Next Article in Journal
Nutrition Interventions for Pediatric Obesity Prevention: An Umbrella Review of Systematic Reviews
Next Article in Special Issue
Nutrition at the Intersection between Gut Microbiota Eubiosis and Effective Management of Type 2 Diabetes
Previous Article in Journal
The Impact of an 8-Week Supplementation with Fermented and Non-Fermented Aronia Berry Pulp on Cardiovascular Risk Factors in Individuals with Type 2 Diabetes
Previous Article in Special Issue
Efficacy of Time-Restricted Eating and Behavioral Economic Intervention in Reducing Fasting Plasma Glucose, HbA1c, and Cardiometabolic Risk Factors in Patients with Impaired Fasting Glucose: A Randomized Controlled Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nutritional Strategies for the Management of Type 2 Diabetes Mellitus: A Narrative Review

by
Tatiana Palotta Minari
1,*,
Lúcia Helena Bonalume Tácito
2,
Louise Buonalumi Tácito Yugar
3,
Sílvia Elaine Ferreira-Melo
4,
Carolina Freitas Manzano
1,
Antônio Carlos Pires
2,
Heitor Moreno
4,
José Fernando Vilela-Martin
1,
Luciana Neves Cosenso-Martin
2 and
Juan Carlos Yugar-Toledo
1
1
Department of Hypertension, State Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
2
Department of Endocrinology, State Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
3
School of Medical Sciences, State University of Campinas (UNICAMP), Campinas 13083-887, SP, Brazil
4
Cardiovascular Pharmacology & Hypertension Laboratory, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas 13083-887, SP, Brazil
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(24), 5096; https://doi.org/10.3390/nu15245096
Submission received: 20 November 2023 / Revised: 1 December 2023 / Accepted: 5 December 2023 / Published: 13 December 2023
(This article belongs to the Special Issue Dietary Interventions for Patients with Type 2 Diabetes)

Abstract

:
Background: Thinking about greater adherence to dietary planning, it is extremely important to be aware of all nutritional strategies and dietary prescriptions available in the literature, and of which of them is the most efficient for the management of T2DM. Methods: A search was carried out in 2023 for randomized clinical trials, systematic reviews, meta-analyses, and guidelines in the following databases: Pubmed, Scielo, Web of Science, CrossRef and Google Scholar. In total, 202 articles were collected and analyzed. The period of publications was 1983–2023. Results: There is still no consensus on what the best nutritional strategy or ideal dietary prescription is, and individuality is necessary. In any case, these references suggest that Mediterranean Diet may of greater interest for the management of T2DM, with the following recommended dietary prescription: 40–50% carbohydrates; 15–25% proteins; 25–35% fats (<7% saturated, 10% polyunsaturated, and 10% monounsaturated); at least 14 g of fiber for every 1000 kcal consumed; and <2300 mg sodium. Conclusions: Individuality is the gold standard for dietary prescriptions, however, the Mediterranean diet with low levels of carbohydrates and fats seems to be the most promising strategy for the management of T2DM.

Graphical Abstract

1. Introduction

Type 2 diabetes mellitus (T2DM) is a highly prevalent chronic disease worldwide, and represents one of the biggest public health problems of the 21st century. Its high incidence and prevalence are attributed to population aging and lifestyle, which is characterized by physical inactivity and eating habits that predispose individuals to obesity and metabolic syndrome [1].
At the same time, this epidemiological scenario is worsening with the increase in obesity rates [2]. According to the literature, obesity can predispose individuals to the development of chronic noncommunicable diseases such as T2DM, hypertension and cardiovascular diseases that probably reflect changes in lifestyle characterized by an increase in energy intake and a reduction in physical exercise [2,3,4].
For patients with T2DM and who are overweight, progressive weight loss is recommended to improve quality of life and treatment. This recommendation is based on short-term studies that point to the several benefits of the weight loss process, including improvements in glycemic modulation, cardiorespiratory markers, and quality of life [3,5,6,7]. However, the big question is “what is the patient’s motivation to form new healthy habits and control the disease in the long term?” [8].
Thinking about greater adherence to dietary planning, it is important for the nutritionist to be aware of all dietary prescriptions and nutritional strategies available in the literature. Another step would be a critical analysis of them, thus verifying which would be the most efficient in the management of T2DM.

2. Methods

A search was carried out in 2023 for randomized clinical trials, systematic reviews, meta-analyses, and guidelines in the following databases: Pubmed, Scielo, Web of Science, CrossRef, and Google Scholar. In total, 202 articles were collected and analyzed. The period of publications was 1983–2023. The MeSH indexed terms searched were nutritional interventions, nutritional therapy, type 2 diabetes mellitus, nutritional strategies and dietary prescription.

3. Results

3.1. The Importance of Individuality in Nutritional Management of T2DM

The treatment of patients with T2DM can be controlled through structured lifestyle programs, which include dietary education, intensive interdisciplinary therapy and physical exercise [6,9,10,11,12]. The individuality of the treatment and the synergy of interdisciplinary team (nutritionist, endocrinologist, cardiologist, physical educator, and psychologist) are fundamental to increase the patient’s quality of life. Another relevant factor is maintaining empathy and trust between professionals and patients, including the person with T2DM, who is the protagonist of the intervention [9,11,12,13,14,15,16].
The strongest evidence for T2DM prevention includes intensive lifestyle intervention resulting in weight loss, reduced incidence of T2DM in overweight adults, and decreased glucose tolerance over three years [17]. Other studies with lifestyle interventions have shown a reduction of 43% in T2DM progression over 20 years [18,19], of 34% over 10 years [17], and of 27% over 15 years [20], and have demonstrated a reduction in all-cause cardiovascular mortality [21].
Over the years, nutritional interventions have gained increasing prominence in the prevention, treatment, and maintenance of the disease. Strong evidence supports the high effectiveness and cost-effectiveness of nutritional therapy for the treatment of T2DM. Therefore, it is extremely important that all members of the healthcare team know the benefits of improving the quality of life of T2DM patients in the long term [6,22,23,24,25,26].
Nutritional management must consider life cycles, nutritional diagnosis, eating habits, individual characteristics, sociocultural issues, the patient’s economic situation, metabolic profile, use of drugs, physical exercise, and other factors. The dietary prescription, nutritional strategy, and eating plan must be individualized, taking into account the following objectives: to improve glycemic markers, to increase weight loss, and to reduce cardiovascular risk [9,27,28,29,30,31].
A generic healthy living plan is not enough to prevent and control T2DM, being unrealistic and incompatible with the peculiarities of each patient’s clinical case. This review provides clarity about the many dietary choices and patterns that can help people achieve health and a better quality of life [9,27,28,29,30,31]. Individualized nutritional management with a multidisciplinary approach can help reduce glycated hemoglobin (HbA1C), presenting similar or even greater results than would be expected with pharmacological treatment for T2DM. According to evidence, reductions in HbA1C could reach up to 2.0% in patients with T2DM in 3–6 months [32,33]. The cost–effect relationship of various lifestyle interventions for diabetes prevention and control have also been documented in several other studies [32,33,34,35,36].

3.2. Dietary Prescription and Macronutrient Distribution

Evidence suggests that there is no nutritional strategy or ideal percentage of calories from carbohydrates, proteins, and fats for all people with T2DM. Therefore, the type of strategy and distribution of macronutrients should be based on an individualized assessment of current eating patterns, preferences, and metabolic goals. The strategy should include an individualized dietary prescription on carbohydrate intake and glycemic self-monitoring to improve the patient’s meal timing, portioning, and food choices. Health professionals must advise the correct use of medications and regular physical exercise. Moreover, patients with T2DM should also be encouraged to consume fiber, vegetables, legumes (beans, peas, and lentils), fruits, and whole grains for the reduction of HbA1C [9,27,28,36,37,38].

3.2.1. Carbohydrates

Carbohydrate is a source of energy used by the body, and is responsible for the postprandial increase in blood glucose [36,37,38,39]. Foods that contain carbohydrates (sugars, starches, or fiber) have a wide range of effects on the individual glycemic response. Some types of carbohydrate prolong the increase and slow the decrease in blood glucose concentrations, while others cause a rapid rise followed by a rapid fall [40].
Food sources of carbohydrates, especially those made up predominantly of dietary fiber, vitamins, and minerals (those low in added sugars, fats, and sodium) should be highlighted in individualized eating plans [32,36,37,38,41]. The amount of carbohydrates needed for optimal health in patients with T2DM is still uncertain, and studies vary greatly in the types of approaches and percentages of prescriptions. However, the literature indicates that dietary intake of carbohydrates in T2DM should be around 130 g/day or 40–50% of carbohydrates within the total energy value (TEV), aiming mainly to meet brain glucose requirements. The body’s other metabolic processes could be supplied by other energy substrates, such as fatty acids, amino acids (glycogenolysis and gluconeogenesis), and ketone bodies (ketogenesis) [39,40,41,42].
-Glycemic Index and Glycemic Load
The use of glycemic index (GI) and glycemic load (GL) to classify foods rich in carbohydrates according to their effects on glycemia still remains of interest to the scientific community, especially in the management of T2DM. According to studies, GI and GL could provide a prediction of postprandial blood glucose, the glycemic response curve, and may also classify carbohydrate-rich foods according to their postprandial glycemic response [43].
However, two systematic reviews with GI reported no significant impact on HbA1C, and had mixed results on fasting glucose [32,40]. Furthermore, studies use various definitions to classify low- and high-GI foods, leading to uncertainty regarding their usefulness in clinical care, and this would be a too simplistic metric for consideration in the management of T2DM. The authors discuss that several factors can have an impact, such as the following: (1) Individual glycemic response; (2) The preparation method and cooking time of foods that are sources of carbohydrates; (3) The addition of other food types to the meal, such as vegetables, legumes, proteins, and poly and monounsaturated fats; (4) The addition of drinks to the meal; (5) The order of food intake; (6) The gut microbiota [44,45,46,47,48].
A recent study showed that the order of food intake during a meal affects postprandial glucose and insulin peaks in pre-diabetes. The researchers suggest that eating vegetables and protein-rich foods at the beginning of meals and subsequently eating carbohydrate-rich foods may present an innovative and economically accessible behavioral strategy to reduce postprandial glycemic and insulinemic peaks in pre-diabetes, or to prevent T2DM [46].
Another study monitored glucose levels in 800 participants, and their glycemic responses were measured in 46.898 meals. A high individual variability was found in the different responses to the identical meals offered. Some participants had greater postprandial blood sugar spikes after eating cookies, and others did not have such good responses. Other participants did not show an increase in postprandial blood glucose after eating bananas, and others showed an increase. These findings suggest that universal dietary recommendations may have limited usefulness, because there are several factors that can alter these glycemic responses, such as genetics, sex, age, diet, mental health, and even composition of the gut microbiota [45]. Therefore, individuality must be taken into consideration when creating a dietary prescription, especially for T2DM patients who continuously monitor their blood glucose levels [45,46].
According to guidelines, glycemic load has more relevance in treatment of T2DM, as it takes into account the amount of carbohydrates presents within a 100 g portion [9,49]. Counting carbohydrates in the distribution of the patient’s meals is essential when putting together dietary planning. The ‘Carbohydrate Counting Manual’ is a great guide formulated by the Brazilian Diabetes Society that provides a resource for patients with T1DM or T2DM to educate themselves and understand the disease [49]. Therefore, carbohydrate counting, that is, knowing the amount of carbohydrates present in a 100 g portion (glycemic load), seems to be more important for dietary management than analyzing the GI alone. In this way, foods that are sources of carbohydrates (fruits, tubers, cereals, and whole grains) can be included in the dietary plan, as long as they are within the daily carbohydrate quantification and are combined with other foods [9,47,49].

3.2.2. Proteins

The references show a wide disparity in the percentage of protein prescription in the management of T2DM. Some comparisons of protein amounts did not demonstrate differences in results related to T2DM. Prescriptions ranged from 15 to 20% of TEV, but more studies are needed [50,51,52,53,54]. It is worth remembering that in individuals with T1DM and T2DM, the intake of some foods that are sources of protein, such as dairy products, can also increase the postprandial insulin response. Therefore, the use of high-carbohydrate hypercaloric diets in conjunction with high protein should be avoided when treating patients, due to the potential increase insulin [9].
However, high-protein diets are advocated by some researchers as a strategy to facilitate weight loss, compared to other energy restriction strategies. The hypothesis would be that patients with T2DM could benefit from high-protein diets due to better blood glucose control. One study compared 30% protein intake within TEV vs. 15% protein within TEV for 12 weeks. The results showed a reduction in weight, fasting glucose, and medication use in the group that consumed 30% of their TEV in protein [54]. Other studies lasting 4 to 24 weeks reported that high-protein diets (25–32% of TEV) resulted in 2 kg more weight loss and a 0.5% greater improvement in HbA1C, but without significant improvements in fasting blood glucose, total cholesterol or blood pressure [55,56].
A recent meta-analysis analyzed the effect of protein intake on the metabolism of T2DM patients. The results showed that there were no significant differences in relation to HDL and LDL cholesterol in the groups with a high-protein and low-protein diet. The same applies to HbA1C. However, significant reductions in blood pressure and greater weight reduction were observed [56].
Researchers who develop high-protein diets can provide positive regulation of anorectic hormones {cholecystokinin (CCK), peptide YY (PYY) and glucagon-like peptide-1 (GLP-1)}, which suppress brain reactions that are linked to increased appetite. Furthermore, protein consumption also helps to increase gastric emptying time, promoting a greater satietogenic effect. In the long term, reduced appetite may contribute to weight loss, blood pressure, and molecular biomarkers. In any case, a protein intake range of 15% to 20% of TEV is the most prevalent in the literature, and so far appears to be the most fair prescription for the management of T2DM. Additionally, more studies are needed [56].

