by tracking their intake of carbohydrates the type of food with the greatest impact on blood sugar 48 In general people with type 1 diabetes are advised to follow an individualized eating plan rather than a pre decided one 49 There are camps for children to teach them how and when to use or monitor their insulin without parental help 50 As psychological stress may have a negative effect on diabetes a number of measures have been recommended including exercising taking up a new hobby or joining a charity among others 51 Regular exercise is important for maintaining general health though the effect of exercise on blood sugar can be challenging to predict 52 Exogenous insulin can drive down blood sugar leaving those with diabetes at risk of hypoglycemia during and immediately after exercise then again seven to eleven hours after exercise called the lag effect 52 Conversely high intensity exercise can result in a shortage of insulin and consequent hyperglycemia 52 The risk of hypoglycemia can be managed by beginning exercise when blood sugar is relatively high above 100 mg dL ingesting carbohydrates during or shortly after exercise and reducing the amount of injected insulin within two hours of the planned exercise 52 Similarly the risk of exercise induced hyperglycemia can be managed by avoiding exercise when insulin levels are very low when blood sugar is extremely high above 350 mg dL or when one feels unwell 52 Transplant edit In some cases people can receive transplants of the pancreas or isolated islet cells to restore insulin production and alleviate diabetic symptoms Transplantation of the whole pancreas is rare due in part to the few available donor organs and to the need for lifelong immunosuppressive therapy to prevent transplant rejection 53 54 The American Diabetes Association recommends pancreas transplant only in people who also require a kidney transplant or who struggle to perform regular insulin therapy and experience repeated severe side effects of poor blood sugar control 54 Most pancreas transplants are done simultaneously with a kidney transplant with both organs from the same donor 55 The transplanted pancreas continues to function for at least five years in around three quarters of recipients allowing them to stop taking insulin 56 Transplantations of islets alone have become increasingly common 57 Pancreatic islets are isolated from a donor pancreas then injected into the recipient s portal vein from which they implant onto the recipient s liver 58 In nearly half of recipients the islet transplant continues to work well enough that they still do not need exogenous insulin five years after transplantation 59 If a transplant fails recipients can receive subsequent injections of islets from additional donors into the portal vein 58 Like with whole pancreas transplantation islet transplantation requires lifelong immunosuppression and depends on the limited supply of donor organs it is therefore similarly limited to people with severe poorly controlled diabetes and those who have had or are scheduled for a kidney transplant 57 60 Donislecel Lantidra allogeneic donor pancreatic islet cellular therapy was approved for medical use in the United States in June 2023 61 Pathogenesis editType 1 diabetes is a result of the destruction of pancreatic beta cells although what triggers that destruction remains unclear 62 People with type 1 diabetes tend to have more CD8 T cells and B cells that specifically target islet antigens than those without type 1 diabetes suggesting a role for the adaptive immune system in beta cell destruction 62 63 Type 1 diabetics also tend to have reduced regulatory T cell function which may exacerbate autoimmunity 62 Destruction of beta cells results in inflammation of the islet of Langerhans called insulitis These inflamed islets tend to contain CD8 T cells and to a lesser extent CD4 T cells 62 Abnormalities in the pancreas or the beta cells themselves may also contribute to beta cell destruction The pancreases of people with type 1 diabetes tend to be smaller lighter and have abnormal blood vessels nerve innervations and extracellular matrix organization 64 In addition beta cells from people with type 1 diabetes sometimes overexpress HLA class I molecules responsible for signaling to the immune system and have increased endoplasmic reticulum stress and issues with synthesizing and folding new proteins any of which could contribute to their demise 64 The mechanism by which the beta cells actually die likely involves both necroptosis and apoptosis induced or exacerbated by CD8 T cells and macrophages 65 Necroptosis can be triggered by activated T cells which secrete toxic granzymes and perforin or indirectly as a result of reduced blood flow or the generation of reactive oxygen species 65 As some beta cells die they may release cellular components that amplify the immune response exacerbating inflammation and cell death 65 Pancreases from people with type 1 diabetes also have signs of beta cell apoptosis linked to activation of the janus kinase and TYK2 pathways 65 Partial ablation of beta cell function is enough to cause diabetes at diagnosis people with type 1 diabetes often still have detectable beta cell function Once insulin therapy is started many people experience a resurgence in beta cell function and can go some time with little to no insulin treatment called the honeymoon phase 64 This eventually fades as beta cells continue to be destroyed and insulin treatment is required again 64 Beta cell destruction is not always complete as 30 80 of type 1 diabetics produce small amounts of insulin years or decades after diagnosis 64 Alpha cell dysfunction edit Onset of autoimmune diabetes is accompanied by impaired ability to regulate the hormone glucagon 66 which acts in antagonism with insulin to regulate blood sugar and metabolism Progressive beta cell destruction leads to dysfunction in the neighboring alpha cells which secrete glucagon exacerbating excursions away from euglycemia in both directions overproduction of glucagon after meals causes sharper hyperglycemia and failure to stimulate glucagon upon hypoglycemia prevents a glucagon mediated rescue of glucose levels 67 Hyperglucagonemia edit Onset of type 1 diabetes is followed by an increase in glucagon secretion after meals Increases have been measured up to 37 during the first year of diagnosis while c peptide levels indicative of islet derived insulin decline by up to 45 68 Insulin production will continue to fall as the immune system destroys beta cells and islet derived insulin will continue to be replaced by therapeutic exogenous insulin Simultaneously there is measurable alpha cell hypertrophy and hyperplasia in the