Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Review Article
  • Open access
  • Published: 30 November 2020

Effect of the ketogenic diet on glycemic control, insulin resistance, and lipid metabolism in patients with T2DM: a systematic review and meta-analysis

  • Xiaojie Yuan 1   na1 ,
  • Jiping Wang 1   na1 ,
  • Shuo Yang 2   na1 ,
  • Mei Gao 2 ,
  • Lingxia Cao 2 ,
  • Xumei Li 1 ,
  • Dongxu Hong 1 ,
  • Suyan Tian 3 &
  • Chenglin Sun   ORCID: orcid.org/0000-0003-3570-1918 1 , 2  

Nutrition & Diabetes volume  10 , Article number:  38 ( 2020 ) Cite this article

37k Accesses

114 Citations

495 Altmetric

Metrics details

  • Type 2 diabetes

At present, the beneficial effect of the ketogenic diet (KD) on weight loss in obese patients is generally recognized. However, a systematic research on the role of KD in the improvement of glycemic and lipid metabolism of patients with diabetes is still found scarce.

This meta-study employed the meta-analysis model of random effects or of fixed effects to analyze the average difference before and after KD and the corresponding 95% CI, thereby evaluating the effect of KD on T2DM.

After KD intervention, in terms of glycemic control, the level of fasting blood glucose decreased by 1.29 mmol/L (95% CI: −1.78 to −0.79) on average, and glycated hemoglobin A1c by 1.07 (95% CI: −1.37 to −0.78). As for lipid metabolism, triglyceride was decreased by 0.72 (95% CI: −1.01 to −0.43) on average, total cholesterol by 0.33 (95% CI: −0.66 to −0.01), and low-density lipoprotein by 0.05 (95% CI: −0.25 to −0.15); yet, high-density lipoprotein increased by 0.14 (95% CI: 0.03−0.25). In addition, patients’ weight decreased by 8.66 (95% CI: −11.40 to −5.92), waist circumference by 9.17 (95% CI: −10.67 to −7.66), and BMI by 3.13 (95% CI: −3.31 to −2.95).

KD not only has a therapeutic effect on glycemic and lipid control among patients with T2DM but also significantly contributes to their weight loss.

Similar content being viewed by others

ketogenic diet thesis statement

Taurine reduces the risk for metabolic syndrome: a systematic review and meta-analysis of randomized controlled trials

ketogenic diet thesis statement

The effects of vegetarian diets on glycemia and lipid parameters in adult patients with overweight and obesity: a systematic review and meta-analysis

ketogenic diet thesis statement

The effects of low-fat, high-carbohydrate diets vs. low-carbohydrate, high-fat diets on weight, blood pressure, serum liquids and blood glucose: a systematic review and meta-analysis

Introduction.

Diabetes mellitus (DM) is the world’s leading cause for motility and morbidity, and the disease has become a major public health burden worldwide. It is estimated that the prevalence of diabetes in adults worldwide is over 300 million, and it will increase by 55% by 2035 1 . Obesity or overweight is one of the essential risk factors for diabetes and contributes to a twice-higher risk to develop DM 2 , 3 . Thus, dietary therapy aiming at weight loss is typically recommended in clinical practice 4 . Due to the fact that diabetes and its complications affect many aspects of physiology, the benefits of weight reduction are not limited to glycemic control but are also related to many cardiovascular risk factors such as blood pressure, high-density lipoprotein (HDL), total cholesterol (TC) and triglyceride (TG) 2 .

Medical nutrition, as part of the comprehensive treatment of DM with obesity with a primary goal of weight reduction, is the most simple, effective and economical choice of intervention. The dietary approach for body weight reduction can be obtained from many strategies, including a low-calorie diet, a very low-calorie diet, high-protein diet, and so on. Ketogenic diet (KD), which contains a very low level of carbohydrates (<55 g/d) with the main energy sources of lipid and protein, and which causes ketosis and simulates the physiological state of fasting, has been well reported to be effective for weight loss and glycemic control 4 , 5 , 6 , 7 , 8 , 9 . Previous meta-analyses have proved the efficacy of KD in body weight reduction 2 , 10 , 11 ; however, systemic reviews on the effect of KD on weight reduction and glycolipid metabolism in patients with DM are still limited. Westman et al. 12 and Partsalaki et al. 13 demonstrated that KD improved type 2 diabetes mellitus (T2DM) by reducing the glycemic response caused by carbohydrate and improving potential insulin resistance. Leonetti et al. 14 and Walton et al. 15 reported reduced TG and TC with increased HDL levels after KD consumption for a lipid profile. However, controversies are still existing; studies revealed that a low-carbohydrate, high-fat diet may exacerbate the lipid profile in patients with diabetes, although glycemic control improved with hypoglycemic medications 16 , 17 , 18 . Therefore, the purpose of the current review was to conduct a meta-analysis on the effects of a KD in patients with diabetes.

Considering the potential benefits of KD in diabetes management and weight reduction, and considering fasting blood glucose and glycated hemoglobin A1c (HbA1c) as common biomarkers for long-term glycemic control, HDL, LDL, TC, and TG levels are included in the current analysis to determine the changes of metabolic disorders in glucose and lipid metabolism. In addition, the homeostatic model assessment of insulin resistance (HOMA-IR) is considered as a reflection of insulin resistance reversal.

Materials and methods

Literature search.

In this meta-analysis, only studies published in English were considered, which were identified by searching the PubMed and MEDLINE databases. The keywords used for this literature search are T2DM or diabetes mellitus, ketogenic diet, obesity, and human. The search was finished on September 20, 2019. This meta-analysis was planned and performed according to the Preferred Reporting Items for Systemic Reviews (PRISM) guideline (Fig. 1 ).

figure 1

Only studies published in English were considered, which were identified by searching the PubMed and MEDLINE databases. The keywords used for this literature search are T2DM or diabetes mellitus, ketogenic diet, obesity, and human. The search was finished on September 20, 2019.

Inclusive/exclusive criteria

Studies that met the following inclusive criteria were included: (1) the disease of interest is type II diabetes; (2) the therapeutic diet under consideration is KD; (3) the study was carried out on humans; animal experiments are not included; and (4) the summary statistics of the mean difference between before and after KD (if both means for before and after measurements are available, then we took the difference of these two statistics to obtain the desired mean difference), their corresponding standard error or 95% CI (according to this, the standard error was calculated) or p values (according to this, the corresponding t statistics and subsequently the standard error were calculated) are available.

Exclusive criteria: (1) case report studies were excluded; (2) meta-analysis or review studies were excluded; (3) studies on other diseases rather than type II diabetes were excluded; and (4) if only the respective mean and standard errors were available, such studies were excluded given it is hard to get an accurate estimation for the standard error of mean difference (since both measurements were on the same patient, they should be correlated to each other, and hence it is impossible to estimate this correlation).

Statistical analysis

The effects of KD on type II diabetes were estimated by the mean difference after KD versus before KD and their corresponding 95% CIs in random-effects meta-analysis models or fixed-effect meta-analysis models. To determine which model should be used, heterogeneity among studies was evaluated by the Cochrane’s Q statistic corresponding p values and the I 2 statistics. If the p value was <0.05 and I 2  > 0.5, a random-effect meta-analysis model was used. Otherwise, a fixed-effect meta-analysis model was chosen. Additionally, potential bias was assessed by using funnel plots, in which effect sizes versus standard errors were diagrammed. All statistical analysis was carried out in the R software, version 3.5 ( www.r-project.org ) 19 , 20 , 21 .

There are 13 studies included in this meta-analysis; the details of these 13 studies are presented in Table 1 . In total, 567 subjects were included in the final meta-analysis. From the perspective of glucose metabolism, lipid metabolism, and weight control, the effects of KD on T2DM were systemically reviewed by comparing the after-intervention measures with before-intervention measures of several biomarkers for the same patient. The variables used to surrogate for carbohydrate metabolism are included fasting glucose level and HbA1c; for lipid metabolism TC, TG, HDL and LDL; and for weight loss body weight, BMI and waist circumference. For all variables except BMI and waist, random-effect models were adopted according to the Q statistic p value and I 2 statistics.

Using the meta-analysis method, we found that the fasting blood glucose level was decreased 1.29 mmol/l (95% CI: −1.78 to −0.79) after the intervention of KD, compared to before such an intervention (based on ten articles that have the summary statistics for the difference between after- and before-intervention measures). As far as HbA1c is concerned, we found that the reduced proportion of HbA1c is more significant after the KD implementation, with a difference of −1.07% (95% CI: −1.37 to −0.78), which is regarded as the ideal therapeutic effect of drugs that is possible to be achieved on HbA1c. The forest plots for these two carbohydrates metabolism indices are given in Fig. 2 .

figure 2

The reduced proportion of HbA1c is more significant after the KD implementation, which is regarded as the ideal therapeutic effect of drugs that is possible to be achieved on HbA1c.

In this study, eight articles investigated the effect of KD on the lipid metabolism of diabetic patients, but only five papers analyzed total cholesterol. It can be seen that after KD consumption, TG decreased by 0.72 mmol/L (95% CI: −1.01 to −0.43), TC decreased by 0.33 mmol/L (95% CI: −0.66 to −0.01), and LDL decreased by 0.05 mmol/L (95% CI: −0.25 to −0.15). On the other hand, HDL increased by 0.14 mmol/L (95% CI: 0.03−0.25). The forest plots for these four biomarkers are shown in Fig. 3 .

figure 3

It can be seen that after KD consumption, TG, TC, and LDL decreased. On the other hand, HDL increased.

Regarding weight loss, many studies have demonstrated that KD has a positive effect by providing effective control over obesity. The results of our meta-analysis are consistent with previous results. Specifically, the average weight decreased by 8.66 kg (95% CI: −11.40 to −5.92), waist circumference reduced by 9.17 cm (95% CI: −10.67 to −7.66) and BMI reduced by 3.13 kg/m 2 (95% CI: −3.31 to −2.95), as shown in Fig. 4 .

figure 4

Many studies have demonstrated that KD has a positive effect by providing effective control over obesity; our findings were consistent with the previous reports.

The American Diabetes Society (ADA) recommended physical activity, dietary management, and medical intake and other approaches should be applied simultaneously to manage blood glucose levels, and other abnormal metabolic factors. KD showed numerous health benefits to patients with T2DM 22 , 23 . KD provides energy through fat oxidation. When the human body experienced extreme hunger or very limited carbohydrate, the ketone body was produced and released to circulation by hepatic transformation of fatty acids 24 , 25 . Nutritional ketosis is different from severe pathological diabetic ketosis; the blood ketone body remained at 0.5−3.0 mmol/L with reduced blood glucose and normal blood pH, with no symptoms in nutritional ketosis 26 .

The possible mechanism for the health benefit of KD on patients with T2DM is that the extreme restriction of carbohydrate reduces the intestinal absorption of mono-saccharide, which leads to lower blood glucose level and reduces the fluctuation of blood glucose, and its effectiveness on regulating glucose metabolism was confirmed by a large body of evidence 27 , 28 . The current study analyzed 13 studies from literature focusing on diabetic patients; the results showed that the reduction of blood glucose ranges from 0.62 to 5.61 mmol/L. Higher reduction amplitudes were reported by Dashti 29 and Leonetti et al. 14 of 5.61 mmol/L (weight random 3.0%) and 3.87 mmol/L (weight random 1.2%), respectively; other reductions in blood glucose were all lower than 1.8 mmol/L. The possible reason for the higher reduction found in these two studies could be the higher blood glucose level included in the studies, and also that the average blood glucose concentration was above 10.0 mmol/L, leading to the possibility of a larger reduction; however, their contribution to the overall effect estimations in the meta-analysis was low. The average changes in fasting blood glucose after the KD consumption among the selected studies were −1.29 mmol/L, indicating the effectiveness of the KD in lowering fasting blood glucose.

No studies included in this meta-analysis evaluated the effect of KD on postprandial glucose level; unlike medications, dietetic therapy showed a long-term effect on glucose regulation 4 , 16 , and HbA1c was analyzed to evaluate the long-term effect of KD. HbA1c effectively reflects the blood glucose control in the past 2−3 months in patients with diabetes. It is reported that the risk of cardiac infarction and micro-vascular complications reduced by 14% and 37%, respectively, when HbA1c reduced by 1%. Therefore, the HbA1c level showed essential clinical significance in evaluating the blood glucose control, revealing the potential problems in the treatment and thereby guiding the therapeutic schedule 30 , 31 . Eight of the selected studies showed a reduction of HbA1c after KD consumption, the changes ranging from −0.6% to −3.3%; HbA1c reduced <1.5% in the majority of the studies included in the current analysis besides the study conducted by Walton (−3.3%; weight random 5.1%) 15 . The possible explanation for such strong improvement of HbA1c could be that Walton’s study had enrolled a limited number of patients and thus the compliance of patients to KD therapy can be guaranteed. Moreover, the studied subjects were newly diagnosed diabetic patients who were under dietary management without taking glucose-lowering medications; newly diagnosed subjects persist well in the study. Considering the causal factors comprehensively, the above study showed an ideal reduction in HbA1c. The average reduction of HbA1c was 1.07 in the current analysis of the selected eight studies, indicating that dietary management may also achieve the ideal therapeutic effects of medication.

HOMA-IR is considered as an indicator to evaluate the status of insulin resistance. Insulin resistance as a clinical characteristic of T2DM is closely related to obesity. Improving insulin resistance is one of the major targets in diabetes treatment 32 , 33 , 34 . However, studies focusing on the role of KD in the improvement of insulin resistance in patients with diabetes are very limited; most of the studies focused on the effect in obese subjects 35 , 36 . For instance, a controlled clinical trial aiming at the effects of KD consumption in obese people without diabetes revealed that HOMA-IR decreased by about 2.0 after KD consumption for 6 weeks 37 . The current analysis showed consistent changes in the studies that included HOMA-IR evaluation, with reduction ranging from −0.4 to −3.4; the reason for the significant reduction of 3.4 in the study by Tay et al. 38 is that the population included was obese diabetic patients with BMI higher than 30 kg/m 2 . Obesity is closely related to insulin resistance; KD consumption is confirmed to be effective in reducing body weight, and it is expected that KD may improve insulin resistance in obese diabetic patients 39 . During the ketogenesis, the sensitivity of the insulin receptor is promoted; therefore, KD not only ensures the supply of basic nutrients but also maintains a negative balance of energy, and reduces the fluctuation and reduction of insulin secretion caused by reduced carbohydrate intake as well, which eventually leads to improved insulin sensitivity 40 , 41 , 42 , 43 .