3.2.3. Fats

The Dietary Guidelines for Americans defines a dietary fat prescription with a 25 to 35% TEV fat range. Dietary patterns that recommend replacing saturated fats with polyunsaturated and monounsaturated fats have demonstrated positive results in reducing blood glucose, triglycerides, LDL cholesterol, and increasing HDL cholesterol. The types or quality of fats in dietary plans can influence results associated with a greater likelihood of developing cardiovascular diseases [57,58,59,60].
The American Heart Association (AHA) recommends a low-fat diet, given that the world population consumes, on average, 36–46% of their TEV in fats. This high intake has been associated in several studies with increased cardiovascular risk. Additionally, only for individuals with hypercholesterolemia and T2DM, the American College of Cardiology (ACC) and the AHA recommend limiting the TEV of saturated fats to 5% to 6%. European and Brazilian guidelines recommend limiting the consumption of saturated fats to <7% of the TEV and total fats to <35% of the TEV to control dyslipidemia and T2DM [16,61,62,63].
High-fat diets, especially those rich in saturated fatty acids, are capable of altering the composition of the gut microbiota, causing a decrease in bacterial diversity and an increase in intestinal permeability. This process raises lipopolysaccharides (LPS) and activation of TLR4 (toll-like receptor 4), generating metabolic endotoxemia and low-grade systemic inflammation. This process add to development of several chronic diseases such as obesity, diabetes, and atherosclerosis [64]. In addition, patients also pay attention to trans fatty acids. Foods containing trans fatty acids (hydrogenated vegetable fat) should be minimized as much as possible from the diet [16,33]. Excess trans and saturated fat intake above the recommendation has been associated with a higher risk of T2DM [16,61,64,65], and can also increase the inflammatory response in the gut microbiota [65].
-Cholesterol
The Dietary Guidelines for Americans concluded that the available evidence does not support the recommendation to limit dietary cholesterol for the general population; the exact recommendations for patients with chronic noncommunicable diseases, such as T2DM, are still less clear. Some researchers argue that the body produces enough cholesterol to perform its physiological and structural functions, so that people do not need to obtain it from food [33]. The studies also indicate that dietary cholesterol intake is correlated with an increase in total cholesterol levels, but this is not linked to a greater likelihood of cardiovascular diseases. More research is needed on the relationship between dietary cholesterol, blood cholesterol, and cardiovascular events in people with T2DM [16,60,66].
-Saturated fat
Guidelines recommend that patients with dyslipidemia and T2DM consume an average of <7% of their TEV of saturated fat [16,33,62,63,67]. The scientific justification for reducing saturated fat in the diet is based on the significant effect of saturated fat intake on increasing LDL-C, a factor that contributes to the development and increased risk of atherosclerosis [68]. The American Heart Association concluded that reducing saturated fat intake and replacing it with unsaturated fats, especially polyunsaturated fats, reduces the incidence of cardiovascular disease [69].
A meta-analysis showed a 17% reduction in the risk of cardiovascular events in studies that reduced saturated fat intake from 9% of the TEV, but no reductions in stroke, cardiovascular mortality, or all-cause mortality were found. Benefits have also been seen when replacing saturated fat with polyunsaturated fat, but not with carbohydrates or proteins [70]. In another study, with patients with T2DM, intake of food sources of monounsaturated and polyunsaturated fats was associated with a lower risk of CVD and death, while intake of saturated fat and trans fats was associated with a higher risk of cardiovascular disease. Replacing saturated fat with monounsaturated or polyunsaturated fat and replacing trans fat with monounsaturated fat have been associated with a reduced risk of developing cardiovascular disease [71,72].
In general, replacing saturated fat with unsaturated fats, especially polyunsaturated fat, significantly reduces total cholesterol and LDL-C, and replacing it with monounsaturated fat from plant sources such as olive oil and nuts reduces the risk of cardiovascular disease [67]. Replacing saturated fat with carbohydrates also lowers total cholesterol and LDL-C, but significantly increases triglycerides and lowers HDL-C [68,73].
Recently, there has been an increase in the consumption and prescription of foods that are sources of saturated fats by doctors and nutritionists, so guidelines have taken a strong stance against the use of coconut oil, lard, bacon, and other foods rich in saturated fatty acids. They have high levels of lauric, myristic, and palmitic acid, so daily consumption should be avoided; they should be replaced by vegetable oils rich in monounsaturated and polyunsaturated fatty acids, such as olive oil, canola oil, and sunflower oil [48].
A systematic review and meta-analysis analyzed the effect of coconut oil consumption on LDL-C, HDL-C and other cardiovascular risk factors compared to other cooking oils. The results showed that coconut oil consumption significantly increased LDL-C +10.47 mg/dL and minimally increased HDL-C +4.00 mg/dL. The authors concluded that even when HDL-C is increased, its daily consumption should be avoided, as it results in higher LDL-C than other vegetable oils [74]. Coconut oil is rich in saturated fatty acids, especially lauric acid (12 carbon atoms), myristic acid (14 carbon atoms), and palmitic acid (16 carbon atoms). These types of fatty acids can contribute to increasing concentrations of total cholesterol and LDL-C [74].
Another meta-analysis also demonstrated that all these saturated fatty acids (lauric, myristic, and palmitic acid were included in items such as coconut oil, hydrogenated vegetable fat, and palm oil) increase LDL cholesterol. Therefore, the authors suggest that coconut oil should be replaced with other unsaturated vegetable oils (olive oil, canola oil, and sunflower oil). The results are extremely relevant for nutritional and dietary guidelines [75].
However, other studies did not show an association between intake of some foods that contain a small portion of saturated fat, such as dairy products and eggs, with an increased risk of diabetes and cardiovascular diseases. Dairy products also have carbohydrates and proteins in their composition, and their saturated fat contents would not be too high compared to other sources of saturated fat. Studies suggest that dairy products can be included in the diet of T2DM, as long as they are quantified in <7% of saturated fats in the TEV. Therefore, there should also be more interest in giving preference to low-fat dairy products to ensure that the daily intake of saturated fats does not exceed 7% of the TEV [76,77,78].
A similar result was found for eggs. Although they contain fat, they are also rich in proteins, water, phytochemicals, carotenoids, and micronutrients. Their consumption was not associated with cardiovascular risk, as long as it is properly quantified within the energy need for saturated fats within the TEV (<7% for T2DM). Unfortunately, there is still no consensus on the amount of egg intake per day or week, as there is great divergence in the literature regarding the intake protocols used [51,62,63,68].
Some research has shown an increase in the risk of T2DM in individuals who consumed 3 to 4 eggs per week, and an increase in those who consumed more than 5 eggs per week. Separation of intake by sex was also studied, illustrating that an intake of 5 eggs per week in men and above 7 eggs per week in women could increase the risk of T2DM. Opposite results were observed in other studies, as higher egg consumption was associated with a lower risk of T2DM. In systematic reviews and meta-analyses that evaluated healthy individuals, there was also no consensus on the association between egg consumption and a higher risk of cardiovascular disease or T2DM. Confounding factors between saturated fat intake and the amount of calories ingested per day (which favor weight gain and the development of metabolic syndrome) can limit study results. In any case, more long-term research is needed [62,63].
-Monounsaturated fats
Monounsaturated fatty acids have been recommended in the literature for their great capacity to reduce inflammatory response and cardiovascular risk. Two clinical trials applied the Mediterranean Diet, which is rich in food sources of monounsaturated fatty acids (extra virgin olive oil and walnut oil), and showed a reduction in the incidence of disease, cardiovascular risk, blood glucose, and body weight in T2DM [79,80].
A systematic review and meta-analysis including 1460 participants compared the effect of eating plans high in monounsaturated fat with eating plans high in carbohydrates. Eating plans high in monounsaturated fat showed significant reductions in fasting glucose, triglycerides, body weight and systolic blood pressure, along with significant increases in HDL-C. The researchers also compared eating plans rich in polyunsaturated fat with monounsaturated fat, and found a significant reduction in fasting plasma glucose in both [57].
Canola oil is another excellent source of monounsaturated fatty acids (omega 9), and unfortunately has a bad reputation due to distorted information published in the media. Moreover, it is an economically accessible source of fat, and rich in bioactive compounds. Its consumption is safe, and is assessed through the extremely rigorous processing quality control standards of regulatory bodies in all countries. A systematic review and meta-analysis published in the Journal of American College Of Nutrition analyzed the lipid profile performance of 1359 participants who ingested canola oil. The results showed that its consumption reduced total cholesterol (−7.24 mg/dL) and LDL-C (−6.4 mg/dL) compared to sunflower oil and coconut oil. There were no effects on HDL -C, triglycerides, Apo B and Apo A. These results are extremely relevant for delaying the progression of heart disease [81].
-Polyunsaturated fats
Recommendations suggest increasing intake of food sources of omega-3 polyunsaturated fatty acids (long-chain) containing a good proportion of EPA and DHA, such as salmon, tuna, mackerel, prego fish, anchovies, sardines, hake, algae, seeds, vegetable oils and others. Omega-3 and are recommended for T2DM because they have an anti-inflammatory and antioxidant effects on cytokines and low-density lipoproteins (LDL), and help to prevent heart disease. For vegetarians or vegans, omega-3 α-linoleic acid (ALA) is found in plant-based foods such as olive oil, canola oil, avocado oil, walnut oil, seeds, and algae. They are substitutes for foods high in saturated fat, and provide benefits for preventing and decreasing the risk of cardiovascular disease [16,33,82,83].
Large epidemiological studies have shown the consumption of polyunsaturated fatty acids is associated with a lower risk of T2DM [84]. Furthermore, in a randomized clinical trial with cardiac patients with T2DM, researchers demonstrated the benefits of the addition of 2 g of omega-3, twice a day, for patients on statin therapy, resulting in lower mortality rates from cardiovascular diseases [85,86].
In any case, many studies do not recommend omega-3 supplementation for the prevention or treatment of cardiovascular events and T2DM. A recent clinical trial found that omega-3 supplementation at a dose of 1 g/day did not reduce cardiovascular risk in people with T2DM [86,87,88]. Another research showed that vitamin D supplementation with 1 g of omega-3 in patients with T2DM also did not result in a lower incidence and fewer predictors of major adverse cardiovascular events [89]. Studies are inconclusive, and did not present solid evidence for the reduction of cardiovascular diseases events or mortality, but this may be useful in people who require a reduction in triglycerides and attenuation of inflammatory response [87,89].
A recent study carried out 4 g/day omega-3 supplementation in 823 participants diagnosed with T2DM. The results showed a significant reduction in cardiovascular events by 25% when compared to placebo. However, this dose is very high, which makes this dietary supplementation more expensive. Therefore, further research is needed [16,86].
-Trans fats
The use of trans fats in industry provides advantages to food factories, such as cost reduction, longer shelf life, high melting point, and wide potential for use. However, the literature shows an association between intake and an increase in systemic inflammatory response and cardiovascular risk. Furthermore, a meta-analysis showed that trans fat intake resulted in an increase in total cholesterol and LDL-C, and a decrease in HDL-C concentration [90]. Trans fats have also been associated with all-cause mortality and cardiovascular disease mortality. Therefore, the intake of trans fatty acids should be avoided and excluded from the diets of T2DM patients [13,16,63,71].

3.2.4. Fibers

The Dietary Guidelines for Americans 2020–2025 recommend that patients with T2DM consume at least 14 g of fiber per 1000 kcal (or 28 g per 2000 kcal), which must come from the consumption of grains, whole grains, vegetables, legumes, fruits and legumes (beans, peas, and lentils) [33]. Regular intake of dietary fiber is associated with reduced all-cause mortality in T2DM, and should therefore be encouraged, as it also provides additional benefits in obtaining micronutrients and phytochemicals [36,37,39]. Some studies have shown a reduction in total cholesterol, LDL cholesterol and HbA1C with an intake of 25 g of fiber per day. However, this excessive intake may cause flatulence, bloating, and diarrhea. Therefore, supplementation should occur in specific cases, and is not recommended in the long term [39,91].

3.2.5. Sodium

Many groups of Health Sciences researchers recognize that the average sodium intake of the population is greater than 3500 mg per day, and must be reduced to prevent and control hypertension [26,33,39,92,93,94]. Reducing sodium to the general recommended intake of 2300 mg/day (5 g of table salt) demonstrates positive effects on blood pressure and for patients with T2DM, especially those with hypertension and heart disease [31,95].
However, high reductions in sodium should be avoided, as some studies have shown an increased risk of mortality associated with very low sodium intake. This can be explained by the increased excretion of sodium in urine in T2DM. Therefore, sodium intake targets below 2300 mg/day should be avoided and considered individually according to dietary preference and palatability, using medical monitoring [96,97,98,99].

3.2.6. Alcohol

Guidelines suggest moderation for adults with T2DM who drink alcoholic beverages. It is recommended that healthcare professionals warn patients about the signs, symptoms and self-care of reactive hypoglycemia after drinking alcohol, especially when using hypoglycemic medications, so monitoring glucose after drinking alcoholic beverages should also be encouraged [9,16,33,63].
Some studies demonstrate that moderate alcohol consumption has a minimal, if any, effect on blood glucose in T2DM [100,101,102,103]. The maximum daily intake recommendation is defined as 15 g for women and 30 g for men. This 15 g a day may be represented by a 12-ounce (355 mL) bottle of beer, a 5-ounce (150 mL) glass of wine, or a 1.5-ounce (45 mL) portion of distilled beverages [9,33]. Excessive alcohol use (more than 3 drinks per day or 21 drinks per week for men, and more than 2 drinks per day or 14 drinks per week for women) may contribute to hyperglycemia [9,104,105].
However, studies have shown glycemic and cardiovascular benefits from moderate alcohol consumption, but this needs to be analyzed carefully, because chronic intake can put people with T2DM at risk of reactive hypoglycemia [103,106,107,108,109]. This effect may be the result of inhibition of gluconeogenesis, reduced perception of hypoglycemia due to the cerebral effects of alcohol, and reduced counterregulatory response to hypoglycemia. This is relevant for patients who use insulin secretagogues and may have a fasting hypoglycemia following alcohol consumption at night [32,33]. To minimize the risk of nocturnal hypoglycemia, it is suggested that patients eat some food when they drink alcohol [32,33,109]. It is essential that people with T2DM receive education about recognizing the symptoms and managing reactive hypoglycemia, as well as engaging in frequent blood glucose monitoring after alcohol consumption [32,33,110].
Other reviews and meta-analyses also suggest a protective effect of moderate alcohol intake on the risk of developing T2DM [101,111,112]. Moderate alcohol intake ranging from 6–48 g/day (0.5–3.4 drinks) has been associated with a 30–56% lower incidence of T2DM [32,33,101,110,111,112]. Some studies have found an intake of 20 to 30 g of alcohol per day from wine or beer to decrease the incidence of T2DM by 20% for wine and 9% for beer. However, the authors do not advise that patients who do not drink alcohol should start consuming it [113]. In any case, alcohol consumption is an individual choice, but additional factors such as history, religion, genetic factors, mental health, and drug interactions, should be considered before alcohol use [33].

3.2.7. Sweeteners

The Dietary Guidelines for Americans suggest replacing sugar-sweetened beverages (non-diet soft drinks/sodas, flavored juice drinks, sports drinks, sweetened tea, coffee drinks, energy drinks, and electrolyte replacement drinks) with water [33]. One study found that replacing sugary drinks with an equal amount of water reduced the risk of T2DM by 7–8% [114]. When low-energy sweeteners are used to reduce overall calorie and carbohydrate intake, people should be advised to avoid compensatory behaviors like additional calorie intake from other food sources [33].
Consumption of sugary drinks by the general population contributes to a significantly increased risk of T2DM, weight gain, heart disease, kidney disease, non-alcoholic liver disease, and tooth decay [115]. A meta-analysis reported that consuming one serving of a sugary drink per day increased the risk of T2DM in adults with prediabetes by 26% [116]. Another study showed that regular soda intake increased the risk of T2DM by 13%, while diet soda consumption increased the risk of T2DM by 8% [117].
The Food and Drug Administration (FDA) and Health Surveillance Agency have reviewed the safety of ingesting various types of sweeteners, approving them for consumption by the general public, including T2DM patients. The term “sweeteners” refers to high-intensity sweeteners, artificial sweeteners, non-nutritive sweeteners, and low-calorie sweeteners. These include saccharin, neotame, acesulfame-K, aspartame, sucralose, advantame, stevia, and others. Replacing table sugar and sweetened beverages with sweeteners can reduce carbohydrate and calorie intake. These dietary changes can beneficially affect blood glucose, weight, and cardiometabolic control [118,119].
Unfortunately, there is not enough evidence to determine whether the use of sweeteners could really contribute to weight loss, reduced cardiometabolic risk, and attenuation of glycemia in the long term [118]. Moreover, it may be said that the use of sweeteners is interesting, as long as individuals do not compensate for calories throughout the day. Studies conclude that sweeteners can be useful in reducing caloric intake, especially carbohydrates, but more research is needed [120,121]. Regarding adverse effects, studies have investigated hypotheses regarding (1) changing the sensation of hunger and satiety; (2) excessive use of dietary products and consequent dysregulation of the intestinal microbiota; and (3) reduced perception of calorie intake [118,122]. Therefore, for people who looking to reduce intake of sugary drinks, drinking water is more encouraged than substituting diet drinks [118].
Sugar alcohols/polyols represent a separate category of sweeteners, and have been approved by the FDA for consumption by the general public and in T2DM. Sugar alcohols have fewer calories per gram than sugars, and they are not as sweet. Therefore, a larger amount is needed to match the sweetness of the sugars, generally raising caloric content to a level similar to sugars [123]. The use of sugar alcohols should be moderate, as they can cause gastrointestinal effects in sensitive individuals. Currently, there is little research on the benefits of sugar alcohols for people with T2DM [9,124].

3.2.8. Micronutrients and Supplements

The benefits of multivitamins or mineral supplements on glycemia and cardiovascular disease risk in T2DM have little literary support, and routine use is not recommended for people who are not deficient [22,125,126]. However, metformin is associated with vitamin B12 (B12) deficiency, and annual monitoring of blood levels are recommended for people who are undergoing therapy, especially those who are anemic or have peripheral neuropathy [127], because metformin can reduce absorption of B12 [128,129,130,131].
The use of micronutrient and nutraceutical supplements, such as chromium, chromium picolinate, L-carnitine, zinc, propolis, spirulina, chorella, vitamin D, cinnamon, curcumin, aloe vera, coconut oil, cardamom oil or any other supplements to improve blood glucose in T2DM is not supported by evidence and is not recommended [9,16,48,125,126,131]. Patients who do not meet glucose targets may be at increased risk of micronutrient deficiencies [126]. Therefore, it is essential to maintain a balanced intake of food sources that provide at least the Recommended Dietary Allowance [22]. For pregnant, celiac, elderly, and vegetarian patients and those who wish to lose weight, supplementation may be fair and adjusted for each clinical case [126,132,133].
A systematic review on the effect of chromium supplementation on glucose and lipid metabolism concluded that the evidence is limited by poor-quality primary studies and heterogeneity in methodology [133,134]. Evidence from clinical studies evaluating magnesium and vitamin D supplementation to improve blood glucose in T2DM is also equally contradictory [134,135,136,137,138]. Clinical trials and meta-analyses that evaluated vitamin D supplementation to improve glycemia in T2DM have concluded that prescription is unnecessary in patients who are not deficient. Those who are deficient must be supplemented and monitored every 6 months [137,138,139,140,141,142,143,144,145,146].
Creatine has been gaining prominence in the literature within the treatment of T2DM. Evidence suggests that creatine supplementation alone or in combination with exercise training can reduce glucose intolerance and insulin resistance in T2DM. The mechanism of the effects is an increase in glucose transport into the muscle cells through translocation of glucose transporter type 4 (GLUT-4) to the sarcolemma. It is believed that creatine increases the amount and speed of translocation too. The evidence is considered promising, but more research is needed [147].
In any case, some nutritional supplements, herbal, and nutraceutical products are not yet regulated by supervisors and standard-setting bodies. Health professionals should consider the real costs, benefits, adverse effects, and drug interactions. The variability of herbal and micronutrient supplements makes research challenging, making it difficult to conclude on their effectiveness. Unfortunately, there is limited evidence supporting the use of supplements and nutraceuticals to control blood glucose [148,149].

