Exp Clin Endocrinol Diabetes 2022; 130(S 01): S151-S184
DOI: 10.1055/a-1624-5095
German Diabetes Association: Clinical Practice Guidelines

Dietary recommendations for persons with type 2 diabetes mellitus

Thomas Skurk
1   ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
2   Else Kröner-Fresenius-Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
,
Anja Bosy-Westphal
3   Institute for Human Nutrition, Faculty of Agricultural and Nutritional Sciences, Christian-Albrechts-University of Kiel, Kiel, Germany
,
Arthur Grünerbel
4   Diabetes Center Munich South, Munich, Germany
,
Stefan Kabisch
5   German Institute of Human Nutrition Potsdam-Rehbrücke, Potsdam, Germany
6   Department of Endocrinology, Diabetes and Nutritional Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
7   German Center for Diabetes Research (DZD), Munich, Germany
,
Winfried Keuthage
8   Focus Practice for Diabetes and Nutritional Medicine, Münster, Germany
,
Peter Kronsbein
9   Department of Ecotrophology, Niederrhein University of Applied Sciences, Mönchengladbach Campus, Germany
,
Karsten Müssig
10   Department of Internal Medicine, Gastroenterology and Diabetology, Niels Stensen Hospitals, Franziskus Hospital Harderberg, Georgsmarienhütte, Germany
,
Andreas F.H. Pfeiffer
6   Department of Endocrinology, Diabetes and Nutritional Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
,
Marie-Christine Simon
11   Institute of Nutrition and Food Sciences, Rheinische Friedrich-Wilhelms University, Bonn, Germany
,
Astrid Tombek
12   Diabetes Center Bad Mergentheim, Bad Mergentheim
,
Katharina S. Weber
13   Institute of Epidemiology, Christian-Albrechts-University of Kiel, Kiel, Germany
,
Diana Rubin
14   Vivantes Hospital Spandau, Berlin, Germany
15   Vivantes Humboldt Hospital, Berlin, Germany
› Author Affiliations
 

Preface

This practice guideline is aimed at all professional groups caring for people with type 2 diabetes mellitus (T2Dm). In addition to the multifaceted aspects of nutrition in diabetes, there is a particular call for individualization of therapy, counseling, empowerment, and diabetes self-management [1] [2] [3]. Therefore, the Nutrition Committee of the DDG has set the goal to compile practice guidelines on nutrition as target group-specific as possible with the highest available evidence. In doing so, it is considered necessary to treatment forms separately presentation since the therapeutic significance of nutrition differs significantly in each case and must be seen against the background of different drug therapy components.

T2Dm is characterized by a progressive course in terms of β-cell insufficiency, which progresses at different rates in different individuals [4] [5] [6] [7]. Against this background, patients with T2Dm have both quite different characteristics and treatment regimens [8].

For patients with special life circumstances, e. g., sarcopenia and need for long-term care, diets must be designed taking strong consideration of personal preferences and with an emphasis on meeting protein requirements.

Overall, as a result, nutritional therapy needs to be highly individualized to realize its full potential.

The option of individualized nutritional counseling, including via telemedicine, should therefore be used more widely and intensively in people with T2Dm. The general goals are to promote balanced eating habits, provide training on appropriate portion sizes, and address individual dietary needs while maintaining enjoyment of food and providing practical tools for meal planning. Individualized nutrition counseling sessions include evidence-based topics that should be provided by qualified and appropriately certified nutrition professionals (dietitian, nutritionist or ecotrophologist).

The nutritional therapy plan must also be coordinated and continuously aligned with the overall management strategy, including medications administered, physical activity, etc.

In addition, people with prediabetes and excess weight/obesity should be referred to an intensive lifestyle intervention program that includes individualized goal-setting components, as defined, for example, by the S3 Guideline Prevention and Therapy of Obesity (S3-Leitlinie Prävention und Therapie der Adipositas). Since this service is not yet a standard benefit of the statutory health insurance, at minimum individualized nutrition counseling should be provided with partial cost coverage according to § 43 German Social Security Code (SGB).

Another important recommendation is the referral of adults with diabetes to comprehensive diabetes self-management training and support (Diabetes-Selbstmanagementschulung und -unterstützung - DSMES) according to national standards.

This practice guideline represents the summary and evaluation of the literature by the Nutrition Committee of the DDG on selected nutritional aspects in the management of T2Dm. Regular updating and, if necessary, supplementation is planned. In doing so, the evidence - if available - was assessed in the context of literature research based on systematic reviews or meta-analyses. Original papers were also used for topics without the availability of such reviews.


#

Body weight recommendations

General recommendations

RECOMMENDATION
  • In cases of excess weight, the goal of weight reduction should generally be pursued.

  • Weight cycling should be avoided.

Comment

With age comes a weight gain leading to an increase in BMI of 5 points and is associated with a 3-fold (weight gain between 18 and 24 years) or 2-fold (weight gain≥25 years) higher risk of T2Dm [9]. Obesity alone is an independent risk factor also for coronary heart disease (CHD). Moderate weight reduction, on the other hand (5–10% of current weight), reduces risks such as insulin resistance, hyperglycemia, and dyslipidemia [10] and can reduce secondary complications. A very-low-calorie diet (VLCD; 624 kcal/d) for 8 weeks can also lead to a temporary diabetes remission of at least 6 months [11]. The effectiveness of a VLCD diet is greater with a shorter duration of diabetes and with higher fasting insulin and C-peptide levels [12]. Intensive weight management with a mixed diet and lifestyle intervention also leads to sustained remission [13]. In this context, a stable body weight seems to be associated with a better cardiovascular outcome than a high weight variability [14] [15] [16]. Weight gain or weight variability in T2Dm is associated with higher mortality [15] [17].

However, especially in elderly patients, greater weight loss (>25%) is associated with loss of muscle mass [18]. Studies also show that individuals with T2Dm with a normal weight have higher mortality than those with higher body weight [19] [20], which has been repeatedly described as the obesity paradox [21]. A possible explanation for this effect is a larger, more metabolically-active muscle mass in obese patients [22]; this must be factored into weight goals and, if necessary, included in a physical activity program for muscle maintenance [23].


#
#

Quantitative statements on targeted weight reduction, diabetes remission

RECOMMENDATION
  • The extent of weight reduction is based on individual therapy goals. For diabetes remission, a target weight reduction of 15 kg of baseline overweight/obesity weight should be aimed for.

Comment

The association of obesity with all components of the metabolic syndrome makes weight reduction a priority therapeutic goal. The normal and realistic consensus was a 3–5 kg weight reduction in the context of dietary and exercise behavior modification. Achieving these goals allowed a reduction in T2Dm manifestation of about 60% in people with prediabetes and has been demonstrated in large studies [24]. A greater weight loss of 10 kg was significantly more effective and prevented diabetes manifestation in over 90% of study participants [25] over 3 years.

Remission of T2Dm after an average of 5 years of diabetes duration and 1 year of intensive lifestyle modification program with 8.9% weight reduction (baseline BMI 35 kg/m²) was 11.5% in the Look Ahead trial. After 4 years, weight reduction was still 4.7% of baseline weight, and 7.3% showed remission defined as fasting blood glucose below 126 mg/dl without diabetes medications [26].

In the DIRECT study, a weight reduction of 15 kg with formula diets resulted in an 86% remission of T2Dm after a maximum of 6 years of previous diabetes. Lower weight loss resulted in greatly lower success rates, but only a few patients were able to achieve significant weight loss. The data shows a quantitative effect of weight loss on diabetes remission [13]. Patients should therefore be offered appropriate therapy as early as possible after diagnosis of T2Dm [21].

What is the role of the weight loss strategy of a formula diet versus slow moderate weight loss? In the long term, the likelihood of regaining weight after cessation of the diet program is more than 80%. Formula diets result in faster and more significant weight loss and still show greater weight loss in the long term [27].

Weight loss leads to rapid improvement in hepatic insulin resistance, so that blood glucose levels decrease rapidly while the insulin secretory capacity remains unchanged. With insulin therapy and insulin resistance, insulin levels must be reduced rapidly (1–5 days), often by two-thirds of the initial dose. The patient must either be prepared for this or the therapy should be performed as an inpatient for the first few days, and as an outpatient only with daily patient contact.


#
#

Using telemedicine for type 2 diabetes mellitus

RECOMMENDATION
  • Telemedicine applications can support the implementation of behavioral modifications recommended in the treatment of T2Dm.

  • Telemedicine can increase adherence to weight loss programs and accessibility.

Comment

The COVID-19 pandemic has increased the need for digital consultation methods in the therapy of diabetes mellitus. Telemedicine refers to the use of audio-visual communication technologies for the purpose of diagnosis, consultation, and emergency medical services [28]. Telemedicine care for diabetes patients had already been used before the COVID-19 pandemic and has established itself as a proven form of therapy.

As part of a telemedicine program, therapy-relevant data (e. g., blood glucose level, insulin dose, body weight) is transmitted to the healthcare professional, whereupon the patient receives feedback. A distinction is made between telemedical therapy via text messages/e-mail and via telephone/video conferencing.

A meta-analysis by Su et al. from 2015 with 92 included studies showed a significant reduction of the HbA1c value in type 1 and type 2 diabetes patients through telemedical nutrition therapy [29]. However, no significant difference was found between telemedicine programs via messaging (cell phone or email) and a face-to-face consultation (telephone call or video conference).

In Germany, a randomized controlled trial by Kempf et al. reported a 0.6% lower HbA1c value and a 5 kg greater weight reduction at the 1-year follow-up in the telemedicine-assisted group vs. standard therapy [30].

Telemedical applications can be prescribed by physicians and psychotherapists and reimbursed by the statutory health insurance companies if they are included in the Federal Institute of Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte - BfArM) directory as digital health applications (Digitale Gesundheitsanwendungen - DiGA). This is regulated in the Digital Health Care Act (Digitales Versorgungsgesetz - DVG), which came into effect in December 2019. Digital health applications are usually used by the patients on their own. However, it is also possible for patients and providers to make use of digital health applications together, for example in the form of teleconsultation or chats. At the time of publication of these practice guidelines, no digital health applications with the indication “diabetes” are listed in the BfArM directory, but several diabetes digital health applications (Diabetes-DiGA) are currently being evaluated.

The DiGA “Zanadio” with the indication “obesity” is provisionally included in the BfArM directory. Zanadio works on the basis of the guideline recommendations for the therapy of obesity and supports a conservative obesity therapy consisting of exercise, diet and behavioral change. Zanadio includes telemedicine elements in that users are supported by a dietitian via a chat function.

An example of a telemedicine application - though not approved as a digital health application - is the TeLiPro telemedicine lifestyle intervention program. In this program, patients are provided with an app that is used to monitor lifestyle activities. Bluetooth-compatible blood glucose meters, scales, blood pressure monitors and pedometers are used for this purpose. A cloud enables the diabetes coach (diabetes advisor) to view the data and interact directly with the patient via a chat function or by telephone.

In the TeLiPro study, both groups received the app, scales, pedometers, blood glucose and blood pressure monitors. However, the groups differed in that a diabetes coach was only available to patients in the intervention group [29].

As a result, it can be seen that the intervention group, in contrast to the control group, had a significant reduction in HbA1c (mean±SD −1.1±1.2% vs. −0.2±0.8%; P<0.0001). There was also a reduction in weight (TeLiPro −6.2±4.6 kg vs. control −1.0±3.4 kg,

BMI (−2.1±1.5 kg/m2 vs. −0.3±1.1 kg/m2). Furthermore, the intervention group reported a generally better quality of life as well as a better nutritional status [30].


#
#
#

Strategies for weight reduction and weight maintenance

RECOMMENDATION
  • Weight reduction must be clearly indicated before it is recommended. Higher age is a risk factor for sarcopenia and cardiometabolic disadvantages from hypocaloric diets.

  • Close follow-up with dietary counseling is necessary to facilitate good long-term adherence.

  • The weight loss strategy should match the preferences of the overweight person (individualized nutrition therapy).

  • The strategy for sustainable stabilization of a reduced body weight should be coordinated on an individual basis with the affected person.

  • To date, no dietary pattern is clearly superior to other dietary patterns in weight reduction.

Comment

Various forms of hypocaloric dietary modification - ranging from long-term useful procedures to procedures limited to short interventions - lead to a reduction in body weight in T2Dm patients and often also to an improvement in metabolic status and other cardiovascular risk factors. However, only a few patients succeed in achieving significant long-term weight loss, both with complex lifestyle intervention and with formula diets. Thus, to date, the true goal – diabetes remission, as well as reduction in actual long-term risk for cardiovascular morbidity and mortality – remains achievable at best for subgroups that are difficult to define [13] [31]. Bariatric procedures are also successful in diabetes remission, but are also subject to strict criteria for indication [32].

Numerous strategies have evolved for weight loss, differing in approach in terms of daily energy intake (low-calorie diet [LCD]/VLCD), nutrient ratio (low-fat/low-carb), consistency (common foods/formula drinks), preference for an omnivorous or vegetarian/vegan diet, as well as the limiting of fasting and eating times (intermittent fasting).

The effects of these respective approaches are continually published and championed. However, there is no strategy that is fundamentally superior to another. It depends on personal preference as to which method (or combination of methods) the person wanting to lose weight prefers and which method motivates him or her to implement it sustainably in everyday life [2] [3].

In most studies, it has not been conclusively clarified as to how the targeted, and ultimately achieved, weight reduction is actually decisive or necessary for the obtaining results [33]. Dietary modifications without weight reduction also sometimes achieve dramatic improvements. A systematic head-to-head comparison of hypo- and isocaloric diets with the same macronutrient ratio is rarely described in literature. Meta-analyses find little long-term metabolic benefit for a primary weight loss intervention compared with standard therapy, albeit with considerable heterogeneity among studies [34].

Maintenance of long-term weight loss is strongly influenced by the ability to adhere to the diet program. Behavioral support can significantly improve outcomes. There are individual differences in response to each diet that are greater than the difference in mean weight loss between comparison diets.


#

Interaction between diet and physical activity

RECOMMENDATION
  • A high level of low-intensity physical activity (e. g., brisk walking) after meals improves body weight regulation and is beneficial for glycemic control.

Comment

While inactivity or a predominantly sedentary lifestyle pose a risk for excessive caloric intake and thus for the development of obesity [35] [36] [37], a high level of physical activity even at a low intensity (e. g., fast walking) ensures a better adaptation of appetite to energy demand [38] [39] and thus improves the regulation of body weight even independently of a higher caloric expenditure [38].

Additionally, exercise type, intensity, and timing (fasting or postprandial) have an impact on glycemic regulation [40]. In this regard, the intensity of physical activity correlates positively with the improvement of insulin sensitivity, and the best results are obtained by a combination of strength and endurance training [40]. There is evidence that high-intensity exercise (e. g., high-intensity interval training-HIIT) best improves glycemic control when fasting (i. e., when substrate availability is low) [40]. However, the effectiveness and safety of this method in patients with T2Dm need further investigation. In contrast, low-intensity physical activity is safe and effective in improving glycemia in patients with T2Dm, especially when substrate availability is high. Accordingly, fast walking after eating has a beneficial effect on postprandial glycemia by improving insulin-independent glucose uptake [41] [42] [43] [44] [45] [46].


#
#

Reducing carbohydrates (low-carb)

RECOMMENDATION
  • For weight reduction, a moderate reduction in carbohydrates is recommended as a possible method, especially in the short term (e. g., traditional Mediterranean, plant-based).

  • Carbohydrates should preferably be consumed in the form of whole grains, legumes, and nuts.

  • For weight maintenance, low-carb are probably on par with low-fat diets and should be chosen according to individual preference.

  • Low-carb diets in particular can only be implemented in individuals with insulin therapy under close therapy monitoring.

Comment

Carbohydrates account for an average of about 45% of energy intake in the diet of Germans, including about 90 grams of sugar (=18 energy percent [E%]) and often mainly rapidly metabolized polysaccharides. Epidemiologically, there is increased mortality with carbohydrate intakes greater than and less than 50% (the latter only with animal-emphasized protein sources) [47]. Reducing carbohydrates as part of a dietary intervention almost invariably leads to weight loss and metabolic changes. The scientific literature mostly considers low-carbohydrate diets in juxtaposition to low-fat diets. Carbohydrate reduction can be classified as moderate-carb, low-carb, or very-low carb, depending on the intensity; according to this, the traditional Mediterranean diet is also a low-carbohydrate diet [42].

American Diabetes Association (ADA) and European Association for the Study of Diabetes (EASD) classify low-carb as a dietary therapeutic option, but classify the Mediterranean diet as superior [48]. This consensus reflects the state of knowledge from recent meta-analyses: among all dietary models defined in terms of food quality examined in randomized controlled trials (RCTs), the traditional Mediterranean diet performs best for fasting glucose and lipid profile, and is among the top 3 diets for HbA1c levels, blood pressure, and weight reduction, respectively. Low-carb is the most effective method for reducing HbA1c levels and body weight; in reducing fasting glucose, blood pressure, and blood lipids, this diet is also very successful and more effective than low-fat [49] [50] [51]. However, with prolonged use, low-carb and low-fat converge in their effect; whether this is due to waning adherence or a failure of the metabolic response cannot be answered at present [52].

A very recent meta-analysis also highlights that low-carb (<26 E% or<130 g carbohydrate [carbs]/day) may be superior to low-fat in diabetes remission. After 6 months, significantly more patients achieve HbA1c levels below 6.5% with low-carb; the differences are not significant when the additional criterion of no medication or longer intervention is applied [53].

Looking at the effect of specific food groups on the overall metabolic picture of all cardiovascular risk parameters, among 66 food categories, nuts, legumes, and whole grains (all of which are carbohydrate carriers) performed best [50]. Isocaloric replacement alone of different digestible carbohydrates with each other produces only relatively small effects on fasting glucose and low-density lipoprotein (LDL) cholesterol (sugar replaced with starch), as well as homeostasis model assessment-insulin resistance (HOMA-IR) and uric acid (fructose replaced with glucose). The evidence of these results is, however, considered to be low [54]. Effects on inflammatory parameters are not observed [55].

Overall, the traditional Mediterranean diet is to be regarded as a specific representative of “low-carb” as an optimal dietary form. More generally, “low-carb” and “low-fat” are metabolically equivalent after a few months of intervention at the latest [55]. According to current knowledge, there is no clear long-term optimum for the energy content of carbohydrates. Patients whose personal preference strongly leans toward one of these dietary variants can use it. However, depending on the intensity and dynamics, additional intermediate metabolic controls are recommended to detect an individually unpredictable derailment of glycemia and insulin resistance, lipid metabolism or uric acid levels at an early stage [50].


#
#

Reducing fats (low-fat)

RECOMMENDATION
  • A low-fat diet cannot be recommended to individuals with T2Dm as a generalization.

Comment

As described in the section on carbohydrate reduction, reducing dietary fats alone is associated with inferior outcomes compared with all low-carbohydrate diets for weight reduction, blood pressure reduction, and optimization of triglycerides and glycemic parameters [48] [51] [52] [56]. Compared to diabetes prevention by complex lifestyle intervention and a low-fat approach has been consistently shown [24], the chance of diabetes remission by low-fat dietary change as a sole intervention is comparatively small [26] [57].

Trans fats

RECOMMENDATION
  • Industrially-produced trans fats should continue to be avoided; natural trans fats are probably not problematic.


#
#

Comment

The quality of the fat also has a relevant influence on the glycemic metabolic state. In observational studies, industrially-produced trans fats have been shown to increase mortality, in particular by increasing the risk of CHD. An increased risk of diabetes is not described [58].

Natural trans fats, such as those found in beef and dairy products, are associated with decreased diabetes risk in epidemiological studies and do not affect the risk of cardiovascular mortality or morbidity [58].

Saturated fats

RECOMMENDATION
  • Foods with a natural saturated fat content are safe if consumed in moderation. Highly processed products with added saturated fats should be avoided.


#
#

Comment

The discourse on saturated fats has not reached a definitive conclusion even in 2021. The criticism of saturated fats (sometimes even erroneously all fats) fueled by the Seven Countries Study and many epidemiological follow-up surveys is no longer justified in recent meta-analyses on cohort studies [58]. The evidence regarding a potential for harm from saturated fat is insufficient [59]. Even butter, as a typical food with very high levels of saturated and total fat, epidemiologically only increases mortality minimally, but does not affect cardiovascular risk and is more associated with lower diabetes risk [60]. Other high-fat or low-fat dairy products also have little adverse effect on metabolic outcomes [61].

RCTs on low-fat diets show a mean slight reduction in body weight, BMI, body fat percentage, and waist circumference [62], but no effect on CHD, cardiovascular mortality, or all-cause mortality [63]. A reduction in saturated fat consistently has a beneficial effect on the inflammatory phenotype [31] [64]. It has also been shown to lower LDL levels but worsen HDL and triglyceride levels [65].

Unsaturated fats

RECOMMENDATION
  • High levels of unsaturated fatty acids should be targeted in patients with T2Dm, regardless of total fat, through intake of natural foods but not supplements.


#
#

Comment

Observational studies describe clear diabetes- and cardioprotective associations for monounsaturated and polyunsaturated fatty acids, especially for linoleic acid and alpha-linolenic acid [66] [67] [68].

In intervention studies, evidence of cardio-protection and mortality reduction is lacking for polyunsaturated fatty acids (PUFAs) omega-6 fatty acids and non-long-chain plant omega-3 PUFAs [69] [70]. Moreover, in meta-analyses of randomized controlled trials, no glycemic benefit is seen for unsaturated fatty acids when compared against saturated fatty acids [71]. Compared with carbohydrates, monounsaturated fatty acids (MUFAs) are beneficial in all metabolic axes except for blood pressure [72] [73]. Compared to saturated fats or placebos, there is a benefit in waist circumference, inflammation, triglyceride levels, platelet aggregation, and probably fatty liver (omega-3 fatty acids) [74] [75] [76] [77] [78]. A high omega-3/omega-6 fatty acid ratio may play a beneficial role in people with diabetes and during prolonged intervention, particularly in lowering insulin but not glucose levels [79] [80]. Women appear to benefit more markedly than men [81]. There is no clear interventional benefit for alpha-linolenic acid with respect to diabetic metabolic status [82].


#
#

Intermittent fasting/interval fasting

RECOMMENDATION
  • Intermittent fasting can be used as a means of weight reduction under medical supervision.

  • No general recommendation can be made for any form of interval fasting.

Comment

In addition to the qualitative adjustment of the diet through a modified nutrient profile or targeted redistribution of food groups, meal frequency is also considered a starting point for weight reduction and metabolic improvement.

Randomized trials of daily meal frequency show a small benefit in favor of less frequent meals (1–2 vs. 6–8) with respect to body weight, fat mass, and waist circumference. However, these effects are of low overall evidence [82].

Less frequent food intake prolongs lifespan in some animal models. Observational studies in humans (e. g., in the context of Ramadan) see only relatively small metabolic changes in healthy individuals, and moreover, these changes are transitory [83] [84] [85]. In diabetics, metabolic deterioration is also described. Further cohort studies describe a less frequent occurrence of coronary heart disease and [86] [87].

Targeted, long-term, regular skipping of meals according to a fixed chronological pattern (interval fasting) comprises different variants: alternate day fasting, 5:2 fasting, and time-restricted eating (e. g., 16:8 fasting). These are sometimes compared in literature together with continuous caloric restriction or even unchanged control diets.

In all meta-analyses on interval fasting (8 meta-analyses on over 40 RCTs), no superiority of interval fasting over continuous calorie restriction is found. Compared with an unchanged control diet, there is a significantly greater reduction in body weight, waist circumference, blood pressure and triglycerides, but not in LDL cholesterol, fasting glucose or HbA1c [88] [89] [90] [91] [92]. RCTs with T2Dm patients are scarce. These show the same pattern of desired outcomes as in the aforementioned meta-analyses, but an increased risk of hypoglycemia [93] [94] [95] [96] [97].


#
#

Meal replacements/formula diets (with/without multimodal program)

RECOMMENDATION
  • Low-calorie formula diets allow clinically-relevant weight loss in people with T2Dm, associated with significant improvement in glucose and lipid metabolism and reduction in other cardiovascular risk factors.

Comment

Replacing meals with low-calorie formulary diets is a safe and effective weight loss intervention in overweight and obese individuals with T2Dm compared with conventional calorie-restricted diets. In addition to favorably affecting anthropometric parameters such as waist circumference and body fat mass, formula diets also improve other cardiometabolic risk parameters such as blood pressure, fasting glucose, HbA1c level, and lipid metabolism [98] [99] [100] [101]. In weight loss programs, the use of formula diets results in pronounced weight loss similar to that seen after bariatric surgery, associated with sustained diabetes remission. However, only 25% achieve a weight reduction of>15% at which remission is very likely to occur [13] [102].


#

Scientific background

Obesity is one of the most important risk factors for the development of T2Dm [103]. Overweight or obesity exists in 60–90% of patients with T2Dm [104] [105]. In contrast, weight loss leads to an improvement in glucose and lipid metabolism and a decrease in elevated blood pressure levels. Thus, weight loss in patients with T2Dm represents one of the most important therapeutic measures [105]. However, weight loss, already challenging for people without diabetes, is often further complicated in people with T2Dm because of genetic and metabolic differences, fear of hypoglycemia, glucose-lowering therapies that promote weight gain, decreased physical activity, and diet fatigue. Low-calorie diets have the potential to result in weight loss similar to bariatric surgery in people with T2Dm. A meta-analysis of 9 studies examining the effects of very-low-energy diets (VLED) in a total of 192 obese people with T2Dm found that participants had lost 9.6% of baseline weight after 6 weeks and fasting glucose had reduced by 50% after only 2 weeks [106]. However, many people with T2Dm find it difficult to make longer-term lifestyle changes aimed at weight loss, and motivation may be rapidly lost in the absence of short-term intervention success. Low-calorie formula diets have now been shown in numerous studies to be a safe and effective treatment option to improve cardiometabolic endpoints such as waist circumference, body fat mass, blood pressure, and HbA1c levels in obese patients with T2Dm [98] [99] [100] [101]. A meta-analysis including 4 studies with a total of more than 500 study participants found that weight loss resulting from low-calorie formula diets, providing between 300 and 1000 kcal of energy per day, was similar for both people with T2Dm and without diabetes, with a mean weight loss between 8 and 21% of baseline weight after a treatment period of 4–52 weeks. There was also no difference in the rate of weight loss between people with (−0.6 kg per week) and without T2Dm (−0.5 kg per week) [107]. In another study, there was also no difference in weight loss after starting a low-calorie formula diet between patients with and without diabetes. One fifth of the participants achieved a weight loss of more than 15 kg after 12 months. Among participants who continued the weight management program beyond one year, nearly 40% had a weight loss of at least 15 kg after 24 months [108]. Weight loss resulting from temporary use of a low-calorie formula diet is associated with longer-term improvement in glucose and lipid metabolism and blood pressure [109]. Also, in patients with inadequate metabolic control, meal replacement with a formula diet can lead to a clinically-relevant decrease in HbA1c and a substantial reduction in insulin doses in patients on intensified conventional insulin therapy [110] [111]. Diabetes remission also appears possible as a result of strict caloric restriction, as suggested by the results of the Diabetes Remission Clinical Trial (DiRECT) [112]. Nearly half of the overweight and obese patients with T2Dm who initially received only a formula diet of 825 to 853 kcal per day for 3–5 months achieved diabetes remission in contrast to only 4% of patients who received only standard therapy from their primary care physician [13]. After 12 months, one quarter of the intervention group had achieved the stated goal of losing 15 kg or more and no participant in the control group. Diabetes remission was very closely associated with weight loss. While remission did not occur in any of the patients who gained weight, the remission rate was 86% in participants who lost at least 15 kg. Two years after the intervention, more than one-third of patients with T2Dm were still in remission. In participants who had lost more than 10%, the remission rate was as high as 64% [102]. Even in people with an increased risk of diabetes due to overweight or obesity and at least one other metabolic syndrome comorbidity, additional meal replacement with a decreasing frequency formula diet over the study period was superior to lifestyle intervention alone in terms of weight loss and improvement of cardiometabolic risk factors [113]. In addition, conversion from prediabetes to normoglycemia was achieved in half of the participants who also received a formula diet, whereas this was the case in less than one-third of the participants treated with lifestyle intervention alone [114].


#
#

Additional aspects of weight reduction in insulin-treated T2Dm

RECOMMENDATION
  • Insulin therapy should be limited to what is necessary due to the anabolic effect of the hormone. Weight loss under insulin therapy is more difficult.

Comment

In addition, insulin therapy often leads to weight gain in patients with diabetes, most of whom are already overweight: the United Kingdom Prospective Diabetes Study (UKPDS), in which T2Dm patients treated with insulin were randomized, showed an average weight gain of 6.5 kilograms [115]. Despite insulin therapy, lifestyle intervention remains a very important therapeutic component [116].

However, another study showed that the higher the baseline BMI of the patients, the lower the weight gain. When the HbA1c value decreased by one percentage point, weight increased by an average of 1.24 kg in those with normal weight (BMI less than 25 kg/m2), but weight decreased by as much as 0.32 kg in those with severe obesity (BMI greater than 40 kg/m2) [117].


#
#
#

Summary evaluation and outlook

There are a number of methods to choose from for weight reduction or weight stabilization. There is more or less good evidence for each of these methods. In our view, the focus must be placed on the individual preferences of the patients, which strengthen adherence to the respective therapy method regardless of the outcome.


#

Dietary patterns

RECOMMENDATION
  • For diabetes management and reduction of the risk of cardiovascular complications in individuals with T2Dm, a variety of dietary patterns is acceptable, such as a Mediterranean, vegetarian, or vegan diet.

  • There is currently insufficient evidence to recommend the DASH diet, the Nordic dietary pattern, and the Paleo diet specifically for the treatment of T2Dm.

  • Until additional evidence is available on the superiority of a specific dietary pattern related to diabetes therapy target parameters, individuals with T2Dm should be guided by the commonalities of the dietary patterns mentioned: choosing non-starchy vegetables and low-processed foods and avoiding refined sugars and highly-processed grains.

Comment

Based on current evidence, there is no dietary pattern that could be universally recommended for all affected individuals with T2Dm. Instead, according to the recommendations of professional societies, different dietary patterns such as the Mediterranean diet or a vegetarian or vegan diet are suitable to achieve the target parameters of diabetes therapy [2] [118] [119] [120]. While the evidence for the effects of the Mediterranean diet in individuals with T2Dm is primarily based on RCTs (including several larger trials and longitudinal studies) and their systematic reviews and meta-analyses [121], the RCTs on vegetarian and vegan diets mostly have small case numbers and short study durations [121] [122] [123] [124]. The currently-available evidence on the DASH diet, the Nordic dietary pattern [125] [126] [127], the Paleo diet [2], and the macrobiotic diet [124] [128] in individuals with T2Dm is small and partly contradictory, so that further studies are needed to support observed beneficial effects of these dietary patterns for diabetes management in T2Dm.

In individuals with newly-diagnosed T2Dm, the Mediterranean diet achieved a weight loss of≥5%, which was considered clinically relevant [129]. Similarly, further meta-analyses from RCTs in individuals with T2Dm found significantly greater weight loss for the Mediterranean diet compared with the respective control diets [130] [131] [132]. Adherence to a vegetarian or vegan diet also resulted in weight loss in individuals with and without T2Dm [133] [134] [135] [136].

Based on a network meta-analysis of 56 RCTs and 9 dietary patterns [136] and evidence from several meta-analyses of RCTs [131] [132] [137], the Mediterranean diet is superior to the respective control diets in reducing HbA1c and most effective after low-carb diets in reducing HbA1c and fasting blood glucose, followed by the Paleo diet and vegetarian diet [49] [131] [132] [137]. Other systematic reviews and meta-analyses confirmed the positive effects of vegetarian and vegan diets on glycemic control in individuals with and without T2D [123]. However, all other diets studied in the network meta-analysis also significantly reduced HbA1c and fasting blood glucose in individuals with T2Dm compared with control diets, and the overall results were rated with very low to moderate credibility and strength of evidence because of significant inconsistencies [49]. Thus, superiority of one dietary regimen over the others in terms of glucose parameter reduction cannot be inferred at this time [138]. In addition, further studies are needed to confirm the effects of dietary patterns on glycemic control in individuals with T2Dm independent of weight loss [120] [122] [138] [139] and which examine differences between vegetarian and vegan dietary patterns [122] [123].

In addition to positive effects on weight loss and glycemic control, dietary patterns could also reduce the incidence and mortality of various cardiovascular outcomes and improve individual cardiometabolic risk factors such as dyslipidemia and arterial hypertension in individuals with and without T2Dm [122] [123] [131] [132] [140]. The available evidence on this is low to moderate for the Mediterranean diet and very low to low (incidence and mortality) and low to moderate (risk factors) for the vegetarian/vegan diet and the Dietary Approaches to Stop Hypertension (DASH) diet, respectively. For the Nordic dietary pattern, only a preliminary study assessment is available to date, indicating very low evidence for reduction in incidence and mortality from coronary heart disease [123] [140]. A meta-analysis based on 52 RCTs and 9 dietary patterns concluded that, with low to moderate evidence, the Mediterranean diet was most effective in increasing HDL cholesterol and reducing triglycerides compared with control diets, whereas the vegetarian diet was most effective in reducing LDL cholesterol compared with control diets [51]. For effects of the Mediterranean, vegan, and vegetarian diets on microvascular complications associated with T2Dm, the evidence is limited to a few studies with small subject numbers. Based on surrogate parameters, improvements are suggested for nephropathy and retinopathy with adherence to the above dietary patterns, whereas the evidence for risk of microvascular complications is insufficient and results for neuropathy are inconsistent [122]. Overall, based on the available evidence, it is thus difficult to draw solid conclusions for the effects of dietary patterns on microvascular and macrovascular complications in individuals with T2Dm [122].

Based on the available evidence, because no dietary pattern is superior to others, individualized meal planning focusing on dietary patterns rather than individual nutrients or individual foods (or the factors common to dietary patterns) is recommended [2] [3] [118].


#
#

Singular effects of individual nutrients

Protein

Effect on glycemia

RECOMMENDATIONS

  • We recommend a protein intake of 10–25% of dietary energy (%E) for patients with T2Dm younger than 60 years and 15–25% for those older than 60 years with intact renal function (glomerular filtration rate [GFR]>60 ml/min/m²) and a constant weight.

  • In impaired renal function of any stage, protein reduction to less than 0.8 g/kg body weight is unlikely to be beneficial and should be avoided because of the risk for malnutrition, especially in higher-grade renal failure.


#

Comment

A detailed AWMF S3 guideline on protein intake in T2Dm can be found at the following Internet address: [141]. A meta-analysis has been published and is freely available [142].

Protein is required as a supplier of amino acids in a minimum amount of about 0.8 g/kg body weight or 10 E% to avoid malnutrition and sarcopenia. The lower limit of 0.8 g/kg/day may be insufficient for the elderly because of decreasing efficiency of protein synthesis [143], so a higher protein intake of at least 1 g/kg body weight/day is recommended [144].

The importance of a higher protein intake is controversial. Arguments for higher protein intake include better satiety and higher energy expenditure through postprandial thermogenesis, which may counteract weight gain. Protein metabolism requires considerably less insulin than carbohydrates, which facilitates blood glucose control and may simplify insulin dosing. However, a certain amount of insulin is required because of the protein-induced release of glucagon [145]. Elderly people often experience significant muscle loss due to disease, glucocorticoid therapy, immobility, or loss of appetite, which is why geriatricians also recommend a higher protein intake [146].

Arguments against higher protein intake arise from observational epidemiological studies describing higher mortality [143] [147] and diabetes incidence [148] with higher protein intake. Because they do not adequately account for lifestyles and other variables, the conclusions of these observational studies have been called into doubt in Cochrane meta-analyses [148] [149]. Intervention studies consistently show positive effects of higher protein intake in overweight individuals without diabetes [150] [151]. High protein intake above 20 E% versus below 20 E%, for example, approximately 1.2–1.6 g/kg body weight, did not increase the risk of diabetes or other diseases in prediabetes patients in a large European-Australian prospective randomized intervention trial over 3 years [150].

Recommendation for chronic renal insufficiency

Historically, low-protein dietary plans have been recommended to reduce albuminuria and prevent progression of (diabetic) nephropathy.

Recent meta-analyses are available on the issue of protein intake in individuals with diabetes mellitus and chronic renal failure, showing that protein restriction to 0.6–0.8 g/kg body weight does not provide a demonstrable improvement in renal function [152]. Currently, it is still recommended by nephrology societies [153], but not in the consensus paper of the Nutrition Working Group of the American Diabetes Association [2].

Substantial protein restriction to 0.3–0.4 g/kg body weight showed a significant but small reduction in end-stage renal disease (ESRD) but no effect on mortality in the Cochrane analysis [154] [155]. Carrying out such a dietary regimen is exceptionally difficult, leads to a significant deterioration in quality of life, and carries a high risk of malnutrition and sarcopenia, which are associated with increased mortality in stages of ESRD [156]. In addition, the amino acid preparations (keto analogues) to be used as supplements for this extreme form of nutrition cannot be prescribed in Germany.

The consensus paper of the Nutrition Working Group of the American Diabetes Association also does not recommend restricting protein intake in renal insufficiency [2].


#

Recommendation for weight reduction

RECOMMENDATION
  • In the context of weight reduction diets of up to 12 months duration, the protein content can be increased to 23–32% of the total energy intake.


#
#

Comment

Hypocaloric weight reduction diets usually contain a relatively high protein content. Because of the overall reduction in calories, it is usually in the normal range of 0.9–1.2 g/kg body weight, i. e., in the normal to slightly higher range. Numerous comparative studies of higher and lower protein content are available for these diets. Overall, moderate differences in cardiometabolic risk factors due to higher versus lower protein levels have been shown in previous meta-analyses [130] [142] [157]. Although higher protein diets only modestly enhance weight loss, they moderately improve fasting blood glucose levels and systolic blood pressure. Overall, the higher protein diets perform slightly better and show no disadvantages [142].


#
#

Quality of carbohydrates, glycemic index, sugars in highly-processed foods

RECOMMENDATION
  • Selecting low glycemic index (GI) carbohydrates contributes to an improvement of health risks in patients with T2Dm.

  • The influence of GI or glycemic load (GL) in this context is proportionally independent of glycemic regulation and also results in, for example, improved plasma lipids and a higher intake of healthy content such as fiber, micronutrients, and secondary phytochemicals, with lower consumption of detrimental content from highly processed foods with high GI/GL.