Consumption of KD not only improved glucose metabolism, but a large body of evidence has reported that KD improved lipid metabolism as well. Hussain et al. 4 reported that KD reduced TG and TC, and increased HDL level, thus ameliorating the status of dyslipidemia. In the present study, eight studies included showed results of lipid metabolism in diabetic patients after KD consumption; however, only five analyzed the TC levels. The current results showed the mean reduction of TG was 0.72 mmol/L, TC was 0.33 mmol/L, and LDL was 0.05 mmol/L, while the increase of HDL was 0.14 mmol/L. The higher amplitude of variation occurred in the Dashti et al. study 29 . This study reported that TG reduced by 3.67 mmol/L, TC reduced by 1.88 mmol/L, and LDL reduced by 1.78 mmol/L, while HDL increased by 0.14 mmol/L. Changes in the amplitude of the lipid biomarkers were all at the higher end in the above study. Both glucose and lipid metabolism showed great improvement after KD consumption in such a study; the characteristics of subjects recruited were closely correlated. The study recruited 31 obese subjects with hyperglycemia, dyslipidemia, and BMI over 30 kg/m 2 . The baseline TG, TC, and LDL were higher than those of typical patients with T2DM, which may contribute to the significant changes after the intervention. Consumption of KD showed a significant therapeutic effect in common patients with T2DM, including the Dashti 29 study. Disorders of lipid metabolism are particularly strong among patients with insulin resistance in T2DM. Dyslipidemia is lipotoxic to cells, leading to and/or aggravating insulin resistance. Its typical manifestation is the increase of TG and free fatty acid (FFA) 44 , 45 , 46 , 47 . Increased FFA is an independent pathogenic factor for insulin resistance and can possibly increase the risk for cardiovascular diseases 48 , 49 . Therefore, the improvement of dyslipidemia is beneficial for not only regulating insulin sensitivity but also controlling the occurrence and progression of diabetic complications 50 , 51 .

Numerous studies have confirmed the role of KD consumption in weight reduction in obese patients 35 , 36 , 37 , 40 , 41 , 42 , 43 , 52 ; the current meta-analysis focused on the effect of KD on weight reduction in obese diabetic patients. The results showed the average reduction of body weight was 8.66 kg, waist circumference was 9.17 cm, and BMI was 3.22 kg/m 2 , which were consistent with previous studies in nondiabetic patients. We also found that KD reduced systolic blood pressure by 4.30 (95% CI: −7.02 to 1.58) and diastolic blood pressure by 5.14 (95% CI: −10.18 to 0.10) in patients with T2DM, which possibly benefit from the improvement of body weight 51 .

Besides the mediation of glucose and lipid metabolism, KD may also benefit other clinical symptoms in diabetic patients, including insomnia, chills, constipation, pruritus, numbness of limbs, hypopsia, fatty liver, hypertension, and reduced cardiac function.

The potential side effects of KD were only mentioned in two of the studies 14 , 41 included in the meta-analysis; thus it is impossible to perform a systematic review in terms of the risks associated with KD consumption. Specifically, Goday and Leonetti’s 14 , 41 study investigated the adverse reactions of KD. Goday et al. 41 mentioned that fatigue, headache, nausea and vomiting were more common in the KD diet group after a 2-week intervention, while constipation and orthostatic hypotension were more common after 10 weeks. It was revealed by Leonetti et al. 14 that in the early stages of applying the KD, patients reported a sense of hunger, but it could be significantly alleviated with the progress of the intervention. Even though headache, nausea, vomiting, constipation, diarrhea, and other symptoms were reported during the study, the symptoms were mild and lasted for a short time, not relating to clinical practice.

Limitations

Only 13 studies were included in the current analysis, with limited studies focusing on the effect of KD in patients with T2DM worldwide. For instance, no analysis was conducted on HOMA-IR even though there was a trend of improvement; also, very limited literature was available. All studies included in this meta-analysis were carried out among Caucasian diabetic patients (no East Asians included); however, the majority of East Asian diabetic patients showed insulin resistance with central obesity and defect in insulin secretion. Therefore, clinical trials conducted among East Asians are highly desirable to confirm whether there is an improvement in the secretion function of islet cells other than improved regulation of glucose and lipid metabolism. Moreover, the current study analyzed the data without assigning studies into time duration due to the limited number of studies and the missing data of insulin and lipid biomarkers; in addition, the duration of the follow-up was decentralized into days, months, and years. The available studies concerning the effects of ketogenic diet in patients with diabetes are very limited; it is impossible to summarize a similar follow-up interval for statistical analysis of time points. However, the current results suggested that ketogenic diet consumption contributed to therapeutic effects despite the length of the term of intervention. The analysis of the difference before and after the intervention may also give credit to the clinical efficacy of the diet therapy. In current clinical practice, a majority of the patients have to use a combination of multiple drugs to improve their glycolipid metabolism. Drug therapy is a heavy mental and economical burden to patients. Given the fact that most of the patients are confused regarding a proper dietary therapy plan, it is essential to recommend a feasible dietary therapy plan to transmit a positive message to both patients with diabetes and physicians majored in the area of diabetes.

Based on a meta-analysis that systematically reviewed 13 relevant studies, we found that ketogenic diet can not only control fasting blood glucose and reduce glycosylated hemoglobin, but also improve lipid metabolism. Additionally, ketogenic diet can reduce BMI and body weight. Therefore, ketogenic diet may be used as part of the integrated management of type 2 diabetes.

Guariguata, L. et al. Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res. Clin. Pr. 103 , 139–149 (2014).

Google Scholar  

Yu, Z. et al. Effects of high-protein diet on glycemic control, insulin resistance and blood pressure in T2DM: a systematic review and metaanalysis of randomized controlled trials. Clin. Nutr. 39 , 1724–1734 (2020).

Article   CAS   Google Scholar  

Guh, D. P. et al. The incidence of co-morbidities related to obesity and overweight: a systematic review and meta-analysis. BMC Public Health 9 , 88 (2009).

Article   Google Scholar  

Hussain, T. A. et al. Effect of low-calorie versus low-carbohydrate ketogenic diet in T2DM. Nutrition 28 , 1061–1021 (2012).

Hall, K. D. et al. Energy expenditure and body composition changes after an isocaloric ketogenic diet in overweight and obese men. Am. J. Clin. Nutr. 104 , 324–333 (2016).

Hamdy, O. et al. Fat versus carbohydrate-based energy-restricted diets for weight loss in patients with T2DM. Curr. Diab. Rep. 18 , 128 (2018).

Joshi, S., Ostfeld, R. J. & McMacken, M. The ketogenic diet for obesity and diabetes-enthusiasm outpaces evidence. JAMA Intern. Med. 179 , 1163–1164 (2019).

Saslow, L. R. et al. An online intervention comparing a very low-carbohydrate ketogenic diet and lifestyle recommendations versus a plate method diet in overweight individuals with T2DM: a randomized controlled trial. J. Med. Internet Res. 19 , e36 (2017).

Saslow, L. R. et al. Twelve-month outcomes of a randomized trial of a moderate-carbohydrate versus very low-carbohydrate diet in overweight adults with T2DM mellitus or prediabetes. Nutr. Diabetes 7 , 304 (2017).

Meng, Y. et al. Efficacy of low carbohydrate diet for T2DM mellitus management: a systematic review and meta-analysis of randomized controlled trials. Diabetes Res. Clin. Pr. 131 , 124–131 (2017).

Castellana, M. et al. Efficacy and safety of very low calorie ketogenic diet (VLCKD) in patients with overweight and obesity: a systematic review and meta-analysis. Rev. Endocr. Metab. Disord. 21 , 5–16 (2019).

Westman, E. C., Yancy, W. S. Jr., Mavropoulos, J. C., Marquart, M. & McDuffie, J. R. The effect of a low-carbohydrate, ketogenic diet versus a low-glycemic index diet on glycemic control in T2DM mellitus. Nutr. Metab. 5 , 36 (2008).

Partsalaki, I., Karvela, A. & Spiliotis, B. E. Metabolic impact of a ketogenic diet compared to a hypocaloric diet in obese children and adolescents. J. Pediatr. Endocrinol. Metab. 25 , 697–704 (2012).

Leonetti, F. et al. Very low-carbohydrate ketogenic diet before bariatric surgery: prospective evaluation of a sequential diet. Obes. Surg. 25 , 64–71 (2015).

Walton, C. M., Perry, K., Hart, R. H., Berry, S. L. & Bikman, B. T. Improvement in glycemic and lipid profiles in type 2 diabetics with a 90-day ketogenic diet. J. Diabetes Res. 14 , 8681959 (2019).

Leow, Z. Z. X., Guelfi, K. J., Davis, E. A., Jones, T. W. & Fournier, P. A. The glycaemic benefits of a very-low-carbohydrate ketogenic diet in adults with Type 1 diabetes mellitus may be opposed by increased hypoglycaemia risk and dyslipidaemia. Diabet. Med. 35 , 9 (2018).

Azevedo, D. L. P. et al. Effect of classic ketogenic diet treatment on lipoprotein subfractions in children and adolescents with refractory epilepsy. Nutrition 33 , 271–277 (2017).

Krebs, J. D. et al. Improvements in glucose metabolism and insulin sensitivity with a low-carbohydrate diet in obese patients with T2DM. J. Am. Coll. Nutr. 32 , 11–17 (2013).

Lau, J., Ioannidis, J. P. & Schmid, C. H. Quantitative synthesis in systematic reviews. Ann. Intern. Med. 127 , 820–826 (1997).

Higgins, J. P. & Thompson, S. G. Quantifying heterogeneity in a meta-analysis. Stat. Med. 21 , 1539–1558 (2002).

DerSimonian, R. & Laird, N. Meta-analysis in clinical trials. Control Clin. Trials 7 , 177–188 (1986).

McKenzie, A. L. et al. A Novel Intervention Including Individualized Nutritional Recommendations Reduces Hemoglobin A1c Level, Medication Use, and Weight in Type 2 Diabetes. JMIR Diabetes 2 , e5 (2017).

Yancy, W. S. et al. A low-carbohydrate, ketogenic diet to treat type 2 diabetes. Nutr. Metab. (Lond) 2 , 34 (2005).

Athinarayanan, S. J. et al. Long-term effects of a novel continuous remote care intervention including nutritional ketosis for the management of T2DM: a 2-year non-randomized clinical trial. Front. Endocrinol. 10 , 348 (2019).

Laffel, L. Ketone bodies: a review of physiology, pathophysiology and application of monitoring to diabetes. Diabetes Metab. Res. Rev. 15 , 412–426 (1999).

Gershuni, V. M., Yan, S. L. & Medici, V. Nutritional ketosis for weight management and reversal of metabolic syndrome. Curr. Nutr. Rep. 7 , 97–106 (2018).

Yancy, W. S., Vernon, M. C. & Westman, E. C. A pilot trial of a low-carbohydrate, ketogenic diet in patients with T2DM. Metab. Syndr. Relat. Disord. 1 , 239–243 (2003).

Bolla, A. M., Caretto, A., Laurenzi, A., Scavini, M. & Piemonti, L. Low-carb and ketogenic diets in type 1 and type 2 diabetes. Nutrients 26 , 962 (2019).

Dashti, H. M. et al . Beneficial effects of ketogenic diet in obese diabetic subjects. Mol. Cell Biochem . 302 , 249–256 (2007).

Yancy, W. S. Jr. et al. A randomized trial of a low-carbohydrate diet vs orlistat plus a low-fat diet for weight loss. Arch. Intern. Med . 170 , 136–145 (2010).

Stratton, I. M. et al. Association of glycaemia with macrovascular and microvascular complications of T2DM (UKPDS 35): prospective observational study. BMJ 321 , 405–412 (2000).

Nakanishi, S. et al. Comparison of HbA1c levels and body mass index for prevention of diabetic kidney disease: a retrospective longitudinal study using outpatient clinical data in Japanese patients with T2DM mellitus. Diabetes Res. Clin. Pr . 155 , 107807 (2019).

Dehghan, P. & Abbasalizad, F. M. Dietary acid load, blood pressure, fasting blood sugar and biomarkers of insulin resistance among adults: findings from an updated systematic review and meta-analysis. Int. J. Clin. Pr. 74 , 4 (2019).

Avtanski, D., Pavlov, V. A., Tracey, K. J. & Poretsky, L. Characterization of inflammation and insulin resistance in high-fat diet-induced male C57BL/6J mouse model of obesity. Anim. Model Exp. Med . 2 , 252–258 (2019).

Saslow, L. R. et al. A randomized pilot trial of a moderate carbohydrate diet compared to a very low carbohydrate diet in overweight or obese individuals with type 2 diabetes mellitus or prediabetes. PLoS One 9 , e91027 (2014).

Stocker, R. K., Reber, A. E., Bally, L., Nuoffer, J. M. & Stanga, Z. Ketogenic diet and its evidence-based therapeutic implementation in endocrine diseases. Praxis 108 , 541–553 (2019).

Handley, R. T., Bentley, R. E., Brown, T. L. & Annan, A. A. Successful treatment of obesity and insulin resistance via ketogenic diet status post Roux-en-Y. BMJ Case Rep. 2018 , bcr2018225643 (2018).

Tay, J. et al. Comparison of low- and high-carbohydrate diets for type 2 diabetes management: a randomized trial. Am. J. Clin. Nutr . 102 , 780–790 (2015).

Kinzig, K. P., Honors, M. A. & Hargrave, S. L. Insulin sensitivity and glucose tolerance are altered by maintenance on a ketogenic diet. Endocrinology 151 , 3105–3114 (2010).

Cox, N., Gibas, S., Salisbury, M., Gomer, J. & Gibas, K. Ketogenic diets potentially reverse Type II diabetes and ameliorate clinical depression: a case study. Diabetes Metab. Syndr . 13 , 1475–1479 (2019).

Goday, A. et al. Short-term safety, tolerability and efficacy of a very low-calorie-ketogenic diet interventional weight loss program versus hypocaloric diet in patients with type 2 diabetes mellitus. Nutr Diabetes 6 , e230 (2016).

Cohen, C. W. et al. A ketogenic diet reduces central obesity and serum Insulin in women with ovarian or endometrial cancer. J. Nutr . 148 , 1253–1260 (2018).

Myette-Cote, E. et al. The effect of a short-term low-carbohydrate, high-fat diet with or without postmeal walks on glycemic control and inflammation in type 2 diabetes: a randomized trial. Am. J. Physiol. Regul. Integr. Comp. Physiol. 315 , R1210–R1219 (2018).

Zhu, B. et al. Lipid oversupply induces CD36 sarcolemmal translocation via dual modulation of PKCzeta and TBC1D1: an early event prior to insulin resistance. Theranostics 10 , 1332–1354 (2020).

Akhtar, D. H., Iqbal, U., Vazquez-Montesino, L. M., Dennis, B. B. & Ahmed, A. Pathogenesis of insulin resistance and atherogenic dyslipidemia in nonalcoholic fatty liver disease. J. Clin. Transl. Hepatol. 7 , 362–370 (2019).

Iqbal, J., Al Qarni, A., Hawwari, A., Alghanem, A. F. & Ahmed, G. Metabolic syndrome, dyslipidemia and regulation of lipoprotein metabolism. Curr. Diabetes Rev . 14 , 427–433 (2018).

Laakso, M. & Kuusisto, J. Insulin resistance and hyperglycaemia in cardiovascular disease development. Nat. Rev. Endocrinol . 10 , 293–302 (2014).

Han, L. et al. Free fatty acid can induce cardiac dysfunction and alter insulin signaling pathways in the heart. Lipids Health Dis. 17 , 185 (2018).

Yu, S. et al. Treatment with adipose tissue-derived mesenchymal stem cells exerts anti-diabetic effects, improves long-term complications, and attenuates inflammation in type 2 diabetic rats. Stem Cell Res. Ther . 10 , 333 (2019).

Ponce, A. J. et al. Low prolactin levels are associated with visceral adipocyte hypertrophy and insulin resistance in humans. Endocrine 67 , 331–343 (2020).

Karasek, D. & Vaverkova, H. Diabetic dyslipidemia and microvascular complications of diabetes. Vnitr. Lek. 64 , 17–24 (2018).