3.3. Nutritional Strategies

Many types of dietary patterns for the nutritional management of T2DM have been studied, but the scientific community always highlights: (1) increased intake of fruits, vegetables, and legumes; (2) minimizing added sugars and refined grains; (3) choosing natural and minimally processed foods rather than ultra-processed foods [13,22,132].

3.3.1. Mediterranean Diet

The Mediterranean diet is an intervention that has the highest level of scientific evidence, and consists of foods of high nutritional quality, such as fruits, vegetables, legumes, seeds, whole grains, fish, lean meats, skimmed dairy products, and olive oil. Several epidemiological studies have reported the protective effect of this diet on metabolic disorders, chronic diseases, and mental health. This diet has good adherence in the long term, and is recommended within the prescription of individualized, flexible, and balanced eating plans that match the patient’s goals [9,16,132,150].
Studies show that the Mediterranean lifestyle with a low-fat or low-carb diet offers multiple benefits to T2DM patients [79,150,151,152,153]. A randomized controlled trial compared the Mediterranean diet (low-carb) to the low-fat diet, aiming to analyze the prevention of T2DM. The results showed that the Mediterranean diet resulted in a 30% lower relative risk compared to the low-fat diet [80]. Moreover, epidemiological studies correlate the Mediterranean, vegetarian and DASH (Diet Approach to Stop Hypertension) diets with a lower risk of developing T2DM, with no effect shown for the Ketogenic diet in the long term [115,154,155,156,157,158,159,160,161,162,163,164,165]. Large clinical trials show that a low-fat diet optimizes weight loss and improves glucose tolerance, and causes a decrease in the incidence of T2DM [151,152,160,161].
Studies have also pointed out that the Mediterranean diet may have a mixed effect on HbA1C, weight and cholesterol. In a clinical trial, obese patients with T2DM were divided in to three groups: the Mediterranean diet with calorie restriction vs. a low-fat diet with calorie restriction vs. a very low-carbohydrate diet (28% of TEV carbohydrates) with calorie restriction. The results showed that fasting glucose was lower in the Mediterranean diet than in the low-fat and very low-carb groups [165].
Another study compared the Mediterranean diet with the low-fat diet for 4 years. The results showed an improvement in the control of the glycemic profile in the Mediterranean diet, and the need for the use of hypoglycemic medications was lower too [166]. Another study showed that a Mediterranean diet with olive oil and nuts significantly reduced the incidence of cardiovascular disease in T2DM patients [29].
A randomized crossover study evaluated the impact of the Ketogenic diet vs. the Mediterranean diet on T2DM. Both diets incorporate the inclusion of non-starchy vegetables, the restriction of added sugars, and limitation of refined grains. The main differences are the consumption of legumes, fruits, and whole grains only in the Mediterranean. The authors concluded that both diets produced beneficial effects for individuals and a significant reduction in HbA1c, but the ketogenic diet increased LDL, making it impossible to rule out the potential cardiovascular risks that this change presents; in addition, it reduced intake of vitamins and minerals, which is closely correlated with the restriction of important food groups. Although controlling and reducing carbohydrate intake is beneficial and recommended for controlling prediabetes and T2DM, more studies are needed [167].

3.3.2. Dash (Dietary Approaches to Stop Hypertension)

The DASH diet is an intervention whose objective is weight loss and consequently a reduction in blood pressure. It has characteristics similar to the Mediterranean pattern, but alcohol consumption is not encouraged, and there is also sodium restriction. One study found that the DASH diet can reduce HbA1C, blood pressure, total cholesterol and weight levels in T2DM, but without significant differences in triglycerides [132,168].
Another study compared the DASH diet with conventional restriction. The results showed that blood pressure and weight were lower in the DASH group, but HbA1C and lipids showed no statistically significant differences [30,31,95]. In any case, guidelines have stated that DASH and the Mediterranean diet can be effective in the management of T2DM, especially for those who also have hypertension and coronary heart disease [16,63].

3.3.3. Low-Carb Diet and Ketogenic Diet (Very Low-Carb Diet)

Low-carb diets, especially the very low-carb (Ketogenic) diet were initially proposed for the management of patients with epilepsy and autism. Over the years, there has been interest from the scientific community in expanding these diets to treat other diseases, especially T2DM. Despite several different types of protocol that define the low-carb diet, the standard that most prevails in the literature has wide variability in the consumption of fresh and minimally processed foods, with high nutritional quality. Dietary prescription is also variable, but in general is <40% TEV carbohydrates, 20–25% TEV proteins, 30–40% TEV fats, and a limit of <10% saturated fats in most studies [9,16,132].
Part of the challenge in interpreting low-carb research is the wide range of definitions of a low-carb diet. Weight reduction is also an objective outlined in many studies, which further complicates the assessment of the distinct contribution of dietary patterns, as weight loss can also have positive effects on patients with T2DM [9]. Clinical trials have shown that this diet could reduce HbA1C and the need for antihyperglycemic medications. A meta-analysis compared the low-carb (≤45% of carbohydrate TEV) and high-carbohydrate diets (>45% of carbohydrate TEV). Both groups restricted saturated fat intake to <10% of TEV. The results showed that the benefits of improving HbA1C were more pronounced in individuals who followed the low-carb diet [168,169].
Other interventions also showed benefits in improving HbA1C in patients with T2DM who followed a Ketogenic diet (<26% of TEV in carbohydrates) for 3 and 6 months. However, the same results were not found at 12 and 24 months. The authors discuss that the major limitations of these strategies would be low adherence to the diet and the increased likelihood of long-term loss of lean mass. Food sources of carbohydrates are hyperpalatable, as are fats, and alongside providing energy substrates for physiological and metabolic functions, they also have an important emotional role. The production of ketone bodies would not be sufficient to sustain the myocyte’s energy demand during long-term muscle contraction. Therefore, researchers suggest individualized adjustments and flexibility after 3–6 months of following this type of intervention, for greater adherence to lifestyle changes [9,170].
Another meta-analysis compared the low-carb (<40% of the TEV from carbohydrates) and low-fat diets (<30% TEV from fat) for 6 months. All groups restricted saturated fat intake to <10% of the TEV. The results showed that the low-carb diet reduced HbA1C, triglycerides, blood pressure, and the use of hypoglycemic drugs, and increased HDL-C [171]. Another study compared the low-carb diet and high-carbohydrate diet (both groups restricted saturated fat intake to <10% of TEV), and the results showed a reduction in HbA1C, but this was not sustained in the long term [172].
However, the ketogenic diet is not recommended for people with T2DM who have chronic kidney disease, people with eating disorders, or women who are pregnant. More studies and literary support are needed before recommending this diet. Adopting the very low-carb diet can cause diuresis and quickly reduce blood glucose levels. Therefore, multidisciplinary team management is necessary to prevent dehydration and hypoglycemia [9,16,132].
Reducing carbohydrate intake in T2DM appears improve glucose metabolism and molecular markers, thereby becoming a viable nutritional strategy. However, studies on ketogenic diets generally indicate challenges to long-term sustainability. Therefore, it is important to reevaluate and individualize eating plan guidance regularly for those interested in this approach. It is worth remembering that insulin and other medications may need to be adjusted to prevent hypoglycemia and blood pressure [9,132].
In any case, no randomized trials have been performed with people with T2DM who increased saturated fat intake on low-carb or ketogenic diets to examine the effects on blood glucose and risk factors for cardiovascular disease [167,171,172,173,174]. Guidelines recommend the restriction of saturated fats, comprising 5–7% of the TEV for patients with T2DM [9,16,47,48,49,68]. Therefore, more evidence is needed to analyze the real efficacy, adherence, improvement of biochemical and cardiovascular parameters in the long term [167,170,171,172,173,174].

3.3.4. Low-Fat Diet

The Look AHEAD (Action for Health in Diabetes) research group showed in studies that patients who followed low-fat calorie restrictions achieved positive scores compared to a conventional calorie-restricted diet [175,176]. The evidence has shown the several benefits of a low-fat diet in improving biochemical markers (glucose, glycated hemoglobin, and LDL cholesterol), cardiovascular markers, and even quality of life [3,6,7,8,11,16]. However, according to some studies, reducing total fat intake did not improve blood glucose and cardiovascular disease risk factors in people with T2DM compared to conventional calorie restriction [177,178,179,180,181]. The benefit of the low-fat diet appears to be primarily related to weight loss [16,175,176].
The American Heart Association, the Brazilian Society of Cardiology, and the European Guidelines emphasize that low-fat diets, especially a Mediterranean or vegetarian dietary patterns, are very important for adapting the consumption of fats and fiber by the population; they are potentially efficient strategies, especially for patients who have increased LDL cholesterol. Currently, individuals consume a large amount of fat (>36–46% of TEV), and this excess has important consequences, in particular increased cardiovascular risk and weight gain. Therefore, this strategy is also viable for the management of T2DM [16,60,61,62,63,64,74,75,132].

3.3.5. Ornish and Pritikin Diets (Very Low-Fat Diet)

Both of these types of diets are very low-fat eating patterns. The Ornish program emphasizes a low-fat, whole-food, plant-based eating plan (70% TEV carbohydrates, 10% TEV fat, 20% TEV protein, and 60 g fiber per day) predominantly made up of vegetables, legumes, fruits, grains, skimmed dairy, and egg whites [182,183,184]. In the same way, the Pritikin diet recommends the consumption of 77% carbohydrates, 10% fat, 13% protein within the TEV, and 30–40 g of fiber without caloric restriction during a 26-day stay in a treatment center (SPA). A meta-analysis with 652 participants showed that these diets can have positive effects on glucose levels, weight, blood pressure, and HDL-C in the long term [182,183,184,185]. Unfortunately, these studies are old, and interest in studying these types of interventions has reduced over time, so more evidence is needed [16,132].

3.3.6. Vegetarian and Vegan Diet (Plant-Based Diet)

Studies of vegetarian or vegan eating plans over 12 to 74 weeks have demonstrated positive results on blood glucose, risk of cardiovascular disease, and weight loss. Two meta-analyses concluded that vegetarian and vegan diets can reduce HbA1C, weight, waist circumference and LDL-C in patients with T2DM. No significant effects were found on HDL-C, insulin, triglycerides, and blood pressure [16,132,184,185,186,187,188,189,190,191].
Studies have demonstrated the benefits of plant-based diets in treating T2DM and reducing macro and microvascular complications. The type and source of carbohydrates (unrefined vs. refined), fats (monounsaturated and polyunsaturated vs. saturated and trans), and proteins (plant vs. animal) play an important role in preventing and controlling disease, and have multiple benefits such as improving insulin resistance, reducing body weight, increasing fiber and phytonutrients, increasing interactions between food and the microbiome, decreasing saturated fat, and advancing glycation end-products and nitrosamines [16,132,184,185,186,187,188,189,190,191].

3.3.7. Paleolithic Diet (Paleo Diet)

The Paleolithic diet is an intervention made to mimic the eating habits of our ancestors. Unfortunately, Westernization makes it difficult to follow this strategy, and it is not applicable in the 21st century. It is worth remembering the individuals who lived in the Paleozoic era were hunters, gatherers, and nomadic peoples. They consumed raw meat, fruits, vegetables, and roots, and they survived from what nature could offer and spent long periods fasting [192,193,194].
This lifestyle has become impossible, especially due to eating raw meat and high levels of food contamination. Studies using the Paleolithic diet in T2DM are scarce. They have a short duration (3 months), few participants (n = 13–29), and other methodological limitations. The most studies findings found mixed effects on HbA1C, weight, and lipids [193,194]. Due to the lack of literature, this dietary pattern is not recommended by guidelines [16,47,132].

3.3.8. Intermittent Fasting

Intermittent fasting is not an eating pattern by definition, but it was included in this literature review due to increased interest from the scientific community. Fasting means going without eating and drinking and abstaining from foods and drinks that contain macronutrients and calories. People fast for any reasons, such as rapid weight loss, and religious or spiritual practices. Intermittent fasting is a strategy that focuses more on time, i.e., “when” you eat, rather than “what” you eat. Daily caloric intake occurs within a defined dietary window during the day [195].
Studies on intermittent fasting with T2DM demonstrate a variety of protocols: (1) restriction of food intake for 18–20 h a day; (2) fasting all day (24 h fasting with days of normocaloric intake); and (3) severe calorie restriction (intake < 1000 kcal/day) for up to 8 consecutive days or more [196]. Some clinical trials with few participants (≤63 participants) and a short duration (≤20 weeks) have demonstrated that fasting protocols on consecutive days with severe caloric restriction (<1000 kcal/day) and fasting > 16 h per day can result in weight loss in T2DM. However, there were no improvements in HbA1C compared with conventional calorie restriction. One of the studies showed modest reductions in HbA1C, weight, and medication doses when patients underwent 2 days of severe energy restriction compared to conventional energy restriction [195,196,197].
A study has analyzed the applicability of fasting in patients with pre-diabetes. The individuals were divided into two groups: (1) intervention with a dietary window of just 6 h (last meal at 3 p.m.); and (2) a control group with a 12 h food window. The results showed improved insulin sensitivity, greater responsiveness of β cells, and a reduction in blood pressure and oxidative stress in the intervention group. However, the study did not control diet and exercise [197].
The results of intermittent fasting in T2DM are still controversial, and have limitations (short-term studies, few participants, low long-term adherence, lack of control over diet and physical exercise). Therefore, its practice requires great caution, especially for patients who use hypoglycemic medications [197]. The safety of fasting in other specific subgroups, including pregnant women and patients with eating disorders, has not yet been studied and is also not recommended [16,47,132].

3.3.9. Mindful Eating Program

The ‘Mindful Eating Program’ has also shown strong evidence for the treatment of different diseases, even T2DM. This therapy mixes the science of nutrition, body awareness, and self-control. Studies have demonstrated significant improvement in eating regulation in T2DM, as prioritizing self-control can be a great alternative to severely restrictive diets. The practices can also help to increase knowledge of the factors (physiological, environmental, or emotional) that dictate excess food intake and train patients in intuitive eating (respecting hunger/satiety signals and chewing food). The program not only emphasizes the quantity and quality eaten, but also the enjoyment of the experience, thus changing the reward value of the food without restricting it [198,199].
The Mindful Eating intervention has been gaining increasing prominence in the scientific community, as it is closely linked to mental health care [198,199]. The American Diabetes Association emphasizes that the psychological factors of patients with T2DM must be respected and valued by health professionals. Therefore, it is extremely important to welcome and understand what happens on an emotional, physiological and metabolic level. More evidence is needed on applicability in T2DM in the long term [16,132,198,199].

3.4. Summary of Dietary Patterns for the Management of T2DM

Table 1 below summarizes all descriptions of dietary patterns, their level of evidence, adherence, and comments.

3.5. Nutritional Management of T2DM in the COVID-19 Pandemic

The COVID-19 lockdown clearly affected the lifestyle of the population and entailed changes in their daily habits. These transformations involved potential health consequences, especially in T2DM. Recent studies have examined the impact of lockdown on eating habits, exercise, and the psychological effects in T2DM, and showed that there was an increase in the consumption of foods high in sugar and sandwiches during the pandemic. An association between food craving levels and sandwich consumption was also found. The research also showed a high prevalence of physical inactivity before lockdown, which was even higher during home confinement. These findings emphasize the great importance of developing new research with larger samples, aiming to expand public health policies that promote a healthy lifestyle in T2DM, especially after lockdown [200].
However, another study shows the more than 60% of participants with T2DM and T1DM started eating more nutritious meals at the pandemic, and increased their personal and environmental hygiene habits, especially through the use of hand sanitizers. Additionally, 40% of all respondents stated that their glycemic self-management had improved. These positive results may be related to patients’ fear of knowing that they belong to a risk group, thus having a high probability of suffering with COVID-19. However, the data collection has limitations, because they were obtained through patient reports [201].
Another study found no changes in physical activity and adherence to diet in more than 80% of participants. There was an increase in the consumption of vegetables (81%) and fruits (43%), and a decrease unhealthy snacks (63%). No significant changes were observed in HbA1c or body weight before and after lockdown. The majority watched television and spent time with their families. However, the quality of food intake and glycemic control worsened in those with mental stress, insufficient sleep and physical inactivity [202]. Therefore, it is concluded that measures to promote healthy lifestyle practices, along with ways to reduce psychosocial stress, should be implemented to better management T2DM after the pandemic [200,202].