Comment

The glycemic index (GI) and the glycemic load (GL) describe the influence of carbohydrate-rich foods on glycemia. The GI indicates how quickly the carbohydrates of a food are digested, absorbed and thus become effective as blood glucose, while the GL adjusts the GI for the amount of carbohydrate consumed. Thus, the blood glucose response of a food depends primarily on characteristics of the food itself (e. g., degree of processing and fat content) [158]. Phenotype characteristics of patients such as the composition of the intestinal microbiome are thought to play a minor role [159] [160] although individual influences have also been observed [161]. A dietary GI≤40 or≤55 is considered low and a GI≥70 is considered high [162]. Prospective observational studies find a positive influence of a low GI/GL diet on the prevention of T2Dm [163] [164]. In patients with T2Dm, a high consumption of low GI foods such as legumes and oats can improve glycemic control, increase insulin sensitivity, and thus reduce insulin requirements [165]. These effects are now explained partly by a positive influence of poorly digestible carbohydrates on the microbiome [166]. To date, the extent to which the benefits of a low-GI diet are explained by its higher fiber content remains controversial. A 6-month intervention with a low-GI diet was slightly better at reducing HbA1c compared with a diet rich in grain fiber (0.5 vs. 0.18%) [167]. However, this study had significant weaknesses because the high-fiber group was asked to avoid high-GI foods, and the low-GI group ultimately had higher fiber consumption than the high-fiber group. In fact, a 12-week substitution of high glycemic carbohydrates with isomaltulose (low-GI) resulted in a reduction in HbA1c and HOMA index in patients with T2Dm [168], indicating an influence of GI independent of dietary fiber content.

Despite convincing evidence on diabetes prevention from observational studies and plausible mechanistic explanations, systematic reviews based on randomized controlled trials on the influence of GI/GL of the diet in patients with T2Dm come to contradictory results. They show both positive [169] [170] and no effects [171] [172] on relevant outcome parameters such as HbA1c level and fasting blood glucose level.

In turn, the result of prospective cohort studies investigating the influence of GI/GL on complications of diabetes is clearer. The risk of CHD showed a clear and dose-dependent relationship with dietary GL or GI [164]. In the group of overweight subjects, the risk for cardiovascular events or mortality due to a high GI is particularly high in this regard [173]. These findings fit with previous results showing a higher risk of fatal and nonfatal cardiovascular events with increasing postprandial glycemia [174] [175]. Characteristic of the dyslipidemia associated with T2Dm are high triglyceride levels, low HDL cholesterol levels, and a high proportion of small dense LDL particles. This lipid pattern can be positively influenced not only by reducing carbohydrate consumption but also by lowering GI/GL [176].

The discrepancy between the results from observational and intervention studies is proportionally explained by the fact that the health assessment of foods based on the GI is inadequate. Carbohydrate quality using GI correlates not only with fiber content, but also with micronutrient content and phytochemical content. At the same time, high carbohydrate quality is associated with lower consumption of highly-processed foods and thus, for example, a lower intake of sugars and saturated fats. High carbohydrate quality therefore has long-term effects on the prevention of diabetes and its complications, independent of the regulation of glycemia.


#
#

Dietary fiber

Dietary fiber, in general

RECOMMENDATION
  • Various dietary fibers from natural sources should be consumed daily.

  • Although there is little evidence to date to support the recommendation of 30 g of dietary fiber per day (15 g/1000 kcal), this represents a valid target for nutritional counseling.


#

Comment

In cohort studies, high intakes of insoluble dietary fiber, particularly grains, are associated with decreased risk of T2Dm, CHD, cancer, and other diseases [51] [177] [178] [179]. Patients with T2Dm also show a dose-dependent reduction in mortality risk [180]. Thus, for T2Dm, whole grain products (bread, rice, pasta) in particular represent a protective food group. Meta-analyses show significant benefits of a higher fiber diet or fiber supplements for body weight, glycemia and insulin resistance, lipid profile and inflammatory status [181], sometimes also for blood pressure [182] even under isocaloric conditions. Even though dietary fiber lowers the glycemic index, it appears to be too imprecise an indicator of recommended foods [181]. Emphasizing “whole grains,” or even better the actual fiber intake, is the most effective and meaningful. Based on an average dietary pattern with 20 grams of fiber, an increase of 15 grams to 35 grams per day is targeted [181].

However, due to the heterogeneity of the studies, resulting among others from the variety of dietary fibers, fiber-containing foods, cohorts and interventions (whole grains, non-grain products, fortified foods, supplements, etc.), a further differentiation of these results is necessary [51] [181].

Insoluble dietary fiber

RECOMMENDATION
  • Carbohydrates should preferably be obtained from high-fiber foods, especially whole-grain products. The benefit of supplementation has not yet been proven.


#
#

Comment

Intervention studies with whole grain products show a glycemic benefit at minimum for rice, but not for wheat and rye products [183]. Apart from a small effect on body weight, no cardiometabolic benefits that can be clearly attributed to whole grain products have been described in meta-analyses [184]. Studies explicitly examining insoluble dietary fiber in an intervention setting are few [185] [186] [187], but none, to date, in patients with T2Dm. Previous data suggests that the tighter the metabolic restriction, the more pronounced the efficacy of dietary fiber [187] [188].

Soluble dietary fiber

RECOMMENDATION
  • High-fiber foods, especially whole grains, but also vegetables, legumes, and low-sugar fruits are recommended in T2Dm and are likely metabolically beneficial. The long-term benefit of supplementation is not established despite consistent short-term effects for glycemia, lipid status, and possibly blood pressure.


#
#

Comment

For soluble fiber, there is insufficient epidemiological evidence for long-term benefit, both in terms of morbidity and mortality.

In contrast to insoluble fiber, however, research on soluble fiber is much more advanced, particularly in the form of supplementation studies. For beta-glucans and psyllium, a minimum of a short- to medium-term (weeks to months) benefit on blood glucose and insulin resistance has been demonstrated; however, long-term data is lacking [189]. Beneficial effects on glycemia and insulinemia have also been systematically described for inulin (specific fructans), especially for women and obese people with T2Dm [190] [191]. However, such studies of a duration of more than 3 months intervention duration are scarce.

The glycemic benefits of inulin and psyllium are probably due to fermentation to short-chain fatty acids, not weight reduction [192]. A mixed effect may be present for beta-glucans [193].

Psyllium, konjac glucomannan, and also beta-glucans also moderately lower LDL cholesterol and triglyceride levels and may therefore provide secondary benefits in T2Dm [194] [195] [196] [197]. No clear metabolic benefits have been demonstrated for other soluble fibers (guar, pectin) [198].

Antihypertensive effects have been described on average for all viscous fibers, but are most expected for psyllium. The effect of 2 mmHg systolic and 0.5 mmHg diastolic is hardly clinically relevant [199].


#
#
#

Nutritional aspects of special populations

Geriatric patients

RECOMMENDATION
  • The nutrition therapy goals for geriatric patients should focus on maintaining independence and avoiding malnutrition and hypoglycemia.

  • Obesity is associated with reduced mortality in this group of individuals and should not be reduced.

Comment

In principle, the nutritional recommendations for elderly people with T2Dm do not differ from those for older metabolically-healthy people or younger people with T2Dm. At the same time, the general nutritional recommendations for this patient group apply to geriatric patients with T2Dm. The consequences of malnutrition in old age, especially in functionally-dependent patients, are severe and should also be in focus for patients with T2Dm. For example, the loss of muscle mass associated with weight loss exacerbates age-related sarcopenia and frailty, thereby promoting disability and loss of independence.

The S2k guideline “Diagnosis, Therapy and Follow-up of Diabetes in the Elderly” (Diagnostik, Therapie und Verlaufskontrolle des Diabetes im Alter) contains very detailed recommendations also on nutritional therapy for older persons with diabetes. It makes clear that therapy goals - also with regard to nutrition - can often change in elderly and especially geriatric patients, but do not have to. Functionality and maintenance of independence are paramount.

Although an improvement in insulin sensitivity could also be achieved in the elderly through intentional weight reduction [200], strict dietary prescriptions should be avoided in the elderly with excess weight or obesity because of the risk of malnutrition. Dietary restrictions that may limit food intake are potentially harmful and should be avoided. If weight loss is considered, dietary measures should be combined with physical activity whenever possible and should focus on meeting protein intake requirements. A significant increase in mortality was found in those over 65 years of age only above a BMI of 30 kg/m2 [200]. Restrictions on the consumption of familiar and favorite foods lead to a reduction in the subjectively perceived quality of life. This aspect is of decisive importance, especially for people of advanced age.

The risk of potential malnutrition is present when there is a persistent reduced food intake (approximately<50% of requirements for more than 3 days) or when several risk factors are present simultaneously that either reduce the amount of food eaten or significantly increase energy and nutrient requirements. The risk of malnutrition can be assessed, for example, using the Mini Nutritional Assessment (MNA) or the corresponding short form (SF-MNA); both screening methods are well evaluated [201] [202]. In underweight patients, the causes should be clarified and corrected if possible.

Nutritional therapy should also focus on the prevention of hypoglycemia, with initial emphasis on medication adjustment for this purpose.

For further discussion, especially for persons with diabetes in nursing homes and when artificial nutrition is required, reference is made to the S2k guideline “Diagnosis, Therapy and Follow-up of Diabetes Mellitus in Older Adults” and the S3 guideline “Clinical Nutrition in Geriatrics” [203] [204] [205].

Due to the complexity of geriatric patients, who are often multimorbid, the planning and implementation of disease-specific diets should, if necessary, be carried out by a multi-professional team including nutrition-specific expertise.


#

Migrants

RECOMMENDATION
  • Medical professionals should ensure that patients have understood the dietary instructions and that their nuclear families are included in the therapy.

  • Medical professionals should ascertain and take into account the individual nutritional concept of the patient and his or her environment (for example, religious aspects, cultural beliefs, the fasting month of Ramadan, pregnancy).


#

Comment

Reference is made to the specific therapy and nutritional aspects of migrants in the DDG Clinical Practice Guideline Diabetes and Migration [206].

There are some very individual eating habits in the context of different cultures and regions. Eating culture is shaped by geographic, historical, sociological, economic, and psychological characteristics of a society and is shared by the members of a given community. Culture represents a fundamental determinant to “what we eat” [207]. Migrants often have different dietary behaviors than natives. They sometimes prefer different foods, often eat more carbohydrates, have different meal concepts, a different understanding of portions, and different food preparation methods and food combinations. Their dietary concepts are usually based on their own traditional cuisine, personal habits, and they also adopt the eating habits of the native population, often resulting in a new “mixed cuisine” [208]. It is not uncommon for special foods to be obtained from the home countries. Migrants from some cultures may have little use for the weights in local recipes when cooking. People have a highly variable postprandial glucose response to identical foods. Individualized culturally sensitive counseling improves adherence [209]. In this context, fasting during Ramadan - food choices and fasting rules influenced by religion - pregnancy, and shift work play a special role. In everyday practice, knowledge of the main sources of carbohydrates and in what form and when carbohydrates are eaten is vital. The practice tool on nutritional patterns [206] [210] of migrants, which was created by the Diabetes und Migration Working Group of the DDG, is intended to provide initial information and assistance. A pragmatic regional breakdown with information on common cuisine forms the basis. In addition to the type (hot/cold) and number of meals, the main sources of carbohydrates and other regional characteristics are presented. Cuisines are quite diverse around the world, and there are further great diversities regionally. It should be taken into account that many beverages have, in the meantime, made their way into many food cultures worldwide, for example soft drinks, energy drinks, various sweetener-enriched beverages and some types of beer.

Possible language barriers and culturally-sensitive communication should be considered when providing nutritional counseling [206]. Individualized, culturally-sensitive counseling therefore improves compliance and treatment success.


#
#
#

Nutritional aspects of special foods and food supplements

Beverages

RECOMMENDATION
  • Individuals with T2Dm should minimize their intake of sugar-sweetened beverages.

Comment

Current evidence-based guidelines from the American and British Diabetes Societies generally recommend a reduction in the consumption of sugar-sweetened beverages for individuals with diabetes to control blood glucose levels and body weight and reduce the risk of cardiovascular disease and fatty liver (Levels of Evidence B and 2, respectively) [2] [118] [119]. Reducing the consumption of sugar-sweetened beverages is also generally desirable, as it contributes to increased micronutrient density, a reduction in the intake of added sugars, and thus a more balanced diet overall [211].

However, the evidence for the association between sugar-sweetened beverage consumption, glycemic control, and insulin sensitivity/resistance is deemed insufficient for adults (regardless of diabetes status) based on cohort studies and RCTs, so that no robust conclusions can be drawn [212]. A meta-analysis of 11 cohort studies shows an association between higher sugar-sweetened beverage intake and higher fasting blood glucose and insulin concentrations after adjustment for potential confounders for individuals without diabetes [213]. Two systematic reviews and meta-analyses specific to the effects of sugar-sweetened beverages containing fructose on glycemic control and serum lipid concentrations examined the effects of isocaloric substitution of glucose or sucrose with fructose in beverages and solid foods. Both short-term and long-term (study duration 2–10 weeks) substitution showed no adverse effects of fructose on either the maximum postprandial blood glucose, insulin or triglyceride concentrations, or the fasting blood glucose, insulin or triglyceride concentrations in subjects with normoglycemia, prediabetes, and T2Dm [214] [215]. However, when interpreting these results, it should be noted that a subgroup analysis was only performed for short-term substitution in normoglycemic individuals for the effect of sugar-sweetened beverages vs. sugar-sweetened foods [215], and the subgroup analyses for individuals with T2Dm in both studies were based on only a very small number of trials [214] [215].

The evidence for the association between sugar-sweetened beverage consumption and diabetes-associated macrovascular complications such as coronary events, stroke, hypertension, and dyslipidemia is also generally rated as insufficient for adults [212]. Systematic reviews (and meta-analyses) based on 4–11 prospective cohort studies indicate associations between sugar-sweetened beverage consumption and vascular risk factors (hypertension, hyperlipidemia), coronary heart disease, stroke, and mitral valve regurgitation [216] [217] [218]. To note, however, is that the results are not specific to individuals with T2Dm [216] [217] [218]. For the association between sugar-sweetened beverages and coronary heart disease, no significant effects were observed in the two studies of individuals with diabetes [217], and analyses for diabetes as a mediator for the association between sugar-sweetened beverages and vascular risk factors yielded inconsistent results [216].

With regards to diabetes-associated microvascular disease, another meta-analysis based on 5 study populations (also not exclusively individuals with T2Dm) found a significant association between chronic consumption of sugar-sweetened beverages and chronic kidney disease. However, the included studies were very heterogeneous, and evidence for publication bias was present [219].

Two systematic reviews and meta-analyses based on 4 and 12 cohort studies (including some individuals with T2Dm) on the association between sugar-sweetened beverage consumption and nonalcoholic fatty liver showed a significantly higher risk of nonalcoholic fatty liver disease (NAFLD) for the highest vs. lowest intake category of sugar-sweetened beverages [220] [221]. Even the lowest intake of<1 glass/week was associated with a 14% increase in the relative risk for NAFLD, and consumption of sugar-sweetened beverages showed a dose-dependent effect on the risk for NAFLD [220].

In conclusion, in line with the recommendation for the general population, a reduction in sugar-sweetened beverage intake should be targeted as part of a balanced diet for individuals with T2Dm to reduce the risk of cardiometabolic comorbidities [2] [118] [119] [211] [212]


#

Scientific background

When interpreting the data on the effects of sugar-sweetened beverages on individual diabetes-related target parameters, the following points should be considered: 1) the majority of studies do not exclusively examine individuals with T2Dm, so further studies in this patient group are needed to confirm the transferability of the results; 2) most associations for sugar-sweetened beverages are significant only for the comparison of extreme consumption categories, but not for moderate intake levels, which, however, roughly correspond to the mean estimated global intake of sugar-sweetened beverages [222]. On the one hand, the effects of additional sugar consumption on target parameters seem to depend on the energy balance and, on the other hand, on the sugar source as sugar-sweetened beverages providing excess energy seem to have a particularly negative effect on, for example, fasting blood glucose and insulin concentrations [223]. Furthermore, the direct association between the consumption of beverages containing fructose and sugar-sweetened beverages with the increased risk of, for example, the incidence of metabolic syndrome and other cardiometabolic risk factors and events seems to be limited to sugar-sweetened beverages and not transferable to the consumption of sugars from other sources (e. g., fruit, yogurt, fruit juices) [222] [224]. Possible explanations for this observation are that 1) the effect of sugar-sweetened beverages appears to be strongly mediated by the additional energy intake and resulting weight gain; that 2) other sources of fructose or sugar contain additional potentially health-promoting ingredients (which is not true of sugar-sweetened beverages) and that 3) sugar-sweetened beverages represent a marker of an overall unhealthier lifestyle [222].


#

Whole grains

RECOMMENDATIONS

  • In overweight patients with T2Dm, a diet rich in whole grains can help reduce the total energy intake and thus support targeted weight loss.

  • Consumption of low-processed whole grain products with a high proportion of whole grains results in a less pronounced postprandial blood glucose response, which may be a nonpharmacologic treatment option, particularly for people with T2Dm without insulin resistance.

  • Patients with T2Dm on insulin treatment should primarily consider consumption of whole grain products in terms of quantity according to carbohydrate content and additionally according to glycemic index, and adjust it to their insulin therapy.

  • Highly processed whole grain products show no additional beneficial effects on postprandial blood glucose response


#

Comment

For the general population, choosing whole grain products is recommended [225]. This is justified by their higher content of vitamins, minerals, and secondary phytochemicals, as well as beneficial effects on digestion and intestinal health due to the associated higher fiber intake. In addition, long-term cohort studies [226] [227] and numerous meta-analyses/reviews of cohort studies show associations between a significantly increased whole grain consumption and an up to 20% reduced risk of cardiovascular disease and mortality [228] [229] [230] [231] [232] [233] [234]. This results in recommendations by authors that even “moderate increases in whole grain consumption could reduce the risk of premature death” [232]. However, a causal relationship has not yet been established. In studies, the underlying dietary data is often based on only one survey (3-day protocol or Food Frequency Questionnaire at beginning of the cohort study), and classifications of foods as “whole grain foods” are inconsistent.

With regard to diabetes management, the degree to which whole grain foods are processed is important. As early as 1988, Jenkins et al. published results on the postprandial blood glucose response after consumption of whole grain breads with varying ratios of whole grain flour and whole grain content. The blood glucose response is determined less by the overall whole grain property of a milled grain product (whole meal) than by the proportion of whole grains (whole grain) it contains [235]. The higher the proportion of whole grains, the lower the blood glucose response, because the fruit and seed hulls form a physical barrier to the action of amylase on the endosperm.

Thirty years later, these results regarding the influence of the degree of processing have recently been confirmed under experimental [236] as well as everyday conditions [237]. No positive effects on the diabetes treatment have been shown for the mere addition of wheat bran to the usual meals with the aim of increasing the dietary fiber content [238].

For people with T2Dm, the recommendations are differentiated for the different treatment situations and forms:

For overweight patients with T2Dm: a meta-analysis on fiber and whole grain consumption in diabetes management included 42 intervention studies. Increased fiber/whole grain consumption (compared to control groups) was shown to result in a ½ kg lower body weight and a resulting 0.2% (2 mmol/mol) reduction in HbA1c [180]. Shortcomings of this analysis are the heterogeneous designs of the included studies, including diabetes medication, study duration, diabetes diagnosis, and type of whole grain consumption.

In non-insulin-treated, normal-weight patients with T2Dm (without insulin resistance), consumption of minimally-processed whole grain products with a high proportion of whole grains may result in a less pronounced postprandial blood glucose response. Positive effects of such a dietary measure on the achievement of the therapeutic goal depend, among other things, on the patient's acceptance of this dietary form and, in the medium term, on the continuing existence of residual β-cell function.

Insulin-treated individuals with T2Dm should assess how their diets affect increases in blood glucose in order to adjust insulin dosage to match. Accordingly, they should primarily consider consumption of whole grain products in terms of quantity according to carbohydrate content and additionally according to glycemic index, and adjust it to their insulin therapy. Whole grain products can be consumed according to personal preference.

Highly processed whole grain products show no additional beneficial effects on postprandial blood glucose response.


#
#

Fruits and vegetables

RECOMMENDATION
  • In the dietary planning for overweight patients with T2Dm, increased vegetable consumption in particular can support a targeted weight reduction.

  • In the dietary planning for normal-weight patients with T2Dm, consumption of large portions of fruit (products) and starchy vegetables (potatoes, corn, rice, grains, etc.) should be avoided.

  • Patients with T2Dm treated with insulin should consider the consumption of fruit in terms of quantity according to carbohydrate content and adjust it to their insulin therapy.

  • Separation into recommended and non-recommended fruits is not considered useful.

Comment

For the general population, a daily intake of at least 3 servings of vegetables (400 g) and 2 servings of fruit (250 g) is recommended under the slogan “5 a day” [225]. Recent results from the PURE study [239] and meta-analyses/reviews of cohort studies [233] [240] [241] [242] show associations of increased fruit and vegetable consumption with a 5–20% reduced risk with respect to cardiovascular disease and all-cause mortality. However, a cause-and-effect relationship has not yet been established, and data is inconsistent regarding effective fruit and vegetable varieties, minimum daily consumption levels, and the extent of clinical relevance with respect to specific diseases and mortalities. Beyond individual health, comparable recommendations for vegetable and fruit consumption supplemented with approximately 100 g of legumes/soy products daily are provided by the EAT-Lancet Commission as part of a Planetary Health Diet for environmental and social reasons [243].

For people with T2Dm, the recommendation is differentiated for the different treatment situations and forms:

In overweight patients with T2Dm, fruit and vegetable consumption should be seen as a supportive component for weight reduction. When energy-dense foods are replaced by judicious consumption of fruits and increased consumption of vegetables, this can sustain weight reduction. Intervention studies on the singular effects of individual food (groups) on physical or blood parameters do not exist or do not allow causal statements to be made because of the many additional influencing factors. However, intervention studies in people with T2Dm on the effects of an overall plant-rich diet – rich in fruits and especially vegetables – have shown a significant reduction in body weight, with corresponding positive effects on glycemia [122] [244] [245].

In non-insulin-treated normal-weight patients with T2Dm, large amounts of carbohydrates at individual meals should be avoided to prevent strong postprandial blood glucose responses. Therefore, large amounts of fruit, fruit juices, and starchy vegetables are not recommended (clinical experience). For non-starchy vegetables, there is no limiting quantity recommendation for consumption.

Insulin-treated patients with T2Dm should assess how their diets affect increases in blood glucose in order to adjust insulin dosage to match. Accordingly, the consumption of fruits and starchy vegetables (potatoes, sweet potatoes) should be assessed for carbohydrate content according to carbohydrate units and the individual form of insulin therapy should be adjusted. Fruits and vegetables can be consumed according to personal preferences.

In general, it should be noted that large amounts of carbohydrates can be absorbed in a short time through fruit juices, smoothies, and dried fruit - compared to unprocessed fresh fruit.

Based on the data available, there is no evidence for a blanket separation into recommended and non-recommended types of fruit, which is repeatedly popularized in lay publications because of the different carbohydrate content.


#
#

Fish

RECOMMENDATION
  • Oily fish may contribute to lowering blood lipids and the inflammatory phenotype and thus possibly the cardiovascular risk.

  • Evidence to recommend fish oil supplements in T2Dm is insufficient.

  • Sustainable fishing/fish farming should be considered when selecting fish meals [246].

Comment

Dietary patterns that include fish have been linked to a lower risk of diabetes in observational studies [247]. However, consumption of fish per se, as well as fish oils (long-chain omega-3 fatty acids such as docosahexaenoic acid [DHA] and eicosapentaenoic acid [EPA]), is inconsistently associated epidemiologically with diabetes risk. In Western regions (North America, Europe), there is a trend toward increased risk, whereas in the Pacific region, there is a trend toward decreased risk [248] [249] [250] [251]. These associations are in discrepancy with cohort studies linking fish consumption in a dose-dependent manner to a significantly lower risk of visceral obesity [252] and indicating lower cardiovascular risk and cardiovascular and all-cause mortality [241] [253] [254]. There is no significant relationship to hypertension [50].

The benefit in terms of cardiovascular risk is also controversial. Meta-analyses of RCTs see slight or nonsignificant effects [255] [256]. A meta-analysis specific to T2Dm patients has not yet been published.

In intervention studies, the specific effects of fish consumption are poorly studied. Fish oils appear to improve insulin sensitivity in patients with metabolic syndrome–but not in healthy individuals [257]. This effect has been shown to be gender-specific in women, but data is lacking for men [81]. Data on diabetes incidence is not available. Glycemic parameters do not improve with supplementation [258].

Metabolic benefits from fish oil supplementation are most likely with respect to triglycerides and C-reactive protein (CRP) [75] [258]. For non-inflammatory benefits, a high EPA/DHA ratio is advantageous [258].


#
#

Meat

RECOMMENDATION
  • In part, high-protein diets prove to be beneficial to possibly superior with regard to glycemia (see above). The replacement of carbohydrates with protein sources carried out in these diets can also be covered in part from animal sources, including meat of all kinds.

  • From an environmental point of view (e. g., to reduce demands on land or greenhouse gas emissions), meat consumption should also be reduced to the recommended level of the German Nutrition Society (DGE) [246] [259]

Comment

A diet heavy in meat and thus generally low in carbohydrates is associated with increased (cardiovascular) mortality in observational studies [260]. Epidemiologically, there are also moderate associations with cancer, CHD, and T2Dm. These associations are particularly strong with red meat, especially processed red meat [149].

Intervention studies show improvement in numerous metabolic parameters when reducing the amount of meat consumed daily. Causality for the benefits of low meat diets is also unclear in RCTs because in these studies the reduction is either isocalorically compensated with other potentially beneficial foods (e. g., whole grains, vegetables, legumes, nuts) or a meat avoidance is implemented in a hypocaloric setting.

RCTs on meat exchanges (red vs. white meat) mostly present the same confounding variable (e. g., red meat=standard diet vs. white meat=Mediterranean diet). A relevant intervention effect on mortality and morbidity (including T2Dm incidence) is questionable [260]. Only 6 RCTs have explicitly compared red and white meat and show no metabolic difference in the non-diabetic subjects studied [261] [262] [263] [264] [265] [266].

Based on this data, in 2019, the NutriRECS Consortium concluded not to recommend meat reduction due to lack of evidence [267]. However, the NutriRECS Consortium's assessment of the available nutritional evidence reveals the common, although flawed, assumption that medical and nutritional research should be evaluated according to the same criteria. For example, less value is systematically placed on observational studies and RCTs are rated very highly. However, long-term RCTs with food, especially with blinding and placebo control, are very difficult to conduct in the nutrition field. Overall, the recommendation to avoid (red) meat is currently still much better justified from an ecological and animal ethics perspective than by metabolic research.


#
#

Cinnamon

RECOMMENDATION
  • Consumption of cinnamon cannot be recommended to people with T2Dm as a component of successful diabetes therapy.

Comment

Over the past 15 years, numerous intervention studies have been published on the effects of cinnamon consumption on fasting blood glucose and HbA1c levels in people with T2Dm. Despite inconsistent study results, beneficial effects of cinnamon consumption on treatment outcomes in T2Dm have been consistently disseminated. Two meta-analyses in 2011 and 2012 postulated positive effects for cinnamon on fasting blood glucose [268] [269], and HbA1c [269] in their abstracts, while simultaneously concluding that the majority of the studies examined showed no relevant therapeutic effect on glycemia in people with T2Dm. Two meta-analyses in subsequent years have included the available studies up to early 2012 in their investigations, with the Cochrane paper [270] excluding studies of questionable quality from the analysis. Both studies found no significant effect of cinnamon consumption on HbA1c levels. Allen et al. [271] showed positive treatment effects on fasting blood glucose, but put this into perspective due to clear methodological deficits in the studies examined. Two other recent reviews [272] [273] conclude that the use of cinnamon (as an adjuvant) in the treatment of T2Dm cannot be recommended in view of the current study situation. Methodological problems extraordinarily limit the validity and comparability of the studies: for example, although the daily cinnamon doses used in the intervention groups of the studies are always given (0.1 to 6.0 g/day), there are no, incomplete, or inconsistent details on the cinnamon variety investigated (C. cassia, C. aromaticum, C. zeylanium), the form of application (cinnamon powder, cinnamon extract, capsules, tablets), the amount of active cinnamon ingredient tested, the drop-out rate of the subject collective or the intention-to-treat analysis, and other influencing factors (body weight, diabetes medication) that may have affected the target glycemic parameters studied (especially fasting blood glucose and HbA1c level) during the study period (4–18 weeks).


#
#

Artificial sweeteners

RECOMMENDATION
  • The consumption of artificial sweeteners in T2Dm mellitus is harmless to health if the respective maximum amounts are observed and may be useful if used occasionally as part of diabetes therapy.

  • In children and adolescents with T2Dm, the lower acceptable daily intake (ADI value) must be taken into account due to the lower body weight.

Comment

Artificial sweeteners are always the subject of controversial discussions in literature. According to one hypothesis, sweeteners could have an appetite-increasing effect due to their intense sweetness (e. g., [274]). However, when sweeteners were ingested (in the form of a beverage), compared with water, no appetite-increasing effect was found either in healthy, normal-weight subjects [275] [276] [277] or in metabolically healthy, overweight subjects. Sweeteners are said to have an orexigenic effect comparable to that of water [275].

The extent to which sweetener consumption affects glucose metabolism in patients diagnosed with T2Dm mellitus has been assessed in several clinical trials. No effect of sweetener consumption on the concentration of the parameters glucose, insulin or C-peptide, glucagon-like peptide-1 (GLP1), glucose-dependent insulinotropic peptide (GIP), peptide YY (PYY), glucagon, as well as HbA1c could be detected [278] [279] [280] [281] [282] [283]. Accordingly, the consumption of sweeteners does not seem to have a negative effect on glucose and insulin regulation in T2Dm.

The low cariogenic effect of sweeteners in contrast to conventional sugar is undisputed. In the case of saccharin, sucralose, aspartame as well as stevia, there is an additional bacteriostatic effect on oral flora [284] [285]. The extent to which sweeteners influence the intestinal microbiota has not yet been adequately clarified. In an intervention study, a change in the intestinal microbiota was observed in about half of the subjects (4/7) as a result of saccharin administration [286]. However, these results have not yet been confirmed.

The earlier reservation that sweeteners were carcinogenic has now been refuted. According to current knowledge, there is no evidence of a carcinogenic effect of sweeteners if the ADI value is not exceeded [287].


#

Scientific background

Sweeteners are synthetically produced or naturally-occurring compounds with high sweetness intensity, which are metabolized independently of insulin and are not cariogenic. Compared to sugar (sucrose), sweeteners have a sweetening power that is many times higher (30 to 20,000 times) and are therefore only used in very small quantities (milligram range), which are negligible in terms of calorie intake. As additives, sweeteners are subject to a health assessment by the European Food Safety Authority (EFSA) prior to approval, which derives acceptable daily intakes (ADI). The ADI value indicates the amount of an additive that can be ingested daily per kilogram of body weight over a lifetime without posing health risks. After approval, sweeteners are reassessed if necessary and re-evaluated at regular intervals [288].


#
#

Probiotics

RECOMMENDATION
  • Consumption of probiotics or synbiotics may have a beneficial effect on glucose regulation and lipid profile of T2Dm.

  • A multi-strain preparation usually achieves a stronger effect than a single-strain preparation.

  • Evidence is insufficient to date to recommend probiotic or synbiotic supplementation.

Comment

The effect of probiotic supplementation on T2Dm mellitus has been extensively studied. Various meta-analyses show a significant reduction in fasting blood glucose in T2Dm by probiotic supplementation, compared with placebo administration [289] [290] [291] [292] [293] [294]. A significant reduction in insulin resistance (Homeostasis Model Assessment [HOMA]) index) was also observed in subjects with T2Dm as a result of probiotic administration, compared with the control group, in several meta-analyses [291] [295]. However, a long-term change, measured by the HbA1c value, could not be detected by probiotic or synbiotic therapy (min. 12 weeks) [289] [290].

The results of meta-analyses regarding the effect of probiotic supplementation on lipid status in patients with T2Dm are heterogeneous. Two recent meta-analyses show, compared with placebo administration, a significant reduction in total cholesterol as well as triglyceride concentration (TG) in T2Dm as a result of 1 to 6 months of probiotic or synbiotic supplementation [289] [296]. In Mahboobi et al. (2018) [293], a significant improvement in TG, LDL, and HDL cholesterol concentrations was recorded as a result of synbiotic but not probiotic supplementation. Another meta-analysis did not find any association in this regard [297].

A recently-published randomized controlled intervention trial in a cross-over design by Palacios et al. (2020) [298] investigated the effect of probiotic administration as an adjunct to metformin therapy. After 12 weeks of administration of a multi-strain probiotic, improvement in glucose regulation (measured by fasting blood glucose concentration, HbA1c level, and HOMA index) and gut barrier function (measured by zonulin), as well as increased plasma butyrate concentration, were observed compared with placebo administration.

There is the following to consider with probiotic supplementation: probiotics may have antibiotic resistance in mobile genes that can be transferred to other, potentially pathogenic bacteria through interbacterial exchange [299]. Examination of various commercially-available probiotics revealed that the probiotic bacteria tested were resistant to several broad-spectrum antibiotics [300].


#

Scientific background

In Germany, probiotics are defined as “living microorganisms that enter the intestine in sufficient quantities in an active form and thereby achieve positive health effects” [301]. Primarily, the genera Lactobacillus and Bifidobacterium are used for the formulation in probiotics. Furthermore, specific lactic acid-producing species of other genera, e. g. Enterococcus faecalis, Streptococcus thermophilus or probiotic yeasts (Saccharomyces boulardii) are used. The dose varies between 108 and 1011 colony-forming units, and the use of above genera or species is considered safe [302].

The gut microbiota can have a strong influence on glucose metabolism mainly by modulating insulin sensitivity [303] and insulin synthesis [304]. According to a postulated mechanism based on the mouse model, microbially-synthesized short-chain fatty acids (acetate, propionate, and butyrate) bind to G-protein-coupled receptors (GRP43), inducing the secretion of the peptide hormone GLP1 [305]. GLP1 stimulates insulin synthesis in both glucose-tolerant individuals and T2Dm patients [306].

Large-scale studies show that an altered gut microbiome (also called dysbiosis) is present in T2Dm sufferers [307] [308] [309]. However, because T2Dm medication, for example metformin, has been shown to modulate the gut microbiota [310] [311] [312], it is often unclear whether the change is due to the disease or the therapy. Therefore, it has not yet been possible to identify a characteristic T2Dm microbiome. However, some studies suggest that the microbiome in T2Dm is characterized by a lower proportion of butyrate-producing bacteria [307] [308] [312]. A loss of butyrate producers is discussed as a predictor of the transition of prediabetes to T2Dm [313], which is why supplementation by probiotics or synbiotics may be a relevant aspect.


#

Sucrose/fructose

RECOMMENDATION
  • Fructose can be consumed in natural foods (e. g., fruit) as part of a balanced diet.

  • Beverages sweetened with fructose should be avoided, especially if the daily recommended energy intake is exceeded.


#

Comment

According to the recommendations of the American and Canadian diabetes societies, the intake of mono- and disaccharides should not exceed 10% and 12% of the daily energy intake, respectively [314] [315]. Isocaloric replacement of carbohydrates such as starch and sucrose with fructose has no adverse effects on body weight [316], blood pressure [317], fasting triglycerides [318], postprandial triglycerides [319], fatty liver markers [320], or uric acid [321]. In people with diabetes, isocaloric replacement with fructose could lower fasting glucose and HbA1c levels [322], especially when consumed in small amounts and in the form of fruit [323]. In contrast, fructose, especially in doses greater than 60 g per day or 10 E% of daily energy requirements, potentially causes mild triglyceride increases in people with T2Dm [318] [324]. Hypercaloric intake of fructose further leads to weight gain [316], uric acid increase [321], hepatic insulin resistance, hepatic fatty acidosis, and transaminases increase [320] [325] with the excessive caloric intake as the presumed cause. For this reason, people with diabetes should minimize the consumption of sugar-sweetened beverages to prevent weight gain and improve the cardiometabolic risk profile [2].


#

Scientific background

High fructose corn syrup (HFCS) has been used to sweeten beverages since the 1970s in the USA and increasingly in other countries. Countries with higher HFCS consumption have 20% higher diabetes prevalence compared with countries with lower HFCS consumption, independent of total sugar consumption and obesity prevalence [326].

Contrary to this epidemiologic association, prospective cohort studies of the effect of fructose on metabolism reached inconsistent results. For example, a meta-analysis of 15 prospective cohort studies did not indicate an association between fructose intake and increased risk of T2Dm that was independent of food type [327]. In a meta-analysis of 51 isocaloric studies and 8 hypercaloric studies, fructose only had unfavorable effects on lipid metabolism in terms of apolipoprotein B and triglyceride increases when offered as additional calories to an existing diet, whereas isocaloric replacement with fructose did not negatively affect lipid metabolism [318]. Consistent with this finding, fructose associated with increased energy intake, but not isocaloric fructose replacement, increased postprandial triglycerides in a meta-analysis of 14 isocaloric and 2 hypercaloric studies [328]. Similarly, in a meta-analysis of 24 controlled intervention studies, consumption of more than 100 g of fructose per day increased low-density lipoprotein (LDL) cholesterol and triglycerides, with no effect on serum lipids when fructose intake was less than 100 g per day [329]. A meta-analysis of 16 studies investigating isocaloric carbohydrate replacement with fructose in patients with T2Dm found heterogeneous effects on lipid metabolism with an increase in triglycerides and a decrease in total cholesterol without affecting LDL cholesterol [324].

Moreover, as shown in a meta-analysis of 21 studies, hypercaloric fructose consumption led to an increase in uric acid only in metabolically-healthy participants, whereas uric acid levels remained unchanged after isocaloric fructose intake in people both with and without diabetes [321]. A recent network meta-analysis indicated that replacement of fructose with starch resulted in decreased LDL cholesterol and replacement of fructose with glucose favorably affected insulin sensitivity and uric acid levels [330]. In contrast, in a meta-analysis of 18 studies in patients with T1Dm and T2Dm, isocaloric replacement with fructose resulted in a clinically-relevant decrease in HbA1c of 0.53% [322]. A similar HbA1c value drop occurred in a meta-analysis of 6 controlled dietary intervention trials after consumption of up to 36 g fructose per day in the form of fruit, without affecting body weight or triglyceride, insulin, and uric acid levels [323]. Consistent with this finding, in patients with recently diagnosed T2Dm, consumption of fructose from sugary beverages, but not from fruit, had an unfavorable effect on peripheral and hepatic insulin sensitivity [331].

In a meta-analysis of 29 papers, short-term fructose consumption, both as an isocaloric exchange for other carbohydrates and as a hypercaloric supplement, led to the development of hepatic insulin resistance in normal-weight, overweight, and obese participants without affecting peripheral or muscle insulin sensitivity [332]. In a meta-analysis of 13 studies, isocaloric replacement with fructose did not favor the development of nonalcoholic fatty liver disease (NAFLD). In contrast, there was an increase in intrahepatocellular lipids and glutamate pyruvate transaminase as a result of increased fructose consumption [320]. Consistent with this finding, another meta-analysis of 6 observational studies and 21 intervention studies also found an increase in liver fat and glutamate oxaloacetate transaminase as a result of hypercaloric fructose intake [325].