early stage of the disease leading to expanded alpha cell mass This together with failing beta cell insulin secretion begins to account for rising glucagon levels that contribute to hyperglycemia 67 Some researchers believe glucagon dysregulation to be the primary cause of early stage hyperglycemia 69 Leading hypotheses for the cause of postprandial hyperglucagonemia suggest that exogenous insulin therapy is inadequate to replace the lost intraislet signalling to alpha cells previously mediated by beta cell derived pulsatile insulin secretion 70 71 Under this working hypothesis intensive insulin therapy has attempted to mimic natural insulin secretion profiles in exogenous insulin infusion therapies 72 In young people with type 1 diabetes unexplained deaths could be due to nighttime hypoglycemia triggering abnormal heart rhythms or cardiac autonomic neuropathy damage to nerves that control the function of the heart Hypoglycemic glucagon impairment edit Glucagon secretion is normally increased upon falling glucose levels but normal glucagon response to hypoglycemia is blunted in type 1 diabetics 73 74 Beta cell glucose sensing and subsequent suppression of administered insulin secretion is absent leading to islet hyperinsulinemia which inhibits glucagon release 73 75 Autonomic inputs to alpha cells are much more important for glucagon stimulation in the moderate to severe ranges of hypoglycemia yet the autonomic response is blunted in a number of ways Recurrent hypoglycemia leads to metabolic adjustments in the glucose sensing areas of the brain shifting the threshold for counter regulatory activation of the sympathetic nervous system to lower glucose concentration 75 This is known as hypoglycemic unawareness Subsequent hypoglycemia is met with impairment in sending of counter regulatory signals to the islets and adrenal cortex This accounts for the lack of glucagon stimulation and epinephrine release that would normally stimulate and enhance glucose release and production from the liver rescuing the diabetic from severe hypoglycemia coma and death Numerous hypotheses have been produced in the search for a cellular mechanism of hypoglycemic unawareness and a consensus has yet to be reached 76 The major hypotheses are summarized in the following table 77 75 76 Mechanisms of hypoglycemic unawarenessGlycogen supercompensation Increased glycogen stores in astrocytes might contribute supplementary glycosyl units for metabolism counteracting the central nervous system perception of hypoglycemia Enhanced glucose metabolism Altered glucose transport and enhanced metabolic efficiency upon recurring hypoglycemia relieves oxidative stress that would activate sympathetic response Alternative fuel hypothesis Decreased reliance on glucose supplementation of lactate from astrocytes or ketones meet metabolic demands and reduce stress to brain Brain neuronal communication Hypothalamic inhibitory GABA normally decreases during hypoglycemia disinhibiting signals for sympathetic tone Recurrent episodes of hypoglycemia result in increased basal GABA which fails to decrease normally during subsequent hypoglycemia Inhibitory tone remains and sympathetic tone is not increased In addition autoimmune diabetes is characterized by a loss of islet specific sympathetic innervation 78 This loss constitutes an 80 90 reduction of islet sympathetic nerve endings happens early in the progression of the disease and is persistent though the life of the patient 79 It is linked to the autoimmune aspect of type 1 diabetics and fails to occur in type 2 diabetics Early in the autoimmune event the axon pruning is activated in islet sympathetic nerves Increased BDNF and ROS that result from insulitis and beta cell death stimulate the p75 neurotrophin receptor p75NTR which acts to prune off axons Axons are normally protected from pruning by activation of tropomyosin receptor kinase A Trk A receptors by NGF which in islets is primarily produced by beta cells Progressive autoimmune beta cell destruction therefore causes both the activation of pruning factors and the loss of protective factors to the islet sympathetic nerves This unique form of neuropathy is a hallmark of type 1 diabetes and plays a part in the loss of glucagon rescue of severe hypoglycemia 78 Complications editFurther information Complications of diabetes The most pressing complication of type 1 diabetes are the always present risks of poor blood sugar control severe hypoglycemia and diabetic ketoacidosis Hypoglycemia typically blood sugar below 70 mg dL triggers the release of epinephrine and can cause people to feel shaky anxious or irritable 80 People with hypoglycemia may also experience hunger nausea sweats chills dizziness and a fast heartbeat 80 Some feel lightheaded sleepy or weak 80 Severe hypoglycemia can develop rapidly causing confusion coordination problems loss of consciousness and seizure 80 81 On average people with type 1 diabetes experience a hypoglycemia event that requires assistance of another 16 20 times in 100 person years and an event leading to unconsciousness or seizure 2 8 times per 100 person years 81 The American Diabetes Association recommends treating hypoglycemia by the 15 15 rule eat 15 grams of carbohydrates then wait 15 minutes before checking blood sugar repeat until blood sugar is at least 70 mg dL 80 Severe hypoglycemia that impairs someone s ability to eat is typically treated with injectable glucagon which triggers glucose release from the liver into the bloodstream 80 People with repeated bouts of hypoglycemia can develop hypoglycemia unawareness where the blood sugar threshold at which they experience symptoms of hypoglycemia decreases increasing their risk of severe hypoglycemic events 82 Rates of severe hypoglycemia have generally declined due to the advent of rapid acting and long acting insulin products in the 1990s and early 2000s 43 however acute hypoglycemic still causes 4 10 of type 1 diabetes related deaths 81 The other persistent risk is diabetic ketoacidosis a state where lack of insulin results in cells burning fat rather than sugar producing toxic ketones as a byproduct 19 Ketoacidosis symptoms can develop rapidly with frequent urination excessive thirst nausea vomiting and severe abdominal pain all common 83 More severe ketoacidosis can result in labored breathing and loss of consciousness due to cerebral edema 83 People with type 1 diabetes experience diabetic ketoacidosis 1 5 times per 100 person years the majority of which result in hospitalization 84 13 19 of type 1 diabetes related deaths are caused by ketoacidosis 81 making ketoacidosis the leading cause of death in people with type 1 diabetes less than 58 years old 84 Long term complications