4. Authors’ Comments—The Best Nutritional Strategies and Dietary Prescription Recommended for the Management of T2DM

Based on the data analyzed, the Mediterranean Diet is the nutritional strategy with the highest level of evidence in the literature, and can bring greater benefits to patients with T2DM in the long term. The best recommended dietary prescription is composed of 40–50% carbohydrates, 15–25% proteins, 25–35% fats (<7% saturated, 10% polyunsaturated, and 10% monounsaturated), at least 14 g of fiber for each 1000 kcal consumed, and <2300 mg of sodium. In any case, it is necessary to individualize the diet based on the stage of the disease, the patient’s goals, socioeconomic situation, and preferences, aiming for greater adherence and significant changes in lifestyle.

5. Conclusions

There is no consensus on what the ideal nutritional strategy and percentages of calories, carbohydrates, proteins, and fats for patients with T2DM. Therefore, the type of strategy and distribution of macronutrients should be based on an individualized assessment of current eating patterns, preferences, and metabolic goals. However, several references show that Mediterranean diet may bring greater benefits in the long term, with the following recommended dietary prescription: 40–50% carbohydrates; 15–25% proteins; 25–35% fats (<7% saturated, 10% polyunsaturated, and 10% monounsaturated); at least 14 g of fiber for every 1000 kcal consumed; and <2300 mg sodium.