Sajoux, I. et al. Effect of a very-low-calorie ketogenic diet on circulating myokine levels compared with the effect of bariatric surgery or a low-calorie diet in patients with obesity. Nutrients 11 , 2368 (2019).

Download references

Acknowledgements

This study was supported by the Science Technology Department of Jilin Province (20180623006TC and 20200404213YY) and the Interdisciplinary Project of First Hospital of Jilin University (JDYYJC010) and Transformation Project of First Hospital of Jilin University (JDYYZH-1902019) and Education Department of Jilin Province (JJKH20190032KJ and JJKH20201081KJ).

Author information

These authors contributed equally: Xiaojie Yuan, Jiping Wang, Shuo Yang

Authors and Affiliations

Department of Clinical Nutrition, First Hospital of Jilin University, 1 Xinmin Street, 130021, Changchun, Jilin, China

Xiaojie Yuan, Jiping Wang, Xumei Li, Dongxu Hong & Chenglin Sun

Department of Endocrinology and Metabolism, First Hospital of Jilin University, 1 Xinmin Street, 130021, Changchun, Jilin, China

Shuo Yang, Mei Gao, Lingxia Cao & Chenglin Sun

Division of Clinical Research, First Hospital of Jilin University, 1 Xinmin Street, 130021, Changchun, Jilin, China

You can also search for this author in PubMed   Google Scholar

Contributions

X.Y., J.W., S.Y., S.T. and C.S. were responsible for writing, M.G., L.C., X.L. and S.Y. were responsible for the literature collection and data management, D.H. and S.T. for statistical analysis, C.S. and S.T. are in charge of the overall research design and supervision.

Corresponding authors

Correspondence to Suyan Tian or Chenglin Sun .

Ethics declarations

Conflict of interest.

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ .

Reprints and permissions

About this article

Cite this article.

Yuan, X., Wang, J., Yang, S. et al. Effect of the ketogenic diet on glycemic control, insulin resistance, and lipid metabolism in patients with T2DM: a systematic review and meta-analysis. Nutr. Diabetes 10 , 38 (2020). https://doi.org/10.1038/s41387-020-00142-z

Download citation

Received : 06 July 2020

Revised : 19 October 2020

Accepted : 06 November 2020

Published : 30 November 2020

DOI : https://doi.org/10.1038/s41387-020-00142-z

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Metagenomic and metabolomic analysis showing the adverse risk–benefit trade-off of the ketogenic diet.

  • Hongyan Qiu
  • Chengxia Kan
  • Junfeng Shi

Lipids in Health and Disease (2024)

A randomized feasibility trial of medium chain triglyceride-supplemented ketogenic diet in people with Parkinson's disease

  • Alexander H. Choi
  • Melanie Delgado
  • Debra J. Ehrlich

BMC Neurology (2024)

Very Low Calorie Ketogenic Diet: What Effects on Lipid Metabolism?

  • Rosario Suarez
  • Sebastián Chapela
  • Giovanna Muscogiuri

Current Nutrition Reports (2024)

The Ketogenic Diet as a Treatment for Mood Disorders

  • Virginie-Anne Chouinard
  • Christopher M. Palmer

Current Treatment Options in Psychiatry (2024)

Effects of ketogenic diet on health outcomes: an umbrella review of meta-analyses of randomized clinical trials

  • Chanthawat Patikorn
  • Pantakarn Saidoung
  • Nathorn Chaiyakunapruk

BMC Medicine (2023)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

ketogenic diet thesis statement

  • Research article
  • Open access
  • Published: 25 May 2023

Effects of ketogenic diet on health outcomes: an umbrella review of meta-analyses of randomized clinical trials

  • Chanthawat Patikorn 1 , 2 ,
  • Pantakarn Saidoung 1 ,
  • Tuan Pham 3 ,
  • Pochamana Phisalprapa 4 ,
  • Yeong Yeh Lee 5 ,
  • Krista A. Varady 6 ,
  • Sajesh K. Veettil 1 &
  • Nathorn Chaiyakunapruk 1 , 7  

BMC Medicine volume  21 , Article number:  196 ( 2023 ) Cite this article

19k Accesses

20 Citations

92 Altmetric

Metrics details

Systematic reviews and meta-analyses of randomized clinical trials (RCTs) have reported the benefits of ketogenic diets (KD) in various participants such as patients with epilepsy and adults with overweight or obesity . Nevertheless, there has been little synthesis of the strength and quality of this evidence in aggregate.

To grade the evidence from published meta-analyses of RCTs that assessed the association of KD, ketogenic low-carbohydrate high-fat diet (K-LCHF), and very low-calorie KD (VLCKD) with health outcomes, PubMed, EMBASE, Epistemonikos, and Cochrane database of systematic reviews were searched up to February 15, 2023. Meta-analyses of RCTs of KD were included. Meta-analyses were re-performed using a random-effects model. The quality of evidence per association provided in meta-analyses was rated by the GRADE (Grading of Recommendations, Assessment, Development, and Evaluations) criteria as high, moderate, low, and very low.

We included 17 meta-analyses comprising 68 RCTs (median [interquartile range, IQR] sample size of 42 [20–104] participants and follow-up period of 13 [8–36] weeks) and 115 unique associations. There were 51 statistically significant associations (44%) of which four associations were supported by high-quality evidence (reduced triglyceride ( n  = 2), seizure frequency ( n  = 1) and increased low-density lipoprotein cholesterol (LDL-C) ( n  = 1)) and four associations supported by moderate-quality evidence (decrease in body weight, respiratory exchange ratio (RER), hemoglobin A 1c , and increased total cholesterol). The remaining associations were supported by very low (26 associations) to low (17 associations) quality evidence. In overweight or obese adults, VLCKD was significantly associated with improvement in anthropometric and cardiometabolic outcomes without worsening muscle mass, LDL-C, and total cholesterol. K-LCHF was associated with reduced body weight and body fat percentage, but also reduced muscle mass in healthy participants.

Conclusions

This umbrella review found beneficial associations of KD supported by moderate to high-quality evidence on seizure and several cardiometabolic parameters. However, KD was associated with a clinically meaningful increase in LDL-C. Clinical trials with long-term follow-up are warranted to investigate whether the short-term effects of KD will translate to beneficial effects on clinical outcomes such as cardiovascular events and mortality.

Peer Review reports

Ketogenic diets (KD) have received substantial attention from the public primarily due to their ability to produce rapid weight loss in the short run [ 1 , 2 ]. The KD eating pattern severely restricts carbohydrate intake to less than 50 g/day while increasing protein and fat intake [ 3 , 4 , 5 , 6 ]. Carbohydrate deprivation leads to an increase in circulating ketone bodies by breaking down fatty acids and ketogenic amino acids. Ketones are an alternative energy source from carbohydrates that alter physiological adaptations. These adaptions have been shown to produce weight loss with beneficial health effects by improving glycemic and lipid profiles [ 7 , 8 ]. KD has also been recommended as a nonpharmacological treatment for medication-refractory epilepsy in children and adults [ 8 , 9 ]. Evidence suggests that KD has reduced seizure frequency in patients with medication-refractory epilepsy, and even allowing some patients to reach complete and sustained remission. 11 However, the exact anticonvulsive mechanism of KD remains unclear [ 10 , 11 ].

Several systematic reviews and meta-analyses of randomized clinical trials (RCTs) have reported on the use of KD in patients with obesity or type 2 diabetes mellitus (T2DM) to control weight and improve cardiometabolic parameters [ 1 , 12 , 13 , 14 , 15 ], in patients with refractory epilepsy to reduce seizure frequency [ 16 ], and in athletes to control weight and improve performance [ 17 ]. To date, there has been little synthesis of the strength and quality of this evidence in aggregate. This umbrella review therefore aims to systematically identify relevant meta-analyses of RCTs of KD, summarize their findings, and assess the strength of evidence of the effects of KD on health outcomes.

The protocol of this study was registered with PROSPERO (CRD42022334717). We reported following the 2020 Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) (Additional file 1 ) [ 18 ]. Difference from the original review protocol is described with rationale in Additional file 2 : Table S1.

Search strategy and eligibility criteria

We searched PubMed, EMBASE, Epistemonikos, and the Cochrane database of systematic reviews (CDSR) from the database inception to February 15, 2023 (Additional file 2 : Table S2). No language restriction was applied. Study selection was independently performed in EndNote by two reviewers (C.P. and PS). After removing duplicates, the identified articles' titles and abstracts were screened for relevance. Full-text articles of the potentially eligible articles were retrieved and selected against the eligibility criteria. Any discrepancies were resolved by discussion with the third reviewer (SKV).

We included studies that met the following eligibility criteria: systematic reviews and meta-analyses of RCTs investigating the effects of any type of KD on any health outcomes in participants with or without any medical conditions compared with any comparators. When more than 1 meta-analysis was available for the same research question, we selected the meta-analysis with the largest data set [ 19 , 20 , 21 ]. Articles without full-text and meta-analyses that provided insufficient or inadequate data for quantitative synthesis were excluded.

Data extraction and quality assessment

Two reviewers (CP and PS) independently performed data extraction and quality assessment (Additional file 2 : Method S1). Discrepancies were resolved with consensus by discussing with the third reviewer (SKV). We used AMSTAR- 2 -A Measurement Tool to Assess Systematic Reviews- to grade the quality of meta-analyses as high, moderate, low, or critically low by assessing the following elements, research question, a priori protocol, search, study selection, data extraction, quality assessment, data analysis, interpretation, heterogeneity, publication bias, source of funding, conflict of interest [ 22 ].

Data synthesis

For each association, we extracted effect sizes (mean difference [MD], the standardized mean difference [SMD], and risk ratio [RR]) of individual studies included in each meta-analysis and performed the meta-analyses to calculate the pooled effect sizes and 95% CIs using a random-effects model under DerSimonian and Laird [ 23 ], or the Hartung-Knapp- Sidik-Jonkman approach for meta-analyses with less than five studies [ 24 ].  p  < 0.05 was considered statistically significant in 2-sided tests. Heterogeneity was evaluated using the I 2 statistic. The evidence for small-study effects was assessed by the Egger regression asymmetry test [ 25 ]. Statistical analyses were conducted using Stata version 16.0 (StataCorp). We presented effect sizes of statistically significant associations with the known or estimated minimally clinically important difference (MCID) thresholds for health outcomes [ 14 , 26 , 27 , 28 , 29 , 30 ].

We assessed the quality of evidence per association by applying the GRADE criteria (Grading of Recommendations, Assessment, Development, and Evaluations) in five domains, including (1) risk of bias in the individual studies, (2) inconsistency, (3) indirectness, (4) imprecision, and (5) publication bias [ 31 ]. We graded the strength of evidence (high, moderate, low, and very low) using GRADEpro version 3.6.1 (McMaster University).

Sensitivity analyses

Sensitivity analyses were performed by excluding small-size studies (< 25 th percentile) [ 32 ] and excluding primary studies having a high risk of bias rated by the Cochrane’s risk of bias 2 tool (RoB 2) for RCTs from the identified associations [ 19 , 20 , 21 , 33 ].

Seventeen meta-analyses were included (Fig.  1 and Additional file 2 : Table S3) [ 1 , 2 , 15 , 16 , 17 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 ]. These meta-analyses comprised 68 unique RCTs with a median (interquartile range, IQR) sample size per RCT of 42 (20–104) participants and a median (IQR) follow-up period of 13 (8–36) weeks. The quality of meta-analyses assessed using AMSTAR-2 found that none were rated as high confidence, 2 (12%) as moderate confidence, 2 (12%) as low confidence, and 13 (76.0%) as critically low confidence (Table 1 and Additional file 2 : Table S4).

figure 1

Study selection flow of meta-analyses. Abbreviation: CDSR, Cochrane database of systematic review

Types of KD identified in this umbrella review were categorized as (1) KD, which limits carbohydrate intake to < 50 g/day or < 10% of the total energy intake (TEI) [ 35 ], (2) ketogenic low-carbohydrate, high-fat diet (K-LCHF), which limits carbohydrate intake to < 50 g/day or < 10% of TEI with high amount of fat intake (60–80% of TEI) [ 38 , 46 ], (3) very low-calorie KD (VLCKD), which limits carbohydrate intake to < 30–50 g/day or 13–25% of TEI with TEI < 700–800 kcal/day, and (4) modified Atkins diet (MAD), which generally limits carbohydrate intake to < 10 g/day while encouraging high-fat foods [ 15 , 47 ]. Meta-analyses of long-chain triglyceride KD, medium-chain triglyceride KD, and low glycemic index treatment were not identified.

Description and summary of associations

We identified 115 unique associations of KD with health outcomes (Additional file 2 : Table S5). The median (IQR) number of studies per association was 3 [ 4 , 5 , 6 ], and the median (IQR) sample size was 244 (127–430) participants. Outcomes were associated with KD types, including 40 (35%) KD, 18 (16%) K-LCHF, 13 (11%) VLCKD, 25 (22%) KD or K-LCHF, 5 (4%) KD or VLCKD, 1 (1%) KD or MAD, and 13 (11%) KD, K-LCHF, or VLCKD.

The associations involved 40 (35%) anthropometric measures (i.e., body weight, body mass index [BMI] [calculated as weight in kilograms divided by height in meters squared], waist circumference, muscle mass, fat mass, body fat percentage, and visceral adipose tissue), 37 (32%) lipid profile outcomes (i.e., triglyceride, total cholesterol, high-density lipoprotein cholesterol [HDL-C], and low-density lipoprotein cholesterol [LDL-C]), 22 (19%) glycemic profile outcomes (i.e., hemoglobin A 1c [HbA 1c ], fasting plasma glucose, fasting insulin, and homeostatic model assessment of insulin resistance [HOMA-IR]), 6 (5%) exercise performance (i.e., maximal heart rate, respiratory exchange ratio [RER], maximal oxygen consumption (VO 2 max), 5 (4%) blood pressure outcomes (i.e., systolic blood pressure [SBP], diastolic blood pressure [DBP], and heart rate), 1 (1%) outcome associated with seizure frequency reduction ≥ 50% from baseline, and 3 other outcomes (i.e., serum creatinine, C-peptide, and C-reactive protein). In addition, there is 1 association (1%) of adverse events.

Participants in the identified associations included 68 (59%) associations in adults with overweight or obesity with or without T2DM or dyslipidemia, 15 (13%) athletes or resistance-trained adults, 12 (10%) adults with T2DM, 11 (10%) healthy participants ≥ 16 years old, 8 (7%) cancer patients, and 1 (1%) in children and adolescents with epilepsy.

Using GRADE, 115 associations were supported by very low strength of evidence ( n  = 66, 57%), with the remaining being low ( n  = 36, 31%), moderate ( n  = 9, 8%), and high quality of evidence ( n  = 4, 3%) (Additional file 2 : Table S5). Almost half, or 44% (51 associations), were statistically significant based on a random-effects model, of which 51% (26 associations) were supported by a very low level of evidence, followed by low (17 associations [33%]), moderate (4 associations [8%]), and high (4 associations [8%]) levels of evidence. Overall beneficial outcomes associated with KD were BMI [ 37 , 42 ], body weight [ 1 , 2 , 35 , 36 , 37 , 41 ], waist circumference [ 37 , 42 ], fat mass [ 37 , 42 ], body fat percentage [ 38 , 40 ], visceral adipose tissue [ 37 ], triglyceride [ 1 , 2 , 36 , 42 ], HDL-C [ 1 , 2 , 42 ], HbA 1c  [ 2 , 34 , 35 ],  HOMA-IR [ 2 , 42 ], DBP [ 1 ], seizure frequency reduction ≥ 50% from baseline [ 16 ], and respiratory exchange ratio [ 17 , 39 ]. Adverse outcomes associated with KD were reduced muscle mass [ 37 , 38 ], and increased LDL-C [ 2 , 35 ], and total cholesterol [ 2 , 17 ]. In terms of safety, one association showed no significant increase in adverse events (e.g., constipation, abdominal pain, and nausea) with KD [ 44 ].