edit In addition to the acute complications of diabetes long term hyperglycemia results in damage to the small blood vessels throughout the body This damage tends to manifest particularly in the eyes nerves and kidneys causing diabetic retinopathy diabetic neuropathy and diabetic nephropathy respectively 82 In the eyes prolonged high blood sugar causes the blood vessels in the retina to become fragile 85 People with type 1 diabetes also have increased risk of cardiovascular disease which is estimated to shorten the life of the average type 1 diabetic by 8 13 years 86 Cardiovascular disease 87 as well as neuropathy 88 may have an autoimmune basis as well Women with type 1 DM have a 40 higher risk of death as compared to men with type 1 DM 89 About 12 percent of people with type 1 diabetes have clinical depression 90 About 6 percent of people with type 1 diabetes also have celiac disease but in most cases there are no digestive symptoms 7 91 or are mistakenly attributed to poor control of diabetes gastroparesis or diabetic neuropathy 91 In most cases celiac disease is diagnosed after onset of type 1 diabetes The association of celiac disease with type 1 diabetes increases the risk of complications such as retinopathy and mortality This association can be explained by shared genetic factors and inflammation or nutritional deficiencies caused by untreated celiac disease even if type 1 diabetes is diagnosed first 7 Urinary tract infection edit People with diabetes show an increased rate of urinary tract infection 92 The reason is bladder dysfunction is more common in people with diabetes than people without diabetes due to diabetes nephropathy When present nephropathy can cause a decrease in bladder sensation which in turn can cause increased residual urine a risk factor for urinary tract infections 93 Sexual dysfunction edit Sexual dysfunction in people with diabetes is often a result of physical factors such as nerve damage and poor circulation and psychological factors such as stress and or depression caused by the demands of the disease 94 The most common sexual issues in males with diabetes are problems with erections and ejaculation With diabetes blood vessels supplying the penis s erectile tissue can get hard and narrow preventing the adequate blood supply needed for a firm erection The nerve damage caused by poor blood glucose control can also cause ejaculate to go into the bladder instead of through the penis during ejaculation called retrograde ejaculation When this happens semen leaves the body in the urine Another cause of erectile dysfunction is reactive oxygen species created as a result of the disease Antioxidants can be used to help combat this 95 Sexual problems are common in women who have diabetes 94 including reduced sensation in the genitals dryness difficulty inability to orgasm pain during sex and decreased libido Diabetes sometimes decreases estrogen levels in females which can affect vaginal lubrication Less is known about the correlation between diabetes and sexual dysfunction in females than in males 94 Oral contraceptive pills can cause blood sugar imbalances in women who have diabetes Dosage changes can help address that at the risk of side effects and complications 94 Women with type 1 diabetes show a higher than normal rate of polycystic ovarian syndrome PCOS 96 The reason may be that the ovaries are exposed to high insulin concentrations since women with type 1 diabetes can have frequent hyperglycemia 97 Autoimmune disorders edit People with type 1 diabetes are at an increased risk for developing several autoimmune disorders particularly thyroid problems around 20 of people with type 1 diabetes have hypothyroidism or hyperthyroidism typically caused by Hashimoto thyroiditis or Graves disease respectiveley 98 81 Celiac disease affects 2 8 of people with type 1 diabetes and is more common in those who were younger at diabetes diagnosis and in white people 98 Type 1 diabetics are also at increased risk of rheumatoid arthritis lupus autoimmune gastritis pernicious anemia vitiligo and Addison s disease 81 Conversely complex autoimmune syndromes caused by mutations in the immunity related genes AIRE causing autoimmune polyglandular syndrome FoxP3 causing IPEX syndrome or STAT3 include type 1 diabetes in their effects 99 Prevention editThere is no way to prevent type 1 diabetes 100 however the development of diabetes symptoms can be delayed in some people who are at high risk of developing the disease In 2022 the FDA approved an intravenous injection of teplizumab to delay the progression of type 1 diabetes in those older than eight who have already developed diabetes related autoantibodies and problems with blood sugar control In that population the anti CD3 monoclonal antibody teplizumab can delay the development of type 1 diabetes symptoms by around two years 101 In addition to anti CD3 antibodies several other immunosuppressive agents have been trialled with the aim of preventing beta cell destruction Large trials of cyclosporine treatment suggested that cyclosporine could improve insulin secretion in those recently diagnosed with type 1 diabetes however people who stopped taking cyclosporine rapidly stopped making insulin and cyclosporine s kidney toxicity and increased risk of cancer prevented people from using it long term 102 Several other immunosuppressive agents prednisone azathioprine anti thymocyte globulin mycophenolate and antibodies against CD20 and IL2 receptor a have been the subject of research but none have provided lasting protection from development of type 1 diabetes 102 There have also been clinical trials attempting to induce immune tolerance by vaccination with insulin GAD65 and various short peptides targeted by immune cells during type 1 diabetes none have yet delayed or prevented development of disease 103 Several trials have attempted dietary interventions with the hope of reducing the autoimmunity that leads to type 1 diabetes Trials that withheld cow s milk or gave infants formula free of bovine insulin decreased the development of b cell targeted antibodies but did not prevent the development of type 1 diabetes 104 Similarly trials that gave high risk individuals injected insulin oral insulin or nicotinamide did not prevent diabetes development 104 Epidemiology editType 1 diabetes makes up an estimated 10 15 of all diabetes cases 22 or 11 22 million cases worldwide 105 Symptoms can begin at any age but onset is most common in children with diagnoses slightly more common in 5 to 7 year olds and much more common around the age of puberty 106 15 In contrast to most autoimmune diseases type 1 diabetes is slightly more common in males than in females 106 In 2006 type 1 