Author Contributions

All individuals contributed significantly to the conception, design, and data collection of the work, and participated sufficiently in the writing of this critical review article to establish ownership of the intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financed in part by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—Brasil (CAPES)—Finance Code 001.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to the State Faculty of Medicine of São José do Rio Preto (FAMERP), State University of Campinas (UNICAMP), and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—Brasil (CAPES) for making this work possible.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Moulis, G.; Ibañez, B.; Palmaro, A.; Aizpuru, F.; Millan, E.; Lapeyre-Mestre, M.; Sailler, L.; Cambra, K. Cross-national health care database utilization between Spain and France: Results from the EPICHRONIC study assessing the prevalence of type 2 diabetes mellitus. Clin. Epidemiol. 2018, 10, 863–874. [Google Scholar] [CrossRef]
  2. Al-Jawaldeh, A.; Hammerich, A.; Doggui, R.; Engesveen, K.; Lang, K.; McColl, K. Implementation of WHO Recommended Policies and Interventions on Healthy Diet in the Countries of the Eastern Mediterranean Region: From Policy to Action. Nutrients 2020, 12, 3700. [Google Scholar] [CrossRef]
  3. Berger, S.E.; Huggins, G.S.; McCaffery, J.M.; Jacques, P.F.; Lichtenstein, A.H. Change in Cardiometabolic Risk Factors Associated with Magnitude of Weight Regain 3 Years after a 1-Year Intensive Lifestyle Intervention in Type 2 Diabetes Mellitus: The Look AHEAD Trial. J. Am. Heart Assoc. 2019, 8, e010951. [Google Scholar] [CrossRef]
  4. Magliano, D.J.; Boyko, E.J. IDF Diabetes Atlas 10th Edition Scientific Committee, 10th ed.; International Diabetes Federation: Brussels, Belgium, 2021. Available online: https://www.ncbi.nlm.nih.gov/books/NBK581934/ (accessed on 20 August 2023).
  5. Belalcazar, L.M.; Ballantyne, C.M. Looking Back at Look AHEAD through the Lens of Recent Diabetes Outcome Trials. Circulation 2017, 135, 720–723. [Google Scholar] [CrossRef]
  6. Berger, S.E.; Huggins, G.S.; McCaffery, J.M.; Lichtenstein, A.H. Comparison among criteria to define successful weight-loss maintainers and regainers in the Action for Health in Diabetes (Look AHEAD) and Diabetes Prevention Program trials. Am. J. Clin. Nutr. 2017, 106, 1337–1346. [Google Scholar] [CrossRef]
  7. Look AHEAD Research Group. Impact of intensive lifestyle intervention on depression and health-related quality of life in type 2 diabetes: The Look AHEAD Trial. Diabetes Care 2014, 37, 1544–1553. [Google Scholar] [CrossRef] [PubMed]
  8. Look AHEAD Research Group. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N. Engl. J. Med. 2013, 369, 145–154, Erratum in N. Engl. J. Med. 2014, 370, 1866. [Google Scholar] [CrossRef]
  9. ElSayed, N.A.; Aleppo, G.; Aroda, V.R.; Bannuru, R.R.; Brown, F.M.; Bruemmer, D.; Collins, B.S.; Hilliard, M.E.; Isaacs, D.; Johnson, E.L.; et al. 2. Classification and Diagnosis of Diabetes: Standards of Care in Diabetes-2023. Diabetes Care 2023, 46 (Suppl. S1), S19–S40, Erratum in Diabetes Care 2023, 46, 1715. [Google Scholar] [CrossRef]
  10. Apolzan, J.W.; Venditti, E.M.; Edelstein, S.L.; Knowler, W.C.; Dabelea, D.; Boyko, E.J.; Pi-Sunyer, X.; Kalyani, R.R.; Franks, P.W.; Srikanthan, P.; et al. Long-Term Weight Loss with Metformin or Lifestyle Intervention in the Diabetes Prevention Program Outcomes Study. Ann. Intern. Med. 2019, 170, 682–690, Erratum in Ann. Intern. Med. 2020, 173, 508. [Google Scholar] [CrossRef]
  11. Gregg, E.W.; Jakicic, J.M.; Blackburn, G.; Bloomquist, P.; Bray, G.A.; Clark, J.M. Look AHEAD Research Group. Association of the magnitude of weight loss and changes in physical fitness with long-term cardiovascular disease outcomes in overweight or obese people with type 2 diabetes: A post-hoc analysis of the Look AHEAD randomised clinical trial. Lancet Diabetes Endocrinol. 2016, 4, 913–921. [Google Scholar] [CrossRef]
  12. Williams, B.; Mancia, G.; Spiering, W.; Agabiti Rosei, E.; Azizi, M.; Burnier, M.; Clement, D.L.; Coca, A.; de Simone, G.; Dominiczak, A.; et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension: The Task Force for the management of arterial hypertension of the European Society of Cardiology and the European Society of Hypertension. J. Hypertens. 2018, 36, 1953–2041, Erratum in J. Hypertens. 2019, 37, 226. [Google Scholar] [CrossRef]
  13. Brazil, M.d.S. Food Guide for the Brazilian Population Promoting Healthy Eating. Standards and Technical Manuals. 2014, Brasília, 1–158. Available online: http://www.cfn.org.br/wp-content/uploads/2015/12/guia-alimentar-da-populacaobrasileira.pdf (accessed on 20 August 2023).
  14. Ivers, N.M.; Jiang, M.; Alloo, J.; Singer, A.; Ngui, D.; Casey, C.G.; Yu, C.H. Diabetes Canada 2018 clinical practice guidelines: Key messages for family physicians caring for patients living with type 2 diabetes. Can. Fam. Physician 2019, 65, 14–24. [Google Scholar]
  15. Connor, H.; Annan, F.; Bunn, E.; Frost, G.; McGough, N.; Sarwar, T.; Thomas, B. Nutrition Subcommittee of the Diabetes Care Advisory Committee of Diabetes, U.K. The implementation of nutritional advice for people with diabetes. Diabet. Med. 2003, 20, 786–807, Erratum in Diabet Med. 2004, 21, 200. [Google Scholar] [CrossRef]
  16. Marx, N.; Federici, M.; Schütt, K.; Müller-Wieland, D.; Ajjan, R.A.; Antunes, M.J.; Christodorescu, R.M.; Crawford, C.; Di Angelantonio, E.; Eliasson, B.; et al. ESC Scientific Document Group; 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes. Eur. Heart J. 2023, 44, 4043–4140, Erratum in Eur. Heart J. 2023, 44, 4043–4140. [Google Scholar] [CrossRef] [PubMed]
  17. Diabetes Prevention Program Research Group. 10-year follow-up of diabetes incidence and weight loss in the Diabetes Prevention Program Outcomes Study. Lancet 2009, 374, 1677–1686, Erratum in Lancet 2009, 374, 2054. [Google Scholar] [CrossRef]
  18. Li, G.; Zhang, P.; Wang, J.; Gregg, E.W.; Yang, W.; Gong, Q.; Li, H.; Li, H.; Jiang, Y.; An, Y.; et al. The long-term effect of lifestyle interventions to prevent diabetes in the China Da Qing Diabetes Prevention Study: A 20-year follow-up study. Lancet 2008, 371, 1783–1789. [Google Scholar] [CrossRef] [PubMed]
  19. Lindström, J.; Ilanne-Parikka, P.; Peltonen, M.; Aunola, S.; Eriksson, J.G.; Hemiö, K.; Hämäläinen, H.; Härkönen, P.; Keinänen-Kiukaanniemi, S.; Laakso, M.; et al. Sustained reduction in the incidence of type 2 diabetes by lifestyle intervention: Follow-up of the Finnish Diabetes Prevention Study. Lancet 2006, 368, 1673–1679. [Google Scholar] [CrossRef]
  20. Diabetes Prevention Program Research Group. Long-term effects of lifestyle intervention or metformin on diabetes development and microvascular complications over 15-year follow-up: The Diabetes Prevention Program Outcomes Study. Lancet Diabetes Endocrinol. 2015, 3, 866–875. [Google Scholar] [CrossRef]
  21. Li, G.; Zhang, P.; Wang, J.; An, Y.; Gong, Q.; Gregg, E.W.; Yang, W.; Zhang, B.; Shuai, Y.; Hong, J.; et al. Cardiovascular mortality, all-cause mortality, and diabetes incidence after lifestyle intervention for people with impaired glucose tolerance in the Da Qing Diabetes Prevention Study: A 23-year follow-up study. Lancet Diabetes Endocrinol. 2014, 2, 474–480. [Google Scholar] [CrossRef]
  22. Evert, A.B.; Dennison, M.; Gardner, C.D.; Garvey, W.T.; Lau, K.H.K.; MacLeod, J.; Mitri, J.; Pereira, R.F.; Rawlings, K.; Robinson, S.; et al. Nutrition Therapy for Adults with Diabetes or Prediabetes: A Consensus Report. Diabetes Care 2019, 42, 731–754. [Google Scholar] [CrossRef]
  23. Carbone, S.; Dixon, D.L.; Buckley, L.F.; Abbate, A. Glucose-Lowering Therapies for Cardiovascular Risk Reduction in Type 2 Diabetes Mellitus: State-of-the-Art Review. Mayo Clin. Proc. 2018, 93, 1629–1647, Erratum in Mayo Clin. Proc. 2019, 94, 554. [Google Scholar] [CrossRef]
  24. Franz, M.J.; Bantle, J.P.; Beebe, C.A.; Brunzell, J.D.; Chiasson, J.L.; Garg, A.; Holzmeister, L.A.; Hoogwerf, B.; Mayer-Davis, E.; Mooradian, A.D.; et al. American Diabetes Association. Evidence-based nutrition principles and recommendations for the treatment and prevention of diabetes and related complications. Diabetes Care 2003, 26 (Suppl. S1), S51–S61. [Google Scholar] [CrossRef]
  25. Sartorelli, D.S.; Sciarra, E.C.; Franco, L.J.; Cardoso, M.A. Primary prevention of type 2 diabetes through nutritional counseling. Diabetes Care 2004, 27, 3019. [Google Scholar] [CrossRef]
  26. World Health Organization. Guideline: Sodium Intake for Adults and Children. 2012. Available online: http://www.ncbi.nlm.nih.gov/books/NBK133309/ (accessed on 20 September 2023).
  27. Inzucchi, S.E.; Bergenstal, R.M.; Buse, J.B.; Diamant, M.; Ferrannini, E.; Nauck, M.; Peters, A.L.; Tsapas, A.; Wender, R.; Matthews, D.R.; et al. Management of hyperglycemia in type 2 diabetes, 2015: A patient-centered approach: Update to a position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2015, 38, 140–149. [Google Scholar] [CrossRef]
  28. Powers, M.A.; Bardsley, J.; Cypress, M.; Duker, P.; Funnell, M.M.; Fischl, A.H.; Maryniuk, M.D.; Siminerio, L.; Vivian, E. Diabetes Self-Management Education and Support in Type 2 Diabetes: A Joint Position Statement of the American Diabetes Association, the American Association of Diabetes Educators, and the Academy of Nutrition and Dietetics. J. Acad. Nutr. Diet. 2015, 115, 1323–1334. [Google Scholar] [CrossRef]
  29. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. PREDIMED Study Investigators. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N. Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef] [PubMed]
  30. Drehmer, M.; Odegaard, A.O.; Schmidt, M.I.; Duncan, B.B.; Cardoso, L.O.; Matos, S.M.A.; Molina, M.D.C.B.; Barreto, S.M.; Pereira, M.A. Brazilian dietary patterns and the dietary approaches to stop hypertension (DASH) diet-relationship with metabolic syndrome and newly diagnosed diabetes in the ELSA-Brasil study. Diabetol. Metab. Syndr. 2017, 9, 13. [Google Scholar] [CrossRef]
  31. Paula, T.P.; Viana, L.V.; Neto, A.T.; Leitão, C.B.; Gross, J.L.; Azevedo, M.J. Effects of the DASH Diet and Walking on Blood Pressure in Patients with Type 2 Diabetes and Uncontrolled Hypertension: A Randomized Controlled Trial. J. Clin. Hypertens. 2015, 17, 895–901. [Google Scholar] [CrossRef] [PubMed]
  32. Franz, M.J.; MacLeod, J.; Evert, A.; Brown, C.; Gradwell, E.; Handu, D.; Reppert, A.; Robinson, M. 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. [Google Scholar] [CrossRef] [PubMed]
  33. Snetselaar, L.G.; de Jesus, J.M.; DeSilva, D.M.; Stoody, E.E. Dietary Guidelines for Americans, 2020-2025: Understanding the Scientific Process, Guidelines, and Key Recommendations. Nutr. Today 2021, 56, 287–295. [Google Scholar] [CrossRef]
  34. Davidson, P.; Ross, T.; Castor, C. Academy of Nutrition and Dietetics: Revised 2017 Standards of Practice and Standards of Professional Performance for Registered Dietitian Nutritionists (Competent, Proficient, and Expert) in Diabetes Care. J. Acad. Nutr. Die. 2018, 118, 932–946.e48. [Google Scholar] [CrossRef]
  35. Briggs, E.K.; Stanley, K. Position of the Academy of Nutrition and Dietetics: The Role of Medical Nutrition Therapy and Registered Dietitian Nutritionists in the Prevention and Treatment of Prediabetes and Type 2 Diabetes. J. Acad. Nutr. Diet 2018, 118, 343–353. [Google Scholar] [CrossRef] [PubMed]
  36. He, M.; van Dam, R.M.; Rimm, E.; Hu, F.B.; Qi, L. Whole-grain, cereal fiber, bran, and germ intake and the risks of all-cause and cardiovascular disease-specific mortality among women with type 2 diabetes mellitus. Circulation 2010, 121, 2162–2168. [Google Scholar] [CrossRef] [PubMed]
  37. Burger, K.N.; Beulens, J.W.; van der Schouw, Y.T.; Sluijs, I.; Spijkerman, A.M.; Sluik, D.; Boeing, H.; Kaaks, R.; Teucher, B.; Dethlefsen, C.; et al. Dietary fiber, carbohydrate quality and quantity, and mortality risk of individuals with diabetes mellitus. PLoS ONE 2012, 7, e43127. [Google Scholar] [CrossRef] [PubMed]
  38. Jenkins, D.J.; Kendall, C.W.; Augustin, L.S.; Mitchell, S.; Sahye-Pudaruth, S.; Blanco Mejia, S.; Chiavaroli, L.; Mirrahimi, A.; Ireland, C.; Bashyam, B.; 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. [Google Scholar] [CrossRef] [PubMed]
  39. Trumbo, P.; Schlicker, S.; Yates, A.A.; Poos, M.; Food and Nutrition Board of the Institute of Medicine; The National Academies. Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty acids, cholesterol, protein and amino acids. J. Am. Diet. Assoc. 2002, 102, 1621–1630, Erratum in J. Am. Diet Assoc. 2003, 103, 563. [Google Scholar] [CrossRef] [PubMed]
  40. Vega-López, S.; Venn, B.J.; Slavin, J.L. Relevance of the Glycemic Index and Glycemic Load for Body Weight, Diabetes, and Cardiovascular Disease. Nutrients 2018, 10, 1361. [Google Scholar] [CrossRef]
  41. Evert, A.B.; Boucher, J.L.; Cypress, M.; Dunbar, S.A.; Franz, M.J.; Mayer-Davis, E.J.; Neumiller, J.J.; Nwankwo, R.; Verdi, C.L.; Urbanski, P.; et al. Nutrition therapy recommendations for the management of adults with diabetes. Diabetes Care 2014, 37 (Suppl. S1), S120–S143. [Google Scholar] [CrossRef]
  42. Oza-Frank, R.; Cheng, Y.J.; Narayan, K.M.; Gregg, E.W. Trends in nutrient intake among adults with diabetes in the United States: 1988–2004. J. Am. Diet Assoc. 2009, 109, 1173–1178. [Google Scholar] [CrossRef]
  43. Brand-Miller, J.C.; Stockmann, K.; Atkinson, F.; Petocz, P.; Denyer, G. Glycemic index, postprandial glycemia, and the shape of the curve in healthy subjects: Analysis of a database of more than 1,000 foods. Am. J. Clin. Nutr. 2009, 89, 97–105. [Google Scholar] [CrossRef]
  44. Wheeler, M.L.; Dunbar, S.A.; Jaacks, L.M.; Karmally, W.; Mayer-Davis, E.J.; Wylie-Rosett, J.; Yancy, W.S., Jr. Macronutrients, food groups, and eating patterns in the management of diabetes: A systematic review of the literature, 2010. Diabetes Care 2012, 35, 434–445. [Google Scholar] [CrossRef]
  45. Zeevi, D.; Korem, T.; Zmora, N.; Israeli, D.; Rothschild, D.; Weinberger, A.; Ben-Yacov, O.; Lador, D.; Avnit-Sagi, T.; Lotan-Pompan, M.; et al. Personalized Nutrition by Prediction of Glycemic Responses. Cell 2015, 163, 1079–1094. [Google Scholar] [CrossRef]
  46. Shukla, A.P.; Dickison, M.; Coughlin, N.; Karan, A.; Mauer, E.; Truong, W.; Casper, A.; Emiliano, A.B.; Kumar, R.B.; Saunders, K.H.; et al. The impact of food order on postprandial glycaemic excursions in prediabetes. Diabetes Obes. Metab. 2019, 21, 377–381. [Google Scholar] [CrossRef]
  47. Brazilian Diabetes Society (SBD). Official Position on Therapeutic Conduct for People with Diabetes and Hypertension. Brazilian Diabetes Society (SBD). N03/2020. Arq. Bras Cardiol. 2020, 4, 1–20. Available online: https://profissional.diabetes.org.br/wp-content/uploads/2021/06/CONDUTA-TERAPEUTICA-HIPERTENSAO-ARTERIAL.pdf (accessed on 20 September 2023).
  48. Official Position of SBEM and ABESO on the Use of Coconut Oil to Lose Weight. Brazilian Society of Endocrinology and Metabology (SBEM); Brazilian Association for the Study of Obesity and Metabolic Syndrome (ABESO & SBEM). 2020. Available online: https://www.endocrino.org.br/media/uploads/posicionamento_oficial_%C3%B3leo_de_coco_sbem_e_abeso.pdf (accessed on 20 September 2023).
  49. Brazilian Diabetes Society. Carbohydrate Counting Manual; Department of Nutrition of the Brazilian Diabetes Society (SBD): Rio De Janeiro, Brazil, 2021; Available online: https://diabetes.org.br/wp-content/uploads/2021/05/manual-de-contagem-de-carbo.pdf (accessed on 20 September 2023).
  50. Gross, J.L.; Zelmanovitz, T.; Moulin, C.C.; De Mello, V.; Perassolo, M.; Leitão, C.; Hoefel, A.; Paggi, A.; Azevedo, M.J. Effect of a chicken-based diet on renal function and lipid profile in patients with type 2 diabetes: A randomized crossover trial. Diabetes Care 2002, 25, 645–651. [Google Scholar] [CrossRef] [PubMed]
  51. Fuller, N.R.; Caterson, I.D.; Sainsbury, A.; Denyer, G.; Fong, M.; Gerofi, J.; Baqleh, K.; Williams, K.H.; Lau, N.S.; Markovic, T.P. The effect of a high-egg diet on cardiovascular risk factors in people with type 2 diabetes: The Diabetes and Egg (DIABEGG) study—A 3-mo randomized controlled trial. Am. J. Clin. Nutr. 2015, 101, 705–713. [Google Scholar] [CrossRef]
  52. Malik, V.S.; Li, Y.; Tobias, D.K.; Pan, A.; Hu, F.B. Dietary Protein Intake and Risk of Type 2 Diabetes in US Men and Women. Am. J. Epidemiol. 2016, 183, 715–728. [Google Scholar] [CrossRef] [PubMed]
  53. Vuksan, V.; Jenkins, A.L.; Brissette, C.; Choleva, L.; Jovanovski, E.; Gibbs, A.L.; Bazinet, R.P.; Au-Yeung, F.; Zurbau, A.; Ho, H.V.; et al. Salba-chia (Salvia hispanica L.) in the treatment of overweight and obese patients with type 2 diabetes: A double-blind randomized controlled trial. Nutr. Metab. Cardiovasc. Dis. 2017, 27, 138–146. [Google Scholar] [CrossRef]
  54. Luger, M.; Holstein, B.; Schindler, K.; Kruschitz, R.; Ludvik, B. Feasibility and efficacy of an isocaloric high-protein vs. standard diet on insulin requirement, body weight and metabolic parameters in patients with type 2 diabetes on insulin therapy. Exp. Clin. Endocrinol. Diabetes 2013, 121, 286–294. [Google Scholar] [CrossRef]
  55. Dong, J.Y.; Zhang, Z.L.; Wang, P.Y.; Qin, L.Q. 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. [Google Scholar] [CrossRef]