In a meta-analysis of 31 isocaloric and 10 hypercaloric prospective cohort studies, fructose administration had no effect on body weight in the isocaloric studies, whereas, in contrast, intake of large amounts of fructose resulted in weight gain [316].

In summary, when assessing studies on the effect of fructose on metabolism, it is important to distinguish whether fructose was ingested isocalorically in exchange for other carbohydrates or hypercalorically as additional energy. Hypercaloric studies indicate the unfavorable effects of fructose on metabolism, which can probably be attributed to the intake of additional energy. Unfavorable effects of isocaloric fructose intake cannot be substantiated with the available studies. It is possible that fructose, consumed in small amounts and in the form of fruit, has beneficial effects on glucose metabolism.


#

Alcohol

RECOMMENDATION
  • People with T2Dm should limit the amount of alcohol consumed to that recommended for the general population. Moderate, low-risk alcohol intake is consistent with good metabolic control and diabetes prognosis.

  • People with diabetes with high-risk alcohol use or dependence need to be educated about the dangers of alcohol, specifically including worsened metabolic control, and the risk of secondary diseases.

  • It must be pointed out in general that the risk of severe, especially nocturnal hypoglycemia under insulin therapy increases when larger amounts of alcohol are consumed and that this risk is reduced by consuming food during the period of alcohol consumption and raising the target blood glucose at night.


#

Comment

Differentiated content on the management of alcohol for people with diabetes mellitus can be found in the S2 guideline Psychosocial and Diabetes [333].

People with T2Dm should be counselled about the effects of alcohol consumption on blood glucose levels and, if alcohol is consumed, encouraged to consume at low risk levels. The Deutsche Hauptstelle für Suchtfragen (DHS) e. V. (German Centre for Addiction Issues) defines 12 g of alcohol per day for women and 24 g of alcohol per day for men as thresholds for low-risk consumption. The World Health Organization (WHO) defines a consumption of 10 g of alcohol per day for women and 20 g of alcohol per day for men as low-risk. These amounts also apply to people with T2Dm.


#

Alcohol and glucose metabolism

In people with diabetes, a linear and inverse relationship is shown between regular alcohol consumption and HbA1c levels [334] (level IIb). Consumption of one glass of wine per day (150 ml or 13 g alcohol) over a 3-month period resulted in a significant reduction in fasting glucose without increasing postprandial glucose levels compared with a control group consuming one glass of nonalcoholic beer per day. A positive effect on HbA1c was greatest in the group with the higher baseline HbA1c. In another controlled study, consumption of 1–2 glasses of wine per day (120–240 ml or 18 g of alcohol) over a 4-week period showed no negative effect on metabolic parameters (fasting glucose, lipids) but a significant positive effect on fasting serum insulin levels [335].

Alcohol consumption may impair blood glucose counterregulation and thus increase the risk of hypoglycemia under insulin therapy or insulinotropic oral antidiabetic agents [336] [337] [338].

Alcohol consumption is the cause of about one in 5 severe hypoglycemias leading to hospitalization [339]. However, the main effect of alcohol is likely to be the impairment of awareness, which leads to impaired perception of hypoglycemia and prevents affected individuals from responding appropriately [340].

Excessive consumption of alcohol interferes with diabetes management. Patients with excessive or risky alcohol consumption are less likely to implement therapy recommendations on exercise behavior, diet, medication intake, blood glucose self-monitoring or regular HbA1c value monitoring. There is a linear relationship: the more alcohol consumed, the less frequently therapy recommendations implemented [341].

According to the S2k guideline Psychosocial and Diabetes, alcohol consumption should be assessed regularly - at least once a year - in people with diabetes, and help should be offered when risky alcohol consumption is an issue.


#

Nutritional supplements

RECOMMENDATION
  • Individuals with T2Dm should meet their nutritional needs through a balanced diet. Routine micronutrient supplementation is not recommended.

  • In patients with T2Dm and established vitamin D deficiency, vitamin D supplementation may improve insulin resistance.


#

Comment

The American, Canadian, and British Diabetes Societies summarize the evidence on supplementation in general for persons with diabetes as follows: there is no clear evidence that supplementation with vitamins, minerals (e. g., chromium or vitamin D), herbs, or spices (e. g., cinnamon or aloe vera) improves metabolic control in persons without underlying nutritional deficiencies, and they are not generally recommended to improve glycemic control [2] [118] [119] [120]. Routine supplementation with antioxidants (e. g., vitamins E, C, or carotene) is not recommended because of a lack of evidence of efficacy and concerns about long-term safety. However, multivitamin supplementation might be necessary in special groups, e. g., pregnant or lactating women, elderly persons, vegetarians, or persons with a very low-calorie or low-carbohydrate diet [2] [118]. Vitamin B12 deficiency may occur with metformin use, so regular testing of vitamin B12 levels should be considered in individuals with T2Dm and taking metformin, especially in the additional presence of anemia or peripheral neuropathy, and possible vitamin B12 deficiency could be compensated with supplementation [2] [118] (1, 4). In the case of supplement use, possible adverse side effects and drug interactions must be considered [2] [118] [342]. Rather than a general recommendation of routine nutritional supplementation, individuals with diabetes should be encouraged to meet their nutrient needs through a balanced diet [120]. It should be kept in mind that individuals with diabetes who do not achieve their metabolic goals may be at increased risk for micronutrient deficiencies, thus adherence to a balanced diet that provides the minimum daily recommended daily intake of nutrients, and micronutrients in particular, is essential [2].

Due to the large number of available nutritional supplements, the following scientific background will highlight a selection of substances – namely n-3 PUFAs, vitamin D, magnesium, chromium, zinc, antioxidants (vitamin C, E) and polyphenols – with regard to their potential efficacy in individuals with T2Dm. Criteria for the selection of these supplements were the relevance of the potential effects of supplementation on diabetes management and a relatively “good” data situation, primarily based on systematic reviews and meta-analyses.


#

Scientific background

Consumption of n-3 PUFAs is discussed in the context of positive effects on glycemic control and cardiovascular disease prevention in individuals with T2Dm [118]. A systematic review from the Cochrane Library (23 RCTs, n=1075 T2Dm) showed a significant reduction in triglyceride (moderate effect) and VLDL concentrations (significant in subgroup analyses only for individuals with hypertriglyceridemia) and a significant increase in LDL cholesterol concentrations after supplementation with n-3 PUFAs vs. vegetable oils or placebo. Supplementation had no effects on total or HDL cholesterol concentrations, HbA1c levels, fasting blood glucose or fasting insulin concentrations, or body weight compared with control [343]. An increase in LDL cholesterol concentration after supplementation with n-3 PUFAs vs. control was also confirmed in another systematic review and meta-analysis (24 RCTs, n=1533 T2Dm) by Hartweg et al. [344]. However, supplementation did not show any change in LDL particle size, which characterizes diabetic dyslipoproteinemia along with changes in trigylceride and HDL cholesterol concentrations [344]. Furthermore, in both papers, the increase in LDL cholesterol concentrations by n-3 PUFA supplementation was not significant in the subgroup of individuals with hypertriglyceridemia [343] [344]. A recent systematic review with meta-analysis (45 RCTs, n=2674 T2Dm) confirmed the protective effects of n-3 PUFA supplementation vs. placebo on lipid metabolism and reported a significant reduction in LDL-cholesterol, VLDL-cholesterol, and triglyceride concentrations by supplementation with n-3 PUFAs vs. placebo [345]. Furthermore, O'Mahoney et al. showed a reduction in HbA1c and no effects on fasting blood glucose, fasting insulin concentrations, and HOMA-IR by supplementation with n-3 PUFAs vs. placebo [345]. Brown et al. (83 RCTs, n=121 070 with and without T2Dm) examined effects of higher vs. lower intakes of n-3, n-6, and total PUFA on diabetes risk, they also examined their effects on glycemic control and insulin resistance and found no effects of higher vs. lower n-3 PUFA intake on HbA1c, fasting blood glucose, fasting insulin concentration, and HOMA-IR [71]. Furthermore, there is evidence that high-dose supplementation with long-chain n-3 PUFAs (>4.4 g/d) may worsen glucose metabolism [71].

Overall, the American Diabetes Association summarizes the evidence on n-3 PUFAs for individuals with T2Dm with a recommendation of consumption of foods high in long-chain n-3 fatty acids from, for example, fish, nuts, and seeds for the prevention and treatment of cardiovascular disease (level of evidence B) [118]. However, benefits of routine n-3 PUFA supplementation are not supported based on current evidence (level of evidence A), as supplements do not appear to have the same beneficial effects as the corresponding whole foods on glycemic control and primary and secondary prevention of cardiovascular disease [118]. Furthermore, studies of n-3 PUFA supplementation with vascular events, cardiovascular disease, or mortality as an end point in individuals with T2Dm are lacking [343] [344].

Vitamin D deficiency is associated with alterations in glucose metabolism and insulin secretion [346]. However, evidence on effects of vitamin D supplementation on glycemic control is conflicting based on the systematic reviews and meta-analyses by Li et al. (20 RCTs, n=2703 T2Dm) and Mirhosseini et al. (24 RCTs, n=1528 T2Dm) [346] [347]. While both reviews confirmed a significant increase in serum 25-OH vitamin D levels and a reduction in HOMA-IR after supplementation with vitamin D compared with placebo [346] [347], a reduction in fasting blood glucose concentration and HbA1c value after supplementation with vitamin D compared with placebo was significant only in Mirhosseini et al. [346] [347]. These positive effects on parameters of glycemic control and insulin resistance were particularly measurable with a high daily vitamin D dose (≥4000 IU/d) and a long intervention duration (7 months on average) [346]. According to Li et al., compared with placebo, vitamin D supplementation reduces fasting insulin concentrations only in nonobese subjects with T2Dm and fasting blood glucose concentrations only with short-term supplementation, doses>2000 IU/d, and in subjects with vitamin D deficiency and good HbA1c control at baseline [347]. Other systematic reviews and meta-analyses examined the effects of vitamin D supplementation compared with placebo on blood pressure, serum lipid concentrations, and chronic subclinical inflammation [348] [349] [350] [351]. For blood pressure (15 RCTs, n=1134 T2Dm), vitamin D supplementation vs. placebo showed a significant but small reduction in diastolic blood pressure and no change in systolic blood pressure [350]. Similarly, with respect to serum lipid concentrations (17 RCTS, n=1365 T2Dm), vitamin D supplementation vs. placebo showed significant reductions in serum total, LDL, and HDL cholesterol concentrations, but these effects were small [348]. Furthermore, supplementation with vitamin D vs. placebo resulted in reductions in individual biomarkers of chronic subclinical inflammation such as CRP (20 RCTs, n=1270 T2 D and 13 RCTs, n=875 T2Dm) [349] [351]. While the recommendations for fracture prevention for individuals with T2Dm are identical to those for individuals in the general population and include supplementation with vitamin D [118], the quality of the evidence for the other outcomes considered, and the quality of the studies included in the reviews for them, have been found to be very heterogeneous by the authors. Further high-quality and long-term RCTs are thus needed to make a recommendation on vitamin D supplementation for individuals with T2Dm - beyond fracture prevention [346] [347] [349] [350] [351].

Magnesium, an essential mineral, is involved in intracellular carbohydrate metabolism, insulin secretion and signaling cascade, lipid metabolism, and regulation of blood pressure, among others [352]. Evidence on the effect of magnesium supplementation on glycemic control and blood pressure in individuals with T2Dm is conflicting [352] [353] [354]. After supplementation with magnesium vs. placebo (28 RCTs, n=1694 T2Dm), there were significant improvements in fasting blood glucose concentration and systolic blood pressure, with more pronounced effects in individuals with hypomagnesemia at baseline, but no changes in fasting insulin concentration, HbA1c value, and diastolic blood pressure [352]. In another systematic review with meta-analysis (21 RCTs, n=1362 subjects with and without diabetes), supplementation with magnesium compared with control showed only a significant reduction in HOMA-IR in the overall population and stratified by diabetes status, but not in HbA1c value or insulin concentration. Fasting blood glucose concentration was significantly reduced only with magnesium supplementation≥4 months. In the stratified analysis by diabetes status, magnesium supplementation had no significant effects on fasting blood glucose, insulin concentration, or HbA1c levels in subjects with diabetes compared with controls [353]. Asbaghi et al. examined the effects of magnesium supplementation (11 RCTs, n=673 T2Dm) on blood pressure and anthropometric parameters [354]. Magnesium supplementation compared with placebo resulted in significant reductions in systolic and diastolic blood pressure, especially with supplementation>12 weeks with≥300 mg/d inorganic magnesium. However, there were no effects of magnesium supplementation vs. placebo on anthropometric parameters [354]. In addition to the effects of magnesium supplementation on glycemic control and blood pressure, Verma and Garg also investigated its effect on serum lipids and demonstrated a significant increase in HDL cholesterol concentration and a reduction in LDL cholesterol and triglyceride concentration compared with control [352]. Further long-term RCTs with good study quality in individuals with T2Dm are needed to make evidence-based recommendations on magnesium supplementation.

The essential trace element chromium plays an important role in carbohydrate and lipid metabolism [355]. Supplementation with chromium compared with placebo (23 RCTs, n=1350 T2Dm and T1Dm [T1Dm included only in 1 RCT in addition to T2Dm]) resulted in significant reductions in fasting blood glucose and insulin concentrations, HbA1c levels, and HOMA-IR. Based on subgroup analysis, these effects were more pronounced with longer-term supplementation of at least 12 weeks, but showed no dependence on the chromium dose used. All included studies were rated as good quality, but the meta-analysis did not stratify results according to the chromium formulation used (chromium picolinate, chromium chloride, chromium from brewer's yeast) [356]. Based on 2 previous systematic reviews and meta-analyses (22 RCTs, n=1332 T2Dm and 14 RCTs, n=875 T2Dm), the effects of chromium supplementation compared with placebo on fasting blood glucose concentrations were most pronounced when chromium picolinate was used or only significant when chromium from brewer's yeast was used [357] [358]. Increases in HDL cholesterol and reductions in triglyceride levels were also achieved, particularly with supplementation with chromium picolinate or chromium from brewer's yeast compared with placebo [358], so that further research is needed on the optimal formulation of chromium supplements for individuals with T2Dm.

The essential trace element zinc plays an important role in the synthesis, storage, and secretion of insulin [359]. The zinc deficiency and hyperglycemia observed in individuals with T2Dm may be interrelated [360]. Based on 11 observational studies, in individuals with T2Dm compared with metabolically healthy controls, whole blood zinc concentrations decreased with each additional year of diabetes. This inverse relationship was generally not explained by lower nutritive zinc intake, as only individuals with T2Dm and complications who were dependent on nutritional therapy (e. g., nephropathy) had significantly lower zinc intake [361]. A subgroup analysis of a systematic review with meta-analysis (32 RCTs and n=1700 total, 19 RCTs with individuals with T2Dm) showed a significant reduction in fasting blood glucose concentration with supplementation with zinc vs. control for individuals with T2Dm. In the overall study population, which also included individuals at increased risk for T2Dm, supplementation with zinc additionally resulted in significant reductions in 2-h postprandial blood glucose concentration, fasting insulin concentration, HOMA-IR, HbA1c level, and high-sensitivity (hs)CRP compared with control [362]. Furthermore, supplementation with zinc vs. placebo (9 RCTs, n=424 T2Dm) reduced serum concentrations of triglycerides and total cholesterol. For LDL cholesterol concentrations, only positive effects of zinc supplementation compared with placebo were seen for stratified analyses by LDL cholesterol concentration and HbA1c level at baseline and for an intervention duration<12 weeks with dosing<100 mg/d. An increase in HDL cholesterol concentration was shown only for individuals with HDL cholesterol concentrations in the normal range and elevated HbA1c at baseline as well as stratified by intervention duration and zinc dosage [363]. Due to significant heterogeneity between the included studies and varying quality of the studies, further investigations are necessary before zinc supplementation can be recommended as an adjunctive therapy for T2Dm [362] [363].

Oxidative stress plays an important role in the pathogenesis of diabetes and its complications, so that supplementation with antioxidants could be expected to have beneficial effects on diabetes management [364]. In terms of glycemic control, a subgroup analysis of a systematic review and a meta-analysis based on 22 RCTs in individuals with and without T2Dm (n=597 T2Dm) for supplementation with vitamin C compared with placebo showed a significant reduction in blood glucose concentrations, but not HbA1c values and insulin concentrations for individuals with T2Dm, in older individuals and intervention duration≥30 days [365]. A subgroup analysis of a systematic review with meta-analysis based on 14 RCTs in individuals with T2Dm (n=714) showed significant reductions in HbA1c and fasting blood glucose concentrations with vitamin E supplementation compared with control for individuals with low baseline vitamin E status and poor glycemic control [366]. Neither supplementation with vitamin C or vitamin E alone nor a combination of both antioxidants showed significant effects on HOMA-IR (14 RCTs, n=735 T2Dm) [367]. Supplementation with the antioxidants vitamin C and vitamin E compared with placebo showed no overall effects on endothelial function in another study (10 RCTs, n=296 T2Dm), but a significant improvement in endothelial function after intervention for nonobese individuals with T2Dm (BMI≤29.45 kg/m2) in a subgroup analysis [368]. Individuals with T2Dm and diabetic retinopathy compared with individuals with T2Dm without retinopathy had lower serum concentrations of antioxidants and higher concentrations of oxidative stress biomarkers based on 14 observational studies and 7 RCTs (n=256 259). Due to strong methodological heterogeneity, only a qualitative synthesis of the included RCTs was performed, indicating beneficial effects of supplementation with antioxidants in diabetic retinopathy [369]. Overall, the reported effects of supplementation with antioxidants in individuals with T2Dm are primarily based on studies of low to moderate quality, so that the evidence for supplementation to improve metabolic control and endothelial function is currently insufficient [365] [366] [367] [368].

Resveratrol or polyphenols in general are also antioxidants and thus could have positive effects on diabetes management [370]. Supplementation with polyphenols (36 RCTs, n=1954 total, n=1426 T2Dm) resulted in a significant reduction in HbA1c compared with control (mean HbA1c at baseline: 7.03%). Subgroup analysis showed that this reduction was significant for individuals with T2Dm (mean HbA1c value at baseline: 7.44%), whereas no effects of supplementation were evident in individuals without diabetes and with prediabetes compared with controls [371]. In contrast, a systematic review from the Cochrane Library (3 RCTs, n=50 T2Dm) showed no effects of supplementation with resveratrol on HbA1c levels, fasting blood glucose concentrations, or insulin resistance. Overall, the available evidence from the included RCTs was rated as very low, so that the currently-available evidence on the safety and efficacy of supplementation with resveratrol was also rated as highly insufficient for it to be recommended for the treatment of T2Dm [370]. On systolic and diastolic blood pressure and mean arterial pressure or pulse pressure, supplementation with resveratrol showed no effects compared with control in the overall study population (17 RCTs, n=681 total, n=262 T2Dm). In subgroup analyses, resveratrol supplementation significantly reduced systolic blood pressure, mean arterial pressure, and pulse pressure in subjects with T2Dm compared with control [372].

Overall, due to, e. g., poor quality of included studies, heterogeneity in the method and results of the studies, an insufficient number of conducted studies or missing data on selected endpoints, long-term effects and long-term safety, there is still a need for further research on all considered dietary supplements before they can be recommended as an adjunct to the therapy of T2Dm. Although for individual cases or specific groups of individuals with T2Dm, compensating for nutrient deficiency by taking a nutritional supplement may be considered on an individual basis, taking into account potential adverse side effects and drug interactions, in general, individuals with T2Dm should meet their nutrient needs through a balanced diet and routine supplementation with micronutrients is not recommended.


#
#
#

Particularity of inpatient therapy or special diets to reduce insulin requirements

RECOMMENDATION
  • In the inpatient setting, 2-day oat or fiber days are highly recommended to break severe insulin resistance. These must be hypocaloric and contain a high fiber content. Oat days are very effective in this regard. Alternatively, other fiber diets may be chosen.

  • Blood glucose levels do not rise as much after eating high-fiber oat products compared to other meals with a comparable amount of carbohydrates, and less insulin secretion is induced.

Comment

Multiple studies have shown that insulin resistance in people with T2Dm could be significantly reduced by a specific diet for several days. These diets have always been hypocaloric and high in fiber. Oat days performed best, with regard to the HOMA index. The amount of soluble fiber is particularly high in oats [373]. The special effect of oats is thought to lie in its composition. Oats contain β-glucan and, at about 7.8%, the amount contained is particularly high [374]. In addition, an inhibitory effect of oat β-glucan on the expression of SGLT1 receptors as well as glucose transporter 2 (GLUT-2) in intestinal cells has been shown in vitro [375]. Furthermore, an inhibitory effect on dipeptidyl peptidase 4 (DDP4) was shown in vitro for certain oat proteins. This was somewhat stronger than the effect of buckwheat and barley [376]. It was also seen that oat β-glucan inhibited alpha-glucosidase [377].

Under inpatient conditions, a total of 14 patients were given oatmeal for 2 consecutive days, each with approximately 1100 calories per day. Mean blood glucose, adiponectin, and mean insulin dose were recorded before, 2 days after, and 4 weeks after the intervention. The mean insulin dose was reduced by 47% and this effect could still be seen 4 weeks after the intervention. The authors hypothesized effects on the microbiome as a result of the oat days [378].

In the cross-over study “OatMeal And Insulin Resistance (OMA-IR)” in people with inadequately-controlled T2Dm, the insulin requirement on the 3rd and 4th day decreased very significantly as a result of 2 oat days compared to a diabetes-adapted diet only. At the same time, over the course of 4 weeks after the oat days, HbA1c levels also decreased [379]. The study shows that oatβ-glucan is able to bind bile acids and lower blood cholesterol levels. Moreover, a close correlation was observed between the decrease in total bile acids as well as the decrease in proinsulin levels after oat days [380] [381] [382] [383] [384] [385] [386].

The European Food Safety Authority (EFSA) Panel on Dietetic Products, Nutrition and Allergies (NDA) considers it proven on the basis of studies that: the consumption of beta-glucan from oats […] leads to a reduction in the glucose rise after a meal [387]. Subsequently, the European Commission of the EU published the Health Claim: consumption of beta-glucans from oats […] as part of a meal contributes to the reduction of post-meal blood glucose levels [388].

In a meta-analysis of 103 comparative studies with 538 study participants, the addition of oat β-glucan to meals containing carbohydrates was shown to be associated with a reduced glucose and insulin response [389].

β-Glucan increases viscosity in the small intestine, delays gastric emptying and the release and absorption of food components, especially carbohydrates, thereby causing blood glucose to rise more slowly and resulting in a lower insulin response [390] [391].


#
#

German Diabetes Association: Clinical Practice Guidelines

This is a translation of the DDG clinical practice guideline published in Diabetologie 2021; 16 (Suppl 2): S255–S289. DOI 10.1055/a-1543-1293


#
#

Conflict of Interest

Thomas Skurk: has received lecture fees from Novo Nordisk. Diana Rubin: has received lecture fees from DGVS and Kaiserin-Friedrich-Stiftung. Anja Bosy-Westphal: none. Arthur Grünerbel: has received fees from Bavarian Medical Association, KV Bayern, Lilly, Novartis. Stefan Kabisch: has received fees and travel expenses from Sanofi, Berlin Chemie and Lilly; travel expenses and research support from J. Rettenmaier & Söhne, Holzmühle; further research support from Beneo Südzucker and California Walnut Commission. Peter Kronsbein: none. Karsten Müssig: none. Marie-Christine Simon: none. Astrid Tombek: none

  • References

  • 1 Beck J, Greenwood DA, Blanton L. et al. 2017 National Standards for Diabetes Self-Management Education and Support. Diabetes care 2017; 40: 1409-1419 DOI: 10.2337/dci17-0025.
  • 2 Evert AB, Dennison M, Gardner CD. et al. Nutrition Therapy for Adults With Diabetes or Prediabetes: A Consensus Report. Diabetes care 2019; 42: 731-754 DOI: 10.2337/dci19-0014.
  • 3 https://www.deutsche-diabetes-gesellschaft.de/politik/stellungnahmen/stellungnahme-des-ausschuss-ernaehrung-der-ddg-zum-consensusreport-nutrition-therapy-for-adults-with-diabetes-or-prediabetes Ausschuss Ernährung der DDG. Stellungnahme des Ausschuss Ernährung der DDG zum Consensus Report: Nutrition Therapy for Adults with Diabetes or Prediabetes: [Evert AB et al. Diabetes Care 2019;42:731-54.] 2019
  • 4 DeFronzo RA, Bonadonna RC, Ferrannini E. Pathogenesis of NIDDM. A balanced overview. Diabetes care 1992; 15: 318-368 DOI: 10.2337/diacare.15.3.318.
  • 5 DeFronzo RA, Eldor R, Abdul-Ghani M. Pathophysiologic approach to therapy in patients with newly diagnosed type 2 diabetes. Diabetes care 2013; 36: S127-S138 DOI: 10.2337/dcS13-2011.
  • 6 Lencioni C, Lupi R, Del Prato S. Beta-cell failure in type 2 diabetes mellitus. Curr Diab Rep 2008; 8: 179-184 DOI: 10.1007/s11892-008-0031-0.
  • 7 [Anonymous]. U.K. prospective diabetes study 16. Overview of 6 years’ therapy of type II diabetes: a progressive disease. U.K. Prospective Diabetes Study Group. Diabetes 1995; 44: 1249-1258
  • 8 Zaharia OP, Strassburger K, Strom A. et al. Risk of diabetes-associated diseases in subgroups of patients with recent-onset diabetes: a 5-year follow-up study. Lancet Diabetes Endocrinol 2019; 7: 684-694 DOI: 10.1016/S2213-8587(19)30187-1.
  • 9 Kodama S, Horikawa C, Fujihara K. et al. Quantitative relationship between body weight gain in adulthood and incident type 2 diabetes: a meta-analysis. Obes Rev 2014; 15: 202-214 DOI: 10.1111/obr.12129.
  • 10 Wing RR, Lang W, Wadden TA. et al. Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes care 2011; 34: 1481-1486 DOI: 10.2337/dc10-2415.
  • 11 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: 808-815 DOI: 10.2337/dc15-1942.
  • 12 Jazet IM, Pijl H, Frölich M. et al. Factors predicting the blood glucose lowering effect of a 30-day very low calorie diet in obese Type 2 diabetic patients. Diabetic medicine: a journal of the British Diabetic Association 2005; 22: 52-55 DOI: 10.1111/j.1464-5491.2004.01357.x.
  • 13 Lean MEJ, Leslie WS, Barnes AC. et al. Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, clusterrandomised trial. The Lancet 2018; 391: 541-551 DOI: 10.1016/S0140-6736(17)33102-1.
  • 14 Bangalore S, Fayyad R, DeMicco DA. et al. Body Weight Variability and Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus. Circ Cardiovasc Qual Outcomes 2018; 11: e004724 DOI: 10.1161/CIRCOUT-COMES.118.004724.
  • 15 Yeboah P, Hsu FC, Bertoni AG. et al. Body Mass Index, Change in Weight, Body Weight Variability and Outcomes in Type 2 Diabetes Mellitus (from the ACCORD Trial). The American journal of cardiology 2019; 123: 576-581 DOI: 10.1016/j.amjcard.2018.11.016.
  • 16 Pagidipati NJ, Zheng Y, Green JB. et al. Association of obesity with cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease: Insights from TECOS. Am Heart J 2020; 219: 47-57 DOI: 10.1016/j.ahj.2019.09.016.
  • 17 Bodegard J, Sundström J, Svennblad B. et al. Changes in body mass index following newly diagnosed type 2 diabetes and risk of cardiovascular mortality: a cohort study of 8486 primary-care patients. Diabetes Metab 2013; 39: 306-313 DOI: 10.1016/j.diabet.2013.05.004.
  • 18 Weinheimer EM, Sands LP, Campbell WW. A systematic review of the separate and combined effects of energy restriction and exercise on fatfree mass in middle-aged and older adults: implications for sarcopenic obesity. Nutr Rev 2010; 68: 375-388 DOI: 10.1111/j.1753-4887.2010.00298.x.
  • 19 Zaccardi F, Dhalwani NN, Papamargaritis D. et al. Nonlinear association of BMI with all-cause and cardiovascular mortality in type 2 diabetes mellitus: a systematic review and meta-analysis of 414587 participants in prospective studies. Diabetologia 2017; 60: 240-248 DOI: 10.1007/s00125-016-4162-6.
  • 20 Salehidoost R, Mansouri A, Amini M. et al. Body mass index and the all-cause mortality rate in patients with type 2 diabetes mellitus. Acta Diabetol 2018; 55: 569-577 DOI: 10.1007/s00592-018-1126-y.
  • 21 Hainer V, Aldhoon-Hainerová I. Obesity paradox does exist. Diabetes care 2013; 36: S276-S281 DOI: 10.2337/dcS13-2023.
  • 22 Murphy RA, Reinders I, Garcia ME. et al. Adipose tissue, muscle, and function: potential mediators of associations between body weight and mortality in older adults with type 2 diabetes. Diabetes care 2014; 37: 3213-3219 DOI: 10.2337/dc14-0293.
  • 23 Bales CW, Porter Starr KN. Obesity Interventions for Older Adults: Diet as a Determinant of Physical Function. Adv Nutr 2018; 9: 151-159 DOI: 10.1093/advances/nmx016.
  • 24 Uusitupa M, Khan TA, Viguiliouk E. et al. Prevention of Type 2 Diabetes by Lifestyle Changes: A Systematic Review and Meta-Analysis. Nutrients 2019; 11 DOI: 10.3390/nu11112611.
  • 25 Raben A, Vestentoft PS, Brand-Miller J. et al. The PREVIEW intervention study: Results from a 3-year randomized 2 × 2 factorial multinational trial investigating the role of protein, glycaemic index and physical activity for prevention of type 2 diabetes. Diabetes Obes Metab 2021; 23: 324-337 DOI: 10.1111/dom.14219.
  • 26 Gregg EW, Chen H, Wagenknecht LE. et al. Association of an intensive lifestyle intervention with remission of type 2 diabetes. JAMA 2012; 308: 2489-2496 DOI: 10.1001/jama.2012.67929.
  • 27 Anderson JW, Konz EC, Frederich RC. et al. Long-term weight-loss maintenance: a meta-analysis of US studies. Am J Clin Nutr 2001; 74: 579-584 DOI: 10.1093/ajcn/74.5.579.
  • 28 Bundesgesundheitsministerium 2015. Telemedizin. Im Internet (Stand: 09.04.2021) https://www.bundesgesundheitsministerium.de/service/begriffe-von-a-z/t/telemedizin.html
  • 29 Su D, McBride C, Zhou J. et al. Does nutritional counseling in telemedicine improve treatment outcomes for diabetes? A systematic review and meta-analysis of results from 92 studies. J Telemed Telecare 2016; 22: 333-347 DOI: 10.1177/1357633X15608297.
  • 30 Kempf K, Altpeter B, Berger J. et al. Efficacy of the Telemedical Lifestyle intervention Program TeLiPro in Advanced Stages of Type 2 Diabetes: A Randomized Controlled Trial. Diabetes care 2017; 40: 863-871 DOI: 10.2337/dc17-0303.
  • 31 Belalcazar LM, Haffner SM, Lang W. et al. Lifestyle intervention and/or statins for the reduction of C-reactive protein in type 2 diabetes: from the look AHEAD study. Obesity (Silver Spring) 2013; 21: 944-950 DOI: 10.1002/oby.20431.
  • 32 Colquitt JL, Pickett K, Loveman E. et al. Surgery for weight loss in adults. Cochrane Database Syst Rev 2014; DOI: 10.1002/14651858.CD003641.pub4.
  • 33 Patel KV, Bahnson JL, Gaussoin SA. et al. Association of Baseline and Longitudinal Changes in Body Composition Measures With Risk of Heart Failure and Myocardial Infarction in Type 2 Diabetes: Findings From the Look AHEAD Trial. Circulation 2020; 142: 2420-2430 DOI: 10.1161/CIRCULATIONAHA.120.050941.
  • 34 Franz MJ, Boucher JL, Rutten-Ramos S. et al. Lifestyle weight-loss intervention outcomes in overweight and obese adults with type 2 diabetes: a systematic review and meta-analysis of randomized clinical trials. J Acad Nutr Diet 2015; 115: 1447-1463 DOI: 10.1016/j.jand.2015.02.031.
  • 35 Murgatroyd PR, Goldberg GR, Leahy FE. et al. Effects of inactivity and diet composition on human energy balance. Int J Obes Relat Metab Disord 1999; 23: 1269-1275 DOI: 10.1038/sj.ijo.0801062.
  • 36 Stubbs RJ, Sepp A, Hughes DA. et al. The effect of graded levels of exercise on energy intake and balance in free-living women. Int J Obes Relat Metab Disord 2002; 26: 866-869 DOI: 10.1038/sj.ijo.0801874.
  • 37 Granados K, Stephens BR, Malin SK. et al. Appetite regulation in response to sitting and energy imbalance. Appl Physiol Nutr Metab 2012; 37: 323-333 DOI: 10.1139/h2012-002.
  • 38 Hägele FA, Büsing F, Nas A. et al. Appetite Control Is Improved by Acute Increases in Energy Turnover at Different Levels of Energy Balance. J Clin Endocrinol Metab 2019; 104: 4481-4491 DOI: 10.1210/jc.2019-01164.
  • 39 Douglas JA, King JA, Clayton DJ. et al. Acute effects of exercise on appetite, ad libitum energy intake and appetite-regulatory hormones in lean and overweight/obese men and women. Int J Obes (Lond) 2017; 41: 1737-1744 DOI: 10.1038/ijo.2017.181.
  • 40 Savikj M, Zierath JR. Train like an athlete: applying exercise interventions to manage type 2 diabetes. Diabetologia 2020; 63: 1491-1499 DOI: 10.1007/s00125-020-05166-9.
  • 41 Büsing F, Hägele FA, Nas A. et al. Impact of energy turnover on the regulation of glucose homeostasis in healthy subjects. Nutr Diabetes 2019; 9: 22 DOI: 10.1038/s41387-019-0089-6.
  • 42 Larsen JJ, Dela F, Kjaer M. et al. The effect of moderate exercise on postprandial glucose homeostasis in NIDDM patients. Diabetologia 1997; 40: 447-453 DOI: 10.1007/s001250050699.
  • 43 Heden TD, Winn NC, Mari A. et al. Postdinner resistance exercise improves postprandial risk factors more effectively than predinner resistance exercise in patients with type 2 diabetes. J Appl Physiol (1985) 2015; 118: 624-634 DOI: 10.1152/japplphysiol.00917.2014.
  • 44 Reynolds AN, Mann JI, Williams S. et al. Advice to walk after meals is more effective for lowering postprandial glycaemia in type 2 diabetes mellitus than advice that does not specify timing: a randomised crossover study. Diabetologia 2016; 59: 2572-2578 DOI: 10.1007/s00125-016-4085-2.
  • 45 Gaudet-Savard T, Ferland A, Broderick TL. et al. Safety and magnitude of changes in blood glucose levels following exercise performed in the fasted and the postprandial state in men with type 2 diabetes. Eur J Cardiovasc Prev Rehabil 2007; 14: 831-836 DOI: 10.1097/HJR.0b013e3282efaf38.
  • 46 DiPietro L, Gribok A, Stevens MS. et al. Three 15-min bouts of moderate postmeal walking significantly improves 24-h glycemic control in older people at risk for impaired glucose tolerance. Diabetes care 2013; 36: 3262-3268 DOI: 10.2337/dc13-0084.
  • 47 Seidelmann SB, Claggett B, Cheng S. et al. Dietary carbohydrate intake and mortality: a prospective cohort study and meta-analysis. Lancet Public Health 2018; 3: e419-e428 DOI: 10.1016/S2468-2667(18)30135-X.
  • 48 Davies MJ, D’Alessio DA, Fradkin J. et al. Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes care 2018; 41: 2669-2701 DOI: 10.2337/dci18-0033.
  • 49 Schwingshackl L, Chaimani A, Hoffmann G. et al. A network meta-analysis on the comparative efficacy of different dietary approaches on glycaemic control in patients with type 2 diabetes mellitus. Eur J Epidemiol 2018; 33: 157-170 DOI: 10.1007/s10654-017-0352-x.
  • 50 Schwingshackl L, Hoffmann G, Iqbal K. et al. Food groups and intermediate disease markers: a systematic review and network meta-analysis of randomized trials. Am J Clin Nutr 2018; 108: 576-586 DOI: 10.1093/ajcn/nqy151.
  • 51 Neuenschwander M, Ballon A, Weber KS. et al. Role of diet in type 2 diabetes incidence: umbrella review of meta-analyses of prospective observational studies. BMJ 2019; 366: l2368 DOI: 10.1136/bmj.l2368.
  • 52 Ge L, Sadeghirad B, Ball GDC. et al. Comparison of dietary macronutrient patterns of 14 popular named dietary programmes for weight and cardiovascular risk factor reduction in adults: systematic review and network meta-analysis of randomised trials. BMJ 2020; 369: m696 DOI: 10.1136/bmj.m696.
  • 53 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 DOI: 10.1136/bmj.m4743.
  • 54 Schwingshackl L, Nitschke K, Zähringer J. et al. Impact of Meal Frequency on Anthropometric Outcomes: A Systematic Review and Network Meta-Analysis of Randomized Controlled Trials. Adv Nutr 2020; 11: 1108-1122 DOI: 10.1093/advances/nmaa056.
  • 55 Della Corte KW, Perrar I, Penczynski KJ. et al. Effect of Dietary Sugar Intake on Biomarkers of Subclinical Inflammation: A Systematic Review and Meta-Analysis of Intervention Studies. Nutrients 2018; 10 DOI: 10.3390/nu10050606.
  • 56 Schwingshackl L, Chaimani A, Schwedhelm C. et al. Comparative effects of different dietary approaches on blood pressure in hypertensive and pre-hypertensive patients: A systematic review and network meta-analysis. Crit Rev Food Sci Nutr 2019; 59: 2674-2687 DOI: 10.1080/10408398.2018.1463967.
  • 57 Thom G, Messow CM, Leslie WS. et al. Predictors of type 2 diabetes remission in the Diabetes Remission Clinical Trial (DiRECT). Diabetic medicine: a journal of the British Diabetic Association 2020; e14395 DOI: 10.1111/dme.14395.
  • 58 de Souza RJ, Mente A, Maroleanu A. et al. Intake of saturated and trans unsaturated fatty acids and risk of all cause mortality, cardiovascular disease, and type 2 diabetes: systematic review and meta-analysis of observational studies. BMJ 2015; 351: h3978 DOI: 10.1136/bmj.h3978.
  • 59 Astrup A, Magkos F, Bier DM. et al. Saturated Fats and Health: A Reassessment and Proposal for Food-Based Recommendations: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 76: 844-857 DOI: 10.1016/j.jacc.2020.05.077.
  • 60 Pimpin L, Wu JHY, Haskelberg H. et al. Is Butter Back? A Systematic Review and Meta-Analysis of Butter Consumption and Risk of Cardiovascular Disease, Diabetes, and Total Mortality. PLoS One 2016; 11: e0158118 DOI: 10.1371/journal.pone.0158118.
  • 61 Benatar JR, Sidhu K, Stewart RAH. Effects of high and low fat dairy food on cardio-metabolic risk factors: a meta-analysis of randomized studies. PLoS One 2013; 8: e76480 DOI: 10.1371/journal.pone.0076480.
  • 62 Hooper L, Abdelhamid AS, Jimoh OF. et al. Effects of total fat intake on body fatness in adults. Cochrane Database Syst Rev 2020; 6: CD013636 DOI: 10.1002/14651858.CD013636.
  • 63 Hooper L, Martin N, Jimoh OF. et al. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst Rev 2020; 8: CD011737 DOI: 10.1002/14651858.CD011737.pub3.
  • 64 Belalcazar LM, Reboussin DM, Haffner SM. et al. A 1-year lifestyle intervention for weight loss in individuals with type 2 diabetes reduces high C-reactive protein levels and identifies metabolic predictors of change: from the Look AHEAD (Action for Health in Diabetes) study. Diabetes care 2010; 33: 2297-2303 DOI: 10.2337/dc10-0728.
  • 65 Lu M, Wan Y, Yang B. et al. Effects of low-fat compared with high-fat diet on cardiometabolic indicators in people with overweight and obesity without overt metabolic disturbance: a systematic review and metaanalysis of randomised controlled trials. Br J Nutr 2018; 119: 96-108 DOI: 10.1017/S0007114517002902.
  • 66 Wu JHY, Marklund M, Imamura F. et al. Omega-6 fatty acid biomarkers and incident type 2 diabetes: pooled analysis of individual-level data for 39 740 adults from 20 prospective cohort studies. Lancet Diabetes Endocrinol 2017; 5: 965-974 DOI: 10.1016/S2213-8587(17)30307-8.
  • 67 Li J, Guasch-Ferré M, Li Y. et al. Dietary intake and biomarkers of linoleic acid and mortality: systematic review and meta-analysis of prospective cohort studies. Am J Clin Nutr 2020; 112: 150-167 DOI: 10.1093/ajcn/nqz349.
  • 68 Pan A, Chen M, Chowdhury R. et al. α-Linolenic acid and risk of cardiovascular disease: a systematic review and meta-analysis. Am J Clin Nutr 2012; 96: 1262-1273 DOI: 10.3945/ajcn.112.044040.
  • 69 Abdelhamid AS, Martin N, Bridges C. et al. Polyunsaturated fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11: CD012345 DOI: 10.1002/14651858.CD012345.pub3.
  • 70 Abdelhamid AS, Brown TJ, Brainard JS. et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2020; 3: CD003177 DOI: 10.1002/14651858.CD003177.pub5.
  • 71 Brown TJ, Brainard J, Song F. et al. Omega-3, omega-6, and total dietary polyunsaturated fat for prevention and treatment of type 2 diabetes mellitus: systematic review and meta-analysis of randomised controlled trials. BMJ 2019; 366: l4697 DOI: 10.1136/bmj.l4697.
  • 72 Qian F, Korat AA, Malik V. et al. Metabolic Effects of Monounsaturated Fatty Acid-Enriched Diets Compared With Carbohydrate or Polyunsaturated Fatty Acid-Enriched Diets in Patients With Type 2 Diabetes: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Diabetes care 2016; 39: 1448-1457 DOI: 10.2337/dc16-0513.
  • 73 Jovanovski E, de Castro Ruiz Marques A, Li D. et al. Effect of high-carbohydrate or high-monounsaturated fatty acid diets on blood pressure: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 2019; 77: 19-31 DOI: 10.1093/nutrit/nuy040.
  • 74 Zhang YY, Liu W, Zhao TY. et al. Efficacy of Omega-3 Polyunsaturated Fatty Acids Supplementation in Managing Overweight and Obesity: A Meta-Analysis of Randomized Clinical Trials. J Nutr Health Aging 2017; 21: 187-192 DOI: 10.1007/s12603-016-0755-5.
  • 75 Lin N, Shi JJ, Li YM. et al What is the impact of n-3 PUFAs on inflammation markers in Type 2 diabetic mellitus populations?: a systematic review and meta-analysis of randomized controlled trials. Lipids Health Dis 2016; 15: 133 DOI: 10.1186/s12944-016-0303-7.
  • 76 Reis CEG, Landim KC, Nunes ACS. et al. Safety in the hypertriglyceridemia treatment with N-3 polyunsaturated fatty acids on glucose metabolism in subjects with type 2 diabetes mellitus. Nutr Hosp 2014; 31: 570-576 DOI: 10.3305/nh.2015.31.2.7845.
  • 77 Gao L, Cao J, Mao Q. et al. Influence of omega-3 polyunsaturated fatty acid-supplementation on platelet aggregation in humans: a meta-analysis of randomized controlled trials. Atherosclerosis 2013; 226: 328-334 DOI: 10.1016/j.atherosclerosis.2012.10.056.
  • 78 He XX, Wu XL, Chen RP. et al. Effectiveness of Omega-3 Polyunsaturated Fatty Acids in Non-Alcoholic Fatty Liver Disease: A Meta-Analysis of Randomized Controlled Trials. PLoS One 2016; 11: e0162368 DOI: 10.1371/journal.pone.0162368.
  • 79 Li N, Yue H, Jia M. et al. Effect of low-ratio n-6/n-3 PUFA on blood glucose: a meta-analysis. Food Funct 2019; 10: 4557-4565 DOI: 10.1039/c9fo00323a.
  • 80 Wanders AJ, Blom WAM, Zock PL. et al. Plant-derived polyunsaturated fatty acids and markers of glucose metabolism and insulin resistance: a meta-analysis of randomized controlled feeding trials. BMJ Open Diabetes Res Care 2019; 7: e000585 DOI: 10.1136/bmjdrc-2018-000585.
  • 81 Abbott KA, Burrows TL, Thota RN. et al. Do ω-3 PUFAs affect insulin resistance in a sex-specific manner? A systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2016; 104: 1470-1484 DOI: 10.3945/ajcn.116.138172.
  • 82 Jovanovski E, Li D, Thanh Ho HV. et al. The effect of alpha-linolenic acid on glycemic control in individuals with type 2 diabetes: A systematic review and meta-analysis of randomized controlled clinical trials. Medicine (Baltimore) 2017; 96: e6531 DOI: 10.1097/MD.0000000000006531.
  • 83 Faris MAI, Jahrami H, BaHammam A. et al. A systematic review, meta-analysis, and meta-regression of the impact of diurnal intermittent fasting during Ramadan on glucometabolic markers in healthy subjects. Diabetes Res Clin Pract 2020; 165: 108226 DOI: 10.1016/j.diabres.2020.108226.
  • 84 Mirmiran P, Bahadoran Z, Gaeini Z. et al. Effects of Ramadan intermittent fasting on lipid and lipoprotein parameters: An updated meta-analysis. Nutr Metab Cardiovasc Dis 2019; 29: 906-915 DOI: 10.1016/j.numecd.2019.05.056.
  • 85 Fernando HA, Zibellini J, Harris RA. et al. Effect of Ramadan Fasting on Weight and Body Composition in Healthy Non-Athlete Adults: A Systematic Review and Meta-Analysis. Nutrients 2019; 11 DOI: 10.3390/nu11020478.
  • 86 Horne BD, May HT, Anderson JL. et al. Usefulness of routine periodic fasting to lower risk of coronary artery disease in patients undergoing coronary angiography. The American journal of cardiology 2008; 102: 814-819 DOI: 10.1016/j.amjcard.2008.05.021.
  • 87 Horne BD, Muhlestein JB, May HT. et al. Relation of routine, periodic fasting to risk of diabetes mellitus, and coronary artery disease in patients undergoing coronary angiography. The American journal of cardiology 2012; 109: 1558-1562 DOI: 10.1016/j.amjcard.2012.01.379.
  • 88 Schwingshackl L, Zähringer J, Nitschke K. et al. Impact of intermittent energy restriction on anthropometric outcomes and intermediate disease markers in patients with overweight and obesity: systematic review and meta-analyses. Crit Rev Food Sci Nutr 2021; 61: 1293-1304 DOI: 10.1080/10408398.2020.1757616.
  • 89 Park J, Seo YG, Paek YJ. et al. Effect of alternate-day fasting on obesity and cardiometabolic risk: A systematic review and meta-analysis. Metabolism 2020; 111: 154336 DOI: 10.1016/j.metabol.2020.154336.
  • 90 Harris L, Hamilton S, Azevedo LB. et al. Intermittent fasting interventions for treatment of overweight and obesity in adults: a systematic review and meta-analysis. JBI Database System Rev Implement Rep 2018; 16: 507-547 DOI: 10.11124/JBISRIR-2016-003248.
  • 91 Seimon RV, Roekenes JA, Zibellini J. et al. Do intermittent diets provide physiological benefits over continuous diets for weight loss? A systematic review of clinical trials. Mol Cell Endocrinol 2015; 418: 153-172 DOI: 10.1016/j.mce.2015.09.014.
  • 92 Horne BD, Muhlestein JB, Anderson JL. Health effects of intermittent fasting: hormesis or harm? A systematic review. Am J Clin Nutr 2015; 102: 464-470 DOI: 10.3945/ajcn.115.109553.
  • 93 Borgundvaag E, Mak J, Kramer CK. Metabolic Impact of Intermittent Fasting in Patients With Type 2 Diabetes Mellitus: A Systematic Review and Meta-analysis of Interventional Studies. J Clin Endocrinol Metab 2021; 106: 902-911 DOI: 10.1210/clinem/dgaa926.
  • 94 Parr EB, Devlin BL, Lim KHC. et al. Time-Restricted Eating as a Nutrition Strategy for Individuals with Type 2 Diabetes: A Feasibility Study. Nutrients 2020; 12 DOI: 10.3390/nu12113228.
  • 95 Carter S, Clifton PM, Keogh JB. The effects of intermittent compared to continuous energy restriction on glycaemic control in type 2 diabetes; a pragmatic pilot trial. Diabetes Res Clin Pract 2016; 122: 106-112 DOI: 10.1016/j.diabres.2016.10.010.
  • 96 Carter S, Clifton PM, Keogh JB. The effect of intermittent compared with continuous energy restriction on glycaemic control in patients with type 2 diabetes: 24-month follow-up of a randomised noninferiority trial. Diabetes Res Clin Pract 2019; 151: 11-19 DOI: 10.1016/j.diabres.2019.03.022.
  • 97 Corley BT, Carroll RW, Hall RM. et al. Intermittent fasting in Type 2 diabetes mellitus and the risk of hypoglycaemia: a randomized controlled trial. Diabetic medicine: a journal of the British Diabetic Association 2018; 35: 588-594 DOI: 10.1111/dme.13595.
  • 98 Henry RR, Wiest-Kent TA, Scheaffer L. et al. Metabolic consequences of very-low-calorie diet therapy in obese non-insulin-dependent diabetic and nondiabetic subjects. Diabetes 1986; 35: 155-164 DOI: 10.2337/diab.35.2.155.
  • 99 Amatruda JM, Richeson JF, Welle SL. et al. The safety and efficacy of a controlled low-energy (‘very-low-calorie’) diet in the treatment of noninsulin-dependent diabetes and obesity. Arch Intern Med 1988; 148: 873-877
  • 100 Rotella CM, Cresci B, Mannucci E. et al. Short cycles of very low calorie diet in the therapy of obese type II diabetes mellitus. J Endocrinol Invest 1994; 17: 171-179 DOI: 10.1007/BF03347709.
  • 101 Dhindsa P, Scott AR, Donnelly R. Metabolic and cardiovascular effects of very-low-calorie diet therapy in obese patients with Type 2 diabetes in secondary failure: outcomes after 1 year. Diabetic medicine: a journal of the British Diabetic Association 2003; 20: 319-324 DOI: 10.1046/j.1464-5491.2003.00937.x.
  • 102 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: 344-355 DOI: 10.1016/S2213-8587(19)30068-3.
  • 103 Maggio CA, Pi-Sunyer FX. Obesity and type 2 diabetes. Endocrinol Metab Clin North Am 2003; 32: 805-822 viii DOI: 10.1016/s0889-8529(03)00071-9.
  • 104 Wolf AM, Colditz GA. Current estimates of the economic cost of obesity in the United States. Obes Res 1998; 6: 97-106 DOI: 10.1002/j.1550-8528.1998.tb00322.x.
  • 105 Colditz GA, Willett WC, Rotnitzky A. et al. Weight gain as a risk factor for clinical diabetes mellitus in women. Ann Intern Med 1995; 122: 481-486 DOI: 10.7326/0003-4819-122-7-199504010-00001.
  • 106 Anderson JW, Kendall CWC, Jenkins DJA. Importance of weight management in type 2 diabetes: review with meta-analysis of clinical studies. J Am Coll Nutr 2003; 22: 331-339 DOI: 10.1080/07315724.2003.10719316.
  • 107 Leslie WS, Taylor R, Harris L. et al. Weight losses with low-energy formula diets in obese patients with and without type 2 diabetes: systematic review and meta-analysis. Int J Obes (Lond) 2017; 41: 96-101 DOI: 10.1038/ijo.2016.175.
  • 108 McCombie L, Brosnahan N, Ross H. et al. Filling the intervention gap: service evaluation of an intensive nonsurgical weight management programme for severe and complex obesity. J Hum Nutr Diet 2019; 32: 329-337 DOI: 10.1111/jhn.12611.
  • 109 Jazet IM, de Craen AJ, van Schie EM. et al. Sustained beneficial metabolic effects 18 months after a 30-day very low calorie diet in severely obese, insulin-treated patients with type 2 diabetes. Diabetes Res Clin Pract 2007; 77: 70-76 DOI: 10.1016/j.diabres.2006.10.019.
  • 110 Kempf K, Schloot NC, Gärtner B. et al. Meal replacement reduces insulin requirement, HbA1c and weight long-term in type 2 diabetes patients with 100 U insulin per day. J Hum Nutr Diet 2014; 27: 21-27 DOI: 10.1111/jhn.12145.
  • 111 Kempf K, Röhling M, Niedermeier K. et al. Individualized Meal Replacement Therapy Improves Clinically Relevant Long-Term Glycemic Control in Poorly Controlled Type 2 Diabetes Patients. Nutrients 2018; 10 DOI: 10.3390/nu10081022.
  • 112 Taylor R, Leslie WS, Barnes AC. et al. Clinical and metabolic features of the randomised controlled Diabetes Remission Clinical Trial (DiRECT) cohort. Diabetologia 2018; 61: 589-598 DOI: 10.1007/s00125-017-4503-0.
  • 113 Halle M, Röhling M, Banzer W. et al. Meal replacement by formula diet reduces weight more than a lifestyle intervention alone in patients with overweight or obesity and accompanied cardiovascular risk factors-the ACOORH trial. Eur J Clin Nutr 2021; 75: 661-669 DOI: 10.1038/s41430-020-00783-4.
  • 114 Röhling M, Kempf K, Banzer W. et al. Prediabetes Conversion to Normoglycemia Is Superior Adding a Low-Carbohydrate and Energy Deficit Formula Diet to Lifestyle Intervention-A 12-Month Subanalysis of the ACOORH Trial. Nutrients 2020; 12 DOI: 10.3390/nu12072022.
  • 115 Holman RR, Paul SK, Bethel MA. et al. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359: 1577-1589 DOI: 10.1056/NEJMoa0806470.
  • 116 Haslacher H, Fallmann H, Waldhäusl C. et al. Type 2 diabetes care: Improvement by standardization at a diabetes rehabilitation clinic. An observational report. PLoS One 2019; 14: e0226132 DOI: 10.1371/journal.pone.0226132.
  • 117 Paul SK, Shaw JE, Montvida O. et al. Weight gain in insulin-treated patients by body mass index category at treatment initiation: new evidence from real-world data in patients with type 2 diabetes. Diabetes Obes Metab 2016; 18: 1244-1252 DOI: 10.1111/dom.12761.
  • 118 [Anonymous]. 5. Facilitating Behavior Change and Well-being to Improve Health Outcomes: Standards of Medical Care in Diabetes-2020. Diabetes care 2020; 43: S48-S65 DOI: 10.2337/dc20-S005.
  • 119 Dyson PA, Twenefour D, Breen C. et al. Diabetes UK evidence-based nutrition guidelines for the prevention and management of diabetes. Diabetic medicine: a journal of the British Diabetic Association 2018; 35: 541-547 DOI: 10.1111/dme.13603.
  • 120 Dworatzek PD, Arcudi K, Gougeon R. et al. Nutrition therapy. Can J Diabetes 2013; 37: S45-S55 DOI: 10.1016/j.jcjd.2013.01.019.
  • 121 Hallberg SJ, Dockter NE, Kushner JA. et al. Improving the scientific rigour of nutritional recommendations for adults with type 2 diabetes: A comprehensive review of the American Diabetes Association guidelinerecommended eating patterns. Diabetes Obes Metab 2019; 21: 1769-1779 DOI: 10.1111/dom.13736.
  • 122 Salas-Salvadó J, Becerra-Tomás N, Papandreou C. et al. Dietary Patterns Emphasizing the Consumption of Plant Foods in the Management of Type 2 Diabetes: A Narrative Review. Adv Nutr 2019; 10: S320-S331 DOI: 10.1093/advances/nmy102.
  • 123 Viguiliouk E, Kendall CW, Kahleová H. et al. Effect of vegetarian dietary patterns on cardiometabolic risk factors in diabetes: A systematic review and meta-analysis of randomized controlled trials. Clin Nutr 2019; 38: 1133-1145 DOI: 10.1016/j.clnu.2018.05.032.
  • 124 Papamichou D, Panagiotakos DB, Itsiopoulos C. Dietary patterns and management of type 2 diabetes: A systematic review of randomised clinical trials. Nutr Metab Cardiovasc Dis 2019; 29: 531-543 DOI: 10.1016/j.numecd.2019.02.004.
  • 125 Ohlsson B. An Okinawan-based Nordic diet improves glucose and lipid metabolism in health and type 2 diabetes, in alignment with changes in the endocrine profile, whereas zonulin levels are elevated. Exp Ther Med 2019; 17: 2883-2893 DOI: 10.3892/etm.2019.7303.
  • 126 Daneshzad E, Emami S, Darooghegi Mofrad M. et al. Association of modified Nordic diet with cardiovascular risk factors among type 2 diabetes patients: a cross-sectional study. J Cardiovasc Thorac Res 2018; 10: 153-161 DOI: 10.15171/jcvtr.2018.25.
  • 127 Via MA, Mechanick JI. Nutrition in Type 2 Diabetes and the Metabolic Syndrome. Med Clin North Am 2016; 100: 1285-1302 DOI: 10.1016/j.mcna.2016.06.009.
  • 128 Porrata-Maury C, Hernández-Triana M, Ruiz-Álvarez V. et al. Ma-Pi 2 macrobiotic diet and type 2 diabetes mellitus: pooled analysis of shortterm intervention studies. Diabetes Metab Res Rev 2014; 30: 55-66 DOI: 10.1002/dmrr.2519.
  • 129 Garvey WT, Mechanick JI, Brett EM. et al. American Association of Clinical Endocrinologists and American College of Endocrinology Comprehensive CLINICAL Practice Guidelines for Medical Care of Patients with Obesity. Endocr Pract 2016; 22: 1-203 DOI: 10.4158/EP161365.GL.
  • 130 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: 505-516 DOI: 10.3945/ajcn.112.042457.
  • 131 Huo R, Du T, Xu Y. et al. Effects of Mediterranean-style diet on glycemic control, weight loss and cardiovascular risk factors among type 2 diabetes individuals: a meta-analysis. Eur J Clin Nutr 2015; 69: 1200-1208 DOI: 10.1038/ejcn.2014.243.
  • 132 Pan B, Wu Y, Yang Q. et al. The impact of major dietary patterns on glycemic control, cardiovascular risk factors, and weight loss in patients with type 2 diabetes: A network meta-analysis. J Evid Based Med 2019; 12: 29-39 DOI: 10.1111/jebm.12312.
  • 133 Johannesen CO, Dale HF, Jensen C. et al. Effects of Plant-Based Diets on Outcomes Related to Glucose Metabolism: A Systematic Review. Diabetes Metab Syndr Obes 2020; 13: 2811-2822 DOI: 10.2147/DMSO.S265982.
  • 134 Toumpanakis A, Turnbull T, Alba-Barba I. Effectiveness of plant-based diets in promoting well-being in the management of type 2 diabetes: a systematic review. BMJ Open Diabetes Res Care 2018; 6: e000534 DOI: 10.1136/bmjdrc-2018-000534.
  • 135 Tran E, Dale HF, Jensen C. et al. Effects of Plant-Based Diets on Weight Status: A Systematic Review. Diabetes Metab Syndr Obes 2020; 13: 3433-3448 DOI: 10.2147/DMSO.S272802.
  • 136 Medawar E, Huhn S, Villringer A. et al. The effects of plant-based diets on the body and the brain: a systematic review. Transl Psychiatry 2019; 9: 226 DOI: 10.1038/s41398-019-0552-0.
  • 137 Esposito K, Maiorino MI, Bellastella G. et al. A journey into a Mediterranean diet and type 2 diabetes: a systematic review with meta-analyses. BMJ Open 2015; 5: e008222 DOI: 10.1136/bmjopen-2015-008222.
  • 138 Carter P, Achana F, Troughton J. et al. A Mediterranean diet improves HbA1c but not fasting blood glucose compared to alternative dietary strategies: a network meta-analysis. J Hum Nutr Diet 2014; 27: 280-297 DOI: 10.1111/jhn.12138.
  • 139 Emadian A, Andrews RC, England CY. et al. The effect of macronutrients on glycaemic control: a systematic review of dietary randomised controlled trials in overweight and obese adults with type 2 diabetes in which there was no difference in weight loss between treatment groups. Br J Nutr 2015; 114: 1656-1666 DOI: 10.1017/S0007114515003475.
  • 140 Kahleova H, Salas-Salvadó J, Rahelić D. et al. Dietary Patterns and Cardiometabolic Outcomes in Diabetes: A Summary of Systematic Reviews and Meta-Analyses. Nutrients 2019; 11 DOI: 10.3390/nu11092209.
  • 141 DDG https://www.deutsche-diabetes-gesellschaft.de/fileadmin/user_upload/01_Die_DDG/03_Ausschuesse/02_Ernaehrung/2015-057-025l_S3_Diabetes_mellitus_Empfehlungen_Proteinzufuhr_2015-10.pdfStand: 06.07.2021
  • 142 Pfeiffer AFH, Pedersen E, Schwab U. et al. The Effects of Different Quantities and Qualities of Protein Intake in People with Diabetes Mellitus. Nutrients 2020; 12 DOI: 10.3390/nu12020365.
  • 143 Mittendorfer B, Klein S, Fontana L. A word of caution against excessive protein intake. Nat Rev Endocrinol 2020; 16: 59-66 DOI: 10.1038/s41574-019-0274-7.
  • 144 Labonte CC, Chevalier S, Marliss EB. et al. Effect of 10% dietary protein intake on whole body protein kinetics in type 2 diabetic adults. Clin Nutr 2015; 34: 1115-1121 DOI: 10.1016/j.clnu.2014.12.022.
  • 145 Markova M, Hornemann S, Sucher S. et al. Rate of appearance of amino acids after a meal regulates insulin and glucagon secretion in patients with type 2 diabetes: a randomized clinical trial. Am J Clin Nutr 2018; 108: 279-291 DOI: 10.1093/ajcn/nqy100.
  • 146 Volkert D. Aktuelle ESPEN-Leitlinie Klinische Ernährung und Hydration in der Geriatrie. Dtsch Med Wochenschr 2020; 145: 1306-1314 DOI: 10.1055/a-0986-2892.
  • 147 Song M, Fung TT, Hu FB. et al. Association of Animal and Plant Protein Intake With All-Cause and Cause-Specific Mortality. JAMA Intern Med 2016; 176: 1453-1463 DOI: 10.1001/jamainternmed.2016.4182.
  • 148 Ye J, Yu Q, Mai W. et al. Dietary protein intake and subsequent risk of type 2 diabetes: a dose-response meta-analysis of prospective cohort studies. Acta Diabetol 2019; 56: 851-870 DOI: 10.1007/s00592-019-01320-x.
  • 149 Vernooij RWM, Zeraatkar D, Han MA. et al. Patterns of Red and Processed Meat Consumption and Risk for Cardiometabolic and Cancer Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann Intern Med 2019; 171: 732-741 DOI: 10.7326/M19-1583.
  • 150 Vogtschmidt YD, Raben A, Faber I. et al. Is protein the forgotten ingredient: Effects of higher compared to lower protein diets on cardiometabolic risk factors. A systematic review and meta-analysis of randomised controlled trials. Atherosclerosis 2021; DOI: 10.1016/j.atherosclerosis.2021.05.011.
  • 151 Clifton PM, Condo D, Keogh JB. Long term weight maintenance after advice to consume low carbohydrate, higher protein diets-a systematic review and meta analysis. Nutr Metab Cardiovasc Dis 2014; 24: 224-235 DOI: 10.1016/j.numecd.2013.11.006.
  • 152 Hahn D, Hodson EM, Fouque D. Low protein diets for non-diabetic adults with chronic kidney disease. Cochrane Database Syst Rev 2020; 10: CD001892 DOI: 10.1002/14651858.CD001892.pub5.
  • 153 Ikizler TA, Burrowes JD, Byham-Gray LD. et al. KDOQI Clinical Practice Guideline for Nutrition in CKD: 2020 Update. Am J Kidney Dis 2020; 76: S1-S107 DOI: 10.1053/j.ajkd.2020.05.006.
  • 154 Menon V, Kopple JD, Wang X. et al. Effect of a very low-protein diet on outcomes: long-term follow-up of the Modification of Diet in Renal Disease (MDRD) Study. Am J Kidney Dis 2009; 53: 208-217 DOI: 10.1053/j.ajkd.2008.08.009.
  • 155 Jiang Z. Effect of restricted protein diet supplemented with keto analogues in end-stage renal disease: a systematic review and meta-analysis. International urology and nephrology 2017; 1-8 DOI: 10.1007/s11255-017-1713-9.
  • 156 Fiaccadori E, Sabatino A, Barazzoni R. et al. ESPEN guideline on clinical nutrition in hospitalized patients with acute or chronic kidney disease. Clin Nutr 2021; 40: 1644-1668 DOI: 10.1016/j.clnu.2021.01.028.
  • 157 Dong JY, Zhang ZL, Wang PY. et al. Effects of high-protein diets on body weight, glycaemic control, blood lipids and blood pressure in type 2 diabetes: meta-analysis of randomised controlled trials. Br J Nutr 2013; 110: 781-789 DOI: 10.1017/S0007114513002055.
  • 158 [Anonymous]. Carbohydrates in human nutrition. Report of a Joint FAO/WHO Expert Consultation. FAO Food Nutr Pap 1998; 66: 1-140
  • 159 Wolever TMS. Personalized nutrition by prediction of glycaemic responses: fact or fantasy?. Eur J Clin Nutr 2016; 70: 411-413 DOI: 10.1038/ejcn.2016.31.
  • 160 Berry SE, Valdes AM, Drew DA. et al. Human postprandial responses to food and potential for precision nutrition. Nat Med 2020; 26: 964-973 DOI: 10.1038/s41591-020-0934-0.
  • 161 Zeevi D, Korem T, Zmora N. et al. Personalized Nutrition by Prediction of Glycemic Responses. Cell 2015; 163: 1079-1094 DOI: 10.1016/j.cell.2015.11.001.
  • 162 Jung CH, Choi KM. Impact of High-Carbohydrate Diet on Metabolic Parameters in Patients with Type 2 Diabetes. Nutrients 2017; 9 DOI: 10.3390/nu9040322.
  • 163 Livesey G, Taylor R, Livesey H. et al. Is there a dose-response relation of dietary glycemic load to risk of type 2 diabetes? Meta-analysis of prospective cohort studies. Am J Clin Nutr 2013; 97: 584-596 DOI: 10.3945/ajcn.112.041467.
  • 164 Livesey G, Livesey H. Coronary Heart Disease and Dietary Carbohydrate, Glycemic Index, and Glycemic Load: Dose-Response Meta-analyses of Prospective Cohort Studies. Mayo Clin Proc Innov Qual Outcomes 2019; 3: 52-69 DOI: 10.1016/j.mayocpiqo.2018.12.007.
  • 165 Thomas DE, Elliott EJ. The use of low-glycaemic index diets in diabetes control. Br J Nutr 2010; 104: 797-802 DOI: 10.1017/S0007114510001534.
  • 166 Xu B, Fu J, Qiao Y. et al. Higher intake of microbiota-accessible carbohydrates and improved cardiometabolic risk factors: a meta-analysis and umbrella review of dietary management in patients with type 2 diabetes. Am J Clin Nutr 2021; DOI: 10.1093/ajcn/nqaa435.
  • 167 Jenkins DJA, Kendall CWC, McKeown-Eyssen G. et al. Effect of a lowglycemic index or a high-cereal fiber diet on type 2 diabetes: a randomized trial. JAMA 2008; 300: 2742-2753 DOI: 10.1001/jama.2008.808.
  • 168 Holub I, Gostner A, Hessdörfer S. et al. Improved metabolic control after 12-week dietary intervention with low glycaemic isomalt in patients with type 2 diabetes mellitus. Horm Metab Res 2009; 41: 886-892 DOI: 10.1055/s-0029-1234107.
  • 169 Brand-Miller J, Hayne S, Petocz P. et al. Low-glycemic index diets in the management of diabetes: a meta-analysis of randomized controlled trials. Diabetes care 2003; 26: 2261-2267 DOI: 10.2337/diacare.26.8.2261.
  • 170 Ojo O, Ojo OO, Adebowale F. et al. 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 DOI: 10.3390/nu10030373.
  • 171 Franz MJ, MacLeod J, Evert A. et al. Academy of Nutrition and Dietetics Nutrition Practice Guideline for Type 1 and Type 2 Diabetes in Adults: Systematic Review of Evidence for Medical Nutrition Therapy Effectiveness and Recommendations for Integration into the Nutrition Care Process. J Acad Nutr Diet 2017; 117: 1659-1679 DOI: 10.1016/j.jand.2017.03.022.
  • 172 Vega-López S, Venn BJ, Slavin JL. Relevance of the Glycemic Index and Glycemic Load for Body Weight, Diabetes, and Cardiovascular Disease. Nutrients 2018; 10 DOI: 10.3390/nu10101361.
  • 173 Jenkins DJA, Dehghan M, Mente A. et al. Glycemic Index, Glycemic Load, and Cardiovascular Disease and Mortality. N Engl J Med 2021; 384: 1312-1322 DOI: 10.1056/NEJMoa2007123.
  • 174 Coutinho M, Gerstein HC, Wang Y. et al. The relationship between glucose and incident cardiovascular events. A metaregression analysis of published data from 20 studies of 95783 individuals followed for 12.4 years. Diabetes care 1999; 22: 233-240 DOI: 10.2337/diacare.22.2.233.
  • 175 Levitan EB, Song Y, Ford ES. et al. Is nondiabetic hyperglycemia a risk factor for cardiovascular disease? A meta-analysis of prospective studies. Arch Intern Med 2004; 164: 2147-2155 DOI: 10.1001/archinte.164.19.2147.
  • 176 Siri PW, Krauss RM. Influence of dietary carbohydrate and fat on LDL and HDL particle distributions. Curr Atheroscler Rep 2005; 7: 455-459 DOI: 10.1007/s11883-005-0062-9.
  • 177 Aune D, Norat T, Romundstad P. et al. Whole grain and refined grain consumption and the risk of type 2 diabetes: a systematic review and dose-response meta-analysis of cohort studies. Eur J Epidemiol 2013; 28: 845-858 DOI: 10.1007/s10654-013-9852-5.
  • 178 [Anonymous]. Dietary fibre and incidence of type 2 diabetes in eight European countries: the EPIC-InterAct Study and a meta-analysis of prospective studies. Diabetologia 2015; 58: 1394-1408 DOI: 10.1007/s00125-015-3585-9.
  • 179 Kim Y, Je Y. Dietary fibre intake and mortality from cardiovascular disease and all cancers: A meta-analysis of prospective cohort studies. Arch Cardiovasc Dis 2016; 109: 39-54 DOI: 10.1016/j.acvd.2015.09.005.
  • 180 Reynolds AN, Akerman AP, Mann J. Dietary fibre and whole grains in diabetes management: Systematic review and meta-analyses. PLoS Med 2020; 17: e1003053 DOI: 10.1371/journal.pmed.1003053.
  • 181 Da Silva Borges D, Fernandes R, Thives Mello A. et al. Prebiotics may reduce serum concentrations of C-reactive protein and ghrelin in overweight and obese adults: a systematic review and meta-analysis. Nutr Rev 2020; 78: 235-248 DOI: 10.1093/nutrit/nuz045.
  • 182 Reynolds A, Mann J, Cummings J. et al. Carbohydrate quality and human health: a series of systematic reviews and meta-analyses. Lancet 2019; 393: 434-445 DOI: 10.1016/S0140-6736(18)31809-9.
  • 183 Musa-Veloso K, Poon T, Harkness LS. et al. The effects of whole-grain compared with refined wheat, rice, and rye on the postprandial blood glucose response: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2018; 108: 759-774 DOI: 10.1093/ajcn/nqy112.
  • 184 Wang W, Li J, Chen X. et al. Whole grain food diet slightly reduces cardiovascular risks in obese/overweight adults: a systematic review and meta-analysis. BMC Cardiovasc Disord 2020; 20: 82 DOI: 10.1186/s12872-020-01337-z.
  • 185 Weickert MO, Roden M, Isken F. et al. Effects of supplemented isoenergetic diets differing in cereal fiber and protein content on insulin sensitivity in overweight humans. Am J Clin Nutr 2011; 94: 459-471 DOI: 10.3945/ajcn.110.004374.
  • 186 Honsek C, Kabisch S, Kemper M. et al. Fibre supplementation for the prevention of type 2 diabetes and improvement of glucose metabolism: the randomised controlled Optimal Fibre Trial (OptiFiT). Diabetologia 2018; 61: 1295-1305 DOI: 10.1007/s00125-018-4582-6.
  • 187 Kabisch S, Meyer NMT, Honsek C. et al. Fasting Glucose State Determines Metabolic Response to Supplementation with Insoluble Cereal Fibre: A Secondary Analysis of the Optimal Fibre Trial (OptiFiT). Nutrients 2019; 11 DOI: 10.3390/nu11102385.
  • 188 Hjorth MF, Ritz C, Blaak EE. et al. Pretreatment fasting plasma glucose and insulin modify dietary weight loss success: results from 3 randomized clinical trials. Am J Clin Nutr 2017; 106: 499-505 DOI: 10.3945/ajcn.117.155200.
  • 189 Xiao Z, Chen H, Zhang Y. et al. The effect of psyllium consumption on weight, body mass index, lipid profile, and glucose metabolism in diabetic patients: A systematic review and dose-response meta-analysis of randomized controlled trials. Phytother Res 2020; 34: 1237-1247 DOI: 10.1002/ptr.6609.
  • 190 Wang L, Yang H, Huang H. et al. Inulin-type fructans supplementation improves glycemic control for the prediabetes and type 2 diabetes populations: results from a GRADE-assessed systematic review and doseresponse meta-analysis of 33 randomized controlled trials. J Transl Med 2019; 17: 410 DOI: 10.1186/s12967-019-02159-0.
  • 191 Rao M, Gao C, Xu L. et al. Effect of Inulin-Type Carbohydrates on Insulin Resistance in Patients with Type 2 Diabetes and Obesity: A Systematic Review and Meta-Analysis. J Diabetes Res 2019; 2019: 5101423 DOI: 10.1155/2019/5101423.
  • 192 Darooghegi Mofrad M, Mozaffari H, Mousavi SM. et al. The effects of psyllium supplementation on body weight, body mass index and waist circumference in adults: A systematic review and dose-response metaanalysis of randomized controlled trials. Crit Rev Food Sci Nutr 2020; 60: 859-872 DOI: 10.1080/10408398.2018.1553140.
  • 193 Rahmani J, Miri A, Černevičiūtė R. et al. Effects of cereal beta-glucan consumption on body weight, body mass index, waist circumference and total energy intake: A meta-analysis of randomized controlled trials. Complement Ther Med 2019; 43: 131-139 DOI: 10.1016/j.ctim.2019.01.018.
  • 194 Ho HVT, Sievenpiper JL, Zurbau A. et al. The effect of oat β-glucan on LDL-cholesterol, non-HDL-cholesterol and apoB for CVD risk reduction: a systematic review and meta-analysis of randomised-controlled trials. Br J Nutr 2016; 116: 1369-1382 DOI: 10.1017/S000711451600341X.
  • 195 Jovanovski E, Yashpal S, Komishon A. et al. Effect of psyllium (Plantago ovata) fiber on LDL cholesterol and alternative lipid targets, non-HDL cholesterol and apolipoprotein B: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2018; 108: 922-932 DOI: 10.1093/ajcn/nqy115.
  • 196 Brum J, Ramsey D, McRorie J. et al. Meta-Analysis of Usefulness of Psyllium Fiber as Adjuvant Antilipid Therapy to Enhance Cholesterol Lowering Efficacy of Statins. The American journal of cardiology 2018; 122: 1169-1174 DOI: 10.1016/j.amjcard.2018.06.040.
  • 197 Ho HVT, Jovanovski E, Zurbau A. et al. A systematic review and metaanalysis of randomized controlled trials of the effect of konjac glucomannan, a viscous soluble fiber, on LDL cholesterol and the new lipid targets non-HDL cholesterol and apolipoprotein B. Am J Clin Nutr 2017; 105: 1239-1247 DOI: 10.3945/ajcn.116.142158.
  • 198 Pittler MH, Ernst E. Guar gum for body weight reduction: meta-analysis of randomized trials. Am J Med 2001; 110: 724-730 DOI: 10.1016/s0002-9343(01)00702-1.
  • 199 Khan K, Jovanovski E, Ho HVT. et al. The effect of viscous soluble fiber on blood pressure: A systematic review and meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis 2018; 28: 3-13 DOI: 10.1016/j.numecd.2017.09.007.
  • 200 Thinggaard M, Jacobsen R, Jeune B. et al. Is the relationship between BMI and mortality increasingly U-shaped with advancing age? A 10-year follow-up of persons aged 70-95 years. J Gerontol A Biol Sci Med Sci 2010; 65: 526-531 DOI: 10.1093/gerona/glp214.
  • 201 Guigoz Y, Vellas B. Malnutrition in the elderly: the Mini Nutritional Assessment (MNA). Ther Umsch 1997; 54: 345-350
  • 202 Rubenstein LZ, Harker JO, Salvà A. et al. Screening for undernutrition in geriatric practice: developing the short-form mini-nutritional assessment (MNA-SF). J Gerontol A Biol Sci Med Sci 2001; 56: M366-M372 DOI: 10.1093/gerona/56.6.m366.
  • 203 [Anonym]. S2k-Leitlinie Diagnostik, Therapie und Verlaufskontrolle des Diabetes mellitus im Alter. 2. Auflage 2018 – AWMF-Register-Nr. 057-017. Diabetologie und Stoffwechsel 2018; 13: 423-489 DOI: 10.1055/a-0666-0820.
  • 204 Volkert D, Bauer J, Frühwald T. et al. S3-Leitlinie der Deutschen Gesellschaft für Ernährungsmedizin (DGEM) in Zusammenarbeit mit der GESKES, der AKE und der DGG Klinische Ernährung in der Geriatrie. Aktuelle Ernährungsmedizin 2013; 38: e1-e48
  • 205 Zeyfang A, Wernecke J, Bahrmann A. Diabetes mellitus im Alter. Diabetologie 2020; 15: S112-S119
  • 206 Şat S, Aydınkoç-Tuzcu K, Berger F. et al. Diabetes und Migration. Diabetologie und Stoffwechsel 2019; 14: S306-S317 DOI: 10.1055/a-0899-2742.
  • 207 Diker O, Deniz T, Çetinkaya A. History of Turkish Cuisine Culture and the Influence of the Balkans. Journal of Humanities And Social Science 2016; 10: 1-6
  • 208 Schmid B. Ernährung und Migration [Zugl.: München, Techn. Univ., Diss., 2003]. München: Utz, Wiss; c; 2003
  • 209 Magni P, Bier DM, Pecorelli S. et al. Perspective: Improving Nutritional Guidelines for Sustainable Health Policies: Current Status and Perspectives. Adv Nutr 2017; 8: 532-545 DOI: 10.3945/an.116.014738.
  • 210 Praxistool zur Ernährung. Orientierungshilfe für die Diabetesberatung nach geografischen Räumen. Im Internet (Stand: 15.07.2021) https://migration.deutsche-diabetes-gesellschaft.de/fileadmin/user_upload/01_Die_DDG/05_Arbeitsgemeinschaften/AG_Migranten/Microsite/200417_Ernaehrungstoo_DDG-GB19-Einleger_04.pdf
  • 211 European Commission. Health Promotion and Disease Prevention Knowledge Gateway: Sugars and Sweeteners. Im Internet (Stand: 27.01.2021) https://ec.europa.eu/jrc/en/health-knowledge-gateway/promotion-prevention/nutrition/sugars-sweeteners
  • 212 Scientific Advisory Committee on Nutrition. Carbohydrates and Health report, 2015. Im Internet (Stand: 26.01.2021) https://www.gov.uk/government/publications/
  • 213 McKeown NM, Dashti HS, Ma J. et al. Sugar-sweetened beverage intake associations with fasting glucose and insulin concentrations are not modified by selected genetic variants in a ChREBP-FGF21 pathway: a meta-analysis. Diabetologia 2018; 61: 317-330 DOI: 10.1007/s00125-017-4475-0.
  • 214 Evans RA, Frese M, Romero J. et al. Chronic fructose substitution for glucose or sucrose in food or beverages has little effect on fasting blood glucose, insulin, or triglycerides: a systematic review and metaanalysis. Am J Clin Nutr 2017; 106: 519-529 DOI: 10.3945/ajcn.116.145169.
  • 215 Evans RA, Frese M, Romero J. et al. Fructose replacement of glucose or sucrose in food or beverages lowers postprandial glucose and insulin without raising triglycerides: a systematic review and meta-analysis. Am J Clin Nutr 2017; 106: 506-518 DOI: 10.3945/ajcn.116.145151.
  • 216 Keller A, Heitmann BL, Olsen N. Sugar-sweetened beverages, vascular risk factors and events: a systematic literature review. Public Health Nutr 2015; 18: 1145-1154 DOI: 10.1017/S1368980014002122.
  • 217 Huang C, Huang J, Tian Y. et al. Sugar sweetened beverages consumption and risk of coronary heart disease: a meta-analysis of prospective studies. Atherosclerosis 2014; 234: 11-16 DOI: 10.1016/j.atherosclerosis.2014.01.037.
  • 218 Narain A, Kwok CS, Mamas MA. Soft drinks and sweetened beverages and the risk of cardiovascular disease and mortality: a systematic review and meta-analysis. Int J Clin Pract 2016; 70: 791-805 DOI: 10.1111/ijcp.12841.
  • 219 Cheungpasitporn W, Thongprayoon C, O’Corragain OA. et al. Associations of sugar-sweetened and artificially sweetened soda with chronic kidney disease: a systematic review and meta-analysis. Nephrology (Carlton) 2014; 19: 791-797 DOI: 10.1111/nep.12343.
  • 220 Chen H, Wang J, Li Z. et al. Consumption of Sugar-Sweetened Beverages Has a Dose-Dependent Effect on the Risk of Non-Alcoholic Fatty Liver Disease: An Updated Systematic Review and Dose-Response Meta-Analysis. Int J Environ Res Public Health 2019; 16 DOI: 10.3390/ijerph16122192.
  • 221 Asgari-Taee F, Zerafati-Shoae N, Dehghani M. et al. Association of sugar sweetened beverages consumption with non-alcoholic fatty liver disease: a systematic review and meta-analysis. Eur J Nutr 2019; 58: 1759-1769 DOI: 10.1007/s00394-018-1711-4.
  • 222 Khan TA, Sievenpiper JL. Controversies about sugars: results from systematic reviews and meta-analyses on obesity, cardiometabolic disease and diabetes. Eur J Nutr 2016; 55: 25-43 DOI: 10.1007/s00394-016-1345-3.
  • 223 Choo VL, Viguiliouk E, Blanco Mejia S. et al. Food sources of fructosecontaining sugars and glycaemic control: systematic review and metaanalysis of controlled intervention studies. BMJ 2018; 363: k4644 DOI: 10.1136/bmj.k4644.
  • 224 Semnani-Azad Z, Khan TA, Blanco Mejia S. et al. Association of Major Food Sources of Fructose-Containing Sugars With Incident Metabolic Syndrome: A Systematic Review and Meta-analysis. JAMA Netw Open 2020; 3: e209993 DOI: 10.1001/jamanetworkopen.2020.9993.
  • 225 Bechthold A. Vollwertig essen und trinken nach den 10 Regeln der DGE. Bonn: Deutsche Gesellschaft für Ernährung e. V. (DGE); 2018
  • 226 Wu H, Flint AJ, Qi Q. et al. Association between dietary whole grain intake and risk of mortality: two large prospective studies in US men and women. JAMA Intern Med 2015; 175: 373-384 DOI: 10.1001/jamainternmed.2014.6283.
  • 227 Johnsen NF, Frederiksen K, Christensen J. et al. Whole-grain products and whole-grain types are associated with lower all-cause and causespecific mortality in the Scandinavian HELGA cohort. Br J Nutr 2015; 114: 608-623 DOI: 10.1017/S0007114515001701.
  • 228 Wei H, Gao Z, Liang R. et al. Whole-grain consumption and the risk of all-cause, CVD and cancer mortality: a meta-analysis of prospective cohort studies – CORRIGENDUM. Br J Nutr 2016; 116: 952 DOI: 10.1017/S0007114516002543.
  • 229 Chen GC, Tong X, Xu JY. et al. Whole-grain intake and total, cardiovascular, and cancer mortality: a systematic review and meta-analysis of prospective studies. Am J Clin Nutr 2016; 104: 164-172 DOI: 10.3945/ajcn.115.122432.
  • 230 Benisi-Kohansal S, Saneei P, Salehi-Marzijarani M. et al. Whole-Grain Intake and Mortality from All Causes, Cardiovascular Disease, and Cancer: A Systematic Review and Dose-Response Meta-Analysis of Prospective Cohort Studies. Adv Nutr 2016; 7: 1052-1065 DOI: 10.3945/an.115.011635.
  • 231 Zong G, Gao A, Hu FB. et al. Whole Grain Intake and Mortality From All Causes, Cardiovascular Disease, and Cancer: A Meta-Analysis of Prospective Cohort Studies. Circulation 2016; 133: 2370-2380 DOI: 10.1161/CIRCULATIONAHA.115.021101.
  • 232 Aune D, Keum N, Giovannucci E. et al. Whole grain consumption and risk of cardiovascular disease, cancer, and all cause and cause specific mortality: systematic review and dose-response meta-analysis of prospective studies. BMJ 2016; 353: i2716 DOI: 10.1136/bmj.i2716.
  • 233 Aune D. Plant Foods, Antioxidant Biomarkers, and the Risk of Cardiovascular Disease, Cancer, and Mortality: A Review of the Evidence. Adv Nutr 2019; 10: S404-S421 DOI: 10.1093/advances/nmz042.
  • 234 Zhang B, Zhao Q, Guo W. et al. Association of whole grain intake with all-cause, cardiovascular, and cancer mortality: a systematic review and dose-response meta-analysis from prospective cohort studies. Eur J Clin Nutr 2018; 72: 57-65 DOI: 10.1038/ejcn.2017.149.
  • 235 Jenkins DJ, Wesson V, Wolever TM. et al. Wholemeal versus wholegrain breads: proportion of whole or cracked grain and the glycaemic response. BMJ 1988; 297: 958-960 DOI: 10.1136/bmj.297.6654.958.
  • 236 Reynolds AN, Mann J, Elbalshy M. et al. Wholegrain Particle Size Influences Postprandial Glycemia in Type 2 Diabetes: A Randomized Crossover Study Comparing Four Wholegrain Breads. Diabetes care 2020; 43: 476-479 DOI: 10.2337/dc19-1466.
  • 237 Åberg S, Mann J, Neumann S. et al. Whole-Grain Processing and Glycemic Control in Type 2 Diabetes: A Randomized Crossover Trial. Diabetes care 2020; 43: 1717-1723 DOI: 10.2337/dc20-0263.
  • 238 Jenkins DJA, Kendall CWC, Augustin LSA. et al. Effect of wheat bran on glycemic control and risk factors for cardiovascular disease in type 2 diabetes. Diabetes care 2002; 25: 1522-1528 DOI: 10.2337/diacare.25.9.1522.
  • 239 Miller V, Mente A, Dehghan M. et al. Fruit, vegetable, and legume intake, and cardiovascular disease and deaths in 18 countries (PURE): a prospective cohort study. Lancet 2017; 390: 2037-2049 DOI: 10.1016/S0140-6736(17)32253-5.
  • 240 Aune D, Giovannucci E, Boffetta P. et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality-a systematic review and dose-response meta-analysis of prospective studies. Int J Epidemiol 2017; 46: 1029-1056 DOI: 10.1093/ije/dyw319.
  • 241 Bechthold A, Boeing H, Schwedhelm C. et al. Food groups and risk of coronary heart disease, stroke and heart failure: A systematic review and dose-response meta-analysis of prospective studies. Crit Rev Food Sci Nutr 2019; 59: 1071-1090 DOI: 10.1080/10408398.2017.1392288.
  • 242 Zhan J, Liu YJ, Cai LB. et al. Fruit and vegetable consumption and risk of cardiovascular disease: A meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr 2017; 57: 1650-1663 DOI: 10.1080/10408398.2015.1008980.
  • 243 Willett W, Rockström J, Loken B. et al. Food in the Anthropocene: the EAT-Lancet Commission on healthy diets from sustainable food systems. Lancet 2019; 393: 447-492 DOI: 10.1016/S0140-6736(18)31788-4.
  • 244 Barnard ND, Cohen J, Jenkins DJA. et al. A low-fat vegan diet improves glycemic control and cardiovascular risk factors in a randomized clinical trial in individuals with type 2 diabetes. Diabetes care 2006; 29: 1777-1783 DOI: 10.2337/dc06-0606.
  • 245 Jenkins DJA, Kendall CWC, Augustin LSA. et al. Effect of legumes as part of a low glycemic index diet on glycemic control and cardiovascular risk factors in type 2 diabetes mellitus: a randomized controlled trial. Arch Intern Med 2012; 172: 1653-1660 DOI: 10.1001/2013.jamainternmed.70.
  • 246 Renner B, Arens-Azevêdo U, Watzl B. et al. DGE-Positionspapier zur nachhaltigeren Ernährung. ernährungsumschau 2021; 68: 144-154
  • 247 Jannasch F, Kröger J, Schulze MB. Dietary Patterns and Type 2 Diabetes: A Systematic Literature Review and Meta-Analysis of Prospective Studies. J Nutr 2017; 147: 1174-1182 DOI: 10.3945/jn.116.242552.
  • 248 Wallin A, Di Giuseppe D, Orsini N. et al. Fish consumption, dietary longchain n-3 fatty acids, and risk of type 2 diabetes: systematic review and meta-analysis of prospective studies. Diabetes care 2012; 35: 918-929 DOI: 10.2337/dc11-1631.
  • 249 Xun P, He K. Fish Consumption and Incidence of Diabetes: meta-analysis of data from 438000 individuals in 12 independent prospective cohorts with an average 11-year follow-up. Diabetes care 2012; 35: 930-938 DOI: 10.2337/dc11-1869.
  • 250 Schwingshackl L, Hoffmann G, Lampousi AM. et al. Food groups and risk of type 2 diabetes mellitus: a systematic review and meta-analysis of prospective studies. Eur J Epidemiol 2017; 32: 363-375 DOI: 10.1007/s10654-017-0246-y.
  • 251 Muley A, Muley P, Shah MALA. fatty fish or marine n-3 fatty acids for preventing DM?: a systematic review and meta-analysis. Curr Diabetes Rev 2014; 10: 158-165 DOI: 10.2174/1573399810666140515113137.
  • 252 Schlesinger S, Neuenschwander M, Schwedhelm C. et al. Food Groups and Risk of Overweight, Obesity, and Weight Gain: A Systematic Review and Dose-Response Meta-Analysis of Prospective Studies. Adv Nutr 2019; 10: 205-218 DOI: 10.1093/advances/nmy092.
  • 253 Micha R, Shulkin ML, Peñalvo JL. et al. Etiologic effects and optimal intakes of foods and nutrients for risk of cardiovascular diseases and diabetes: Systematic reviews and meta-analyses from the Nutrition and Chronic Diseases Expert Group (NutriCoDE). PLoS One 2017; 12: e0175149 DOI: 10.1371/journal.pone.0175149.
  • 254 Jayedi A, Shab-Bidar S, Eimeri S. et al. Fish consumption and risk of allcause and cardiovascular mortality: a dose-response meta-analysis of prospective observational studies. Public Health Nutr 2018; 21: 1297-1306 DOI: 10.1017/S1368980017003834.
  • 255 Abdelhamid AS, Brown TJ, Brainard JS. et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11: CD003177 DOI: 10.1002/14651858.CD003177.pub4.
  • 256 Hu Y, Hu FB, Manson JE. Marine Omega-3 Supplementation and Cardiovascular Disease: An Updated Meta-Analysis of 13 Randomized Controlled Trials Involving 127 477 Participants. J Am Heart Assoc 2019; 8: e013543 DOI: 10.1161/JAHA.119.013543.
  • 257 Gao H, Geng T, Huang T. et al. Fish oil supplementation and insulin sensitivity: a systematic review and meta-analysis. Lipids Health Dis 2017; 16: 131 DOI: 10.1186/s12944-017-0528-0.
  • 258 Chen C, Yu X, Shao S. Effects of Omega-3 Fatty Acid Supplementation on Glucose Control and Lipid Levels in Type 2 Diabetes: A Meta-Analysis. PLoS One 2015; 10: e0139565 DOI: 10.1371/journal.pone.0139565.
  • 259 DGE – Deutsche Gesellschaft für Ernährung. Vollwertig essen und trinken nach den 10 Regeln der DGE. Im Internet (Stand: 13.07.2021) https://www.dge.de/ernaehrungspraxis/vollwertige-ernaehrung/10-regeln-der-dge/
  • 260 Zeraatkar D, Han MA, Guyatt GH. et al. Red and Processed Meat Consumption and Risk for All-Cause Mortality and Cardiometabolic Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann Intern Med 2019; 171: 703-710 DOI: 10.7326/M19-0655.
  • 261 Davidson MH, Hunninghake D, Maki KC. et al. Comparison of the effects of lean red meat vs lean white meat on serum lipid levels among freeliving persons with hypercholesterolemia: a long-term, randomized clinical trial. Arch Intern Med 1999; 159: 1331-1338 DOI: 10.1001/archinte.159.12.1331.
  • 262 Hunninghake DB, Maki KC, Kwiterovich PO. et al. Incorporation of lean red meat into a National Cholesterol Education Program Step I diet: a long-term, randomized clinical trial in free-living persons with hypercholesterolemia. J Am Coll Nutr 2000; 19: 351-360 DOI: 10.1080/07315724.2000.10718931.
  • 263 Bergeron N, Chiu S, Williams PT. et al. Effects of red meat, white meat, and nonmeat protein sources on atherogenic lipoprotein measures in the context of low compared with high saturated fat intake: a randomized controlled trial. Am J Clin Nutr 2019; 110: 24-33 DOI: 10.1093/ajcn/nqz035.
  • 264 Charlton K, Walton K, Batterham M. et al. Pork and Chicken Meals Similarly Impact on Cognitive Function and Strength in Community-Living Older Adults: A Pilot Study. J Nutr Gerontol Geriatr 2016; 35: 124-145 DOI: 10.1080/21551197.2016.1162259.
  • 265 Murphy KJ, Parker B, Dyer KA. et al. A comparison of regular consumption of fresh lean pork, beef and chicken on body composition: a randomized cross-over trial. Nutrients 2014; 6: 682-696 DOI: 10.3390/nu6020682.
  • 266 Murphy KJ, Thomson RL, Coates AM. et al. Effects of eating fresh lean pork on cardiometabolic health parameters. Nutrients 2012; 4: 711-723 DOI: 10.3390/nu4070711.
  • 267 Johnston BC, Zeraatkar D, Han MA. et al. Unprocessed Red Meat and Processed Meat Consumption: Dietary Guideline Recommendations From the Nutritional Recommendations (NutriRECS) Consortium. Ann Intern Med 2019; 171: 756-764 DOI: 10.7326/M19-1621.
  • 268 Davis PA, Yokoyama W. Cinnamon intake lowers fasting blood glucose: meta-analysis. J Med Food 2011; 14: 884-889 DOI: 10.1089/jmf.2010.0180.
  • 269 Akilen R, Tsiami A, Devendra D. et al. Cinnamon in glycaemic control: Systematic review and meta analysis. Clin Nutr 2012; 31: 609-615 DOI: 10.1016/j.clnu.2012.04.003.
  • 270 Leach MJ, Kumar S. Cinnamon for diabetes mellitus. Cochrane Database Syst Rev 2012; DOI: 10.1002/14651858.CD007170.pub2.
  • 271 Allen RW, Schwartzman E, Baker WL. et al. Cinnamon use in type 2 diabetes: an updated systematic review and meta-analysis. Ann Fam Med 2013; 11: 452-459 DOI: 10.1370/afm.1517.
  • 272 Costello RB, Dwyer JT, Saldanha L. et al. Do Cinnamon Supplements Have a Role in Glycemic Control in Type 2 Diabetes? A Narrative Review. J Acad Nutr Diet 2016; 116: 1794-1802 DOI: 10.1016/j.jand.2016.07.015.
  • 273 Sierra-Puente D, Abadi-Alfie S, Arakanchi-Altaled K. et al. Cinammon (Cinnamomum Spp.) and Type 2 Diabetes Mellitus. CTNR 2019; 18: 247-255 DOI: 10.37290/ctnr2641-452X.18:247-255.
  • 274 Chan CB, Hashemi Z, Subhan FB. The impact of low and no-caloric sweeteners on glucose absorption, incretin secretion, and glucose tolerance. Appl Physiol Nutr Metab 2017; 42: 793-801 DOI: 10.1139/apnm-2016-0705.
  • 275 Brown AW, Bohan Brown MM, Onken KL. et al. Short-term consumption of sucralose, a nonnutritive sweetener, is similar to water with regard to select markers of hunger signaling and short-term glucose homeostasis in women. Nutr Res 2011; 31: 882-888 DOI: 10.1016/j.nutres.2011.10.004.
  • 276 Ford HE, Peters V, Martin NM. et al. Effects of oral ingestion of sucralose on gut hormone response and appetite in healthy normal-weight subjects. Eur J Clin Nutr 2011; 65: 508-513 DOI: 10.1038/ejcn.2010.291.
  • 277 Steinert RE, Frey F, Töpfer A. et al. Effects of carbohydrate sugars and artificial sweeteners on appetite and the secretion of gastrointestinal satiety peptides. Br J Nutr 2011; 105: 1320-1328 DOI: 10.1017/S000711451000512X.
  • 278 Barriocanal LA, Palacios M, Benitez G. et al. Apparent lack of pharmacological effect of steviol glycosides used as sweeteners in humans. A pilot study of repeated exposures in some normotensive and hypotensive individuals and in Type 1 and Type 2 diabetics. Regul Toxicol Pharmacol 2008; 51: 37-41 DOI: 10.1016/j.yrtph.2008.02.006.
  • 279 Brown RJ, Walter M, Rother KI. Effects of diet soda on gut hormones in youths with diabetes. Diabetes care 2012; 35: 959-964 DOI: 10.2337/dc11-2424.
  • 280 Grotz VL, Henry RR, McGill JB. et al. Lack of effect of sucralose on glucose homeostasis in subjects with type 2 diabetes. J Am Diet Assoc 2003; 103: 1607-1612 DOI: 10.1016/j.jada.2003.09.021.
  • 281 Maki KC, Curry LL, Reeves MS. et al. Chronic consumption of rebaudioside A, a steviol glycoside, in men and women with type 2 diabetes mellitus. Food Chem Toxicol 2008; 46: S47-S53 DOI: 10.1016/j.fct.2008.05.007.
  • 282 Olalde-Mendoza L, Moreno-González YE. Modificación de la glucemia en ayuno en adultos con diabetes mellitus tipo 2 después de la ingesta de refrescos de cola y de dieta en el estado de querétaro, México. Arch Latinoam Nutr 2013; 63: 142-147
  • 283 Temizkan S, Deyneli O, Yasar M. et al. Sucralose enhances GLP-1 release and lowers blood glucose in the presence of carbohydrate in healthy subjects but not in patients with type 2 diabetes. Eur J Clin Nutr 2015; 69: 162-166 DOI: 10.1038/ejcn.2014.208.
  • 284 Ferrazzano GF, Cantile T, Alcidi B. et al. Is Stevia rebaudiana Bertoni a Non Cariogenic Sweetener? A Review. Molecules 2015; 21: E38 DOI: 10.3390/molecules21010038.
  • 285 Prashant GM, Patil RB, Nagaraj T. et al. The antimicrobial activity of the three commercially available intense sweeteners against common periodontal pathogens: an in vitro study. J Contemp Dent Pract 2012; 13: 749-752 DOI: 10.5005/jp-journals-10024-1222.
  • 286 Suez J, Korem T, Zeevi D. et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 2014; 514: 181-186 DOI: 10.1038/nature13793.
  • 287 EFSA 2013. EFSA schließt vollständige Risikobewertung zu Aspartam ab und kommt zu dem Schluss, dass es in den derzeitigen Expositionsmengen sicher ist. Im Internet (Stand: 01.09.2020) https://www.efsa.europa.eu/de/press/news/131210
  • 288 Bundesinstitut für Risikobewertung. Bewertung von Süßstoffen und Zuckeraustauschstoffen. Hintergrundinformation Nr. 025/2014 des BfR vom 1. Juli 2014. Im Internet (Stand: 01.09.2020) www.bfr.bund.de/cm/343/bewertung_von_suessstoffen.pdf
  • 289 Bock PM, Telo GH, Ramalho R. et al. The effect of probiotics, prebiotics or synbiotics on metabolic outcomes in individuals with diabetes: a systematic review and meta-analysis. Diabetologia 2021; 64: 26-41 DOI: 10.1007/s00125-020-05295-1.
  • 290 Rittiphairoj T, Pongpirul K, Janchot K. et al. Probiotics Contribute to Glycemic Control in Patients with Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Adv Nutr 2021; 12: 722-734 DOI: 10.1093/advances/nmaa133.
  • 291 Tao YW, Gu YL, Mao XQ. et al. Effects of probiotics on type II diabetes mellitus: a meta-analysis. J Transl Med 2020; 18: 30 DOI: 10.1186/s12967-020-02213-2.
  • 292 Ardeshirlarijani E, Tabatabaei-Malazy O, Mohseni S. et al. Effect of probiotics supplementation on glucose and oxidative stress in type 2 diabetes mellitus: a meta-analysis of randomized trials. Daru 2019; 27: 827-837 DOI: 10.1007/s40199-019-00302-2.
  • 293 Mahboobi S, Rahimi F, Jafarnejad S. Effects of Prebiotic and Synbiotic Supplementation on Glycaemia and Lipid Profile in Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials. Adv Pharm Bull 2018; 8: 565-574 DOI: 10.15171/apb.2018.065.
  • 294 Akbari V, Hendijani F. Effects of probiotic supplementation in patients with type 2 diabetes: systematic review and meta-analysis. Nutr Rev 2016; 74: 774-784 DOI: 10.1093/nutrit/nuw039.
  • 295 Yao K, Zeng L, He Q. et al. Effect of Probiotics on Glucose and Lipid Metabolism in Type 2 Diabetes Mellitus: A Meta-Analysis of 12 Randomized Controlled Trials. Med Sci Monit 2017; 23: 3044-3053 DOI: 10.12659/msm.902600.
  • 296 Wang C, Zhang C, Li S. et al. Effects of Probiotic Supplementation on Dyslipidemia in Type 2 Diabetes Mellitus: A Meta-Analysis of Randomized Controlled Trials. Foods 2020; 9 DOI: 10.3390/foods9111540.
  • 297 Kasińska MA, Drzewoski J. Effectiveness of probiotics in type 2 diabetes: a meta-analysis. Pol Arch Med Wewn 2015; 125: 803-813 DOI: 10.20452/pamw.3156.
  • 298 Palacios T, Vitetta L, Coulson S. et al. Targeting the Intestinal Microbiota to Prevent Type 2 Diabetes and Enhance the Effect of Metformin on Glycaemia: A Randomised Controlled Pilot Study. Nutrients 2020; 12 DOI: 10.3390/nu12072041.
  • 299 Zheng M, Zhang R, Tian X. et al. Assessing the Risk of Probiotic Dietary Supplements in the Context of Antibiotic Resistance. Front Microbiol 2017; 8: 908 DOI: 10.3389/fmicb.2017.00908.
  • 300 Wong A, Ngu DYS, Dan LA. et al. Detection of antibiotic resistance in probiotics of dietary supplements. Nutr J 2015; 14: 95 DOI: 10.1186/s12937-015-0084-2.
  • 301 BgVV – ehemals: Bundesinstitut für gesundheitlichen Verbraucherschutz und Veterinärmedizin. Abschlussbericht der Arbeitsgruppe „Probiotische Mikroorganismenkulturen in Lebensmitteln“ am BgVV. Im Internet (Stand: 13.07.2021) https://mobil.bfr.bund.de/cm/343/probiot.pdf
  • 302 de Vrese M. Mikrobiologie, Wirkung und Sicherheit von Probiotika. Monatsschrift Kinderheilkunde 2008; 156: 1063-1069 DOI: 10.1007/s00112-008-1829-1.
  • 303 Vrieze A, van Nood E, Holleman F. et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012; 143: 913-916.e7 DOI: 10.1053/j.gastro.2012.06.031.
  • 304 Simon MC, Strassburger K, Nowotny B. et al. Intake of Lactobacillus reuteri improves incretin and insulin secretion in glucose-tolerant humans: a proof of concept. Diabetes care 2015; 38: 1827-1834 DOI: 10.2337/dc14-2690.
  • 305 Tilg H, Moschen AR. Microbiota and diabetes: an evolving relationship. Gut 2014; 63: 1513-1521 DOI: 10.1136/gutjnl-2014-306928.
  • 306 Kjems LL, Holst JJ, Vølund A. et al. The influence of GLP-1 on glucosestimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes 2003; 52: 380-386 DOI: 10.2337/diabetes.52.2.380.
  • 307 Karlsson FH, Tremaroli V, Nookaew I. et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013; 498: 99-103 DOI: 10.1038/nature12198.
  • 308 Qin J, Li Y, Cai Z. et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012; 490: 55-60 DOI: 10.1038/nature11450.
  • 309 Larsen N, Vogensen FK, van den Berg FWJ. et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One 2010; 5: e9085 DOI: 10.1371/journal.pone.0009085.
  • 310 Wu H, Esteve E, Tremaroli V. et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med 2017; 23: 850-858 DOI: 10.1038/nm.4345.
  • 311 Forslund K, Hildebrand F, Nielsen T. et al. Corrigendum: Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2017; 545: 116 DOI: 10.1038/nature22318.
  • 312 Forslund K, Hildebrand F, Nielsen T. et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2015; 528: 262-266 DOI: 10.1038/nature15766.
  • 313 Caesar R. Pharmacologic and Nonpharmacologic Therapies for the Gut Microbiota in Type 2 Diabetes. Can J Diabetes 2019; 43: 224-231 DOI: 10.1016/j.jcjd.2019.01.007.
  • 314 Evert AB, Boucher JL, Cypress M. et al. Nutrition therapy recommendations for the management of adults with diabetes. Diabetes care 2014; 37: S120-S143 DOI: 10.2337/dc14-S120.
  • 315 Sievenpiper JL, Chan CB, Dworatzek PD. et al. Nutrition Therapy. Can J Diabetes 2018; 42: S64-S79 DOI: 10.1016/j.jcjd.2017.10.009.
  • 316 Sievenpiper JL, de Souza RJ, Mirrahimi A. et al. Effect of fructose on body weight in controlled feeding trials: a systematic review and meta-analysis. Ann Intern Med 2012; 156: 291-304 DOI: 10.7326/0003-4819-156-4-201202210-00007.
  • 317 Ha V, Sievenpiper JL, de Souza RJ. et al. Effect of fructose on blood pressure: a systematic review and meta-analysis of controlled feeding trials. Hypertension 2012; 59: 787-795 DOI: 10.1161/HYPERTENSIO-NAHA.111.182311.
  • 318 Chiavaroli L, de Souza RJ, Ha V. et al. Effect of Fructose on Established Lipid Targets: A Systematic Review and Meta-Analysis of Controlled Feeding Trials. J Am Heart Assoc 2015; 4: e001700 DOI: 10.1161/JAHA.114.001700.
  • 319 Wang X, Ouyang Y, Liu J. et al. Fruit and vegetable consumption and mortality from all causes, cardiovascular disease, and cancer: systematic review and dose-response meta-analysis of prospective cohort studies. BMJ 2014; 349: g4490 DOI: 10.1136/bmj.g4490.
  • 320 Chiu S, Sievenpiper JL, de Souza RJ. et al. Effect of fructose on markers of non-alcoholic fatty liver disease (NAFLD): a systematic review and meta-analysis of controlled feeding trials. Eur J Clin Nutr 2014; 68: 416-423 DOI: 10.1038/ejcn.2014.8.
  • 321 Wang DD, Sievenpiper JL, de Souza RJ. et al. The effects of fructose intake on serum uric acid vary among controlled dietary trials. J Nutr 2012; 142: 916-923 DOI: 10.3945/jn.111.151951.
  • 322 Cozma AI, Sievenpiper JL, de Souza RJ. et al. Effect of fructose on glycemic control in diabetes: a systematic review and meta-analysis of controlled feeding trials. Diabetes care 2012; 35: 1611-1620 DOI: 10.2337/dc12-0073.
  • 323 Sievenpiper JL, Chiavaroli L, de Souza RJ. et al. ‘Catalytic’ doses of fructose may benefit glycaemic control without harming cardiometabolic risk factors: a small meta-analysis of randomised controlled feeding trials. Br J Nutr 2012; 108: 418-423 DOI: 10.1017/S000711451200013X.
  • 324 Sievenpiper JL, Carleton AJ, Chatha S. et al. Heterogeneous effects of fructose on blood lipids in individuals with type 2 diabetes: systematic review and meta-analysis of experimental trials in humans. Diabetes care 2009; 32: 1930-1937 DOI: 10.2337/dc09-0619.
  • 325 Chung M, Ma J, Patel K. et al. Fructose, high-fructose corn syrup, sucrose, and nonalcoholic fatty liver disease or indexes of liver health: a systematic review and meta-analysis. Am J Clin Nutr 2014; 100: 833-849 DOI: 10.3945/ajcn.114.086314.
  • 326 Goran MI, Ulijaszek SJ, Ventura EE. High fructose corn syrup and diabetes prevalence: a global perspective. Glob Public Health 2013; 8: 55-64 DOI: 10.1080/17441692.2012.736257.
  • 327 Tsilas CS, de Souza RJ, Mejia SB. et al. Relation of total sugars, fructose and sucrose with incident type 2 diabetes: a systematic review and meta-analysis of prospective cohort studies. CMAJ 2017; 189: E711-E720 DOI: 10.1503/cmaj.160706.
  • 328 David Wang D, Sievenpiper JL, de Souza RJ. et al. Effect of fructose on postprandial triglycerides: a systematic review and meta-analysis of controlled feeding trials. Atherosclerosis 2014; 232: 125-133 DOI: 10.1016/j.atherosclerosis.2013.10.019.
  • 329 Zhang YH, An T, Zhang RC. et al. Very high fructose intake increases serum LDL-cholesterol and total cholesterol: a meta-analysis of controlled feeding trials. J Nutr 2013; 143: 1391-1398 DOI: 10.3945/jn.113.175323.
  • 330 Schwingshackl L, Neuenschwander M, Hoffmann G. et al. Dietary sugars and cardiometabolic risk factors: a network meta-analysis on isocaloric substitution interventions. Am J Clin Nutr 2020; 111: 187-196 DOI: 10.1093/ajcn/nqz273.
  • 331 Weber KS, Simon MC, Strassburger K. et al. Habitual Fructose Intake Relates to Insulin Sensitivity and Fatty Liver Index in Recent-Onset Type 2 Diabetes Patients and Individuals without Diabetes. Nutrients 2018; 10 DOI: 10.3390/nu10060774.
  • 332 ter Horst KW, Schene MR, Holman R. et al. Effect of fructose consumption on insulin sensitivity in nondiabetic subjects: a systematic review and meta-analysis of diet-intervention trials. Am J Clin Nutr 2016; 104: 1562-1576 DOI: 10.3945/ajcn.116.137786.
  • 333 Kulzer B, Albus C, Herpertz S. et al. Psychosoziales und Diabetes. Der Diabetologe 2019; 15: 452-469 DOI: 10.1007/s11428-018-0440-x.
  • 334 Ahmed AT, Karter AJ, Warton EM. et al. The relationship between alcohol consumption and glycemic control among patients with diabetes: the Kaiser Permanente Northern California Diabetes Registry. J Gen Intern Med 2008; 23: 275-282 DOI: 10.1007/s11606-007-0502-z.
  • 335 Bantle AE, Thomas W, Bantle JP. Metabolic effects of alcohol in the form of wine in persons with type 2 diabetes mellitus. Metabolism 2008; 57: 241-245 DOI: 10.1016/j.metabol.2007.09.007.
  • 336 Avogaro A, Beltramello P, Gnudi L. et al. Alcohol intake impairs glucose counterregulation during acute insulin-induced hypoglycemia in IDDM patients. Evidence for a critical role of free fatty acids. Diabetes 1993; 42: 1626-1634
  • 337 Turner BC, Jenkins E, Kerr D. et al. The effect of evening alcohol consumption on next-morning glucose control in type 1 diabetes. Diabetes care 2001; 24: 1888-1893 DOI: 10.2337/diacare.24.11.1888.
  • 338 Richardson T, Weiss M, Thomas P. et al. Day after the night before: influence of evening alcohol on risk of hypoglycemia in patients with type 1 diabetes. Diabetes care 2005; 28: 1801-1802 DOI: 10.2337/diacare.28.7.1801.
  • 339 Pedersen-Bjergaard U, Reubsaet JLE, Nielsen SL. et al. Psychoactive drugs, alcohol, and severe hypoglycemia in insulin-treated diabetes: analysis of 141 cases. Am J Med 2005; 118: 307-310 DOI: 10.1016/j.amjmed.2004.07.054.
  • 340 Frier B, Fisher M. Hrsg Moderators, monitoring and management of hypoglycaemia [101-120]. Chichester: John Wiley & Sons; 2007
  • 341 Ahmed AT, Karter AJ, Liu J. Alcohol consumption is inversely associated with adherence to diabetes self-care behaviours. Diabetic medicine: a journal of the British Diabetic Association 2006; 23: 795-802 DOI: 10.1111/j.1464-5491.2006.01878.x.
  • 342 Nahas R, Goguen J. Natural health products. Can J Diabetes 2013; 37: S97-S99 DOI: 10.1016/j.jcjd.2013.01.029.
  • 343 Hartweg J, Perera R, Montori V. et al. Omega-3 polyunsaturated fatty acids (PUFA) for type 2 diabetes mellitus. Cochrane Database Syst Rev 2008; DOI: 10.1002/14651858.CD003205.pub2.
  • 344 Hartweg J, Farmer AJ, Holman RR. et al. Potenzial impact of omega-3 treatment on cardiovascular disease in type 2 diabetes. Curr Opin Lipidol 2009; 20: 30-38 DOI: 10.1097/mol.0b013e328321b3be.
  • 345 O’Mahoney LL, Matu J, Price OJ. et al. Omega-3 polyunsaturated fatty acids favourably modulate cardiometabolic biomarkers in type 2 diabetes: a meta-analysis and meta-regression of randomized controlled trials. Cardiovasc Diabetol 2018; 17: 98 DOI: 10.1186/s12933-018-0740-x.
  • 346 Mirhosseini N, Vatanparast H, Mazidi M. et al. The Effect of Improved Serum 25-Hydroxyvitamin D Status on Glycemic Control in Diabetic Patients: A Meta-Analysis. J Clin Endocrinol Metab 2017; 102: 3097-3110 DOI: 10.1210/jc.2017-01024.
  • 347 Li X, Liu Y, Zheng Y. et al. The Effect of Vitamin D Supplementation on Glycemic Control in Type 2 Diabetes Patients: A Systematic Review and Meta-Analysis. Nutrients 2018; 10 DOI: 10.3390/nu10030375.
  • 348 Jafari T, Fallah AA, Barani A. Effects of vitamin D on serum lipid profile in patients with type 2 diabetes: A meta-analysis of randomized controlled trials. Clin Nutr 2016; 35: 1259-1268 DOI: 10.1016/j.clnu.2016.03.001.
  • 349 Mousa A, Naderpoor N, Teede H. et al. Vitamin D supplementation for improvement of chronic low-grade inflammation in patients with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 2018; 76: 380-394 DOI: 10.1093/nutrit/nux077.
  • 350 Lee KJ, Lee YJ. Effects of vitamin D on blood pressure in patients with type 2 diabetes mellitus. Int J Clin Pharmacol Ther 2016; 54: 233-242 DOI: 10.5414/CP202493.
  • 351 Yu Y, Tian L, Xiao Y. et al. Effect of Vitamin D Supplementation on Some Inflammatory Biomarkers in Type 2 Diabetes Mellitus Subjects: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Ann Nutr Metab 2018; 73: 62-73 DOI: 10.1159/000490358.
  • 352 Verma H, Garg R. Effect of magnesium supplementation on type 2 diabetes associated cardiovascular risk factors: a systematic review and meta-analysis. J Hum Nutr Diet 2017; 30: 621-633 DOI: 10.1111/jhn.12454.
  • 353 Simental-Mendía LE, Sahebkar A, Rodríguez-Morán M. et al. A systematic review and meta-analysis of randomized controlled trials on the effects of magnesium supplementation on insulin sensitivity and glucose control. Pharmacol Res 2016; 111: 272-282 DOI: 10.1016/j.phrs.2016.06.019.
  • 354 Asbaghi O, Hosseini R, Boozari B. et al. The Effects of Magnesium Supplementation on Blood Pressure and Obesity Measure Among Type 2 Diabetes Patient: a Systematic Review and Meta-analysis of Randomized Controlled Trials. Biol Trace Elem Res 2021; 199: 413-424 DOI: 10.1007/s12011-020-02157-0.
  • 355 Vincent JB. Elucidating a biological role for chromium at a molecular level. Acc Chem Res 2000; 33: 503-510 DOI: 10.1021/ar990073r.
  • 356 Asbaghi O, Fatemeh N, Mahnaz RK. et al. Effects of chromium supplementation on glycemic control in patients with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res 2020; 161: 105098 DOI: 10.1016/j.phrs.2020.105098.
  • 357 Yin RV, Phung OJ. Effect of chromium supplementation on glycated hemoglobin and fasting plasma glucose in patients with diabetes mellitus. Nutr J 2015; 14: 14 DOI: 10.1186/1475-2891-14-14.
  • 358 Suksomboon N, Poolsup N, Yuwanakorn A. Systematic review and meta-analysis of the efficacy and safety of chromium supplementation in diabetes. J Clin Pharm Ther 2014; 39: 292-306 DOI: 10.1111/jcpt.12147.
  • 359 Chimienti F. Zinc, pancreatic islet cell function and diabetes: new insights into an old story. Nutr Res Rev 2013; 26: 1-11 DOI: 10.1017/S0954422412000212.
  • 360 de Carvalho GB. Zinc’s role in the glycemic control of patients with type 2 diabetes: a systematic review. BioMetals 2017; 1-12 DOI: 10.1007/s10534-017-9996-y.
  • 361 Fernández-Cao JC, Warthon-Medina M, Hall Moran V. et al. Dietary zinc intake and whole blood zinc concentration in subjects with type 2 diabetes versus healthy subjects: A systematic review, meta-analysis and meta-regression. J Trace Elem Med Biol 2018; 49: 241-251 DOI: 10.1016/j.jtemb.2018.02.008.
  • 362 Wang X, Wu W, Zheng W. et al. Zinc supplementation improves glycemic control for diabetes prevention and management: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2019; 110: 76-90 DOI: 10.1093/ajcn/nqz041.
  • 363 Asbaghi O, Sadeghian M, Fouladvand F. et al. Effects of zinc supplementation on lipid profile in patients with type 2 diabetes mellitus: A systematic review and meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis 2020; 30: 1260-1271 DOI: 10.1016/j.numecd.2020.03.021.
  • 364 Rahimi R, Nikfar S, Larijani B. et al. A review on the role of antioxidants in the management of diabetes and its complications. Biomed Pharmacother 2005; 59: 365-373 DOI: 10.1016/j.biopha.2005.07.002.
  • 365 Ashor AW, Werner AD, Lara J. et al. Effects of vitamin C supplementation on glycaemic control: a systematic review and meta-analysis of randomised controlled trials. Eur J Clin Nutr 2017; 71: 1371-1380 DOI: 10.1038/ejcn.2017.24.
  • 366 Xu R, Zhang S, Tao A. et al. Influence of vitamin E supplementation on glycaemic control: a meta-analysis of randomised controlled trials. PLoS One 2014; 9: e95008 DOI: 10.1371/journal.pone.0095008.
  • 367 Khodaeian M, Tabatabaei-Malazy O, Qorbani M. et al. Effect of vitamins C and E on insulin resistance in diabetes: a meta-analysis study. Eur J Clin Invest 2015; 45: 1161-1174 DOI: 10.1111/eci.12534.
  • 368 Montero D, Walther G, Stehouwer CDA. et al. Effect of antioxidant vitamin supplementation on endothelial function in type 2 diabetes mellitus: a systematic review and meta-analysis of randomized controlled trials. Obes Rev 2014; 15: 107-116 DOI: 10.1111/obr.12114.
  • 369 Tabatabaei-Malazy O, Ardeshirlarijani E, Namazi N. et al. Dietary antioxidative supplements and diabetic retinopathy; a systematic review. J Diabetes Metab Disord 2019; 18: 705-716 DOI: 10.1007/s40200-019-00434-x.
  • 370 Jeyaraman MM, Al-Yousif NSH, Singh Mann A. et al. Resveratrol for adults with type 2 diabetes mellitus. Cochrane Database Syst Rev 2020; 1: CD011919 DOI: 10.1002/14651858.CD011919.pub2.
  • 371 Palma-Duran SA, Vlassopoulos A, Lean M. et al. Nutritional intervention and impact of polyphenol on glycohemoglobin (HbA1c) in non-diabetic and type 2 diabetic subjects: Systematic review and meta-analysis. Crit Rev Food Sci Nutr 2017; 57: 975-986 DOI: 10.1080/10408398.2014.973932.
  • 372 Fogacci F, Tocci G, Presta V. et al. Effect of resveratrol on blood pressure: A systematic review and meta-analysis of randomized, controlled, clinical trials. Crit Rev Food Sci Nutr 2019; 59: 1605-1618 DOI: 10.1080/10408398.2017.1422480.
  • 373 Drzikova B. Haferprodukte mit modifiziertem Gehalt an β-Glucanen und resistenter Stärke und ihre Effekte auf den Gastrointestinaltrakt unter In-vitro- und In-vivo-Bedingungen. 2005 http://opus.kobv.de/ubp/volltexte/205/592/
  • 374 He L, Zhao J, Huang Y. et al. The difference between oats and beta-glucan extract intake in the management of HbA1c, fasting glucose and insulin sensitivity: ameta-analysis of randomized controlled trials. Food Funct 2016; 7: 1413-1428 DOI: 10.1039/c5fo01364j.
  • 375 Abbasi NN, Purslow PP, Tosh SM. et al. Oat β-glucan depresses SGLT1- and GLUT2-mediated glucose transport in intestinal epithelial cells (IEC-6). Nutr Res 2016; 36: 541-552 DOI: 10.1016/j.nutres.2016.02.004.
  • 376 Wang F, Yu G, Zhang Y. et al. Dipeptidyl Peptidase IV Inhibitory Peptides Derived from Oat (Avena sativa L.), Buckwheat (Fagopyrum esculentum), and Highland Barley (Hordeum vulgare trifurcatum (L.) Trofim) Proteins. J Agric Food Chem 2015; 63: 9543-9549 DOI: 10.1021/acs.jafc.5b04016.
  • 377 Liu M, Zhang Y, Zhang H. et al. The anti-diabetic activity of oat β-d-glucan in streptozotocin-nicotinamide induced diabetic mice. Int J Biol Macromol 2016; 91: 1170-1176 DOI: 10.1016/j.ijbiomac.2016.06.083.
  • 378 Lammert A, Kratzsch J, Selhorst J. et al. Clinical benefit of a short term dietary oatmeal intervention in patients with type 2 diabetes and severe insulin resistance: a pilot study. Exp Clin Endocrinol Diabetes 2008; 116: 132-134 DOI: 10.1055/s-2007-984456.
  • 379 Delgado G, Kleber ME, Krämer BK. et al. Dietary Intervention with Oatmeal in Patients with uncontrolled Type 2 Diabetes Mellitus – A Crossover Study. Exp Clin Endocrinol Diabetes 2019; 127: 623-629 DOI: 10.1055/a-0677-6068.
  • 380 Delgado GE, Krämer BK, Scharnagl H. et al. Bile Acids in Patients with Uncontrolled Type 2 Diabetes Mellitus – The Effect of Two Days of Oatmeal Treatment. Exp Clin Endocrinol Diabetes 2020; 128: 624-630 DOI: 10.1055/a-1069-7330.
  • 381 Behall KM, Scholfield DJ, Hallfrisch J. Comparison of hormone and glucose responses of overweight women to barley and oats. J Am Coll Nutr 2005; 24: 182-188 DOI: 10.1080/07315724.2005.10719464.
  • 382 Braaten JT, Scott FW, Wood PJ. et al. High beta-glucan oat bran and oat gum reduce postprandial blood glucose and insulin in subjects with and without type 2 diabetes. Diabetic medicine: a journal of the British Diabetic Association 1994; 11: 312-318 DOI: 10.1111/j.1464-5491.1994.tb00277.x.
  • 383 Pick ME, Hawrysh ZJ, Gee MI. et al. Oat bran concentrate bread products improve long-term control of diabetes: a pilot study. J Am Diet Assoc 1996; 96: 1254-1261 DOI: 10.1016/S0002-8223(96)00329-X.
  • 384 Tapola N, Karvonen H, Niskanen L. et al. Glycemic responses of oat bran products in type 2 diabetic patients. Nutr Metab Cardiovasc Dis 2005; 15: 255-261 DOI: 10.1016/j.numecd.2004.09.003.
  • 385 Tappy L, Gügolz E, Würsch P. Effects of breakfast cereals containing various amounts of beta-glucan fibers on plasma glucose and insulin responses in NIDDM subjects. Diabetes care 1996; 19: 831-834 DOI: 10.2337/diacare.19.8.831.
  • 386 Wood PJ, Beer MU, Butler G. Evaluation of role of concentration and molecular weight of oat beta-glucan in determining effect of viscosity on plasma glucose and insulin following an oral glucose load. Br J Nutr 2000; 84: 19-23
  • 387 [Anonym]. Scientific Opinion on the substantiation of health claims related to beta glucans and maintenance or achievement of normal blood glucose concentrations (ID 756, 802, 2935) pursuant to Article 13(1) of Regulation (EC) No 1924/2006. EFS2 2010; 8 DOI: 10.2903/j.efsa.2010.1482.
  • 388 Amtsblatt der Europäischen Union 2011 L 136/1 vom 25.5.2012 https://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2012:136:0001:0040:DE:PDFStand: 04.07.2021
  • 389 Zurbau A, Noronha JC, Khan TA. et al. The effect of oat β-glucan on postprandial blood glucose and insulin responses: a systematic review and meta-analysis. Eur J Clin Nutr 2021; DOI: 10.1038/s41430-021-00875-9.
  • 390 Battilana P, Ornstein K, Minehira K. et al. Mechanisms of action of betaglucan in postprandial glucose metabolism in healthy men. Eur J Clin Nutr 2001; 55: 327-333 DOI: 10.1038/sj.ejcn.1601160.
  • 391 Jenkins AL, Jenkins DJA, Zdravkovic U. et al. Depression of the glycemic index by high levels of beta-glucan fiber in two functional foods tested in type 2 diabetes. Eur J Clin Nutr 2002; 56: 622-628 DOI: 10.1038/sj.ejcn.1601367.

Correspondence

Prof. Dr. Diana Rubin
Center for Nutritional Medicine
Vivantes Hospital Spandau
Neue Bergstraße 6
13585 Berlin
Germany   

Publication History

Article published online:
31 March 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Beck J, Greenwood DA, Blanton L. et al. 2017 National Standards for Diabetes Self-Management Education and Support. Diabetes care 2017; 40: 1409-1419 DOI: 10.2337/dci17-0025.
  • 2 Evert AB, Dennison M, Gardner CD. et al. Nutrition Therapy for Adults With Diabetes or Prediabetes: A Consensus Report. Diabetes care 2019; 42: 731-754 DOI: 10.2337/dci19-0014.
  • 3 https://www.deutsche-diabetes-gesellschaft.de/politik/stellungnahmen/stellungnahme-des-ausschuss-ernaehrung-der-ddg-zum-consensusreport-nutrition-therapy-for-adults-with-diabetes-or-prediabetes Ausschuss Ernährung der DDG. Stellungnahme des Ausschuss Ernährung der DDG zum Consensus Report: Nutrition Therapy for Adults with Diabetes or Prediabetes: [Evert AB et al. Diabetes Care 2019;42:731-54.] 2019
  • 4 DeFronzo RA, Bonadonna RC, Ferrannini E. Pathogenesis of NIDDM. A balanced overview. Diabetes care 1992; 15: 318-368 DOI: 10.2337/diacare.15.3.318.
  • 5 DeFronzo RA, Eldor R, Abdul-Ghani M. Pathophysiologic approach to therapy in patients with newly diagnosed type 2 diabetes. Diabetes care 2013; 36: S127-S138 DOI: 10.2337/dcS13-2011.
  • 6 Lencioni C, Lupi R, Del Prato S. Beta-cell failure in type 2 diabetes mellitus. Curr Diab Rep 2008; 8: 179-184 DOI: 10.1007/s11892-008-0031-0.
  • 7 [Anonymous]. U.K. prospective diabetes study 16. Overview of 6 years’ therapy of type II diabetes: a progressive disease. U.K. Prospective Diabetes Study Group. Diabetes 1995; 44: 1249-1258
  • 8 Zaharia OP, Strassburger K, Strom A. et al. Risk of diabetes-associated diseases in subgroups of patients with recent-onset diabetes: a 5-year follow-up study. Lancet Diabetes Endocrinol 2019; 7: 684-694 DOI: 10.1016/S2213-8587(19)30187-1.
  • 9 Kodama S, Horikawa C, Fujihara K. et al. Quantitative relationship between body weight gain in adulthood and incident type 2 diabetes: a meta-analysis. Obes Rev 2014; 15: 202-214 DOI: 10.1111/obr.12129.
  • 10 Wing RR, Lang W, Wadden TA. et al. Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes care 2011; 34: 1481-1486 DOI: 10.2337/dc10-2415.
  • 11 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: 808-815 DOI: 10.2337/dc15-1942.
  • 12 Jazet IM, Pijl H, Frölich M. et al. Factors predicting the blood glucose lowering effect of a 30-day very low calorie diet in obese Type 2 diabetic patients. Diabetic medicine: a journal of the British Diabetic Association 2005; 22: 52-55 DOI: 10.1111/j.1464-5491.2004.01357.x.
  • 13 Lean MEJ, Leslie WS, Barnes AC. et al. Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, clusterrandomised trial. The Lancet 2018; 391: 541-551 DOI: 10.1016/S0140-6736(17)33102-1.
  • 14 Bangalore S, Fayyad R, DeMicco DA. et al. Body Weight Variability and Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus. Circ Cardiovasc Qual Outcomes 2018; 11: e004724 DOI: 10.1161/CIRCOUT-COMES.118.004724.
  • 15 Yeboah P, Hsu FC, Bertoni AG. et al. Body Mass Index, Change in Weight, Body Weight Variability and Outcomes in Type 2 Diabetes Mellitus (from the ACCORD Trial). The American journal of cardiology 2019; 123: 576-581 DOI: 10.1016/j.amjcard.2018.11.016.
  • 16 Pagidipati NJ, Zheng Y, Green JB. et al. Association of obesity with cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease: Insights from TECOS. Am Heart J 2020; 219: 47-57 DOI: 10.1016/j.ahj.2019.09.016.
  • 17 Bodegard J, Sundström J, Svennblad B. et al. Changes in body mass index following newly diagnosed type 2 diabetes and risk of cardiovascular mortality: a cohort study of 8486 primary-care patients. Diabetes Metab 2013; 39: 306-313 DOI: 10.1016/j.diabet.2013.05.004.
  • 18 Weinheimer EM, Sands LP, Campbell WW. A systematic review of the separate and combined effects of energy restriction and exercise on fatfree mass in middle-aged and older adults: implications for sarcopenic obesity. Nutr Rev 2010; 68: 375-388 DOI: 10.1111/j.1753-4887.2010.00298.x.
  • 19 Zaccardi F, Dhalwani NN, Papamargaritis D. et al. Nonlinear association of BMI with all-cause and cardiovascular mortality in type 2 diabetes mellitus: a systematic review and meta-analysis of 414587 participants in prospective studies. Diabetologia 2017; 60: 240-248 DOI: 10.1007/s00125-016-4162-6.
  • 20 Salehidoost R, Mansouri A, Amini M. et al. Body mass index and the all-cause mortality rate in patients with type 2 diabetes mellitus. Acta Diabetol 2018; 55: 569-577 DOI: 10.1007/s00592-018-1126-y.
  • 21 Hainer V, Aldhoon-Hainerová I. Obesity paradox does exist. Diabetes care 2013; 36: S276-S281 DOI: 10.2337/dcS13-2023.
  • 22 Murphy RA, Reinders I, Garcia ME. et al. Adipose tissue, muscle, and function: potential mediators of associations between body weight and mortality in older adults with type 2 diabetes. Diabetes care 2014; 37: 3213-3219 DOI: 10.2337/dc14-0293.
  • 23 Bales CW, Porter Starr KN. Obesity Interventions for Older Adults: Diet as a Determinant of Physical Function. Adv Nutr 2018; 9: 151-159 DOI: 10.1093/advances/nmx016.
  • 24 Uusitupa M, Khan TA, Viguiliouk E. et al. Prevention of Type 2 Diabetes by Lifestyle Changes: A Systematic Review and Meta-Analysis. Nutrients 2019; 11 DOI: 10.3390/nu11112611.
  • 25 Raben A, Vestentoft PS, Brand-Miller J. et al. The PREVIEW intervention study: Results from a 3-year randomized 2 × 2 factorial multinational trial investigating the role of protein, glycaemic index and physical activity for prevention of type 2 diabetes. Diabetes Obes Metab 2021; 23: 324-337 DOI: 10.1111/dom.14219.
  • 26 Gregg EW, Chen H, Wagenknecht LE. et al. Association of an intensive lifestyle intervention with remission of type 2 diabetes. JAMA 2012; 308: 2489-2496 DOI: 10.1001/jama.2012.67929.
  • 27 Anderson JW, Konz EC, Frederich RC. et al. Long-term weight-loss maintenance: a meta-analysis of US studies. Am J Clin Nutr 2001; 74: 579-584 DOI: 10.1093/ajcn/74.5.579.
  • 28 Bundesgesundheitsministerium 2015. Telemedizin. Im Internet (Stand: 09.04.2021) https://www.bundesgesundheitsministerium.de/service/begriffe-von-a-z/t/telemedizin.html
  • 29 Su D, McBride C, Zhou J. et al. Does nutritional counseling in telemedicine improve treatment outcomes for diabetes? A systematic review and meta-analysis of results from 92 studies. J Telemed Telecare 2016; 22: 333-347 DOI: 10.1177/1357633X15608297.
  • 30 Kempf K, Altpeter B, Berger J. et al. Efficacy of the Telemedical Lifestyle intervention Program TeLiPro in Advanced Stages of Type 2 Diabetes: A Randomized Controlled Trial. Diabetes care 2017; 40: 863-871 DOI: 10.2337/dc17-0303.
  • 31 Belalcazar LM, Haffner SM, Lang W. et al. Lifestyle intervention and/or statins for the reduction of C-reactive protein in type 2 diabetes: from the look AHEAD study. Obesity (Silver Spring) 2013; 21: 944-950 DOI: 10.1002/oby.20431.
  • 32 Colquitt JL, Pickett K, Loveman E. et al. Surgery for weight loss in adults. Cochrane Database Syst Rev 2014; DOI: 10.1002/14651858.CD003641.pub4.
  • 33 Patel KV, Bahnson JL, Gaussoin SA. et al. Association of Baseline and Longitudinal Changes in Body Composition Measures With Risk of Heart Failure and Myocardial Infarction in Type 2 Diabetes: Findings From the Look AHEAD Trial. Circulation 2020; 142: 2420-2430 DOI: 10.1161/CIRCULATIONAHA.120.050941.
  • 34 Franz MJ, Boucher JL, Rutten-Ramos S. et al. Lifestyle weight-loss intervention outcomes in overweight and obese adults with type 2 diabetes: a systematic review and meta-analysis of randomized clinical trials. J Acad Nutr Diet 2015; 115: 1447-1463 DOI: 10.1016/j.jand.2015.02.031.
  • 35 Murgatroyd PR, Goldberg GR, Leahy FE. et al. Effects of inactivity and diet composition on human energy balance. Int J Obes Relat Metab Disord 1999; 23: 1269-1275 DOI: 10.1038/sj.ijo.0801062.
  • 36 Stubbs RJ, Sepp A, Hughes DA. et al. The effect of graded levels of exercise on energy intake and balance in free-living women. Int J Obes Relat Metab Disord 2002; 26: 866-869 DOI: 10.1038/sj.ijo.0801874.
  • 37 Granados K, Stephens BR, Malin SK. et al. Appetite regulation in response to sitting and energy imbalance. Appl Physiol Nutr Metab 2012; 37: 323-333 DOI: 10.1139/h2012-002.
  • 38 Hägele FA, Büsing F, Nas A. et al. Appetite Control Is Improved by Acute Increases in Energy Turnover at Different Levels of Energy Balance. J Clin Endocrinol Metab 2019; 104: 4481-4491 DOI: 10.1210/jc.2019-01164.
  • 39 Douglas JA, King JA, Clayton DJ. et al. Acute effects of exercise on appetite, ad libitum energy intake and appetite-regulatory hormones in lean and overweight/obese men and women. Int J Obes (Lond) 2017; 41: 1737-1744 DOI: 10.1038/ijo.2017.181.
  • 40 Savikj M, Zierath JR. Train like an athlete: applying exercise interventions to manage type 2 diabetes. Diabetologia 2020; 63: 1491-1499 DOI: 10.1007/s00125-020-05166-9.
  • 41 Büsing F, Hägele FA, Nas A. et al. Impact of energy turnover on the regulation of glucose homeostasis in healthy subjects. Nutr Diabetes 2019; 9: 22 DOI: 10.1038/s41387-019-0089-6.
  • 42 Larsen JJ, Dela F, Kjaer M. et al. The effect of moderate exercise on postprandial glucose homeostasis in NIDDM patients. Diabetologia 1997; 40: 447-453 DOI: 10.1007/s001250050699.
  • 43 Heden TD, Winn NC, Mari A. et al. Postdinner resistance exercise improves postprandial risk factors more effectively than predinner resistance exercise in patients with type 2 diabetes. J Appl Physiol (1985) 2015; 118: 624-634 DOI: 10.1152/japplphysiol.00917.2014.
  • 44 Reynolds AN, Mann JI, Williams S. et al. Advice to walk after meals is more effective for lowering postprandial glycaemia in type 2 diabetes mellitus than advice that does not specify timing: a randomised crossover study. Diabetologia 2016; 59: 2572-2578 DOI: 10.1007/s00125-016-4085-2.
  • 45 Gaudet-Savard T, Ferland A, Broderick TL. et al. Safety and magnitude of changes in blood glucose levels following exercise performed in the fasted and the postprandial state in men with type 2 diabetes. Eur J Cardiovasc Prev Rehabil 2007; 14: 831-836 DOI: 10.1097/HJR.0b013e3282efaf38.
  • 46 DiPietro L, Gribok A, Stevens MS. et al. Three 15-min bouts of moderate postmeal walking significantly improves 24-h glycemic control in older people at risk for impaired glucose tolerance. Diabetes care 2013; 36: 3262-3268 DOI: 10.2337/dc13-0084.
  • 47 Seidelmann SB, Claggett B, Cheng S. et al. Dietary carbohydrate intake and mortality: a prospective cohort study and meta-analysis. Lancet Public Health 2018; 3: e419-e428 DOI: 10.1016/S2468-2667(18)30135-X.
  • 48 Davies MJ, D’Alessio DA, Fradkin J. et al. Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes care 2018; 41: 2669-2701 DOI: 10.2337/dci18-0033.
  • 49 Schwingshackl L, Chaimani A, Hoffmann G. et al. A network meta-analysis on the comparative efficacy of different dietary approaches on glycaemic control in patients with type 2 diabetes mellitus. Eur J Epidemiol 2018; 33: 157-170 DOI: 10.1007/s10654-017-0352-x.
  • 50 Schwingshackl L, Hoffmann G, Iqbal K. et al. Food groups and intermediate disease markers: a systematic review and network meta-analysis of randomized trials. Am J Clin Nutr 2018; 108: 576-586 DOI: 10.1093/ajcn/nqy151.
  • 51 Neuenschwander M, Ballon A, Weber KS. et al. Role of diet in type 2 diabetes incidence: umbrella review of meta-analyses of prospective observational studies. BMJ 2019; 366: l2368 DOI: 10.1136/bmj.l2368.
  • 52 Ge L, Sadeghirad B, Ball GDC. et al. Comparison of dietary macronutrient patterns of 14 popular named dietary programmes for weight and cardiovascular risk factor reduction in adults: systematic review and network meta-analysis of randomised trials. BMJ 2020; 369: m696 DOI: 10.1136/bmj.m696.
  • 53 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 DOI: 10.1136/bmj.m4743.
  • 54 Schwingshackl L, Nitschke K, Zähringer J. et al. Impact of Meal Frequency on Anthropometric Outcomes: A Systematic Review and Network Meta-Analysis of Randomized Controlled Trials. Adv Nutr 2020; 11: 1108-1122 DOI: 10.1093/advances/nmaa056.
  • 55 Della Corte KW, Perrar I, Penczynski KJ. et al. Effect of Dietary Sugar Intake on Biomarkers of Subclinical Inflammation: A Systematic Review and Meta-Analysis of Intervention Studies. Nutrients 2018; 10 DOI: 10.3390/nu10050606.
  • 56 Schwingshackl L, Chaimani A, Schwedhelm C. et al. Comparative effects of different dietary approaches on blood pressure in hypertensive and pre-hypertensive patients: A systematic review and network meta-analysis. Crit Rev Food Sci Nutr 2019; 59: 2674-2687 DOI: 10.1080/10408398.2018.1463967.
  • 57 Thom G, Messow CM, Leslie WS. et al. Predictors of type 2 diabetes remission in the Diabetes Remission Clinical Trial (DiRECT). Diabetic medicine: a journal of the British Diabetic Association 2020; e14395 DOI: 10.1111/dme.14395.
  • 58 de Souza RJ, Mente A, Maroleanu A. et al. Intake of saturated and trans unsaturated fatty acids and risk of all cause mortality, cardiovascular disease, and type 2 diabetes: systematic review and meta-analysis of observational studies. BMJ 2015; 351: h3978 DOI: 10.1136/bmj.h3978.
  • 59 Astrup A, Magkos F, Bier DM. et al. Saturated Fats and Health: A Reassessment and Proposal for Food-Based Recommendations: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 76: 844-857 DOI: 10.1016/j.jacc.2020.05.077.
  • 60 Pimpin L, Wu JHY, Haskelberg H. et al. Is Butter Back? A Systematic Review and Meta-Analysis of Butter Consumption and Risk of Cardiovascular Disease, Diabetes, and Total Mortality. PLoS One 2016; 11: e0158118 DOI: 10.1371/journal.pone.0158118.
  • 61 Benatar JR, Sidhu K, Stewart RAH. Effects of high and low fat dairy food on cardio-metabolic risk factors: a meta-analysis of randomized studies. PLoS One 2013; 8: e76480 DOI: 10.1371/journal.pone.0076480.
  • 62 Hooper L, Abdelhamid AS, Jimoh OF. et al. Effects of total fat intake on body fatness in adults. Cochrane Database Syst Rev 2020; 6: CD013636 DOI: 10.1002/14651858.CD013636.
  • 63 Hooper L, Martin N, Jimoh OF. et al. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst Rev 2020; 8: CD011737 DOI: 10.1002/14651858.CD011737.pub3.
  • 64 Belalcazar LM, Reboussin DM, Haffner SM. et al. A 1-year lifestyle intervention for weight loss in individuals with type 2 diabetes reduces high C-reactive protein levels and identifies metabolic predictors of change: from the Look AHEAD (Action for Health in Diabetes) study. Diabetes care 2010; 33: 2297-2303 DOI: 10.2337/dc10-0728.
  • 65 Lu M, Wan Y, Yang B. et al. Effects of low-fat compared with high-fat diet on cardiometabolic indicators in people with overweight and obesity without overt metabolic disturbance: a systematic review and metaanalysis of randomised controlled trials. Br J Nutr 2018; 119: 96-108 DOI: 10.1017/S0007114517002902.
  • 66 Wu JHY, Marklund M, Imamura F. et al. Omega-6 fatty acid biomarkers and incident type 2 diabetes: pooled analysis of individual-level data for 39 740 adults from 20 prospective cohort studies. Lancet Diabetes Endocrinol 2017; 5: 965-974 DOI: 10.1016/S2213-8587(17)30307-8.
  • 67 Li J, Guasch-Ferré M, Li Y. et al. Dietary intake and biomarkers of linoleic acid and mortality: systematic review and meta-analysis of prospective cohort studies. Am J Clin Nutr 2020; 112: 150-167 DOI: 10.1093/ajcn/nqz349.
  • 68 Pan A, Chen M, Chowdhury R. et al. α-Linolenic acid and risk of cardiovascular disease: a systematic review and meta-analysis. Am J Clin Nutr 2012; 96: 1262-1273 DOI: 10.3945/ajcn.112.044040.
  • 69 Abdelhamid AS, Martin N, Bridges C. et al. Polyunsaturated fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11: CD012345 DOI: 10.1002/14651858.CD012345.pub3.
  • 70 Abdelhamid AS, Brown TJ, Brainard JS. et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2020; 3: CD003177 DOI: 10.1002/14651858.CD003177.pub5.
  • 71 Brown TJ, Brainard J, Song F. et al. Omega-3, omega-6, and total dietary polyunsaturated fat for prevention and treatment of type 2 diabetes mellitus: systematic review and meta-analysis of randomised controlled trials. BMJ 2019; 366: l4697 DOI: 10.1136/bmj.l4697.
  • 72 Qian F, Korat AA, Malik V. et al. Metabolic Effects of Monounsaturated Fatty Acid-Enriched Diets Compared With Carbohydrate or Polyunsaturated Fatty Acid-Enriched Diets in Patients With Type 2 Diabetes: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Diabetes care 2016; 39: 1448-1457 DOI: 10.2337/dc16-0513.
  • 73 Jovanovski E, de Castro Ruiz Marques A, Li D. et al. Effect of high-carbohydrate or high-monounsaturated fatty acid diets on blood pressure: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 2019; 77: 19-31 DOI: 10.1093/nutrit/nuy040.
  • 74 Zhang YY, Liu W, Zhao TY. et al. Efficacy of Omega-3 Polyunsaturated Fatty Acids Supplementation in Managing Overweight and Obesity: A Meta-Analysis of Randomized Clinical Trials. J Nutr Health Aging 2017; 21: 187-192 DOI: 10.1007/s12603-016-0755-5.
  • 75 Lin N, Shi JJ, Li YM. et al What is the impact of n-3 PUFAs on inflammation markers in Type 2 diabetic mellitus populations?: a systematic review and meta-analysis of randomized controlled trials. Lipids Health Dis 2016; 15: 133 DOI: 10.1186/s12944-016-0303-7.
  • 76 Reis CEG, Landim KC, Nunes ACS. et al. Safety in the hypertriglyceridemia treatment with N-3 polyunsaturated fatty acids on glucose metabolism in subjects with type 2 diabetes mellitus. Nutr Hosp 2014; 31: 570-576 DOI: 10.3305/nh.2015.31.2.7845.
  • 77 Gao L, Cao J, Mao Q. et al. Influence of omega-3 polyunsaturated fatty acid-supplementation on platelet aggregation in humans: a meta-analysis of randomized controlled trials. Atherosclerosis 2013; 226: 328-334 DOI: 10.1016/j.atherosclerosis.2012.10.056.
  • 78 He XX, Wu XL, Chen RP. et al. Effectiveness of Omega-3 Polyunsaturated Fatty Acids in Non-Alcoholic Fatty Liver Disease: A Meta-Analysis of Randomized Controlled Trials. PLoS One 2016; 11: e0162368 DOI: 10.1371/journal.pone.0162368.
  • 79 Li N, Yue H, Jia M. et al. Effect of low-ratio n-6/n-3 PUFA on blood glucose: a meta-analysis. Food Funct 2019; 10: 4557-4565 DOI: 10.1039/c9fo00323a.
  • 80 Wanders AJ, Blom WAM, Zock PL. et al. Plant-derived polyunsaturated fatty acids and markers of glucose metabolism and insulin resistance: a meta-analysis of randomized controlled feeding trials. BMJ Open Diabetes Res Care 2019; 7: e000585 DOI: 10.1136/bmjdrc-2018-000585.
  • 81 Abbott KA, Burrows TL, Thota RN. et al. Do ω-3 PUFAs affect insulin resistance in a sex-specific manner? A systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2016; 104: 1470-1484 DOI: 10.3945/ajcn.116.138172.
  • 82 Jovanovski E, Li D, Thanh Ho HV. et al. The effect of alpha-linolenic acid on glycemic control in individuals with type 2 diabetes: A systematic review and meta-analysis of randomized controlled clinical trials. Medicine (Baltimore) 2017; 96: e6531 DOI: 10.1097/MD.0000000000006531.
  • 83 Faris MAI, Jahrami H, BaHammam A. et al. A systematic review, meta-analysis, and meta-regression of the impact of diurnal intermittent fasting during Ramadan on glucometabolic markers in healthy subjects. Diabetes Res Clin Pract 2020; 165: 108226 DOI: 10.1016/j.diabres.2020.108226.
  • 84 Mirmiran P, Bahadoran Z, Gaeini Z. et al. Effects of Ramadan intermittent fasting on lipid and lipoprotein parameters: An updated meta-analysis. Nutr Metab Cardiovasc Dis 2019; 29: 906-915 DOI: 10.1016/j.numecd.2019.05.056.
  • 85 Fernando HA, Zibellini J, Harris RA. et al. Effect of Ramadan Fasting on Weight and Body Composition in Healthy Non-Athlete Adults: A Systematic Review and Meta-Analysis. Nutrients 2019; 11 DOI: 10.3390/nu11020478.
  • 86 Horne BD, May HT, Anderson JL. et al. Usefulness of routine periodic fasting to lower risk of coronary artery disease in patients undergoing coronary angiography. The American journal of cardiology 2008; 102: 814-819 DOI: 10.1016/j.amjcard.2008.05.021.
  • 87 Horne BD, Muhlestein JB, May HT. et al. Relation of routine, periodic fasting to risk of diabetes mellitus, and coronary artery disease in patients undergoing coronary angiography. The American journal of cardiology 2012; 109: 1558-1562 DOI: 10.1016/j.amjcard.2012.01.379.
  • 88 Schwingshackl L, Zähringer J, Nitschke K. et al. Impact of intermittent energy restriction on anthropometric outcomes and intermediate disease markers in patients with overweight and obesity: systematic review and meta-analyses. Crit Rev Food Sci Nutr 2021; 61: 1293-1304 DOI: 10.1080/10408398.2020.1757616.
  • 89 Park J, Seo YG, Paek YJ. et al. Effect of alternate-day fasting on obesity and cardiometabolic risk: A systematic review and meta-analysis. Metabolism 2020; 111: 154336 DOI: 10.1016/j.metabol.2020.154336.
  • 90 Harris L, Hamilton S, Azevedo LB. et al. Intermittent fasting interventions for treatment of overweight and obesity in adults: a systematic review and meta-analysis. JBI Database System Rev Implement Rep 2018; 16: 507-547 DOI: 10.11124/JBISRIR-2016-003248.
  • 91 Seimon RV, Roekenes JA, Zibellini J. et al. Do intermittent diets provide physiological benefits over continuous diets for weight loss? A systematic review of clinical trials. Mol Cell Endocrinol 2015; 418: 153-172 DOI: 10.1016/j.mce.2015.09.014.
  • 92 Horne BD, Muhlestein JB, Anderson JL. Health effects of intermittent fasting: hormesis or harm? A systematic review. Am J Clin Nutr 2015; 102: 464-470 DOI: 10.3945/ajcn.115.109553.
  • 93 Borgundvaag E, Mak J, Kramer CK. Metabolic Impact of Intermittent Fasting in Patients With Type 2 Diabetes Mellitus: A Systematic Review and Meta-analysis of Interventional Studies. J Clin Endocrinol Metab 2021; 106: 902-911 DOI: 10.1210/clinem/dgaa926.
  • 94 Parr EB, Devlin BL, Lim KHC. et al. Time-Restricted Eating as a Nutrition Strategy for Individuals with Type 2 Diabetes: A Feasibility Study. Nutrients 2020; 12 DOI: 10.3390/nu12113228.
  • 95 Carter S, Clifton PM, Keogh JB. The effects of intermittent compared to continuous energy restriction on glycaemic control in type 2 diabetes; a pragmatic pilot trial. Diabetes Res Clin Pract 2016; 122: 106-112 DOI: 10.1016/j.diabres.2016.10.010.
  • 96 Carter S, Clifton PM, Keogh JB. The effect of intermittent compared with continuous energy restriction on glycaemic control in patients with type 2 diabetes: 24-month follow-up of a randomised noninferiority trial. Diabetes Res Clin Pract 2019; 151: 11-19 DOI: 10.1016/j.diabres.2019.03.022.
  • 97 Corley BT, Carroll RW, Hall RM. et al. Intermittent fasting in Type 2 diabetes mellitus and the risk of hypoglycaemia: a randomized controlled trial. Diabetic medicine: a journal of the British Diabetic Association 2018; 35: 588-594 DOI: 10.1111/dme.13595.
  • 98 Henry RR, Wiest-Kent TA, Scheaffer L. et al. Metabolic consequences of very-low-calorie diet therapy in obese non-insulin-dependent diabetic and nondiabetic subjects. Diabetes 1986; 35: 155-164 DOI: 10.2337/diab.35.2.155.
  • 99 Amatruda JM, Richeson JF, Welle SL. et al. The safety and efficacy of a controlled low-energy (‘very-low-calorie’) diet in the treatment of noninsulin-dependent diabetes and obesity. Arch Intern Med 1988; 148: 873-877
  • 100 Rotella CM, Cresci B, Mannucci E. et al. Short cycles of very low calorie diet in the therapy of obese type II diabetes mellitus. J Endocrinol Invest 1994; 17: 171-179 DOI: 10.1007/BF03347709.
  • 101 Dhindsa P, Scott AR, Donnelly R. Metabolic and cardiovascular effects of very-low-calorie diet therapy in obese patients with Type 2 diabetes in secondary failure: outcomes after 1 year. Diabetic medicine: a journal of the British Diabetic Association 2003; 20: 319-324 DOI: 10.1046/j.1464-5491.2003.00937.x.
  • 102 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: 344-355 DOI: 10.1016/S2213-8587(19)30068-3.
  • 103 Maggio CA, Pi-Sunyer FX. Obesity and type 2 diabetes. Endocrinol Metab Clin North Am 2003; 32: 805-822 viii DOI: 10.1016/s0889-8529(03)00071-9.
  • 104 Wolf AM, Colditz GA. Current estimates of the economic cost of obesity in the United States. Obes Res 1998; 6: 97-106 DOI: 10.1002/j.1550-8528.1998.tb00322.x.
  • 105 Colditz GA, Willett WC, Rotnitzky A. et al. Weight gain as a risk factor for clinical diabetes mellitus in women. Ann Intern Med 1995; 122: 481-486 DOI: 10.7326/0003-4819-122-7-199504010-00001.
  • 106 Anderson JW, Kendall CWC, Jenkins DJA. Importance of weight management in type 2 diabetes: review with meta-analysis of clinical studies. J Am Coll Nutr 2003; 22: 331-339 DOI: 10.1080/07315724.2003.10719316.
  • 107 Leslie WS, Taylor R, Harris L. et al. Weight losses with low-energy formula diets in obese patients with and without type 2 diabetes: systematic review and meta-analysis. Int J Obes (Lond) 2017; 41: 96-101 DOI: 10.1038/ijo.2016.175.
  • 108 McCombie L, Brosnahan N, Ross H. et al. Filling the intervention gap: service evaluation of an intensive nonsurgical weight management programme for severe and complex obesity. J Hum Nutr Diet 2019; 32: 329-337 DOI: 10.1111/jhn.12611.
  • 109 Jazet IM, de Craen AJ, van Schie EM. et al. Sustained beneficial metabolic effects 18 months after a 30-day very low calorie diet in severely obese, insulin-treated patients with type 2 diabetes. Diabetes Res Clin Pract 2007; 77: 70-76 DOI: 10.1016/j.diabres.2006.10.019.
  • 110 Kempf K, Schloot NC, Gärtner B. et al. Meal replacement reduces insulin requirement, HbA1c and weight long-term in type 2 diabetes patients with 100 U insulin per day. J Hum Nutr Diet 2014; 27: 21-27 DOI: 10.1111/jhn.12145.
  • 111 Kempf K, Röhling M, Niedermeier K. et al. Individualized Meal Replacement Therapy Improves Clinically Relevant Long-Term Glycemic Control in Poorly Controlled Type 2 Diabetes Patients. Nutrients 2018; 10 DOI: 10.3390/nu10081022.
  • 112 Taylor R, Leslie WS, Barnes AC. et al. Clinical and metabolic features of the randomised controlled Diabetes Remission Clinical Trial (DiRECT) cohort. Diabetologia 2018; 61: 589-598 DOI: 10.1007/s00125-017-4503-0.
  • 113 Halle M, Röhling M, Banzer W. et al. Meal replacement by formula diet reduces weight more than a lifestyle intervention alone in patients with overweight or obesity and accompanied cardiovascular risk factors-the ACOORH trial. Eur J Clin Nutr 2021; 75: 661-669 DOI: 10.1038/s41430-020-00783-4.
  • 114 Röhling M, Kempf K, Banzer W. et al. Prediabetes Conversion to Normoglycemia Is Superior Adding a Low-Carbohydrate and Energy Deficit Formula Diet to Lifestyle Intervention-A 12-Month Subanalysis of the ACOORH Trial. Nutrients 2020; 12 DOI: 10.3390/nu12072022.
  • 115 Holman RR, Paul SK, Bethel MA. et al. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359: 1577-1589 DOI: 10.1056/NEJMoa0806470.
  • 116 Haslacher H, Fallmann H, Waldhäusl C. et al. Type 2 diabetes care: Improvement by standardization at a diabetes rehabilitation clinic. An observational report. PLoS One 2019; 14: e0226132 DOI: 10.1371/journal.pone.0226132.
  • 117 Paul SK, Shaw JE, Montvida O. et al. Weight gain in insulin-treated patients by body mass index category at treatment initiation: new evidence from real-world data in patients with type 2 diabetes. Diabetes Obes Metab 2016; 18: 1244-1252 DOI: 10.1111/dom.12761.
  • 118 [Anonymous]. 5. Facilitating Behavior Change and Well-being to Improve Health Outcomes: Standards of Medical Care in Diabetes-2020. Diabetes care 2020; 43: S48-S65 DOI: 10.2337/dc20-S005.
  • 119 Dyson PA, Twenefour D, Breen C. et al. Diabetes UK evidence-based nutrition guidelines for the prevention and management of diabetes. Diabetic medicine: a journal of the British Diabetic Association 2018; 35: 541-547 DOI: 10.1111/dme.13603.
  • 120 Dworatzek PD, Arcudi K, Gougeon R. et al. Nutrition therapy. Can J Diabetes 2013; 37: S45-S55 DOI: 10.1016/j.jcjd.2013.01.019.
  • 121 Hallberg SJ, Dockter NE, Kushner JA. et al. Improving the scientific rigour of nutritional recommendations for adults with type 2 diabetes: A comprehensive review of the American Diabetes Association guidelinerecommended eating patterns. Diabetes Obes Metab 2019; 21: 1769-1779 DOI: 10.1111/dom.13736.
  • 122 Salas-Salvadó J, Becerra-Tomás N, Papandreou C. et al. Dietary Patterns Emphasizing the Consumption of Plant Foods in the Management of Type 2 Diabetes: A Narrative Review. Adv Nutr 2019; 10: S320-S331 DOI: 10.1093/advances/nmy102.
  • 123 Viguiliouk E, Kendall CW, Kahleová H. et al. Effect of vegetarian dietary patterns on cardiometabolic risk factors in diabetes: A systematic review and meta-analysis of randomized controlled trials. Clin Nutr 2019; 38: 1133-1145 DOI: 10.1016/j.clnu.2018.05.032.
  • 124 Papamichou D, Panagiotakos DB, Itsiopoulos C. Dietary patterns and management of type 2 diabetes: A systematic review of randomised clinical trials. Nutr Metab Cardiovasc Dis 2019; 29: 531-543 DOI: 10.1016/j.numecd.2019.02.004.
  • 125 Ohlsson B. An Okinawan-based Nordic diet improves glucose and lipid metabolism in health and type 2 diabetes, in alignment with changes in the endocrine profile, whereas zonulin levels are elevated. Exp Ther Med 2019; 17: 2883-2893 DOI: 10.3892/etm.2019.7303.
  • 126 Daneshzad E, Emami S, Darooghegi Mofrad M. et al. Association of modified Nordic diet with cardiovascular risk factors among type 2 diabetes patients: a cross-sectional study. J Cardiovasc Thorac Res 2018; 10: 153-161 DOI: 10.15171/jcvtr.2018.25.
  • 127 Via MA, Mechanick JI. Nutrition in Type 2 Diabetes and the Metabolic Syndrome. Med Clin North Am 2016; 100: 1285-1302 DOI: 10.1016/j.mcna.2016.06.009.
  • 128 Porrata-Maury C, Hernández-Triana M, Ruiz-Álvarez V. et al. Ma-Pi 2 macrobiotic diet and type 2 diabetes mellitus: pooled analysis of shortterm intervention studies. Diabetes Metab Res Rev 2014; 30: 55-66 DOI: 10.1002/dmrr.2519.
  • 129 Garvey WT, Mechanick JI, Brett EM. et al. American Association of Clinical Endocrinologists and American College of Endocrinology Comprehensive CLINICAL Practice Guidelines for Medical Care of Patients with Obesity. Endocr Pract 2016; 22: 1-203 DOI: 10.4158/EP161365.GL.
  • 130 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: 505-516 DOI: 10.3945/ajcn.112.042457.
  • 131 Huo R, Du T, Xu Y. et al. Effects of Mediterranean-style diet on glycemic control, weight loss and cardiovascular risk factors among type 2 diabetes individuals: a meta-analysis. Eur J Clin Nutr 2015; 69: 1200-1208 DOI: 10.1038/ejcn.2014.243.
  • 132 Pan B, Wu Y, Yang Q. et al. The impact of major dietary patterns on glycemic control, cardiovascular risk factors, and weight loss in patients with type 2 diabetes: A network meta-analysis. J Evid Based Med 2019; 12: 29-39 DOI: 10.1111/jebm.12312.
  • 133 Johannesen CO, Dale HF, Jensen C. et al. Effects of Plant-Based Diets on Outcomes Related to Glucose Metabolism: A Systematic Review. Diabetes Metab Syndr Obes 2020; 13: 2811-2822 DOI: 10.2147/DMSO.S265982.
  • 134 Toumpanakis A, Turnbull T, Alba-Barba I. Effectiveness of plant-based diets in promoting well-being in the management of type 2 diabetes: a systematic review. BMJ Open Diabetes Res Care 2018; 6: e000534 DOI: 10.1136/bmjdrc-2018-000534.
  • 135 Tran E, Dale HF, Jensen C. et al. Effects of Plant-Based Diets on Weight Status: A Systematic Review. Diabetes Metab Syndr Obes 2020; 13: 3433-3448 DOI: 10.2147/DMSO.S272802.
  • 136 Medawar E, Huhn S, Villringer A. et al. The effects of plant-based diets on the body and the brain: a systematic review. Transl Psychiatry 2019; 9: 226 DOI: 10.1038/s41398-019-0552-0.
  • 137 Esposito K, Maiorino MI, Bellastella G. et al. A journey into a Mediterranean diet and type 2 diabetes: a systematic review with meta-analyses. BMJ Open 2015; 5: e008222 DOI: 10.1136/bmjopen-2015-008222.
  • 138 Carter P, Achana F, Troughton J. et al. A Mediterranean diet improves HbA1c but not fasting blood glucose compared to alternative dietary strategies: a network meta-analysis. J Hum Nutr Diet 2014; 27: 280-297 DOI: 10.1111/jhn.12138.
  • 139 Emadian A, Andrews RC, England CY. et al. The effect of macronutrients on glycaemic control: a systematic review of dietary randomised controlled trials in overweight and obese adults with type 2 diabetes in which there was no difference in weight loss between treatment groups. Br J Nutr 2015; 114: 1656-1666 DOI: 10.1017/S0007114515003475.
  • 140 Kahleova H, Salas-Salvadó J, Rahelić D. et al. Dietary Patterns and Cardiometabolic Outcomes in Diabetes: A Summary of Systematic Reviews and Meta-Analyses. Nutrients 2019; 11 DOI: 10.3390/nu11092209.
  • 141 DDG https://www.deutsche-diabetes-gesellschaft.de/fileadmin/user_upload/01_Die_DDG/03_Ausschuesse/02_Ernaehrung/2015-057-025l_S3_Diabetes_mellitus_Empfehlungen_Proteinzufuhr_2015-10.pdfStand: 06.07.2021
  • 142 Pfeiffer AFH, Pedersen E, Schwab U. et al. The Effects of Different Quantities and Qualities of Protein Intake in People with Diabetes Mellitus. Nutrients 2020; 12 DOI: 10.3390/nu12020365.
  • 143 Mittendorfer B, Klein S, Fontana L. A word of caution against excessive protein intake. Nat Rev Endocrinol 2020; 16: 59-66 DOI: 10.1038/s41574-019-0274-7.
  • 144 Labonte CC, Chevalier S, Marliss EB. et al. Effect of 10% dietary protein intake on whole body protein kinetics in type 2 diabetic adults. Clin Nutr 2015; 34: 1115-1121 DOI: 10.1016/j.clnu.2014.12.022.
  • 145 Markova M, Hornemann S, Sucher S. et al. Rate of appearance of amino acids after a meal regulates insulin and glucagon secretion in patients with type 2 diabetes: a randomized clinical trial. Am J Clin Nutr 2018; 108: 279-291 DOI: 10.1093/ajcn/nqy100.
  • 146 Volkert D. Aktuelle ESPEN-Leitlinie Klinische Ernährung und Hydration in der Geriatrie. Dtsch Med Wochenschr 2020; 145: 1306-1314 DOI: 10.1055/a-0986-2892.
  • 147 Song M, Fung TT, Hu FB. et al. Association of Animal and Plant Protein Intake With All-Cause and Cause-Specific Mortality. JAMA Intern Med 2016; 176: 1453-1463 DOI: 10.1001/jamainternmed.2016.4182.
  • 148 Ye J, Yu Q, Mai W. et al. Dietary protein intake and subsequent risk of type 2 diabetes: a dose-response meta-analysis of prospective cohort studies. Acta Diabetol 2019; 56: 851-870 DOI: 10.1007/s00592-019-01320-x.
  • 149 Vernooij RWM, Zeraatkar D, Han MA. et al. Patterns of Red and Processed Meat Consumption and Risk for Cardiometabolic and Cancer Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann Intern Med 2019; 171: 732-741 DOI: 10.7326/M19-1583.
  • 150 Vogtschmidt YD, Raben A, Faber I. et al. Is protein the forgotten ingredient: Effects of higher compared to lower protein diets on cardiometabolic risk factors. A systematic review and meta-analysis of randomised controlled trials. Atherosclerosis 2021; DOI: 10.1016/j.atherosclerosis.2021.05.011.
  • 151 Clifton PM, Condo D, Keogh JB. Long term weight maintenance after advice to consume low carbohydrate, higher protein diets-a systematic review and meta analysis. Nutr Metab Cardiovasc Dis 2014; 24: 224-235 DOI: 10.1016/j.numecd.2013.11.006.
  • 152 Hahn D, Hodson EM, Fouque D. Low protein diets for non-diabetic adults with chronic kidney disease. Cochrane Database Syst Rev 2020; 10: CD001892 DOI: 10.1002/14651858.CD001892.pub5.
  • 153 Ikizler TA, Burrowes JD, Byham-Gray LD. et al. KDOQI Clinical Practice Guideline for Nutrition in CKD: 2020 Update. Am J Kidney Dis 2020; 76: S1-S107 DOI: 10.1053/j.ajkd.2020.05.006.
  • 154 Menon V, Kopple JD, Wang X. et al. Effect of a very low-protein diet on outcomes: long-term follow-up of the Modification of Diet in Renal Disease (MDRD) Study. Am J Kidney Dis 2009; 53: 208-217 DOI: 10.1053/j.ajkd.2008.08.009.
  • 155 Jiang Z. Effect of restricted protein diet supplemented with keto analogues in end-stage renal disease: a systematic review and meta-analysis. International urology and nephrology 2017; 1-8 DOI: 10.1007/s11255-017-1713-9.
  • 156 Fiaccadori E, Sabatino A, Barazzoni R. et al. ESPEN guideline on clinical nutrition in hospitalized patients with acute or chronic kidney disease. Clin Nutr 2021; 40: 1644-1668 DOI: 10.1016/j.clnu.2021.01.028.
  • 157 Dong JY, Zhang ZL, Wang PY. et al. Effects of high-protein diets on body weight, glycaemic control, blood lipids and blood pressure in type 2 diabetes: meta-analysis of randomised controlled trials. Br J Nutr 2013; 110: 781-789 DOI: 10.1017/S0007114513002055.
  • 158 [Anonymous]. Carbohydrates in human nutrition. Report of a Joint FAO/WHO Expert Consultation. FAO Food Nutr Pap 1998; 66: 1-140
  • 159 Wolever TMS. Personalized nutrition by prediction of glycaemic responses: fact or fantasy?. Eur J Clin Nutr 2016; 70: 411-413 DOI: 10.1038/ejcn.2016.31.
  • 160 Berry SE, Valdes AM, Drew DA. et al. Human postprandial responses to food and potential for precision nutrition. Nat Med 2020; 26: 964-973 DOI: 10.1038/s41591-020-0934-0.
  • 161 Zeevi D, Korem T, Zmora N. et al. Personalized Nutrition by Prediction of Glycemic Responses. Cell 2015; 163: 1079-1094 DOI: 10.1016/j.cell.2015.11.001.
  • 162 Jung CH, Choi KM. Impact of High-Carbohydrate Diet on Metabolic Parameters in Patients with Type 2 Diabetes. Nutrients 2017; 9 DOI: 10.3390/nu9040322.
  • 163 Livesey G, Taylor R, Livesey H. et al. Is there a dose-response relation of dietary glycemic load to risk of type 2 diabetes? Meta-analysis of prospective cohort studies. Am J Clin Nutr 2013; 97: 584-596 DOI: 10.3945/ajcn.112.041467.
  • 164 Livesey G, Livesey H. Coronary Heart Disease and Dietary Carbohydrate, Glycemic Index, and Glycemic Load: Dose-Response Meta-analyses of Prospective Cohort Studies. Mayo Clin Proc Innov Qual Outcomes 2019; 3: 52-69 DOI: 10.1016/j.mayocpiqo.2018.12.007.
  • 165 Thomas DE, Elliott EJ. The use of low-glycaemic index diets in diabetes control. Br J Nutr 2010; 104: 797-802 DOI: 10.1017/S0007114510001534.
  • 166 Xu B, Fu J, Qiao Y. et al. Higher intake of microbiota-accessible carbohydrates and improved cardiometabolic risk factors: a meta-analysis and umbrella review of dietary management in patients with type 2 diabetes. Am J Clin Nutr 2021; DOI: 10.1093/ajcn/nqaa435.
  • 167 Jenkins DJA, Kendall CWC, McKeown-Eyssen G. et al. Effect of a lowglycemic index or a high-cereal fiber diet on type 2 diabetes: a randomized trial. JAMA 2008; 300: 2742-2753 DOI: 10.1001/jama.2008.808.
  • 168 Holub I, Gostner A, Hessdörfer S. et al. Improved metabolic control after 12-week dietary intervention with low glycaemic isomalt in patients with type 2 diabetes mellitus. Horm Metab Res 2009; 41: 886-892 DOI: 10.1055/s-0029-1234107.
  • 169 Brand-Miller J, Hayne S, Petocz P. et al. Low-glycemic index diets in the management of diabetes: a meta-analysis of randomized controlled trials. Diabetes care 2003; 26: 2261-2267 DOI: 10.2337/diacare.26.8.2261.
  • 170 Ojo O, Ojo OO, Adebowale F. et al. 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 DOI: 10.3390/nu10030373.
  • 171 Franz MJ, MacLeod J, Evert A. et al. Academy of Nutrition and Dietetics Nutrition Practice Guideline for Type 1 and Type 2 Diabetes in Adults: Systematic Review of Evidence for Medical Nutrition Therapy Effectiveness and Recommendations for Integration into the Nutrition Care Process. J Acad Nutr Diet 2017; 117: 1659-1679 DOI: 10.1016/j.jand.2017.03.022.
  • 172 Vega-López S, Venn BJ, Slavin JL. Relevance of the Glycemic Index and Glycemic Load for Body Weight, Diabetes, and Cardiovascular Disease. Nutrients 2018; 10 DOI: 10.3390/nu10101361.
  • 173 Jenkins DJA, Dehghan M, Mente A. et al. Glycemic Index, Glycemic Load, and Cardiovascular Disease and Mortality. N Engl J Med 2021; 384: 1312-1322 DOI: 10.1056/NEJMoa2007123.
  • 174 Coutinho M, Gerstein HC, Wang Y. et al. The relationship between glucose and incident cardiovascular events. A metaregression analysis of published data from 20 studies of 95783 individuals followed for 12.4 years. Diabetes care 1999; 22: 233-240 DOI: 10.2337/diacare.22.2.233.
  • 175 Levitan EB, Song Y, Ford ES. et al. Is nondiabetic hyperglycemia a risk factor for cardiovascular disease? A meta-analysis of prospective studies. Arch Intern Med 2004; 164: 2147-2155 DOI: 10.1001/archinte.164.19.2147.
  • 176 Siri PW, Krauss RM. Influence of dietary carbohydrate and fat on LDL and HDL particle distributions. Curr Atheroscler Rep 2005; 7: 455-459 DOI: 10.1007/s11883-005-0062-9.
  • 177 Aune D, Norat T, Romundstad P. et al. Whole grain and refined grain consumption and the risk of type 2 diabetes: a systematic review and dose-response meta-analysis of cohort studies. Eur J Epidemiol 2013; 28: 845-858 DOI: 10.1007/s10654-013-9852-5.
  • 178 [Anonymous]. Dietary fibre and incidence of type 2 diabetes in eight European countries: the EPIC-InterAct Study and a meta-analysis of prospective studies. Diabetologia 2015; 58: 1394-1408 DOI: 10.1007/s00125-015-3585-9.
  • 179 Kim Y, Je Y. Dietary fibre intake and mortality from cardiovascular disease and all cancers: A meta-analysis of prospective cohort studies. Arch Cardiovasc Dis 2016; 109: 39-54 DOI: 10.1016/j.acvd.2015.09.005.
  • 180 Reynolds AN, Akerman AP, Mann J. Dietary fibre and whole grains in diabetes management: Systematic review and meta-analyses. PLoS Med 2020; 17: e1003053 DOI: 10.1371/journal.pmed.1003053.
  • 181 Da Silva Borges D, Fernandes R, Thives Mello A. et al. Prebiotics may reduce serum concentrations of C-reactive protein and ghrelin in overweight and obese adults: a systematic review and meta-analysis. Nutr Rev 2020; 78: 235-248 DOI: 10.1093/nutrit/nuz045.
  • 182 Reynolds A, Mann J, Cummings J. et al. Carbohydrate quality and human health: a series of systematic reviews and meta-analyses. Lancet 2019; 393: 434-445 DOI: 10.1016/S0140-6736(18)31809-9.
  • 183 Musa-Veloso K, Poon T, Harkness LS. et al. The effects of whole-grain compared with refined wheat, rice, and rye on the postprandial blood glucose response: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2018; 108: 759-774 DOI: 10.1093/ajcn/nqy112.
  • 184 Wang W, Li J, Chen X. et al. Whole grain food diet slightly reduces cardiovascular risks in obese/overweight adults: a systematic review and meta-analysis. BMC Cardiovasc Disord 2020; 20: 82 DOI: 10.1186/s12872-020-01337-z.
  • 185 Weickert MO, Roden M, Isken F. et al. Effects of supplemented isoenergetic diets differing in cereal fiber and protein content on insulin sensitivity in overweight humans. Am J Clin Nutr 2011; 94: 459-471 DOI: 10.3945/ajcn.110.004374.
  • 186 Honsek C, Kabisch S, Kemper M. et al. Fibre supplementation for the prevention of type 2 diabetes and improvement of glucose metabolism: the randomised controlled Optimal Fibre Trial (OptiFiT). Diabetologia 2018; 61: 1295-1305 DOI: 10.1007/s00125-018-4582-6.
  • 187 Kabisch S, Meyer NMT, Honsek C. et al. Fasting Glucose State Determines Metabolic Response to Supplementation with Insoluble Cereal Fibre: A Secondary Analysis of the Optimal Fibre Trial (OptiFiT). Nutrients 2019; 11 DOI: 10.3390/nu11102385.
  • 188 Hjorth MF, Ritz C, Blaak EE. et al. Pretreatment fasting plasma glucose and insulin modify dietary weight loss success: results from 3 randomized clinical trials. Am J Clin Nutr 2017; 106: 499-505 DOI: 10.3945/ajcn.117.155200.
  • 189 Xiao Z, Chen H, Zhang Y. et al. The effect of psyllium consumption on weight, body mass index, lipid profile, and glucose metabolism in diabetic patients: A systematic review and dose-response meta-analysis of randomized controlled trials. Phytother Res 2020; 34: 1237-1247 DOI: 10.1002/ptr.6609.
  • 190 Wang L, Yang H, Huang H. et al. Inulin-type fructans supplementation improves glycemic control for the prediabetes and type 2 diabetes populations: results from a GRADE-assessed systematic review and doseresponse meta-analysis of 33 randomized controlled trials. J Transl Med 2019; 17: 410 DOI: 10.1186/s12967-019-02159-0.
  • 191 Rao M, Gao C, Xu L. et al. Effect of Inulin-Type Carbohydrates on Insulin Resistance in Patients with Type 2 Diabetes and Obesity: A Systematic Review and Meta-Analysis. J Diabetes Res 2019; 2019: 5101423 DOI: 10.1155/2019/5101423.
  • 192 Darooghegi Mofrad M, Mozaffari H, Mousavi SM. et al. The effects of psyllium supplementation on body weight, body mass index and waist circumference in adults: A systematic review and dose-response metaanalysis of randomized controlled trials. Crit Rev Food Sci Nutr 2020; 60: 859-872 DOI: 10.1080/10408398.2018.1553140.
  • 193 Rahmani J, Miri A, Černevičiūtė R. et al. Effects of cereal beta-glucan consumption on body weight, body mass index, waist circumference and total energy intake: A meta-analysis of randomized controlled trials. Complement Ther Med 2019; 43: 131-139 DOI: 10.1016/j.ctim.2019.01.018.
  • 194 Ho HVT, Sievenpiper JL, Zurbau A. et al. The effect of oat β-glucan on LDL-cholesterol, non-HDL-cholesterol and apoB for CVD risk reduction: a systematic review and meta-analysis of randomised-controlled trials. Br J Nutr 2016; 116: 1369-1382 DOI: 10.1017/S000711451600341X.
  • 195 Jovanovski E, Yashpal S, Komishon A. et al. Effect of psyllium (Plantago ovata) fiber on LDL cholesterol and alternative lipid targets, non-HDL cholesterol and apolipoprotein B: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2018; 108: 922-932 DOI: 10.1093/ajcn/nqy115.
  • 196 Brum J, Ramsey D, McRorie J. et al. Meta-Analysis of Usefulness of Psyllium Fiber as Adjuvant Antilipid Therapy to Enhance Cholesterol Lowering Efficacy of Statins. The American journal of cardiology 2018; 122: 1169-1174 DOI: 10.1016/j.amjcard.2018.06.040.
  • 197 Ho HVT, Jovanovski E, Zurbau A. et al. A systematic review and metaanalysis of randomized controlled trials of the effect of konjac glucomannan, a viscous soluble fiber, on LDL cholesterol and the new lipid targets non-HDL cholesterol and apolipoprotein B. Am J Clin Nutr 2017; 105: 1239-1247 DOI: 10.3945/ajcn.116.142158.
  • 198 Pittler MH, Ernst E. Guar gum for body weight reduction: meta-analysis of randomized trials. Am J Med 2001; 110: 724-730 DOI: 10.1016/s0002-9343(01)00702-1.
  • 199 Khan K, Jovanovski E, Ho HVT. et al. The effect of viscous soluble fiber on blood pressure: A systematic review and meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis 2018; 28: 3-13 DOI: 10.1016/j.numecd.2017.09.007.
  • 200 Thinggaard M, Jacobsen R, Jeune B. et al. Is the relationship between BMI and mortality increasingly U-shaped with advancing age? A 10-year follow-up of persons aged 70-95 years. J Gerontol A Biol Sci Med Sci 2010; 65: 526-531 DOI: 10.1093/gerona/glp214.
  • 201 Guigoz Y, Vellas B. Malnutrition in the elderly: the Mini Nutritional Assessment (MNA). Ther Umsch 1997; 54: 345-350
  • 202 Rubenstein LZ, Harker JO, Salvà A. et al. Screening for undernutrition in geriatric practice: developing the short-form mini-nutritional assessment (MNA-SF). J Gerontol A Biol Sci Med Sci 2001; 56: M366-M372 DOI: 10.1093/gerona/56.6.m366.
  • 203 [Anonym]. S2k-Leitlinie Diagnostik, Therapie und Verlaufskontrolle des Diabetes mellitus im Alter. 2. Auflage 2018 – AWMF-Register-Nr. 057-017. Diabetologie und Stoffwechsel 2018; 13: 423-489 DOI: 10.1055/a-0666-0820.
  • 204 Volkert D, Bauer J, Frühwald T. et al. S3-Leitlinie der Deutschen Gesellschaft für Ernährungsmedizin (DGEM) in Zusammenarbeit mit der GESKES, der AKE und der DGG Klinische Ernährung in der Geriatrie. Aktuelle Ernährungsmedizin 2013; 38: e1-e48
  • 205 Zeyfang A, Wernecke J, Bahrmann A. Diabetes mellitus im Alter. Diabetologie 2020; 15: S112-S119
  • 206 Şat S, Aydınkoç-Tuzcu K, Berger F. et al. Diabetes und Migration. Diabetologie und Stoffwechsel 2019; 14: S306-S317 DOI: 10.1055/a-0899-2742.
  • 207 Diker O, Deniz T, Çetinkaya A. History of Turkish Cuisine Culture and the Influence of the Balkans. Journal of Humanities And Social Science 2016; 10: 1-6
  • 208 Schmid B. Ernährung und Migration [Zugl.: München, Techn. Univ., Diss., 2003]. München: Utz, Wiss; c; 2003
  • 209 Magni P, Bier DM, Pecorelli S. et al. Perspective: Improving Nutritional Guidelines for Sustainable Health Policies: Current Status and Perspectives. Adv Nutr 2017; 8: 532-545 DOI: 10.3945/an.116.014738.
  • 210 Praxistool zur Ernährung. Orientierungshilfe für die Diabetesberatung nach geografischen Räumen. Im Internet (Stand: 15.07.2021) https://migration.deutsche-diabetes-gesellschaft.de/fileadmin/user_upload/01_Die_DDG/05_Arbeitsgemeinschaften/AG_Migranten/Microsite/200417_Ernaehrungstoo_DDG-GB19-Einleger_04.pdf
  • 211 European Commission. Health Promotion and Disease Prevention Knowledge Gateway: Sugars and Sweeteners. Im Internet (Stand: 27.01.2021) https://ec.europa.eu/jrc/en/health-knowledge-gateway/promotion-prevention/nutrition/sugars-sweeteners
  • 212 Scientific Advisory Committee on Nutrition. Carbohydrates and Health report, 2015. Im Internet (Stand: 26.01.2021) https://www.gov.uk/government/publications/
  • 213 McKeown NM, Dashti HS, Ma J. et al. Sugar-sweetened beverage intake associations with fasting glucose and insulin concentrations are not modified by selected genetic variants in a ChREBP-FGF21 pathway: a meta-analysis. Diabetologia 2018; 61: 317-330 DOI: 10.1007/s00125-017-4475-0.
  • 214 Evans RA, Frese M, Romero J. et al. Chronic fructose substitution for glucose or sucrose in food or beverages has little effect on fasting blood glucose, insulin, or triglycerides: a systematic review and metaanalysis. Am J Clin Nutr 2017; 106: 519-529 DOI: 10.3945/ajcn.116.145169.
  • 215 Evans RA, Frese M, Romero J. et al. Fructose replacement of glucose or sucrose in food or beverages lowers postprandial glucose and insulin without raising triglycerides: a systematic review and meta-analysis. Am J Clin Nutr 2017; 106: 506-518 DOI: 10.3945/ajcn.116.145151.
  • 216 Keller A, Heitmann BL, Olsen N. Sugar-sweetened beverages, vascular risk factors and events: a systematic literature review. Public Health Nutr 2015; 18: 1145-1154 DOI: 10.1017/S1368980014002122.
  • 217 Huang C, Huang J, Tian Y. et al. Sugar sweetened beverages consumption and risk of coronary heart disease: a meta-analysis of prospective studies. Atherosclerosis 2014; 234: 11-16 DOI: 10.1016/j.atherosclerosis.2014.01.037.
  • 218 Narain A, Kwok CS, Mamas MA. Soft drinks and sweetened beverages and the risk of cardiovascular disease and mortality: a systematic review and meta-analysis. Int J Clin Pract 2016; 70: 791-805 DOI: 10.1111/ijcp.12841.
  • 219 Cheungpasitporn W, Thongprayoon C, O’Corragain OA. et al. Associations of sugar-sweetened and artificially sweetened soda with chronic kidney disease: a systematic review and meta-analysis. Nephrology (Carlton) 2014; 19: 791-797 DOI: 10.1111/nep.12343.
  • 220 Chen H, Wang J, Li Z. et al. Consumption of Sugar-Sweetened Beverages Has a Dose-Dependent Effect on the Risk of Non-Alcoholic Fatty Liver Disease: An Updated Systematic Review and Dose-Response Meta-Analysis. Int J Environ Res Public Health 2019; 16 DOI: 10.3390/ijerph16122192.
  • 221 Asgari-Taee F, Zerafati-Shoae N, Dehghani M. et al. Association of sugar sweetened beverages consumption with non-alcoholic fatty liver disease: a systematic review and meta-analysis. Eur J Nutr 2019; 58: 1759-1769 DOI: 10.1007/s00394-018-1711-4.
  • 222 Khan TA, Sievenpiper JL. Controversies about sugars: results from systematic reviews and meta-analyses on obesity, cardiometabolic disease and diabetes. Eur J Nutr 2016; 55: 25-43 DOI: 10.1007/s00394-016-1345-3.
  • 223 Choo VL, Viguiliouk E, Blanco Mejia S. et al. Food sources of fructosecontaining sugars and glycaemic control: systematic review and metaanalysis of controlled intervention studies. BMJ 2018; 363: k4644 DOI: 10.1136/bmj.k4644.
  • 224 Semnani-Azad Z, Khan TA, Blanco Mejia S. et al. Association of Major Food Sources of Fructose-Containing Sugars With Incident Metabolic Syndrome: A Systematic Review and Meta-analysis. JAMA Netw Open 2020; 3: e209993 DOI: 10.1001/jamanetworkopen.2020.9993.
  • 225 Bechthold A. Vollwertig essen und trinken nach den 10 Regeln der DGE. Bonn: Deutsche Gesellschaft für Ernährung e. V. (DGE); 2018
  • 226 Wu H, Flint AJ, Qi Q. et al. Association between dietary whole grain intake and risk of mortality: two large prospective studies in US men and women. JAMA Intern Med 2015; 175: 373-384 DOI: 10.1001/jamainternmed.2014.6283.
  • 227 Johnsen NF, Frederiksen K, Christensen J. et al. Whole-grain products and whole-grain types are associated with lower all-cause and causespecific mortality in the Scandinavian HELGA cohort. Br J Nutr 2015; 114: 608-623 DOI: 10.1017/S0007114515001701.
  • 228 Wei H, Gao Z, Liang R. et al. Whole-grain consumption and the risk of all-cause, CVD and cancer mortality: a meta-analysis of prospective cohort studies – CORRIGENDUM. Br J Nutr 2016; 116: 952 DOI: 10.1017/S0007114516002543.
  • 229 Chen GC, Tong X, Xu JY. et al. Whole-grain intake and total, cardiovascular, and cancer mortality: a systematic review and meta-analysis of prospective studies. Am J Clin Nutr 2016; 104: 164-172 DOI: 10.3945/ajcn.115.122432.
  • 230 Benisi-Kohansal S, Saneei P, Salehi-Marzijarani M. et al. Whole-Grain Intake and Mortality from All Causes, Cardiovascular Disease, and Cancer: A Systematic Review and Dose-Response Meta-Analysis of Prospective Cohort Studies. Adv Nutr 2016; 7: 1052-1065 DOI: 10.3945/an.115.011635.
  • 231 Zong G, Gao A, Hu FB. et al. Whole Grain Intake and Mortality From All Causes, Cardiovascular Disease, and Cancer: A Meta-Analysis of Prospective Cohort Studies. Circulation 2016; 133: 2370-2380 DOI: 10.1161/CIRCULATIONAHA.115.021101.
  • 232 Aune D, Keum N, Giovannucci E. et al. Whole grain consumption and risk of cardiovascular disease, cancer, and all cause and cause specific mortality: systematic review and dose-response meta-analysis of prospective studies. BMJ 2016; 353: i2716 DOI: 10.1136/bmj.i2716.
  • 233 Aune D. Plant Foods, Antioxidant Biomarkers, and the Risk of Cardiovascular Disease, Cancer, and Mortality: A Review of the Evidence. Adv Nutr 2019; 10: S404-S421 DOI: 10.1093/advances/nmz042.
  • 234 Zhang B, Zhao Q, Guo W. et al. Association of whole grain intake with all-cause, cardiovascular, and cancer mortality: a systematic review and dose-response meta-analysis from prospective cohort studies. Eur J Clin Nutr 2018; 72: 57-65 DOI: 10.1038/ejcn.2017.149.
  • 235 Jenkins DJ, Wesson V, Wolever TM. et al. Wholemeal versus wholegrain breads: proportion of whole or cracked grain and the glycaemic response. BMJ 1988; 297: 958-960 DOI: 10.1136/bmj.297.6654.958.
  • 236 Reynolds AN, Mann J, Elbalshy M. et al. Wholegrain Particle Size Influences Postprandial Glycemia in Type 2 Diabetes: A Randomized Crossover Study Comparing Four Wholegrain Breads. Diabetes care 2020; 43: 476-479 DOI: 10.2337/dc19-1466.
  • 237 Åberg S, Mann J, Neumann S. et al. Whole-Grain Processing and Glycemic Control in Type 2 Diabetes: A Randomized Crossover Trial. Diabetes care 2020; 43: 1717-1723 DOI: 10.2337/dc20-0263.
  • 238 Jenkins DJA, Kendall CWC, Augustin LSA. et al. Effect of wheat bran on glycemic control and risk factors for cardiovascular disease in type 2 diabetes. Diabetes care 2002; 25: 1522-1528 DOI: 10.2337/diacare.25.9.1522.
  • 239 Miller V, Mente A, Dehghan M. et al. Fruit, vegetable, and legume intake, and cardiovascular disease and deaths in 18 countries (PURE): a prospective cohort study. Lancet 2017; 390: 2037-2049 DOI: 10.1016/S0140-6736(17)32253-5.
  • 240 Aune D, Giovannucci E, Boffetta P. et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality-a systematic review and dose-response meta-analysis of prospective studies. Int J Epidemiol 2017; 46: 1029-1056 DOI: 10.1093/ije/dyw319.
  • 241 Bechthold A, Boeing H, Schwedhelm C. et al. Food groups and risk of coronary heart disease, stroke and heart failure: A systematic review and dose-response meta-analysis of prospective studies. Crit Rev Food Sci Nutr 2019; 59: 1071-1090 DOI: 10.1080/10408398.2017.1392288.
  • 242 Zhan J, Liu YJ, Cai LB. et al. Fruit and vegetable consumption and risk of cardiovascular disease: A meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr 2017; 57: 1650-1663 DOI: 10.1080/10408398.2015.1008980.
  • 243 Willett W, Rockström J, Loken B. et al. Food in the Anthropocene: the EAT-Lancet Commission on healthy diets from sustainable food systems. Lancet 2019; 393: 447-492 DOI: 10.1016/S0140-6736(18)31788-4.
  • 244 Barnard ND, Cohen J, Jenkins DJA. et al. A low-fat vegan diet improves glycemic control and cardiovascular risk factors in a randomized clinical trial in individuals with type 2 diabetes. Diabetes care 2006; 29: 1777-1783 DOI: 10.2337/dc06-0606.
  • 245 Jenkins DJA, Kendall CWC, Augustin LSA. et al. Effect of legumes as part of a low glycemic index diet on glycemic control and cardiovascular risk factors in type 2 diabetes mellitus: a randomized controlled trial. Arch Intern Med 2012; 172: 1653-1660 DOI: 10.1001/2013.jamainternmed.70.
  • 246 Renner B, Arens-Azevêdo U, Watzl B. et al. DGE-Positionspapier zur nachhaltigeren Ernährung. ernährungsumschau 2021; 68: 144-154
  • 247 Jannasch F, Kröger J, Schulze MB. Dietary Patterns and Type 2 Diabetes: A Systematic Literature Review and Meta-Analysis of Prospective Studies. J Nutr 2017; 147: 1174-1182 DOI: 10.3945/jn.116.242552.
  • 248 Wallin A, Di Giuseppe D, Orsini N. et al. Fish consumption, dietary longchain n-3 fatty acids, and risk of type 2 diabetes: systematic review and meta-analysis of prospective studies. Diabetes care 2012; 35: 918-929 DOI: 10.2337/dc11-1631.
  • 249 Xun P, He K. Fish Consumption and Incidence of Diabetes: meta-analysis of data from 438000 individuals in 12 independent prospective cohorts with an average 11-year follow-up. Diabetes care 2012; 35: 930-938 DOI: 10.2337/dc11-1869.
  • 250 Schwingshackl L, Hoffmann G, Lampousi AM. et al. Food groups and risk of type 2 diabetes mellitus: a systematic review and meta-analysis of prospective studies. Eur J Epidemiol 2017; 32: 363-375 DOI: 10.1007/s10654-017-0246-y.
  • 251 Muley A, Muley P, Shah MALA. fatty fish or marine n-3 fatty acids for preventing DM?: a systematic review and meta-analysis. Curr Diabetes Rev 2014; 10: 158-165 DOI: 10.2174/1573399810666140515113137.
  • 252 Schlesinger S, Neuenschwander M, Schwedhelm C. et al. Food Groups and Risk of Overweight, Obesity, and Weight Gain: A Systematic Review and Dose-Response Meta-Analysis of Prospective Studies. Adv Nutr 2019; 10: 205-218 DOI: 10.1093/advances/nmy092.
  • 253 Micha R, Shulkin ML, Peñalvo JL. et al. Etiologic effects and optimal intakes of foods and nutrients for risk of cardiovascular diseases and diabetes: Systematic reviews and meta-analyses from the Nutrition and Chronic Diseases Expert Group (NutriCoDE). PLoS One 2017; 12: e0175149 DOI: 10.1371/journal.pone.0175149.
  • 254 Jayedi A, Shab-Bidar S, Eimeri S. et al. Fish consumption and risk of allcause and cardiovascular mortality: a dose-response meta-analysis of prospective observational studies. Public Health Nutr 2018; 21: 1297-1306 DOI: 10.1017/S1368980017003834.
  • 255 Abdelhamid AS, Brown TJ, Brainard JS. et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11: CD003177 DOI: 10.1002/14651858.CD003177.pub4.
  • 256 Hu Y, Hu FB, Manson JE. Marine Omega-3 Supplementation and Cardiovascular Disease: An Updated Meta-Analysis of 13 Randomized Controlled Trials Involving 127 477 Participants. J Am Heart Assoc 2019; 8: e013543 DOI: 10.1161/JAHA.119.013543.