diabetes affected 440 000 children under 14 years of age and was the primary cause of diabetes in those less than 15 years of age 107 22 Rates vary widely by country and region Incidence is highest in Scandinavia at 30 60 new cases per 100 000 children per year intermediate in the U S and Southern Europe at 10 20 cases per 100 000 per year and lowest in China much of Asia and South America at 1 3 cases per 100 000 per year 26 In the United States type 1 and 2 diabetes affected about 208 000 youths under the age of 20 in 2015 Over 18 000 youths are diagnosed with Type 1 diabetes every year Every year about 234 051 Americans die due to diabetes type I or II or diabetes related complications with 69 071 having it as the primary cause of death 108 In Australia about one million people have been diagnosed with diabetes and of this figure 130 000 people have been diagnosed with type 1 diabetes Australia ranks 6th highest in the world with children under 14 years of age Between 2000 and 2013 31 895 new cases were established with 2 323 in 2013 a rate of 10 13 cases per 100 00 people each year Aboriginals and Torres Strait Islander people are less affected 109 110 Since the 1950s the incidence of type 1 diabetes has been gradually increasing across the world by an average 3 4 per year 26 The increase is more pronounced in countries that began with a lower incidence of type 1 diabetes 26 A single 2023 study suggested a relationship between COVID 19 infection and the incidence of type 1 diabetes in children 111 confirmatory studies have not appeared to date History editMain article History of diabetes The connection between diabetes and pancreatic damage was first described by the German pathologist Martin Schmidt who in a 1902 paper noted inflammation around the pancreatic islet of a child who had died of diabetes 112 The connection between this inflammation and diabetes onset was further developed through the 1920s by Shields Warren and the term insulitis was coined by Hanns von Meyenburg in 1940 to describe the phenomenon 112 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 113 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 114 The name juvenile diabetes was used earlier as it often first is diagnosed in childhood Society and culture editSee also List of people with type 1 diabetes Type 1 and 2 diabetes 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 115 In the United States 245 billion every year is attributed to diabetes Individuals diagnosed with diabetes have 2 3 times the health care costs as individuals who do not have diabetes One in ten health care dollars are spent on individuals with type 1 and 2 diabetes 108 Research editFunding 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 Institutes of Health and in the UK via the National Institute for Health and Care Research or the Medical Research Council The Juvenile Diabetes Research Foundation JDRF founded by parents of children with type 1 diabetes is the world s largest provider of charity based funding for type 1 diabetes research citation needed Other charities include the American Diabetes Association Diabetes UK Diabetes Research and Wellness Foundation 116 Diabetes Australia and the Canadian Diabetes Association Organ replacement edit Pluripotent stem cells can be used to generate beta cells but previously these cells did not function as well as normal beta cells 117 In 2014 more mature beta cells were produced which released insulin in response to blood sugar when transplanted into mice 118 119 Before these techniques can be used in humans more evidence of safety and effectiveness is needed 117 There has also been substantial effort to develop a fully automated insulin delivery system or artificial pancreas that could sense glucose levels and inject appropriate insulin without conscious input from the user 120 Current hybrid closed loop systems use a continuous glucose monitor to sense blood sugar levels and a subcutaneous insulin pump to deliver insulin however due to the delay between insulin injection and its action current systems require the user to initiate insulin before taking meals 121 Several improvements to these systems are currently undergoing clinical trials in humans including a dual hormone system that injects glucagon in addition to insulin and an implantable device that injects insulin intraperitoneally where it can be absorbed more quickly 122 Disease models edit Various animal models of disease are used to understand the pathogenesis and etiology of type 1 diabetes Currently available models of T1D can be divided into spontaneously autoimmune chemically induced virus induced and genetically induced 123 The nonobese diabetic NOD mouse is the most widely studied model of type 1 diabetes 123 It is an inbred strain that spontaneously develops type 1 diabetes in 30 100 of female mice depending on housing conditions 124 Diabetes in NOD mice is caused by several genes primarily MHC genes involved in antigen presentation 124 Like diabetic humans NOD mice develop islet autoantibodies and inflammation in the islet followed by reduced insulin production and hyperglycemia 124 125 Some features of human diabetes are exaggerated in NOD mice namely the mice have more severe islet inflammation than humans and have a much more pronounced sex bias with females developing diabetes far more frequently than males 124 In NOD mice the onset of insulitis occurs at 3 4 weeks of age The islets of Langerhans are infiltrated by CD4 CD8 T lymphocytes NK cells B lymphocytes dendritic cells macrophages and neutrophils similar to the disease process in humans 126 In addition to sex breeding conditions gut microbiome composition or diet also influence the onset of T1D 127 The BioBreeding Diabetes Prone BB rat is another widely used spontaneous experimental model for T1D The onset of diabetes occurs in up to 90 of individuals regardless of sex at 8 16 weeks of age 126 During insulitis the pancreatic islets are infiltrated by T lymphocytes B lymphocytes macrophages and NK cells with the difference from the human course of insulitis being that CD4 T lymphocytes are markedly reduced and CD8 T lymphocytes are almost absent The aforementioned lymphopenia is the major drawback of this model The disease is characterized by hyperglycemia hypoinsulinemia weight loss ketonuria and the need for insulin therapy for survival 126 BB Rats are used to study the genetic aspects of T1D and are also used for interventional studies and diabetic nephropathy studies 128 LEW 1AR1 iddm rats are derived from congenital Lewis rats and represent a rarer spontaneous model for T1D These rats develop diabetes at about 8 9 weeks of age with no sex differences unlike NOD mice 129 In