  56. Pfeiffer, A.F.H.; Pedersen, E.; Schwab, U.; Risérus, U.; Aas, A.M.; Uusitupa, M.; Thanopoulou, A.; Kendall, C.; Sievenpiper, J.L.; Kahleová, H.; et al. The Effects of Different Quantities and Qualities of Protein Intake in People with Diabetes Mellitus. Nutrients 2020, 12, 365. [Google Scholar] [CrossRef] [PubMed]
  57. Qian, F.; Korat, A.A.; Malik, V.; Hu, F.B. 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. [Google Scholar] [CrossRef] [PubMed]
  58. Bendsen, N.T.; Christensen, R.; Bartels, E.M.; Astrup, A. Consumption of industrial and ruminant trans fatty acids and risk of coronary heart disease: A systematic review and meta-analysis of cohort studies. Eur. J. Clin. Nutr. 2011, 65, 773–783. [Google Scholar] [CrossRef] [PubMed]
  59. Berger, S.; Raman, G.; Vishwanathan, R.; Jacques, P.F.; Johnson, E.J. Dietary cholesterol and cardiovascular disease: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2015, 102, 276–294. [Google Scholar] [CrossRef]
  60. Grundy, S.M.; Stone, N.J.; Bailey, A.L.; Beam, C.; Birtcher, K.K.; Blumenthal, R.S.; Braun, L.T.; de Ferranti, S.; Faiella-Tommasino, J.; Forman, D.E.; et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 2019, 73, 3168–3209, Erratum in J. Am. Coll. Cardiol. 2019, 73, 3234–3237. [Google Scholar] [CrossRef] [PubMed]
  61. Eckel, R.H.; Jakicic, J.M.; Ard, J.D.; de Jesus, J.M.; Houston Miller, N.; Hubbard, V.S.; Lee, I.M.; Lichtenstein, A.H.; Loria, C.M.; Millen, B.E.; et al. American College of Cardiology/American Heart Association Task Force on Practice Guidelines. 2013 AHA/ACC guideline on lifestyle management to reduce cardiovascular risk: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation 2014, 129 (Suppl. S2), S76–S99, Erratum in Circulation 2015, 131, e326. [Google Scholar] [CrossRef]
  62. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A.; et al. ESC Scientific Document Group. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Eur. Heart J. 2020, 41, 111–188, Erratum in Eur. Heart J. 2020, 41, 4255. [Google Scholar] [CrossRef]
  63. Izar, M.C.O.; Lottenberg, A.M.; Giraldez, V.Z.R.; Santos Filho, R.D.D.; Machado, R.M.; Bertolami, A.; Assad, M.H.V.; Saraiva, J.F.K.; Faludi, A.A.; Moreira, A.S.B.; et al. Position Statement on Fat Consumption and Cardiovascular Health—2021. Arq. Bras. Cardiol. 2021, 116, 160–212, Erratum in Arq. Bras. Cardiol. 2021, 116, 855. [Google Scholar] [CrossRef]
  64. Guasch-Ferré, M.; Becerra-Tomás, N.; Ruiz-Canela, M.; Corella, D.; Schröder, H.; Estruch, R.; Ros, E.; Arós, F.; Gómez-Gracia, E.; Fiol, M.; et al. Total and subtypes of dietary fat intake and risk of type 2 diabetes mellitus in the Prevención con Dieta Mediterránea (PREDIMED) study. Am. J. Clin. Nutr. 2017, 105, 723–735. [Google Scholar] [CrossRef]
  65. Salazar, J.; Angarita, L.; Morillo, V.; Navarro, C.; Martínez, M.S.; Chacín, M.; Torres, W.; Rajotia, A.; Rojas, M.; Cano, C.; et al. Microbiota and Diabetes Mellitus: Role of Lipid Mediators. Nutrients 2020, 12, 3039. [Google Scholar] [CrossRef]
  66. McNamara, D.J. Dietary cholesterol, heart disease risk and cognitive dissonance. Proc. Nutr. Soc. 2014, 73, 161–166. [Google Scholar] [CrossRef]
  67. Guasch-Ferré, M.; Babio, N.; Martínez-González, M.A.; Corella, D.; Ros, E.; Martín-Peláez, S.; Estruch, R.; Arós, F.; Gómez-Gracia, E.; Fiol, M.; et al. PREDIMED Study Investigators. Dietary fat intake and risk of cardiovascular disease and all-cause mortality in a population at high risk of cardiovascular disease. Am. J. Clin. Nutr. 2015, 102, 1563–1573. [Google Scholar] [CrossRef] [PubMed]
  68. Mensink, R.P. Effects of Saturated Fatty Acids on Serum Lipids and Lipoproteins: A Systematic Review and Regression Analysis; World Health Organization: Geneva, Switzerland, 2016. [Google Scholar]
  69. Sacks, F.M.; Lichtenstein, A.H.; Wu, J.H.Y.; Appel, L.J.; Creager, M.A.; Kris-Etherton, P.M.; Miller, M.; Rimm, E.B.; Rudel, L.L.; Robinson, J.G.; et al. American Heart Association. Dietary Fats and Cardiovascular Disease: A Presidential Advisory from the American Heart Association. Circulation 2017, 136, e1–e23, Erratum in Circulation 2017, 136, e195. [Google Scholar] [CrossRef]
  70. Hooper, L.; Martin, N.; Jimoh, O.F.; Kirk, C.; Foster, E.; Abdelhamid, A.S. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst. Rev. 2020, 8, CD011737. [Google Scholar] [CrossRef] [PubMed]
  71. de Souza, R.J.; Mente, A.; Maroleanu, A.; Cozma, A.I.; Ha, V.; Kishibe, T.; Uleryk, E.; Budylowski, P.; Schünemann, H.; Beyene, J.; 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. [Google Scholar] [CrossRef]
  72. Dehghan, M.; Mente, A.; Zhang, X.; Swaminathan, S.; Li, W.; Mohan, V.; Iqbal, R.; Kumar, R.; Wentzel-Viljoen, E.; Rosengren, A.; et al. Prospective Urban Rural Epidemiology (PURE) study investigators. Associations of fats and carbohydrate intake with cardiovascular disease and mortality in 18 countries from five continents (PURE): A prospective cohort study. Lancet 2017, 390, 2050–2062. [Google Scholar] [CrossRef]
  73. McGuire, S. Scientific Report of the 2015 Dietary Guidelines Advisory Committee. Washington, DC: US Departments of Agriculture and Health and Human Services, 2015. Adv. Nutr. 2016, 7, 202–204. [Google Scholar] [CrossRef] [PubMed]
  74. Neelakantan, N.; Seah, J.Y.H.; van Dam, R.M. The Effect of Coconut Oil Consumption on Cardiovascular Risk Factors: A Systematic Review and Meta-Analysis of Clinical Trials. Circulation 2020, 141, 803–814. [Google Scholar] [CrossRef]
  75. Sacks, F.M. Coconut Oil and Heart Health: Fact or Fiction? Circulation 2020, 141, 815–817. [Google Scholar] [CrossRef]
  76. Gijsbers, L.; Ding, E.L.; Malik, V.S.; de Goede, J.; Geleijnse, J.M.; Soedamah-Muthu, S.S. Consumption of dairy foods and diabetes incidence: A dose-response meta-analysis of observational studies. Am. J. Clin. Nutr. 2016, 103, 1111–1124. [Google Scholar] [CrossRef]
  77. Schwingshackl, L.; Bogensberger, B.; Hoffmann, G. Diet Quality as Assessed by the Healthy Eating Index, Alternate Healthy Eating Index, Dietary Approaches to Stop Hypertension Score, and Health Outcomes: An Updated Systematic Review and Meta-Analysis of Cohort Studies. J. Acad. Nutr. Diet. 2018, 118, 74–100.e11. [Google Scholar] [CrossRef] [PubMed]
  78. Ericson, U.; Hellstrand, S.; Brunkwall, L.; Schulz, C.A.; Sonestedt, E.; Wallström, P.; Gullberg, B.; Wirfält, E.; Orho-Melander, M. Food sources of fat may clarify the inconsistent role of dietary fat intake for incidence of type 2 diabetes. Am. J. Clin. Nutr. 2015, 101, 1065–1080. [Google Scholar] [CrossRef] [PubMed]
  79. Huo, R.; Du, T.; Xu, Y.; Xu, W.; Chen, X.; Sun, K.; Yu, X. 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. [Google Scholar] [CrossRef] [PubMed]
  80. Salas-Salvadó, J.; Bulló, M.; Estruch, R.; Ros, E.; Covas, M.I.; Ibarrola-Jurado, N.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; et al. Prevention of diabetes with Mediterranean diets: A subgroup analysis of a randomized trial. Ann. Intern. Med. 2014, 160, 1–10, Erratum in Ann. Intern. Med. 2018, 169, 271–272. [Google Scholar] [CrossRef]
  81. Ghobadi, S.; Hassanzadeh-Rostami, Z.; Mohammadian, F.; Zare, M.; Faghih, S. Effects of Canola Oil Consumption on Lipid Profile: A Systematic Review and Meta-Analysis of Randomized Controlled Clinical Trials. J. Am. Coll. Nutr. 2019, 38, 185–196. [Google Scholar] [CrossRef]
  82. O’Mahoney, L.L.; Matu, J.; Price, O.J.; Birch, K.M.; Ajjan, R.A.; Farrar, D.; Tapp, R.; West, D.J.; Deighton, K.; Campbell, M.D. 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. [Google Scholar] [CrossRef]
  83. Bosch, J.; Gerstein, H.C.; Dagenais, G.R.; Díaz, R.; Dyal, L.; Jung, H.; Maggiono, A.P.; Probstfield, J.; Ramachandran, A.; Riddle, M.C. n-3 fatty acids and cardiovascular outcomes in patients with dysglycemia. N. Engl. J. Med. 2012, 367, 309–318. [Google Scholar] [CrossRef]
  84. Wu, J.H.Y.; Marklund, M.; Imamura, F.; Tintle, N.; Ardisson Korat, A.V.; de Goede, J.; Zhou, X.; Yang, W.S.; de Oliveira Otto, M.C.; Kröger, J.; et al. Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Fatty Acids and Outcomes Research Consortium (FORCE). Omega-6 fatty acid biomarkers and incident type 2 diabetes: Pooled analysis of individual-level data for 39740 adults from 20 prospective cohort studies. Lancet Diabetes Endocrinol. 2017, 5, 965–974. [Google Scholar] [CrossRef]
  85. Sawada, T.; Tsubata, H.; Hashimoto, N.; Takabe, M.; Miyata, T.; Aoki, K.; Yamashita, S.; Oishi, S.; Osue, T.; Yokoi, K.; et al. Effects of 6-month eicosapentaenoic acid treatment on postprandial hyperglycemia, hyperlipidemia, insulin secretion ability, and concomitant endothelial dysfunction among newly-diagnosed impaired glucose metabolism patients with coronary artery disease. An open label, single blinded, prospective randomized controlled trial. Cardiovasc. Diabetol. 2016, 15, 121. [Google Scholar] [CrossRef]
  86. Bhatt, D.L.; Steg, P.G.; Miller, M.; Brinton, E.A.; Jacobson, T.A.; Ketchum, S.B.; Doyle, R.T., Jr.; Juliano, R.A.; Jiao, L.; Granowitz, C.; et al. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N. Engl. J. Med. 2019, 380, 11–22. [Google Scholar] [CrossRef]
  87. ASCEND Study Collaborative Group. Effects of n-3 Fatty Acid Supplements in Diabetes Mellitus. N. Engl. J. Med. 2018, 379, 1540–1550. [Google Scholar] [CrossRef] [PubMed]
  88. Manson, J.E.; Cook, N.R.; Lee, I.M.; Christen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Albert, C.M.; Gordon, D.; Copeland, T.; et al. VITAL Research Group. Marine n-3 Fatty Acids and Prevention of Cardiovascular Disease and Cancer. N. Engl. J. Med. 2019, 380, 23–32. [Google Scholar] [CrossRef] [PubMed]
  89. 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. [Google Scholar] [CrossRef] [PubMed]
  90. Aronis, K.N.; Khan, S.M.; Mantzoros, C.S. Effects of trans fatty acids on glucose homeostasis: A meta-analysis of randomized, placebo-controlled clinical trials. Am. J. Clin. Nutr. 2012, 96, 1093–1099. [Google Scholar] [CrossRef] [PubMed]
  91. Post, R.E.; Mainous, A.G., 3rd; King, D.E.; Simpson, K.N. Dietary fiber for the treatment of type 2 diabetes mellitus: A meta-analysis. J. Am. Board Fam. Med. 2012, 25, 16–23. [Google Scholar] [CrossRef]
  92. Zhang, Z.; Cogswell, M.E.; Gillespie, C.; Fang, J.; Loustalot, F.; Dai, S.; Carriquiry, A.L.; Kuklina, E.V.; Hong, Y.; Merritt, R.; et al. Association between usual sodium and potassium intake and blood pressure and hypertension among U.S. adults: NHANES 2005–2010. PLoS ONE 2013, 8, e75289. [Google Scholar] [CrossRef] [PubMed]
  93. Centers for Disease Control and Prevention (CDC). CDC grand rounds: Dietary sodium reduction—Time for choice. Morb. Mortal. Wkly. Rep. 2012, 61, 89–91. [Google Scholar]
  94. Appel, L.J.; Frohlich, E.D.; Hall, J.E.; Pearson, T.A.; Sacco, R.L.; Seals, D.R.; Sacks, F.M.; Smith, S.C., Jr.; Vafiadis, D.K.; Van Horn, L.V. The importance of population-wide sodium reduction as a means to prevent cardiovascular disease and stroke: A call to action from the American Heart Association. Circulation 2011, 123, 1138–1143. [Google Scholar] [CrossRef]
  95. Azadbakht, L.; Fard, N.R.; Karimi, M.; Baghaei, M.H.; Surkan, P.J.; Rahimi, M.; Esmaillzadeh, A.; Willett, W.C. Effects of the Dietary Approaches to Stop Hypertension (DASH) eating plan on cardiovascular risks among type 2 diabetic patients: A randomized crossover clinical trial. Diabetes Care 2011, 34, 55–57. [Google Scholar] [CrossRef]
  96. Ekinci, E.I.; Clarke, S.; Thomas, M.C.; Moran, J.L.; Cheong, K.; MacIsaac, R.J.; Jerums, G. Dietary salt intake and mortality in patients with type 2 diabetes. Diabetes Care 2011, 34, 703–709. [Google Scholar] [CrossRef]
  97. Thomas, M.C.; Moran, J.; Forsblom, C.; Harjutsalo, V.; Thorn, L.; Ahola, A.; Wadén, J.; Tolonen, N.; Saraheimo, M.; Gordin, D.; et al. The association between dietary sodium intake, ESRD, and all-cause mortality in patients with type 1 diabetes. Diabetes Care 2011, 34, 861–866. [Google Scholar] [CrossRef] [PubMed]
  98. Dunkler, D.; Dehghan, M.; Teo, K.K.; Heinze, G.; Gao, P.; Kohl, M.; Clase, C.M.; Mann, J.F.; Yusuf, S.; Oberbauer, R. ONTARGET Investigators. Diet and kidney disease in high-risk individuals with type 2 diabetes mellitus. JAMA Intern. Med. 2013, 173, 1682–1692. [Google Scholar] [CrossRef] [PubMed]
  99. Maillot, M.; Drewnowski, A. A conflict between nutritionally adequate diets and meeting the 2010 dietary guidelines for sodium. Am. J. Prev. Med. 2012, 42, 174–179. [Google Scholar] [CrossRef] [PubMed]
  100. Shai, I.; Wainstein, J.; Harman-Boehm, I.; Raz, I.; Fraser, D.; Rudich, A.; Stampfer, M.J. Glycemic effects of moderate alcohol intake among patients with type 2 diabetes: A multicenter, randomized, clinical intervention trial. Diabetes Care 2007, 30, 3011–3016. [Google Scholar] [CrossRef]
  101. Ahmed, A.T.; Karter, A.J.; Warton, E.M.; Doan, J.U.; Weisner, C.M. 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. [Google Scholar] [CrossRef] [PubMed]
  102. Bantle, A.E.; Thomas, W.; Bantle, J.P. Metabolic effects of alcohol in the form of wine in persons with type 2 diabetes mellitus. Metabolism 2008, 57, 241–245. [Google Scholar] [CrossRef]
  103. Schrieks, I.C.; Heil, A.L.; Hendriks, H.F.; Mukamal, K.J.; Beulens, J.W. The effect of alcohol consumption on insulin sensitivity and glycemic status: A systematic review and meta-analysis of intervention studies. Diabetes Care 2015, 38, 723–732. [Google Scholar] [CrossRef]
  104. Howard, A.A.; Arnsten, J.H.; Gourevitch, M.N. Effect of alcohol consumption on diabetes mellitus: A systematic review. Ann. Intern. Med. 2004, 140, 211–219. [Google Scholar] [CrossRef]
  105. Timko, C.; Kong, C.; Vittorio, L.; Cucciare, M.A. Screening and brief intervention for unhealthy substance use in patients with chronic medical conditions: A systematic review. J. Clin. Nurs. 2016, 25, 3131–3143. [Google Scholar] [CrossRef]
  106. Gepner, Y.; Golan, R.; Harman-Boehm, I.; Henkin, Y.; Schwarzfuchs, D.; Shelef, I.; Durst, R.; Kovsan, J.; Bolotin, A.; Leitersdorf, E.; et al. Effects of Initiating Moderate Alcohol Intake on Cardiometabolic Risk in Adults with Type 2 Diabetes: A 2-Year Randomized, Controlled Trial. Ann. Intern. Med. 2015, 163, 569–579. [Google Scholar] [CrossRef]
  107. Gepner, Y.; Henkin, Y.; Schwarzfuchs, D.; Golan, R.; Durst, R.; Shelef, I.; Harman-Boehm, I.; Spitzen, S.; Witkow, S.; Novack, L.; et al. Differential Effect of Initiating Moderate Red Wine Consumption on 24-h Blood Pressure by Alcohol Dehydrogenase Genotypes: Randomized Trial in Type 2 Diabetes. Am. J. Hypertens. 2016, 29, 476–483. [Google Scholar] [CrossRef] [PubMed]
  108. Shimomura, T.; Wakabayashi, I. Inverse associations between light-to-moderate alcohol intake and lipid-related indices in patients with diabetes. Cardiovasc. Diabetol. 2013, 12, 104. [Google Scholar] [CrossRef] [PubMed]
  109. Tetzschner, R.; Nørgaard, K.; Ranjan, A. Effects of alcohol on plasma glucose and prevention of alcohol-induced hypoglycemia in type 1 diabetes-A systematic review with GRADE. Diabetes Metab. Res. Rev. 2018, 34, e2965. [Google Scholar] [CrossRef] [PubMed]
  110. Barnard, K.D.; Dyson, P.; Sinclair, J.M.; Lawton, J.; Anthony, D.; Cranston, M.; Holt, R.I. Alcohol health literacy in young adults with type 1 diabetes and its impact on diabetes management. Diabet. Med. 2014, 31, 1625–1630. [Google Scholar] [CrossRef] [PubMed]
  111. Baliunas, D.O.; Taylor, B.J.; Irving, H.; Roerecke, M.; Patra, J.; Mohapatra, S.; Rehm, J. Alcohol as a risk factor for type 2 diabetes: A systematic review and meta-analysis. Diabetes Care 2009, 32, 2123–2132. [Google Scholar] [CrossRef] [PubMed]
  112. Knott, C.; Bell, S.; Britton, A. Alcohol Consumption and the Risk of Type 2 Diabetes: A Systematic Review and Dose-Response Meta-analysis of More Than 1.9 Million Individuals from 38 Observational Studies. Diabetes Care 2015, 38, 1804–1812. [Google Scholar] [CrossRef] [PubMed]
  113. Huang, J.; Wang, X.; Zhang, Y. Specific types of alcoholic beverage consumption and risk of type 2 diabetes: A systematic review and meta-analysis. J. Diabetes Investig. 2017, 8, 56–68. [Google Scholar] [CrossRef]
  114. Pan, A.; Malik, V.S.; Schulze, M.B.; Manson, J.E.; Willett, W.C.; Hu, F.B. Plain-water intake and risk of type 2 diabetes in young and middle-aged women. Am. J. Clin. Nutr. 2012, 95, 1454–1460. [Google Scholar] [CrossRef]
  115. Malik, V.S. Sugar sweetened beverages and cardiometabolic health. Curr. Opin. Cardiol. 2017, 32, 572–579. [Google Scholar] [CrossRef]
  116. Malik, V.S.; Hu, F.B. Fructose and Cardiometabolic Health: What the Evidence from Sugar-Sweetened Beverages Tells Us. J. Am. Coll. Cardiol. 2015, 66, 1615–1624. [Google Scholar] [CrossRef]
  117. Imamura, F.; O’Connor, L.; Ye, Z.; Mursu, J.; Hayashino, Y.; Bhupathiraju, S.N.; Forouhi, N.G. Consumption of sugar sweetened beverages, artificially sweetened beverages, and fruit juice and incidence of type 2 diabetes: Systematic review, meta-analysis, and estimation of population attributable fraction. BMJ 2015, 351, h3576. [Google Scholar] [CrossRef]
  118. Richardson, I.L.; Frese, S.A. Non-nutritive sweeteners and their impacts on the gut microbiome and host physiology. Front. Nutr. 2022, 9, 988144. [Google Scholar] [CrossRef]
