Skip to main content
Erschienen in: Diabetes Therapy 4/2022

Open Access 10.03.2022 | Review

Could Dietary Modification Independent of Energy Balance Influence the Underlying Pathophysiology of Type 2 Diabetes? Implications for Type 2 Diabetes Remission

verfasst von: Nicola D. Guess

Erschienen in: Diabetes Therapy | Ausgabe 4/2022

Abstract

High-quality clinical trial data demonstrate that remission is possible for people living with type 2 diabetes (T2D) if they lose a large amount of weight (≥ 10 kg). Durable remission appears predicated on the long-term maintenance of weight loss. Unfortunately, long-term follow-up data from lifestyle-based weight loss programmes show that, on average, most people regain at least some of the weight lost. In addition, restoration of a diminished first-phase insulin response also appears necessary for durable remission, and this becomes less likely as T2D progresses. A pragmatic approach to enhance the effects of weight loss on durable remission is to consider whether dietary components could help control blood glucose, independent of caloric balance. This manuscript reviews current evidence on weight-neutral effects of diet on blood glucose, including high-protein, low-carbohydrate, high-fibre and plant-based diets, with a particular focus on the effect of nutrition on the underlying pathophysiology of T2D, including the first-phase insulin response. The importance of mechanistic data in enhancing our understanding of dietary strategies in T2D remission is described, and suggestions are made for future advances in remission research.
Key Summary Points
Remission of type 2 diabetes (T2D) is now possible for many people living with T2D if they are able to lose and maintain > 10% weight loss and have T2D of short duration.
This excludes many people if they do not want to, or are unable to lose and maintain that much weight loss. It also potentially excludes people who have T2D of longer duration. Dietary strategies which could help lower glycaemia independent of weight loss could help more people achieve normalisation of glucose concentrations.
This manuscript reviews the evidence for different weight-neutral dietary strategies to help support optimal management of T2D and describes a way forward for remission research.

Introduction

The aim of type 2 diabetes (T2D) management is to control cardiovascular (CV) risk factors, including glucose, altered lipid profile and blood pressure. The pinnacle of T2D management can be considered remission [1]—the normalisation or achievement of blood glucose concentrations under the diabetic threshold in the absence of hypoglycaemic medications, which is often associated with reduction in blood pressure and improvement in the lipid profile [1, 2].
It is currently accepted that remission can be achieved via marked weight loss following caloric restriction achieved through diet or bariatric surgery [1, 3]. In the UK, a total diet replacement approach used in the seminal Diabetes Remission Clinical Trial (DiRECT) is being piloted across 12 sites in the UK [4]. While the evidence shows that body weight (an imperfect proxy for adiposity) is the most important factor in T2D development [5], remission [6] and relapse [6], other dietary strategies could help lower blood glucose [7, 8] and manage some T2D risk factors [9, 10] in ways that could be independent of energy balance. The use of such strategies could be a useful addition to T2D remission programmes, but also to prevention programmes.
It is important to note that dietary interventions often require an individual to make changes to nearly all aspects of their lives—from shopping, to cooking, to eating—and that these changes may need to be continued for most meals that person eats. For these and other reasons, no one diet will ever be appropriate for everyone, regardless of whether it is physiologically efficacious or not. Nevertheless, a better quality evidence base which can allow us to make more precise statements about what nutrients and diets can and cannot do will enable us to inform our patients of what works, how it works and possibly in whom it works.
The dearth of mechanistic and well-controlled studies in individuals with T2D to date mean that we are largely unable to give patients more specific guidance than “lose weight in whatever way you are able to sustain” [11]. Even the most up-to-date guidance for T2D is based on free-living weight loss interventions which use different dietary patterns to achieve this weight loss. As such, recommendations for specific dietary strategies are understandably weak [12]. As I described in this review, there is growing evidence that there are dietary strategies which can indeed influence key aspects of T2D physiology and lower blood glucose independent of caloric balance.
This manuscript will examine how dietary strategies could influence the underlying pathophysiology of T2D beyond caloric restriction and describe a way forward for T2D remission research. Since this article is based on previously conducted studies and does not contain any new studies with human participants, ethical approval was not sought.

History of Diet-Induced Medication Withdrawal/Remission in T2D

Following observations from bariatric surgery that normalisation of blood glucose and medication withdrawal was possible in patients with T2D before any significant weight loss had occurred, multiple investigative groups examined the independent effect of marked caloric restriction alone on T2D pathophysiology.
A series of mechanistic physiological studies [1317] demonstrated that marked energy restriction (intake of 400–800 kcal/day) over the short term (2 days to 8 weeks) favourably alters key aspects of T2D pathophysiology and leads to substantial reductions in or even normalisation of blood glucose. However, in all of these studies, the follow-up tests were carried out immediately following the energy restriction, and the durability of these glucoregulatory improvements was unknown [18]. Further research demonstrated that the remission achieved with marked weight loss (in the order of 10–15 kg) can be maintained up to 2 years in a large proportion of people, provided weight regain is limited or prevented [2, 6, 19].
Another key aspect of our understanding of remission is that durable remission achieved with diet-induced marked weight loss is dependent upon the return of a key aspect of beta-cell function, called the first-phase insulin response [19, 20]. This marked post-meal insulin spike is critical physiologically to suppress hepatic glucose output and lipolysis of peripheral adipose tissue, and to promote the uptake of glucose and fatty acids into skeletal muscle [21]. The first-phase insulin response declines over the course of T2D, and data from lifestyle trials and bariatric surgery consistently show an inverse relationship between duration of T2D and likelihood of remission [1921].
In summary, our current understanding is that remission of T2D via diet can be achieved contingent on: (1) the return of the first-phase insulin response, which itself is reliant on achieving and sustaining significant weight loss, and (2) T2D being of relatively short duration.
The next sections review how other dietary strategies could optimise T2D remission programmes, with a focus on how diet affects the underlying physiology of the disease. The discussion points are summarised in Table 1 and in Fig. 1.
Table 1
Summary of the emerging evidence for the effect of dietary components on glucose and the physiological drivers underlying glucose concentrations in type 2 diabetes independent of calorie restriction
Dietary components
First-phase/early insulin response
Hepatic glucose output
Increase glucose uptake
Glucose
Increasing dietary protein
Increases
No acute effect, long-term effect unknown
Conflicting data
Lowers, may need to be combined with carbohydrate restriction
Reducing dietary carbohydrate
Unknown
May reduce output, but likely depends on degree of reduction
Unknown—likely depends on the degree of reduction
Unknown—likely depends on the degree of reduction
Increasing dietary ketones
Unknown
Reduces
Unknown
Lowers
Increasing fibre
Increasesa
Reducesa
Increasesa
Reducesa
Increasing polyunsaturated fatty acids:saturated fatty acids ratio
Unknown
Reduces acutely
Unknown
No effect
Increasing plant-based components eg phytonutrients
Unknown
Unknown
Unknown
Unknown
aDepends on the physicochemical properties of the fibres, and the dose. Large differences in fibre are needed to see a significant effect

High Protein

In healthy individuals, the pancreas is able to precisely control blood glucose by releasing just the right amount of insulin required to keep blood glucose within a narrow range [22]. One of the most important qualitative properties of the insulin secretory response is the ability of the beta-cells [22] to respond to the rapid rises in blood glucose following a carbohydrate load by acutely and rapidly increasing the amount of insulin released [21]. In clinical terms, this “insulin spike” is referred to as the “early” or “first-phase” insulin response [21, 23].
The physiological importance of the first-phase insulin response is demonstrated by studies in which its experimental reduction results in impaired suppression of hepatic glucose output, hyperglucongaemia, post-prandial lipaemia, higher maximal glucose concentrations and prolonged hyperglycaemia which may last for several hours [21, 24, 25]. Conversely, experimental restoration of the absent early insulin response in T2D normalises glucose tolerance without increasing the overall insulin demand [21, 25].
Protein has long been known to potentiate the insulin response to a carbohydrate load [26], such that increasing the amount of protein in a meal acutely can more than double the amount of insulin produced [2729], and lead to a reduction in post-prandial glucose concentrations [26, 30, 31]. This insulinogenic response likely occurs via direct stimulation of amino acids on the beta-cell [32, 33] and an enhanced incretin effect [34].
Of particular relevance to T2D, while glucose-stimulated insulin secretion declines markedly as T2D progresses, amino acid-stimulated insulin secretion appears to remain relatively intact [35]. In fact, the insulinotrophic effect of protein may be greater in people with T2D [28, 29] than in people with normal glucose tolerance. Given that a potent prandial insulin response is necessary for optimal control of post-prandial glucose concentrations [20, 25], and that the glucose-stimulated insulin response declines in T2D [21], this raises the question of whether increasing the amount of protein in the diet could help lower blood glucose concentrations, independent of weight loss.
To date, five well-controlled studies [8, 10, 3638] by two independent research groups have shown that increasing the protein content of the diet from 15% to 30% kcal lowers blood glucose in T2D. These studies carefully controlled body weight, provided full food provision throughout each dietary intervention and measured multiple parameters of glycaemic control. The reduction in glucose is clinically significant: up to a reduction of 2–5 mmol/L during the 4-h period after a meal. In each of these studies the carbohydrate was also reduced and represented between 20 and 40%kcal. Therefore, the relative contribution of increased protein and carbohydrate restriction to the improvements in glycaemia are unknown. It is also important to reference studies [39, 40] which tested a similar dietary composition and did not find any superiority on glucose lowering while also noting that these studies did not provide the dietary protocol or control the confounding variables, including body weight.
As convincingly shown by the DiRECT trial investigators [2], marked weight loss can restore the first-phase insulin response in people with T2D of short duration (< 4 and < 6 years)[19, 20]. However, therapeutically it is important to understand if there are other ways to restore the early insulin response that could be effective in people with T2D of longer duration, or without necessarily requiring such marked weight loss. If the beta-cells are able to produce a large post-prandial insulin response to amino acids, this could be a clinically useful strategy to help manage glycaemia in T2D of long duration.
With respect to the effect of additional protein on other underlying drivers of glucose homeostasis, evidence suggests high intakes of protein do not increase hepatic glucose output acutely [41], but the longer-term impact is not known. There is conflicting data on whether high-protein diets influence peripheral glucose uptake [42, 43]. However, the reduction in blood glucose with a high-protein (alongside a reduced-carbohydrate) diet is large enough that if there are changes in hepatic glucose output or glucose uptake, these may not be significant enough to counteract the glucose-lowering effect of the enhanced insulin secretion.

