Insulin resistance










Insulin resistance
Specialty Endocrinology

Insulin resistance (IR) is considered as a pathological condition in which cells fail to respond normally to the hormone insulin.[1] To prevent hyperglycemia and noticeable organ damage over time,[2] the body produces insulin when glucose starts to be released into the bloodstream, primarily from the digestion of carbohydrates in the diet. Under normal conditions of insulin reactivity, this insulin response triggers glucose being taken into body cells, to be used for energy, and inhibits the body from using fat for energy, thereby causing the concentration of glucose in the blood to decrease as a result, staying within the normal range even when a large amount of carbohydrates is consumed. Carbohydrates comprise simple sugars, i.e. monosaccharides, such as glucose and fructose, disaccharides, such as cane sugar, and polysaccharides, e.g. starches. Fructose, which is metabolised into triglycerides in the liver, stimulates insulin production through another mechanism, and can have a more potent effect than other carbohydrates. A habitually high intake of carbohydrates, and particularly fructose, e.g. with sweetened beverages, contributes to insulin resistance and has been linked to weight gain and obesity.[3][4][5] If excess blood sugar is not sufficiently absorbed by cells even in the presence of insulin, the increase in the level of blood sugar can result in the classic hyperglycemic triad of polyphagia (increased appetite), polydipsia (increased thirst), and polyuria (increased urination). Avoiding carbohydrates and sugars, a no-carbohydrate diet or fasting can reverse insulin resistance.[6][7]




Contents






  • 1 Overview


  • 2 Associated risk factors


  • 3 Cause


    • 3.1 Molecular mechanism


    • 3.2 Diet


    • 3.3 Sedentary lifestyle


    • 3.4 Protease inhibitors


    • 3.5 Cellular level


    • 3.6 Molecular


    • 3.7 Disease


    • 3.8 HCV and insulin resistance


    • 3.9 Genetics




  • 4 Pathophysiology


  • 5 Diagnosis


    • 5.1 Fasting insulin levels


    • 5.2 Glucose tolerance testing


    • 5.3 Measuring insulin resistance


      • 5.3.1 Hyperinsulinemic euglycemic clamp


      • 5.3.2 Modified insulin suppression test




    • 5.4 Alternatives




  • 6 Management


  • 7 History


    • 7.1 Adaptive Explanations




  • 8 See also


  • 9 References


  • 10 Further reading


  • 11 External links





Overview


When the body produces insulin under conditions of insulin resistance, the cells are resistant to the insulin and are unable to use it as effectively, leading to high blood sugar. Beta cells in the pancreas subsequently increase their production of insulin, further contributing to a high blood insulin level. This often remains undetected and can contribute to the development of type 2 diabetes, obesity or latent autoimmune diabetes of adults.[8] Although this type of chronic insulin resistance is harmful, during acute illness it is actually a well-evolved protective mechanism.


Recent investigations have revealed that insulin resistance helps to conserve the brain's glucose supply by preventing muscles from taking up excessive glucose.[1] In theory, insulin resistance should even be strengthened under harsh metabolic conditions such as pregnancy, during which the expanding fetal brain demands more glucose.


People who develop type 2 diabetes usually pass through earlier stages of insulin resistance and prediabetes, although those often go undiagnosed. Insulin resistance is a syndrome (a set of signs and symptoms) resulting from reduced insulin activity; it is also part of a larger constellation of symptoms called the metabolic syndrome.


Insulin resistance may also develop in patients who have recently experienced abdominal or bariatric procedures.[9] This acute form of insulin resistance that may result post-operatively tends to increase over the short term, with sensitivity to insulin typically returning to patients after about five days.[10]



Associated risk factors


Several associated risk factors include the following:



  • Genetic factors (inherited component):

    • Family history of type 2 diabetes


      • Insulin receptor mutations (Donohue syndrome)


      • LMNA mutations (familial partial lipodystrophy)[citation needed][11][12]



    • Cultural variables, such as diet varying with race and class; factors related to stress, socio-economic status and history have been shown to activate the stress response, which increases the production of glucose and insulin resistance, as well as inhibiting pancreatic function and thus might be of importance,[13][14] although it is not fully corroborated by the scientific evidence.



  • Particular physiological conditions and environmental factors:

    • Age 40–45 years or older[13][14]


    • Obesity[13][14]

    • The tendency to store fat preferentially in the abdomen (also known as "abdominal obesity)", as opposed to storing it in hips and thighs[14]

    • Sedentary lifestyle, lack of physical exercise[13][14]

    • Hypertension[13]

    • High triglyceride level (hypertriglyceridemia)[13]

    • Low level of high-density lipoprotein (also known as HDL cholesterol or "good cholesterol")[13]


    • Prediabetes, blood glucose levels have been too high in the past, i.e. the patient's body has previously shown slight problems with its production and usage of insulin ("previous evidence of impaired glucose homeostasis")[13][14]

    • Having developed gestational diabetes during past pregnancies[13][14]

    • Giving birth to a baby weighing more than 9 pounds (a bit over 4 kilograms)[13][14]



  • Pathology:

    • Obesity and overweight (BMI > 25)

    • Metabolic syndrome (hyperlipidemia + HDL cholesterol level < 0.90 mmol/L or triglyceride level > 2.82 mmol/L), hypertension (> 140/90 mmHg), or arteriosclerosis

    • Liver pathologies

    • Infection (Hepatitis C[15])

    • Hemochromatosis

    • Gastroparesis


    • Polycystic ovary syndrome (PCOS)




  • Hypercortisolism (e.g., Cushing's syndrome, glucocorticoid therapy)[16]


  • Medications (e.g., glucosamine, rifampicin, isoniazid, olanzapine, risperidone, progestogens, glucocorticoids, methadone, many antiretrovirals)



Cause



Molecular mechanism


Insulin resistance implies that the body's cells (primarily muscle) lose sensitivity to insulin, a hormone secreted by the pancreas to promote glucose utilization. At the molecular level, a cell senses insulin through insulin receptors, with the signal propagating through a cascade of molecules collectively known as PI3K/Akt/mTOR signaling pathway.[17] Recent studies suggested that the pathway may operate as a bistable switch under physiologic conditions for certain types of cells, and insulin response may well be a threshold phenomenon.[1][17][18] The pathway's sensitivity to insulin may be blunted by many factors such as free fatty acids,[19] causing insulin resistance. From a broader perspective, however, sensitivity tuning (including sensitivity reduction) is a common practice for an organism to adapt to the changing environment or metabolic conditions.[20] Pregnancy, for example, is a prominent change of metabolic conditions, under which the mother has to reduce her muscles' insulin sensitivity to spare more glucose for the brains (the mother's brain and the fetal brain). This can be achieved through raising the response threshold (i.e., postponing the onset of sensitivity) by secreting placental growth factor to interfere with the interaction between insulin receptor substrate (IRS) and PI3K, which is the essence of the so-called adjustable threshold hypothesis of insulin resistance.[1]



Diet


It is well known that insulin regulates the conversion of carbohydrates into fats (triglycerides) by promoting the absorption of, especially, glucose from the blood into fat cells.[21] The intake of simple sugars, and particularly fructose, is a factor that contributes to insulin resistance.[3] Fructose is metabolized by the liver into triglycerides, and, as mentioned above, tends to raise their levels in the blood stream.