  119. Brazil. Resolution No. 818, 28 September 2023. Provides for the Health Requirements for Tabletop Sweeteners and Dietary Sweeteners. Published in the DOU—Official Gazette of the Union; Executive Branch, No. 188, of October 2, 2023. Available online: https://antigo.anvisa.gov.br/documents/10181/6661634/RDC_818_2023_.pdf/0b734a91-e7ff-43d6-b29f-98528d2b0a7d#:~:text=Disp%C3%B5e%20sobre%20os%20requisitos%20sanit%C3%A1rios,que%20lhe%20conferem%20os%20arts. (accessed on 29 October 2023).
  120. Johnson, R.K.; Appel, L.J.; Brands, M.; Howard, B.V.; Lefevre, M.; Lustig, R.H. American Heart Association Nutrition Committee of the Council on Nutrition, Physical Activity and Metabolism, Council on Arteriosclerosis, Thrombosis and Vascular Biology, Council on Cardiovascular Disease in the Young; American Diabetes Association. Nonnutritive sweeteners: Current use and health perspectives: A scientific statement from the American Heart Association and the American Diabetes Association. Diabetes Care 2012, 35, 1798–1808. [Google Scholar] [CrossRef]
  121. Johnson, R.K.; Lichtenstein, A.H.; Anderson, C.A.M.; Carson, J.A.; Després, J.P.; Hu, F.B.; American Heart Association Nutrition Committee of the Council on Lifestyle and Cardiometabolic Health; Council on Cardiovascular and Stroke Nursing; Council on Clinical Cardiology; Council on Quality of Care and Outcomes Research; et al. Low-Calorie Sweetened Beverages and Cardiometabolic Health: A Science Advisory from the American Heart Association. Circulation 2018, 138, e126–e140. [Google Scholar] [CrossRef] [PubMed]
  122. Nichol, A.D.; Holle, M.J.; An, R. Glycemic impact of non-nutritive sweeteners: A systematic review and meta-analysis of randomized controlled trials. Eur. J. Clin. Nutr. 2018, 72, 796–804. [Google Scholar] [CrossRef] [PubMed]
  123. Sylvetsky, A.C.; Rother, K.I. Nonnutritive Sweeteners in Weight Management and Chronic Disease: A Review. Obesity 2018, 26, 635–640. [Google Scholar] [CrossRef] [PubMed]
  124. Wiebe, N.; Padwal, R.; Field, C.; Marks, S.; Jacobs, R.; Tonelli, M. A systematic review on the effect of sweeteners on glycemic response and clinically relevant outcomes. BMC Med. 2011, 9, 123. [Google Scholar] [CrossRef] [PubMed]
  125. Sesso, H.D.; Christen, W.G.; Bubes, V.; Smith, J.P.; MacFadyen, J.; Schvartz, M.; Manson, J.E.; Glynn, R.J.; Buring, J.E.; Gaziano, J.M. Multivitamins in the prevention of cardiovascular disease in men: The Physicians’ Health Study II randomized controlled trial. JAMA 2012, 308, 1751–1760. [Google Scholar] [CrossRef] [PubMed]
  126. Macpherson, H.; Pipingas, A.; Pase, M.P. Multivitamin-multimineral supplementation and mortality: A meta-analysis of randomized controlled trials. Am. J. Clin. Nutr. 2013, 97, 437–444. [Google Scholar] [CrossRef] [PubMed]
  127. Aroda, V.R.; Edelstein, S.L.; Goldberg, R.B.; Knowler, W.C.; Marcovina, S.M.; Orchard, T.J.; Bray, G.A.; Schade, D.S.; Temprosa, M.G.; White, N.H.; et al. Long-term Metformin Use and Vitamin B12 Deficiency in the Diabetes Prevention Program Outcomes Study. J. Clin. Endocrinol. Metab. 2016, 101, 1754–1761. [Google Scholar] [CrossRef]
  128. Buvat, D.R. Use of metformin is a cause of vitamin B12 deficiency. Am. Fam. Physician 2004, 69, 264. [Google Scholar] [PubMed]
  129. Bauman, W.A.; Shaw, S.; Jayatilleke, E.; Spungen, A.M.; Herbert, V. Increased intake of calcium reverses vitamin B12 malabsorption induced by metformin. Diabetes Care 2000, 23, 1227–1231. [Google Scholar] [CrossRef]
  130. Wang, H.; Li, L.; Qin, L.L.; Song, Y.; Vidal-Alaball, J.; Liu, T.H. Oral vitamin B12 versus intramuscular vitamin B12 for vitamin B12 deficiency. Cochrane Database Syst. Rev. 2018, 3, CD004655. [Google Scholar] [CrossRef] [PubMed]
  131. Butler, C.C.; Vidal-Alaball, J.; Cannings-John, R.; McCaddon, A.; Hood, K.; Papaioannou, A.; McDowell, I.; Goringe, A. Oral vitamin B12 versus intramuscular vitamin B12 for vitamin B12 deficiency: A systematic review of randomized controlled trials. Fam. Pract. 2006, 23, 279–285. [Google Scholar] [CrossRef] [PubMed]
  132. ElSayed, N.A.; Aleppo, G.; Aroda, V.R.; Bannuru, R.R.; Brown, F.M.; Bruemmer, D.; Collins, B.S.; Hilliard, M.E.; Isaacs, D.; Johnson, E.L.; et al. 5. Facilitating Positive Health Behaviors and Well-being to Improve Health Outcomes: Standards of Care in Diabetes-2023. Diabetes Care 2023, 46 (Suppl. S1), S68–S96. [Google Scholar] [CrossRef] [PubMed]
  133. Balk, E.M.; Tatsioni, A.; Lichtenstein, A.H.; Lau, J.; Pittas, A.G. Effect of chromium supplementation on glucose metabolism and lipids: A systematic review of randomized controlled trials. Diabetes Care 2007, 30, 2154–2163. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, Y.; Cotillard, A.; Vatier, C.; Bastard, J.P.; Fellahi, S.; Stévant, M.; Allatif, O.; Langlois, C.; Bieuvelet, S.; Brochot, A.; et al. A Dietary Supplement Containing Cinnamon, Chromium and Carnosine Decreases Fasting Plasma Glucose and Increases Lean Mass in Overweight or Obese Pre-Diabetic Subjects: A Randomized, Placebo-Controlled Trial. PLoS ONE 2015, 10, e0138646, Erratum in PLoS ONE 2015, 10, e0145315. [Google Scholar] [CrossRef]
  135. Veronese, N.; Watutantrige-Fernando, S.; Luchini, C.; Solmi, M.; Sartore, G.; Sergi, G.; Manzato, E.; Barbagallo, M.; Maggi, S.; Stubbs, B. Effect of magnesium supplementation on glucose metabolism in people with or at risk of diabetes: A systematic review and meta-analysis of double-blind randomized controlled trials. Eur. J. Clin. Nutr. 2016, 70, 1354–1359, Erratum in Eur. J. Clin. Nutr. 2016, 70, 1463. [Google Scholar] [CrossRef] [PubMed]
  136. De Valk, H.W.; Verkaaik, R.; van Rijn, H.J.; Geerdink, R.A.; Struyvenberg, A. Oral magnesium supplementation in insulin-requiring Type 2 diabetic patients. Diabet. Med. 1998, 15, 503–507. [Google Scholar] [CrossRef]
  137. Jorde, R.; Figenschau, Y. Supplementation with cholecalciferol does not improve glycaemic control in diabetic subjects with normal serum 25-hydroxyvitamin D levels. Eur. J. Nutr. 2009, 48, 349–354. [Google Scholar] [CrossRef]
  138. Patel, P.; Poretsky, L.; Liao, E. Lack of effect of subtherapeutic vitamin D treatment on glycemic and lipid parameters in Type 2 diabetes: A pilot prospective randomized trial. J. Diabetes. 2010, 2, 36–40. [Google Scholar] [CrossRef] [PubMed]
  139. Parekh, D.; Sarathi, V.; Shivane, V.K.; Bandgar, T.R.; Menon, P.S.; Shah, N.S. Pilot study to evaluate the effect of short-term improvement in vitamin D status on glucose tolerance in patients with type 2 diabetes mellitus. Endocr. Pract. 2010, 16, 600–608. [Google Scholar] [CrossRef] [PubMed]
  140. Nikooyeh, B.; Neyestani, T.R.; Farvid, M.; Alavi-Majd, H.; Houshiarrad, A.; Kalayi, A.; Shariatzadeh, N.; Gharavi, A.; Heravifard, S.; Tayebinejad, N.; et al. Daily consumption of vitamin D- or vitamin D + calcium-fortified yogurt drink improved glycemic control in patients with type 2 diabetes: A randomized clinical trial. Am. J. Clin. Nutr. 2011, 93, 764–771. [Google Scholar] [CrossRef] [PubMed]
  141. Soric, M.M.; Renner, E.T.; Smith, S.R. Effect of daily vitamin D supplementation on HbA1c in patients with uncontrolled type 2 diabetes mellitus: A pilot study. J. Diabetes 2012, 4, 104–105. [Google Scholar] [CrossRef] [PubMed]
  142. Alkharfy, K.M.; Al-Daghri, N.M.; Sabico, S.B.; Al-Othman, A.; Moharram, O.; Alokail, M.S.; Al-Saleh, Y.; Kumar, S.; Chrousos, G.P. Vitamin D supplementation in patients with diabetes mellitus type 2 on different therapeutic regimens: A one-year prospective study. Cardiovasc. Diabetol. 2013, 12, 113. [Google Scholar] [CrossRef] [PubMed]
  143. Sadiya, A.; Ahmed, S.M.; Carlsson, M.; Tesfa, Y.; George, M.; Ali, S.H.; Siddieg, H.H.; Abusnana, S. Vitamin D3 supplementation and body composition in persons with obesity and type 2 diabetes in the UAE: A randomized controlled double-blinded clinical trial. Clin. Nutr. 2016, 35, 77–82. [Google Scholar] [CrossRef] [PubMed]
  144. Mousa, A.; Naderpoor, N.; de Courten, M.P.; Teede, H.; Kellow, N.; Walker, K.; Scragg, R.; de Courten, B. Vitamin D supplementation has no effect on insulin sensitivity or secretion in vitamin D-deficient, overweight or obese adults: A randomized placebo-controlled trial. Am. J. Clin. Nutr. 2017, 105, 1372–1381. [Google Scholar] [CrossRef]
  145. Moreira-Lucas, T.S.; Duncan, A.M.; Rabasa-Lhoret, R.; Vieth, R.; Gibbs, A.L.; Badawi, A.; Wolever, T.M. Effect of vitamin D supplementation on oral glucose tolerance in individuals with low vitamin D status and increased risk for developing type 2 diabetes (EVIDENCE): A double-blind, randomized, placebo-controlled clinical trial. Diabetes Obes. Metab. 2017, 19, 133–141. [Google Scholar] [CrossRef]
  146. Tabesh, M.; Azadbakht, L.; Faghihimani, E.; Tabesh, M.; Esmaillzadeh, A. Effects of calcium-vitamin D co-supplementation on metabolic profiles in vitamin D insufficient people with type 2 diabetes: A randomised controlled clinical trial. Diabetologia 2014, 57, 2038–2047. [Google Scholar] [CrossRef]
  147. Solis, M.Y.; Artioli, G.G.; Gualano, B. Potential of Creatine in Glucose Management and Diabetes. Nutrients 2021, 13, 570. [Google Scholar] [CrossRef]
  148. Chedid, V.; Dhalla, S.; Clarke, J.O.; Roland, B.C.; Dunbar, K.B.; Koh, J.; Justino, E.; Tomakin, E.; Mullin, G.E. Herbal therapy is equivalent to rifaximin for the treatment of small intestinal bacterial overgrowth. Glob. Adv. Health Med. 2014, 3, 16–24. [Google Scholar] [CrossRef]
  149. US Food and Drug Administration. Dietary Supplements. 2018. Available online: https://www.fda.gov/food/dietarysupplements/ (accessed on 1 October 2023).
  150. Hidalgo-Mora, J.J.; García-Vigara, A.; Sánchez-Sánchez, M.L.; García-Pérez, M.Á.; Tarín, J.; Cano, A. The Mediterranean diet: A historical perspective on food for health. Maturitas 2020, 132, 65–69. [Google Scholar] [CrossRef] [PubMed]
  151. Diabetes Prevention Program Research Group. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N. Engl. J. Med. 2002, 346, 393–403. [Google Scholar] [CrossRef] [PubMed]
  152. Tuomilehto, J.; Lindström, J.; Eriksson, J.G.; Valle, T.T.; Hämäläinen, H.; Ilanne-Parikka, P.; Keinänen-Kiukaanniemi, S.; Laakso, M.; Louheranta, A.; Rastas, M.; et al. Finnish Diabetes Prevention Study Group. Prevention of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired glucose tolerance. N. Engl. J. Med. 2001, 344, 1343–1350. [Google Scholar] [CrossRef] [PubMed]
  153. Stentz, F.B.; Brewer, A.; Wan, J.; Garber, C.; Daniels, B.; Sands, C.; Kitabchi, A.E. Remission of pre-diabetes to normal glucose tolerance in obese adults with high protein versus high carbohydrate diet: Randomized control trial. BMJ Open Diabetes Res. Care 2016, 4, e000258. [Google Scholar] [CrossRef] [PubMed]
  154. Esposito, K.; Chiodini, P.; Maiorino, M.I.; Bellastella, G.; Panagiotakos, D.; Giugliano, D. Which diet for prevention of type 2 diabetes? A meta-analysis of prospective studies. Endocrine 2014, 47, 107–116. [Google Scholar] [CrossRef] [PubMed]
  155. Chiu, T.H.T.; Pan, W.H.; Lin, M.N.; Lin, C.L. Vegetarian diet, change in dietary patterns, and diabetes risk: A prospective study. Nutr. Diabetes. 2018, 8, 12. [Google Scholar] [CrossRef]
  156. Becerra-Tomás, N.; Díaz-López, A.; Rosique-Esteban, N.; Ros, E.; Buil-Cosiales, P.; Corella, D.; Estruch, R.; Fitó, M.; Serra-Majem, L.; Arós, F.; et al. PREDIMED Study Investigators. Legume consumption is inversely associated with type 2 diabetes incidence in adults: A prospective assessment from the PREDIMED study. Clin. Nutr. 2018, 37, 906–913. [Google Scholar] [CrossRef]
  157. Lee, Y.; Park, K. Adherence to a Vegetarian Diet and Diabetes Risk: A Systematic Review and Meta-Analysis of Observational Studies. Nutrients 2017, 9, 603. [Google Scholar] [CrossRef]
  158. Schwingshackl, L.; Chaimani, A.; Hoffmann, G.; Schwedhelm, C.; Boeing, H. 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. [Google Scholar] [CrossRef]
  159. Noto, H.; Goto, A.; Tsujimoto, T.; Noda, M. Low-carbohydrate diets and all-cause mortality: A systematic review and meta-analysis of observational studies. PLoS ONE 2013, 8, e55030, Erratum in PLoS ONE 2019, 14, e0212203. [Google Scholar] [CrossRef] [PubMed]
  160. Pan, X.R.; Li, G.W.; Hu, Y.H.; Wang, J.X.; Yang, W.Y.; An, Z.X.; Hu, Z.X.; Lin, J.; Xiao, J.Z.; Cao, H.B.; et al. Effects of diet and exercise in preventing NIDDM in people with impaired glucose tolerance. The Da Qing IGT and Diabetes Study. Diabetes Care 1997, 20, 537–544. [Google Scholar] [CrossRef]
  161. Anderssen, S.A.; Hjermann, I.; Urdal, P.; Torjesen, P.A.; Holme, I. Improved carbohydrate metabolism after physical training and dietary intervention in individuals with the “atherothrombogenic syndrome’. Oslo Diet and Exercise Study (ODES). A randomized trial. J. Intern. Med. 1996, 240, 203–209. [Google Scholar] [CrossRef] [PubMed]
  162. Rodríguez-Villar, C.; Pérez-Heras, A.; Mercadé, I.; Casals, E.; Ros, E. Comparison of a high-carbohydrate and a high-monounsaturated fat, olive oil-rich diet on the susceptibility of LDL to oxidative modification in subjects with Type 2 diabetes mellitus. Diabet. Med. 2004, 21, 142–149. [Google Scholar] [CrossRef] [PubMed]
  163. Itsiopoulos, C.; Brazionis, L.; Kaimakamis, M.; Cameron, M.; Best, J.D.; O’Dea, K.; Rowley, K. Can the Mediterranean diet lower HbA1c in type 2 diabetes? Results from a randomized cross-over study. Nutr. Metab. Cardiovasc. Dis. 2011, 21, 740–747. [Google Scholar] [CrossRef] [PubMed]
  164. Toobert, D.J.; Glasgow, R.E.; Strycker, L.A.; Barrera, M., Jr.; Radcliffe, J.L.; Wander, R.C.; Bagdade, J.D. Biologic and quality-of-life outcomes from the Mediterranean Lifestyle Program: A randomized clinical trial. Diabetes Care 2003, 26, 2288–2293. [Google Scholar] [CrossRef]
  165. Elhayany, A.; Lustman, A.; Abel, R.; Attal-Singer, J.; Vinker, S. A low carbohydrate Mediterranean diet improves cardiovascular risk factors and diabetes control among overweight patients with type 2 diabetes mellitus: A 1-year prospective randomized intervention study. Diabetes Obes. Metab. 2010, 12, 204–209. [Google Scholar] [CrossRef]
  166. Esposito, K.; Maiorino, M.I.; Ciotola, M.; Di Palo, C.; Scognamiglio, P.; Gicchino, M.; Petrizzo, M.; Saccomanno, F.; Beneduce, F.; Ceriello, A.; et al. Effects of a Mediterranean-style diet on the need for antihyperglycemic drug therapy in patients with newly diagnosed type 2 diabetes: A randomized trial. Ann. Intern. Med. 2009, 151, 306–314, Erratum in Ann. Intern. Med. 2009, 151, 591. [Google Scholar] [CrossRef]
  167. Gardner, C.D.; Landry, M.J.; Perelman, D.; Petlura, C.; Durand, L.R.; Aronica, L.; Crimarco, A.; Cunanan, K.M.; Chang, A.; Dant, C.C.; et al. Effect of a ketogenic diet versus Mediterranean diet on glycated hemoglobin in individuals with prediabetes and type 2 diabetes mellitus: The interventional Keto-Med randomized crossover trial. Am. J. Clin. Nutr. 2022, 116, 640–652, Erratum in Am. J. Clin. Nutr. 2022, 116, 1904. [Google Scholar] [CrossRef]
  168. Tay, J.; Thompson, C.H.; Luscombe-Marsh, N.D.; Wycherley, T.P.; Noakes, M.; Buckley, J.D.; Wittert, G.A.; Yancy, W.S., Jr.; Brinkworth, G.D. Effects of an energy-restricted low-carbohydrate, high unsaturated fat/low saturated fat diet versus a high-carbohydrate, low-fat diet in type 2 diabetes: A 2-year randomized clinical trial. Diabetes Obes. Metab. 2018, 20, 858–871. [Google Scholar] [CrossRef]
  169. Sainsbury, E.; Kizirian, N.V.; Partridge, S.R.; Gill, T.; Colagiuri, S.; Gibson, A.A. Effect of dietary carbohydrate restriction on glycemic control in adults with diabetes: A systematic review and meta-analysis. Diabetes Res. Clin. Pract. 2018, 139, 239–252. [Google Scholar] [CrossRef] [PubMed]
  170. Van Zuuren, E.J.; Fedorowicz, Z.; Kuijpers, T.; Pijl, H. Effects of low-carbohydrate- compared with low-fat-diet interventions on metabolic control in people with type 2 diabetes: A systematic review including GRADE assessments. Am. J. Clin. Nutr. 2018, 108, 300–331. [Google Scholar] [CrossRef]
  171. Snorgaard, O.; Poulsen, G.M.; Andersen, H.K.; Astrup, A. Systematic review and meta-analysis of dietary carbohydrate restriction in patients with type 2 diabetes. BMJ Open Diabetes Res. Care 2017, 5, e000354. [Google Scholar] [CrossRef] [PubMed]
  172. Bhanpuri, N.H.; Hallberg, S.J.; Williams, P.T.; McKenzie, A.L.; Ballard, K.D.; Campbell, W.W.; McCarter, J.P.; Phinney, S.D.; Volek, J.S. Cardiovascular disease risk factor responses to a type 2 diabetes care model including nutritional ketosis induced by sustained carbohydrate restriction at 1 year: An open label, non-randomized, controlled study. Cardiovasc. Diabetol. 2018, 17, 56. [Google Scholar] [CrossRef] [PubMed]
  173. Tay, J.; Luscombe-Marsh, N.D.; Thompson, C.H.; Noakes, M.; Buckley, J.D.; Wittert, G.A.; Yancy, W.S., Jr.; Brinkworth, G.D. Comparison of low- and high-carbohydrate diets for type 2 diabetes management: A randomized trial. Am. J. Clin. Nutr. 2015, 102, 780–790. [Google Scholar] [CrossRef]
  174. Wycherley, T.P.; Thompson, C.H.; Buckley, J.D.; Luscombe-Marsh, N.D.; Noakes, M.; Wittert, G.A.; Brinkworth, G.D. Long-term effects of weight loss with a very-low carbohydrate, low saturated fat diet on flow mediated dilatation in patients with type 2 diabetes: A randomised controlled trial. Atherosclerosis 2016, 252, 28–31. [Google Scholar] [CrossRef] [PubMed]