LEW mice diabetes presents with hyperglycemia glycosuria ketonuria and polyuria 130 126 The advantage of the model is the progression of the prediabetic phase which is very similar to human disease with infiltration of islet by immune cells about a week before hyperglycemia is observed This model is suitable for intervention studies or for the search for predictive biomarkers It is also possible to observe individual phases of pancreatic infiltration by immune cells The advantage of congenic LEW mice is also the good viability after the manifestation of T1D compared to NOD mice and BB rats 131 Chemically induced edit The chemical compounds aloxan and streptozotocin STZ are commonly used to induce diabetes and destroy b cells in mouse rat animal models 126 In both cases it is a cytotoxic analog of glucose that passes GLUT2 transport and accumulates in b cells causing their destruction The chemically induced destruction of b cells leads to decreased insulin production hyperglycemia and weight loss in the experimental animal 132 The animal models prepared in this way are suitable for research into blood sugar lowering drugs and therapies e g for testing new insulin preparations They are also the most commonly used genetically induced T1D model is the so called AKITA mouse originally C57BL 6NSIc mouse The development of diabetes in AKITA mice is caused by a spontaneous point mutation in the Ins2 gene which is responsible for the correct composition of insulin in the endoplasmic reticulum Decreased insulin production is then associated with hyperglycemia polydipsia and polyuria If severe diabetes develops within 3 4 weeks AKITA mice survive no longer than 12 weeks without treatment intervention The description of the etiology of the disease shows that unlike spontaneous models the early stages of the disease are not accompanied by insulitis 133 AKITA mice are used to test drugs targeting endoplasmic reticulum stress reduction to test islet transplants and to study diabetes related complications such as nephropathy sympathetic autonomic neuropathy and vascular disease 126 134 for testing transplantation therapies Their advantage is mainly the low cost the disadvantage is the cytotoxicity of the chemical compounds 135 Genetically induced edit Virally induced edit Viral infections play a role in the development of a number of autoimmune diseases including human type 1 diabetes However the mechanisms by which viruses are involved in the induction of type 1 DM are not fully understood Virus induced models are used to study the etiology and pathogenesis of the disease in particular the mechanisms by which environmental factors contribute to or protect against the occurrence of type 1 DM 136 Among the most commonly used are Coxsackie virus lymphocytic choriomeningitis virus encephalomyocarditis virus and Kilham rat virus Examples of virus induced animals include NOD mice infected with coxsackie B4 that developed type 1 DM within two weeks 137 References edit a b c d e f g h Causes of Diabetes NIDDK August 2014 Archived from the original on 10 August 2016 Retrieved 31 July 2016 a b c d e f g Types of Diabetes NIDDK February 2014 Archived from the original on 16 August 2016 Retrieved 31 July 2016 Diabetes Blue Circle Symbol International Diabetes Federation 17 March 2006 Archived from the original on 5 August 2007 a b c d e f g h i j k Diabetes Fact sheet N 312 WHO November 2016 Archived from the original on 26 August 2013 Retrieved 29 May 2017 Diabetes mellitus Type 1 Autoimmune Registry Inc Retrieved 15 June 2022 a b c d e f g h i Chiang JL Kirkman MS Laffel LM Peters AL July 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 PMC 5865481 PMID 24935775 a b c Elfstrom P Sundstrom J Ludvigsson JF November 2014 Systematic review with meta analysis associations between coeliac disease and type 1 diabetes Alimentary Pharmacology amp Therapeutics 40 10 1123 1132 doi 10 1111 apt 12973 PMID 25270960 S2CID 25468009 a b Diagnosis of Diabetes and Prediabetes NIDDK May 2015 Archived from the original on 16 August 2016 Retrieved 31 July 2016 a b Daneman D March 2006 Type 1 diabetes Lancet 367 9513 847 858 doi 10 1016 S0140 6736 06 68341 4 PMID 16530579 S2CID 21485081 Torpy JM Lynm C Glass RM September 2007 JAMA patient page Type 1 diabetes JAMA 298 12 1472 doi 10 1001 jama 298 12 1472 PMID 17895465 Alternative Devices for Taking Insulin NIDDK July 2016 Archived from the original on 16 August 2016 Retrieved 31 July 2016 Fast Facts Data and Statistics about Diabetes American Diabetes Association Archived from the original on 16 December 2015 Retrieved 25 July 2014 Global report on diabetes PDF World Health Organization 2016 pp 26 27 ISBN 978 92 4 156525 7 Archived PDF from the original on 7 October 2016 Retrieved 31 July 2016 Skyler J 2012 Atlas of diabetes 4th ed New York Springer pp 67 68 ISBN 978 1 4614 1028 7 Archived from the original on 8 September 2017 a b Atkinson et al 2020 Table 36 1 a b Wolsdorf amp Garvey 2016 Type 1 Diabetes a b c Atkinson et al 2020 Clinical presentation a b DiMeglio Evans Molina amp Oram 2018 p 2449 a b DKA Ketoacidosis amp Ketones American Diabetes Association Retrieved 28 July 2021 a b c Delli amp Lernmark 2016 Signs and symptoms a b c d Katsarou et al 2017 p 1 a b c Katsarou et al 2017 Epidemiology Katsarou et al 2017 Introduction Knip M Veijola R Virtanen SM Hyoty H Vaarala O Akerblom HK December 2005 Environmental triggers and determinants of type 1 diabetes Diabetes 54 Suppl 2 S125 S136 doi 10 2337 diabetes 54 suppl 2 S125 PMID 16306330 a b c d e Norris Johnson amp Stene 2020 Environmental factors a b c d Norris Johnson amp Stene 2020 Trends in epidemiology a b Norris Johnson amp Stene 2020 Infections Norris Johnson amp Stene 2020 The hygiene hypothesis and proxies of microbial exposures a b c d DiMeglio Evans Molina amp Oram 2018 p 2450 a b c Repaske 2016 Additional medications that decrease insulin release Repaske 2016 A common medication that decreases insulin release Thayer KA Heindel JJ Bucher JR Gallo MA June 2012 Role of environmental chemicals in diabetes and obesity a National Toxicology Program workshop review Environmental Health Perspectives Review 120 6 779 789 doi 10 1289 ehp 1104597 PMC 3385443 PMID 22296744 Pyriminil U S National Library of Medicine Archived from the original on 4 July 2013 Lucier Jessica Tabatabai Laila 8 May 2020 SAT 677 A Case of Opdivo Induced Type 1 Diabetes Journal of the Endocrine Society 4 Suppl 1 SAT 677 doi 10 1210 jendso bvaa046 469 PMC 7208640 Definition and Diagnosis of Diabetes Mellitus and Intermediate Hyperglycemia PDF Geneva World Health Organization 2006 p 1 ISBN 978 92 4 159493 6 Retrieved 28 July 2021 American Diabetes Association January 2021 2 