Once established, insulin resistance would result in increased circulating levels of insulin. Since insulin is the primary hormonal signal for energy storage into fat cells, which tend to retain their sensitivity in the face of hepatic and skeletal muscle resistance, IR stimulates the formation of new fatty tissue and accelerates weight gain.[22]


Insulin resistance and type 2 diabetes are associated with excess body weight.[23] A possible explanation is that both insulin resistance and obesity often have the same cause, systematic overeating, which has the potential to lead to insulin resistance and obesity due to repeated administration of excess levels of glucose, which stimulate insulin secretion; excess levels of fructose, which raise triglyceride levels in the bloodstream; and fats, which may be absorbed easily by the adipose cells, and tend to end up as fatty tissue in a hypercaloric diet.[citation needed] Some scholars go as far as to claim that neither insulin resistance, nor obesity really are metabolic disorders per se, but simply adaptive responses to sustained caloric surplus, intended to protect bodily organs from lipotoxicity (unsafe levels of lipids in the bloodstream and tissues): "Obesity should therefore not be regarded as a pathology or disease, but rather as the normal, physiologic response to sustained caloric surplus... As a consequence of the high level of lipid accumulation in insulin target tissues including skeletal muscle and liver, it has been suggested that exclusion of glucose from lipid-laden cells is a compensatory defense against further accumulation of lipogenic substrate."[24]


Fast food meals combined with drinks containing sugar typically possess several characteristics, all of which have independently been linked to IR: they are sugar rich, palatable, and cheap, increasing risk of overeating and leptin resistance; simultaneously, they are high in dietary fat and fructose, and low in omega-3 and fiber; and they usually have high glycemic indices.[22] Overconsumption of cheap sugar rich meals and beverages have been proposed as a fundamental factor behind the metabolic syndrome epidemic and all its constituents.


Vitamin D deficiency also is associated with insulin resistance.[25]


Elevated levels of free fatty acids and triglycerides in the blood stream and tissues also have been found in many studies to occur in states of insulin resistance.[26][27][28][29] Triglyceride levels are driven by a variety of dietary factors.


In studies on animals caloric intake that is far in excess of animals' energy needs results in rapid weight gain and significant insulin resistance after just three weeks (in rats).[30][26]



Sedentary lifestyle


Sedentary lifestyle increases the likelihood of development of insulin resistance.[31][32] It has been estimated that each 500 kcal/week increment in physical activity related energy expenditure, reduces the lifetime risk of type 2 diabetes by 9%.[33] A different study found that vigorous exercise at least once a week reduced the risk of type 2 diabetes in women by 33%.[34]



Protease inhibitors


Protease inhibitors found in HIV drugs are linked to insulin resistance.[35]



Cellular level


At the cellular level, much of the variance in insulin sensitivity between untrained, non-diabetic humans may be explained by two mechanisms: differences in phospholipid profiles of skeletal muscle cell membranes, and in intramyocellular lipid (ICML) stores within these cells.[36] High levels of lipids in the bloodstream have the potential to result in accumulation of triglycerides and their derivatives within muscle cells, which activate proteins Kinase C-ε and C-θ, ultimately reducing the glucose uptake at any given level of insulin.[27][37] This mechanism is quite fast-acting and may induce insulin resistance within days or even hours in response to a large lipid influx.[38] Draining the intracellular reserves, on the other hand, is more challenging: moderate caloric restriction alone, even over a period of several months, appears to be ineffective,[39][40] and it must be combined with physical exercise to have any effect.


In the long term, diet has the potential to change the ratio of polyunsaturated to saturated phospholipids in cell membranes, correspondingly changing cell membrane fluidity; full impact of such changes is not fully understood, but it is known that the percentage of polyunsaturated phospholipids is strongly inversely correlated with insulin resistance.[41] It is hypothesized that increasing cell membrane fluidity by increasing PUFA concentration might result in an enhanced number of insulin receptors, an increased affinity of insulin to its receptors, and a reduced insulin resistance, and vice versa.[42]


Many stressing factors may lead to increased cortisol in the bloodstream. Cortisol counteracts insulin, contributes to hyperglycemia-causing hepatic gluconeogenesis,[43] and inhibits the peripheral utilization of glucose, which eventually leads to insulin resistance.[43] It does this by decreasing the translocation of glucose transporters (especially GLUT4) to the cell membrane.[44][45]


Inflammation by itself also seems to be implicated in causing insulin resistance. Mice without JNK1-signaling do not develop insulin resistance under dietary conditions that normally produce it.[46][47] Recent study have found out the specific role of the MLK family of protein in the activation of JNK during obesity and insulin resistance.[48]


Rare type 2 diabetes cases sometimes use high levels of exogenous insulin. As short-term overdosing of insulin causes short-term insulin resistance, it has been hypothesized that chronic high dosing contributes to more permanent insulin resistance.[citation needed]



Molecular


At a molecular level, insulin resistance has been proposed to be a reaction to excess nutrition by superoxide dismutase in cell mitochondria that acts as an antioxidant defense mechanism. This link seems to exist under diverse causes of insulin resistance. It also is based on the finding that insulin resistance may be reversed rapidly by exposing cells to mitochondrial uncouplers, electron transport chain inhibitors, or mitochondrial superoxide dismutase mimetics.[49]



Disease


Recent research and experimentation has uncovered a non-obesity related connection to insulin resistance and type 2 diabetes. It has long been observed that patients who have had some kinds of bariatric surgery have increased insulin sensitivity and even remission of type 2 diabetes. It was discovered that diabetic/insulin resistant non-obese rats whose duodenum has been removed surgically, also experienced increased insulin sensitivity[50] and remission of type 2 diabetes. This suggested similar surgery in humans, and early reports in prominent medical journals[51] are that the same effect is seen in humans, at least the small number who have participated in the experimental surgical program. The speculation is, that some substance is produced in the mucosa of that initial portion of the small intestine that signals body cells to become insulin resistant. If the producing tissue is removed, the signal ceases and body cells revert to normal insulin sensitivity. No such substance has been found as yet, and the existence of such a substance remains speculative.[citation needed]


Insulin resistance is associated with PCOS.[52]



HCV and insulin resistance


Hepatitis C also makes people three to four times more likely to develop type 2 diabetes and insulin resistance.[15] In addition, "people with Hepatitis C who develop diabetes probably have susceptible insulin-producing cells, and probably would get it anyway, but much later in life.[15] The extra insulin resistance caused by Hepatitis C apparently brings on diabetes at age 35 or 40, instead of 65 or 70."[15]



Genetics


Variation in the NAT2 gene is associated with insulin resistance by affecting insulin sensitivity. Specifically, it is believed that the rs1208 loci near the NAT2 gene plays a role in insulin resistance. One study showed that when the expression of the NAT1 gene, the mouse equivalent of NAT2, is reduced there was a decrease in insulin stimulated glucose uptake and therefore decreases insulin sensitivity.  Further research has shown that loci near the GCKR and IGFI genes are linked to insulin resistance. Several other loci have also been determined to be associated with insulin insensitivity. These loci however, are estimated to only account for 25-44% of the genetic component of insulin resistance.[53]



Pathophysiology


One of insulin's functions is to regulate delivery of glucose into cells to provide them with energy.[54] Insulin resistant cells cannot take in glucose, amino acids and fatty acids. Thus, glucose, fatty acids and amino acids 'leak' out of the cells. A decrease in insulin/glucagon ratio inhibits glycolysis which in turn decreases energy production. The resulting increase in blood glucose may raise levels outside the normal range and cause adverse health effects, depending on dietary conditions.[55] Certain cell types such as fat and muscle cells require insulin to absorb glucose. When these cells fail to respond adequately to circulating insulin, blood glucose levels rise. The liver helps regulate glucose levels by reducing its secretion of glucose in the presence of insulin. This normal reduction in the liver's glucose production may not occur in people with insulin resistance.[56]


Insulin resistance in muscle and fat cells reduces glucose uptake (and also local storage of glucose as glycogen and triglycerides, respectively), whereas insulin resistance in liver cells results in reduced glycogen synthesis and storage and also a failure to suppress glucose production and release into the blood. Insulin resistance normally refers to reduced glucose-lowering effects of insulin. However, other functions of insulin can also be affected. For example, insulin resistance in fat cells reduces the normal effects of insulin on lipids and results in reduced uptake of circulating lipids and increased hydrolysis of stored triglycerides. Increased mobilization of stored lipids in these cells elevates free fatty acids in the blood plasma. Elevated blood fatty-acid concentrations (associated with insulin resistance and diabetes mellitus Type 2), reduced muscle glucose uptake, and increased liver glucose production all contribute to elevated blood glucose levels. High plasma levels of insulin and glucose due to insulin resistance are a major component of the metabolic syndrome. If insulin resistance exists, more insulin needs to be secreted by the pancreas. If this compensatory increase does not occur, blood glucose concentrations increase and type 2 diabetes or latent autoimmune diabetes of adults occurs.[55][57]