  175. Gibbs, B.B.; Brancati, F.L.; Chen, H.; Coday, M.; Jakicic, J.M.; Lewis, C.E.; Stewart, K.J.; Clark, J.M.; Look AHEAD Research Group. Effect of improved fitness beyond weight loss on cardiovascular risk factors in individuals with type 2 diabetes in the Look AHEAD study. Eur. J Prev. Cardiol. 2014, 21, 608–617. [Google Scholar] [CrossRef]
  176. Pi-Sunyer, X.; Blackburn, G.; Brancati, F.L.; Bray, G.A.; Bright, R.; Clark, J.M.; Curtis, J.M.; Espeland, M.A.; Foreyt, J.P.; Graves, K.; et al. Look AHEAD Research Group. Reduction in weight and cardiovascular disease risk factors in individuals with type 2 diabetes: One-year results of the look AHEAD trial. Diabetes Care 2007, 30, 1374–1383. [Google Scholar] [CrossRef]
  177. Brehm, B.J.; Lattin, B.L.; Summer, S.S.; Boback, J.A.; Gilchrist, G.M.; Jandacek, R.J.; D’Alessio, D.A. One-year comparison of a high-monounsaturated fat diet with a high-carbohydrate diet in type 2 diabetes. Diabetes Care 2009, 32, 215–220. [Google Scholar] [CrossRef]
  178. Davis, N.J.; Tomuta, N.; Schechter, C.; Isasi, C.R.; Segal-Isaacson, C.J.; Stein, D.; Zonszein, J.; Wylie-Rosett, J. Comparative study of the effects of a 1-year dietary intervention of a low-carbohydrate diet versus a low-fat diet on weight and glycemic control in type 2 diabetes. Diabetes Care 2009, 32, 1147–1152. [Google Scholar] [CrossRef]
  179. Guldbrand, H.; Dizdar, B.; Bunjaku, B.; Lindström, T.; Bachrach-Lindström, M.; Fredrikson, M.; Ostgren, C.J.; Nystrom, F.H. In type 2 diabetes, randomisation to advice to follow a low-carbohydrate diet transiently improves glycaemic control compared with advice to follow a low-fat diet producing a similar weight loss. Diabetologia 2012, 55, 2118–2127. [Google Scholar] [CrossRef]
  180. Papakonstantinou, E.; Triantafillidou, D.; Panagiotakos, D.B.; Koutsovasilis, A.; Saliaris, M.; Manolis, A.; Melidonis, A.; Zampelas, A. A high-protein low-fat diet is more effective in improving blood pressure and triglycerides in calorie-restricted obese individuals with newly diagnosed type 2 diabetes. Eur. J. Clin. Nutr. 2010, 64, 595–602. [Google Scholar] [CrossRef]
  181. Kodama, S.; Saito, K.; Tanaka, S.; Maki, M.; Yachi, Y.; Sato, M.; Sugawara, A.; Totsuka, K.; Shimano, H.; Ohashi, Y.; et al. Influence of fat and carbohydrate proportions on the metabolic profile in patients with type 2 diabetes: A meta-analysis. Diabetes Care 2009, 32, 959–965. [Google Scholar] [CrossRef] [PubMed]
  182. Barnard, R.J.; Massey, M.R.; Cherny, S.; O’Brien, L.T.; Pritikin, N. Long-term use of a high-complex-carbohydrate, high-fiber, low-fat diet and exercise in the treatment of NIDDM patients. Diabetes Care 1983, 6, 268–273. [Google Scholar] [CrossRef]
  183. Barnard, R.J.; Jung, T.; Inkeles, S.B. Diet and exercise in the treatment of NIDDM. The need for early emphasis. Diabetes Care. 1994, 17, 1469–1472. [Google Scholar] [CrossRef] [PubMed]
  184. Barnard, N.D.; Cohen, J.; Jenkins, D.J.; Turner-McGrievy, G.; Gloede, L.; Jaster, B.; Seidl, K.; Green, A.A.; Talpers, S. 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. [Google Scholar] [CrossRef] [PubMed]
  185. Nicholson, A.S.; Sklar, M.; Barnard, N.D.; Gore, S.; Sullivan, R.; Browning, S. Toward improved management of NIDDM: A randomized, controlled, pilot intervention using a lowfat, vegetarian diet. Prev. Med. 1999, 29, 87–91. [Google Scholar] [CrossRef]
  186. Tonstad, S.; Butler, T.; Yan, R.; Fraser, G.E. Type of vegetarian diet, body weight, and prevalence of type 2 diabetes. Diabetes Care 2009, 32, 791–796. [Google Scholar] [CrossRef]
  187. Kahleova, H.; Matoulek, M.; Malinska, H.; Oliyarnik, O.; Kazdova, L.; Neskudla, T.; Skoch, A.; Hajek, M.; Hill, M.; Kahle, M.; et al. Vegetarian diet improves insulin resistance and oxidative stress markers more than conventional diet in subjects with Type 2 diabetes. Diabet. Med. 2011, 28, 549–559. [Google Scholar] [CrossRef]
  188. Barnard, N.D.; Cohen, J.; Jenkins, D.J.; Turner-McGrievy, G.; Gloede, L.; Green, A.; Ferdowsian, H. A low-fat vegan diet and a conventional diabetes diet in the treatment of type 2 diabetes: A randomized, controlled, 74-wk clinical trial. Am. J. Clin. Nutr. 2009, 89, 1588S–1596S. [Google Scholar] [CrossRef]
  189. Hosseinpour-Niazi, S.; Mirmiran, P.; Hedayati, M.; Azizi, F. Substitution of red meat with legumes in the therapeutic lifestyle change diet based on dietary advice improves cardiometabolic risk factors in overweight type 2 diabetes patients: A cross-over randomized clinical trial. Eur. J. Clin. Nutr. 2015, 69, 592–597. [Google Scholar] [CrossRef]
  190. McMacken, M.; Shah, S. A plant-based diet for the prevention and treatment of type 2 diabetes. J. Geriatr. Cardiol. 2017, 14, 342–354. [Google Scholar] [CrossRef] [PubMed]
  191. Pollakova, D.; Andreadi, A.; Pacifici, F.; Della-Morte, D.; Lauro, D.; Tubili, C. The Impact of Vegan Diet in the Prevention and Treatment of Type 2 Diabetes: A Systematic Review. Nutrients 2021, 13, 2123. [Google Scholar] [CrossRef] [PubMed]
  192. Jönsson, T.; Granfeldt, Y.; Ahrén, B.; Branell, U.C.; Pålsson, G.; Hansson, A.; Söderström, M.; Lindeberg, S. Beneficial effects of a Paleolithic diet on cardiovascular risk factors in type 2 diabetes: A randomized cross-over pilot study. Cardiovasc. Diabetol. 2009, 8, 35. [Google Scholar] [CrossRef] [PubMed]
  193. Masharani, U.; Sherchan, P.; Schloetter, M.; Stratford, S.; Xiao, A.; Sebastian, A.; Nolte Kennedy, M.; Frassetto, L. Metabolic and physiologic effects from consuming a hunter-gatherer (Paleolithic)-type diet in type 2 diabetes. Eur. J. Clin. Nutr. 2015, 69, 944–948. [Google Scholar] [CrossRef] [PubMed]
  194. Lindeberg, S.; Jönsson, T.; Granfeldt, Y.; Borgstrand, E.; Soffman, J.; Sjöström, K.; Ahrén, B. A Palaeolithic diet improves glucose tolerance more than a Mediterranean-like diet in individuals with ischaemic heart disease. Diabetologia 2007, 50, 1795–1807. [Google Scholar] [CrossRef] [PubMed]
  195. Carter, S.; Clifton, P.M.; Keogh, J.B. Effect of Intermittent Compared with Continuous Energy Restricted Diet on Glycemic Control in Patients with Type 2 Diabetes: A Randomized Noninferiority Trial. JAMA Netw. Open. 2018, 1, e180756. [Google Scholar] [CrossRef]
  196. Corley, B.T.; Carroll, R.W.; Hall, R.M.; Weatherall, M.; Parry-Strong, A.; Krebs, J.D. Intermittent fasting in Type 2 diabetes mellitus and the risk of hypoglycaemia: A randomized controlled trial. Diabet. Med. 2018, 35, 588–594. [Google Scholar] [CrossRef]
  197. Borgundvaag, E.; Mak, J.; Kramer, C.K. 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. [Google Scholar] [CrossRef]
  198. Miller, C.K. Mindful Eating with Diabetes. Diabetes Spectr. 2017, 30, 89–94. [Google Scholar] [CrossRef]
  199. Mason, A.E.; Saslow, L.; Moran, P.J.; Kim, S.; Wali, P.K.; Abousleiman, H.; Hartman, A.; Richler, R.; Schleicher, S.; Hartogensis, W.; et al. Examining the Effects of Mindful Eating Training on Adherence to a Carbohydrate-Restricted Diet in Patients with Type 2 Diabetes (the DELISH Study): Protocol for a Randomized Controlled Trial. JMIR Res. Protoc. 2019, 8, e11002, Erratum in JMIR Res. Protoc. 2020, 9, e17226. [Google Scholar] [CrossRef] [PubMed]
  200. Ruiz-Roso, M.B.; Knott-Torcal, C.; Matilla-Escalante, D.C.; Garcimartín, A.; Sampedro-Nuñez, M.A.; Dávalos, A.; Marazuela, M. COVID-19 Lockdown and Changes of the Dietary Pattern and Physical Activity Habits in a Cohort of Patients with Type 2 Diabetes Mellitus. Nutrients 2020, 12, 2327. [Google Scholar] [CrossRef] [PubMed]
  201. Grabia, M.; Markiewicz-Żukowska, R.; Puścion-Jakubik, A.; Bielecka, J.; Nowakowski, P.; Gromkowska-Kępka, K.; Mielcarek, K.; Socha, K. The Nutritional and Health Effects of the COVID-19 Pandemic on Patients with Diabetes Mellitus. Nutrients 2020, 12, 3013. [Google Scholar] [CrossRef] [PubMed]
  202. Sankar, P.; Ahmed, W.N.; Mariam Koshy, V.; Jacob, R.; Sasidharan, S. Effects of COVID-19 lockdown on type 2 diabetes, lifestyle and psychosocial health: A hospital-based cross-sectional survey from South India. Diabetes Metab. Syndr. 2020, 14, 1815–1819. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of dietary patterns for the management of T2DM.
Table 1. Summary of dietary patterns for the management of T2DM.
Dietary PatternsDescriptionLevel of
Evidence
Adherence and Comments
Mediterranean DietMediterranean diet encourages consumption of foods with high nutritional quality, such as fruits, vegetables, legumes, whole grains, cereals, tubers, roots fish, low-fat dairy products, vegetable oils, nuts, seed oils, and wine in moderation [16,132,163,164,165,166,167].High(1) High long-term adherence [16,132,163,164,165,166,167].
(2) There is a lot of evidence in the literature for the improvement of molecular markers, glycemic control, cardiovascular health, and weight loss [16,132,163,164,165,166,167].
Dash Diet
(Dietary Approaches to Stop Hypertension)
DASH diet is similar to the Mediterranean pattern, but alcohol consumption is not encouraged and there is also sodium restriction. It consists of fruits, vegetables, legumes, whole grains, cereals, tubers, roots, fish, chicken breast, lean meats, low-fat dairy products, vegetable oils, nuts, oilseeds and sodium restriction (<2300 mg or 1500 mg for severe heart disease) [16,30,31,63,95,132].High(1) High long-term adherence [16,30,31,63,95,132].
(2) There is a lot of evidence in the literature for the improvement of molecular markers, glycemic control, cardiovascular health, reduce blood pressure and weight loss [16,30,31,63,95,132].
(3) Severe sodium restriction (1500 mg) requires caution and monitoring, and is indicated for some specific cases [16,30,31,63,94,95,96,97,98,99,132].
Ketogenic Diet
(very low-carb diet)
A dietary pattern composed of 5–10% carbohydrates, 15–25% proteins and 60–70% fats of the total energy value (TEV). Includes raw vegetables, very low-carb fruits (avocado and strawberries), all types of meat (beef, pork, fish and chicken), full-fat dairy products, vegetable oils, animal fat and eggs. It has a low intake of fruits, legumes, whole grains, cereals, roots and tubers [16,132,167,173].Low/
Moderate
(1) Low long-term adherence [16,132,167,168,169,170,171,172,173,174].
(2) There are many controversies in the literature [16,132,167,168,169,170,171,172,173,174].
(3) No advantages over other nutritional strategies [16,132,167].
(4) The severe carbohydrate restriction may not really be necessary and safe [16,132,167].
(5) New evidence shows results that it improves weight loss and glycemic control in the short term, but increases markers of cardiovascular risk, such as LDL cholesterol [16,132,167].
(6) This dietary pattern also can increase the risk of dehydration and hypoglycemia; there is a high probability of food monotony, low fiber and micronutrients intake [16,132,167].
(7) More studies are needed in patients with diabetes, especially in the long term [16,132,167].
Low-Carb DietLow-carb diet promotes reducing the consumption of ultra-processed foods. The carbohydrate intake range is 40–45% of the TEV. Encourages the consumption of fruits, vegetables, legumes, whole grains, tubers, fish, lean meats, skimmed dairy products, vegetable oils, nuts, avocados, eggs and seed oils. Carbohydrates of high nutritional quality are allowed, but without excess [16,132,167,168,169,170,171,172,173,174].High(1) High long-term adherence [16,132,167].
(2) In recent years, good evidence has been published in the literature for the modulation of molecular markers, glycemic control, cardiovascular health and weight loss [16,132,167,168,169,170,171,172,173,174].
(3) There are several types of protocols, and those that restrict saturated fats (<7% of TEV) and prioritize sources of polyunsaturated and monounsaturated fats show good results [16,132,167,168,169,170,171,172,173,174].
Low-FatThis dietary pattern involves ingesting 25–30% fats within the TEV. It encourages the consumption of fruits, vegetables, legumes, whole grains, tubers, fish, chicken breast, lean meats, and skimmed dairy products. This eating pattern is similar to DASH [175,176,177,178,179,180,181].High(1) High long-term adherence [175,176,177,178,179,180,181].
(2) There is a lot of evidence in the literature for the modulation of molecular markers, glycemic control, cardiovascular health, reduced blood pressure, and weight loss [175,176,177,178,179,180,181].
Ornish and
Pritikins
(very low-fat diet)
Both eating patterns have very low fat consumption (10% fat of the TEV). They encourage the consumption of whole foods, vegetables, legumes, fruits, grains, low-fat dairy products, and egg whites [16,132,182,183,184,185].Low(1) Low long-term adherence [16,132,182,183,184,185].
(2) Over the years, the scientific community has lost interest in studying this type of diet, since it has low adherence, small palatability, and can possibly cause metabolic damage (hormone production, protection, and energy storage) [16,132,182,183,184,185].
Plant-Based Diet
(or vegetarian/
vegan diet)
Plant-based diet consisting eat of foods with high nutritional quality, such as fruits, vegetables, legumes, whole grains, cereal, tubers, roots, vegetable oils, nuts and seed oils. In this diet there is no intake of any type of food from an animal source (intake of high-fiber foods). It encourages questioning about food choices, autonomy, and ethical and cultural issues [16,132,184,185,186,187,188,189,190,191].High(1) Moderate long-term adherence [16,132,184,185,186,187,188,189,190,191].
(2) There is a lot of evidence in the literature for the modulation of molecular markers, glycemic control, cardiovascular health, reduced blood pressure, and weight loss (intake of high-fiber foods) [16,132,184,185,186,187,188,189,190,191].
(3) However, this diet needs constant nutritional monitoring, because in the long term it can reduce intake of some micronutrients, such as iron, calcium, and vitamin B12 [16,132,184,185,186,187,188,189,190,191].
Paleolithic DietThe Paleolithic diet consists of following similar eating habits as our ancestors. It encourages the consumption of all types of meat, animal fat, fruits, vegetables, roots, raw foods and all types of food that nature can offer. The habit of fasting is also recommended in this type of dietary pattern [16,132,192,193,194].Low(1) Low long-term adherence [16,132,192,193,194].
(2) It has become technically impossible to follow a diet identical to the Paleolithic period in Westernized society [16,132,192,193,194].
(3) More studies are needed in patients with Diabetes [16,132,192,193,194].
Intermittent
Fasting
Fasting means abstaining from foods and drinks that contain macronutrients and calories. Daily caloric intake occurs within a defined eating window during the day, and there are several types of fasting protocols (16 to 24 h), aiming to enhance the production of ketone bodies [16,132,195,196,197].Low(1) Low long-term adherence [16,132,195,196,197].
(2) It can increase the risk of dehydration, headache, hypoglycemia, and lack of glycemic control [16,132,195,196,197].
(3) It is strongly discouraged by guidelines for patients with diabetes, mainly in diabetic ketoacidosis [16,132,195,196,197].
(4) More studies are needed in patients with diabetes [16,132,195,196,197].
Mindful EatingThe practices of Mindful Eating can help increase knowledge of the factors (physiological, environmental, or emotional) that lead to excessive food consumption and training in intuitive eating (respecting signs of hunger/satiety and chewing food). The program not only values the quantity and quality consumed, but also the pleasure of the experience, thereby changing the value of the food reward without restricting it [16,132,198,199].Moderate(1) Moderate long-term adherence [16,132,198,199].
(2) It is a type of nutritional strategy that has been gaining prominence in the literature with good results [16,132,198,199].
(3) However, more studies are needed to evaluate the impact of mindful eating on weight loss, glycemic control, and improvement in cardiovascular markers [16,132,198,199].
(4) Seems to be a good strategy to take care of the subjective issues of nutritional management [16,132,198,199].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Minari, T.P.; Tácito, L.H.B.; Yugar, L.B.T.; Ferreira-Melo, S.E.; Manzano, C.F.; Pires, A.C.; Moreno, H.; Vilela-Martin, J.F.; Cosenso-Martin, L.N.; Yugar-Toledo, J.C. Nutritional Strategies for the Management of Type 2 Diabetes Mellitus: A Narrative Review. Nutrients 2023, 15, 5096. https://doi.org/10.3390/nu15245096

AMA Style

Minari TP, Tácito LHB, Yugar LBT, Ferreira-Melo SE, Manzano CF, Pires AC, Moreno H, Vilela-Martin JF, Cosenso-Martin LN, Yugar-Toledo JC. Nutritional Strategies for the Management of Type 2 Diabetes Mellitus: A Narrative Review. Nutrients. 2023; 15(24):5096. https://doi.org/10.3390/nu15245096

Chicago/Turabian Style

Minari, Tatiana Palotta, Lúcia Helena Bonalume Tácito, Louise Buonalumi Tácito Yugar, Sílvia Elaine Ferreira-Melo, Carolina Freitas Manzano, Antônio Carlos Pires, Heitor Moreno, José Fernando Vilela-Martin, Luciana Neves Cosenso-Martin, and Juan Carlos Yugar-Toledo. 2023. "Nutritional Strategies for the Management of Type 2 Diabetes Mellitus: A Narrative Review" Nutrients 15, no. 24: 5096. https://doi.org/10.3390/nu15245096

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop