Skip to main content
Erschienen in: BMC Medicine 1/2018

Open Access 01.12.2018 | Review

Insulin translates unfavourable lifestyle into obesity

verfasst von: Hubert Kolb, Michael Stumvoll, Werner Kramer, Kerstin Kempf, Stephan Martin

Erschienen in: BMC Medicine | Ausgabe 1/2018

Abstract

Lifestyle factors conferring increased diabetes risk are associated with elevated basal insulin levels (hyperinsulinaemia). The latter predicts later obesity in children and adolescents.
A causal role of hyperinsulinaemia for adipose tissue growth is probable because pharmacological reduction of insulin secretion lowers body weight in people who are obese. Genetic inactivation of insulin gene alleles in mice also lowers their systemic insulin levels and prevents or ameliorates high-fat diet-induced obesity. Hyperinsulinaemia causes weight gain because of a physiological property of insulin. Insulin levels that are on the high side of normal, or which are slightly elevated, are sufficient to suppress lipolysis and promote lipogenesis in adipocytes. The effect of insulin on glucose transport or hepatic glucose production requires six or two times higher hormone levels, respectively.
It seems justified to suggest a lifestyle that avoids high insulin levels in order to limit anabolic fat tissue activity.
Abkürzungen
BMI
Body mass index
EC50
Mean insulin concentration for 50% effect
FFAs
Free fatty acids
T2DM
Type 2 diabetes mellitus

Background

Lifestyle, systemic inflammation and hyperinsulinaemia

There are now more overweight people in the world than those who are underweight [1]. There has also been a major increase in the global prevalence of type 2 diabetes mellitus (T2DM) [2]. Lifestyle changes are commonly held responsible for these epidemics, with energy-dense western-style diets and little physical activity being major risk factors. However, other lifestyle factors may confer risks of both obesity and T2DM [3]. All factors are associated with moderately elevated systemic levels of pro-inflammatory mediators, increased fasting levels of insulin and decreased insulin sensitivity. Calorie-rich diets caused postprandial inflammation and hyperinsulinaemia [4, 5]. Continuous excess nutrition more than doubled basal insulin levels within 4 days, but did not cause elevated basal glucose levels [6]. Increased physical activity or reallocation of sedentary time to physical activity lowers fasting insulin concentrations and the level of systemic inflammation [7]. Conversely, short-term decreased physical activity, with increased sedentary behaviour, increased whole-body insulin resistance [8]. In an experimental setting, exposure to road traffic-associated fine particulate matter was associated with higher levels of inflammatory markers, insulin and insulin resistance [9]. Sleep deprivation, even for one night, increases systemic insulin resistance [10, 11] and is accompanied by systemic inflammation [12] (Table 1). Although only studied using epidemiological approaches, a positive association has been observed between smoking, depression, stress or low socioeconomic status and inflammation or hyperinsulinaemia/insulin resistance [1318].
Table 1
Association of obesity risk factors with low-grade systemic inflammation and hyperinsulinaemia
Lifestyle factor
Inflammation
Hyperinsulinaemia/ insulin resistance
References
Calorie-rich diets
Yes a
Yes a
[4, 5, 114]
Sedentary time
Yes a
Yes a
[7, 8, 115]
Road traffic
Yes a
Yes a
[9, 116, 117]
Sleep deprivation
Yes a
Yes a
[1012]
Smoking
Yes
Yes
[14, 15]
Depression, stress
Yes
Yes
[13, 16]
Low socioeconomic status
Yes
Yes
[17, 18]
a Randomised controlled trials indicate causal relationship
Largely independent of obesity status, inflammation appears to be a rapid response to an unfavourable lifestyle [19, 20] and may be responsible for metabolic deterioration. For instance, low levels of pro-inflammatory cytokines such as interleukin-1 increased insulin secretion by ß-cells [21]. Pro-inflammatory cytokines interfere with insulin signalling; for example, the induction of IĸB kinase ß, which phosphorylates serine residues of IRS-1 and thereby interferes with insulin signalling [22, 23].
Several other pathways may be involved in promoting obesity by environmental/lifestyle factors; e.g., the hypothalamic–pituitary–adrenal axis, because increased cortisol exposure enhances fat accumulation in visceral depots [24]. Hypothalamic circuits contribute to appetite regulation and energy homeostasis [25]. Recently, glial and endothelial cells have reportedly contributed to metabolic disorders and obesity [26, 27]. Genetic studies confirm the association between neurodevelopmental loci and obesity [28]. Another player is the microbiota [29]. Lipid fluxes and the liver are expected to affect the development of hepatosteatosis and obesity [30].
Independent of the initial effect of lifestyle factors, the pathway to obesity requires hyperinsulinaemia as a critical mediator in translating an unfavourable lifestyle into body weight gain.

Main text

Hyperinsulinaemia versus obesity: epidemiological findings

Prospective studies exploring whether hyperinsulinaemia precedes and predicts later obesity have mostly been conducted in children and adolescents. Several studies found fasting hyperinsulinaemia and insulin resistance to be risk factors for weight gain in later years [3135]. In a recent cohort that included 39% obese children, fasting hyperinsulinaemia did not predict change in body mass index (BMI), except for more weight gain in obese children [36]. Studies in adults do not offer consistent results. Fasting hyperinsulinaemia predicted weight gain in postmenopausal women, except in the most obese [37]. By contrast, high fasting insulin levels were associated with lower rates of weight gain in cohorts with a mean BMI of 26 kg/m2 [38, 39] and in obese people [40, 41].
These results indicate that insulin levels may predict obesity in children and adolescents. Conclusions drawn from adult studies are less clear. However, these observational studies did not document and control for all lifestyle-dependent factors of obesity risk, all of which impact insulin secretion (Table 1). Only one study analysed dietary intake and an interaction was found between fasting insulin, total calories consumed, and fat percentage in predicting weight gain [42].

Hyperinsulinaemia versus obesity: intervention trials

A more direct approach for assessing the role of fasting (and diurnal) levels of insulin in weight gain includes interventions targeting insulin secretion. Insulin secretion can be partially inhibited with the potent ß-cell KATP channel opener diazoxide [43]. In a randomised controlled trial, diazoxide in conjunction with a hypocaloric diet for 8 weeks led to greater weight loss in obese people than those in the control group treated by diet alone [44]. In the diazoxide group, insulin levels decreased by 36% (fasting) and ~55% (post intravenous glucose) without differences in blood glucose levels compared with the control. In a similar trial, diazoxide did not induce more weight loss than the hypocaloric diet alone in the control group [45]. Unfortunately, the baseline fasting insulin levels in this study were significantly higher in the diazoxide group (by 32%) compared to the diet-alone group and the decrease in insulin secretion was not different between the diazoxide and control groups after 8 weeks of treatment. Taken together, body weight was reduced in all trials in which diazoxide achieved a lowering of basal and postchallenge blood glucose levels.
Insulin secretion can also be lowered by the long-acting somatostatin analogue octreotide. This peptide binds with high affinity to somatostatin receptors 5 and 2, effectively suppressing hormone production in ß-cells and several other endocrine cells, such as those in the pituitary or gut [43]. Severely obese adults were treated for 24 weeks with a dose of octreotide that significantly suppressed insulin production (and possibly other hormones). Stimulated insulin indices were reduced by 57% and there was significant weight loss (–3.5 kg/–2.8%) [46]. Secondary analyses showed that insulin was not significantly decreased in the subgroup with weight gain. By contrast, the remaining subgroup exhibited both a decrease in insulin secretion and substantial weight loss (–5.6 kg). In a subsequent similar study with three doses of long-acting octreotide and a randomised placebo control, the two higher doses caused significant weight loss (–2.1 kg/–1.9%) compared with the control group (–0.1 kg) [47]. As in the diazoxide trials, body weight reduction was only observed in association with lowered insulin levels.
Both compounds used to decrease insulin secretion have other pharmacological effects that may contribute to the weight loss observed. Diazoxide causes smooth muscle relaxation and fluid retention, while octreotide has a low risk of cardiac, hepatic and renal toxicity [48, 49]. However, the two drugs represent quite different pharmacological approaches, which share an insulin lowering effect but not adverse effects. Body weight reduction was only noted in conjunction with decreased insulin secretion.
In T2DM, treatment with exogenous insulin increases systemic insulin levels and this may support fat tissue growth [50]. The heterogeneous nature of T2DM means it is difficult to draw conclusions about the role of insulin or hyperinsulinaemia in a healthy metabolic state. Therefore, insulin therapy in type 1 or type 2 diabetes is not discussed here.
Increasing insulin concentrations in the brain appears to have opposite effects. Cerebral insulin is an anorexic hormone, but its actions are impaired in obese people because of brain insulin resistance [51]. Intranasal insulin delivery suppresses food intake and enhances postprandial thermogenesis, with concurrent lowering of postprandial systemic insulin levels [52, 53].

Hyperinsulinaemia versus obesity: genetic studies

Currently, selective lowering of circulating insulin levels without pharmacological effects in other organs can only be achieved by genetically manipulation. Mice harbour two insulin genes, Ins1 and Ins2. The tissue distribution pattern of Ins2 resembles that of the human insulin gene, while Ins1 is expressed in ß-cells only. Glucose homeostasis remains normal after ablation of either insulin gene. After disrupting Ins2 and one allele of Ins1, fasting insulin was substantially reduced (>50%) without persistent effects on glucose homeostasis [54]. When fed an obesogenic high-fat diet, these mice did not become obese, did not develop hepatic steatosis and increased energy expenditure in association with browning of white adipose tissue. In a second study, female mice with a deleted Ins1 gene and only one intact Ins2 allele exhibited lower insulin levels, but only during the first 6 months. However, this was sufficient to protect them from high-fat diet-induced obesity over one year of observation [55]. In a recent study using mice expressing only Ins2 (both alleles), mice exhibited compensatory high insulin production from Ins2 genes and developed obesity on a high fat diet. The Ins2 gene had been modified to allow reduction of the insulin gene dosage by the Cre-loxP system. Partial ablation of Ins2 alleles in adult obese mice led to significant weight loss, with a specific effect on visceral adipose tissue [56]. A moderate reduction in fasting insulin levels was seen (30%) and did not affect glucose tolerance, insulin sensitivity, glucose-induced insulin secretion or body growth under a low or moderate fat diet. There were no differences in the levels of several other circulating hormones, including leptin, resistin, ghrelin, GIP, GLP-1, IL-6, and PYY.
The role of insulin in adipose tissue growth was also tested by selectively disrupting the insulin receptor gene in fat cells of white and brown adipose tissue. Such mice grew normally and their glucose tolerance was not different from control littermates. Basal glucose uptake into adipocytes was unchanged, but insulin-stimulated glucose uptake reduced by ~90%. Mice with such selective insulin resistance of adipose tissue had low fat mass and were protected from age-related obesity [57]. In summary, four different approaches to lowering insulin secretion had the same consequence: prevention or remission of obesity (Fig. 1).
Mutations of the human insulin gene have been described that affect insulin secretion. However, in all cases, insulin secretion was impaired to an extent that resulted in increased fasting glucose levels or diabetes [5860].

Hyperinsulinaemia: old findings reappraised

Weight gain leading to an overweight BMI is usually caused by fat tissue growth rather than muscle. Ectopic storage of triglycerides in many other cell types, including liver and muscle cells, also contributes [61].
Insulin’s lipogenic activity has been well studied and and is identical in concentration dependence to its inhibitory action on lipolysis; i.e., signalling through the insulin receptor on adipocytes causes simultaneous inhibition of lipolysis and storage of triglycerides [62]. One observation, first made in the 1980s, is that lower concentrations of insulin are required to inhibit lipolysis in adipocytes than are needed to promote glucose influx into peripheral tissue. In hyperinsulinaemic–euglycaemic studies, the concentrations of plasma insulin required to lower plasma levels of the products of triglyceride metabolism by 50% were 42–120 pmol/l (mean = 78 pmol/l), in non-obese subjects [6369]. The mean fasting insulin level of all study groups combined was 48 pmol/l; i.e., people with fasting insulin levels above the mean had substantially inhibited lipolysis. In another hyperinsulinaemic–euglycaemic study published in 1999, the inhibitory action of systemic insulin was similar between adipose and muscle tissue. Increasing insulin concentrations from 50 to 63 pmol/l already significantly inhibited glycerol release by around 20% [70].
Since most of these studies were North American, we compared figures with the normal range of fasting serum insulin levels of the representative National Health and Nutrition Examination Survey 1988–1994. Geometric mean fasting serum insulin levels for non-obese nondiabetic people were ~46 pmol/l [71]. This suggests that more than half of the adult non-obese population in the USA had fasting insulin concentrations in the range required to inhibit ≥ 50% of lipolysis (Fig. 2). At the individual level, the relationship between insulin concentrations and lipolysis will be difficult to predict because of the substantial variation in fasting insulin concentrations (or insulin resistance) between non-obese people with varying genetic backgrounds and lifestyles [72]. Increased insulin resistance may attenuate increased lipolysis inhibition in people with higher fasting insulin levels. Such an interrelationship indeed exists, but two-thirds of the individual variation of insulin resistance cannot be explained by fasting insulin levels [73].
More important is the observation that stimulating glucose uptake by insulin requires much higher hormone concentrations than is needed to inhibit lipolysis, even in the same individual. In five clamp studies, the mean insulin concentrations required to have a 50% effect (EC50) on the stimulation of peripheral glucose uptake were ~720, 480, 348, 360 and 360 pmol/l (mean = 454 pmol/l) [64, 65, 67, 74, 75]. Half maximal stimulation of glucose uptake required an insulin concentration that was about six times higher compared with that required for 50% inhibition of lipolysis (Fig. 2).
Suppression of hepatic glucose production also required higher insulin concentrations than lipolysis inhibition – again, in part measured in the same individuals. EC50 values for insulin were ~300, 156, 156, 144, 120, 120 pmol/l (mean = 170 pmol/l) [6365, 67, 74]. Suppression of hepatic glucose output by 50% thus required more than twice the insulin concentration needed for half maximal inhibition of lipolysis (Fig. 2). To obtain the latter findings, insulin was infused at a peripheral vein to deliver a steady concentration of exogenous insulin to the liver via the arterial circulation. Usually, insulin is released from ß-cells in discrete pulses, about every 5 minutes, with an amplitude of 0.5–1 nmol/l insulin in the fasted state and up to 5 nmol/l after a meal [76]. Of these high amounts of insulin, 50–80% is taken up by hepatocytes and does not therefore reach the peripheral circulation. Since hepatocytes can modulate the extent of insulin clearance, peripheral insulin levels are not only determined by ß-cell function [76, 77].
Taken together, evidence is compelling that insulin levels on the high side of normal, or which are slightly elevated, substantially inhibit lipolysis in the absence of relevant insulin actions on hepatic glucose production or on glucose transport into muscle. Even in high fasting plasma insulin concentrations, lipolysis inhibition is the most sensitive response to insulin (Fig. 2). Since lipolysis inhibition is equivalent to lipogenesis promotion, the effect of insulin on adipocytes is probably responsible for the observed association between hyperinsulinaemia and incident obesity. It also fits with the anti-obesity effects of lowering insulin levels with diazoxide or octreotide, and with the prevention or remission of obesity with genetic downregulation of insulin production or insulin receptor expression on adipocytes in mice. Studies in rodents also suggest that reducing circulating insulin levels by inactivating insulin genes or with diazoxide increases the basal metabolic rate by enhanced heat production from mitochondria during fat oxidation by uncoupling protein 1 [54, 78].

Hyperinsulinaemia: the bigger picture

Inhibition of lipolysis/promotion of lipogenesis, hepatic gluconeogenesis and glucose uptake into insulin-sensitive cells via upregulation of glucose transporter GLUT4 all require different levels of insulin to signal effectively via their receptors. Currently, the most probable explanation is the activation of different insulin signalling pathways in the different tissues, such as the PI3K-Akt pathway versus the MAP kinase pathway [79]. Insulin stimulates glucose transport via the canonical PI3K-Akt pathway, whereas lipolysis is suppressed via Akt-independent suppression of protein kinase A [80]. A further concept is that of selective insulin resistance. Indeed, insulin resistance affects glucose uptake but does not interfere with ChREBP-ß-dependent de novo lipogenesis [79, 81].
It is not currently possible to disentangle the insulin-dependent regulatory network controlling body weight and weight increase. Insulin modifies its own activity [82] and interacts with other regulatory factors, such as other hormones, neuronal activity or gut function [8385]. Genetic components, such as putative thrifty genes and DNA sequences associated with obesity risk, add further complexity [86, 87]. It is therefore almost surprising that modulation of the systemic level of a single hormone, insulin, has such profound consequences on the risk of becoming obese. Insulin is our dominant anabolic hormone and, during an anabolic state of metabolism, cell stress is increased [20]. Insulin resistance is therefore considered a physiological defence to limit damage [88]. Low insulin levels extend the lifespan – at least in mice, possibly because of lower oxidative stress [8991]. This effect was seen in the absence of altered IGF-1 levels and was associated with lower fasting glucose and improved insulin sensitivity.
The inverse association between insulin-mediated lipolysis and lipogenesis in adipocytes [62] means that decreased adipose tissue growth is accompanied by increased release of non-esterified free fatty acids (FFAs) from adipocyte triglycerides because of increased lipolysis. Systemic FFAs mostly come from upper body subcutaneous fat and do not reflect visceral adiposity [92]. Individual fasting FFA concentrations vary substantially – even if measured on consecutive days (coefficient of variation, 45%, versus 4.8% for fasting glucose) [93]. One reason may be that the half-life of FFAs in the circulation is only 2–4 minutes [94].
The epidemiological association between increased FFA levels after an overnight fast with metabolic and cardiovascular outcomes is not convincing, because the opposite has also been reported [9496]. Women have FFA levels that are approximately 20% higher than men, yet they have similar insulin sensitivity [92, 94]. Fasting FFA concentrations were not associated with several measures of insulin resistance or with liver fat accumulation [97].
In lean people, mean overnight fasting FFA concentrations ranges between 300 and 600 μmol/l [94]. Obese people with manifold higher fat mass exhibit marginally higher FFAs (difference ~70 μmol/l, mean of 43 studies) in the circulation [94]. This indicates that fat tissue releases fewer FFAs in obesity, attributed to downregulation of enzymes involved in the breakdown of triglycerides [98]. By contrast, FFA levels increased by ~60% after 24 hours of severe energy restriction (2.3 MJ) and reached around 1300 μmol/l after a 72-hour fast [94, 99]. Severe energy restriction of a similar magnitude (2.5 MJ/day) in T2DM patients has reportedly substantially improved metabolic health and even reverted clinical T2DM [100]. As expected, there was a decrease in plasma insulin levels by approximately one-third, reaching the level of non-diabetic controls, and a concomitant increase in systemic FFAs by ~40%. However, elevated FFA concentrations reverted to baseline levels within 8 weeks. Similarly, bariatric surgery lowered fasting insulin levels, accompanied by an increase of FFA concentrations, but FFA levels returned to baseline or lower after several months [101]. These findings indicate that humans adapt to lower insulin levels by normalising initially increased FFA levels.

Conclusions

Barbara Corkey introduced the concept of hyperinsulinaemia as a risk factor for obesity [102]. She suggested that environmental agents, such as food additives, toxins or excess iron, which have entered the food chain since 1980, might cause insulin hypersecretion [102, 103]. We report here that all lifestyle characteristics known to confer a risk of obesity are associated with hyperinsulinaemia. Any relationship between unfavourable lifestyle factors and high consumption of food additives or toxins at a global level remains unknown. Here, we suggest a mechanism for the association between hyperinsulinaemia and obesity, based on work mostly published in the 1980s. These studies concur in that much lower concentrations of insulin are required to inhibit lipolysis compared with gluconeogenesis or the promotion of glucose uptake (Fig. 2).
Interestingly, fasting insulin levels were much lower in lean vegetarians (mean = 30 pmol/l) than in a lean case control group with similar energy and major nutrient intake (mean = 44 pmol/l) and there was only a minor difference in fasting glucose values (means = 4.47 versus 4.71 mmol/l) [104]. Although insulin acts in virtually all tissues of the body, the low insulin levels of vegetarians are apparently sufficient to maintain the hormonal effects of insulin in the body. The very low basal insulin concentrations in vegetarians indicate that fasting insulin levels are modified by type of diet. The strongest effects are seen in people who follow very low calorie diets [100], intermittent fasting [105], or undergo bariatric surgery [106]. Dietary interventions are complicated by a diurnal pattern of insulin resistance, being lowest in the morning [107, 108]. Therefore, skipping breakfast has less favourable consequences than skipping dinner [109]. Physical exercise also reduces fasting insulin levels [7] (independently of weight change because it can be observed after a single exercise session) [110].
It may be insufficient to recognise prolonged hyperinsulinaemia by monitoring diurnal glucose levels. For instance, people with higher fasting insulin levels but normal glycaemia respond with higher postprandial insulin secretion than people with low basal insulin [111]. In healthy adults, oral glucose tolerance was not affected by the level of physical activity on the preceding day, but serum insulin levels during the glucose tolerance test were lower after high physical activity [112].
Taken together, the data presented justify the recommendation of a lifestyle that avoids high insulin levels for much of the day to limit the period of anabolic fat tissue activity (Box 1).

Box 1: Key points

• All known lifestyle-dependent obesity risk factors are associated with, or give rise to, hyperinsulinaemia.
• Insulin levels that are on the high side of normal, or that are slightly elevated, predict later obesity in children and adolescents but not in adults.
• Pharmacological lowering of insulin secretion by diazoxide or octreotide causes weight loss.
• In mice, genetic lowering of insulin levels or selective genetic disruption of the insulin receptor in adipocytes causes prevention or remission of obesity.
• Lipolysis inhibition is the most sensitive metabolic action of insulin. Consequently, fasting insulin levels that are on the high side of normal, or that are slightly elevated, are sufficient to substantially inhibit lipolysis and the promotion of concomitant lipogenesis in adipocytes.
• Insulin concentrations that are six times higher than normal are required to stimulate glucose uptake and two times higher than normal to inhibit gluconeogenesis.
• It seems justified to suggest a lifestyle that avoids high insulin levels for much of the day to limit the period of anabolic fat tissue activity. Appropriate measures include low calorie diets, intermittent fasting or physical activity.

Acknowledgements

We thank Dr Fraser W Scott, University of Ottawa, for reading the manuscript.

Funding

Not applicable.

Availability of data and materials

Data for this review were identified by searching MEDLINE, PubMed and references from relevant articles using the search terms “lipolysis”, “hyperinsulinemia”, “adipocyte lipogenesis”, “free fatty acids” OR “non esterified fatty acids”, and “obesity”. To limit the number of references, more recently published papers referring to several previously published articles were cited, if possible. Only articles published in English were selected.
Not applicable.
Not applicable.

Competing interests

SM has received non-restricted support to conduct trials of lifestyle change in people who have, or are at risk of, T2DM by Novartis, Boehringer Ingelheim, Almased Wellness, Nintendo of Europe, HMM Holding, Gesellschaft von Freunden und Förderern der Heinrich-Heine-Universität Düsseldorf. Sponsors played no role in writing this manuscript or the decision to submit it for publication.
HK, MS, WK and KK declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults. Lancet. 2017;390:2627–42.CrossRef NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults. Lancet. 2017;390:2627–42.CrossRef
2.
Zurück zum Zitat NCD Risk Factor Collaboration (NCD-RisC). Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2 million participants. Lancet. 2016;387:1377–96.CrossRef NCD Risk Factor Collaboration (NCD-RisC). Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2 million participants. Lancet. 2016;387:1377–96.CrossRef
4.
Zurück zum Zitat Herieka M, Erridge C. High-fat meal induced postprandial inflammation. Mol Nutr Food Res. 2014;58:136–46.PubMedCrossRef Herieka M, Erridge C. High-fat meal induced postprandial inflammation. Mol Nutr Food Res. 2014;58:136–46.PubMedCrossRef
5.
Zurück zum Zitat de Vries MA, Klop B, Janssen HW, Njo TL, Westerman EM, Castro CM. Postprandial inflammation: targeting glucose and lipids. Adv Exp Med Biol. 2014;824:161–70.PubMedCrossRef de Vries MA, Klop B, Janssen HW, Njo TL, Westerman EM, Castro CM. Postprandial inflammation: targeting glucose and lipids. Adv Exp Med Biol. 2014;824:161–70.PubMedCrossRef
6.
Zurück zum Zitat Boden G, Homko C, Barrero CA, Stein TP, Chen X, Cheung P, et al. Excessive caloric intake acutely causes oxidative stress, GLUT4 carbonylation, and insulin resistance in healthy men. Sci Transl Med. 2015;7:304re7.PubMedCrossRefPubMedCentral Boden G, Homko C, Barrero CA, Stein TP, Chen X, Cheung P, et al. Excessive caloric intake acutely causes oxidative stress, GLUT4 carbonylation, and insulin resistance in healthy men. Sci Transl Med. 2015;7:304re7.PubMedCrossRefPubMedCentral
7.
Zurück zum Zitat Jelleyman C, Yates T, O'Donovan G, Gray LJ, King JA, Khunti K, et al. The effects of high-intensity interval training on glucose regulation and insulin resistance: a meta-analysis. Obes Rev. 2015;16:942–61.PubMedCrossRef Jelleyman C, Yates T, O'Donovan G, Gray LJ, King JA, Khunti K, et al. The effects of high-intensity interval training on glucose regulation and insulin resistance: a meta-analysis. Obes Rev. 2015;16:942–61.PubMedCrossRef
8.
Zurück zum Zitat Bowden Davies KA, Sprung VS, Norman JA, Thompson A, Mitchell KL, Halford JCG, et al. Short-term decreased physical activity with increased sedentary behaviour causes metabolic derangements and altered body composition: effects in individuals with and without a first-degree relative with type 2 diabetes. Diabetologia. 2018;61:1282–94.PubMedCrossRef Bowden Davies KA, Sprung VS, Norman JA, Thompson A, Mitchell KL, Halford JCG, et al. Short-term decreased physical activity with increased sedentary behaviour causes metabolic derangements and altered body composition: effects in individuals with and without a first-degree relative with type 2 diabetes. Diabetologia. 2018;61:1282–94.PubMedCrossRef
9.
Zurück zum Zitat Li H, Cai J, Chen R, Zhao Z, Ying Z, Wang L, et al. Particulate matter exposure and stress hormone levels: a randomized, double-blind, crossover trial of air purification. Circulation. 2017;136:618–27.PubMedCrossRef Li H, Cai J, Chen R, Zhao Z, Ying Z, Wang L, et al. Particulate matter exposure and stress hormone levels: a randomized, double-blind, crossover trial of air purification. Circulation. 2017;136:618–27.PubMedCrossRef
10.
Zurück zum Zitat Donga E, van Dijk M, van Dijk JG, Biermasz NR, Lammers GJ, van Kralingen KW, et al. A single night of partial sleep deprivation induces insulin resistance in multiple metabolic pathways in healthy subjects. J Clin Endocrinol Metab. 2010;95:2963–8.PubMedCrossRef Donga E, van Dijk M, van Dijk JG, Biermasz NR, Lammers GJ, van Kralingen KW, et al. A single night of partial sleep deprivation induces insulin resistance in multiple metabolic pathways in healthy subjects. J Clin Endocrinol Metab. 2010;95:2963–8.PubMedCrossRef
11.
Zurück zum Zitat Rao MN, Neylan TC, Grunfeld C, Mulligan K, Schambelan M, Schwarz JM. Subchronic sleep restriction causes tissue-specific insulin resistance. J Clin Endocrinol Metab. 2015;100:1664–71.PubMedCrossRefPubMedCentral Rao MN, Neylan TC, Grunfeld C, Mulligan K, Schambelan M, Schwarz JM. Subchronic sleep restriction causes tissue-specific insulin resistance. J Clin Endocrinol Metab. 2015;100:1664–71.PubMedCrossRefPubMedCentral
12.
Zurück zum Zitat Wright KP Jr, Drake AL, Frey DJ, Fleshner M, Desouza CA, Gronfier C, et al. Influence of sleep deprivation and circadian misalignment on cortisol, inflammatory markers, and cytokine balance. Brain Behav Immun. 2015;47:24–34.PubMedCrossRefPubMedCentral Wright KP Jr, Drake AL, Frey DJ, Fleshner M, Desouza CA, Gronfier C, et al. Influence of sleep deprivation and circadian misalignment on cortisol, inflammatory markers, and cytokine balance. Brain Behav Immun. 2015;47:24–34.PubMedCrossRefPubMedCentral
13.
Zurück zum Zitat Rao MN, Chau A, Madden E, Inslicht S, Talbot L, Richards A, et al. Hyperinsulinemic response to oral glucose challenge in individuals with posttraumatic stress disorder. Psychoneuroendocrinology. 2014;49:171–81.PubMedCrossRefPubMedCentral Rao MN, Chau A, Madden E, Inslicht S, Talbot L, Richards A, et al. Hyperinsulinemic response to oral glucose challenge in individuals with posttraumatic stress disorder. Psychoneuroendocrinology. 2014;49:171–81.PubMedCrossRefPubMedCentral
14.
15.
Zurück zum Zitat Piazzolla G, Castrovilli A, Liotino V, Vulpi MR, Fanelli M, Mazzocca A, et al. Metabolic syndrome and Chronic Obstructive Pulmonary Disease (COPD): the interplay among smoking, insulin resistance and vitamin D. PLoS One. 2017;12:e0186708.PubMedCrossRefPubMedCentral Piazzolla G, Castrovilli A, Liotino V, Vulpi MR, Fanelli M, Mazzocca A, et al. Metabolic syndrome and Chronic Obstructive Pulmonary Disease (COPD): the interplay among smoking, insulin resistance and vitamin D. PLoS One. 2017;12:e0186708.PubMedCrossRefPubMedCentral
16.
Zurück zum Zitat Webb M, Davies M, Ashra N, Bodicoat D, Brady E, Webb D, et al. The association between depressive symptoms and insulin resistance, inflammation and adiposity in men and women. PLoS One. 2017;12:e0187448.PubMedCrossRefPubMedCentral Webb M, Davies M, Ashra N, Bodicoat D, Brady E, Webb D, et al. The association between depressive symptoms and insulin resistance, inflammation and adiposity in men and women. PLoS One. 2017;12:e0187448.PubMedCrossRefPubMedCentral
17.
Zurück zum Zitat Bonaccio M, Di Castelnuovo A, Pounis G, De Curtis A, Costanzo S, Persichillo M, et al. Relative contribution of health-related behaviours and chronic diseases to the socioeconomic patterning of low-grade inflammation. Int J Public Health. 2017;62:551–62.PubMedCrossRef Bonaccio M, Di Castelnuovo A, Pounis G, De Curtis A, Costanzo S, Persichillo M, et al. Relative contribution of health-related behaviours and chronic diseases to the socioeconomic patterning of low-grade inflammation. Int J Public Health. 2017;62:551–62.PubMedCrossRef
18.
Zurück zum Zitat Volaco A, Cavalcanti AM, Filho RP, Precoma DB. Socioeconomic status: the missing link between obesity and diabetes mellitus? Curr Diabetes Rev. 2018;14:321–6.PubMedCrossRef Volaco A, Cavalcanti AM, Filho RP, Precoma DB. Socioeconomic status: the missing link between obesity and diabetes mellitus? Curr Diabetes Rev. 2018;14:321–6.PubMedCrossRef
19.
Zurück zum Zitat Kolb H, Mandrup-Poulsen T. The global diabetes epidemic as a consequence of lifestyle-induced low-grade inflammation. Diabetologia. 2010;53:10–20.PubMedCrossRef Kolb H, Mandrup-Poulsen T. The global diabetes epidemic as a consequence of lifestyle-induced low-grade inflammation. Diabetologia. 2010;53:10–20.PubMedCrossRef
20.
Zurück zum Zitat Kolb H, Eizirik DL. Resistance to type 2 diabetes mellitus: a matter of hormesis? Nat Rev Endocrinol. 2011;8:183–92.PubMedCrossRef Kolb H, Eizirik DL. Resistance to type 2 diabetes mellitus: a matter of hormesis? Nat Rev Endocrinol. 2011;8:183–92.PubMedCrossRef
21.
Zurück zum Zitat Spinas GA, Palmer JP, Mandrup-Poulsen T, Andersen H, Nielsen JH, Nerup J. The bimodal effect of interleukin 1 on rat pancreatic beta-cells--stimulation followed by inhibition--depends upon dose, duration of exposure, and ambient glucose concentration. Acta Endocrinol. 1988;119:307–11.CrossRef Spinas GA, Palmer JP, Mandrup-Poulsen T, Andersen H, Nielsen JH, Nerup J. The bimodal effect of interleukin 1 on rat pancreatic beta-cells--stimulation followed by inhibition--depends upon dose, duration of exposure, and ambient glucose concentration. Acta Endocrinol. 1988;119:307–11.CrossRef
22.
Zurück zum Zitat Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, et al. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med. 2005;11:191–8.PubMedCrossRef Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, et al. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med. 2005;11:191–8.PubMedCrossRef
23.
Zurück zum Zitat Cai D, Yuan M, Frantz DF, Melendez PA, Hansen L, Lee J, et al. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat Med. 2005;11:183–90.PubMedCrossRefPubMedCentral Cai D, Yuan M, Frantz DF, Melendez PA, Hansen L, Lee J, et al. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat Med. 2005;11:183–90.PubMedCrossRefPubMedCentral
24.
Zurück zum Zitat Anagnostis P, Athyros VG, Tziomalos K, Karagiannis A, Mikhailidis DP. Clinical review: The pathogenetic role of cortisol in the metabolic syndrome: a hypothesis. J Clin Endocrinol Metab. 2009;94:2692–701.PubMedCrossRef Anagnostis P, Athyros VG, Tziomalos K, Karagiannis A, Mikhailidis DP. Clinical review: The pathogenetic role of cortisol in the metabolic syndrome: a hypothesis. J Clin Endocrinol Metab. 2009;94:2692–701.PubMedCrossRef
25.
26.
Zurück zum Zitat Douglass JD, Dorfman MD, Thaler JP. Glia: silent partners in energy homeostasis and obesity pathogenesis. Diabetologia. 2017;60:226–36.PubMedCrossRef Douglass JD, Dorfman MD, Thaler JP. Glia: silent partners in energy homeostasis and obesity pathogenesis. Diabetologia. 2017;60:226–36.PubMedCrossRef
27.
Zurück zum Zitat Graupera M, Claret M. Endothelial Cells: New players in obesity and related metabolic disorders. Trends Endocrinol Metab. 2018;29:781–94.PubMedCrossRef Graupera M, Claret M. Endothelial Cells: New players in obesity and related metabolic disorders. Trends Endocrinol Metab. 2018;29:781–94.PubMedCrossRef
28.
Zurück zum Zitat Mace A, Tuke MA, Deelen P, Kristiansson K, Mattsson H, Noukas M, et al. CNV-association meta-analysis in 191,161 European adults reveals new loci associated with anthropometric traits. Nat Commun. 2017;8:744.PubMedCrossRefPubMedCentral Mace A, Tuke MA, Deelen P, Kristiansson K, Mattsson H, Noukas M, et al. CNV-association meta-analysis in 191,161 European adults reveals new loci associated with anthropometric traits. Nat Commun. 2017;8:744.PubMedCrossRefPubMedCentral
29.
Zurück zum Zitat Mulders RJ, de Git KCG, Schele E, Dickson SL, Sanz Y, Adan RAH. Microbiota in obesity: interactions with enteroendocrine, immune and central nervous systems. Obes Rev. 2018;19:435–51.PubMedCrossRef Mulders RJ, de Git KCG, Schele E, Dickson SL, Sanz Y, Adan RAH. Microbiota in obesity: interactions with enteroendocrine, immune and central nervous systems. Obes Rev. 2018;19:435–51.PubMedCrossRef
30.
Zurück zum Zitat Ertunc ME, Hotamisligil GS. Lipid signaling and lipotoxicity in metaflammation: indications for metabolic disease pathogenesis and treatment. J Lipid Res. 2016;57:2099–114.PubMedCrossRefPubMedCentral Ertunc ME, Hotamisligil GS. Lipid signaling and lipotoxicity in metaflammation: indications for metabolic disease pathogenesis and treatment. J Lipid Res. 2016;57:2099–114.PubMedCrossRefPubMedCentral
31.
Zurück zum Zitat Odeleye OE, de Courten M, Pettitt DJ, Ravussin E. Fasting hyperinsulinemia is a predictor of increased body weight gain and obesity in Pima Indian children. Diabetes. 1997;46:1341–5.PubMedCrossRef Odeleye OE, de Courten M, Pettitt DJ, Ravussin E. Fasting hyperinsulinemia is a predictor of increased body weight gain and obesity in Pima Indian children. Diabetes. 1997;46:1341–5.PubMedCrossRef
32.
Zurück zum Zitat Morrison JA, Glueck CJ, Horn PS, Schreiber GB, Wang P. Pre-teen insulin resistance predicts weight gain, impaired fasting glucose, and type 2 diabetes at age 18–19 y: a 10-y prospective study of black and white girls. Am J Clin Nutr. 2008;88:778–88.PubMedCrossRef Morrison JA, Glueck CJ, Horn PS, Schreiber GB, Wang P. Pre-teen insulin resistance predicts weight gain, impaired fasting glucose, and type 2 diabetes at age 18–19 y: a 10-y prospective study of black and white girls. Am J Clin Nutr. 2008;88:778–88.PubMedCrossRef
33.
Zurück zum Zitat Adam TC, Toledo-Corral C, Lane CJ, Weigensberg MJ, Spruijt-Metz D, Davies JN, et al. Insulin sensitivity as an independent predictor of fat mass gain in Hispanic adolescents. Diabetes Care. 2009;32:2114–5.PubMedCrossRefPubMedCentral Adam TC, Toledo-Corral C, Lane CJ, Weigensberg MJ, Spruijt-Metz D, Davies JN, et al. Insulin sensitivity as an independent predictor of fat mass gain in Hispanic adolescents. Diabetes Care. 2009;32:2114–5.PubMedCrossRefPubMedCentral
34.
Zurück zum Zitat Labayen I, Ruiz JR, Ortega FB, Harro J, Merenakk L, Oja L, et al. Insulin sensitivity at childhood predicts changes in total and central adiposity over a 6-year period. Int J Obes. 2011;35:1284–8.CrossRef Labayen I, Ruiz JR, Ortega FB, Harro J, Merenakk L, Oja L, et al. Insulin sensitivity at childhood predicts changes in total and central adiposity over a 6-year period. Int J Obes. 2011;35:1284–8.CrossRef
35.
Zurück zum Zitat Chen YY, Wang JP, Jiang YY, Li H, Hu YH, Lee KO, et al. Fasting plasma insulin at 5 years of age predicted subsequent weight increase in early childhood over a 5-year period – the Da Qing Children Cohort Study. PLoS One. 2015;10:e0127389.PubMedCrossRefPubMedCentral Chen YY, Wang JP, Jiang YY, Li H, Hu YH, Lee KO, et al. Fasting plasma insulin at 5 years of age predicted subsequent weight increase in early childhood over a 5-year period – the Da Qing Children Cohort Study. PLoS One. 2015;10:e0127389.PubMedCrossRefPubMedCentral
36.
Zurück zum Zitat Sedaka NM, Olsen CH, Yannai LE, Stutzman WE, Krause AJ, Sherafat-Kazemzadeh R, et al. A longitudinal study of serum insulin and insulin resistance as predictors of weight and body fat gain in African American and Caucasian children. Int J Obes. 2017;41:61–70.CrossRef Sedaka NM, Olsen CH, Yannai LE, Stutzman WE, Krause AJ, Sherafat-Kazemzadeh R, et al. A longitudinal study of serum insulin and insulin resistance as predictors of weight and body fat gain in African American and Caucasian children. Int J Obes. 2017;41:61–70.CrossRef
37.
Zurück zum Zitat Howard BV, Adams-Campbell L, Allen C, Black H, Passaro M, Rodabough RJ, et al. Insulin resistance and weight gain in postmenopausal women of diverse ethnic groups. Int J Obes Relat Metab Disord. 2004;28:1039–47.PubMedCrossRef Howard BV, Adams-Campbell L, Allen C, Black H, Passaro M, Rodabough RJ, et al. Insulin resistance and weight gain in postmenopausal women of diverse ethnic groups. Int J Obes Relat Metab Disord. 2004;28:1039–47.PubMedCrossRef
38.
Zurück zum Zitat Hoag S, Marshall JA, Jones RH, Hamman RF. High fasting insulin levels associated with lower rates of weight gain in persons with normal glucose tolerance: the San Luis Valley Diabetes Study. Int J Obes Relat Metab Disord. 1995;19:175–80.PubMed Hoag S, Marshall JA, Jones RH, Hamman RF. High fasting insulin levels associated with lower rates of weight gain in persons with normal glucose tolerance: the San Luis Valley Diabetes Study. Int J Obes Relat Metab Disord. 1995;19:175–80.PubMed
39.
Zurück zum Zitat Wedick NM, Snijder MB, Dekker JM, Heine RJ, Stehouwer CD, Nijpels G, et al. Prospective investigation of metabolic characteristics in relation to weight gain in older adults: the Hoorn Study. Obesity. 2009;17:1609–14.PubMedCrossRef Wedick NM, Snijder MB, Dekker JM, Heine RJ, Stehouwer CD, Nijpels G, et al. Prospective investigation of metabolic characteristics in relation to weight gain in older adults: the Hoorn Study. Obesity. 2009;17:1609–14.PubMedCrossRef
40.
Zurück zum Zitat Swinburn BA, Nyomba BL, Saad MF, Zurlo F, Raz I, Knowler WC, et al. Insulin resistance associated with lower rates of weight gain in Pima Indians. J Clin Invest. 1991;88:168–73.PubMedCrossRefPubMedCentral Swinburn BA, Nyomba BL, Saad MF, Zurlo F, Raz I, Knowler WC, et al. Insulin resistance associated with lower rates of weight gain in Pima Indians. J Clin Invest. 1991;88:168–73.PubMedCrossRefPubMedCentral
41.
Zurück zum Zitat Valdez R, Mitchell BD, Haffner SM, Hazuda HP, Morales PA, Monterrosa A, et al. Predictors of weight change in a bi-ethnic population. The San Antonio Heart Study. Int J Obes Relat Metab Disord. 1994;18:85–91.PubMed Valdez R, Mitchell BD, Haffner SM, Hazuda HP, Morales PA, Monterrosa A, et al. Predictors of weight change in a bi-ethnic population. The San Antonio Heart Study. Int J Obes Relat Metab Disord. 1994;18:85–91.PubMed
42.
Zurück zum Zitat Morrison JA, Glueck CJ, Wang P. Preteen insulin levels interact with caloric intake to predict increases in obesity at ages 18 to 19 years: a 10-year prospective study of black and white girls. Metabolism. 2010;59:718–27.PubMedCrossRef Morrison JA, Glueck CJ, Wang P. Preteen insulin levels interact with caloric intake to predict increases in obesity at ages 18 to 19 years: a 10-year prospective study of black and white girls. Metabolism. 2010;59:718–27.PubMedCrossRef
43.
Zurück zum Zitat Doyle ME, Egan JM. Pharmacological agents that directly modulate insulin secretion. Pharmacol Rev. 2003;55:105–31.PubMedCrossRef Doyle ME, Egan JM. Pharmacological agents that directly modulate insulin secretion. Pharmacol Rev. 2003;55:105–31.PubMedCrossRef
44.
Zurück zum Zitat Alemzadeh R, Langley G, Upchurch L, Smith P, Slonim AE. Beneficial effect of diazoxide in obese hyperinsulinemic adults. J Clin Endocrinol Metab. 1998;83:1911–5.PubMed Alemzadeh R, Langley G, Upchurch L, Smith P, Slonim AE. Beneficial effect of diazoxide in obese hyperinsulinemic adults. J Clin Endocrinol Metab. 1998;83:1911–5.PubMed
45.
Zurück zum Zitat Due A, Flint A, Eriksen G, Moller B, Raben A, Hansen JB, et al. No effect of inhibition of insulin secretion by diazoxide on weight loss in hyperinsulinaemic obese subjects during an 8-week weight-loss diet. Diabetes Obes Metab. 2007;9:566–74.PubMedCrossRef Due A, Flint A, Eriksen G, Moller B, Raben A, Hansen JB, et al. No effect of inhibition of insulin secretion by diazoxide on weight loss in hyperinsulinaemic obese subjects during an 8-week weight-loss diet. Diabetes Obes Metab. 2007;9:566–74.PubMedCrossRef
46.
Zurück zum Zitat Velasquez-Mieyer PA, Cowan PA, Arheart KL, Buffington CK, Spencer KA, Connelly BE, et al. Suppression of insulin secretion is associated with weight loss and altered macronutrient intake and preference in a subset of obese adults. Int J Obes Relat Metab Disord. 2003;27:219–26.PubMedCrossRefPubMedCentral Velasquez-Mieyer PA, Cowan PA, Arheart KL, Buffington CK, Spencer KA, Connelly BE, et al. Suppression of insulin secretion is associated with weight loss and altered macronutrient intake and preference in a subset of obese adults. Int J Obes Relat Metab Disord. 2003;27:219–26.PubMedCrossRefPubMedCentral
47.
Zurück zum Zitat Lustig RH, Greenway F, Velasquez-Mieyer P, Heimburger D, Schumacher D, Smith D, et al. A multicenter, randomized, double-blind, placebo-controlled, dose-finding trial of a long-acting formulation of octreotide in promoting weight loss in obese adults with insulin hypersecretion. Int J Obes. 2006;30:331–41.CrossRef Lustig RH, Greenway F, Velasquez-Mieyer P, Heimburger D, Schumacher D, Smith D, et al. A multicenter, randomized, double-blind, placebo-controlled, dose-finding trial of a long-acting formulation of octreotide in promoting weight loss in obese adults with insulin hypersecretion. Int J Obes. 2006;30:331–41.CrossRef
48.
Zurück zum Zitat Hu S, Xu Z, Yan J, Liu M, Sun B, Li W, et al. The treatment effect of diazoxide on 44 patients with congenital hyperinsulinism. J Pediatr Endocrinol Metab. 2012;25:1119–22.PubMedCrossRef Hu S, Xu Z, Yan J, Liu M, Sun B, Li W, et al. The treatment effect of diazoxide on 44 patients with congenital hyperinsulinism. J Pediatr Endocrinol Metab. 2012;25:1119–22.PubMedCrossRef
49.
Zurück zum Zitat Pivonello R, Muscogiuri G, Holder G, Paul M, Sarp S, Lesogor A, et al. Long-term safety of long-acting octreotide in patients with diabetic retinopathy: results of pooled data from 2 randomized, double-blind, placebo-controlled phase 3 studies. Endocrine. 2018;60:65–72.PubMedCrossRef Pivonello R, Muscogiuri G, Holder G, Paul M, Sarp S, Lesogor A, et al. Long-term safety of long-acting octreotide in patients with diabetic retinopathy: results of pooled data from 2 randomized, double-blind, placebo-controlled phase 3 studies. Endocrine. 2018;60:65–72.PubMedCrossRef
50.
Zurück zum Zitat Hodish I. Insulin therapy, weight gain and prognosis. Diabetes Obes Metab. 2018;20:2085–92.PubMedCrossRef Hodish I. Insulin therapy, weight gain and prognosis. Diabetes Obes Metab. 2018;20:2085–92.PubMedCrossRef
51.
Zurück zum Zitat Kullmann S, Heni M, Hallschmid M, Fritsche A, Preissl H, Haring HU. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiol Rev. 2016;96:1169–209.PubMedCrossRef Kullmann S, Heni M, Hallschmid M, Fritsche A, Preissl H, Haring HU. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiol Rev. 2016;96:1169–209.PubMedCrossRef
52.
Zurück zum Zitat Jauch-Chara K, Friedrich A, Rezmer M, Melchert UH, Scholand-Engler G, Hallschmid M, et al. Intranasal insulin suppresses food intake via enhancement of brain energy levels in humans. Diabetes. 2012;61:2261–8.PubMedCrossRefPubMedCentral Jauch-Chara K, Friedrich A, Rezmer M, Melchert UH, Scholand-Engler G, Hallschmid M, et al. Intranasal insulin suppresses food intake via enhancement of brain energy levels in humans. Diabetes. 2012;61:2261–8.PubMedCrossRefPubMedCentral
53.
Zurück zum Zitat Benedict C, Brede S, Schioth HB, Lehnert H, Schultes B, Born J, et al. Intranasal insulin enhances postprandial thermogenesis and lowers postprandial serum insulin levels in healthy men. Diabetes. 2011;60:114–8.PubMedCrossRef Benedict C, Brede S, Schioth HB, Lehnert H, Schultes B, Born J, et al. Intranasal insulin enhances postprandial thermogenesis and lowers postprandial serum insulin levels in healthy men. Diabetes. 2011;60:114–8.PubMedCrossRef
54.
Zurück zum Zitat Mehran AE, Templeman NM, Brigidi GS, Lim GE, Chu KY, Hu X, et al. Hyperinsulinemia drives diet-induced obesity independently of brain insulin production. Cell Metab. 2012;16:723–37.PubMedCrossRef Mehran AE, Templeman NM, Brigidi GS, Lim GE, Chu KY, Hu X, et al. Hyperinsulinemia drives diet-induced obesity independently of brain insulin production. Cell Metab. 2012;16:723–37.PubMedCrossRef
55.
Zurück zum Zitat Templeman NM, Clee SM, Johnson JD. Suppression of hyperinsulinaemia in growing female mice provides long-term protection against obesity. Diabetologia. 2015;58:2392–402.PubMedCrossRefPubMedCentral Templeman NM, Clee SM, Johnson JD. Suppression of hyperinsulinaemia in growing female mice provides long-term protection against obesity. Diabetologia. 2015;58:2392–402.PubMedCrossRefPubMedCentral
56.
Zurück zum Zitat Page MM, Skovso S, Cen H, Chiu AP, Dionne DA, Hutchinson DF, et al. Reducing insulin via conditional partial gene ablation in adults reverses diet-induced weight gain. FASEB J. 2018;32:1196–206.PubMedCrossRef Page MM, Skovso S, Cen H, Chiu AP, Dionne DA, Hutchinson DF, et al. Reducing insulin via conditional partial gene ablation in adults reverses diet-induced weight gain. FASEB J. 2018;32:1196–206.PubMedCrossRef
57.
Zurück zum Zitat Bluher M, Michael MD, Peroni OD, Ueki K, Carter N, Kahn BB, et al. Adipose tissue selective insulin receptor knockout protects against obesity and obesity-related glucose intolerance. Dev Cell. 2002;3:25–38.PubMedCrossRef Bluher M, Michael MD, Peroni OD, Ueki K, Carter N, Kahn BB, et al. Adipose tissue selective insulin receptor knockout protects against obesity and obesity-related glucose intolerance. Dev Cell. 2002;3:25–38.PubMedCrossRef
58.
Zurück zum Zitat Edghill EL, Flanagan SE, Patch AM, Boustred C, Parrish A, Shields B, et al. Insulin mutation screening in 1,044 patients with diabetes: mutations in the INS gene are a common cause of neonatal diabetes but a rare cause of diabetes diagnosed in childhood or adulthood. Diabetes. 2008;57:1034–42.PubMedCrossRef Edghill EL, Flanagan SE, Patch AM, Boustred C, Parrish A, Shields B, et al. Insulin mutation screening in 1,044 patients with diabetes: mutations in the INS gene are a common cause of neonatal diabetes but a rare cause of diabetes diagnosed in childhood or adulthood. Diabetes. 2008;57:1034–42.PubMedCrossRef
59.
Zurück zum Zitat Molven A, Ringdal M, Nordbo AM, Raeder H, Stoy J, Lipkind GM, et al. Mutations in the insulin gene can cause MODY and autoantibody-negative type 1 diabetes. Diabetes. 2008;57:1131–5.PubMedCrossRef Molven A, Ringdal M, Nordbo AM, Raeder H, Stoy J, Lipkind GM, et al. Mutations in the insulin gene can cause MODY and autoantibody-negative type 1 diabetes. Diabetes. 2008;57:1131–5.PubMedCrossRef
60.
Zurück zum Zitat Kleinberger JW, Copeland KC, Gandica RG, Haymond MW, Levitsky LL, Linder B, et al. Monogenic diabetes in overweight and obese youth diagnosed with type 2 diabetes: the TODAY clinical trial. Genet Med. 2018;20:583–90.PubMedCrossRef Kleinberger JW, Copeland KC, Gandica RG, Haymond MW, Levitsky LL, Linder B, et al. Monogenic diabetes in overweight and obese youth diagnosed with type 2 diabetes: the TODAY clinical trial. Genet Med. 2018;20:583–90.PubMedCrossRef
62.
Zurück zum Zitat Thomas SH, Wisher MH, Brandenburg D, Sonksen PH. Insulin action on adipocytes. Evidence that the anti-lipolytic and lipogenic effects of insulin are mediated by the same receptor. Biochem J. 1979;184:355–60.PubMedCrossRefPubMedCentral Thomas SH, Wisher MH, Brandenburg D, Sonksen PH. Insulin action on adipocytes. Evidence that the anti-lipolytic and lipogenic effects of insulin are mediated by the same receptor. Biochem J. 1979;184:355–60.PubMedCrossRefPubMedCentral
63.
Zurück zum Zitat Nurjhan N, Campbell PJ, Kennedy FP, Miles JM, Gerich JE. Insulin dose-response characteristics for suppression of glycerol release and conversion to glucose in humans. Diabetes. 1986;35:1326–31.PubMedCrossRef Nurjhan N, Campbell PJ, Kennedy FP, Miles JM, Gerich JE. Insulin dose-response characteristics for suppression of glycerol release and conversion to glucose in humans. Diabetes. 1986;35:1326–31.PubMedCrossRef
64.
Zurück zum Zitat Groop LC, Bonadonna RC, DelPrato S, Ratheiser K, Zyck K, Ferrannini E, et al. Glucose and free fatty acid metabolism in non-insulin-dependent diabetes mellitus. Evidence for multiple sites of insulin resistance. J Clin Invest. 1989;84:205–13.PubMedCrossRefPubMedCentral Groop LC, Bonadonna RC, DelPrato S, Ratheiser K, Zyck K, Ferrannini E, et al. Glucose and free fatty acid metabolism in non-insulin-dependent diabetes mellitus. Evidence for multiple sites of insulin resistance. J Clin Invest. 1989;84:205–13.PubMedCrossRefPubMedCentral
65.
Zurück zum Zitat Bonadonna RC, Groop L, Kraemer N, Ferrannini E, Del Prato S, DeFronzo RA. Obesity and insulin resistance in humans: a dose-response study. Metabolism. 1990;39:452–9.PubMedCrossRef Bonadonna RC, Groop L, Kraemer N, Ferrannini E, Del Prato S, DeFronzo RA. Obesity and insulin resistance in humans: a dose-response study. Metabolism. 1990;39:452–9.PubMedCrossRef
66.
Zurück zum Zitat Groop LC, Bonadonna RC, Simonson DC, Petrides AS, Shank M, DeFronzo RA. Effect of insulin on oxidative and nonoxidative pathways of free fatty acid metabolism in human obesity. Am J Physiol. 1992;263:E79–84.PubMed Groop LC, Bonadonna RC, Simonson DC, Petrides AS, Shank M, DeFronzo RA. Effect of insulin on oxidative and nonoxidative pathways of free fatty acid metabolism in human obesity. Am J Physiol. 1992;263:E79–84.PubMed
67.
Zurück zum Zitat Campbell PJ, Mandarino LJ, Gerich JE. Quantification of the relative impairment in actions of insulin on hepatic glucose production and peripheral glucose uptake in non-insulin-dependent diabetes mellitus. Metabolism. 1988;37:15–21.PubMedCrossRef Campbell PJ, Mandarino LJ, Gerich JE. Quantification of the relative impairment in actions of insulin on hepatic glucose production and peripheral glucose uptake in non-insulin-dependent diabetes mellitus. Metabolism. 1988;37:15–21.PubMedCrossRef
68.
Zurück zum Zitat Campbell PJ, Carlson MG, Hill JO, Nurjhan N. Regulation of free fatty acid metabolism by insulin in humans: role of lipolysis and reesterification. Am J Physiol. 1992;263:E1063–9.PubMed Campbell PJ, Carlson MG, Hill JO, Nurjhan N. Regulation of free fatty acid metabolism by insulin in humans: role of lipolysis and reesterification. Am J Physiol. 1992;263:E1063–9.PubMed
69.
Zurück zum Zitat Stumvoll M, Jacob S. Multiple sites of insulin resistance: muscle, liver and adipose tissue. Exp Clin Endocrinol Diabetes. 1999;107:107–10.PubMedCrossRef Stumvoll M, Jacob S. Multiple sites of insulin resistance: muscle, liver and adipose tissue. Exp Clin Endocrinol Diabetes. 1999;107:107–10.PubMedCrossRef
70.
Zurück zum Zitat Jacob S, Hauer B, Becker R, Artzner S, Grauer P, Loblein K, et al. Lipolysis in skeletal muscle is rapidly regulated by low physiological doses of insulin. Diabetologia. 1999;42:1171–4.PubMedCrossRef Jacob S, Hauer B, Becker R, Artzner S, Grauer P, Loblein K, et al. Lipolysis in skeletal muscle is rapidly regulated by low physiological doses of insulin. Diabetologia. 1999;42:1171–4.PubMedCrossRef
71.
Zurück zum Zitat Li C, Ford ES, McGuire LC, Mokdad AH, Little RR, Reaven GM. Trends in hyperinsulinemia among nondiabetic adults in the U.S. Diabetes Care. 2006;29:2396–402.PubMedCrossRef Li C, Ford ES, McGuire LC, Mokdad AH, Little RR, Reaven GM. Trends in hyperinsulinemia among nondiabetic adults in the U.S. Diabetes Care. 2006;29:2396–402.PubMedCrossRef
73.
Zurück zum Zitat Yeni-Komshian H, Carantoni M, Abbasi F, Reaven GM. Relationship between several surrogate estimates of insulin resistance and quantification of insulin-mediated glucose disposal in 490 healthy nondiabetic volunteers. Diabetes Care. 2000;23:171–5.PubMedCrossRef Yeni-Komshian H, Carantoni M, Abbasi F, Reaven GM. Relationship between several surrogate estimates of insulin resistance and quantification of insulin-mediated glucose disposal in 490 healthy nondiabetic volunteers. Diabetes Care. 2000;23:171–5.PubMedCrossRef
74.
Zurück zum Zitat DeFronzo RA, Ferrannini E, Hendler R, Felig P, Wahren J. Regulation of splanchnic and peripheral glucose uptake by insulin and hyperglycemia in man. Diabetes. 1983;32:35–45.PubMedCrossRef DeFronzo RA, Ferrannini E, Hendler R, Felig P, Wahren J. Regulation of splanchnic and peripheral glucose uptake by insulin and hyperglycemia in man. Diabetes. 1983;32:35–45.PubMedCrossRef
75.
Zurück zum Zitat Prager R, Wallace P, Olefsky JM. In vivo kinetics of insulin action on peripheral glucose disposal and hepatic glucose output in normal and obese subjects. J Clin Invest. 1986;78:472–81.PubMedCrossRefPubMedCentral Prager R, Wallace P, Olefsky JM. In vivo kinetics of insulin action on peripheral glucose disposal and hepatic glucose output in normal and obese subjects. J Clin Invest. 1986;78:472–81.PubMedCrossRefPubMedCentral
77.
Zurück zum Zitat Jung SH, Jung CH, Reaven GM, Kim SH. Adapting to insulin resistance in obesity: role of insulin secretion and clearance. Diabetologia. 2018;61:681–7.PubMedCrossRef Jung SH, Jung CH, Reaven GM, Kim SH. Adapting to insulin resistance in obesity: role of insulin secretion and clearance. Diabetologia. 2018;61:681–7.PubMedCrossRef
78.
Zurück zum Zitat Alemzadeh R, Karlstad MD, Tushaus K, Buchholz M. Diazoxide enhances basal metabolic rate and fat oxidation in obese Zucker rats. Metabolism. 2008;57:1597–607.PubMedCrossRef Alemzadeh R, Karlstad MD, Tushaus K, Buchholz M. Diazoxide enhances basal metabolic rate and fat oxidation in obese Zucker rats. Metabolism. 2008;57:1597–607.PubMedCrossRef
79.
Zurück zum Zitat Williams KJ, Wu X. Imbalanced insulin action in chronic over nutrition: Clinical harm, molecular mechanisms, and a way forward. Atherosclerosis. 2016;247:225–82.PubMedCrossRef Williams KJ, Wu X. Imbalanced insulin action in chronic over nutrition: Clinical harm, molecular mechanisms, and a way forward. Atherosclerosis. 2016;247:225–82.PubMedCrossRef
80.
Zurück zum Zitat Choi SM, Tucker DF, Gross DN, Easton RM, DiPilato LM, Dean AS, et al. Insulin regulates adipocyte lipolysis via an Akt-independent signaling pathway. Mol Cell Biol. 2010;30:5009–20.PubMedCrossRefPubMedCentral Choi SM, Tucker DF, Gross DN, Easton RM, DiPilato LM, Dean AS, et al. Insulin regulates adipocyte lipolysis via an Akt-independent signaling pathway. Mol Cell Biol. 2010;30:5009–20.PubMedCrossRefPubMedCentral
81.
Zurück zum Zitat Eissing L, Scherer T, Todter K, Knippschild U, Greve JW, Buurman WA, et al. De novo lipogenesis in human fat and liver is linked to ChREBP-beta and metabolic health. Nat Commun. 2013;4:1528.PubMedCrossRef Eissing L, Scherer T, Todter K, Knippschild U, Greve JW, Buurman WA, et al. De novo lipogenesis in human fat and liver is linked to ChREBP-beta and metabolic health. Nat Commun. 2013;4:1528.PubMedCrossRef
82.
Zurück zum Zitat Del Prato S, Leonetti F, Simonson DC, Sheehan P, Matsuda M, DeFronzo RA. Effect of sustained physiologic hyperinsulinaemia and hyperglycaemia on insulin secretion and insulin sensitivity in man. Diabetologia. 1994;37:1025–35.PubMedCrossRef Del Prato S, Leonetti F, Simonson DC, Sheehan P, Matsuda M, DeFronzo RA. Effect of sustained physiologic hyperinsulinaemia and hyperglycaemia on insulin secretion and insulin sensitivity in man. Diabetologia. 1994;37:1025–35.PubMedCrossRef
83.
Zurück zum Zitat Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542:177–85.PubMedCrossRef Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542:177–85.PubMedCrossRef
84.
Zurück zum Zitat Torres-Fuentes C, Schellekens H, Dinan TG, Cryan JF. The microbiota–gut–brain axis in obesity. Lancet Gastroenterol Hepatol. 2017;2:747–56.PubMedCrossRef Torres-Fuentes C, Schellekens H, Dinan TG, Cryan JF. The microbiota–gut–brain axis in obesity. Lancet Gastroenterol Hepatol. 2017;2:747–56.PubMedCrossRef
85.
Zurück zum Zitat Pan WW, Myers MG Jr. Leptin and the maintenance of elevated body weight. Nat Rev Neurosci. 2018;19:95–105.PubMedCrossRef Pan WW, Myers MG Jr. Leptin and the maintenance of elevated body weight. Nat Rev Neurosci. 2018;19:95–105.PubMedCrossRef
87.
Zurück zum Zitat Qasim A, Turcotte M, de Souza RJ, Samaan MC, Champredon D, Dushoff J, et al. On the origin of obesity: identifying the biological, environmental and cultural drivers of genetic risk among human populations. Obes Rev. 2018;19:121–49.PubMedCrossRef Qasim A, Turcotte M, de Souza RJ, Samaan MC, Champredon D, Dushoff J, et al. On the origin of obesity: identifying the biological, environmental and cultural drivers of genetic risk among human populations. Obes Rev. 2018;19:121–49.PubMedCrossRef
88.
Zurück zum Zitat Nolan CJ, Ruderman NB, Kahn SE, Pedersen O, Prentki M. Insulin resistance as a physiological defense against metabolic stress: implications for the management of subsets of type 2 diabetes. Diabetes. 2015;64:673–86.PubMedCrossRefPubMedCentral Nolan CJ, Ruderman NB, Kahn SE, Pedersen O, Prentki M. Insulin resistance as a physiological defense against metabolic stress: implications for the management of subsets of type 2 diabetes. Diabetes. 2015;64:673–86.PubMedCrossRefPubMedCentral
90.
Zurück zum Zitat Azzu V, Valencak TG. Energy metabolism and ageing in the mouse: a mini-review. Gerontology. 2017;63:327–36.PubMedCrossRef Azzu V, Valencak TG. Energy metabolism and ageing in the mouse: a mini-review. Gerontology. 2017;63:327–36.PubMedCrossRef
91.
Zurück zum Zitat Templeman NM, Flibotte S, Chik JHL, Sinha S, Lim GE, Foster LJ, et al. Reduced circulating insulin enhances insulin sensitivity in old mice and extends lifespan. Cell Rep. 2017;20:451–63.PubMedCrossRef Templeman NM, Flibotte S, Chik JHL, Sinha S, Lim GE, Foster LJ, et al. Reduced circulating insulin enhances insulin sensitivity in old mice and extends lifespan. Cell Rep. 2017;20:451–63.PubMedCrossRef
92.
Zurück zum Zitat Shadid S, Kanaley JA, Sheehan MT, Jensen MD. Basal and insulin-regulated free fatty acid and glucose metabolism in humans. Am J Physiol Endocrinol Metab. 2007;292:E1770–4.PubMedCrossRef Shadid S, Kanaley JA, Sheehan MT, Jensen MD. Basal and insulin-regulated free fatty acid and glucose metabolism in humans. Am J Physiol Endocrinol Metab. 2007;292:E1770–4.PubMedCrossRef
93.
Zurück zum Zitat Widjaja A, Morris RJ, Levy JC, Frayn KN, Manley SE, Turner RC. Within- and between-subject variation in commonly measured anthropometric and biochemical variables. Clin Chem. 1999;45:561–6.PubMed Widjaja A, Morris RJ, Levy JC, Frayn KN, Manley SE, Turner RC. Within- and between-subject variation in commonly measured anthropometric and biochemical variables. Clin Chem. 1999;45:561–6.PubMed
95.
Zurück zum Zitat Il'yasova D, Wang F, D’Agostino RB Jr, Hanley A, Wagenknecht LE. Prospective association between fasting NEFA and type 2 diabetes: impact of post-load glucose. Diabetologia. 2010;53:866–74.PubMedCrossRefPubMedCentral Il'yasova D, Wang F, D’Agostino RB Jr, Hanley A, Wagenknecht LE. Prospective association between fasting NEFA and type 2 diabetes: impact of post-load glucose. Diabetologia. 2010;53:866–74.PubMedCrossRefPubMedCentral
96.
Zurück zum Zitat Byrne CD, Maison P, Halsall D, Martensz N, Hales CN, Wareham NJ. Cross-sectional but not longitudinal associations between non-esterified fatty acid levels and glucose intolerance and other features of the metabolic syndrome. Diabet Med. 1999;16:1007–15.PubMedCrossRef Byrne CD, Maison P, Halsall D, Martensz N, Hales CN, Wareham NJ. Cross-sectional but not longitudinal associations between non-esterified fatty acid levels and glucose intolerance and other features of the metabolic syndrome. Diabet Med. 1999;16:1007–15.PubMedCrossRef
97.
Zurück zum Zitat Finucane FM, Sharp SJ, Hatunic M, Sleigh A, De Lucia RE, Sayer AA, et al. Intrahepatic lipid content and insulin resistance are more strongly associated with impaired NEFA suppression after oral glucose loading than with fasting NEFA levels in healthy older individuals. Int J Endocrinol. 2013;2013:870487.PubMedCrossRefPubMedCentral Finucane FM, Sharp SJ, Hatunic M, Sleigh A, De Lucia RE, Sayer AA, et al. Intrahepatic lipid content and insulin resistance are more strongly associated with impaired NEFA suppression after oral glucose loading than with fasting NEFA levels in healthy older individuals. Int J Endocrinol. 2013;2013:870487.PubMedCrossRefPubMedCentral
98.
Zurück zum Zitat Jocken JW, Langin D, Smit E, Saris WH, Valle C, Hul GB, et al. Adipose triglyceride lipase and hormone-sensitive lipase protein expression is decreased in the obese insulin-resistant state. J Clin Endocrinol Metab. 2007;92:2292–9.PubMedCrossRef Jocken JW, Langin D, Smit E, Saris WH, Valle C, Hul GB, et al. Adipose triglyceride lipase and hormone-sensitive lipase protein expression is decreased in the obese insulin-resistant state. J Clin Endocrinol Metab. 2007;92:2292–9.PubMedCrossRef
99.
Zurück zum Zitat Clayton DJ, Burrell K, Mynott G, Creese M, Skidmore N, Stensel DJ, et al. Effect of 24-h severe energy restriction on appetite regulation and ad libitum energy intake in lean men and women. Am J Clin Nutr. 2016;104:1545–53.PubMedCrossRef Clayton DJ, Burrell K, Mynott G, Creese M, Skidmore N, Stensel DJ, et al. Effect of 24-h severe energy restriction on appetite regulation and ad libitum energy intake in lean men and women. Am J Clin Nutr. 2016;104:1545–53.PubMedCrossRef
100.
Zurück zum Zitat Lim EL, Hollingsworth KG, Aribisala BS, Chen MJ, Mathers JC, Taylor R. Reversal of type 2 diabetes: normalisation of beta cell function in association with decreased pancreas and liver triacylglycerol. Diabetologia. 2011;54:2506–14.PubMedCrossRefPubMedCentral Lim EL, Hollingsworth KG, Aribisala BS, Chen MJ, Mathers JC, Taylor R. Reversal of type 2 diabetes: normalisation of beta cell function in association with decreased pancreas and liver triacylglycerol. Diabetologia. 2011;54:2506–14.PubMedCrossRefPubMedCentral
101.
Zurück zum Zitat Carswell KA, Belgaumkar AP, Amiel SA, Patel AG. A systematic review and meta-analysis of the effect of gastric bypass surgery on plasma lipid levels. Obes Surg. 2016;26:843–55.PubMedCrossRef Carswell KA, Belgaumkar AP, Amiel SA, Patel AG. A systematic review and meta-analysis of the effect of gastric bypass surgery on plasma lipid levels. Obes Surg. 2016;26:843–55.PubMedCrossRef
102.
Zurück zum Zitat Corkey BE. Banting lecture 2011: hyperinsulinemia: cause or consequence? Diabetes. 2012;61:4–13.PubMedCrossRef Corkey BE. Banting lecture 2011: hyperinsulinemia: cause or consequence? Diabetes. 2012;61:4–13.PubMedCrossRef
104.
Zurück zum Zitat Valachovicova M, Krajcovicova-Kudlackova M, Blazicek P, Babinska K. No evidence of insulin resistance in normal weight vegetarians. A case control study. Eur J Nutr. 2006;45:52–4.PubMedCrossRef Valachovicova M, Krajcovicova-Kudlackova M, Blazicek P, Babinska K. No evidence of insulin resistance in normal weight vegetarians. A case control study. Eur J Nutr. 2006;45:52–4.PubMedCrossRef
106.
Zurück zum Zitat Pories WJ, Dohm GL. Diabetes: have we got it all wrong? Hyperinsulinism as the culprit: surgery provides the evidence. Diabetes Care. 2012;35:2438–42.PubMedCrossRefPubMedCentral Pories WJ, Dohm GL. Diabetes: have we got it all wrong? Hyperinsulinism as the culprit: surgery provides the evidence. Diabetes Care. 2012;35:2438–42.PubMedCrossRefPubMedCentral
107.
Zurück zum Zitat Saad A, Dalla MC, Nandy DK, Levine JA, Bharucha AE, Rizza RA, et al. Diurnal pattern to insulin secretion and insulin action in healthy individuals. Diabetes. 2012;61:2691–700.PubMedCrossRefPubMedCentral Saad A, Dalla MC, Nandy DK, Levine JA, Bharucha AE, Rizza RA, et al. Diurnal pattern to insulin secretion and insulin action in healthy individuals. Diabetes. 2012;61:2691–700.PubMedCrossRefPubMedCentral
108.
Zurück zum Zitat Basse AL, Dalbram E, Larsson L, Gerhart-Hines Z, Zierath JR, Treebak JT. Skeletal muscle insulin sensitivity show circadian rhythmicity which is independent of exercise training status. Front Physiol. 2018;9:1198.PubMedCrossRefPubMedCentral Basse AL, Dalbram E, Larsson L, Gerhart-Hines Z, Zierath JR, Treebak JT. Skeletal muscle insulin sensitivity show circadian rhythmicity which is independent of exercise training status. Front Physiol. 2018;9:1198.PubMedCrossRefPubMedCentral
109.
Zurück zum Zitat Nas A, Mirza N, Hagele F, Kahlhofer J, Keller J, Rising R, et al. Impact of breakfast skipping compared with dinner skipping on regulation of energy balance and metabolic risk. Am J Clin Nutr. 2017;105:1351–61.PubMed Nas A, Mirza N, Hagele F, Kahlhofer J, Keller J, Rising R, et al. Impact of breakfast skipping compared with dinner skipping on regulation of energy balance and metabolic risk. Am J Clin Nutr. 2017;105:1351–61.PubMed
111.
Zurück zum Zitat Garnett SP, Srinivasan S, Birt SG, Ambler GR, Lawrie EA, Cowell CT, et al. Evaluation of glycaemic status in young people with clinical insulin resistance; fasting glucose, fasting insulin or an oral glucose tolerance test? Clin Endocrinol. 2010;72:475–80.CrossRef Garnett SP, Srinivasan S, Birt SG, Ambler GR, Lawrie EA, Cowell CT, et al. Evaluation of glycaemic status in young people with clinical insulin resistance; fasting glucose, fasting insulin or an oral glucose tolerance test? Clin Endocrinol. 2010;72:475–80.CrossRef
112.
113.
Zurück zum Zitat Bonadonna RC, Groop LC, Zych K, Shank M, DeFronzo RA. Dose-dependent effect of insulin on plasma free fatty acid turnover and oxidation in humans. Am J Physiol. 1990;259:E736–50.PubMed Bonadonna RC, Groop LC, Zych K, Shank M, DeFronzo RA. Dose-dependent effect of insulin on plasma free fatty acid turnover and oxidation in humans. Am J Physiol. 1990;259:E736–50.PubMed
114.
Zurück zum Zitat Iggman D, Rosqvist F, Larsson A, Arnlov J, Beckman L, Rudling M, et al. Role of dietary fats in modulating cardiometabolic risk during moderate weight gain: a randomized double-blind overfeeding trial (LIPOGAIN study). J Am Heart Assoc. 2014;3:e001095.PubMedCrossRefPubMedCentral Iggman D, Rosqvist F, Larsson A, Arnlov J, Beckman L, Rudling M, et al. Role of dietary fats in modulating cardiometabolic risk during moderate weight gain: a randomized double-blind overfeeding trial (LIPOGAIN study). J Am Heart Assoc. 2014;3:e001095.PubMedCrossRefPubMedCentral
115.
Zurück zum Zitat Nilsson A, Bergens O, Kadi F. Physical activity alters inflammation in older adults by different intensity levels. Med Sci Sports Exerc. 2018;50:1502–7.PubMedCrossRef Nilsson A, Bergens O, Kadi F. Physical activity alters inflammation in older adults by different intensity levels. Med Sci Sports Exerc. 2018;50:1502–7.PubMedCrossRef
116.
Zurück zum Zitat Jiang S, Bo L, Gong C, Du X, Kan H, Xie Y, et al. Traffic-related air pollution is associated with cardio-metabolic biomarkers in general residents. Int Arch Occup Environ Health. 2016;89:911–21.PubMedCrossRef Jiang S, Bo L, Gong C, Du X, Kan H, Xie Y, et al. Traffic-related air pollution is associated with cardio-metabolic biomarkers in general residents. Int Arch Occup Environ Health. 2016;89:911–21.PubMedCrossRef
117.
Zurück zum Zitat Cai Y, Hansell AL, Blangiardo M, Burton PR, de Hoogh K, Doiron D, et al. Long-term exposure to road traffic noise, ambient air pollution, and cardiovascular risk factors in the HUNT and lifelines cohorts. Eur Heart J. 2017;38:2290–6.PubMedCrossRefPubMedCentral Cai Y, Hansell AL, Blangiardo M, Burton PR, de Hoogh K, Doiron D, et al. Long-term exposure to road traffic noise, ambient air pollution, and cardiovascular risk factors in the HUNT and lifelines cohorts. Eur Heart J. 2017;38:2290–6.PubMedCrossRefPubMedCentral
Metadaten
Titel
Insulin translates unfavourable lifestyle into obesity
verfasst von
Hubert Kolb
Michael Stumvoll
Werner Kramer
Kerstin Kempf
Stephan Martin
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2018
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-018-1225-1

Weitere Artikel der Ausgabe 1/2018

BMC Medicine 1/2018 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 24 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Update Allgemeinmedizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.