Any food or drink containing glucose (or the digestible carbohydrates that contain it, such as sucrose, starch, etc.) causes blood glucose levels to increase. In normal metabolism, the elevated blood glucose level instructs beta (β) cells in the Islets of Langerhans, located in the pancreas, to release insulin into the blood. The insulin, in turn, makes insulin-sensitive tissues in the body (primarily skeletal muscle cells, adipose tissue, and liver) absorb glucose, and thereby lower the blood glucose level. The beta cells reduce insulin output as the blood glucose level falls, allowing blood glucose to settle at a constant of approximately 5 mmol/L (mM) (90 mg/dL). In an insulin-resistant person, normal levels of insulin do not have the same effect in controlling blood glucose levels. During the compensated phase on insulin resistance, insulin levels are higher, and blood glucose levels are still maintained. If compensatory insulin secretion fails, then either fasting (impaired fasting glucose) or postprandial (impaired glucose tolerance) glucose concentrations increase. Eventually, type 2 diabetes or latent autoimmune diabetes occurs when glucose levels become higher throughout the day as the resistance increases and compensatory insulin secretion fails. The elevated insulin levels also have additional effects (see insulin) that cause further abnormal biological effects throughout the body.[citation needed]


The most common type of insulin resistance is associated with overweight and obesity in a condition known as the metabolic syndrome. Insulin resistance often progresses to full Type 2 diabetes mellitus (T2DM) or latent autoimmune diabetes of adults.[58][59] This often is seen when hyperglycemia develops after a meal, when pancreatic β-cells are unable to produce sufficient insulin to maintain normal blood sugar levels (euglycemia) in the face of insulin resistance. The inability of the β-cells to produce sufficient insulin in a condition of hyperglycemia is what characterizes the transition from insulin resistance to T2DM.[60]


Various disease states make body tissues more resistant to the actions of insulin. Examples include infection (mediated by the cytokine TNFα) and acidosis. Recent research is investigating the roles of adipokines (the cytokines produced by adipose tissue) in insulin resistance. TNF-α has been found in increased amounts in obese patients. Due to its effects on adipocytes it is possible that excessive TNF-α is one of several factors that leads to insulin resistance. TNF-α can inhibit lipogenesis, promote lipolysis, disrupt insulin signaling, and reduce the expression of GLUT4.[61] Animal studies have tested the theory by reducing the expression of TNF-α and its receptor in obese animals.Beneficial results were found in some studies, supporting the idea that TNF-α plays a role in insulin resistance.[62]


Certain drugs also may be associated with insulin resistance (e.g., glucocorticoids).[citation needed]


The presence of insulin leads to a kind of insulin resistance; every time a cell is exposed to insulin, the production of GLUT4 (Glucose transporter type 4) on the membrane of the cell decreases somewhat.[63] In the presence of a higher than usual level of insulin (generally caused by insulin resistance), this down-regulation acts as a kind of positive feedback, increasing the need for insulin. Exercise reverses this process in muscle tissue,[64] but if it is left unchecked, it may contribute to insulin resistance.


Elevated blood levels of glucose—regardless of cause—lead to increased glycation of proteins with changes, only a few of which are understood in any detail, in protein function throughout the body.[65][66]


Insulin resistance often is found in people with visceral adiposity (i.e., a high degree of fatty tissue within the abdomen—as distinct from subcutaneous adiposity or fat between the skin and the muscle wall, especially elsewhere on the body, such as hips or thighs), hypertension, hyperglycemia, and dyslipidemia involving elevated triglycerides, small dense low-density lipoprotein (sdLDL) particles, and decreased HDL cholesterol levels. With respect to visceral adiposity, a great deal of evidence suggests two strong links with insulin resistance. First, unlike subcutaneous adipose tissue, visceral adipose cells produce significant amounts of proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-a), and Interleukins-1 and −6, etc. In numerous experimental models, these proinflammatory cytokines disrupt normal insulin action in fat and muscle cells, and may be a major factor in causing the whole-body insulin resistance observed in patients with visceral adiposity. Much of the attention on production of proinflammatory cytokines has focused on the IKK-beta/NF-kappa-B pathway, a protein network that enhances transcription of inflammatory markers and mediators that may cause insulin resistance. Second, visceral adiposity is related to an accumulation of fat in the liver, a condition known as non-alcoholic fatty liver disease (NAFLD). The result of NAFLD is an excessive release of free fatty acids into the bloodstream (due to increased lipolysis), and an increase in hepatic glycogenolysis and hepatic glucose production, both of which have the effect of exacerbating peripheral insulin resistance and increasing the likelihood of Type 2 diabetes mellitus.[citation needed]


Also, insulin resistance often is associated with a hypercoagulable state (impaired fibrinolysis) and increased inflammatory cytokine levels.[67]


Another factor that may promote insulin resistance is Leptin, a hormone produced from the ob gene and adipocytes.[68] Its physiological role is to regulate hunger by alerting the body when it is full.[69] From an adaptation perspective, it is possible that leptin resistance was favorable for survival during periods when food was scarce.[69] Today however studies show that lack of leptin causes severe obesity and is strongly linked with insulin resistance.[70] Leptin replacement in mice with obesity and diabetes has been found to quickly decrease glucose and insulin levels and can affect insulin sensitivity.[71] Further research into the molecular mechanisms of these effects would be beneficial for increased understanding.[71]



Diagnosis



Fasting insulin levels


A fasting serum insulin level greater than 25 mU/L or 174 pmol/L is considered insulin resistance. The same levels apply three hours after the last meal.[72]



Glucose tolerance testing


During a glucose tolerance test (GTT), which may be used to diagnose diabetes mellitus, a fasting patient takes a 75 gram oral dose of glucose. Then blood glucose levels are measured over the following two hours.


Interpretation is based on WHO guidelines. After two hours a glycemia less than 7.8 mmol/L (140 mg/dL) is considered normal, a glycemia of between 7.8 and 11.0 mmol/L (140 to 197 mg/dL) is considered as impaired glucose tolerance (IGT), and a glycemia of greater than or equal to 11.1 mmol/L (200 mg/dL) is considered diabetes mellitus.


An oral glucose tolerance test (OGTT) may be normal or mildly abnormal in simple insulin resistance. Often, there are raised glucose levels in the early measurements, reflecting the loss of a postprandial peak (after the meal) in insulin production. Extension of the testing (for several more hours) may reveal a hypoglycemic "dip," that is a result of an overshoot in insulin production after the failure of the physiologic postprandial insulin response.[citation needed]



Measuring insulin resistance



Hyperinsulinemic euglycemic clamp


The gold standard for investigating and quantifying insulin resistance is the "hyperinsulinemic euglycemic clamp," so-called because it measures the amount of glucose necessary to compensate for an increased insulin level without causing hypoglycemia.[73] It is a type of glucose clamp technique. The test is rarely performed in clinical care, but is used in medical research, for example, to assess the effects of different medications. The rate of glucose infusion commonly is referred to in diabetes literature as the GINF value.[74]


The procedure takes about two hours. Through a peripheral vein, insulin is infused at 10–120 mU per m2 per minute. In order to compensate for the insulin infusion, glucose 20% is infused to maintain blood sugar levels between 5 and 5.5 mmol/L. The rate of glucose infusion is determined by checking the blood sugar levels every five to ten minutes.[74]


The rate of glucose infusion during the last thirty minutes of the test determines insulin sensitivity. If high levels (7.5 mg/min or higher) are required, the patient is insulin-sensitive. Very low levels (4.0 mg/min or lower) indicate that the body is resistant to insulin action. Levels between 4.0 and 7.5 mg/min are not definitive, and suggest "impaired glucose tolerance," an early sign of insulin resistance.[74]


This basic technique may be enhanced significantly by the use of glucose tracers. Glucose may be labeled with either stable or radioactive atoms. Commonly used tracers are 3-3H glucose (radioactive), 6,6 2H-glucose (stable) and 1-13C Glucose (stable). Prior to beginning the hyperinsulinemic period, a 3h tracer infusion enables one to determine the basal rate of glucose production. During the clamp, the plasma tracer concentrations enable the calculation of whole-body insulin-stimulated glucose metabolism, as well as the production of glucose by the body (i.e., endogenous glucose production).[74]



Modified insulin suppression test


Another measure of insulin resistance is the modified insulin suppression test developed by Gerald Reaven at Stanford University. The test correlates well with the euglycemic clamp, with less operator-dependent error. This test has been used to advance the large body of research relating to the metabolic syndrome.[74]


Patients initially receive 25 μg of octreotide (Sandostatin) in 5 mL of normal saline over 3 to 5 minutes via intravenous infusion (IV) as an initial bolus, and then, are infused continuously with an intravenous infusion of somatostatin (0.27 μg/m2/min) to suppress endogenous insulin and glucose secretion. Next, insulin and 20% glucose are infused at rates of 32 and 267 mg/m2/min, respectively. Blood glucose is checked at zero, 30, 60, 90, and 120 minutes, and thereafter, every 10 minutes for the last half-hour of the test. These last four values are averaged to determine the steady-state plasma glucose level (SSPG). Subjects with an SSPG greater than 150 mg/dL are considered to be insulin-resistant.[74]



Alternatives


Given the complicated nature of the "clamp" technique (and the potential dangers of hypoglycemia in some patients), alternatives have been sought to simplify the measurement of insulin resistance. The first was the Homeostatic Model Assessment (HOMA), and a more recent method is the Quantitative insulin sensitivity check index (QUICKI). Both employ fasting insulin and glucose levels to calculate insulin resistance, and both correlate reasonably with the results of clamping studies. Wallace et al. point out that QUICKI is the logarithm of the value from one of the HOMA equations.[75]



Management


The primary treatment for insulin resistance is exercise and weight loss. Research shows that a low-carbohydrate diet may help.[76] Both metformin and thiazolidinediones improve insulin resistance, but are approved therapies only for type 2 diabetes, not for insulin resistance. By contrast, growth hormone replacement therapy may be associated with increased insulin resistance.[77]


Metformin has become one of the more commonly prescribed medications for insulin resistance.[78] Unfortunately, Metformin also masks Vitamin B12 deficiency, so accompanying sub-lingual Vitamin B12 tablets are recommended.


Insulin resistance is often associated with abnormalities in lipids particularly high blood triglycerides and low high density lipoprotein.[26][27]


The Diabetes Prevention Program (DPP) showed that exercise and diet were nearly twice as effective as metformin at reducing the risk of progressing to type 2 diabetes.[79] However, the participants in the DPP trial regained about 40% of the weight that they had lost at the end of 2.8 years, resulting in a similar incidence of diabetes development in both the lifestyle intervention and the control arms of the trial.[80] One 2009 study found that carbohydrate deficit after exercise, but not energy deficit, contributed to insulin sensitivity increase.[81]


Resistant starch from high-amylose corn, amylomaize, has been shown to reduce insulin resistance in healthy individuals, in individuals with insulin resistance, and in individuals with type 2 diabetes.[82] Animal studies demonstrate that it cannot reverse damage already done by high glucose levels, but that it reduces insulin resistance and reduces the development of further damage.[83][84][85]


Some types of polyunsaturated fatty acids (omega-3) may moderate the progression of insulin resistance into type 2 diabetes,[86][87][88] however, omega-3 fatty acids appear to have limited ability to reverse insulin resistance, and they cease to be efficacious once type 2 diabetes is established.[89]


Caffeine intake limits insulin action, but not enough to increase blood-sugar levels in healthy persons. People who already have type 2 diabetes may see a small increase in levels if they take 2 or 2-1/2 cups of coffee per day.[90]



History


The concept that insulin resistance may be the underlying cause of diabetes mellitus type 2 was first advanced by Professor Wilhelm Falta and published in Vienna in 1931,[91] and confirmed as contributory by Sir Harold Percival Himsworth of the University College Hospital Medical Centre in London in 1936,[92] however, type 2 diabetes does not occur unless there is concurrent failure of compensatory insulin secretion.[93]



Adaptive Explanations


There is some prevailing thought that insulin resistance can be an evolutionary adaptation. In 1962 James Neel proposed his Thrifty Gene Hypothesis.  


This hypothesis raises the point that if there is a genetic component to insulin resistance and Type 2 Diabetes, these phenotypes should be selected against.[94] Yet, there has been an increase in mean insulin resistance in both the normoglycemic population as well as the diabetic population.[95]


Neel Postulates that originally in times of increased famine in ancient humans ancestors, that genes conferring a mechanism for increased glucose storage would be advantageous. In the modern environment today however this is not the case.[94]


Evidence is contradictory to Neel in studies of the Pima Indians, which indicate that the people with higher insulin sensitives tended to weigh the most and conversely people with insulin resistance tended to weigh less on average in this demographic.[96]


Modern hypotheses suggest that insulin metabolism is a socio-ecological adaptation with insulin being the means for differentiating energy allocation to various components of the body and insulin sensitivity an adaptation to manipulate where the energy is diverted to. The Behavioral Switch Hypothesis posits that insulin resistance results in two methods to alter reproductive strategies and behavioral methods. The two strategies are coined as “r to K” and “soldier to diplomat.” The r to K strategy involves diverting insulin via placenta to the fetus. This has demonstrated weight gain in the fetus, but not the mother indicating a method of increased parental investment (K strategy). In the “soldier to diplomat” the insensitivity of skeletal muscle to insulin could divert the glucose to the brain, which doesn’t require insulin receptors. This has shown increased in cognitive development across various studies.[97]



See also




  • Chronic Somogyi rebound

  • Hyperinsulinemia

  • Resistin

  • Chronic stress

  • Systemic inflammation

  • Circadian rhythm disruption

  • Advanced glycation end-products

  • PCOS




References





  1. ^ abcd Wang G (December 2014). "Raison d'être of insulin resistance: the adjustable threshold hypothesis". Journal of the Royal Society, Interface. 11 (101): 20140892. doi:10.1098/rsif.2014.0892. PMC 4223910. PMID 25320065..mw-parser-output cite.citation{font-style:inherit}.mw-parser-output .citation q{quotes:"""""""'""'"}.mw-parser-output .citation .cs1-lock-free a{background:url("//upload.wikimedia.org/wikipedia/commons/thumb/6/65/Lock-green.svg/9px-Lock-green.svg.png")no-repeat;background-position:right .1em center}.mw-parser-output .citation .cs1-lock-limited a,.mw-parser-output .citation .cs1-lock-registration a{background:url("//upload.wikimedia.org/wikipedia/commons/thumb/d/d6/Lock-gray-alt-2.svg/9px-Lock-gray-alt-2.svg.png")no-repeat;background-position:right .1em center}.mw-parser-output .citation .cs1-lock-subscription a{background:url("//upload.wikimedia.org/wikipedia/commons/thumb/a/aa/Lock-red-alt-2.svg/9px-Lock-red-alt-2.svg.png")no-repeat;background-position:right .1em center}.mw-parser-output .cs1-subscription,.mw-parser-output .cs1-registration{color:#555}.mw-parser-output .cs1-subscription span,.mw-parser-output .cs1-registration span{border-bottom:1px dotted;cursor:help}.mw-parser-output .cs1-ws-icon a{background:url("//upload.wikimedia.org/wikipedia/commons/thumb/4/4c/Wikisource-logo.svg/12px-Wikisource-logo.svg.png")no-repeat;background-position:right .1em center}.mw-parser-output code.cs1-code{color:inherit;background:inherit;border:inherit;padding:inherit}.mw-parser-output .cs1-hidden-error{display:none;font-size:100%}.mw-parser-output .cs1-visible-error{font-size:100%}.mw-parser-output .cs1-maint{display:none;color:#33aa33;margin-left:0.3em}.mw-parser-output .cs1-subscription,.mw-parser-output .cs1-registration,.mw-parser-output .cs1-format{font-size:95%}.mw-parser-output .cs1-kern-left,.mw-parser-output .cs1-kern-wl-left{padding-left:0.2em}.mw-parser-output .cs1-kern-right,.mw-parser-output .cs1-kern-wl-right{padding-right:0.2em}


  2. ^ Diagnosis and Mellitus. (2013). Diabetes Care, 37(Supplement_1), S81–90. https://dx.doi.org/10.2337/dc14-s081


  3. ^ ab Basciano H, Federico L, Adeli K (February 2005). "Fructose, insulin resistance, and metabolic dyslipidemia". Nutrition & Metabolism. 2 (1): 5. doi:10.1186/1743-7075-2-5. PMC 552336. PMID 15723702.