  • 257 Gao H, Geng T, Huang T. et al. Fish oil supplementation and insulin sensitivity: a systematic review and meta-analysis. Lipids Health Dis 2017; 16: 131 DOI: 10.1186/s12944-017-0528-0.
  • 258 Chen C, Yu X, Shao S. Effects of Omega-3 Fatty Acid Supplementation on Glucose Control and Lipid Levels in Type 2 Diabetes: A Meta-Analysis. PLoS One 2015; 10: e0139565 DOI: 10.1371/journal.pone.0139565.
  • 259 DGE – Deutsche Gesellschaft für Ernährung. Vollwertig essen und trinken nach den 10 Regeln der DGE. Im Internet (Stand: 13.07.2021) https://www.dge.de/ernaehrungspraxis/vollwertige-ernaehrung/10-regeln-der-dge/
  • 260 Zeraatkar D, Han MA, Guyatt GH. et al. Red and Processed Meat Consumption and Risk for All-Cause Mortality and Cardiometabolic Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann Intern Med 2019; 171: 703-710 DOI: 10.7326/M19-0655.
  • 261 Davidson MH, Hunninghake D, Maki KC. et al. Comparison of the effects of lean red meat vs lean white meat on serum lipid levels among freeliving persons with hypercholesterolemia: a long-term, randomized clinical trial. Arch Intern Med 1999; 159: 1331-1338 DOI: 10.1001/archinte.159.12.1331.
  • 262 Hunninghake DB, Maki KC, Kwiterovich PO. et al. Incorporation of lean red meat into a National Cholesterol Education Program Step I diet: a long-term, randomized clinical trial in free-living persons with hypercholesterolemia. J Am Coll Nutr 2000; 19: 351-360 DOI: 10.1080/07315724.2000.10718931.
  • 263 Bergeron N, Chiu S, Williams PT. et al. Effects of red meat, white meat, and nonmeat protein sources on atherogenic lipoprotein measures in the context of low compared with high saturated fat intake: a randomized controlled trial. Am J Clin Nutr 2019; 110: 24-33 DOI: 10.1093/ajcn/nqz035.
  • 264 Charlton K, Walton K, Batterham M. et al. Pork and Chicken Meals Similarly Impact on Cognitive Function and Strength in Community-Living Older Adults: A Pilot Study. J Nutr Gerontol Geriatr 2016; 35: 124-145 DOI: 10.1080/21551197.2016.1162259.
  • 265 Murphy KJ, Parker B, Dyer KA. et al. A comparison of regular consumption of fresh lean pork, beef and chicken on body composition: a randomized cross-over trial. Nutrients 2014; 6: 682-696 DOI: 10.3390/nu6020682.
  • 266 Murphy KJ, Thomson RL, Coates AM. et al. Effects of eating fresh lean pork on cardiometabolic health parameters. Nutrients 2012; 4: 711-723 DOI: 10.3390/nu4070711.
  • 267 Johnston BC, Zeraatkar D, Han MA. et al. Unprocessed Red Meat and Processed Meat Consumption: Dietary Guideline Recommendations From the Nutritional Recommendations (NutriRECS) Consortium. Ann Intern Med 2019; 171: 756-764 DOI: 10.7326/M19-1621.
  • 268 Davis PA, Yokoyama W. Cinnamon intake lowers fasting blood glucose: meta-analysis. J Med Food 2011; 14: 884-889 DOI: 10.1089/jmf.2010.0180.
  • 269 Akilen R, Tsiami A, Devendra D. et al. Cinnamon in glycaemic control: Systematic review and meta analysis. Clin Nutr 2012; 31: 609-615 DOI: 10.1016/j.clnu.2012.04.003.
  • 270 Leach MJ, Kumar S. Cinnamon for diabetes mellitus. Cochrane Database Syst Rev 2012; DOI: 10.1002/14651858.CD007170.pub2.
  • 271 Allen RW, Schwartzman E, Baker WL. et al. Cinnamon use in type 2 diabetes: an updated systematic review and meta-analysis. Ann Fam Med 2013; 11: 452-459 DOI: 10.1370/afm.1517.
  • 272 Costello RB, Dwyer JT, Saldanha L. et al. Do Cinnamon Supplements Have a Role in Glycemic Control in Type 2 Diabetes? A Narrative Review. J Acad Nutr Diet 2016; 116: 1794-1802 DOI: 10.1016/j.jand.2016.07.015.
  • 273 Sierra-Puente D, Abadi-Alfie S, Arakanchi-Altaled K. et al. Cinammon (Cinnamomum Spp.) and Type 2 Diabetes Mellitus. CTNR 2019; 18: 247-255 DOI: 10.37290/ctnr2641-452X.18:247-255.
  • 274 Chan CB, Hashemi Z, Subhan FB. The impact of low and no-caloric sweeteners on glucose absorption, incretin secretion, and glucose tolerance. Appl Physiol Nutr Metab 2017; 42: 793-801 DOI: 10.1139/apnm-2016-0705.
  • 275 Brown AW, Bohan Brown MM, Onken KL. et al. Short-term consumption of sucralose, a nonnutritive sweetener, is similar to water with regard to select markers of hunger signaling and short-term glucose homeostasis in women. Nutr Res 2011; 31: 882-888 DOI: 10.1016/j.nutres.2011.10.004.
  • 276 Ford HE, Peters V, Martin NM. et al. Effects of oral ingestion of sucralose on gut hormone response and appetite in healthy normal-weight subjects. Eur J Clin Nutr 2011; 65: 508-513 DOI: 10.1038/ejcn.2010.291.
  • 277 Steinert RE, Frey F, Töpfer A. et al. Effects of carbohydrate sugars and artificial sweeteners on appetite and the secretion of gastrointestinal satiety peptides. Br J Nutr 2011; 105: 1320-1328 DOI: 10.1017/S000711451000512X.
  • 278 Barriocanal LA, Palacios M, Benitez G. et al. Apparent lack of pharmacological effect of steviol glycosides used as sweeteners in humans. A pilot study of repeated exposures in some normotensive and hypotensive individuals and in Type 1 and Type 2 diabetics. Regul Toxicol Pharmacol 2008; 51: 37-41 DOI: 10.1016/j.yrtph.2008.02.006.
  • 279 Brown RJ, Walter M, Rother KI. Effects of diet soda on gut hormones in youths with diabetes. Diabetes care 2012; 35: 959-964 DOI: 10.2337/dc11-2424.
  • 280 Grotz VL, Henry RR, McGill JB. et al. Lack of effect of sucralose on glucose homeostasis in subjects with type 2 diabetes. J Am Diet Assoc 2003; 103: 1607-1612 DOI: 10.1016/j.jada.2003.09.021.
  • 281 Maki KC, Curry LL, Reeves MS. et al. Chronic consumption of rebaudioside A, a steviol glycoside, in men and women with type 2 diabetes mellitus. Food Chem Toxicol 2008; 46: S47-S53 DOI: 10.1016/j.fct.2008.05.007.
  • 282 Olalde-Mendoza L, Moreno-González YE. Modificación de la glucemia en ayuno en adultos con diabetes mellitus tipo 2 después de la ingesta de refrescos de cola y de dieta en el estado de querétaro, México. Arch Latinoam Nutr 2013; 63: 142-147
  • 283 Temizkan S, Deyneli O, Yasar M. et al. Sucralose enhances GLP-1 release and lowers blood glucose in the presence of carbohydrate in healthy subjects but not in patients with type 2 diabetes. Eur J Clin Nutr 2015; 69: 162-166 DOI: 10.1038/ejcn.2014.208.
  • 284 Ferrazzano GF, Cantile T, Alcidi B. et al. Is Stevia rebaudiana Bertoni a Non Cariogenic Sweetener? A Review. Molecules 2015; 21: E38 DOI: 10.3390/molecules21010038.
  • 285 Prashant GM, Patil RB, Nagaraj T. et al. The antimicrobial activity of the three commercially available intense sweeteners against common periodontal pathogens: an in vitro study. J Contemp Dent Pract 2012; 13: 749-752 DOI: 10.5005/jp-journals-10024-1222.
  • 286 Suez J, Korem T, Zeevi D. et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 2014; 514: 181-186 DOI: 10.1038/nature13793.
  • 287 EFSA 2013. EFSA schließt vollständige Risikobewertung zu Aspartam ab und kommt zu dem Schluss, dass es in den derzeitigen Expositionsmengen sicher ist. Im Internet (Stand: 01.09.2020) https://www.efsa.europa.eu/de/press/news/131210
  • 288 Bundesinstitut für Risikobewertung. Bewertung von Süßstoffen und Zuckeraustauschstoffen. Hintergrundinformation Nr. 025/2014 des BfR vom 1. Juli 2014. Im Internet (Stand: 01.09.2020) www.bfr.bund.de/cm/343/bewertung_von_suessstoffen.pdf
  • 289 Bock PM, Telo GH, Ramalho R. et al. The effect of probiotics, prebiotics or synbiotics on metabolic outcomes in individuals with diabetes: a systematic review and meta-analysis. Diabetologia 2021; 64: 26-41 DOI: 10.1007/s00125-020-05295-1.
  • 290 Rittiphairoj T, Pongpirul K, Janchot K. et al. Probiotics Contribute to Glycemic Control in Patients with Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Adv Nutr 2021; 12: 722-734 DOI: 10.1093/advances/nmaa133.
  • 291 Tao YW, Gu YL, Mao XQ. et al. Effects of probiotics on type II diabetes mellitus: a meta-analysis. J Transl Med 2020; 18: 30 DOI: 10.1186/s12967-020-02213-2.
  • 292 Ardeshirlarijani E, Tabatabaei-Malazy O, Mohseni S. et al. Effect of probiotics supplementation on glucose and oxidative stress in type 2 diabetes mellitus: a meta-analysis of randomized trials. Daru 2019; 27: 827-837 DOI: 10.1007/s40199-019-00302-2.
  • 293 Mahboobi S, Rahimi F, Jafarnejad S. Effects of Prebiotic and Synbiotic Supplementation on Glycaemia and Lipid Profile in Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials. Adv Pharm Bull 2018; 8: 565-574 DOI: 10.15171/apb.2018.065.
  • 294 Akbari V, Hendijani F. Effects of probiotic supplementation in patients with type 2 diabetes: systematic review and meta-analysis. Nutr Rev 2016; 74: 774-784 DOI: 10.1093/nutrit/nuw039.
  • 295 Yao K, Zeng L, He Q. et al. Effect of Probiotics on Glucose and Lipid Metabolism in Type 2 Diabetes Mellitus: A Meta-Analysis of 12 Randomized Controlled Trials. Med Sci Monit 2017; 23: 3044-3053 DOI: 10.12659/msm.902600.
  • 296 Wang C, Zhang C, Li S. et al. Effects of Probiotic Supplementation on Dyslipidemia in Type 2 Diabetes Mellitus: A Meta-Analysis of Randomized Controlled Trials. Foods 2020; 9 DOI: 10.3390/foods9111540.
  • 297 Kasińska MA, Drzewoski J. Effectiveness of probiotics in type 2 diabetes: a meta-analysis. Pol Arch Med Wewn 2015; 125: 803-813 DOI: 10.20452/pamw.3156.
  • 298 Palacios T, Vitetta L, Coulson S. et al. Targeting the Intestinal Microbiota to Prevent Type 2 Diabetes and Enhance the Effect of Metformin on Glycaemia: A Randomised Controlled Pilot Study. Nutrients 2020; 12 DOI: 10.3390/nu12072041.
  • 299 Zheng M, Zhang R, Tian X. et al. Assessing the Risk of Probiotic Dietary Supplements in the Context of Antibiotic Resistance. Front Microbiol 2017; 8: 908 DOI: 10.3389/fmicb.2017.00908.
  • 300 Wong A, Ngu DYS, Dan LA. et al. Detection of antibiotic resistance in probiotics of dietary supplements. Nutr J 2015; 14: 95 DOI: 10.1186/s12937-015-0084-2.
  • 301 BgVV – ehemals: Bundesinstitut für gesundheitlichen Verbraucherschutz und Veterinärmedizin. Abschlussbericht der Arbeitsgruppe „Probiotische Mikroorganismenkulturen in Lebensmitteln“ am BgVV. Im Internet (Stand: 13.07.2021) https://mobil.bfr.bund.de/cm/343/probiot.pdf
  • 302 de Vrese M. Mikrobiologie, Wirkung und Sicherheit von Probiotika. Monatsschrift Kinderheilkunde 2008; 156: 1063-1069 DOI: 10.1007/s00112-008-1829-1.
  • 303 Vrieze A, van Nood E, Holleman F. et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012; 143: 913-916.e7 DOI: 10.1053/j.gastro.2012.06.031.
  • 304 Simon MC, Strassburger K, Nowotny B. et al. Intake of Lactobacillus reuteri improves incretin and insulin secretion in glucose-tolerant humans: a proof of concept. Diabetes care 2015; 38: 1827-1834 DOI: 10.2337/dc14-2690.
  • 305 Tilg H, Moschen AR. Microbiota and diabetes: an evolving relationship. Gut 2014; 63: 1513-1521 DOI: 10.1136/gutjnl-2014-306928.
  • 306 Kjems LL, Holst JJ, Vølund A. et al. The influence of GLP-1 on glucosestimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes 2003; 52: 380-386 DOI: 10.2337/diabetes.52.2.380.
  • 307 Karlsson FH, Tremaroli V, Nookaew I. et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013; 498: 99-103 DOI: 10.1038/nature12198.
  • 308 Qin J, Li Y, Cai Z. et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012; 490: 55-60 DOI: 10.1038/nature11450.
  • 309 Larsen N, Vogensen FK, van den Berg FWJ. et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One 2010; 5: e9085 DOI: 10.1371/journal.pone.0009085.
  • 310 Wu H, Esteve E, Tremaroli V. et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med 2017; 23: 850-858 DOI: 10.1038/nm.4345.
  • 311 Forslund K, Hildebrand F, Nielsen T. et al. Corrigendum: Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2017; 545: 116 DOI: 10.1038/nature22318.
  • 312 Forslund K, Hildebrand F, Nielsen T. et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2015; 528: 262-266 DOI: 10.1038/nature15766.
  • 313 Caesar R. Pharmacologic and Nonpharmacologic Therapies for the Gut Microbiota in Type 2 Diabetes. Can J Diabetes 2019; 43: 224-231 DOI: 10.1016/j.jcjd.2019.01.007.
  • 314 Evert AB, Boucher JL, Cypress M. et al. Nutrition therapy recommendations for the management of adults with diabetes. Diabetes care 2014; 37: S120-S143 DOI: 10.2337/dc14-S120.
  • 315 Sievenpiper JL, Chan CB, Dworatzek PD. et al. Nutrition Therapy. Can J Diabetes 2018; 42: S64-S79 DOI: 10.1016/j.jcjd.2017.10.009.
  • 316 Sievenpiper JL, de Souza RJ, Mirrahimi A. et al. Effect of fructose on body weight in controlled feeding trials: a systematic review and meta-analysis. Ann Intern Med 2012; 156: 291-304 DOI: 10.7326/0003-4819-156-4-201202210-00007.
  • 317 Ha V, Sievenpiper JL, de Souza RJ. et al. Effect of fructose on blood pressure: a systematic review and meta-analysis of controlled feeding trials. Hypertension 2012; 59: 787-795 DOI: 10.1161/HYPERTENSIO-NAHA.111.182311.
  • 318 Chiavaroli L, de Souza RJ, Ha V. et al. Effect of Fructose on Established Lipid Targets: A Systematic Review and Meta-Analysis of Controlled Feeding Trials. J Am Heart Assoc 2015; 4: e001700 DOI: 10.1161/JAHA.114.001700.
  • 319 Wang X, Ouyang Y, Liu J. et al. Fruit and vegetable consumption and mortality from all causes, cardiovascular disease, and cancer: systematic review and dose-response meta-analysis of prospective cohort studies. BMJ 2014; 349: g4490 DOI: 10.1136/bmj.g4490.
  • 320 Chiu S, Sievenpiper JL, de Souza RJ. et al. Effect of fructose on markers of non-alcoholic fatty liver disease (NAFLD): a systematic review and meta-analysis of controlled feeding trials. Eur J Clin Nutr 2014; 68: 416-423 DOI: 10.1038/ejcn.2014.8.
  • 321 Wang DD, Sievenpiper JL, de Souza RJ. et al. The effects of fructose intake on serum uric acid vary among controlled dietary trials. J Nutr 2012; 142: 916-923 DOI: 10.3945/jn.111.151951.
  • 322 Cozma AI, Sievenpiper JL, de Souza RJ. et al. Effect of fructose on glycemic control in diabetes: a systematic review and meta-analysis of controlled feeding trials. Diabetes care 2012; 35: 1611-1620 DOI: 10.2337/dc12-0073.
  • 323 Sievenpiper JL, Chiavaroli L, de Souza RJ. et al. ‘Catalytic’ doses of fructose may benefit glycaemic control without harming cardiometabolic risk factors: a small meta-analysis of randomised controlled feeding trials. Br J Nutr 2012; 108: 418-423 DOI: 10.1017/S000711451200013X.
  • 324 Sievenpiper JL, Carleton AJ, Chatha S. et al. Heterogeneous effects of fructose on blood lipids in individuals with type 2 diabetes: systematic review and meta-analysis of experimental trials in humans. Diabetes care 2009; 32: 1930-1937 DOI: 10.2337/dc09-0619.
  • 325 Chung M, Ma J, Patel K. et al. Fructose, high-fructose corn syrup, sucrose, and nonalcoholic fatty liver disease or indexes of liver health: a systematic review and meta-analysis. Am J Clin Nutr 2014; 100: 833-849 DOI: 10.3945/ajcn.114.086314.
  • 326 Goran MI, Ulijaszek SJ, Ventura EE. High fructose corn syrup and diabetes prevalence: a global perspective. Glob Public Health 2013; 8: 55-64 DOI: 10.1080/17441692.2012.736257.
  • 327 Tsilas CS, de Souza RJ, Mejia SB. et al. Relation of total sugars, fructose and sucrose with incident type 2 diabetes: a systematic review and meta-analysis of prospective cohort studies. CMAJ 2017; 189: E711-E720 DOI: 10.1503/cmaj.160706.
  • 328 David Wang D, Sievenpiper JL, de Souza RJ. et al. Effect of fructose on postprandial triglycerides: a systematic review and meta-analysis of controlled feeding trials. Atherosclerosis 2014; 232: 125-133 DOI: 10.1016/j.atherosclerosis.2013.10.019.
  • 329 Zhang YH, An T, Zhang RC. et al. Very high fructose intake increases serum LDL-cholesterol and total cholesterol: a meta-analysis of controlled feeding trials. J Nutr 2013; 143: 1391-1398 DOI: 10.3945/jn.113.175323.
  • 330 Schwingshackl L, Neuenschwander M, Hoffmann G. et al. Dietary sugars and cardiometabolic risk factors: a network meta-analysis on isocaloric substitution interventions. Am J Clin Nutr 2020; 111: 187-196 DOI: 10.1093/ajcn/nqz273.
  • 331 Weber KS, Simon MC, Strassburger K. et al. Habitual Fructose Intake Relates to Insulin Sensitivity and Fatty Liver Index in Recent-Onset Type 2 Diabetes Patients and Individuals without Diabetes. Nutrients 2018; 10 DOI: 10.3390/nu10060774.
  • 332 ter Horst KW, Schene MR, Holman R. et al. Effect of fructose consumption on insulin sensitivity in nondiabetic subjects: a systematic review and meta-analysis of diet-intervention trials. Am J Clin Nutr 2016; 104: 1562-1576 DOI: 10.3945/ajcn.116.137786.
  • 333 Kulzer B, Albus C, Herpertz S. et al. Psychosoziales und Diabetes. Der Diabetologe 2019; 15: 452-469 DOI: 10.1007/s11428-018-0440-x.
  • 334 Ahmed AT, Karter AJ, Warton EM. et al. The relationship between alcohol consumption and glycemic control among patients with diabetes: the Kaiser Permanente Northern California Diabetes Registry. J Gen Intern Med 2008; 23: 275-282 DOI: 10.1007/s11606-007-0502-z.
  • 335 Bantle AE, Thomas W, Bantle JP. Metabolic effects of alcohol in the form of wine in persons with type 2 diabetes mellitus. Metabolism 2008; 57: 241-245 DOI: 10.1016/j.metabol.2007.09.007.
  • 336 Avogaro A, Beltramello P, Gnudi L. et al. Alcohol intake impairs glucose counterregulation during acute insulin-induced hypoglycemia in IDDM patients. Evidence for a critical role of free fatty acids. Diabetes 1993; 42: 1626-1634
  • 337 Turner BC, Jenkins E, Kerr D. et al. The effect of evening alcohol consumption on next-morning glucose control in type 1 diabetes. Diabetes care 2001; 24: 1888-1893 DOI: 10.2337/diacare.24.11.1888.
  • 338 Richardson T, Weiss M, Thomas P. et al. Day after the night before: influence of evening alcohol on risk of hypoglycemia in patients with type 1 diabetes. Diabetes care 2005; 28: 1801-1802 DOI: 10.2337/diacare.28.7.1801.
  • 339 Pedersen-Bjergaard U, Reubsaet JLE, Nielsen SL. et al. Psychoactive drugs, alcohol, and severe hypoglycemia in insulin-treated diabetes: analysis of 141 cases. Am J Med 2005; 118: 307-310 DOI: 10.1016/j.amjmed.2004.07.054.
  • 340 Frier B, Fisher M. Hrsg Moderators, monitoring and management of hypoglycaemia [101-120]. Chichester: John Wiley & Sons; 2007
  • 341 Ahmed AT, Karter AJ, Liu J. Alcohol consumption is inversely associated with adherence to diabetes self-care behaviours. Diabetic medicine: a journal of the British Diabetic Association 2006; 23: 795-802 DOI: 10.1111/j.1464-5491.2006.01878.x.
  • 342 Nahas R, Goguen J. Natural health products. Can J Diabetes 2013; 37: S97-S99 DOI: 10.1016/j.jcjd.2013.01.029.
  • 343 Hartweg J, Perera R, Montori V. et al. Omega-3 polyunsaturated fatty acids (PUFA) for type 2 diabetes mellitus. Cochrane Database Syst Rev 2008; DOI: 10.1002/14651858.CD003205.pub2.
  • 344 Hartweg J, Farmer AJ, Holman RR. et al. Potenzial impact of omega-3 treatment on cardiovascular disease in type 2 diabetes. Curr Opin Lipidol 2009; 20: 30-38 DOI: 10.1097/mol.0b013e328321b3be.
  • 345 O’Mahoney LL, Matu J, Price OJ. et al. Omega-3 polyunsaturated fatty acids favourably modulate cardiometabolic biomarkers in type 2 diabetes: a meta-analysis and meta-regression of randomized controlled trials. Cardiovasc Diabetol 2018; 17: 98 DOI: 10.1186/s12933-018-0740-x.
  • 346 Mirhosseini N, Vatanparast H, Mazidi M. et al. The Effect of Improved Serum 25-Hydroxyvitamin D Status on Glycemic Control in Diabetic Patients: A Meta-Analysis. J Clin Endocrinol Metab 2017; 102: 3097-3110 DOI: 10.1210/jc.2017-01024.
  • 347 Li X, Liu Y, Zheng Y. et al. The Effect of Vitamin D Supplementation on Glycemic Control in Type 2 Diabetes Patients: A Systematic Review and Meta-Analysis. Nutrients 2018; 10 DOI: 10.3390/nu10030375.
  • 348 Jafari T, Fallah AA, Barani A. Effects of vitamin D on serum lipid profile in patients with type 2 diabetes: A meta-analysis of randomized controlled trials. Clin Nutr 2016; 35: 1259-1268 DOI: 10.1016/j.clnu.2016.03.001.
  • 349 Mousa A, Naderpoor N, Teede H. et al. Vitamin D supplementation for improvement of chronic low-grade inflammation in patients with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 2018; 76: 380-394 DOI: 10.1093/nutrit/nux077.
  • 350 Lee KJ, Lee YJ. Effects of vitamin D on blood pressure in patients with type 2 diabetes mellitus. Int J Clin Pharmacol Ther 2016; 54: 233-242 DOI: 10.5414/CP202493.
  • 351 Yu Y, Tian L, Xiao Y. et al. Effect of Vitamin D Supplementation on Some Inflammatory Biomarkers in Type 2 Diabetes Mellitus Subjects: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Ann Nutr Metab 2018; 73: 62-73 DOI: 10.1159/000490358.
  • 352 Verma H, Garg R. Effect of magnesium supplementation on type 2 diabetes associated cardiovascular risk factors: a systematic review and meta-analysis. J Hum Nutr Diet 2017; 30: 621-633 DOI: 10.1111/jhn.12454.
  • 353 Simental-Mendía LE, Sahebkar A, Rodríguez-Morán M. et al. A systematic review and meta-analysis of randomized controlled trials on the effects of magnesium supplementation on insulin sensitivity and glucose control. Pharmacol Res 2016; 111: 272-282 DOI: 10.1016/j.phrs.2016.06.019.
  • 354 Asbaghi O, Hosseini R, Boozari B. et al. The Effects of Magnesium Supplementation on Blood Pressure and Obesity Measure Among Type 2 Diabetes Patient: a Systematic Review and Meta-analysis of Randomized Controlled Trials. Biol Trace Elem Res 2021; 199: 413-424 DOI: 10.1007/s12011-020-02157-0.
  • 355 Vincent JB. Elucidating a biological role for chromium at a molecular level. Acc Chem Res 2000; 33: 503-510 DOI: 10.1021/ar990073r.
  • 356 Asbaghi O, Fatemeh N, Mahnaz RK. et al. Effects of chromium supplementation on glycemic control in patients with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res 2020; 161: 105098 DOI: 10.1016/j.phrs.2020.105098.
  • 357 Yin RV, Phung OJ. Effect of chromium supplementation on glycated hemoglobin and fasting plasma glucose in patients with diabetes mellitus. Nutr J 2015; 14: 14 DOI: 10.1186/1475-2891-14-14.
  • 358 Suksomboon N, Poolsup N, Yuwanakorn A. Systematic review and meta-analysis of the efficacy and safety of chromium supplementation in diabetes. J Clin Pharm Ther 2014; 39: 292-306 DOI: 10.1111/jcpt.12147.
  • 359 Chimienti F. Zinc, pancreatic islet cell function and diabetes: new insights into an old story. Nutr Res Rev 2013; 26: 1-11 DOI: 10.1017/S0954422412000212.
  • 360 de Carvalho GB. Zinc’s role in the glycemic control of patients with type 2 diabetes: a systematic review. BioMetals 2017; 1-12 DOI: 10.1007/s10534-017-9996-y.
  • 361 Fernández-Cao JC, Warthon-Medina M, Hall Moran V. et al. Dietary zinc intake and whole blood zinc concentration in subjects with type 2 diabetes versus healthy subjects: A systematic review, meta-analysis and meta-regression. J Trace Elem Med Biol 2018; 49: 241-251 DOI: 10.1016/j.jtemb.2018.02.008.
  • 362 Wang X, Wu W, Zheng W. et al. Zinc supplementation improves glycemic control for diabetes prevention and management: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2019; 110: 76-90 DOI: 10.1093/ajcn/nqz041.
  • 363 Asbaghi O, Sadeghian M, Fouladvand F. et al. Effects of zinc supplementation on lipid profile in patients with type 2 diabetes mellitus: A systematic review and meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis 2020; 30: 1260-1271 DOI: 10.1016/j.numecd.2020.03.021.
  • 364 Rahimi R, Nikfar S, Larijani B. et al. A review on the role of antioxidants in the management of diabetes and its complications. Biomed Pharmacother 2005; 59: 365-373 DOI: 10.1016/j.biopha.2005.07.002.
  • 365 Ashor AW, Werner AD, Lara J. et al. Effects of vitamin C supplementation on glycaemic control: a systematic review and meta-analysis of randomised controlled trials. Eur J Clin Nutr 2017; 71: 1371-1380 DOI: 10.1038/ejcn.2017.24.
  • 366 Xu R, Zhang S, Tao A. et al. Influence of vitamin E supplementation on glycaemic control: a meta-analysis of randomised controlled trials. PLoS One 2014; 9: e95008 DOI: 10.1371/journal.pone.0095008.
  • 367 Khodaeian M, Tabatabaei-Malazy O, Qorbani M. et al. Effect of vitamins C and E on insulin resistance in diabetes: a meta-analysis study. Eur J Clin Invest 2015; 45: 1161-1174 DOI: 10.1111/eci.12534.
  • 368 Montero D, Walther G, Stehouwer CDA. et al. Effect of antioxidant vitamin supplementation on endothelial function in type 2 diabetes mellitus: a systematic review and meta-analysis of randomized controlled trials. Obes Rev 2014; 15: 107-116 DOI: 10.1111/obr.12114.
  • 369 Tabatabaei-Malazy O, Ardeshirlarijani E, Namazi N. et al. Dietary antioxidative supplements and diabetic retinopathy; a systematic review. J Diabetes Metab Disord 2019; 18: 705-716 DOI: 10.1007/s40200-019-00434-x.
  • 370 Jeyaraman MM, Al-Yousif NSH, Singh Mann A. et al. Resveratrol for adults with type 2 diabetes mellitus. Cochrane Database Syst Rev 2020; 1: CD011919 DOI: 10.1002/14651858.CD011919.pub2.
  • 371 Palma-Duran SA, Vlassopoulos A, Lean M. et al. Nutritional intervention and impact of polyphenol on glycohemoglobin (HbA1c) in non-diabetic and type 2 diabetic subjects: Systematic review and meta-analysis. Crit Rev Food Sci Nutr 2017; 57: 975-986 DOI: 10.1080/10408398.2014.973932.
  • 372 Fogacci F, Tocci G, Presta V. et al. Effect of resveratrol on blood pressure: A systematic review and meta-analysis of randomized, controlled, clinical trials. Crit Rev Food Sci Nutr 2019; 59: 1605-1618 DOI: 10.1080/10408398.2017.1422480.
  • 373 Drzikova B. Haferprodukte mit modifiziertem Gehalt an β-Glucanen und resistenter Stärke und ihre Effekte auf den Gastrointestinaltrakt unter In-vitro- und In-vivo-Bedingungen. 2005 http://opus.kobv.de/ubp/volltexte/205/592/
  • 374 He L, Zhao J, Huang Y. et al. The difference between oats and beta-glucan extract intake in the management of HbA1c, fasting glucose and insulin sensitivity: ameta-analysis of randomized controlled trials. Food Funct 2016; 7: 1413-1428 DOI: 10.1039/c5fo01364j.
  • 375 Abbasi NN, Purslow PP, Tosh SM. et al. Oat β-glucan depresses SGLT1- and GLUT2-mediated glucose transport in intestinal epithelial cells (IEC-6). Nutr Res 2016; 36: 541-552 DOI: 10.1016/j.nutres.2016.02.004.
  • 376 Wang F, Yu G, Zhang Y. et al. Dipeptidyl Peptidase IV Inhibitory Peptides Derived from Oat (Avena sativa L.), Buckwheat (Fagopyrum esculentum), and Highland Barley (Hordeum vulgare trifurcatum (L.) Trofim) Proteins. J Agric Food Chem 2015; 63: 9543-9549 DOI: 10.1021/acs.jafc.5b04016.
  • 377 Liu M, Zhang Y, Zhang H. et al. The anti-diabetic activity of oat β-d-glucan in streptozotocin-nicotinamide induced diabetic mice. Int J Biol Macromol 2016; 91: 1170-1176 DOI: 10.1016/j.ijbiomac.2016.06.083.
  • 378 Lammert A, Kratzsch J, Selhorst J. et al. Clinical benefit of a short term dietary oatmeal intervention in patients with type 2 diabetes and severe insulin resistance: a pilot study. Exp Clin Endocrinol Diabetes 2008; 116: 132-134 DOI: 10.1055/s-2007-984456.
  • 379 Delgado G, Kleber ME, Krämer BK. et al. Dietary Intervention with Oatmeal in Patients with uncontrolled Type 2 Diabetes Mellitus – A Crossover Study. Exp Clin Endocrinol Diabetes 2019; 127: 623-629 DOI: 10.1055/a-0677-6068.
  • 380 Delgado GE, Krämer BK, Scharnagl H. et al. Bile Acids in Patients with Uncontrolled Type 2 Diabetes Mellitus – The Effect of Two Days of Oatmeal Treatment. Exp Clin Endocrinol Diabetes 2020; 128: 624-630 DOI: 10.1055/a-1069-7330.
  • 381 Behall KM, Scholfield DJ, Hallfrisch J. Comparison of hormone and glucose responses of overweight women to barley and oats. J Am Coll Nutr 2005; 24: 182-188 DOI: 10.1080/07315724.2005.10719464.
  • 382 Braaten JT, Scott FW, Wood PJ. et al. High beta-glucan oat bran and oat gum reduce postprandial blood glucose and insulin in subjects with and without type 2 diabetes. Diabetic medicine: a journal of the British Diabetic Association 1994; 11: 312-318 DOI: 10.1111/j.1464-5491.1994.tb00277.x.
  • 383 Pick ME, Hawrysh ZJ, Gee MI. et al. Oat bran concentrate bread products improve long-term control of diabetes: a pilot study. J Am Diet Assoc 1996; 96: 1254-1261 DOI: 10.1016/S0002-8223(96)00329-X.
  • 384 Tapola N, Karvonen H, Niskanen L. et al. Glycemic responses of oat bran products in type 2 diabetic patients. Nutr Metab Cardiovasc Dis 2005; 15: 255-261 DOI: 10.1016/j.numecd.2004.09.003.
  • 385 Tappy L, Gügolz E, Würsch P. Effects of breakfast cereals containing various amounts of beta-glucan fibers on plasma glucose and insulin responses in NIDDM subjects. Diabetes care 1996; 19: 831-834 DOI: 10.2337/diacare.19.8.831.
  • 386 Wood PJ, Beer MU, Butler G. Evaluation of role of concentration and molecular weight of oat beta-glucan in determining effect of viscosity on plasma glucose and insulin following an oral glucose load. Br J Nutr 2000; 84: 19-23
  • 387 [Anonym]. Scientific Opinion on the substantiation of health claims related to beta glucans and maintenance or achievement of normal blood glucose concentrations (ID 756, 802, 2935) pursuant to Article 13(1) of Regulation (EC) No 1924/2006. EFS2 2010; 8 DOI: 10.2903/j.efsa.2010.1482.
  • 388 Amtsblatt der Europäischen Union 2011 L 136/1 vom 25.5.2012 https://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2012:136:0001:0040:DE:PDFStand: 04.07.2021
  • 389 Zurbau A, Noronha JC, Khan TA. et al. The effect of oat β-glucan on postprandial blood glucose and insulin responses: a systematic review and meta-analysis. Eur J Clin Nutr 2021; DOI: 10.1038/s41430-021-00875-9.
  • 390 Battilana P, Ornstein K, Minehira K. et al. Mechanisms of action of betaglucan in postprandial glucose metabolism in healthy men. Eur J Clin Nutr 2001; 55: 327-333 DOI: 10.1038/sj.ejcn.1601160.
  • 391 Jenkins AL, Jenkins DJA, Zdravkovic U. et al. Depression of the glycemic index by high levels of beta-glucan fiber in two functional foods tested in type 2 diabetes. Eur J Clin Nutr 2002; 56: 622-628 DOI: 10.1038/sj.ejcn.1601367.