Carbohydrate Restriction

There has been immense interest in the role of a low-carbohydrate diet on glycaemia because carbohydrate is the macronutrient with the greatest effect on acute glycaemia [44]. Since the greater the glycaemic load of a meal, the greater the post-prandial glucose rise, it has become conventional wisdom that simply reducing the carbohydrate load of the diet will lower blood glucose concentrations. However, this is an oversimplication and ignores physiological changes which occur in the body in response to a reduction in carbohydrate intake, including an increase in gluconeogenesis [45] and possibly the development of insulin resistance [46] and down-regulation of insulin secretion [47]. In fact, some of the earliest studies in T2D show that increasing the amount of carbohydrate in the diet lowers plasma glucose concentrations [48]. Equally, while carbohydrate restriction may alter insulin signalling in some tissues, the net effect on long-term CV risk of an increase in muscle-specific insulin resistance when considered against other homeostatic counter-alterations in glucose homeostasis is unclear [49]. In fact, some studies show improvement in classical CV risk factors with a low-carbohydrate diet [50, 51]. Thus, whether and how physiological changes occur in T2D in response to varying degrees of carbohydrate intake has been largely unexplored [52].
Despite the existence of multiple meta-analyses of trials investigating the effect of dietary carbohydrate restriction on glycaemia in T2D [53, 54], it is important to note that the heterogeneity of these trials makes them very difficult to compare. The trials are confounded by differences in weight loss, protein, fibre, fat type and the presence, type and dose of diabetes medications both within the interventions themselves and between the intervention and control groups. Nevertheless, there is scientific data available which provide insight into the potential role of carbohydrate restriction per se on glycaemia, and this will now be reviewed.
While moderate carbohydrate restriction—independent of protein [36, 55, 56] or weight loss—may not alter glucose concentrations in T2D [57, 58], there is evidence that reducing the carbohydrate intake to levels likely to increase ketogenesis does have weight-independent effects on glucose [45, 59].
In a randomised crossover trial which compared two very low-energy weight loss diets in patients with obesity and T2D that were carefully matched for calorie and protein content, a diet containing 28% kcal from carbohydrate lowered fasting and post-prandial glucose to a greater extent than the diet with 78% kcal carbohydrate [60]. The plasma ketones were 1–2 mmol/L higher in the 28% kcal carbohydrate diet than in the 78% kcal diet, and a significant relationship was observed between plasma ketones and hepatic glucose output. These results support data from experimental physiological studies showing a suppressive effect of plasma ketones on endogenous glucose production and glucose concentrations in people with and without T2D [6163], as well as data showing that a 0% kcal (89% kcal from fat, 11% kcal from protein) carbohydrate diet suppresses glucose appearance to a greater extent compared to an 89% kcal carbohydrate diet (0% kcal from fat, 11% kcal from protein) in T2D [45].
Endogenous glucose production (EGP) is the formation of glucose from non-glucose precursors. EGP prevents hypoglycaemia from occurring during the fasting and interprandial periods. In healthy people, EGP can be suppressed for several hours following the consumption of even a small amount of carbohydrate (e.g. 25 g) [64]. In T2D, EGP is elevated in the fasting state, and its suppression can be impaired following a meal [65], leading to relative hyperglycaemia in the fed and fasting states.
The effect of nutrients per se on EGP may be particularly important because weight loss-induced suppression of EGP may not be durable in the long term: it is known that a reduction in fasting glucose during energy restriction in T2D occurs within the first week [15] and that this is closely correlated to the reduced EGP that can be achieved with caloric restriction [13, 15, 66]. Unfortunately, once a patient returns to a eucaloric diet (as they inevitably must do at some point and which usually occurs in practice after 6–12 months), EGP returns to its baseline rate, even before any weight gain occurs [67]. Christiansen and colleagues even found that the suppression of EGP with caloric restriction is transient only and that it returns to its high baseline rate even during a hypocaloric diet [67].
Weight loss-induced suppression of EGP may be maintained following extreme weight loss (> 50% of excess body weight, approx. 20 kg) [68], but this magnitude of weight loss is difficult to achieve with diet. EGP continued to be suppressed in a cohort of patients with T2D and obesity who had lost a mean of 20 kg over an average of 17 weeks (range: 4–35 weeks). However, the follow-up tests were taken as soon as patient reached their 50% excess weight loss target and therefore may have still been measured in a calorie-deficit state.
In summary, dietary approaches which could suppress endogenous glucose production independent of caloric balance may help enhance durable glycaemic control, and this may be particularly critical as the patient moves from weight loss to weight maintenance.

Whole-Food, Plant-Based Diet

Dietary patterns (Mediterranean, vegetarian etc.) recommended for the management of T2D have in common the inclusion of whole, intact foods from plant-based sources. While these type of diets are also associated with weight loss, there is good evidence they have beneficial effects independent of their energy density. Components of these diets for which there is evidence for weight-neutral effects on glycaemic control include fibre, polyphenolic compounds and a high proportion of saturated versus unsaturated fats.

Fibre

Dietary fibres are a group of carbohydrates which cannot be digested and absorbed in the upper gastrointestinal tract. Various factors contribute to the digestibility of carbohydrates, including the type of starch (amylose or amylopectin), rate of digestion, cooking or processing method and fibre content [69]. If the structure of the carbohydrate is such that the rate or degree of digestion is slowed, the carbohydrate is less available/“glycaemic”. For example, the open structure and absence of fibre secondary to the processing used in the manufacture of white rice renders the glucose monomers rapidly and fully available. Conversely, the rigid and tight structure secondary to the retrogradation of the starch in pasta which has been cooked and cooled makes this glucose less readily available.
The concept of the glycaemic index has formed part of T2D management for decades [70], but the effect size of a low versus high glycaemic index diet in trials is fairly modest (glycated haemoglobin [HbA1c] approx. 0.4–0.5% [4–6 mmol/mol]) [71, 72]. This suggests that “extraglycaemic” properties of carbohydrate play an important role in the relationship between indigestible carbohydrates and T2D control.
Of particular relevance to T2D may be fermentability. Indigestible fibres pass intact to the colon where they are fermented by resident gut bacteria. The products of fermentation include the short-chain fatty acids (SCFA) acetate, butyrate and propionate. These SCFA are ligands for receptors located on L cells within the colon [73, 74] that secrete enteroendocrine hormones, including glucagon-like peptide-1 (GLP-1), which augments glucose-simulated insulin secretion, and peptide YY (PYY), which slows gastric emptying. Despite promising results from animal studies, the data currently available do not suggest a significant effect of fermentable carbohydrate consumption (at 10–35 g/day intakes) on GLP-1 secretion in humans ([75, 76]. Nevertheless, a number of studies using both oral and intravenous measures of insulin secretion suggest that fermentable carbohydrates could help increase the early insulin response in people with and without T2D [77, 78], possibly secondary to a direct effect of SCFA on the beta-cell [79].
Some soluble fibres such as guar gum possess two separate physicochemical properties which could theoretically be of benefit in T2D: their viscosity helps form a gel which slows glucose absorption via the gastrointestinal tract, and the intact fibres are fermentable in the colon. Despite the theoretical promise of such fibres, trials do not consistently show reductions in glucose concentrations in T2D, and again, any effect is small in magnitude [80, 81].
In summary, the effects of individual fibres on glucose concentration appears to be small, and large differences in intakes are probably required to achieve a clinically relevant reduction in glucose concentrations [8286]. Since the physiological effects of these different fibres on glucose homeostasis are distinct, it is perhaps not surprising that combining them might achieve larger reductions in plasma glucose. For example, a low-glycaemic, legume-rich diet with approximately 60 g/day fibre can lower glucose by > 2 mmol/L even compared to a reduced-carbohydrate, high-monounsaturated-fat diet [87].

Non-Nutritive Compounds

Foods which are naturally high in dietary fibre also contain multiple compounds which themselves may help lower glycaemia. For example, bioactive proteins, polyphenolic compounds and other phytochemicals [88] found in fruits, vegetables and legumes have varying effects on glucose metabolism, including inhibiting carbohydrate digestive enzymes, although again the effect size of even concentrates of these compounds when tested in human trials is modest (< 1 mmol/L) [89, 90].

Fatty Acid Composition

Plant-based dietary patterns also tend to be higher in unsaturated fats, including linoleic acid and oleic acid. These fats, and the foods high in them, improve insulin sensitivity when they replace an isocaloric quantity of saturated fat, such as from butter [9193]. Again, it is important to note that these improvements in insulin sensitivity do not necessarily translate into clinically significant reductions in glucose [94], and large differences in intakes of unsaturated versus saturated fat may be needed to observe a difference.

Plant-Based Dietary Patterns

To summarise, it is unlikely that any single element of a plant-based diet can lower glucose on its own to a magnitude which could influence the achievement of remission. However, here it is important to note that some of the proposed mechanisms of effect of the glycaemic index, soluble and fermentable fibres, polyphenolic compounds and unsaturated:saturated fat ratio on glucose homeostasis are distinct. Thus, it might be reasonable to expect that the effects of these foods combined on any given outcome would be additive. If each approach lowers glucose by approximately 0.7 mmol/L, the overall effect of a diet high in plant-based foods independent of weight change could theoretically reach > 3 mmol/L.