Classification and Diagnosis of Diabetes Standards of Medical Care in Diabetes 2021 Diabetes Care American Diabetes Association 44 Suppl 1 S15 S33 doi 10 2337 dc21 S002 PMID 33298413 a b c d Butler amp Misselbrook 2020 What is the next investigation a b c DiMeglio Evans Molina amp Oram 2018 p 2453 Katsarou et al 2017 p 11 a b c d Smith amp Harris 2018 Self monitoring a b American Diabetes Association 6 2021 Glycemic assessment DiMeglio Evans Molina amp Oram 2018 Management of clinical disease a b c d e f Atkinson et al 2020 Insulin Therapy Overview Diabetes in pregnancy management from preconception to the postnatal period Guidance NICE www nice org uk 25 February 2015 Retrieved 31 January 2024 Lee Tara T M Collett Corinne Bergford Simon Hartnell Sara Scott Eleanor M Lindsay Robert S Hunt Katharine F McCance David R Barnard Kelly Katharine Rankin David Lawton Julia Reynolds Rebecca M Flanagan Emma Hammond Matthew Shepstone Lee 26 October 2023 Automated Insulin Delivery in Women with Pregnancy Complicated by Type 1 Diabetes New England Journal of Medicine 389 17 1566 1578 doi 10 1056 NEJMoa2303911 ISSN 0028 4793 Closed loop insulin systems are effective for pregnant women with type 1 diabetes NIHR Evidence 16 January 2024 Atkinson et al 2020 Use of Adjunctive Drugs in T1DM Atkinson et al 2020 Nutrition Therapy Seckold R Fisher E de Bock M King BR Smart CE March 2019 The ups and downs of low carbohydrate diets in the management of Type 1 diabetes a review of clinical outcomes Diabetic Medicine Review 36 3 326 334 doi 10 1111 dme 13845 PMID 30362180 S2CID 53102654 Low carbohydrate diets are of interest for improving glycaemic outcomes in the management of Type 1 diabetes There is limited evidence to support their routine use in the management of Type 1 diabetes Ly TT 2015 Technology and type 1 diabetes Closed loop therapies Current Pediatrics Reports 3 2 170 176 doi 10 1007 s40124 015 0083 y S2CID 68302123 Stress diabetes org American Diabetes Association Archived from the original on 12 November 2014 Retrieved 11 November 2014 a b c d e Atkinson et al 2020 Physical Activity and Exercise Atkinson et al 2020 Pancreas and Islet Cell Transplantation a b Robertson RP Davis C Larsen J Stratta R Sutherland DE April 2006 Pancreas and islet transplantation in type 1 diabetes Diabetes Care 29 4 935 doi 10 2337 diacare 29 04 06 dc06 9908 PMID 16567844 Dean et al 2017 Simultaneous pancreas kidney transplant Dean et al 2017 Outcomes of pancreas transplantation a b Shapiro Pokrywczynska amp Ricordi 2017 Main a b Rickels amp Robertson 2019 Islet allotransplantation for the treatment of type 1 diabetes Rickels amp Robertson 2019 Long term outcomes and comparison with pancreas transplantation Shapiro Pokrywczynska amp Ricordi 2017 Indications for islet transplantation FDA Approves First Cellular Therapy to Treat Patients with Type 1 Diabetes U S Food and Drug Administration FDA Press release 28 June 2023 Retrieved 28 June 2023 nbsp This article incorporates text from this source which is in the public domain a b c d DiMeglio Evans Molina amp Oram 2018 The immune phenotype of type 1 diabetes DiMeglio Evans Molina amp Oram 2018 Diagnosis a b c d e DiMeglio Evans Molina amp Oram 2018 The b cell phenotype of type 1 diabetes a b c d Atkinson et al 2020 Mechanisms of Beta Cell Death in T1DM Farhy LS McCall AL July 2015 Glucagon the new insulin in the pathophysiology of diabetes Current Opinion in Clinical Nutrition and Metabolic Care 18 4 407 414 doi 10 1097 mco 0000000000000192 PMID 26049639 S2CID 19872862 a b Yosten GL February 2018 Alpha cell dysfunction in type 1 diabetes Peptides 100 54 60 doi 10 1016 j peptides 2017 12 001 PMID 29412832 S2CID 46878644 Brown RJ Sinaii N Rother KI July 2008 Too much glucagon too little insulin time course of pancreatic islet dysfunction in new onset type 1 diabetes Diabetes Care 31 7 1403 1404 doi 10 2337 dc08 0575 PMC 2453684 PMID 18594062 Unger RH Cherrington AD January 2012 Glucagonocentric restructuring of diabetes a pathophysiologic and therapeutic makeover The Journal of Clinical Investigation 122 1 4 12 doi 10 1172 JCI60016 PMC 3248306 PMID 22214853 Meier JJ Kjems LL Veldhuis JD Lefebvre P Butler PC April 2006 Postprandial suppression of glucagon secretion depends on intact pulsatile insulin secretion further evidence for the intraislet insulin hypothesis Diabetes 55 4 1051 1056 doi 10 2337 diabetes 55 04 06 db05 1449 PMID 16567528 Cooperberg BA Cryer PE December 2009 Beta cell mediated signaling predominates over direct alpha cell signaling in the regulation of glucagon secretion in humans Diabetes Care 32 12 2275 2280 doi 10 2337 dc09 0798 PMC 2782990 PMID 19729529 Paolisso G Sgambato S Torella R Varricchio M Scheen A D Onofrio F Lefebvre PJ June 1988 Pulsatile insulin delivery is more efficient than continuous infusion in modulating islet cell function in normal subjects and patients with type 1 diabetes The Journal of Clinical Endocrinology and Metabolism 66 6 1220 1226 doi 10 1210 jcem 66 6 1220 PMID 3286673 a b Banarer S McGregor VP Cryer PE April 2002 Intraislet hyperinsulinemia prevents the glucagon response to hypoglycemia despite an intact autonomic response Diabetes 51 4 958 965 doi 10 2337 diabetes 51 4 958 PMID 11916913 Raju B Cryer PE March 2005 Loss of the decrement in intraislet insulin plausibly explains loss of the glucagon response to hypoglycemia in insulin deficient diabetes documentation of the intraislet insulin hypothesis in humans Diabetes 54 3 757 764 doi 10 2337 diabetes 54 3 757 PMID 15734853 a b c Tesfaye N Seaquist ER November 2010 Neuroendocrine responses to hypoglycemia Annals of the New York Academy of Sciences 1212 1 12 28 Bibcode 2010NYASA1212 12T doi 10 1111 j 1749 6632 2010 05820 x PMC 2991551 PMID 21039590 a b Reno CM Litvin M Clark AL Fisher SJ March 2013 Defective counterregulation and hypoglycemia unawareness in diabetes mechanisms and emerging treatments Endocrinology and Metabolism Clinics of North America 42 1 15 38 doi 10 1016 j ecl 2012 11 005 PMC 3568263 PMID 23391237 Martin Timon I Del Canizo Gomez FJ July 2015 Mechanisms of hypoglycemia unawareness and implications in diabetic patients World Journal of Diabetes 6 7 912 926 doi 10 4239 wjd v6 i7 912 PMC 4499525 PMID 26185599 a b Mundinger TO Taborsky GJ October 2016 Early sympathetic islet neuropathy in autoimmune diabetes lessons learned and opportunities for investigation Diabetologia 59 10 2058 2067 doi 10 1007 s00125 016 4026 0 PMC 6214182 PMID 27342407 Mundinger TO Mei Q Foulis AK Fligner CL Hull RL Taborsky GJ August 2016 Human Type 1 Diabetes Is Characterized by an Early Marked Sustained and Islet Selective Loss of Sympathetic Nerves Diabetes 65 8 2322 2330 doi 10 2337 db16 0284 PMC 4955989 PMID 27207540 a b c d e f Hypoglycemia Low blood sugar American Diabetes Association Retrieved 20 March 2022 a b c d e f DiMeglio Evans Molina amp Oram 2018 p 2455 a b DiMeglio Evans Molina amp Oram 2018 Complications of type 1 diabetes a b Cashen amp Petersen 2019 Diagnosis screening and prevention a b Cashen amp Petersen 2019 Epidemiology Brownlee et al 2020 Pathophysiology of diabetic retinopathy DiMeglio Evans Molina amp Oram 2018 p 2456 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 3 774 779 doi 10 2337 diabetes 55 03 06 db05 1417 PMID 16505242 Granberg V Ejskjaer N Peakman M Sundkvist G August 2005 Autoantibodies to autonomic nerves associated with cardiac and peripheral autonomic neuropathy Diabetes Care 28 8 1959 1964 doi 10 2337 diacare 28 8 1959 PMID 16043739 Huxley RR Peters SA Mishra GD Woodward M March 2015 Risk of all cause mortality and vascular events in women versus men with type 1 diabetes a systematic review and meta analysis The Lancet Diabetes amp Endocrinology 3 3 198 206 doi 10 1016 S2213 8587 14 70248 7 PMID 25660575 Roy T Lloyd CE October 2012 Epidemiology of depression and diabetes a systematic review Journal of Affective Disorders 142 Suppl S8 21 doi 10 1016 S0165 0327 12 70004 6 PMID 23062861 a b See JA Kaukinen K Makharia GK Gibson PR Murray JA October 2015 Practical insights into gluten free diets Nature Reviews Gastroenterology amp Hepatology Review 12 10 580 591 doi 10 1038 nrgastro 2015 156 PMID 26392070 S2CID 20270743 Coeliac disease in T1DM is asymptomatic Clinical manifestations of coeliac disease such as abdominal pain gas bloating diarrhoea and weight loss can be present in patients with T1DM but are often attributed to poor control of diabetes gastroparesis or diabetic neuropathy Chen HS Su LT Lin SZ Sung FC Ko MC Li CY January 2012 Increased risk of urinary tract calculi among patients with diabetes mellitus a population based cohort study Urology 79 1 86 92 doi 10 1016 j urology 2011 07 1431 PMID 22119251 James R Hijaz A October 2014 Lower urinary tract symptoms in women with diabetes mellitus a current review Current Urology Reports 15 10 440 doi 10 1007 s11934 014 0440 3 PMID 25118849 S2CID 30653959 a b c d Sexual Dysfunction in Women Diabetes co uk Diabetes Digital Media Ltd Archived from the original on 9 November 2014 Retrieved 28 November 2014 Goswami SK Vishwanath M Gangadarappa SK Razdan R Inamdar MN August 2014 Efficacy of ellagic acid and sildenafil in diabetes induced sexual dysfunction Pharmacognosy Magazine 10 Suppl 3 S581 S587 doi 10 4103 0973 1296 139790 PMC 4189276 PMID 25298678 ProQuest 1610759650 Escobar Morreale HF Roldan B Barrio R Alonso M Sancho J de la Calle H Garcia Robles R November 2000 High prevalence of the polycystic ovary syndrome and hirsutism in women with type 1 diabetes mellitus The Journal of Clinical Endocrinology and Metabolism 85 11 4182 4187 doi 10 1210 jcem 85 11 6931 PMID 11095451 Codner E Escobar Morreale HF April 2007 Clinical review Hyperandrogenism and polycystic ovary syndrome in women with type 1 diabetes mellitus The Journal of Clinical Endocrinology and Metabolism 92 4 1209 1216 doi 10 1210 jc 2006 2641 PMID 17284617 a b Atkinson et al 2020 Other Complications Redondo Steck amp Pugliese 2018 Evidence for the contribution of genetics to type I diabetes What is Type 1 Diabetes Centers for Disease Control and Prevention 11 March 2022 Retrieved 15 February 2023 FDA Approves Tzield Drugs com November 2022 Retrieved 15 February 2023 a b Atkinson et al 2020 Immunosuppresion von Scholten et al 2021 Antigen vaccination a b Dayan et al 2019 Previous prevention trials Diabetes World Health Organization Archived from the original on 26 January 2011 Retrieved 24 January 2011 a b Atkinson et al 2020 Diagnosis Aanstoot HJ Anderson BJ Daneman D Danne T Donaghue K Kaufman F et al October 2007 The global burden of youth diabetes perspectives and potential Pediatric Diabetes 8 8 s8 1 44 doi 10 1111 j 1399 5448 2007 00326 x PMID 17767619 S2CID 222102948 a b Fast Facts PDF American Diabetes Association Archived from the original PDF on 29 April 2015 Australian Institute of Health and Welfare 2015 Incidence of type 1 diabetes in Australia 2000 2013 Archived from the original on 7 October 2016 Retrieved 19 October 2016 Shaw J 2012 diabetes the silent pandemic and its impact on Australia PDF Archived PDF from the original on 7 October 2016 Retrieved 19 October 2016 Weiss A Donnachie E Beyerlein A Ziegler AG Bonifacio E 22 May 2023 Type 1 Diabetes Incidence and Risk in Children With a Diagnosis of COVID 19 PDF JAMA Research Letter 329 23 2089 2091 doi 10 1001 jama 2023 8674 PMC 10203966 PMID 37213115 S2CID 258831851 a b Atkinson et al 2020 Introduction Bottazzo GF Florin Christensen A Doniach D November 1974 Islet cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies Lancet 2 7892 1279 1283 doi 10 1016 s0140 6736 74 90140 8 PMID 4139522 Herold KC Vignali DA Cooke A Bluestone JA April 2013 Type 1 diabetes translating mechanistic observations into effective clinical outcomes Nature Reviews Immunology 13 4 243 256 doi 10 1038 nri3422 PMC 4172461 PMID 23524461 Johnson L 18 November 2008 Study Cost of diabetes 218B USA Today Associated Press Archived from the original on 1 July 2012 About DRWF Diabetes Research amp Wellness Foundation Archived from the original on 11 May 2013 a b Minami K Seino S March 2013 Current status of regeneration of pancreatic b cells Journal of Diabetes Investigation 4 2 131 141 doi 10 1111 jdi 12062 PMC 4019265 PMID 24843642 Pagliuca FW Millman JR Gurtler M Segel M Van Dervort A Ryu JH et al October 2014 Generation of functional human pancreatic b cells in vitro Cell 159 2 428 439 doi 10 1016 j cell 2014 09 040 PMC 4617632 PMID 25303535 Rezania A Bruin JE Arora P Rubin A Batushansky I Asadi A et al November 2014 Reversal of diabetes with insulin producing cells derived in vitro from human pluripotent stem cells Nature Biotechnology 32 11 1121 1133 doi 10 1038 nbt 3033 PMID 25211370 S2CID 205280579 Boughton amp Hovorka 2020 Introduction Boughton amp Hovorka 2020 Regulatory Approval of Closed Loop Systems Ramli R Reddy M Oliver N July 2019 Artificial Pancreas Current Progress and Future Outlook in the Treatment of Type 1 Diabetes Drugs 79 10 1089 1101 doi 10 1007 s40265 019 01149 2 hdl 10044 1 71348 PMID 31190305 S2CID 186207231 a b King AJ 2020 Animal models of diabetes methods and protocols New York NY Humana Press ISBN 978 1 0716 0385 7 OCLC 1149391907 page needed a b c d Atkinson et al 2020 Animal models NOD ShiLtJ The Jackson Laborataory Retrieved 18 January 2022 a b c d e f Pandey S Dvorakova MC 7 January 2020 Future Perspective of Diabetic Animal Models Endocrine Metabolic amp Immune Disorders Drug Targets 20 1 25 38 doi 10 2174 1871530319666190626143832 PMC 7360914 PMID 31241444 Chen D Thayer TC Wen L Wong FS 2020 Mouse Models of Autoimmune Diabetes The Nonobese Diabetic NOD Mouse In King AJ ed Animal Models of Diabetes Methods in Molecular Biology Vol 2128 New York NY Springer US pp 87 92 doi 10 1007 978 1 0716 0385 7 6 ISBN 978 1 0716 0384 0 PMC 8253669 PMID 32180187 Lenzen S Arndt T Elsner M Wedekind D Jorns A 2020 Rat Models of Human Type 1 Diabetes In King AJ ed Animal Models of Diabetes Methods in Molecular Biology Vol 2128 New York NY Springer US pp 69 85 doi 10 1007 978 1 0716 0385 7 5 ISBN 978 1 0716 0384 0 PMID 32180186 S2CID 212741496 Al Awar A Kupai K Veszelka M Szucs G Attieh Z Murlasits Z et al 2016 Experimental Diabetes Mellitus in Different Animal Models Journal of Diabetes Research 2016 9051426 doi 10 1155 2016 9051426 PMC 4993915 PMID 27595114 Lenzen S Tiedge M Elsner M Lortz S Weiss H Jorns A et al September 2001 The LEW 1AR1 Ztm iddm rat a new model of spontaneous insulin dependent diabetes mellitus Diabetologia 44 9 1189 1196 doi 10 1007 s001250100625 PMID 11596676 Lenzen S October 2017 Animal models of human type 1 diabetes for evaluating combination therapies and successful translation to the patient with type 1 diabetes Diabetes Metabolism Research and Reviews 33 7 e2915 doi 10 1002 dmrr 2915 PMID 28692149 S2CID 34331597 Radenkovic M Stojanovic M Prostran M March 2016 Experimental diabetes induced by alloxan and streptozotocin The current state of the art Journal of Pharmacological and Toxicological Methods 78 13 31 doi 10 1016 j vascn 2015 11 004 PMID 26596652 Salpea P Cosentino C Igoillo Esteve M 2020 A Review of Mouse Models of Monogenic Diabetes and ER Stress Signaling In King AJ ed Animal Models of Diabetes Methods in Molecular Biology Vol 2128 New York NY Springer US pp 55 67 doi 10 1007 978 1 0716 0385 7 4 ISBN 978 1 0716 0384 0 PMID 32180185 S2CID 212740474 Chang JH Gurley SB 2012 Assessment of Diabetic Nephropathy in the Akita Mouse In Hans Georg J Hadi AH Schurmann A eds Animal Models in Diabetes Research Methods in Molecular Biology Vol 933 Totowa NJ Humana Press pp 17 29 doi 10 1007 978 1 62703 068 7 2 ISBN 978 1 62703 067 0 PMID 22893398 King AJ Estil Les E Montanya E 2020 Use of Streptozotocin in Rodent Models of Islet Transplantation In King AJ ed Animal Models of Diabetes Methods in Molecular Biology Vol 2128 New York NY Springer US pp 135 147 doi 10 1007 978 1 0716 0385 7 10 ISBN 978 1 0716 0384 0 PMID 32180191 S2CID 212739708 Christoffersson G Flodstrom Tullberg M 2020 Mouse Models of Virus Induced Type 1 Diabetes In King AJ ed Animal Models of Diabetes Methods in Molecular Biology Vol 2128 New York NY Springer US pp 93 105 doi 10 1007 978 1 0716 0385 7 7 ISBN 978 1 0716 0384 0 PMID 32180188 S2CID 212739248 King AJ June 2012 The use of animal models in diabetes research British Journal of Pharmacology 166 3 877 894 doi 10 1111 j 1476 5381 2012 01911 x PMC 3417415 PMID 22352879 Works cited editAmerican Diabetes Association January 2021 6 Glycemic Targets Standards of Medical Care in Diabetes 2021 Diabetes Care 44 Suppl 1 S73 S84 doi 10 2337 dc21 S006 PMID 33298417 S2CID 228087604 Atkinson MA Mcgill DE Dassau E Laffel L 2020 Type 1 diabetes mellitus Williams Textbook of Endocrinology Elsevier pp 1403 1437 Boughton CK Hovorka R August 2020 The artificial pancreas Curr Opin Organ Transplant 25 4 336 342 doi 10 1097 MOT 0000000000000786 PMID 32618719 S2CID 220326946 Brownlee M Aiello LP Sun JK Cooper ME Feldman EL Plutzky J Boulton AJ 2020 Complications of Diabetes Mellitus Williams Textbook of Endocrinology Elsevier pp 1438 1524 ISBN 978 0 323 55596 8 Butler AE Misselbrook D August 2020 Distinguishing between type 1 and type 2 diabetes The BMJ 370 m2998 doi 10 1136 bmj m2998 PMID 32784223 S2CID 221097632 Cashen K Petersen T August 2019 Diabetic Ketoacidosis Pediatr Rev 40 8 412 420 doi 10 1542 pir 2018 0231 PMID 31371634 S2CID 199381655 Dayan CM Korah M Tatovic D Bundy BN Herold KC October 2019 Changing the landscape for type 1 diabetes the first step to prevention Lancet 394 10205 1286 1296 doi 10 1016 S0140 6736 19 32127 0 PMID 31533907 S2CID 202575545 Dean PG Kukla A Stegall MD Kudva YC April 2017 Pancreas transplantation The BMJ 357 j1321 doi 10 1136 bmj j1321 PMID 28373161 S2CID 11374615 Delli AJ Lernmark A 2016 Type 1 insulin dependent diabetes mellitus etiology pathogenesis prediction and prevention In Jameson JL ed Endocrinology Adult and Pediatric 7 ed Saunders pp 672 690 ISBN 978 0 323 18907 1 DiMeglio LA Evans Molina C Oram RA June 2018 Type 1 diabetes Lancet 391 10138 2449 2462 doi 10 1016 S0140 6736 18 31320 5 PMC 6661119 PMID 29916386 Katsarou A Gudbjornsdottir S Rawshani A Dabelea D Bonifacio E Anderson BJ et al March 2017 Type 1 diabetes mellitus Nature Reviews Disease Primers 3 17016 doi 10 1038 nrdp 2017 16 PMID 28358037 S2CID 23127616 Norris JM Johnson RK Stene LC March 2020 Type 1 diabetes early life origins and changing epidemiology The Lancet Diabetes amp Endocrinology 8 3 226 238 doi 10 1016 S2213 8587 19 30412 7 PMC 7332108 PMID 31999944 Redondo MJ Steck AK Pugliese A May 2018 Genetics of type 1 diabetes Pediatric Diabetes 19 3 346 353 doi 10 1111 pedi 12597 PMC 5918237 PMID 29094512 Repaske DR September 2016 Medication induced diabetes mellitus Pediatr Diabetes 17 6 392 7 doi 10 1111 pedi 12406 PMID 27492964 S2CID 4684512 Rickels MR Robertson RP April 2019 Pancreatic Islet Transplantation in Humans Recent Progress and Future Directions Endocr Rev 40 2 631 668 doi 10 1210 er 2018 00154 PMC 6424003 PMID 30541144 Shapiro AM Pokrywczynska M Ricordi C May 2017 Clinical pancreatic islet transplantation Nat Rev Endocrinol 13 5 268 277 doi 10 1038 nrendo 2016 178 PMID 27834384 S2CID 28784928 Smith A Harris C August 2018 Type 1 diabetes Management strategies American Family Physician 98 3 154 156 PMID 30215903 Retrieved 12 January 2022 von Scholten BJ Kreiner FF Gough SC von Herrath M May 2021 Current and future therapies for type 1 diabetes Diabetologia 64 5 1037 1048 doi 10 1007 s00125 021 05398 3 PMC 8012324 PMID 33595677 Wolsdorf Joseph I Garvey Katharine C 2016 Management of Diabetes in Children Endocrinology Adult and Pediatric pp 854 882 e6 doi 10 1016 B978 0 323 18907 1 00049 4 ISBN 978 0 323 18907 1 External links editNational Institute of Diabetes and Digestive and Kidney Diseases NIDDK Archived 25 April 2011 at the Wayback Machine Diabetes in America Textbook PDFs IDF Diabetes Atlas Type 1 Diabetes Archived 30 October 2009 at the Wayback Machine at the American Diabetes Association ADA s Standards of Medical Care in Diabetes 2019 Retrieved from https en wikipedia org w index php title Type 1 diabetes amp oldid 1207292176, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.