  4. ^ Frank Hu Associate Professor of Nutrition and Epidemiology Harvard School of Public Health (20 February 2008). Obesity Epidemiology. Oxford University Press. pp. 283–285. ISBN 978-0-19-971847-4. Retrieved 20 April 2013.


  5. ^ Travis A. Smith (November 2010). Taxing Caloric Sweetened Beverages: Potential Effects on Beverage Consumption, Calorie Intake, and Obesity. DIANE Publishing. pp. 13–14. ISBN 978-1-4379-3593-6. Retrieved 20 April 2013.


  6. ^ Gallagher, James (24 February 2017). "Fasting diet 'regenerates diabetic pancreas'" (Health and science reporter, BBC News website). BBC. Retrieved 6 August 2018.


  7. ^ Sakzewski, Emily (27 Feb 2017). "Fasting diet could regenerate pancreas and reverse diabetes, researchers say". ABC. Retrieved 6 August 2018.


  8. ^ Chiu HK, Tsai EC, Juneja R, Stoever J, Brooks-Worrell B, Goel A, Palmer JP (August 2007). "Equivalent insulin resistance in latent autoimmune diabetes in adults (LADA) and type 2 diabetic patients". Diabetes Research and Clinical Practice. 77 (2): 237–44. doi:10.1016/j.diabres.2006.12.013. PMID 17234296.


  9. ^ Thorell, Anders; Nygren, Jonas; Ljungqvist, Olle (January 1999). "Insulin resistance: a marker of surgical stress : Current Opinion in Clinical Nutrition & Metabolic Care". LWW. 2 (1): 69.


  10. ^ Thorell A, Efendic S, Gutniak M, Häggmark T, Ljungqvist O (January 1994). "Insulin resistance after abdominal surgery". The British Journal of Surgery. 81 (1): 59–63. doi:10.1002/bjs.1800810120. PMID 8313123.


  11. ^ Cao H, Hegele RA (January 2000). "Nuclear lamin A/C R482Q mutation in canadian kindreds with Dunnigan-type familial partial lipodystrophy". Human Molecular Genetics. 9 (1): 109–12. doi:10.1093/hmg/9.1.109. PMID 10587585.


  12. ^ Musso C, Cochran E, Moran SA, Skarulis MC, Oral EA, Taylor S, Gorden P (July 2004). "Clinical course of genetic diseases of the insulin receptor (type A and Rabson-Mendenhall syndromes): a 30-year prospective". Medicine. 83 (4): 209–22. doi:10.1097/01.md.0000133625.73570.54. PMID 15232309.


  13. ^ abcdefghij Mayfield J (October 1998). "Diagnosis and classification of diabetes mellitus: new criteria". American Family Physician. 58 (6): 1355–62, 1369–70. PMID 9803200.


  14. ^ abcdefgh "Type 2 diabetes: Risk factors". Mayo Clinic. Retrieved 21 December 2011.


  15. ^ abcd Milner KL, van der Poorten D, Trenell M, Jenkins AB, Xu A, Smythe G, Dore GJ, Zekry A, Weltman M, Fragomeli V, George J, Chisholm DJ (March 2010). "Chronic hepatitis C is associated with peripheral rather than hepatic insulin resistance". Gastroenterology. 138 (3): 932–41.e1–3. doi:10.1053/j.gastro.2009.11.050. PMID 19962985. Lay summary – Science Daily (March 10, 2010).


  16. ^ Yasuda K, Hines E, Kitabchi AE (November 1982). "Hypercortisolism and insulin resistance: comparative effects of prednisone, hydrocortisone, and dexamethasone on insulin binding of human erythrocytes". The Journal of Clinical Endocrinology and Metabolism. 55 (5): 910–5. doi:10.1210/jcem-55-5-910. PMID 6749880.


  17. ^ ab Wang G (December 2010). "Singularity analysis of the AKT signaling pathway reveals connections between cancer and metabolic diseases". Physical Biology. 7 (4): 046015. Bibcode:2010PhBio...7d6015W. doi:10.1088/1478-3975/7/4/046015. PMID 21178243.


  18. ^ Wang G (October 2012). "Optimal homeostasis necessitates bistable control". Journal of the Royal Society, Interface. 9 (75): 2723–34. doi:10.1098/rsif.2012.0244. PMC 3427521. PMID 22535698.


  19. ^ Lucidi P, Rossetti P, Porcellati F, Pampanelli S, Candeloro P, Andreoli AM, Perriello G, Bolli GB, Fanelli CG (June 2010). "Mechanisms of insulin resistance after insulin-induced hypoglycemia in humans: the role of lipolysis". Diabetes. 59 (6): 1349–57. doi:10.2337/db09-0745. PMC 2874695. PMID 20299466.


  20. ^ Wang G, Zhang M (February 2016). "Tunable ultrasensitivity: functional decoupling and biological insights". Scientific Reports. 6: 20345. Bibcode:2016NatSR...620345W. doi:10.1038/srep20345. PMC 4742884. PMID 26847155.


  21. ^ Stryer, Lubert (1995). Biochemistry (Fourth ed.). New York: W.H. Freeman and Company. pp. 773–774. ISBN 0 7167 2009 4.


  22. ^ ab Isganaitis E, Lustig RH (December 2005). "Fast food, central nervous system insulin resistance, and obesity". Arteriosclerosis, Thrombosis, and Vascular Biology. 25 (12): 2451–62. doi:10.1161/01.ATV.0000186208.06964.91. PMID 16166564.


  23. ^ Kahn SE, Hull RL, Utzschneider KM (December 2006). "Mechanisms linking obesity to insulin resistance and type 2 diabetes". Nature. 444 (7121): 840–6. doi:10.1038/nature05482. PMID 17167471.


  24. ^ Unger RH, Scherer PE (June 2010). "Gluttony, sloth and the metabolic syndrome: a roadmap to lipotoxicity". Trends in Endocrinology and Metabolism. 21 (6): 345–52. doi:10.1016/j.tem.2010.01.009. PMC 2880185. PMID 20223680.


  25. ^ Chiu KC, Chu A, Go VL, Saad MF (May 2004). "Hypovitaminosis D is associated with insulin resistance and beta cell dysfunction". The American Journal of Clinical Nutrition. 79 (5): 820–5. doi:10.1093/ajcn/79.5.820. PMID 15113720.


  26. ^ abc Storlien LH, Jenkins AB, Chisholm DJ, Pascoe WS, Khouri S, Kraegen EW (February 1991). "Influence of dietary fat composition on development of insulin resistance in rats. Relationship to muscle triglyceride and omega-3 fatty acids in muscle phospholipid". Diabetes. 40 (2): 280–9. doi:10.2337/diabetes.40.2.280. PMID 1991575.


  27. ^ abc Schinner S, Scherbaum WA, Bornstein SR, Barthel A (June 2005). "Molecular mechanisms of insulin resistance". Diabetic Medicine. 22 (6): 674–82. doi:10.1111/j.1464-5491.2005.01566.x. PMID 15910615.


  28. ^ Koyama K, Chen G, Lee Y, Unger RH (October 1997). "Tissue triglycerides, insulin resistance, and insulin production: implications for hyperinsulinemia of obesity". The American Journal of Physiology. 273 (4 Pt 1): E708–13. PMID 9357799.


  29. ^ Roden M, Price TB, Perseghin G, Petersen KF, Rothman DL, Cline GW, Shulman GI (June 1996). "Mechanism of free fatty acid-induced insulin resistance in humans". The Journal of Clinical Investigation. 97 (12): 2859–65. doi:10.1172/JCI118742. PMC 507380. PMID 8675698.


  30. ^ Kraegen EW, Clark PW, Jenkins AB, Daley EA, Chisholm DJ, Storlien LH (November 1991). "Development of muscle insulin resistance after liver insulin resistance in high-fat-fed rats". Diabetes. 40 (11): 1397–403. doi:10.2337/diabetes.40.11.1397. PMID 1936601.


  31. ^ Ivy JL (November 1997). "Role of exercise training in the prevention and treatment of insulin resistance and non-insulin-dependent diabetes mellitus". Sports Medicine. Auckland, NZ. 24 (5): 321–36. doi:10.2165/00007256-199724050-00004. PMID 9368278.


  32. ^ Mayer-Davis EJ, D'Agostino R, Karter AJ, Haffner SM, Rewers MJ, Saad M, Bergman RN (March 1998). "Intensity and amount of physical activity in relation to insulin sensitivity: the Insulin Resistance Atherosclerosis Study". JAMA. 279 (9): 669–74. doi:10.1001/jama.279.9.669. PMID 9496984.


  33. ^ Helmrich SP, Ragland DR, Leung RW, Paffenbarger RS (July 1991). "Physical activity and reduced occurrence of non-insulin-dependent diabetes mellitus". The New England Journal of Medicine. 325 (3): 147–52. doi:10.1056/NEJM199107183250302. PMID 2052059.


  34. ^ Manson JE, Rimm EB, Stampfer MJ, Colditz GA, Willett WC, Krolewski AS, Rosner B, Hennekens CH, Speizer FE (September 1991). "Physical activity and incidence of non-insulin-dependent diabetes mellitus in women". Lancet. 338 (8770): 774–8. doi:10.1016/0140-6736(91)90664-B. PMID 1681160.


  35. ^ Fantry, Lori E (2003-03-24). "Protease Inhibitor-Associated Diabetes Mellitus: A Potential Cause of Morbidity and Mortality". Journal of Acquired Immune Deficiency Syndromes. 32 (3){{inconsistent citations}}.


  36. ^ Haag M, Dippenaar NG (December 2005). "Dietary fats, fatty acids and insulin resistance: short review of a multifaceted connection" (PDF). Medical Science Monitor. 11 (12): RA359–67. PMID 16319806. Archived from the original (PDF) on 2011-10-08.


  37. ^ Morino K, Petersen KF, Shulman GI (December 2006). "Molecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction". Diabetes. 55 Suppl 2 (Suppl 2): S9–S15. doi:10.2337/db06-S002. PMC 2995546. PMID 17130651.


  38. ^ Bachmann OP, Dahl DB, Brechtel K, Machann J, Haap M, Maier T, Loviscach M, Stumvoll M, Claussen CD, Schick F, Häring HU, Jacob S (November 2001). "Effects of intravenous and dietary lipid challenge on intramyocellular lipid content and the relation with insulin sensitivity in humans". Diabetes. 50 (11): 2579–84. doi:10.2337/diabetes.50.11.2579. PMID 11679437.


  39. ^ Sato F, Tamura Y, Watada H, Kumashiro N, Igarashi Y, Uchino H, Maehara T, Kyogoku S, Sunayama S, Sato H, Hirose T, Tanaka Y, Kawamori R (August 2007). "Effects of diet-induced moderate weight reduction on intrahepatic and intramyocellular triglycerides and glucose metabolism in obese subjects". The Journal of Clinical Endocrinology and Metabolism. 92 (8): 3326–9. doi:10.1210/jc.2006-2384. PMID 17519317.


  40. ^ Tamura Y, Tanaka Y, Sato F, Choi JB, Watada H, Niwa M, Kinoshita J, Ooka A, Kumashiro N, Igarashi Y, Kyogoku S, Maehara T, Kawasumi M, Hirose T, Kawamori R (June 2005). "Effects of diet and exercise on muscle and liver intracellular lipid contents and insulin sensitivity in type 2 diabetic patients". The Journal of Clinical Endocrinology and Metabolism. 90 (6): 3191–6. doi:10.1210/jc.2004-1959. PMID 15769987.


  41. ^ Haugaard SB, Madsbad S, Høy CE, Vaag A (February 2006). "Dietary intervention increases n-3 long-chain polyunsaturated fatty acids in skeletal muscle membrane phospholipids of obese subjects. Implications for insulin sensitivity". Clinical Endocrinology. 64 (2): 169–78. doi:10.1111/j.1365-2265.2006.02444.x. PMID 16430716.


  42. ^ Russo GL (March 2009). "Dietary n-6 and n-3 polyunsaturated fatty acids: from biochemistry to clinical implications in cardiovascular prevention". Biochemical Pharmacology. 77 (6): 937–46. doi:10.1016/j.bcp.2008.10.020. PMID 19022225.


  43. ^ ab Brown, Dave D (2003). USMLE Step 1 Secrets. p. 63.


  44. ^ King, Michael W (2005). Lange Q&A USMLE Step 1 (6th ed.). New York: McGraw-Hill Medical. p. 82. ISBN 978-0-07-144578-8.


  45. ^ Piroli GG, Grillo CA, Reznikov LR, Adams S, McEwen BS, Charron MJ, Reagan LP (2007). "Corticosterone impairs insulin-stimulated translocation of GLUT4 in the rat hippocampus". Neuroendocrinology. 85 (2): 71–80. doi:10.1159/000101694. PMID 17426391.


  46. ^ Solinas G, Vilcu C, Neels JG, Bandyopadhyay GK, Luo JL, Naugler W, Grivennikov S, Wynshaw-Boris A, Scadeng M, Olefsky JM, Karin M (November 2007). "JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity". Cell Metabolism. 6 (5): 386–97. doi:10.1016/j.cmet.2007.09.011. PMID 17983584.


  47. ^ "UCSD researchers discover inflammation, not obesity, cause of insulin resistance". D life. Archived from the original on 2007-11-21. Retrieved 2008-01-12.


  48. ^ |Kant S, Barrett T, Vertii A, Noh YH, Jung DY, Kim JK, Davis RJ (August 2013). "Role of the mixed-lineage protein kinase pathway in the metabolic stress response to obesity". Cell Reports. 4 (4): 681–8. doi:10.1016/j.celrep.2013.07.019. PMC 3769115. PMID 23954791.


  49. ^ Hoehn KL, Salmon AB, Hohnen-Behrens C, Turner N, Hoy AJ, Maghzal GJ, Stocker R, Van Remmen H, Kraegen EW, Cooney GJ, Richardson AR, James DE (October 2009). "Insulin resistance is a cellular antioxidant defense mechanism". Proceedings of the National Academy of Sciences of the United States of America. 106 (42): 17787–92. Bibcode:2009PNAS..10617787H. doi:10.1073/pnas.0902380106. PMC 2764908. PMID 19805130.


  50. ^ Garrido-Sanchez L, Murri M, Rivas-Becerra J, Ocaña-Wilhelmi L, Cohen RV, Garcia-Fuentes E, Tinahones FJ (2012). "Bypass of the duodenum improves insulin resistance much more rapidly than sleeve gastrectomy". Surgery for Obesity and Related Diseases. 8 (2): 145–50. doi:10.1016/j.soard.2011.03.010. PMID 21570362.


  51. ^ Goodman, Alice (June 1, 2016). "Duodenal resurfacing achieves metabolic benefits in type 2 diabetes". Family Practice News. Retrieved 12 March 2017.


  52. ^ Nafiye Y, Sevtap K, Muammer D, Emre O, Senol K, Leyla M (April 2010). "The effect of serum and intrafollicular insulin resistance parameters and homocysteine levels of nonobese, nonhyperandrogenemic polycystic ovary syndrome patients on in vitro fertilization outcome". Fertility and Sterility. 93 (6): 1864–9. doi:10.1016/j.fertnstert.2008.12.024. PMID 19171332.


  53. ^ Brown AE, Walker M (August 2016). "Genetics of Insulin Resistance and the Metabolic Syndrome". Current Cardiology Reports. 18 (8): 75. doi:10.1007/s11886-016-0755-4. PMC 4911377. PMID 27312935.


  54. ^ "A heavy burden". The Economist. December 15, 2012. Retrieved 10 January 2013.


  55. ^ ab "Insulin resistance". Medicine net.


  56. ^ "Science daily". Jun 2009.


  57. ^ Insulin Resistance Leads to LADA (Report). Diabetes Health. Retrieved Feb 21, 2015.


  58. ^ Behme MT, Dupre J, Harris SB, Hramiak IM, Mahon JL (November 2003). "Insulin resistance in latent autoimmune diabetes of adulthood". Annals of the New York Academy of Sciences. 1005 (1): 374–7. Bibcode:2003NYASA1005..374B. doi:10.1196/annals.1288.062. PMID 14679095.


  59. ^ "Latent Autoimmune Diabetes: A Little Known Type of Diabetes". Pharmacy Times. Retrieved May 30, 2014.


  60. ^ McGarry JD (January 2002). "Banting lecture 2001: dysregulation of fatty acid metabolism in the etiology of type 2 diabetes". Diabetes. 51 (1): 7–18. doi:10.2337/diabetes.51.1.7. PMID 11756317.


  61. ^ Peraldi P, Spiegelman B (May 1998). "TNF-alpha and insulin resistance: summary and future prospects". Molecular and Cellular Biochemistry. 182 (1–2): 169–75. doi:10.1023/A:1006865715292. PMID 9609126.


  62. ^ Hotamisligil GS (June 1999). "The role of TNFalpha and TNF receptors in obesity and insulin resistance". Journal of Internal Medicine. 245 (6): 621–5. doi:10.1046/j.1365-2796.1999.00490.x. PMID 10395191.


  63. ^ Flores-Riveros JR, McLenithan JC, Ezaki O, Lane MD (January 1993). "Insulin down-regulates expression of the insulin-responsive glucose transporter (GLUT4) gene: effects on transcription and mRNA turnover". Proceedings of the National Academy of Sciences of the United States of America. 90 (2): 512–6. Bibcode:1993PNAS...90..512F. doi:10.1073/pnas.90.2.512. PMC 45693. PMID 8421683.


  64. ^ MacLean PS, Zheng D, Jones JP, Olson AL, Dohm GL (March 2002). "Exercise-induced transcription of the muscle glucose transporter (GLUT 4) gene". Biochemical and Biophysical Research Communications. 292 (2): 409–14. doi:10.1006/bbrc.2002.6654. PMID 11906177.


  65. ^ Koga M, Kasayama S (2010). "Clinical impact of glycated albumin as another glycemic control marker". Endocrine Journal. 57 (9): 751–62. doi:10.1507/endocrj.K10E-138. PMID 20724796.


  66. ^ Puddu A, Viviani GL (June 2011). "Advanced glycation endproducts and diabetes. Beyond vascular complications". Endocrine, Metabolic & Immune Disorders Drug Targets. 11 (2): 132–40. doi:10.2174/187153011795564115. PMID 21476962.


  67. ^ Nagaev I, Bokarewa M, Tarkowski A, Smith U (December 2006). Valcarcel J, ed. "Human resistin is a systemic immune-derived proinflammatory cytokine targeting both leukocytes and adipocytes". PLOS One. 1 (1): e31. Bibcode:2006PLoSO...1...31N. doi:10.1371/journal.pone.0000031. PMC 1762367. PMID 17183659.


  68. ^ Friedman JM (April 2000). "Obesity in the new millennium". Nature. 404 (6778): 632–4. doi:10.1038/35007504. PMID 10766249.


  69. ^ ab Flier JS (May 1998). "Clinical review 94: What's in a name? In search of leptin's physiologic role". The Journal of Clinical Endocrinology and Metabolism. 83 (5): 1407–13. doi:10.1210/jcem.83.5.4779. PMID 9589630.


  70. ^ Elmquist JK, Maratos-Flier E, Saper CB, Flier JS (October 1998). "Unraveling the central nervous system pathways underlying responses to leptin". Nature Neuroscience. 1 (6): 445–50. doi:10.1038/2164. PMID 10196541.


  71. ^ ab Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, Lallone RL, Burley SK, Friedman JM (July 1995). "Weight-reducing effects of the plasma protein encoded by the obese gene". Science. 269 (5223): 543–6. doi:10.1126/science.7624777. PMID 7624777.


  72. ^ Insulin at eMedicine


  73. ^ DeFronzo RA, Tobin JD, Andres R (September 1979). "Glucose clamp technique: a method for quantifying insulin secretion and resistance". The American Journal of Physiology. 237 (3): E214–23. doi:10.1152/ajpendo.1979.237.3.e214. PMID 382871.


  74. ^ abcdef Muniyappa R, Lee S, Chen H, Quon MJ (January 2008). "Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usage". American Journal of Physiology. Endocrinology and Metabolism. 294 (1): E15–26. doi:10.1152/ajpendo.00645.2007. PMID 17957034.


  75. ^ Wallace TM, Levy JC, Matthews DR (June 2004). "Use and abuse of HOMA modeling". Diabetes Care. 27 (6): 1487–95. doi:10.2337/diacare.27.6.1487. PMID 15161807.


  76. ^ Boden G, Sargrad K, Homko C, Mozzoli M, Stein TP (March 2005). "Effect of a low-carbohydrate diet on appetite, blood glucose levels, and insulin resistance in obese patients with type 2 diabetes". Annals of Internal Medicine. 142 (6): 403–11. doi:10.7326/0003-4819-142-6-200503150-00006. PMID 15767618.


  77. ^ Bramnert M, Segerlantz M, Laurila E, Daugaard JR, Manhem P, Groop L (April 2003). "Growth hormone replacement therapy induces insulin resistance by activating the glucose-fatty acid cycle". The Journal of Clinical Endocrinology and Metabolism. 88 (4): 1455–63. doi:10.1210/jc.2002-020542. PMID 12679422.


  78. ^ Giannarelli R, Aragona M, Coppelli A, Del Prato S (September 2003). "Reducing insulin resistance with metformin: the evidence today". Diabetes & Metabolism. 29 (4 Pt 2): 6S28–35. doi:10.1016/s1262-3636(03)72785-2. PMID 14502098.


  79. ^ Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, Nathan DM (February 2002). "Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin". The New England Journal of Medicine. 346 (6): 393–403. doi:10.1056/NEJMoa012512. PMC 1370926. PMID 11832527.


  80. ^ Kahn R (January 2012). "Reducing the impact of diabetes: is prevention feasible today, or should we aim for better treatment?". Health Affairs. 31 (1): 76–83. doi:10.1377/hlthaff.2011.1075. PMID 22232097.


  81. ^ Newsom SA, Schenk S, Thomas KM, Harber MP, Knuth ND, Goldenberg N, Horowitz JF (March 2010). "Energy deficit after exercise augments lipid mobilization but does not contribute to the exercise-induced increase in insulin sensitivity". Journal of Applied Physiology. 108 (3): 554–60. doi:10.1152/japplphysiol.01106.2009. PMC 2838634. PMID 20044472.


  82. ^ Keenan MJ, Zhou J, Hegsted M, Pelkman C, Durham HA, Coulon DB, Martin RJ (March 2015). "Role of resistant starch in improving gut health, adiposity, and insulin resistance". Advances in Nutrition. 6 (2): 198–205. doi:10.3945/an.114.007419. PMC 4352178. PMID 25770258.


  83. ^ Higgins JA, Brand Miller JC, Denyer GS (March 1996). "Development of insulin resistance in the rat is dependent on the rate of glucose absorption from the diet". The Journal of Nutrition. 126 (3): 596–602. doi:10.1093/jn/126.3.596. PMID 8598543.


  84. ^
    Byrnes SE, Miller JC, Denyer GS (June 1995). "Amylopectin starch promotes the development of insulin resistance in rats". The Journal of Nutrition. 125 (6): 1430–7. doi:10.1093/jn/125.6.1430 (inactive 2019-02-13). PMID 7782895.



  85. ^
    Wiseman CE, Higgins JA, Denyer GS, Miller JC (February 1996). "Amylopectin starch induces nonreversible insulin resistance in rats". The Journal of Nutrition. 126 (2): 410–5. doi:10.1093/jn/126.2.410. PMID 8632213.



  86. ^ Lovejoy JC (October 2002). "The influence of dietary fat on insulin resistance". Current Diabetes Reports. 2 (5): 435–40. doi:10.1007/s11892-002-0098-y. PMID 12643169.


  87. ^
    Fukuchi S, Hamaguchi K, Seike M, Himeno K, Sakata T, Yoshimatsu H (June 2004). "Role of fatty acid composition in the development of metabolic disorders in sucrose-induced obese rats". Experimental Biology and Medicine. 229 (6): 486–93. doi:10.1177/153537020422900606. PMID 15169967.



  88. ^
    Storlien LH, Baur LA, Kriketos AD, Pan DA, Cooney GJ, Jenkins AB, Calvert GD, Campbell LV (June 1996). "Dietary fats and insulin action". Diabetologia. 39 (6): 621–31. doi:10.1007/BF00418533. PMID 8781757.



  89. ^
    Delarue J, LeFoll C, Corporeau C, Lucas D (2004). "N-3 long chain polyunsaturated fatty acids: a nutritional tool to prevent insulin resistance associated to type 2 diabetes and obesity?". Reproduction, Nutrition, Development. 44 (3): 289–99. doi:10.1051/rnd:2004033. PMID 15460168.



  90. ^ "Does caffeine affect blood sugar?". Mayo Clinic{{inconsistent citations}}


  91. ^ Falta, W.; Boller, R. (1931). "Insulärer und Insulinresistenter Diabetes". Klinische Wochenschrift. 10 (10): 438–43. doi:10.1007/BF01736348.


  92. ^ Himsworth, H (1936). "Diabetes mellitus: its differentiation into insulin-sensitive and insulin insensitive types". The Lancet. 227 (5864): 127–30. doi:10.1016/S0140-6736(01)36134-2.


  93. ^ Nolan CJ (October 2010). "Failure of islet β-cell compensation for insulin resistance causes type 2 diabetes: what causes non-alcoholic fatty liver disease and non-alcoholic steatohepatitis?". Journal of Gastroenterology and Hepatology. 25 (10): 1594–7. doi:10.1111/j.1440-1746.2010.06473.x. PMID 20880166.


  94. ^ ab Neel JV (December 1962). "Diabetes mellitus: a "thrifty" genotype rendered detrimental by "progress"?". American Journal of Human Genetics. 14 (4): 353–62. PMC 1932342. PMID 13937884.


  95. ^ Ioannou GN, Bryson CL, Boyko EJ (2007-11-01). "Prevalence and trends of insulin resistance, impaired fasting glucose, and diabetes". Journal of Diabetes and its Complications. 21 (6): 363–70. doi:10.1016/j.jdiacomp.2006.07.005. PMID 17967708.


  96. ^ Swinburn BA, Nyomba BL, Saad MF, Zurlo F, Raz I, Knowler WC, Lillioja S, Bogardus C, Ravussin E (July 1991). "Insulin resistance associated with lower rates of weight gain in Pima Indians". The Journal of Clinical Investigation. 88 (1): 168–73. doi:10.1172/JCI115274. PMC 296017. PMID 2056116.


  97. ^ Watve MG, Yajnik CS (April 2007). "Evolutionary origins of insulin resistance: a behavioral switch hypothesis". BMC Evolutionary Biology. 7: 61. doi:10.1186/1471-2148-7-61. PMC 1868084. PMID 17437648.




Further reading




  • Reaven GM (2005). "The insulin resistance syndrome: definition and dietary approaches to treatment". Annual Review of Nutrition (review). 25: 391–406. doi:10.1146/annurev.nutr.24.012003.132155. PMID 16011472.


  • Rao G (March 2001). "Insulin resistance syndrome". American Family Physician (review). US. 63 (6): 1159–63, 1165–6. PMID 11277552.



External links











Classification
D
External resources

  • eMedicine: med/1173



  • Insulin resistance at Curlie


  • "The National Insulin Resistance Council". US.


  • "Insulin resistance". Diabetes. US: NIH.










Popular posts from this blog

鏡平學校

ꓛꓣだゔៀៅຸ໢ທຮ໕໒ ,ໂ'໥໓າ໼ឨឲ៵៭ៈゎゔit''䖳𥁄卿' ☨₤₨こゎもょの;ꜹꟚꞖꞵꟅꞛေၦေɯ,ɨɡ𛃵𛁹ޝ޳ޠ޾,ޤޒޯ޾𫝒𫠁သ𛅤チョ'サノބޘދ𛁐ᶿᶇᶀᶋᶠ㨑㽹⻮ꧬ꧹؍۩وَؠ㇕㇃㇪ ㇦㇋㇋ṜẰᵡᴠ 軌ᵕ搜۳ٰޗޮ޷ސޯ𫖾𫅀ल, ꙭ꙰ꚅꙁꚊꞻꝔ꟠Ꝭㄤﺟޱސꧨꧼ꧴ꧯꧽ꧲ꧯ'⽹⽭⾁⿞⼳⽋២៩ញណើꩯꩤ꩸ꩮᶻᶺᶧᶂ𫳲𫪭𬸄𫵰𬖩𬫣𬊉ၲ𛅬㕦䬺𫝌𫝼,,𫟖𫞽ហៅ஫㆔ాఆఅꙒꚞꙍ,Ꙟ꙱エ ,ポテ,フࢰࢯ𫟠𫞶 𫝤𫟠ﺕﹱﻜﻣ𪵕𪭸𪻆𪾩𫔷ġ,ŧآꞪ꟥,ꞔꝻ♚☹⛵𛀌ꬷꭞȄƁƪƬșƦǙǗdžƝǯǧⱦⱰꓕꓢႋ神 ဴ၀க௭எ௫ឫោ ' េㇷㇴㇼ神ㇸㇲㇽㇴㇼㇻㇸ'ㇸㇿㇸㇹㇰㆣꓚꓤ₡₧ ㄨㄟ㄂ㄖㄎ໗ツڒذ₶।ऩछएोञयूटक़कयँृी,冬'𛅢𛅥ㇱㇵㇶ𥄥𦒽𠣧𠊓𧢖𥞘𩔋цѰㄠſtʯʭɿʆʗʍʩɷɛ,əʏダヵㄐㄘR{gỚṖḺờṠṫảḙḭᴮᵏᴘᵀᵷᵕᴜᴏᵾq﮲ﲿﴽﭙ軌ﰬﶚﶧ﫲Ҝжюїкӈㇴffצּ﬘﭅﬈軌'ffistfflſtffतभफɳɰʊɲʎ𛁱𛁖𛁮𛀉 𛂯𛀞నఋŀŲ 𫟲𫠖𫞺ຆຆ ໹້໕໗ๆทԊꧢꧠ꧰ꓱ⿝⼑ŎḬẃẖỐẅ ,ờỰỈỗﮊDžȩꭏꭎꬻ꭮ꬿꭖꭥꭅ㇭神 ⾈ꓵꓑ⺄㄄ㄪㄙㄅㄇstA۵䞽ॶ𫞑𫝄㇉㇇゜軌𩜛𩳠Jﻺ‚Üမ႕ႌႊၐၸဓၞၞၡ៸wyvtᶎᶪᶹစဎ꣡꣰꣢꣤ٗ؋لㇳㇾㇻㇱ㆐㆔,,㆟Ⱶヤマފ޼ޝަݿݞݠݷݐ',ݘ,ݪݙݵ𬝉𬜁𫝨𫞘くせぉて¼óû×ó£…𛅑הㄙくԗԀ5606神45,神796'𪤻𫞧ꓐ㄁ㄘɥɺꓵꓲ3''7034׉ⱦⱠˆ“𫝋ȍ,ꩲ軌꩷ꩶꩧꩫఞ۔فڱێظペサ神ナᴦᵑ47 9238їﻂ䐊䔉㠸﬎ffiﬣ,לּᴷᴦᵛᵽ,ᴨᵤ ᵸᵥᴗᵈꚏꚉꚟ⻆rtǟƴ𬎎

Why https connections are so slow when debugging (stepping over) in Java?