Why Mechanistic Research Matters

Understanding the mechanism of how diet affects glycaemia (and other risk factors) in T2D is important because if a number of dietary changes each influence insulin sensitivity, EGP or insulin secretion via separate mechanisms, their combined effect could then be additive. For example, if a plant-based diet was formulated to also be a high-protein diet, the complementary mechanisms (improved insulin sensitivity, reduced glucose absorption, low glycaemic load of the plant-based diet vs. insulinogenic effects of amino acids) of each of these two dietary approaches could lead to a greater reduction in glucose than each approach alone.
Similarly, understanding how nutrients affect the underlying pathophysiology of disease is important in designing dietary trials. If amino acids stimulate insulin secretion via the same molecular pathways as a sulfonylurea, those participants receiving this medication in a trial may get no benefit from a high-protein diet. Similarly, it is interesting to note that in two inpatient studies, the same low-carbohydrate, high-monounsaturated-fat dietary intervention lowered glucose in people with T2D on insulin [95], but had no effect in diet-controlled T2D [96]. These methodological nuances represent significant barriers in our current understanding of dietary management of T2D.
In addition, understanding the mechanism of effect of foods also enables a dietitian or healthcare practitioner to tailor physiologically effective dietary recommendations to an individual based on what foods they like and can access and afford. This empowers the dietitian to provide guidance beyond the typical advice to reduce portion size in order to reduce calorie intake. This could include simple swaps, such as replacing rice with lentils, or butter with rapeseed oil, or choosing a high-protein snack, such as a yoghurt with fruit, etc.
It might be remarked that changing the macronutrient content of the diet is as challenging as losing weight. However, one of the reasons long-term weight loss maintenance is difficult is because of physiological adaptations to a caloric restriction and weight loss [97, 98]. The same is not true of macronutrient changes. In trials, some people do revert to their normal dietary patterns [99], although not always [100], and within those averages are individuals who are able to maintain dietary changes long-term.
In addition, there have been advances in the development of tools which help maintain behaviour change, including the use of apps to provide people with more intensive support and data-driven algorithms to provide personalised guidance on simple dietary switches. These developments are in their infancy, but the use of these tools could support patients in making more pronounced changes in dietary intake compared to the traditional model of care. Examples of such innovations include the Food4Me study, which used data on dietary intake collected at baseline to generate personalised recommendations for specific dietary switches for each person to make [101], and the Virta programme, which uses biofeedback to encourage patients with T2D to stick to a ketogenic diet [102].
The development of functional foods which have a lower carbohydrate or higher protein and fibre content can also be utilised to help people modify their diets to one which can more effectively help manage their diabetes.
The challenge we have currently is that we do not have data of sufficient quality to provide guidance on precisely which changes might be physiologically effective for people who are able to make larger shifts in their dietary intake. To obtain these data, the nutrition science community needs to focus on trials with greater internal validity.

Advancing Remission of T2D

Some people would argue that we know all we need to know about how to achieve remission of T2D: lose and maintain > 10 kg in body weight. The excellent DiRECT trial showed this can be achieved in primary care [2]. Nevertheless, people who have worked so hard to lose weight do, on average, tend to regain weight regardless of which dietary approach they use [103], even in situations where they receive long-term, high-intensity specialised support [6, 104].
Since the regain of weight is, on average, inevitable and is associated with T2D relapse [6, 105], modifying the diet in such a way as to lower glucose independent of weight change seems a pragmatic approach to try and help people maintain durable glycaemic control. Moreover, some of the physiological mechanisms underlying relapse are beginning to be understood [105], which will help us design dietary and lifestyle-based remission programmes aimed at mitigating these pathophysiological responses to weight regain.
Furthermore, remission becomes much less likely if only modest weight loss (< 10 kg) is achieved [6]. There is an urgent need to develop T2D remission interventions which do not depend on achieving and maintaining > 10 kg weight loss largely because most people find it very difficult to lose and maintain > 10 kg of weight. Only in the most intensive weight loss trials does mean weight loss reach approximately 10 kg, and > 25% of people either do not lose weight or will gain weight across weight loss trials [4, 7, 8]. In addition, it is worth noting that T2D prevalence is highest (15%) in people aged > 60 years [12, 13] and that 7% of people with a body mass index (BMI) < 27 kg/m2 have T2D [13][13]. Many of these older individuals, particularly those with a lower BMI may not feel comfortable losing as much as 10 kg. Therefore, modifying the diet using some of the strategies described in the preceding sections could help augment the effect of more feasible weight loss on glycaemia and other CV disease risk factors. It is worth noting that aspects of the dietary patterns reviewed in this manuscript have also been shown to reduce blood pressure [106, 107], lower liver fat [10, 108] and lower blood lipids independent of weight change. This is important because, of course, lowering glucose while raising apolipoprotein B-containing cholesterol may increase the risk of heart failure in patients who who are already at high risk of CV disease.

Conclusion

As described in this narrative review, there is evidence that elements of diets can meaningfully influence blood glucose and T2D management independent of their effect on body weight. These changes could help more people achieve remission of their T2D, both as an adjunct to traditional weight loss programmes and even as stand-alone glucose-lowering approaches. Given the incidence of weight regain following weight loss interventions, leveraging the weight-neutral effects of diet on T2D is a pragmatic approach to mitigate the pathophysiological consequences of weight regain. A greater understanding of how diet influences blood glucose can help us devise personalised diets that exploit these mechanistic effect of nutrients while meeting the tastes, preferences and needs of the patient.

Acknowledgements

Funding

No funding or sponsorship was received for this study or publication of this article.

Authorship

The named author meets the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, takes responsibility for the integrity of the work as a whole, and has given approval for this version to be published.

Author Contributions

Nicola Guess is the sole author and is responsible for the concept, drafting and refining the man-uscript.

Disclosures

NG has received research funding from the Diabetes Research and Wellness Foundation to research high-protein diets and has received consultancy fees for Fixing Dad (a low carbohydrate app) and Diet Doctor (a low-carbohydrate diet website).

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.
Literatur
1.
Zurück zum Zitat Nagi D, Hambling C, Taylor R. Remission of type 2 diabetes: a position statement from the Association of British Clinical Diabetologists (ABCD) and the Primary Care Diabetes Society (PCDS). Brit J Diabetes. 2019;19(1):73–6.CrossRef Nagi D, Hambling C, Taylor R. Remission of type 2 diabetes: a position statement from the Association of British Clinical Diabetologists (ABCD) and the Primary Care Diabetes Society (PCDS). Brit J Diabetes. 2019;19(1):73–6.CrossRef
2.
Zurück zum Zitat Lean ME, Leslie WS, Barnes AC et al. Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, cluster-randomised trial. Lancet. 2018;391(10120):541–51.PubMedCrossRef Lean ME, Leslie WS, Barnes AC et al. Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, cluster-randomised trial. Lancet. 2018;391(10120):541–51.PubMedCrossRef
4.
Zurück zum Zitat Feinmann J. Type 2 diabetes: 5000 patients to test feasibility of “remission service.” BMJ. 2018;363:k5114.PubMedCrossRef Feinmann J. Type 2 diabetes: 5000 patients to test feasibility of “remission service.” BMJ. 2018;363:k5114.PubMedCrossRef
5.
Zurück zum Zitat Eckel RH, Kahn SE, Ferrannini E et al. Obesity and type 2 diabetes: what can be unified and what needs to be individualized? J Clin Endocrinol Metab. 2011;96(6):1654–63.PubMedPubMedCentralCrossRef Eckel RH, Kahn SE, Ferrannini E et al. Obesity and type 2 diabetes: what can be unified and what needs to be individualized? J Clin Endocrinol Metab. 2011;96(6):1654–63.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Lean MEJ, Leslie WS, Barnes AC et al. Durability of a primary care-led weight-management intervention for remission of type 2 diabetes: 2-year results of the DiRECT open-label, cluster-randomised trial. Lancet Diabetes Endocrinol. 2019;7(5):344–55.PubMedCrossRef Lean MEJ, Leslie WS, Barnes AC et al. Durability of a primary care-led weight-management intervention for remission of type 2 diabetes: 2-year results of the DiRECT open-label, cluster-randomised trial. Lancet Diabetes Endocrinol. 2019;7(5):344–55.PubMedCrossRef
7.
Zurück zum Zitat Simpson HCR, Carter RD, Lousley S, Mann JI. Digestible carbohydrate—an independent effect on diabetic control in type 2 (non-insulin-dependent) diabetic patients? Diabetologia. 1982;23(3):235–9.PubMedCrossRef Simpson HCR, Carter RD, Lousley S, Mann JI. Digestible carbohydrate—an independent effect on diabetic control in type 2 (non-insulin-dependent) diabetic patients? Diabetologia. 1982;23(3):235–9.PubMedCrossRef
8.
Zurück zum Zitat Gannon MC, Nuttall FQ. Effect of a high-protein, low-carbohydrate diet on blood glucose control in people with type 2 diabetes. Diabetes. 2004;53(9):2375–82.PubMedCrossRef Gannon MC, Nuttall FQ. Effect of a high-protein, low-carbohydrate diet on blood glucose control in people with type 2 diabetes. Diabetes. 2004;53(9):2375–82.PubMedCrossRef
9.
Zurück zum Zitat Robertson MD, Bickerton AS, Dennis AL, Vidal H, Frayn KN. Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism. Am J Clin Nutr. 2005;82(3):559–67.PubMedCrossRef Robertson MD, Bickerton AS, Dennis AL, Vidal H, Frayn KN. Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism. Am J Clin Nutr. 2005;82(3):559–67.PubMedCrossRef
10.
Zurück zum Zitat Skytte MJ, Samkani A, Petersen AD et al. A carbohydrate-reduced high-protein diet improves HbA(1c) and liver fat content in weight stable participants with type 2 diabetes: a randomised controlled trial. Diabetologia. 2019;62(11):2066–78.PubMedCrossRef Skytte MJ, Samkani A, Petersen AD et al. A carbohydrate-reduced high-protein diet improves HbA(1c) and liver fat content in weight stable participants with type 2 diabetes: a randomised controlled trial. Diabetologia. 2019;62(11):2066–78.PubMedCrossRef
11.
Zurück zum Zitat Dyson PA, Kelly T, Deakin T et al. Diabetes UK evidence-based nutrition guidelines for the prevention and management of diabetes. Diabet Med. 2011;28(11):1282–8.PubMedCrossRef Dyson PA, Kelly T, Deakin T et al. Diabetes UK evidence-based nutrition guidelines for the prevention and management of diabetes. Diabet Med. 2011;28(11):1282–8.PubMedCrossRef
12.
Zurück zum Zitat Evert AB, Dennison M, Gardner CD et al. Nutrition therapy for adults with diabetes or prediabetes: a consensus report. Diabetes Care. 2019;42(5):731–54.PubMedPubMedCentralCrossRef Evert AB, Dennison M, Gardner CD et al. Nutrition therapy for adults with diabetes or prediabetes: a consensus report. Diabetes Care. 2019;42(5):731–54.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Kelley DE, Wing R, Buonocore C, Sturis J, Polonsky K, Fitzsimmons M. Relative effects of calorie restriction and weight loss in noninsulin-dependent diabetes mellitus. J Clin Endocrinol Metab. 1993;77(5):1287–93.PubMed Kelley DE, Wing R, Buonocore C, Sturis J, Polonsky K, Fitzsimmons M. Relative effects of calorie restriction and weight loss in noninsulin-dependent diabetes mellitus. J Clin Endocrinol Metab. 1993;77(5):1287–93.PubMed
14.
Zurück zum Zitat Jazet IM, Pijl H, Frölich M, Romijn JA, Meinders AE. Two days of a very low calorie diet reduces endogenous glucose production in obese type 2 diabetic patients despite the withdrawal of blood glucose-lowering therapies including insulin. Metabolism. 2005;54(6):705–12.PubMedCrossRef Jazet IM, Pijl H, Frölich M, Romijn JA, Meinders AE. Two days of a very low calorie diet reduces endogenous glucose production in obese type 2 diabetic patients despite the withdrawal of blood glucose-lowering therapies including insulin. Metabolism. 2005;54(6):705–12.PubMedCrossRef
15.
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(10):2506–14.PubMedPubMedCentralCrossRef 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(10):2506–14.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Malandrucco I, Pasqualetti P, Giordani I et al. Very-low-calorie diet: a quick therapeutic tool to improve beta cell function in morbidly obese patients with type 2 diabetes. Am J Clin Nutr. 2012;95(3):609–13.PubMedCrossRef Malandrucco I, Pasqualetti P, Giordani I et al. Very-low-calorie diet: a quick therapeutic tool to improve beta cell function in morbidly obese patients with type 2 diabetes. Am J Clin Nutr. 2012;95(3):609–13.PubMedCrossRef
17.
Zurück zum Zitat Jackness C, Karmally W, Febres G et al. Very low-calorie diet mimics the early beneficial effect of Roux-en-Y gastric bypass on insulin sensitivity and β-cell function in type 2 diabetic patients. Diabetes. 2013;62(9):3027–32.PubMedPubMedCentralCrossRef Jackness C, Karmally W, Febres G et al. Very low-calorie diet mimics the early beneficial effect of Roux-en-Y gastric bypass on insulin sensitivity and β-cell function in type 2 diabetic patients. Diabetes. 2013;62(9):3027–32.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Wing RR. Behavioral treatment of obesity: its application to type II diabetes. Diabetes Care. 1993;16(1):193–9.PubMedCrossRef Wing RR. Behavioral treatment of obesity: its application to type II diabetes. Diabetes Care. 1993;16(1):193–9.PubMedCrossRef
19.
Zurück zum Zitat Steven S, Hollingsworth KG, Al-Mrabeh A et al. Very low-calorie diet and 6 months of weight stability in type 2 diabetes: pathophysiological changes in responders and nonresponders. Diabetes Care. 2016;39(5):808–15.PubMedCrossRef Steven S, Hollingsworth KG, Al-Mrabeh A et al. Very low-calorie diet and 6 months of weight stability in type 2 diabetes: pathophysiological changes in responders and nonresponders. Diabetes Care. 2016;39(5):808–15.PubMedCrossRef
20.
Zurück zum Zitat Taylor R, Al-Mrabeh A, Zhyzhneuskaya S et al. Remission of human type 2 diabetes requires decrease in liver and pancreas fat content but is dependent upon capacity for beta cell recovery. Cell Metab. 2018;28(4):547-556.e3.PubMedCrossRef Taylor R, Al-Mrabeh A, Zhyzhneuskaya S et al. Remission of human type 2 diabetes requires decrease in liver and pancreas fat content but is dependent upon capacity for beta cell recovery. Cell Metab. 2018;28(4):547-556.e3.PubMedCrossRef
21.
Zurück zum Zitat Del Prato S, Tiengo A. The importance of first-phase insulin secretion: implications for the therapy of type 2 diabetes mellitus. Diabetes Metab Res Rev. 2001;17(3):164–74.PubMedCrossRef Del Prato S, Tiengo A. The importance of first-phase insulin secretion: implications for the therapy of type 2 diabetes mellitus. Diabetes Metab Res Rev. 2001;17(3):164–74.PubMedCrossRef
22.
23.
Zurück zum Zitat Garber AJ. The importance of early insulin secretion and its impact on glycaemic regulation. Int J Obes Relat Metab Disord. 2000;24(Suppl 3):S32–7.PubMedCrossRef Garber AJ. The importance of early insulin secretion and its impact on glycaemic regulation. Int J Obes Relat Metab Disord. 2000;24(Suppl 3):S32–7.PubMedCrossRef
24.
Zurück zum Zitat Luzi L, DeFronzo RA. Effect of loss of first-phase insulin secretion on hepatic glucose production and tissue glucose disposal in humans. Am J Physiol. 1989;257(2 Pt 1):E241–6.PubMed Luzi L, DeFronzo RA. Effect of loss of first-phase insulin secretion on hepatic glucose production and tissue glucose disposal in humans. Am J Physiol. 1989;257(2 Pt 1):E241–6.PubMed
25.
Zurück zum Zitat Basu A, Alzaid A, Dinneen S, Caumo A, Cobelli C, Rizza RA. Effects of a change in the pattern of insulin delivery on carbohydrate tolerance in diabetic and nondiabetic humans in the presence of differing degrees of insulin resistance. J Clin Invest. 1996;97(10):2351–61.PubMedPubMedCentralCrossRef Basu A, Alzaid A, Dinneen S, Caumo A, Cobelli C, Rizza RA. Effects of a change in the pattern of insulin delivery on carbohydrate tolerance in diabetic and nondiabetic humans in the presence of differing degrees of insulin resistance. J Clin Invest. 1996;97(10):2351–61.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Gannon MC, Nuttall FQ, Neil BJ, Westphal SA. The insulin and glucose responses to meals of glucose plus various proteins in type II diabetic subjects. Metabolism. 1988;37(11):1081–8.PubMedCrossRef Gannon MC, Nuttall FQ, Neil BJ, Westphal SA. The insulin and glucose responses to meals of glucose plus various proteins in type II diabetic subjects. Metabolism. 1988;37(11):1081–8.PubMedCrossRef
27.
Zurück zum Zitat van Loon LJ, Saris WH, Verhagen H, Wagenmakers AJ. Plasma insulin responses after ingestion of different amino acid or protein mixtures with carbohydrate. Am J Clin Nutr. 2000;72(1):96–105.PubMedCrossRef van Loon LJ, Saris WH, Verhagen H, Wagenmakers AJ. Plasma insulin responses after ingestion of different amino acid or protein mixtures with carbohydrate. Am J Clin Nutr. 2000;72(1):96–105.PubMedCrossRef
28.
Zurück zum Zitat van Loon LJ, Kruijshoop M, Menheere PP, Wagenmakers AJ, Saris WH, Keizer HA. Amino acid ingestion strongly enhances insulin secretion in patients with long-term type 2 diabetes. Diabetes Care. 2003;26(3):625–30.PubMedCrossRef van Loon LJ, Kruijshoop M, Menheere PP, Wagenmakers AJ, Saris WH, Keizer HA. Amino acid ingestion strongly enhances insulin secretion in patients with long-term type 2 diabetes. Diabetes Care. 2003;26(3):625–30.PubMedCrossRef
29.
Zurück zum Zitat Manders RJ, Hansen D, Zorenc AH, Dendale P, Kloek J, Saris WH et al. Protein co-ingestion strongly increases postprandial insulin secretion in type 2 diabetes patients. J Med Food. 2014;17(7):758–63.PubMedCrossRef Manders RJ, Hansen D, Zorenc AH, Dendale P, Kloek J, Saris WH et al. Protein co-ingestion strongly increases postprandial insulin secretion in type 2 diabetes patients. J Med Food. 2014;17(7):758–63.PubMedCrossRef
30.
Zurück zum Zitat Spiller GA, Jensen CD, Pattison TS, Chuck CS, Whittam JH, Scala J. Effect of protein dose on serum glucose and insulin response to sugars. Am J Clin Nutr. 1987;46(3):474–80.PubMedCrossRef Spiller GA, Jensen CD, Pattison TS, Chuck CS, Whittam JH, Scala J. Effect of protein dose on serum glucose and insulin response to sugars. Am J Clin Nutr. 1987;46(3):474–80.PubMedCrossRef
31.
Zurück zum Zitat Manders RJ, Praet SF, Meex RC et al. Protein hydrolysate/leucine co-ingestion reduces the prevalence of hyperglycemia in type 2 diabetic patients. Diabetes Care. 2006;29(12):2721–2.PubMedCrossRef Manders RJ, Praet SF, Meex RC et al. Protein hydrolysate/leucine co-ingestion reduces the prevalence of hyperglycemia in type 2 diabetic patients. Diabetes Care. 2006;29(12):2721–2.PubMedCrossRef
32.
Zurück zum Zitat Schmid R, Schusdziarra V, Schulte-Frohlinde E, Maier V, Classen M. Role of amino acids in stimulation of postprandial insulin, glucagon, and pancreatic polypeptide in humans. Pancreas. 1989;4(3):305–14.PubMedCrossRef Schmid R, Schusdziarra V, Schulte-Frohlinde E, Maier V, Classen M. Role of amino acids in stimulation of postprandial insulin, glucagon, and pancreatic polypeptide in humans. Pancreas. 1989;4(3):305–14.PubMedCrossRef
33.
Zurück zum Zitat Newsholme P, Cruzat V, Arfuso F, Keane K. Nutrient regulation of insulin secretion and action. J Endocrinol. 2014;221(3):R105–20.PubMedCrossRef Newsholme P, Cruzat V, Arfuso F, Keane K. Nutrient regulation of insulin secretion and action. J Endocrinol. 2014;221(3):R105–20.PubMedCrossRef
34.
Zurück zum Zitat van der Klaauw AA, Keogh JM et al. High protein intake stimulates postprandial GLP1 and PYY release. Obesity (Silver Spring). 2013;21(8):1602–7.CrossRef van der Klaauw AA, Keogh JM et al. High protein intake stimulates postprandial GLP1 and PYY release. Obesity (Silver Spring). 2013;21(8):1602–7.CrossRef
35.
Zurück zum Zitat Bock G, Dalla Man C, Campioni M et al. Effects of nonglucose nutrients on insulin secretion and action in people with pre-diabetes. Diabetes. 2007;56(4):1113–9.PubMedCrossRef Bock G, Dalla Man C, Campioni M et al. Effects of nonglucose nutrients on insulin secretion and action in people with pre-diabetes. Diabetes. 2007;56(4):1113–9.PubMedCrossRef
36.
Zurück zum Zitat Gannon MC, Nuttall FQ. Control of blood glucose in type 2 diabetes without weight loss by modification of diet composition. Nutr Metab. 2006;3(1):16.CrossRef Gannon MC, Nuttall FQ. Control of blood glucose in type 2 diabetes without weight loss by modification of diet composition. Nutr Metab. 2006;3(1):16.CrossRef
37.
Zurück zum Zitat Nuttall FQ, Gannon MC. Effect of a LoBAG30 diet on protein metabolism in men with type 2 diabetes. A randomized controlled trial. Nutr Metab (Lond). 2012;9(1):43.CrossRef Nuttall FQ, Gannon MC. Effect of a LoBAG30 diet on protein metabolism in men with type 2 diabetes. A randomized controlled trial. Nutr Metab (Lond). 2012;9(1):43.CrossRef
38.
Zurück zum Zitat Skytte MJ, Samkani A, Astrup A et al. Effects of carbohydrate restriction on postprandial glucose metabolism, beta-cell function, gut hormone secretion, and satiety in patients with type 2 diabetes. Am J Physiol Endocrinol Metab. 2020;320:E7–18.PubMedCrossRef Skytte MJ, Samkani A, Astrup A et al. Effects of carbohydrate restriction on postprandial glucose metabolism, beta-cell function, gut hormone secretion, and satiety in patients with type 2 diabetes. Am J Physiol Endocrinol Metab. 2020;320:E7–18.PubMedCrossRef
39.
Zurück zum Zitat Boden G, Sargrad K, Homko C, Mozzoli M, Stein TP. Effect of a low-carbohydrate diet on appetite, blood glucose levels, and insulin resistance in obese patients with type 2 diabetes. Ann Intern Med. 2005;142(6):403–11.PubMedCrossRef Boden G, Sargrad K, Homko C, Mozzoli M, Stein TP. Effect of a low-carbohydrate diet on appetite, blood glucose levels, and insulin resistance in obese patients with type 2 diabetes. Ann Intern Med. 2005;142(6):403–11.PubMedCrossRef
40.
Zurück zum Zitat Ajala O, English P, Pinkney J. Systematic review and meta-analysis of different dietary approaches to the management of type 2 diabetes. Am J Clin Nutr. 2013;97(3):505–16.PubMedCrossRef Ajala O, English P, Pinkney J. Systematic review and meta-analysis of different dietary approaches to the management of type 2 diabetes. Am J Clin Nutr. 2013;97(3):505–16.PubMedCrossRef
41.
Zurück zum Zitat Gannon MC, Nuttall JA, Damberg G, Gupta V, Nuttall FQ. Effect of protein ingestion on the glucose appearance rate in people with type 2 diabetes. J Clin Endocrinol Metab. 2001;86(3):1040–7.PubMed Gannon MC, Nuttall JA, Damberg G, Gupta V, Nuttall FQ. Effect of protein ingestion on the glucose appearance rate in people with type 2 diabetes. J Clin Endocrinol Metab. 2001;86(3):1040–7.PubMed
42.
Zurück zum Zitat Manders RJ, Wagenmakers AJ, Koopman R et al. Co-ingestion of a protein hydrolysate and amino acid mixture with carbohydrate improves plasma glucose disposal in patients with type 2 diabetes. Am J Clin Nutr. 2005;82(1):76–83.PubMedCrossRef Manders RJ, Wagenmakers AJ, Koopman R et al. Co-ingestion of a protein hydrolysate and amino acid mixture with carbohydrate improves plasma glucose disposal in patients with type 2 diabetes. Am J Clin Nutr. 2005;82(1):76–83.PubMedCrossRef
43.
Zurück zum Zitat Rietman A, Schwarz J, Tomé D, Kok FJ, Mensink M. High dietary protein intake, reducing or eliciting insulin resistance? Eur J Clin Nutr. 2014;68(9):973–9.PubMedCrossRef Rietman A, Schwarz J, Tomé D, Kok FJ, Mensink M. High dietary protein intake, reducing or eliciting insulin resistance? Eur J Clin Nutr. 2014;68(9):973–9.PubMedCrossRef
44.
Zurück zum Zitat Wolever TM, Yang M, Zeng XY, Atkinson F, Brand-Miller JC. Food glycemic index, as given in glycemic index tables, is a significant determinant of glycemic responses elicited by composite breakfast meals. Am J Clin Nutr. 2006;83(6):1306–12.PubMedCrossRef Wolever TM, Yang M, Zeng XY, Atkinson F, Brand-Miller JC. Food glycemic index, as given in glycemic index tables, is a significant determinant of glycemic responses elicited by composite breakfast meals. Am J Clin Nutr. 2006;83(6):1306–12.PubMedCrossRef
45.
Zurück zum Zitat Allick G, Bisschop PH, Ackermans MT et al. A low-carbohydrate/high-fat diet improves glucoregulation in type 2 diabetes mellitus by reducing postabsorptive glycogenolysis. J Clin Endocrinol Metab. 2004;89(12):6193–7.PubMedCrossRef Allick G, Bisschop PH, Ackermans MT et al. A low-carbohydrate/high-fat diet improves glucoregulation in type 2 diabetes mellitus by reducing postabsorptive glycogenolysis. J Clin Endocrinol Metab. 2004;89(12):6193–7.PubMedCrossRef
46.
Zurück zum Zitat Conn JW. Interpretation of the glucose tolerance test. The necessity of a stand ARD preparatory diet. Am J Med Sci. 1940;199:555–64.CrossRef Conn JW. Interpretation of the glucose tolerance test. The necessity of a stand ARD preparatory diet. Am J Med Sci. 1940;199:555–64.CrossRef
47.
Zurück zum Zitat Klein KR, Walker CP, McFerren AL, Huffman H, Frohlich F, Buse JB. Carbohydrate intake prior to oral glucose tolerance testing. J Endocr Soc. 2021;5(5):bvab049.PubMedPubMedCentralCrossRef Klein KR, Walker CP, McFerren AL, Huffman H, Frohlich F, Buse JB. Carbohydrate intake prior to oral glucose tolerance testing. J Endocr Soc. 2021;5(5):bvab049.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Simpson HC, Carter RD, Lousley S, Mann JI. Digestible carbohydrate–an independent effect on diabetic control in type 2 (non-insulin-dependent) diabetic patients? Diabetologia. 1982;23(3):235–9.PubMedCrossRef Simpson HC, Carter RD, Lousley S, Mann JI. Digestible carbohydrate–an independent effect on diabetic control in type 2 (non-insulin-dependent) diabetic patients? Diabetologia. 1982;23(3):235–9.PubMedCrossRef
49.
Zurück zum Zitat Kirk E, Reeds DN, Finck BN, Mayurranjan SM, Patterson BW, Klein S. Dietary fat and carbohydrates differentially alter insulin sensitivity during caloric restriction. Gastroenterology. 2009;136(5):1552–60.PubMedCrossRef Kirk E, Reeds DN, Finck BN, Mayurranjan SM, Patterson BW, Klein S. Dietary fat and carbohydrates differentially alter insulin sensitivity during caloric restriction. Gastroenterology. 2009;136(5):1552–60.PubMedCrossRef
50.
Zurück zum Zitat Hyde PN, Sapper TN, Crabtree CD et al. Dietary carbohydrate restriction improves metabolic syndrome independent of weight loss. JCI Insight. 2019;4(12):e128308.PubMedCentralCrossRef Hyde PN, Sapper TN, Crabtree CD et al. Dietary carbohydrate restriction improves metabolic syndrome independent of weight loss. JCI Insight. 2019;4(12):e128308.PubMedCentralCrossRef
51.
Zurück zum Zitat Ebbeling CB, Knapp A, Johnson A et al. Effects of a low-carbohydrate diet on insulin-resistant dyslipoproteinemia-a randomized controlled feeding trial. Am J Clin Nutr. 2021;115:154–62.PubMedCentralCrossRef Ebbeling CB, Knapp A, Johnson A et al. Effects of a low-carbohydrate diet on insulin-resistant dyslipoproteinemia-a randomized controlled feeding trial. Am J Clin Nutr. 2021;115:154–62.PubMedCentralCrossRef
52.
Zurück zum Zitat Wolever TM, Gibbs AL, Chiasson JL et al. Altering source or amount of dietary carbohydrate has acute and chronic effects on postprandial glucose and triglycerides in type 2 diabetes: Canadian trial of Carbohydrates in Diabetes (CCD). Nutr Metab Cardiovasc Dis. 2013;23(3):227–34.PubMedCrossRef Wolever TM, Gibbs AL, Chiasson JL et al. Altering source or amount of dietary carbohydrate has acute and chronic effects on postprandial glucose and triglycerides in type 2 diabetes: Canadian trial of Carbohydrates in Diabetes (CCD). Nutr Metab Cardiovasc Dis. 2013;23(3):227–34.PubMedCrossRef
53.
Zurück zum Zitat Snorgaard O, Poulsen GM, Andersen HK, Astrup A. Systematic review and meta-analysis of dietary carbohydrate restriction in patients with type 2 diabetes. BMJ Open Diabetes Res Care. 2017;5(1):e000354.PubMedPubMedCentralCrossRef Snorgaard O, Poulsen GM, Andersen HK, Astrup A. Systematic review and meta-analysis of dietary carbohydrate restriction in patients with type 2 diabetes. BMJ Open Diabetes Res Care. 2017;5(1):e000354.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Goldenberg JZ, Day A, Brinkworth GD et al. Efficacy and safety of low and very low carbohydrate diets for type 2 diabetes remission: systematic review and meta-analysis of published and unpublished randomized trial data. BMJ. 2021;372:m4743.PubMedPubMedCentralCrossRef Goldenberg JZ, Day A, Brinkworth GD et al. Efficacy and safety of low and very low carbohydrate diets for type 2 diabetes remission: systematic review and meta-analysis of published and unpublished randomized trial data. BMJ. 2021;372:m4743.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Westman EC, Yancy WS, Mavropoulos JC, Marquart M, McDuffie JR. The effect of a low-carbohydrate, ketogenic diet versus a low-glycemic index diet on glycemic control in type 2 diabetes mellitus. Nutr Metab. 2008;5(1):36.CrossRef Westman EC, Yancy WS, Mavropoulos JC, Marquart M, McDuffie JR. The effect of a low-carbohydrate, ketogenic diet versus a low-glycemic index diet on glycemic control in type 2 diabetes mellitus. Nutr Metab. 2008;5(1):36.CrossRef
56.
Zurück zum Zitat Haimoto H, Sasakabe T, Wakai K, Umegaki H. Effects of a low-carbohydrate diet on glycemic control in outpatients with severe type 2 diabetes. Nutr Metab (Lond). 2009;6:21.CrossRef Haimoto H, Sasakabe T, Wakai K, Umegaki H. Effects of a low-carbohydrate diet on glycemic control in outpatients with severe type 2 diabetes. Nutr Metab (Lond). 2009;6:21.CrossRef
57.
Zurück zum Zitat Wolever TM, Chiasson JL, Josse RG et al. Effects of changing the amount and source of dietary carbohydrates on symptoms and dietary satisfaction over a 1-year period in subjects with type 2 diabetes: Canadian trial of carbohydrates in diabetes (CCD). Can J Diabetes. 2017;41(2):164–76.PubMedCrossRef Wolever TM, Chiasson JL, Josse RG et al. Effects of changing the amount and source of dietary carbohydrates on symptoms and dietary satisfaction over a 1-year period in subjects with type 2 diabetes: Canadian trial of carbohydrates in diabetes (CCD). Can J Diabetes. 2017;41(2):164–76.PubMedCrossRef
58.
Zurück zum Zitat Guess N. A randomised crossover trial: exploring the dose-response effect of carbohydrate restriction on glycaemia in people with type 2 diabetes (D-ROC2). Preprint, 2021. Guess N. A randomised crossover trial: exploring the dose-response effect of carbohydrate restriction on glycaemia in people with type 2 diabetes (D-ROC2). Preprint, 2021.
59.
Zurück zum Zitat Tay J, Luscombe-Marsh ND, Thompson CH et al. A very low-carbohydrate, low-saturated fat diet for type 2 diabetes management: a randomized trial. Diabetes Care. 2014;37(11):2909–18.PubMedCrossRef Tay J, Luscombe-Marsh ND, Thompson CH et al. A very low-carbohydrate, low-saturated fat diet for type 2 diabetes management: a randomized trial. Diabetes Care. 2014;37(11):2909–18.PubMedCrossRef
60.
Zurück zum Zitat Gumbiner B, Wendel JA, McDermott MP. Effects of diet composition and ketosis on glycemia during very-low-energy-diet therapy in obese patients with non-insulin-dependent diabetes mellitus. Am J Clin Nutr. 1996;63(1):110–5.PubMedCrossRef Gumbiner B, Wendel JA, McDermott MP. Effects of diet composition and ketosis on glycemia during very-low-energy-diet therapy in obese patients with non-insulin-dependent diabetes mellitus. Am J Clin Nutr. 1996;63(1):110–5.PubMedCrossRef
61.
Zurück zum Zitat Henry RR, Brechtel G, Lim KH. Effects of ketone bodies on carbohydrate metabolism in non-insulin-dependent (type II) diabetes mellitus. Metabolism. 1990;39(8):853–8.PubMedCrossRef Henry RR, Brechtel G, Lim KH. Effects of ketone bodies on carbohydrate metabolism in non-insulin-dependent (type II) diabetes mellitus. Metabolism. 1990;39(8):853–8.PubMedCrossRef
62.
Zurück zum Zitat Myette-Côté É, Neudorf H, Rafiei H, Clarke K, Little JP. Prior ingestion of exogenous ketone monoester attenuates the glycaemic response to an oral glucose tolerance test in healthy young individuals. J Physiol. 2018;596(8):1385–95.PubMedPubMedCentralCrossRef Myette-Côté É, Neudorf H, Rafiei H, Clarke K, Little JP. Prior ingestion of exogenous ketone monoester attenuates the glycaemic response to an oral glucose tolerance test in healthy young individuals. J Physiol. 2018;596(8):1385–95.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Walsh JJ, Neudorf H, Little JP. 14-days of ketone supplementation lowers glucose and improves vascular function in obesity: a randomized crossover trial. J Clin Endocrinol Metab. 2020;106:1738–54.PubMedCentralCrossRef Walsh JJ, Neudorf H, Little JP. 14-days of ketone supplementation lowers glucose and improves vascular function in obesity: a randomized crossover trial. J Clin Endocrinol Metab. 2020;106:1738–54.PubMedCentralCrossRef
64.
Zurück zum Zitat Kowalski GM, Moore SM, Hamley S, Selathurai A, Bruce CR. The effect of ingested glucose dose on the suppression of endogenous glucose production in humans. Diabetes. 2017;66(9):2400–6.PubMedCrossRef Kowalski GM, Moore SM, Hamley S, Selathurai A, Bruce CR. The effect of ingested glucose dose on the suppression of endogenous glucose production in humans. Diabetes. 2017;66(9):2400–6.PubMedCrossRef
65.
Zurück zum Zitat Singhal P, Caumo A, Carey PE, Cobelli C, Taylor R. Regulation of endogenous glucose production after a mixed meal in type 2 diabetes. Am J Physiol Endocrinol Metab. 2002;283(2):E275–83.PubMedCrossRef Singhal P, Caumo A, Carey PE, Cobelli C, Taylor R. Regulation of endogenous glucose production after a mixed meal in type 2 diabetes. Am J Physiol Endocrinol Metab. 2002;283(2):E275–83.PubMedCrossRef
66.
Zurück zum Zitat Sathananthan M, Shah M, Edens KL et al. Six and 12 weeks of caloric restriction increases beta cell function and lowers fasting and postprandial glucose concentrations in people with type 2 diabetes. J Nutr. 2015;145(9):2046–51.PubMedPubMedCentralCrossRef Sathananthan M, Shah M, Edens KL et al. Six and 12 weeks of caloric restriction increases beta cell function and lowers fasting and postprandial glucose concentrations in people with type 2 diabetes. J Nutr. 2015;145(9):2046–51.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Christiansen MP, Linfoot PA, Neese RA, Hellerstein MK. Effect of dietary energy restriction on glucose production and substrate utilization in type 2 diabetes. Diabetes. 2000;49(10):1691–9.PubMedCrossRef Christiansen MP, Linfoot PA, Neese RA, Hellerstein MK. Effect of dietary energy restriction on glucose production and substrate utilization in type 2 diabetes. Diabetes. 2000;49(10):1691–9.PubMedCrossRef
68.
Zurück zum Zitat Jazet IM, Schaart G, Gastaldelli A et al. Loss of 50% of excess weight using a very low energy diet improves insulin-stimulated glucose disposal and skeletal muscle insulin signalling in obese insulin-treated type 2 diabetic patients. Diabetologia. 2008;51(2):309–19.PubMedCrossRef Jazet IM, Schaart G, Gastaldelli A et al. Loss of 50% of excess weight using a very low energy diet improves insulin-stimulated glucose disposal and skeletal muscle insulin signalling in obese insulin-treated type 2 diabetic patients. Diabetologia. 2008;51(2):309–19.PubMedCrossRef
69.
70.
Zurück zum Zitat Jenkins D, Wolever T, Jenkins A et al. The glycaemic index of foods tested in diabetic patients: a new basis for carbohydrate exchange favouring the use of legumes. Diabetologia. 1983;24(4):257–64.PubMedCrossRef Jenkins D, Wolever T, Jenkins A et al. The glycaemic index of foods tested in diabetic patients: a new basis for carbohydrate exchange favouring the use of legumes. Diabetologia. 1983;24(4):257–64.PubMedCrossRef
71.
Zurück zum Zitat Dyson P, Twenefour D, Breen C et al. Diabetes UK evidence-based nutrition guidelines for the prevention and management of diabetes. Diabet Med. 2018;35(5):541–7.PubMedCrossRef Dyson P, Twenefour D, Breen C et al. Diabetes UK evidence-based nutrition guidelines for the prevention and management of diabetes. Diabet Med. 2018;35(5):541–7.PubMedCrossRef
72.
Zurück zum Zitat Ojo O, Ojo OO, Adebowale F, Wang XH. The effect of dietary glycaemic index on glycaemia in patients with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Nutrients. 2018;10(3):373.PubMedCentralCrossRef Ojo O, Ojo OO, Adebowale F, Wang XH. The effect of dietary glycaemic index on glycaemia in patients with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Nutrients. 2018;10(3):373.PubMedCentralCrossRef
73.
Zurück zum Zitat Wu T, Bound M, Standfield S et al. Effects of rectal administration of taurocholic acid on glucagon-like peptide-1 and peptide YY secretion in healthy humans. Diabetes Obes Metab. 2013;15(5):474–7.PubMedCrossRef Wu T, Bound M, Standfield S et al. Effects of rectal administration of taurocholic acid on glucagon-like peptide-1 and peptide YY secretion in healthy humans. Diabetes Obes Metab. 2013;15(5):474–7.PubMedCrossRef
74.
Zurück zum Zitat Canfora EE, van der Beek CM, Jocken JW et al. Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: a randomized crossover trial. Sci Rep. 2017;7(1):1–12.CrossRef Canfora EE, van der Beek CM, Jocken JW et al. Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: a randomized crossover trial. Sci Rep. 2017;7(1):1–12.CrossRef
75.
Zurück zum Zitat Gee JM, Johnson IT. Dietary lactitol fermentation increases circulating peptide YY and glucagon-like peptide-1 in rats and humans. Nutrition. 2005;21(10):1036–43.PubMedCrossRef Gee JM, Johnson IT. Dietary lactitol fermentation increases circulating peptide YY and glucagon-like peptide-1 in rats and humans. Nutrition. 2005;21(10):1036–43.PubMedCrossRef
76.
Zurück zum Zitat Verhoef SP, Meyer D, Westerterp KR. Effects of oligofructose on appetite profile, glucagon-like peptide 1 and peptide YY3-36 concentrations and energy intake. Br J Nutr. 2011;106(11):1757–62.PubMedCrossRef Verhoef SP, Meyer D, Westerterp KR. Effects of oligofructose on appetite profile, glucagon-like peptide 1 and peptide YY3-36 concentrations and energy intake. Br J Nutr. 2011;106(11):1757–62.PubMedCrossRef
77.
Zurück zum Zitat Weickert MO, Mohlig M, Koebnick C et al. Impact of cereal fibre on glucose-regulating factors. Diabetologia. 2005;48(11):2343–53.PubMedCrossRef Weickert MO, Mohlig M, Koebnick C et al. Impact of cereal fibre on glucose-regulating factors. Diabetologia. 2005;48(11):2343–53.PubMedCrossRef
78.
Zurück zum Zitat Bodinham CL, Smith L, Wright J, Frost GS, Robertson MD. Dietary fibre improves first-phase insulin secretion in overweight individuals. PLoS ONE. 2012;7(7):e40834.PubMedPubMedCentralCrossRef Bodinham CL, Smith L, Wright J, Frost GS, Robertson MD. Dietary fibre improves first-phase insulin secretion in overweight individuals. PLoS ONE. 2012;7(7):e40834.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Priyadarshini M, Wicksteed B, Schiltz GE, Gilchrist A, Layden BT. SCFA receptors in pancreatic β cells: novel diabetes targets? Trends Endocrinol Metab. 2016;27(9):653–64.PubMedPubMedCentralCrossRef Priyadarshini M, Wicksteed B, Schiltz GE, Gilchrist A, Layden BT. SCFA receptors in pancreatic β cells: novel diabetes targets? Trends Endocrinol Metab. 2016;27(9):653–64.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Uusitupa M, Tuomilehto J, Karttunen P, Wolf E. Long term effects of guar gum on metabolic control, serum cholesterol and blood pressure levels in type 2 (non-insulin-dependent) diabetic patients with high blood pressure. Ann Clin Res. 1984;16(Suppl 43):126–31.PubMed Uusitupa M, Tuomilehto J, Karttunen P, Wolf E. Long term effects of guar gum on metabolic control, serum cholesterol and blood pressure levels in type 2 (non-insulin-dependent) diabetic patients with high blood pressure. Ann Clin Res. 1984;16(Suppl 43):126–31.PubMed
81.
Zurück zum Zitat Holman RR, Steemson J, Darling P, Turner RC. No glycemic benefit from guar administration in NIDDM. Diabetes Care. 1987;10(1):68–71.PubMedCrossRef Holman RR, Steemson J, Darling P, Turner RC. No glycemic benefit from guar administration in NIDDM. Diabetes Care. 1987;10(1):68–71.PubMedCrossRef
82.
Zurück zum Zitat Karlström B, Vessby B, Asp N-G et al. Effects of an increased content of cereal fibre in the diet of type 2 (non-insulin-dependent) diabetic patients. Diabetologia. 1984;26(4):272–7.PubMedCrossRef Karlström B, Vessby B, Asp N-G et al. Effects of an increased content of cereal fibre in the diet of type 2 (non-insulin-dependent) diabetic patients. Diabetologia. 1984;26(4):272–7.PubMedCrossRef
83.
Zurück zum Zitat Hagander B, Asp NG, Efendić S, Nilsson-Ehle P, Scherstén B. Dietary fiber decreases fasting blood glucose levels and plasma LDL concentration in noninsulin-dependent diabetes mellitus patients. Am J Clin Nutr. 1988;47(5):852–8.PubMedCrossRef Hagander B, Asp NG, Efendić S, Nilsson-Ehle P, Scherstén B. Dietary fiber decreases fasting blood glucose levels and plasma LDL concentration in noninsulin-dependent diabetes mellitus patients. Am J Clin Nutr. 1988;47(5):852–8.PubMedCrossRef
84.
Zurück zum Zitat Chandalia M, Garg A, Lutjohann D, von Bergmann K, Grundy SM, Brinkley LJ. Beneficial effects of high dietary fiber intake in patients with type 2 diabetes mellitus. N Engl J Med. 2000;342(19):1392–8.PubMedCrossRef Chandalia M, Garg A, Lutjohann D, von Bergmann K, Grundy SM, Brinkley LJ. Beneficial effects of high dietary fiber intake in patients with type 2 diabetes mellitus. N Engl J Med. 2000;342(19):1392–8.PubMedCrossRef
85.
Zurück zum Zitat Gibb RD, McRorie JW, Jr., Russell DA, Hasselblad V, D'Alessio DA. Psyllium fiber improves glycemic control proportional to loss of glycemic control: a meta-analysis of data in euglycemic subjects, patients at risk of type 2 diabetes mellitus, and patients being treated for type 2 diabetes mellitus. Am J Clin Nutr. 2015;102(6):1604–14.PubMedCrossRef Gibb RD, McRorie JW, Jr., Russell DA, Hasselblad V, D'Alessio DA. Psyllium fiber improves glycemic control proportional to loss of glycemic control: a meta-analysis of data in euglycemic subjects, patients at risk of type 2 diabetes mellitus, and patients being treated for type 2 diabetes mellitus. Am J Clin Nutr. 2015;102(6):1604–14.PubMedCrossRef
86.
Zurück zum Zitat Mao T, Huang F, Zhu X, Wei D, Chen L. Effects of dietary fiber on glycemic control and insulin sensitivity in patients with type 2 diabetes: a systematic review and meta-analysis. J Funct Foods. 2021;82:104500.CrossRef Mao T, Huang F, Zhu X, Wei D, Chen L. Effects of dietary fiber on glycemic control and insulin sensitivity in patients with type 2 diabetes: a systematic review and meta-analysis. J Funct Foods. 2021;82:104500.CrossRef
87.
Zurück zum Zitat De Natale C, Annuzzi G, Bozzetto L et al. Effects of a plant-based high-carbohydrate/high-fiber diet versus high–monounsaturated fat/low-carbohydrate diet on postprandial lipids in type 2 diabetic patients. Diabetes Care. 2009;32(12):2168–73.PubMedPubMedCentralCrossRef De Natale C, Annuzzi G, Bozzetto L et al. Effects of a plant-based high-carbohydrate/high-fiber diet versus high–monounsaturated fat/low-carbohydrate diet on postprandial lipids in type 2 diabetic patients. Diabetes Care. 2009;32(12):2168–73.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Moreno-Valdespino CA, Luna-Vital D, Camacho-Ruiz RM, Mojica L. Bioactive proteins and phytochemicals from legumes: mechanisms of action preventing obesity and type-2 diabetes. Food Res Int. 2020;130:108905.PubMedCrossRef Moreno-Valdespino CA, Luna-Vital D, Camacho-Ruiz RM, Mojica L. Bioactive proteins and phytochemicals from legumes: mechanisms of action preventing obesity and type-2 diabetes. Food Res Int. 2020;130:108905.PubMedCrossRef
89.
Zurück zum Zitat Mudra M, Ercan-Fang N, Zhong L, Furne J, Levitt M. Influence of mulberry leaf extract on the blood glucose and breath hydrogen response to ingestion of 75 g sucrose by type 2 diabetic and control subjects. Diabetes Care. 2007;30(5):1272–4.PubMedCrossRef Mudra M, Ercan-Fang N, Zhong L, Furne J, Levitt M. Influence of mulberry leaf extract on the blood glucose and breath hydrogen response to ingestion of 75 g sucrose by type 2 diabetic and control subjects. Diabetes Care. 2007;30(5):1272–4.PubMedCrossRef
90.
Zurück zum Zitat Törrönen R, Sarkkinen E, Tapola N, Hautaniemi E, Kilpi K, Niskanen L. Berries modify the postprandial plasma glucose response to sucrose in healthy subjects. Br J Nutr. 2010;103(8):1094–7.PubMedCrossRef Törrönen R, Sarkkinen E, Tapola N, Hautaniemi E, Kilpi K, Niskanen L. Berries modify the postprandial plasma glucose response to sucrose in healthy subjects. Br J Nutr. 2010;103(8):1094–7.PubMedCrossRef
91.
Zurück zum Zitat Vessby B, Uusitupa M, Hermansen K et al. Substituting dietary saturated for monounsaturated fat impairs insulin sensitivity in healthy men and women: the KANWU study. Diabetologia. 2001;44(3):312–9.PubMedCrossRef Vessby B, Uusitupa M, Hermansen K et al. Substituting dietary saturated for monounsaturated fat impairs insulin sensitivity in healthy men and women: the KANWU study. Diabetologia. 2001;44(3):312–9.PubMedCrossRef
92.
Zurück zum Zitat Summers LK, Fielding BA, Bradshaw HA et al. Substituting dietary saturated fat with polyunsaturated fat changes abdominal fat distribution and improves insulin sensitivity. Diabetologia. 2002;45(3):369–77.PubMedCrossRef Summers LK, Fielding BA, Bradshaw HA et al. Substituting dietary saturated fat with polyunsaturated fat changes abdominal fat distribution and improves insulin sensitivity. Diabetologia. 2002;45(3):369–77.PubMedCrossRef
93.
Zurück zum Zitat Bjermo H, Iggman D, Kullberg J et al. Effects of n-6 PUFAs compared with SFAs on liver fat, lipoproteins, and inflammation in abdominal obesity: a randomized controlled trial. Am J Clin Nutr. 2012;95(5):1003–12.PubMedCrossRef Bjermo H, Iggman D, Kullberg J et al. Effects of n-6 PUFAs compared with SFAs on liver fat, lipoproteins, and inflammation in abdominal obesity: a randomized controlled trial. Am J Clin Nutr. 2012;95(5):1003–12.PubMedCrossRef
94.
Zurück zum Zitat Garg A. High-monounsaturated-fat diets for patients with diabetes mellitus: a meta-analysis. Am J Clin Nutr. 1998;67(3 Suppl):577s–82s.PubMedCrossRef Garg A. High-monounsaturated-fat diets for patients with diabetes mellitus: a meta-analysis. Am J Clin Nutr. 1998;67(3 Suppl):577s–82s.PubMedCrossRef
95.
Zurück zum Zitat Garg A, Bonanome A, Grundy SM, Zhang ZJ, Unger RH. Comparison of a high-carbohydrate diet with a high-monounsaturated-fat diet in patients with non-insulin-dependent diabetes mellitus. N Engl J Med. 1988;319(13):829–34.PubMedCrossRef Garg A, Bonanome A, Grundy SM, Zhang ZJ, Unger RH. Comparison of a high-carbohydrate diet with a high-monounsaturated-fat diet in patients with non-insulin-dependent diabetes mellitus. N Engl J Med. 1988;319(13):829–34.PubMedCrossRef
96.
Zurück zum Zitat Garg A, Grundy SM, Unger RH. Comparison of effects of high and low carbohydrate diets on plasma lipoproteins and insulin sensitivity in patients with mild NIDDM. Diabetes. 1992;41(10):1278–85.PubMedCrossRef Garg A, Grundy SM, Unger RH. Comparison of effects of high and low carbohydrate diets on plasma lipoproteins and insulin sensitivity in patients with mild NIDDM. Diabetes. 1992;41(10):1278–85.PubMedCrossRef
97.
Zurück zum Zitat Sumithran P, Prendergast LA, Delbridge E. Long-term persistence of hormonal adaptations to weight loss. N Engl J Med. 2011;365(17):1597–604.PubMedCrossRef Sumithran P, Prendergast LA, Delbridge E. Long-term persistence of hormonal adaptations to weight loss. N Engl J Med. 2011;365(17):1597–604.PubMedCrossRef
98.
Zurück zum Zitat Fothergill E, Guo J, Howard L et al. Persistent metabolic adaptation 6 years after “The Biggest Loser” competition. Obesity (Silver Spring). 2016;24(8):1612–9.CrossRef Fothergill E, Guo J, Howard L et al. Persistent metabolic adaptation 6 years after “The Biggest Loser” competition. Obesity (Silver Spring). 2016;24(8):1612–9.CrossRef
99.
Zurück zum Zitat Bueno NB, de Melo IS, de Oliveira SL, da Rocha Ataide T. Very-low-carbohydrate ketogenic diet v. low-fat diet for long-term weight loss: a meta-analysis of randomised controlled trials. Br J Nutr. 2013;110(7):1178–87.PubMedCrossRef Bueno NB, de Melo IS, de Oliveira SL, da Rocha Ataide T. Very-low-carbohydrate ketogenic diet v. low-fat diet for long-term weight loss: a meta-analysis of randomised controlled trials. Br J Nutr. 2013;110(7):1178–87.PubMedCrossRef
100.
Zurück zum Zitat Jaacks LM, Ma Y, Davis N et al. Long-term changes in dietary and food intake behaviour in the Diabetes Prevention Program Outcomes Study. Diabet Med. 2014;31(12):1631–42.PubMedPubMedCentralCrossRef Jaacks LM, Ma Y, Davis N et al. Long-term changes in dietary and food intake behaviour in the Diabetes Prevention Program Outcomes Study. Diabet Med. 2014;31(12):1631–42.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Livingstone KM, Celis-Morales C, Navas-Carretero S et al. Personalised nutrition advice reduces intake of discretionary foods and beverages: findings from the Food4Me randomised controlled trial. Int J Behav Nutr Phys Act. 2021;18(1):70.PubMedPubMedCentralCrossRef Livingstone KM, Celis-Morales C, Navas-Carretero S et al. Personalised nutrition advice reduces intake of discretionary foods and beverages: findings from the Food4Me randomised controlled trial. Int J Behav Nutr Phys Act. 2021;18(1):70.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat McKenzie AL, Hallberg SJ, Creighton BC et al. A novel intervention including individualized nutritional recommendations reduces hemoglobin A1c level, medication use, and weight in type 2 diabetes. JMIR Diabetes. 2017;2(1):e5.PubMedPubMedCentralCrossRef McKenzie AL, Hallberg SJ, Creighton BC et al. A novel intervention including individualized nutritional recommendations reduces hemoglobin A1c level, medication use, and weight in type 2 diabetes. JMIR Diabetes. 2017;2(1):e5.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Knowler WC, Barrett-Connor E, Fowler SE et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346(6):393–403.PubMedCrossRef Knowler WC, Barrett-Connor E, Fowler SE et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346(6):393–403.PubMedCrossRef
105.
Zurück zum Zitat Al-Mrabeh A, Zhyzhneuskaya SV, Peters C et al. Hepatic lipoprotein export and remission of human type 2 diabetes after weight loss. Cell Metab. 2020;31(2):233-249.e4.PubMedCrossRef Al-Mrabeh A, Zhyzhneuskaya SV, Peters C et al. Hepatic lipoprotein export and remission of human type 2 diabetes after weight loss. Cell Metab. 2020;31(2):233-249.e4.PubMedCrossRef
106.
Zurück zum Zitat Appel LJ, Sacks FM, Carey VJ et al. Effects of protein, monounsaturated fat, and carbohydrate intake on blood pressure and serum lipidsresults of the omniheart randomized trial. JAMA. 2005;294(19):2455–64.PubMedCrossRef Appel LJ, Sacks FM, Carey VJ et al. Effects of protein, monounsaturated fat, and carbohydrate intake on blood pressure and serum lipidsresults of the omniheart randomized trial. JAMA. 2005;294(19):2455–64.PubMedCrossRef
107.
Zurück zum Zitat Azadbakht L, Fard NR, Karimi M et al. Effects of the Dietary Approaches to Stop Hypertension (DASH) eating plan on cardiovascular risks among type 2 diabetic patients: a randomized crossover clinical trial. Diabetes Care. 2011;34(1):55–7.PubMedCrossRef Azadbakht L, Fard NR, Karimi M et al. Effects of the Dietary Approaches to Stop Hypertension (DASH) eating plan on cardiovascular risks among type 2 diabetic patients: a randomized crossover clinical trial. Diabetes Care. 2011;34(1):55–7.PubMedCrossRef
108.
Zurück zum Zitat Markova M, Pivovarova O, Hornemann S et al. Isocaloric diets high in animal or plant protein reduce liver fat and inflammation in individuals with type 2 diabetes. Gastroenterology. 2017;152(3):571-585.e8.PubMedCrossRef Markova M, Pivovarova O, Hornemann S et al. Isocaloric diets high in animal or plant protein reduce liver fat and inflammation in individuals with type 2 diabetes. Gastroenterology. 2017;152(3):571-585.e8.PubMedCrossRef
Metadaten
Titel
Could Dietary Modification Independent of Energy Balance Influence the Underlying Pathophysiology of Type 2 Diabetes? Implications for Type 2 Diabetes Remission
verfasst von
Nicola D. Guess
Publikationsdatum
10.03.2022
Verlag
Springer Healthcare
Erschienen in
Diabetes Therapy / Ausgabe 4/2022
Print ISSN: 1869-6953
Elektronische ISSN: 1869-6961
DOI
https://doi.org/10.1007/s13300-022-01220-4

Weitere Artikel der Ausgabe 4/2022

Diabetes Therapy 4/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

„Jeder Fall von plötzlichem Tod muss obduziert werden!“

17.05.2024 Plötzlicher Herztod Nachrichten

Ein signifikanter Anteil der Fälle von plötzlichem Herztod ist genetisch bedingt. Um ihre Verwandten vor diesem Schicksal zu bewahren, sollten jüngere Personen, die plötzlich unerwartet versterben, ausnahmslos einer Autopsie unterzogen werden.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Schlechtere Vorhofflimmern-Prognose bei kleinem linken Ventrikel

17.05.2024 Vorhofflimmern Nachrichten

Nicht nur ein vergrößerter, sondern auch ein kleiner linker Ventrikel ist bei Vorhofflimmern mit einer erhöhten Komplikationsrate assoziiert. Der Zusammenhang besteht nach Daten aus China unabhängig von anderen Risikofaktoren.

Semaglutid bei Herzinsuffizienz: Wie erklärt sich die Wirksamkeit?

17.05.2024 Herzinsuffizienz Nachrichten

Bei adipösen Patienten mit Herzinsuffizienz des HFpEF-Phänotyps ist Semaglutid von symptomatischem Nutzen. Resultiert dieser Benefit allein aus der Gewichtsreduktion oder auch aus spezifischen Effekten auf die Herzinsuffizienz-Pathogenese? Eine neue Analyse gibt Aufschluss.

Update Innere Medizin

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