Eight out of 13 associations supported by moderate to high-quality evidence were statistically significant (Table 2 ). There were 4 statistically significant associations supported by high-quality evidence, including the following: (1) KD or MAD for 3–16 months was associated with a higher proportion of children and adolescents with refractory epilepsy achieving seizure frequency reduction ≥ 50% from baseline compared with regular diet (RR, 5.11; 95% CI, 3.18 to 8.21) [ 16 ], (2) KD for 3 months was associated with reduced triglyceride in adults with T2DM compared with regular diet (MD, -18.36 mg/dL; 95% CI, -24.24 to -12.49, MCID threshold 7.96 mg/dL) [ 14 , 35 ], (3) KD for 12 months was associated with reduced triglyceride in adults with T2DM compared with regular diet (MD, -24.10 mg/dL; 95% CI, -33.93 to -14.27, MCID threshold 7.96 mg/dL) [ 14 , 35 ], and (4) KD for 12 months was associated with increased LDL-C in adults with T2DM compared with regular diet (MD, 6.35 mg/dL; 95% CI, 2.02 to 10.69, MCID threshold 3.87 mg/dL) [ 14 , 35 ]. In addition, there were 4 statistically significant associations supported by moderate-quality evidence: (1) KD for 3 months was associated with reduced HbA 1c in adults with T2DM compared with regular diet (MD, -0.61%; 95% CI, -0.82 to -0.40, MCID threshold 0.5%) [ 14 , 35 ], (2) VLCKD for 4–6 weeks was associated with reduced body weight in T2DM adults with overweight or obesity compared with a low-fat diet or regular diet (MD, -9.33 kg; 95% CI, -15.45 to -3.22, MCID threshold 4.40 kg) [ 14 , 15 ], (3) K-LCHF for 4–6 weeks was associated with reduced respiratory exchange ratio in athletes compared with a high-carbohydrate diet (SMD, -2.66; 95% CI, -3.77 to -1.54) [ 39 ], and (4) K-LCHF for 11–24 weeks was associated with increased total cholesterol in athletes compared with regular diet (MD, 1.32 mg/dL; 95% CI, 0.64 to 1.99) [ 14 , 17 ].

Types of KD showed different effects on health outcomes with changes more than the MCID thresholds in different populations (Fig.  2 ). KD or MAD for 3–16 months was associated with a 5-times higher proportion of children and adolescents with refractory epilepsy achieving seizure frequency reduction ≥ 50% from baseline compared with a regular diet (RR, 5.11; 95% CI, 3.18 to 8.21) [ 16 ]. In healthy participants, K-LCHF for 3–12 weeks could reduce body weight by 3.68 kg (95% CI, -4.45 to -2.90) but also significantly reduced muscle mass by 1.27 kg (95% CI, -1.83 to -0.70, MCID threshold 1.10 kg) [ 14 , 26 , 38 ]. In adults with T2DM, KD for 3–12 months was found to have significant associations with changes more than the MCID thresholds, including reduction of triglyceride and HbA 1c ; however, KD for 12 months led to a clinically meaningful increase in LDL-C by 6.35 mg/dL (95% CI, 2.02 to 10.69, MCID threshold 3.87 mg/dL) [ 14 , 35 ]. In adults with overweight or obesity and/or metabolic syndrome, VLCKD for 4–6 weeks demonstrated a clinically meaningful weight loss of 9.33 kg (95% CI, -15.45 to -3.22, MCID threshold 4.40 kg) [ 14 , 15 ]. VLCKD for 3–96 weeks led to a clinically meaningful improvement in BMI, body weight, waist circumference, triglyceride, fat mass, and insulin resistance, while preserving muscle mass [ 42 ].

figure 2

Associations of Types of Ketogenic Diet with Health Outcomes. Abbreviations: BMI, body mass index, DBP, diastolic blood pressure; GRADE, Grading of Recommendations, Assessment, Development, and Evaluations; HbA 1c , hemoglobin A 1c ; HDL-C, high-density lipoprotein cholesterol; HOMA-IR, homeostatic model of insulin resistance; LDL-C, low-density lipoprotein cholesterol; SBP, systolic blood pressure; TEI, total energy intake

Excluding RCTs with small sizes in 7 associations found that the strength of evidence of one association was downgraded to very low quality, i.e., KD for 12 months, and the increase of LDL-C in adults with T2DM compared with a control diet. Another association was downgraded to low quality, i.e., KD for 12 months and the reduction of triglyceride in adults with T2DM compared with the control diet (Additional file 2 : Table S6). The remaining associations retained the same rank.

This umbrella review was performed to systematically assess the potential associations of KD and health outcomes by summarizing the evidence from meta-analyses of RCTs. Sensitivity analyses were performed to provide additional evidence from high-quality RCTs, which further increased the reliability of results. We identified 115 associations of KD with a wide range of outcomes. Most associations were rated as low and very low evidence according to the GRADE criteria because of serious imprecision and large heterogeneity in findings, and indirectness due to a mix of different interventions and comparators.

Our findings showed that KD or MAD resulted in better seizure control in children and adolescents with medication-refractory epilepsy (approximately a third of cases) for up to 16 months [ 10 , 11 , 16 ]. Anti-epileptic mechanisms of KD remain unknown but are likely multifactorial. Enhanced mitochondrial metabolism and an increase in ketone bodies or reduction in glucose across the blood–brain barrier resulted in synaptic stabilization [ 48 , 49 , 50 ]. Other mechanisms include an increase in gamma-aminobutyric acid (GABA) [ 51 ], more beneficial gut microbiome [ 52 ], less pro-inflammatory markers [ 53 ], and epigenetic modifications (e.g. beta-hydroxybutyrate [beta-OHB]) [ 54 ].

In adults, KD was associated with improved anthropometric measures, cardiometabolic parameters, and exercise performance. Our findings, however, demonstrated differences in the level of associations with type of KD. On the one hand, VLCKD is very effective in producing weight loss while preserving muscle mass in adults with overweight or obesity, with specific benefits on anthropometric and cardiometabolic parameters [ 15 , 42 ]. On the other hand, a significant portion of the weight loss seen in K-LCHF was due to muscle mass loss [ 17 , 38 ]. Overall KD was negatively associated with reduced muscle mass and increased LDL-C and total cholesterol.

Our findings demonstrated that KD could induce a rapid weight loss in the initial phase of 6 months, after which time further weight loss was hardly achieved [ 35 ]. Furthermore, weight loss induced by KD is relatively modest and appears comparable to other dietary interventions that are effective for short-term weight loss, e.g., intermittent fastingand Mediterranean diet [ 55 , 56 , 57 ].

KD is one of the dietary interventions employed by individuals to achieve rapid weight loss, which usually comes with reduced muscle mass [ 58 ]. However, KD has been hypothesized to preserve muscle mass following weight loss based on several mechanisms, including the protective effect of ketones and its precursors on muscle tissue [ 59 , 60 , 61 ], and increased growth hormone secretion stimulated by low blood glucose to increase muscle protein synthesis [ 58 , 62 , 63 ].

With regards to KD effects on lipid profiles, our results demonstrate an effective reduction in serum triglyceride levels with 3 months of lowered dietary carbohydrate intake, with even further reduction by month 12 [ 35 ]. Triglyceride levels are consistently shown to decrease after KD. Acute ketosis (beta-OHB ≈ 3 mM) due to ketone supplementation also shows decreases in triglycerides, indicating a potential effect of ketones on triglycerides independent of weight loss. One possible mechanism is the decreased very low-density lipoprotein content in the plasma due to low insulin levels. Due to a lack of insulin, lipolysis increases in fat cells [ 2 , 13 , 15 ]. Of note, the converse has also been observed as a phenomenon known as carbohydrate-induced hypertriglyceridemia, whereby higher dietary carbohydrate intake leads to higher serum triglycerides levels, potentially mediated by changes in triglyceride clearance and hepatic de novo lipogenesis rates [ 64 ]. Though our aggregate results also confirm an increase in LDL-C and total cholesterol with KD and K-LCHF, respectively, it is important to note that an increase in either of these levels does not necessarily signify a potentially deleterious cardiovascular end-point. This qualification derives from the fact that LDL particles are widely heterogeneous in composition and size, with small dense LDL particles being significantly more atherogenic than larger LDL particles [ 65 ]. Our observed aggregate effect of KD on cholesterol levels does not account for the difference in LDL particle size, nor does it distinguish the sources of dietary fat, which can also be a significant effector of LDL particle size distribution and metabolism [ 66 ].

Most RCTs of KD were conducted in patients with a limited group of participants, such as those with overweight, obesity, metabolic syndrome, cancer, and refractory epilepsy. In addition, most outcomes measured were limited to only surrogate outcomes. Thus, more clinical trials with a broader scope in populations and outcomes associated with KD would expand the role of KD in a clinical setting. For example, participant selection could be expanded from previous trials to include elderly patients, nonalcoholic fatty live disease (NAFLD) patients, and polycystic ovarian syndrome patients. Outcomes of interest of could be expanded to include (1) clinical outcomes such as cardiovascular events and liver outcomes, (2) short- and long-term safety outcomes such as adverse events (e.g., gastrointestinal, neurological, hepatic, and renal), eating disorder syndrome, sleep parameters, lipid profiles, and thyroid function and (3) other outcomes such as adherence and quality of life. More importantly, long-term studies are needed to investigate the sustainability of the clinical benefits of KD.

Our findings are useful to support the generation of evidence-based recommendations for clinicians contemplating use of KD in their patients, as well as for the general population. We further emphasize the importance of consultation with healthcare professionals before utilizing KD and any other dietary interventions. We demonstrated the benefits of KD on various outcomes in the short term. However, these improvements may prove difficult to sustain in the long term because of challenges in adherence. As for any diet interventions to achieve sustainable weight loss, factors of success include adherence, negative energy balance, and high-quality foods. Thus, communication and education with KD practitioners are important to ensure their adherence to the diet. Some individuals might benefit from switching from KD to other dietary interventions to maintain long-term weight loss.

Limitations

This umbrella review has several limitations. Firstly, we focused on published meta-analyses which confined us from assessing the associations of KD on outcomes and populations that were not included in existing meta-analyses. Secondly, most of the included meta-analyses were rated with AMSTAR-2 as critically low confidence, mainly due to a lack of study exclusion reasons, unexplained study heterogeneity, and unassessed publication bias. However, these domains unlikely affected our findings. Thirdly, we could not perform a dose–response analysis to understand the effects of different levels of carbohydrate intake on health outcomes because of insufficient details of carbohydrate intake reported in the meta-analyses. Fourthly, most RCTs of KD were limited to a relatively small number of participants with a short-term follow-up period, which limited our assessment of sustained beneficial effects after stopping KD. Lastly, due to decreased adherence, carbohydrate intake most likely increased across the course of the trials. For example, subjects in the KD arm of the A TO Z Weight Loss Study [ 67 ], started with a carbohydrate intake < 10 g/day but ended at 12 months with a carbohydrate intake accounting for 34% of TEI. In the DIRECT trial, subjects in the KD group started with carbohydrate intake of 20 g/day and ended at 12 months with 40% of TEI from carbohydrate intake [ 68 ]. Thus, we cannot be certain how the precise degree of ketosis contributed to the beneficial effects noted.

Beneficial associations of practicing KD were supported by moderate- to high-quality evidence, including weight loss, lower triglyceride levels, decreased HbA 1c , RER, and decreased seizure frequency. However, KD was associated with a clinically meaningful increase in LDL-C. Clinical trials with long-term follow-up are warranted to investigate whether these short-term effects of KD will translate to beneficial effects on more long-term clinical outcomes such as cardiovascular events and mortality.

Availability of data and materials

All data generated or analysed during this study are included in this published article and its supplementary information files.

Abbreviations

Beta-hydroxybutyrate

Body mass index

Diastolic blood pressure

Gamma-aminobutyric acid

High-density lipoprotein cholesterol

Hemoglobin A 1c

Homeostatic model assessment of insulin resistance

Ketogenic low-carbohydrate high-fat diet

Ketogenic diets

Low-density lipoprotein cholesterol

Modified Atkins diet

Minimally clinically important difference

Nonalcoholic fatty liver disease

Randomized clinical trials

Respiratory exchange ratio

Systolic blood pressure

Type 2 diabetes mellitus

Total energy intake

Very low-calorie ketogenic diet

Maximal oxygen consumption

Bueno NB, de Melo IS, de Oliveira SL, da Rocha Ataide T. Very-low-carbohydrate ketogenic diet v. low-fat diet for long-term weight loss: a meta-analysis of randomised controlled trials. Br J Nutr. 2013;110(7):1178–87.

Article   CAS   PubMed   Google Scholar  

Choi YJ, Jeon SM, Shin S. Impact of a ketogenic diet on metabolic parameters in patients with obesity or overweight and with or without type 2 diabetes: a meta-analysis of randomized controlled trials. Nutrients. 2020;12(7):2005.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Kirkpatrick CF, Bolick JP, Kris-Etherton PM, Sikand G, Aspry KE, Soffer DE, et al. Review of current evidence and clinical recommendations on the effects of low-carbohydrate and very-low-carbohydrate (including ketogenic) diets for the management of body weight and other cardiometabolic risk factors: a scientific statement from the National Lipid Association Nutrition and Lifestyle Task Force. J Clin Lipidol. 2019;13(5):689-711.e1.

Article   PubMed   Google Scholar  

Dahlin M, Singleton SS, David JA, Basuchoudhary A, Wickström R, Mazumder R, et al. Higher levels of Bifidobacteria and tumor necrosis factor in children with drug-resistant epilepsy are associated with anti-seizure response to the ketogenic diet. eBioMedicine. 2022;80:104061.

Crosby L, Davis B, Joshi S, Jardine M, Paul J, Neola M, et al. Ketogenic diets and chronic disease: weighing the benefits against the risks. Front Nutr. 2021;8:702802.

Article   PubMed   PubMed Central   Google Scholar  

Paoli A, Rubini A, Volek JS, Grimaldi KA. Beyond weight loss: a review of the therapeutic uses of very-low-carbohydrate (ketogenic) diets. Eur J Clin Nutr. 2013;67(8):789–96.

Gershuni VM, Yan SL, Medici V. Nutritional ketosis for weight management and reversal of metabolic syndrome. Curr Nutr Rep. 2018;7(3):97–106.

Zhu H, Bi D, Zhang Y, Kong C, Du J, Wu X, et al. Ketogenic diet for human diseases: the underlying mechanisms and potential for clinical implementations. Signal Transduct Target Ther. 2022;7(1):11.

Lefevre F, Aronson N. Ketogenic diet for the treatment of refractory epilepsy in children: a systematic review of efficacy. Pediatrics. 2000;105(4):E46.

Kossoff EH, Zupec-Kania BA, Auvin S, Ballaban-Gil KR, Christina Bergqvist AG, Blackford R, et al. Optimal clinical management of children receiving dietary therapies for epilepsy: Updated recommendations of the International Ketogenic Diet Study Group. Epilepsia Open. 2018;3(2):175–92.

Bough KJ, Rho JM. Anticonvulsant mechanisms of the ketogenic diet. Epilepsia. 2007;48(1):43–58.

Yudkoff M, Daikhin Y, Melø TM, Nissim I, Sonnewald U, Nissim I. The ketogenic diet and brain metabolism of amino acids: relationship to the anticonvulsant effect. Annu Rev Nutr. 2007;27:415–30.

Yuan X, Wang J, Yang S, Gao M, Cao L, Li X, et al. Effect of the ketogenic diet on glycemic control, insulin resistance, and lipid metabolism in patients with T2DM: a systematic review and meta-analysis. Nutr Diabetes. 2020;10(1):38.

Goldenberg JZ, Day A, Brinkworth GD, Sato J, Yamada S, Jönsson T, et al. Efficacy and safety of low and very low carbohydrate diets for type 2 diabetes remission: systematic review and meta-analysis of published and unpublished randomized trial data. BMJ. 2021;372:m4743.

Castellana M, Conte E, Cignarelli A, Perrini S, Giustina A, Giovanella L, et al. Efficacy and safety of very low calorie ketogenic diet (VLCKD) in patients with overweight and obesity: a systematic review and meta-analysis. Rev Endocr Metab Disord. 2020;21(1):5–16.

Sourbron J, Klinkenberg S, van Kuijk SMJ, Lagae L, Lambrechts D, Braakman HMH, et al. Ketogenic diet for the treatment of pediatric epilepsy: review and meta-analysis. Childs Nerv Syst. 2020;36(6):1099–109.

Lee HS, Lee J. Influences of ketogenic diet on body fat percentage, respiratory exchange rate, and total cholesterol in athletes: a systematic review and meta-analysis. Int J Environ Res Public Health. 2021;18(6):2912.

Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71.

Dragioti E, Solmi M, Favaro A, Fusar-Poli P, Dazzan P, Thompson T, et al. Association of antidepressant use with adverse health outcomes: a systematic umbrella review. JAMA Psychiat. 2019;76(12):1241–55.

Article   Google Scholar  

Patikorn C, Roubal K, Veettil SK, Chandran V, Pham T, Lee YY, et al. Intermittent Fasting and Obesity-Related Health Outcomes: An Umbrella Review of Meta-analyses of Randomized Clinical Trials. JAMA Netw Open. 2021;4(12):e2139558-e.

Theodoratou E, Tzoulaki I, Zgaga L, Ioannidis JP. Vitamin D and multiple health outcomes: umbrella review of systematic reviews and meta-analyses of observational studies and randomised trials. BMJ. 2014;348: g2035.

Shea BJ, Reeves BC, Wells G, Thuku M, Hamel C, Moran J, et al. AMSTAR 2: a critical appraisal tool for systematic reviews that include randomised or non-randomised studies of healthcare interventions, or both. BMJ. 2017;358:j4008.

DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7(3):177–88.

Higgins JP, Thompson SG, Spiegelhalter DJ. A re-evaluation of random-effects meta-analysis. J R Stat Soc Ser A Stat Soc. 2009;172(1):137–59.

Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ. 1997;315(7109):629–34.

Heymsfield SB, Gonzalez MC, Shen W, Redman L, Thomas D. Weight loss composition is one-fourth fat-free mass: a critical review and critique of this widely cited rule. Obes Rev. 2014;15(4):310–21.

Mulligan AA, Lentjes MA, Luben RN, Wareham NJ, Khaw K-T. Changes in waist circumference and risk of all-cause and CVD mortality: results from the European Prospective Investigation into Cancer in Norfolk (EPIC-Norfolk) cohort study. BMC Cardiovasc Disord. 2019;19(1):1–15.

Jayedi A, Khan TA, Aune D, Emadi A, Shab-Bidar S. Body fat and risk of all-cause mortality: a systematic review and dose-response meta-analysis of prospective cohort studies. Int J Obes. 2022;46(9):1573–81.

Article   CAS   Google Scholar  

Keller HH, Østbye T. Body mass index (BMI), BMI change and mortality in community-dwelling seniors without dementia. J Nutr Health Aging. 2005;9(5):316–20.

CAS   PubMed   Google Scholar  

Kelley GA, Kelley KS, Stauffer BL. Walking and resting blood pressure: an inter-individual response difference meta-analysis of randomized controlled trials. Sci Prog. 2022;105(2):00368504221101636.

Langendam MW, Akl EA, Dahm P, Glasziou P, Guyatt G, Schünemann HJ. Assessing and presenting summaries of evidence in cochrane reviews. Syst Rev. 2013;2(1):81.

Dechartres A, Altman DG, Trinquart L, Boutron I, Ravaud P. Association between analytic strategy and estimates of treatment outcomes in meta-analyses. JAMA. 2014;312(6):623–30.

Sterne JA, Savović J, Page MJ, Elbers RG, Blencowe NS, Boutron I, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ. 2019;366:l4898.

Sainsbury E, Kizirian NV, Partridge SR, Gill T, Colagiuri S, Gibson AA. Effect of dietary carbohydrate restriction on glycemic control in adults with diabetes: a systematic review and meta-analysis. Diabetes Res Clin Pract. 2018;139:239–52.

Rafiullah M, Musambil M, David SK. Effect of a very low-carbohydrate ketogenic diet vs recommended diets in patients with type 2 diabetes: a meta-analysis. Nutr Rev. 2022;80(3):488–502.

Alarim RA, Alasmre FA, Alotaibi HA, Alshehri MA, Hussain SA. Effects of the ketogenic diet on glycemic control in diabetic patients: meta-analysis of clinical trials. Cureus. 2020;12(10):e10796.

PubMed   PubMed Central   Google Scholar  

Amini MR, Aminianfar A, Naghshi S, Larijani B, Esmaillzadeh A. The effect of ketogenic diet on body composition and anthropometric measures: A systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr. 2022;62(13):3644–57.

Ashtary-Larky D, Bagheri R, Asbaghi O, Tinsley GM, Kooti W, Abbasnezhad A, et al. Effects of resistance training combined with a ketogenic diet on body composition: a systematic review and meta-analysis. Crit Rev Food Sci Nutr. 2022;62(21):5717–32.

Cao J, Lei S, Wang X, Cheng S. The effect of a ketogenic low-carbohydrate, high-fat diet on aerobic capacity and exercise performance in endurance athletes: a systematic review and meta-analysis. Nutrients. 2021;13(8):2896.

Lee HS, Lee J. Effects of combined exercise and low carbohydrate ketogenic diet interventions on waist circumference and triglycerides in overweight and obese individuals: a systematic review and meta-analysis. Int J Environ Res Public Health. 2021;18(2):828.

López-Espinoza M, Chacón-Moscoso S, Sanduvete-Chaves S, Ortega-Maureira MJ, Barrientos-Bravo T. Effect of a ketogenic diet on the nutritional parameters of obese patients: a systematic review and meta-analysis. Nutrients. 2021;13(9):2946.

Muscogiuri G, El Ghoch M, Colao A, Hassapidou M, Yumuk V, Busetto L. European guidelines for obesity management in adults with a very low-calorie ketogenic diet: a systematic review and meta-analysis. Obes Facts. 2021;14(2):222–45.

Smith ES, Smith HA, Betts JA, Gonzalez JT, Atkinson G. A Systematic review and meta-analysis comparing heterogeneity in body mass responses between low-carbohydrate and low-fat diets. Obesity. 2020;28(10):1833–42.

Yang YF, Mattamel PB, Joseph T, Huang J, Chen Q, Akinwunmi BO, et al. Efficacy of low-carbohydrate ketogenic diet as an adjuvant cancer therapy: a systematic review and meta-analysis of randomized controlled trials. Nutrients. 2021;13(5):1388.

Vargas-Molina S, Gómez-Urquiza JL, García-Romero J, Benítez-Porres J. Effects of the ketogenic diet on muscle hypertrophy in resistance-trained men and women: a systematic review and meta-analysis. Int J Environ Res Public Health. 2022;19(19):12629.

El-Rashidy OF, Nassar MF, Abdel-Hamid IA, Shatla RH, Abdel-Hamid MH, Gabr SS, et al. Modified Atkins diet vs classic ketogenic formula in intractable epilepsy. Acta Neurol Scand. 2013;128(6):402–8.

Kossoff EH, McGrogan JR, Bluml RM, Pillas DJ, Rubenstein JE, Vining EP. A modified Atkins diet is effective for the treatment of intractable pediatric epilepsy. Epilepsia. 2006;47(2):421–4.

Bough KJ, Wetherington J, Hassel B, Pare JF, Gawryluk JW, Greene JG, et al. Mitochondrial biogenesis in the anticonvulsant mechanism of the ketogenic diet. Ann Neurol. 2006;60(2):223–35.

Kim DY, Simeone KA, Simeone TA, Pandya JD, Wilke JC, Ahn Y, et al. Ketone bodies mediate antiseizure effects through mitochondrial permeability transition. Ann Neurol. 2015;78(1):77–87.

Garriga-Canut M, Schoenike B, Qazi R, Bergendahl K, Daley TJ, Pfender RM, et al. 2-Deoxy-D-glucose reduces epilepsy progression by NRSF-CtBP–dependent metabolic regulation of chromatin structure. Nat Neurosci. 2006;9(11):1382–7.

Wang ZJ, Bergqvist C, Hunter JV, Jin D, Wang DJ, Wehrli S, et al. In vivo measurement of brain metabolites using two-dimensional double-quantum MR spectroscopy—exploration of GABA levels in a ketogenic diet. Magn Reson Med. 2003;49(4):615–9.

Olson CA, Vuong HE, Yano JM, Liang QY, Nusbaum DJ, Hsiao EY. The gut microbiota mediates the anti-seizure effects of the ketogenic diet. Cell. 2018;173(7):1728-41.e13.

Dupuis N, Curatolo N, Benoist JF, Auvin S. Ketogenic diet exhibits anti-inflammatory properties. Epilepsia. 2015;56(7):e95–8.

Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, et al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. 2013;339(6116):211–4.

O’Neill B, Raggi P. The ketogenic diet: Pros and cons. Atherosclerosis. 2020;292:119–26.

Freire R. Scientific evidence of diets for weight loss: different macronutrient composition, intermittent fasting, and popular diets. Nutrition. 2020;69: 110549.

Becker A, Gaballa D, Roslin M, Gianos E, Kane J. Novel nutritional and dietary approaches to weight loss for the prevention of cardiovascular disease: ketogenic diet, intermittent fasting, and bariatric surgery. Curr Cardiol Rep. 2021;23(7):1–9.

Ashtary-Larky D, Bagheri R, Bavi H, Baker JS, Moro T, Mancin L, et al. Ketogenic diets, physical activity and body composition: a review. Br J Nutr. 2022;127(12):1898–920.

Thomsen HH, Rittig N, Johannsen M, Møller AB, Jørgensen JO, Jessen N, et al. Effects of 3-hydroxybutyrate and free fatty acids on muscle protein kinetics and signaling during LPS-induced inflammation in humans: anticatabolic impact of ketone bodies. Am J Clin Nutr. 2018;108(4):857–67.

Koutnik AP, D’Agostino DP, Egan B. Anticatabolic effects of ketone bodies in skeletal muscle. Trends Endocrinol Metab. 2019;30(4):227–9.

Parker BA, Walton CM, Carr ST, Andrus JL, Cheung EC, Duplisea MJ, et al. β-hydroxybutyrate elicits favorable mitochondrial changes in skeletal muscle. Int J Mol Sci. 2018;19(8):2247.

Huang Z, Huang L, Waters MJ, Chen C. Insulin and growth hormone balance: implications for obesity. Trends Endocrinol Metab. 2020;31(9):642–54.

Møller N, Copeland KC, Nair KS. Growth hormone effects on protein metabolism. Endocrinol Metab Clin North Am. 2007;36(1):89–100.

Parks EJ. Effect of dietary carbohydrate on triglyceride metabolism in humans. J Nutr. 2001;131(10):2772S-S2774.

Lamarche B, Tchernof A, Moorjani S, Cantin B, Dagenais GR, Lupien PJ, et al. Small, dense low-density lipoprotein particles as a predictor of the risk of ischemic heart disease in men: prospective results from the Qué bec Cardiovascular Study. Circulation. 1997;95(1):69–75.

Dreon DM, Fernstrom HA, Campos H, Blanche P, Williams PT, Krauss RM. Change in dietary saturated fat intake is correlated with change in mass of large low-density-lipoprotein particles in men. Am J Clin Nutr. 1998;67(5):828–36.

Gardner CD, Kiazand A, Alhassan S, Kim S, Stafford RS, Balise RR, et al. Comparison of the Atkins, Zone, Ornish, and LEARN diets for change in weight and related risk factors among overweight premenopausal women: the A TO Z weight loss study: a randomized trial. JAMA. 2007;297(9):969–77.

Shai I, Schwarzfuchs D, Henkin Y, Shahar DR, Witkow S, Greenberg I, et al. Weight loss with a low-carbohydrate, Mediterranean, or low-fat diet. N Engl J Med. 2008;359(3):229–41.

Download references

Acknowledgements

The authors would like to acknowledge Thunchanok Ingkaprasert and Wachiravit Youngjanin for their editorial assistance.

No funding was obtained for the conduct of this study.

Author information

Authors and affiliations.

Department of Pharmacotherapy, College of Pharmacy, University of Utah, 30 2000 E, Salt Lake City, Utah, 84112, USA

Chanthawat Patikorn, Pantakarn Saidoung, Sajesh K. Veettil & Nathorn Chaiyakunapruk

Department of Social and Administrative Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand

Chanthawat Patikorn

Division of Gastroenterology, Hepatology & Nutrition, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA

Division of Ambulatory Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand

Pochamana Phisalprapa

School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia

Yeong Yeh Lee

Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, USA

Krista A. Varady

IDEAS Center, Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, Utah, USA

Nathorn Chaiyakunapruk

You can also search for this author in PubMed   Google Scholar

Contributions

CP, PS, SKV, and NC conceived and designed the study protocol. CP, PS, and SKV performed a literature review and data analysis. CP, PS, TP, PP, YYL, KAV, SKV, and NC interpreted the study findings. CP and PS were major contributors to writing the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Sajesh K. Veettil or Nathorn Chaiyakunapruk .

Ethics declarations

Ethics approval and consent to participate.

Not applicable.

Consent for publication

Competing interests.

The authors declare that they have no competing interests.

Additional information

Publisher’s note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1..

 PRISMA 2020 Main Checklist. 

Additional file 2:

Method S1. Data extraction. Table S1. Difference from original review protocol. Table S2. Search strategy. Table S3. Excluded studies with reasons.  Table S4. Quality assessment. Table S5. Summary of associations. Table S6. Sensitivity analyses. 

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ . The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Cite this article.

Patikorn, C., Saidoung, P., Pham, T. et al. Effects of ketogenic diet on health outcomes: an umbrella review of meta-analyses of randomized clinical trials. BMC Med 21 , 196 (2023). https://doi.org/10.1186/s12916-023-02874-y

Download citation

Received : 25 November 2022

Accepted : 19 April 2023

Published : 25 May 2023

DOI : https://doi.org/10.1186/s12916-023-02874-y

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Ketegenic diet
  • Umbrella review
  • Systematic review
  • Meta-analysis
  • Weight loss

BMC Medicine

ISSN: 1741-7015

ketogenic diet thesis statement

Ketogenic Diet: Overview Presentation

  • To find inspiration for your paper and overcome writer’s block
  • As a source of information (ensure proper referencing)
  • As a template for you assignment

Diet as the Way to Lose Weight

  • Many people want to lose weight.
  • Every person has their reasons.
  • The issue of weight loss is relevant, but requires careful monitoring.

Often, many people think about losing extra weight. Every person has their reasons for doing so – one wants to lead a healthier life, while others want to attract people. One way or another, the questions of weight loss are acute for humanity; for this reason, quite often among the already known classic methods of weight loss new and radical options that can aggravate human health appear. In order to prevent unnecessary health issues and achieve the desired effect, nutritionists offer patients to use the rules of the popular ketogenic diet.

Diet as the Way to Lose Weight

Everything New is Actually Well-forgotten Old

  • In the early twentieth century, ketogenic diet was used to treat epilepsy.
  • Carbohydrate deficiency in the diet caused insulin reduction.
  • With the advent of drugs, the diet has been relegated to the background.
  • It was only thirty years ago that nutritionists rediscovered the ketogenic diet.

Recently, ketogenic diets have been used by a large number of young and adult people, and this method of starvation has become incredibly popular. However, if one looks at the historical context of ketogenic diets, it is clear that the diet has a rather long and mixed history. This diet was first introduced in the early 20th century to treat diseases of the nervous system, particularly epilepsy (Rho 5). A prerequisite for the appearance of the diet was the use of fasting: when food ceased to arrive, insulin stopped being produced in the body, significantly affecting the central nervous system (Rho 5). Such fasting was indeed valid, but it was impossible to use it for a long time. The situation was particularly difficult for children, as deprivation of food posed a serious health hazard (Rho 6). At that time, a diet similar to healing fasting was developed, but it gave the body energy from fats. It showed excellent results – people with epilepsy practically stopped having seizures. Indeed, the success of the ketogenic diet was not celebrated for long: soon, there were specially developed drugs, as a result of which the diet was forgotten for some time. It was only in the 1990s, when the problem of non-universal antiepileptic drugs for patients matured, that such a diet became discussed again. Information about ketogenic diets started to spread in the media, and the “new” weight-loss remedy began to be popularized.

Everything New is Actually Well-forgotten Old

The Mechanism of the Diet

The rule is simple: lack of carbohydrates leads to burning of fats. The body draws energy from burning its own deposits or with food – as a result, the body weight is reduced.

The meaning of the ketogenic diet is that when carbohydrates are stopped when glucose concentration in blood decreases, the body searches for other sources of energy and burns fat. This process involves both fats from food and the body’s supplies. In the reorganization of diet, the liver produces ketone bodies (acetoacetate), which participate in the oxidation of fatty acids and are used as energy by many organs, including the brain. This condition is called ketosis (Yancy, Mitchell, & Westman 1734). As glucose disappears from the blood, the production of insulin, a hormone that prevents fat burning, is reduced.

The Mechanism of the Diet

Scientific Evidence

  • There are many scientific proofs of the optimality of this model.
  • Zajac et al. confirmed the short-term effect of the diet on athletes.
  • Carbohydrate elimination strengthens muscles, normalizes the biochemical composition and helps to quickly lose weight.

The decision to use any diet must be meaningful, and the effectiveness of starvation has been scientifically proven. For example, Zajac et al. found that a ketogenic diet has a positive effect on the performance of athletes (2496). The study determined what changes would result from changing diets to predominantly fatty young cyclists. Scientists concluded that the short-term effect of a ketogenic diet could preserve muscle structures in athletes after exercise, reduce body weight, and improve biochemical composition. An incredible achievement of Zajac et al. is the discovery that a ketogenic diet has a positive effect on a person’s breathing ability (2498).

Scientific Evidence

Who is Ketogenic Diet Good for

  • Meat lovers;
  • Those who want to save muscle mass;
  • Patients with diabetes mellitus.

Despite the complexity of the first days and strict diet restrictions, for some people’s ketogenic diets are ideal. It is worth trying out for the following categories of people who want to lose weight:

  • Meat lovers. Many diets offer a drastic reduction in meat-eating or complete rejection, but a ketogenic diet does not restrict it.
  • Those who want to save muscle mass. Normally diets can reduce muscle mass, but a ketogenic diet does not destroy muscle fiber structures, which is more suitable for professional athletes.
  • Patients with diabetes mellitus. Ketosis can control blood sugar levels (Yancy, Mitchell, & Westman 1734). It is a type of professional diet, so a doctor’s consultation is necessary.

Who is Ketogenic Diet Good for

Mistakes that are Made

Lack of education can lead to:

  • Carbohydrate consumption.
  • Consumption of large quantities of proteins.

Due to their lack of education, beginners often make typical mistakes when using a ketogenic diet. Most of these errors will neutralize the positive effects of the diet, but others can have adverse effects on health. The most common mistake is when a person neglects the basic rule of diet and eats small amounts of carbohydrates. However, it is essential to understand that even a slice of eaten bread delays the start of ketosis. The second error is the opposite: people often completely give up carbohydrates, but in addition, they increase the amount of protein they consume. Protein poisoning leads to the destruction of the liver, kidneys, and digestive system problems.

Mistakes that are Made

The most challenging part of the keto diet is getting used to an entirely new way of eating, especially the rejection of most foods with high carbohydrate content and the addition of fat. Large amounts of natural fat can be found in vegetable and animal oils. Low-carbohydrate meat products may include beef, poultry and eggs, and fish. Non-starchy foods such as avocados, tomatoes, cabbage, and broccoli are allowed types of vegetables. On a ketogenic diet, it is possible to eat fruits and berries such as raspberries, kiwi, lemons, and blueberries.

Basic Diet

What Does a Person Get: Arguments “For”

  • The scientific validity of diet.
  • Rapidity of fat burning.
  • Comfortable process.
  • Long-term result.
  • Cholesterol Control.
  • Reducing epileptic seizures.
  • Improving skin condition.

Those who decide to use ketogenic diet rules for weight loss should understand the benefits that await them. First, the effectiveness of such a diet has been scientifically proven (Zajac et al. 2493). Secondly, with such a diet, a person burns excess fat very quickly. Also, unlike other diets, the patient has no sense of hunger, so fasting is comfortable. Also, it should be noted that the result obtained is quite long term as the person is not hungry, and the body is not stressed. Several scientific studies demonstrate that a ketogenic diet improves cholesterol levels (“Pros and Cons” 1). This reduces the likelihood of cardiovascular disease. In addition, it should be remembered that a diet is right for people with epilepsy – the number of seizures and convulsions decreases (Feldman 36). In the world of cosmetology, there is a belief that a ketogenic diet has a positive effect on skin health by inhibiting the growth of acne and pimples (Feldman 36).

What Does a Person Get: Arguments “For”

What a Person will Face: Arguments “Against”

  • This diet is unbalanced
  • Like any diet, it requires willpower.
  • It is quite long, can be used for years.
  • Side effects are possible.

It is essential to understand that there are no ideal weight loss models. A ketogenic diet is rich in deficiencies that can significantly worsen a person’s condition (Pros and Cons” 2). For this reason, it is necessary to consult a nutritionist before using such a diet.

  • First, it should be noted that the ketogenic diet is unbalanced – it excludes the intake of several nutrients, vitamins, and trace elements.
  • Secondly, it requires motivation and responsibility – one has to give up favorite dishes and regularly calculate the number of calories.
  • The third disadvantage of the diet is duration: a ketogenic diet can last from 3-4 weeks to a year. There is no point in staying on a diet for less than three weeks because, during this time, the body will go through ketogenic adaptation and only begin to get all the benefits of this diet.
  • It also has many side effects: constipation, nausea and vomiting, growth disorders, kidney stones, and changes in blood lipids.

What a Person will Face: Arguments “Against”

What Else Should be Mentioned

What must be said is :

  • This diet is not suitable for pregnant and sick patients.
  • Long-term effects require careful consideration.
  • Each diet is selected individually and requires analysis by a specialist.

The ketogenic diet has another disadvantage, which, however, cannot be unequivocally attributed to the general disadvantages. The fact is that this type of diet is not suitable for all population groups, and is contraindicated for medical reasons to pregnant women and people with kidney and liver diseases (Feldman 34). The effects of a long-term diet on healthy people have not yet been well understood. Dieticians are advised to choose a diet that can be sustained throughout life. Short-term effects may be successful, but the method is not suitable for everyone. Dietary recommendations are tailored to each individual and following the next trend without consulting a doctor is not recommended.

What Else Should be Mentioned

Works Cited

Feldman, Ellen. “Ketogenic Diet for Refractory Pediatric Seizures.” Integrative Medicine Alert , vol. 22, no. 9, 2019, pp. 32-37.

Mawer, Rudy. “ 10 Graphs That Show the Power of a Ketogenic Diet. ” heathline . 2018. Web.

Mawer, Rudy. “ The Ketogenic Diet: A Detailed Beginner’s Guide to Keto. ” heathline . 2018. Web.

“Pros and Cons of Low-Carb/Ketogenic Diets.” Nutrition Letter , vol. 37, no. 10, 2019, pp. 1-2.

Rho, Jong M. “How Does the Ketogenic Diet Induce Anti-Seizure Effects?” Neuroscience letters , vol. 637, no. 1, 2017, pp. 4-10.

Yancy, William S., Nia S. Mitchell, and Eric C. Westman. “Ketogenic Diet for Obesity and Diabetes.” JAMA Internal Medicine , vol. 179, no. 12, 2019, pp. 1734-1735.

Zajac, Adam, et al. “The Effects of a Ketogenic Diet on Exercise Metabolism and Physical Performance in Off-Road Cyclists.” Nutrients , vol. 6, no. 7, 2014, pp. 2493-2508.

  • How to Making a Healthy Snack
  • Why You Should Not Be a Vegetarian
  • Going Keto: Is It Worth It?
  • Very-Low-Calorie Ketogenic Diet and Kidney Failure
  • Analysis of Low Carb and High-Fat Diets
  • Canada Food Guide Overview
  • Dietary Specifications: Medicinal Meals in China
  • Steps in Ensuring a Healthy Diet in College
  • The Process of Cheese Production
  • Unhealthy Fad Diets: Fact or Hoax
  • Chicago (A-D)
  • Chicago (N-B)

IvyPanda. (2022, July 15). Ketogenic Diet: Overview. https://ivypanda.com/essays/ketogenic-diet-overview/

"Ketogenic Diet: Overview." IvyPanda , 15 July 2022, ivypanda.com/essays/ketogenic-diet-overview/.

IvyPanda . (2022) 'Ketogenic Diet: Overview'. 15 July.

IvyPanda . 2022. "Ketogenic Diet: Overview." July 15, 2022. https://ivypanda.com/essays/ketogenic-diet-overview/.

1. IvyPanda . "Ketogenic Diet: Overview." July 15, 2022. https://ivypanda.com/essays/ketogenic-diet-overview/.

Bibliography

IvyPanda . "Ketogenic Diet: Overview." July 15, 2022. https://ivypanda.com/essays/ketogenic-diet-overview/.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

The Ketogenic Diet and Cardiovascular Diseases

Affiliation.

  • 1 Institute of Health Sciences, Faculty of Medical and Health Sciences, Siedlce University of Natural Sciences and Humanities, 08-110 Siedlce, Poland.
  • PMID: 37571305
  • PMCID: PMC10421332
  • DOI: 10.3390/nu15153368

The most common and increasing causes of death worldwide are cardiovascular diseases (CVD). Taking into account the fact that diet is a key factor, it is worth exploring this aspect of CVD prevention and therapy. The aim of this article is to assess the potential of the ketogenic diet in the prevention and treatment of CVD. The article is a comprehensive, meticulous analysis of the literature in this area, taking into account the most recent studies currently available. The ketogenic diet has been shown to have a multifaceted effect on the prevention and treatment of CVD. Among other aspects, it has a beneficial effect on the blood lipid profile, even compared to other diets. It shows strong anti-inflammatory and cardioprotective potential, which is due, among other factors, to the anti-inflammatory properties of the state of ketosis, the elimination of simple sugars, the restriction of total carbohydrates and the supply of omega-3 fatty acids. In addition, ketone bodies provide "rescue fuel" for the diseased heart by affecting its metabolism. They also have a beneficial effect on the function of the vascular endothelium, including improving its function and inhibiting premature ageing. The ketogenic diet has a beneficial effect on blood pressure and other CVD risk factors through, among other aspects, weight loss. The evidence cited is often superior to that for standard diets, making it likely that the ketogenic diet shows advantages over other dietary models in the prevention and treatment of cardiovascular diseases. There is a legitimate need for further research in this area.

Keywords: CVD; HDL; KD; LDL; blood pressure; cardiomyocytes; cardiovascular disease; cholesterol; diastolic; diet; endothelium; fatty acids; heart; heart metabolism; high fat; inflammation; ketogenic diet; ketone; ketone bodies; lipid profile; low carb; obesity; prevention; reduction; systolic; treatment; triglyceride; weight loss.

PubMed Disclaimer

Conflict of interest statement

The authors declare no conflict of interest.

Four main factors influencing the…

Four main factors influencing the anti-inflammatory, cardioprotective potential of the ketogenic diet. The…

Influence of the ketogenic diet…

Influence of the ketogenic diet on vascular endothelial cells. The above figure was…

Similar articles

  • Activation of G protein-coupled receptors by ketone bodies: Clinical implication of the ketogenic diet in metabolic disorders. Spigoni V, Cinquegrani G, Iannozzi NT, Frigeri G, Maggiolo G, Maggi M, Parello V, Dei Cas A. Spigoni V, et al. Front Endocrinol (Lausanne). 2022 Oct 20;13:972890. doi: 10.3389/fendo.2022.972890. eCollection 2022. Front Endocrinol (Lausanne). 2022. PMID: 36339405 Free PMC article. Review.
  • High-fat ketogenic diets and ketone monoester supplements differentially affect substrate metabolism during aerobic exercise. Margolis LM, Pasiakos SM, Howard EE. Margolis LM, et al. Am J Physiol Cell Physiol. 2023 Oct 1;325(4):C1144-C1153. doi: 10.1152/ajpcell.00359.2023. Epub 2023 Sep 18. Am J Physiol Cell Physiol. 2023. PMID: 37721006 Review.
  • Novel Nutritional and Dietary Approaches to Weight Loss for the Prevention of Cardiovascular Disease: Ketogenic Diet, Intermittent Fasting, and Bariatric Surgery. Becker A, Gaballa D, Roslin M, Gianos E, Kane J. Becker A, et al. Curr Cardiol Rep. 2021 Jun 3;23(7):85. doi: 10.1007/s11886-021-01515-1. Curr Cardiol Rep. 2021. PMID: 34081228 Review.
  • Ketogenic diet and cardiovascular risk - state of the art review. Popiolek-Kalisz J. Popiolek-Kalisz J. Curr Probl Cardiol. 2024 Mar;49(3):102402. doi: 10.1016/j.cpcardiol.2024.102402. Epub 2024 Jan 15. Curr Probl Cardiol. 2024. PMID: 38232923 Review.
  • Ketogenic Diets and Exercise Performance. Harvey KL, Holcomb LE, Kolwicz SC Jr. Harvey KL, et al. Nutrients. 2019 Sep 26;11(10):2296. doi: 10.3390/nu11102296. Nutrients. 2019. PMID: 31561520 Free PMC article. Review.
  • Reply to Najjar, R.S. Comment on "Dyńka et al. The Ketogenic Diet and Cardiovascular Diseases. Nutrients 2023, 15 , 3368". Dyńka D, Kowalcze K, Charuta A, Paziewska A. Dyńka D, et al. Nutrients. 2023 Oct 10;15(20):4312. doi: 10.3390/nu15204312. Nutrients. 2023. PMID: 37892389 Free PMC article.
  • Comment on Dyńka et al. The Ketogenic Diet and Cardiovascular Diseases. Nutrients 2023, 15 , 3368. Najjar RS. Najjar RS. Nutrients. 2023 Oct 10;15(20):4311. doi: 10.3390/nu15204311. Nutrients. 2023. PMID: 37892387 Free PMC article.
  • Relationship between Dietary Decanoic Acid and Coronary Artery Disease: A Population-Based Cross-Sectional Study. Wu Z, Yang W, Li M, Li F, Gong R, Wu Y. Wu Z, et al. Nutrients. 2023 Oct 10;15(20):4308. doi: 10.3390/nu15204308. Nutrients. 2023. PMID: 37892384 Free PMC article.
  • Effects of Incretin-Based Treatment on the Diastolic (Dys)Function in Patients with Uncontrolled Type 2 Diabetes Mellitus: A Prospective Study with 1-Year Follow-Up. Grigorescu ED, Lăcătușu CM, Floria M, Cazac GD, Onofriescu A, Sauciuc LA, Ceasovschih A, Crețu I, Mihai BM, Șorodoc L. Grigorescu ED, et al. Diagnostics (Basel). 2023 Aug 31;13(17):2817. doi: 10.3390/diagnostics13172817. Diagnostics (Basel). 2023. PMID: 37685355 Free PMC article.
  • [(accessed on 4 March 2023)]. Available online: https://www.who.int/health-topics/cardiovascular-diseases#tab=tab_1 .
  • Tsao C.W., Aday A.W., Almarzooq Z.I., Alonso A., Beaton A.Z., Bittencourt M.S., Boehme A.K., Buxton A.E., Carson A.P., Commodore-Mensah Y., et al. Heart Disease and Stroke Statistics-2022 Update: A Report from the American Heart Association. Circulation. 2022;145:e153–e639. doi: 10.1161/CIR.0000000000001052. Erratum in Circulation 2022, 146, e141. - DOI - PubMed
  • Roth G.A., Mensah G.A., Johnson C.O., Addolorato G., Ammirati E., Baddour L.M., Barengo N.C., Beaton A.Z., Benjamin E.J., Benziger C.P., et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019: Update from the GBD 2019 Study. J. Am. Coll. Cardiol. 2020;76:2982–3021. doi: 10.1016/j.jacc.2020.11.010. - DOI - PMC - PubMed
  • [(accessed on 4 March 2023)]. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer .
  • Mathers C.D., Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006;3:e442. doi: 10.1371/journal.pmed.0030442. - DOI - PMC - PubMed

Publication types

  • Search in MeSH

Related information

Grants and funding.

  • Polish Ministry of Science and Higher Education within funds of Institute of Health Sciences, Faculty of Medical and Health Sciences, Siedlce University of Natural Sciences and Humanities./Polish Ministry of Science and Higher Education within funds of Institute of Health Sciences, Faculty of Medical and Health Sciences, Siedlce University of Natural Sciences and Humanities.

LinkOut - more resources

Full text sources.

  • Europe PubMed Central
  • PubMed Central

full text provider logo

  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Information

  • Author Services

Initiatives

You are accessing a machine-readable page. In order to be human-readable, please install an RSS reader.

All articles published by MDPI are made immediately available worldwide under an open access license. No special permission is required to reuse all or part of the article published by MDPI, including figures and tables. For articles published under an open access Creative Common CC BY license, any part of the article may be reused without permission provided that the original article is clearly cited. For more information, please refer to https://www.mdpi.com/openaccess .

Feature papers represent the most advanced research with significant potential for high impact in the field. A Feature Paper should be a substantial original Article that involves several techniques or approaches, provides an outlook for future research directions and describes possible research applications.

Feature papers are submitted upon individual invitation or recommendation by the scientific editors and must receive positive feedback from the reviewers.

Editor’s Choice articles are based on recommendations by the scientific editors of MDPI journals from around the world. Editors select a small number of articles recently published in the journal that they believe will be particularly interesting to readers, or important in the respective research area. The aim is to provide a snapshot of some of the most exciting work published in the various research areas of the journal.

Original Submission Date Received: .

  • Active Journals
  • Find a Journal
  • Proceedings Series
  • For Authors
  • For Reviewers
  • For Editors
  • For Librarians
  • For Publishers
  • For Societies
  • For Conference Organizers
  • Open Access Policy
  • Institutional Open Access Program
  • Special Issues Guidelines
  • Editorial Process
  • Research and Publication Ethics
  • Article Processing Charges
  • Testimonials
  • Preprints.org
  • SciProfiles
  • Encyclopedia

nutrients-logo

Article Menu

ketogenic diet thesis statement

  • Subscribe SciFeed
  • Recommended Articles
  • PubMed/Medline
  • Google Scholar
  • on Google Scholar
  • Table of Contents

Find support for a specific problem in the support section of our website.

Please let us know what you think of our products and services.

Visit our dedicated information section to learn more about MDPI.

JSmol Viewer

The ketogenic diet and cardiovascular diseases.

ketogenic diet thesis statement

Graphical Abstract

1. Introduction

2. the ketogenic diet and blood lipid profile, 2.1. lipid profile and cardiovascular diseases, 2.2. the effect of the ketogenic diet on the blood lipid profile, 3. anti-inflammatory potential of the ketogenic diet in cardiovascular diseases, 3.1. anti-inflammatory, cardioprotective potential of the state of ketosis (ketone bodies), 3.2. anti-inflammatory, cardioprotective effects of elimination of simple sugars, 3.3. anti-inflammatory, cardioprotective effects of total carbohydrate restriction, 3.4. anti-inflammatory, cardioprotective effects of omega-3 fatty acids, 4. ketone bodies and cardiac energy metabolism, 5. the ketogenic diet and the vascular endothelium, 6. the ketogenic diet and blood pressure, 7. the ketogenic diet and weight loss as a factor in cvd prevention and therapy, 8. the effect of the ketogenic diet among patients with cvd and healthy people, 9. conclusions, author contributions, institutional review board statement, informed consent statement, data availability statement, acknowledgments, conflicts of interest.

  • Available online: https://www.who.int/health-topics/cardiovascular-diseases#tab=tab_1 (accessed on 4 March 2023).
  • Tsao, C.W.; Aday, A.W.; Almarzooq, Z.I.; Alonso, A.; Beaton, A.Z.; Bittencourt, M.S.; Boehme, A.K.; Buxton, A.E.; Carson, A.P.; Commodore-Mensah, Y.; et al. Heart Disease and Stroke Statistics-2022 Update: A Report from the American Heart Association. Circulation 2022 , 145 , e153–e639, Erratum in Circulation 2022 , 146 , e141. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; Beaton, A.Z.; Benjamin, E.J.; Benziger, C.P.; et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019: Update from the GBD 2019 Study. J. Am. Coll. Cardiol. 2020 , 76 , 2982–3021. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Available online: https://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 4 March 2023).
  • Mathers, C.D.; Loncar, D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006 , 3 , e442. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Available online: https://www.cdc.gov/chronicdisease/resources/publications/factsheets/heart-disease-stroke.htm (accessed on 4 March 2023).
  • Vancheri, F.; Longo, G.; Vancheri, E.; Henein, M.Y. MentalStress and Cardiovascular Health—Part I. J. Clin. Med. 2022 , 11 , 3353. [ Google Scholar ] [ CrossRef ]
  • Cosentino, N.; Campodonico, J.; Milazzo, V.; De Metrio, M.; Brambilla, M.; Camera, M.; Marenzi, G. Vitamin D and Cardiovascular Disease: Current Evidence and Future Perspectives. Nutrients 2021 , 13 , 3603. [ Google Scholar ] [ CrossRef ]
  • Muzaffar, R.; Khan, M.A.; Mushtaq, M.H.; Nasir, M.; Khan, A.; Haq, I.U.; Muhammad, J. Hyperhomocysteinemia as an Independent Risk Factor for Coronary Heart Disease. Comparison with Conventional Risk Factors. Braz. J. Biol. 2021 , 83 , e249104. [ Google Scholar ] [ CrossRef ]
  • Khan, M.S.; Saeedullah, A.; Andrews, S.C.; Iqbal, K.; Qadir, S.A.; Shahzad, B.; Ahmed, Z.; Shahzad, M. Adolescent Afghan Refugees Display a High Prevalence of Hyperhomocysteinemia and Associated Micronutrients Deficiencies Indicating an Enhanced Risk of Cardiovascular Disease in Later Life. Nutrients 2022 , 14 , 1751. [ Google Scholar ] [ CrossRef ]
  • Xiao, K.; Chen, Y.; Xiao, L.; Sun, H.; He, Z.; Huang, G.; Chen, L.; Xv, L.; Peng, L.; Li, J.; et al. The relationship between hyperhomocysteinemia and total coronary artery occlusion: A cross-sectional study from Southwest China. Coron. Artery Dis. 2023 , 34 , 138–145. [ Google Scholar ] [ CrossRef ]
  • Tyrovola, D.; Soulaidopoulos, S.; Tsioufis, C.; Lazaros, G. The Role of Nutrition in Cardiovascular Disease: Current Concepts and Trends. Nutrients 2023 , 15 , 1064. [ Google Scholar ] [ CrossRef ]
  • Kahleova, H.; Levin, S.; Barnard, N.D. Vegetarian Dietary Patterns and Cardiovascular Disease. Prog. Cardiovasc. Dis. 2018 , 61 , 54–61. [ Google Scholar ] [ CrossRef ]
  • Arnett, D.K.; Blumenthal, R.S.; Albert, M.A.; Buroker, A.B.; Goldberger, Z.D.; Hahn, E.J.; Himmelfarb, C.D.; Khera, A.; Lloyd-Jones, D.; McEvoy, J.W.; et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019 , 140 , 596–646. [ Google Scholar ]
  • Mitrou, P.N.; Kipnis, V.; Thiébaut, A.C.; Reedy, J.; Subar, A.F.; Wirfält, E.; Flood, A.; Mouw, T.; Hollenbeck, A.R.; Leitzmann, M.F.; et al. Mediterranean dietary pattern and prediction of all-cause mortality in a US population: Results from the NIH–AARP diet and health study. Arch. Intern. Med. 2007 , 167 , 2461–2468. [ Google Scholar ] [ CrossRef ]
  • Fung, T.T.; Rexrode, K.M.; Mantzoros, C.S.; Manson, J.E.; Willett, W.C.; Hu, F.B. Mediterranean diet and incidence of and mortality from coronary heart disease and stroke in women. Circulation 2009 , 119 , 1093–1100. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Chiavaroli, L.; Viguiliouk, E.; Nishi, S.K.; Blanco Mejia, S.; Rahelić, D.; Kahleová, H.; Salas-Salvadó, J.; Kendall, C.W.; Sievenpiper, J.L. DASH Dietary Pattern and Cardiometabolic Outcomes: An Umbrella Review of Systematic Reviews and Meta-Analyses. Nutrients 2019 , 11 , 338. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Eckel, R.H.; Jakicic, J.M.; Ard, J.D.; de Jesus, J.M.; Miller, N.H.; Hubbard, V.S.; Lee, I.M.; Lichtenstein, A.H.; Loria, C.M.; Millen, B.E.; et al. 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. J. Am. Coll. Cardiol. 2014 , 63 , 2960–2984. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Wilson, J.; Lowery, R. The Ketogenic Bible ; Victory Belt Publishing Inc.: Las Vegas, NV, USA, 2017; ISBN 13:978-1-628601-04-6. [ Google Scholar ]
  • Gardner, C.D.; Vadiveloo, M.K.; Petersen, K.S.; Anderson, C.A.M.; Springfield, S.; Van Horn, L.; Khera, A.; Lamendola, C.; Mayo, S.M.; Joseph, J.J. American Heart Association Council on Lifestyle and Cardiometabolic Health. Popular Dietary Patterns: Alignment with American Heart Association 2021 Dietary Guidance: A Scientific Statement from the American Heart Association. Circulation 2023 , 147 , 1715–1730. [ Google Scholar ] [ CrossRef ]
  • Du, Z.; Qin, Y. Dyslipidemia and Cardiovascular Disease: Current Knowledge, Existing Challenges, and New Opportunities for Management Strategies. J. Clin. Med. 2023 , 12 , 363. [ Google Scholar ] [ CrossRef ]
  • Jung, E.; Kong, S.Y.; Ro, Y.S.; Ryu, H.H.; Shin, S.D. Serum Cholesterol Levels and Risk of Cardiovascular Death: A Systematic Review and a Dose-Response Meta-Analysis of Prospective Cohort Studies. Int. J. Environ. Res. Public Health 2022 , 19 , 8272. [ Google Scholar ] [ CrossRef ]
  • Dong, J.; Yang, S.; Zhuang, Q.; Sun, J.; Wei, P.; Zhao, X.; Chen, Y.; Chen, X.; Li, M.; Wei, L.; et al. The Associations of Lipid Profiles With Cardiovascular Diseases and Death in a 10-Year Prospective Cohort Study. Front. Cardiovasc. Med. 2021 , 8 , 745539. [ Google Scholar ] [ CrossRef ]
  • Yi, S.W.; Yi, J.J.; Ohrr, H. Total cholesterol and all-cause mortality by sex and age: A prospective cohort study among 12.8 million adults. Sci. Rep. 2019 , 9 , 1596. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • BHF-HEART STATS and WHO-MORTALITY (Adapted). Total Cholesterol Levels vs Mortality Data from 164 Countries, 2005. Available online: https://renegadewellness.files.wordpress.com/2011/02/cholesterol-mortality-chart.pdf (accessed on 23 July 2023).
  • Ravnskov, U.; de Lorgeril, M.; Diamond, D.M.; Hama, R.; Hamazaki, T.; Hammarskjöld, B.; Hynes, N.; Kendrick, M.; Langsjoen, P.H.; Mascitelli, L.; et al. LDL-C does not cause cardiovascular disease: A comprehensive review of the current literature. Expert Rev. Clin. Pharmacol. 2018 , 11 , 959–970. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Borén, J.; Chapman, M.J.; Krauss, R.M.; Packard, C.J.; Bentzon, J.F.; Binder, C.J.; Daemen, M.J.; Demer, L.L.; Hegele, R.A.; Nicholls, S.J.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease: Pathophysiological, genetic, and therapeutic insights: A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2020 , 41 , 2313–2330. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Peng, K.M.; Li, X.; Wang, Z.; Li, M.M.; Yang, Y. Association of low-density lipoprotein cholesterol levels with the risk of mortality and cardiovascular events: A meta-analysis of cohort studies with 1,232,694 participants. Medicine 2022 , 101 , e32003. [ Google Scholar ] [ CrossRef ]
  • Bhargava, S.; de la Puente-Secades, S.; Schurgers, L.; Jankowski, J. Lipids and lipoproteins in cardiovascular diseases: A classification. Trends Endocrinol. Metab. 2022 , 33 , 409–423. [ Google Scholar ] [ CrossRef ]
  • Kim, Y.G.; Jeong, J.H.; Han, K.D.; Roh, S.Y.; Min, K.; Lee, H.S.; Choi, Y.Y.; Shim, J.; Choi, J.I.; Kim, Y.H. Association between low-density lipoprotein cholesterol and sudden cardiac arrest in people with diabetes mellitus. Cardiovasc. Diabetol. 2023 , 22 , 36. [ Google Scholar ] [ CrossRef ]
  • Rong, S.; Li, B.; Chen, L.; Sun, Y.; Du, Y.; Liu, B.; Robinson, J.G.; Bao, W. Association of Low-Density Lipoprotein Cholesterol Levels with More than 20-Year Risk of Cardiovascular and All-Cause Mortality in the General Population. J. Am. Heart Assoc. 2022 , 11 , e023690. [ Google Scholar ] [ CrossRef ]
  • Feingold, K.R. Utility of Advanced Lipoprotein Testing in Clinical Practice ; Feingold, K.R., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., Hofland, J., Eds.; Endotext [Internet]; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [ Google Scholar ]
  • Nicholls, S.J.; Nelson, A.J. HDL and cardiovascular disease. Pathology 2019 , 51 , 142–147. [ Google Scholar ] [ CrossRef ]
  • Trimarco, V.; Izzo, R.; Morisco, C.; Mone, P.; Virginia Manzi, M.; Falco, A.; Pacella, D.; Gallo, P.; Lembo, M.; Santulli, G.; et al. High HDL (High-Density Lipoprotein) Cholesterol Increases Cardiovascular Risk in Hypertensive Patients. Hypertension 2022 , 79 , 2355–2363. [ Google Scholar ] [ CrossRef ]
  • Cho, Y.K.; Jung, C.H. HDL-C and Cardiovascular Risk: You Don’t Need to Worry about Extremely High HDL-C Levels. J. Lipid Atheroscler. 2021 , 10 , 57–61. [ Google Scholar ] [ CrossRef ]
  • Farnier, M.; Zeller, M.; Masson, D.; Cottin, Y. Triglycerides and risk of atherosclerotic cardiovascular disease: An update. Arch Cardiovasc. Dis. 2021 , 114 , 132–139. [ Google Scholar ] [ CrossRef ]
  • Xia, T.L.; Li, Y.M.; Huang, F.Y.; Chai, H.; Huang, B.T.; Li, Q.; Zhao, Z.-G.; Liao, Y.-B.; Zuo, Z.L.; Peng, Y.; et al. The triglyceride paradox in the mortality of coronary artery disease. Lipids Health Dis. 2019 , 18 , 21. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Jain, M.; Jain, A.; Yerragondu, N.; Brown, R.D.; Rabinstein, A.; Jahromi, B.S.; Vaidyanathan, L.; Blyth, B.; Stead, L.G. The triglyceride paradox in stroke survivors: A prospective study. Neurosci. J. 2013 , 2013 , 870608. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kosmas, C.E.; Rodriguez Polanco, S.; Bousvarou, M.D.; Papakonstantinou, E.J.; Peña Genao, E.; Guzman, E.; Kostara, C.E. The Triglyceride/High-Density Lipoprotein Cholesterol (TG/HDL-C) Ratio as a Risk Marker for Metabolic Syndrome and Cardiovascular Disease. Diagnostics 2023 , 13 , 929. [ Google Scholar ] [ CrossRef ]
  • Wu, Z.; Wang, J.; Zhang, H.; Pan, H.; Li, Z.; Liu, Y.; Miao, X.; Han, Z.; Kang, X.; Li, X.; et al. Longitudinal association of remnant cholesterol with joint arteriosclerosis and atherosclerosis progression beyond LDL cholesterol. BMC Med. 2023 , 21 , 42. [ Google Scholar ] [ CrossRef ]
  • Varbo, A.; Freiberg, J.J.; Nordestgaard, B.G. Extreme Nonfasting Remnant Cholesterol vs Extreme LDL Cholesterol as Contributors to Cardiovascular Disease and All-Cause Mortality in 90,000 Individuals from the General Population. Clin. Chem. 2015 , 61 , 533–543. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Tamang, H.K.; Timilsina, U.; Singh, K.P.; Shrestha, S.; Raman, R.K.; Panta, P.; Karna, P.; Khadka, L.; Dahal, C. Apo B/Apo A-I Ratio is Statistically A Better Predictor of Cardiovascular Disease (CVD) than Conventional Lipid Profile: A Study from Kathmandu Valley, Nepal. J. Clin. Diagn. Res. 2014 , 8 , 34–36. [ Google Scholar ] [ CrossRef ]
  • Li, S.; Lin, G.; Chen, J.; Chen, Z.; Xu, F.; Zhu, F.; Zhang, J.; Yuan, S. The effect of periodic ketogenic diet on newly diagnosed overweight or obese patients with type 2 diabetes. BMC Endocr. Disord. 2022 , 22 , 34. [ Google Scholar ] [ CrossRef ]
  • 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 ]
  • Schiavo, L.; Pierro, R.; Asteria, C.; Calabrese, P.; Di Biasio, A.; Coluzzi, I.; Severino, L.; Giovanelli, A.; Pilone, V.; Silecchia, G. Low-Calorie Ketogenic Diet with Continuous Positive Airway Pressure to Alleviate Severe Obstructive Sleep Apnea Syndrome in Patients with Obesity Scheduled for Bariatric/Metabolic Surgery: A Pilot, Prospective, Randomized Multicenter Comparative Study. Obes. Surg. 2022 , 32 , 634–642. [ Google Scholar ] [ CrossRef ]
  • Vidić, V.; Ilić, V.; Toskić, L.; Janković, N.; Ugarković, D. Effects of calorie restricted low carbohydrate high fat ketogenic vs. non-ketogenic diet on strength, body-composition, hormonal and lipid profile in trained middle-aged men. Clin. Nutr. 2021 , 40 , 1495–1502. [ Google Scholar ] [ CrossRef ]
  • Burén, J.; Ericsson, M.; Damasceno, N.R.T.; Sjödin, A. A Ketogenic Low-Carbohydrate High-Fat Diet Increases LDL Cholesterol in Healthy, Young, Normal-Weight Women: A Randomized Controlled Feeding Trial. Nutrients 2021 , 13 , 814. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Ravnskov, U. Is High Cholesterol Deleterious? An Alternative Point of View. Comment on Burén et al. A Ketogenic Low-Carbohydrate High-Fat Diet Increases LDL Cholesterol in Healthy, Young, Normal-Weight Women: A Randomized Controlled Feeding Trial. Nutrients 2021 , 13 , 814, Nutrients 2021 , 13 , 2119. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Basciani, S.; Camajani, E.; Contini, S.; Persichetti, A.; Risi, R.; Bertoldi, L.; Strigari, L.; Prossomariti, G.; Watanabe, M.; Mariani, S.; et al. Very-Low-Calorie Ketogenic Diets with Whey, Vegetable, or Animal Protein in Patients with Obesity: A Randomized Pilot Study. J. Clin. Endocrinol. Metab. 2020 , 105 , dgaa336. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Michalczyk, M.M.; Klonek, G.; Maszczyk, A.; Zajac, A. The Effects of a Low Calorie Ketogenic Diet on Glycaemic Control Variables in Hyperinsulinemic Overweight/Obese Females. Nutrients 2020 , 12 , 1854. [ Google Scholar ] [ CrossRef ]
  • Saslow, L.R.; Daubenmier, J.J.; Moskowitz, J.T.; Kim, S.; Murphy, E.J.; Phinney, S.D.; Ploutz-Snyder, R.; Goldman, V.; Cox, R.M.; Mason, A.E.; et al. Twelve-month outcomes of a randomized trial of a moderate-carbohydrate versus very low-carbohydrate diet in overweight adults with type 2 diabetes mellitus or prediabetes. Nutr. Diabetes 2017 , 7 , 304. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Paoli, A.; Moro, T.; Bosco, G.; Bianco, A.; Grimaldi, K.A.; Camporesi, E.; Mangar, D. Effects of n-3 polyunsaturated fatty acids (ω-3) supplementation on some cardiovascular risk factors with a ketogenic Mediterranean diet. Mar. Drugs. 2015 , 13 , 996–1009. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Partsalaki, I.; Karvela, A.; Spiliotis, B.E. Metabolic impact of a ketogenic diet compared to a hypocaloric diet in obese children and adolescents. J. Pediatr. Endocrinol. Metab. 2012 , 25 , 697–704. [ Google Scholar ] [ CrossRef ]
  • Amanollahi, A.; Khazdouz, M.; Malekahmadi, M.; Klement, R.J.; Lee, D.; Khodabakhshi, A. Effect of Ketogenic Diets on Cardio-Metabolic Outcomes in Cancer Patients: A Systematic Review and Meta-Analysis of Controlled Clinical Trials. Nutr. Cancer 2023 , 75 , 95–111. [ Google Scholar ] [ CrossRef ]
  • Li, M.; Yuan, J. Effects of very low-carbohydrate ketogenic diet on lipid metabolism in patients with type II diabetes mellitus: A meta-analysis. Nutr. Hosp. 2022 , 39 , 916–923, English. [ Google Scholar ] [ CrossRef ]
  • Rafiullah, M.; Musambil, M.; David, S.K. Effect of a very low-carbohydrate ketogenic diet vs recommended diets in patients with type 2 diabetes: A meta-analysis. Nutr. Rev. 2022 , 80 , 488–502. [ Google Scholar ] [ CrossRef ]
  • Yuan, X.; Wang, J.; Yang, S.; Gao, M.; Cao, L.; Li, X.; Hong, D.; Tian, S.; Sun, C. Effect of the ketogenic diet on glycemic control, insulin resistance, and lipid metabolism in patients with T2DM: A systematic review and meta-analysis. Nutr. Diabetes 2020 , 10 , 38. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • López-Espinoza, M.Á.; Chacón-Moscoso, S.; Sanduvete-Chaves, S.; Ortega-Maureira, M.J.; Barrientos-Bravo, T. Effect of a Ketogenic Diet on the Nutritional Parameters of Obese Patients: A Systematic Review and Meta-Analysis. Nutrients 2021 , 13 , 2946. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Muscogiuri, G.; El Ghoch, M.; Colao, A.; Hassapidou, M.; Yumuk, V.; Busetto, L. Obesity Management Task Force (OMTF) of the European Association for the Study of Obesity (EASO). European Guidelines for Obesity Management in Adults with a Very Low-Calorie Ketogenic Diet: A Systematic Review and Meta-Analysis. Obes. Facts. 2021 , 14 , 222–245. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Castellana, M.; Conte, E.; Cignarelli, A.; Perrini, S.; Giustina, A.; Giovanella, L.; Giorgino, F.; Trimboli, P. Efficacy and safety of very low calorie ketogenic diet (VLCKD) in patients with overweight and obesity: A systematic review and meta-analysis. Rev. Endocr. Metab. Disord. 2020 , 21 , 5–16. [ Google Scholar ] [ CrossRef ]
  • Kern, F., Jr. Normal plasma cholesterol in an 88-year-old man who eats 25 eggs a day. Mechanisms of adaptation. N. Engl. J. Med. 1991 , 324 , 896–899. [ Google Scholar ] [ CrossRef ]
  • Fernandez, M.L.; Murillo, A.G. Is There a Correlation between Dietary and Blood Cholesterol? Evidence from Epidemiological Data and Clinical Interventions. Nutrients 2022 , 14 , 2168. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Ravnskov, U.; Diamond, D.M.; Hama, R.; Hamazaki, T.; Hammarskjöld, B.; Hynes, N.; Kendrick, M.; Langsjoen, P.H.; Malhotra, A.; Mascitelli, L.; et al. Lack of an association or an inverse association between low-density-lipoprotein cholesterol and mortality in the elderly: A systematic review. BMJ Open 2016 , 6 , e010401. [ Google Scholar ] [ CrossRef ]
  • Ravnskov, U.; de Lorgeril, M.; Diamond, D.M.; Hama, R.; Hamazaki, T.; Hammarskjöld, B.; Harcombe, Z.; Kendrick, M.; Langsjoen, P.H.; McCully, K.S.; et al. The LDL paradox: Higher LDL-cholesterol is associated with greater longevity. Ann. Epidemiol. Public Health 2020 , 3 , 1040–1047. [ Google Scholar ]
  • Sorriento, D.; Iaccarino, G. Inflammation and Cardiovascular Diseases: The Most Recent Findings. Int. J. Mol. Sci. 2019 , 20 , 3879. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Fiordelisi, A.; Iaccarino, G.; Morisco, C.; Coscioni, E.; Sorriento, D. NFkappaB is a Key Player in the Crosstalk between Inflammation Cardiovascular Diseases. Int. J. Mol. Sci. 2019 , 20 , 1599. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Liccardo, D.; Cannavo, A.; Spagnuolo, G.; Ferrara, N.; Cittadini, A.; Rengo, C.; Rengo, G. Periodontal Disease: A Risk Factor for Diabetes and Cardiovascular Disease. Int. J. Mol. Sci. 2019 , 20 , 1414. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Mercurio, V.; Lobasso, A.; Barbieri, L.; Parrella, P.; Ciervo, D.; Liccardo, B.; Bonaduce, D.; Tocchetti, C.G.; De Paulis, A.; Rossi, F.W. Inflammatory, Serological and Vascular Determinants of Cardiovascular Disease in Systemic Lupus Erythematosus Patients. Int. J. Mol. Sci. 2019 , 20 , 2154. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Varricchi, G.; Loffredo, S.; Borriello, F.; Pecoraro, A.; Rivellese, F.; Genovese, A.; Spadaro, G.; Marone, G. Superantigenic Activation of Human Cardiac Mast Cells. Int. J. Mol. Sci. 2019 , 20 , 1828. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Brigant, B.; Metzinger-Le Meuth, V.; Rochette, J.; Metzinger, L. TRIMming down to TRIM37: Relevance to Inflammation, Cardiovascular Disorders, and Cancer in MULIBREY Nanism. Int. J. Mol. Sci. 2018 , 20 , 67. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017 , 377 , 1119–1131. [ Google Scholar ] [ CrossRef ]
  • Henein, M.Y.; Vancheri, S.; Longo, G.; Vancheri, F. The Role of Inflammation in Cardiovascular Disease. Int. J. Mol. Sci. 2022 , 23 , 12906. [ Google Scholar ] [ CrossRef ]
  • Sklerov, M.; Dayan, E.; Browner, N. Functional neuroimaging of the central autonomic network: Recent developments and clinical implications. Clin. Auton. Res. 2019 , 29 , 555–566. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kraynak, T.E.; Marsland, A.L.; Gianaros, P.J. Neural Mechanisms Linking Emotion with Cardiovascular Disease. Curr. Cardiol. Rep. 2018 , 20 , 128. [ Google Scholar ] [ CrossRef ]
  • Muscatell, K.A.; Dedovic, K.; Slavich, G.M.; Jarcho, M.R.; Breen, E.C.; Bower, J.E.; Irwin, M.R.; Eisenberger, N.I. Greater amygdala activity and dorsomedial prefrontal–amygdala coupling are associated with enhanced inflammatory responses to stress. Brain Behav. Immun. 2015 , 43 , 46–53. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Tracey, K.J. The inflammatory reflex. Nature 2002 , 420 , 853–859. [ Google Scholar ] [ CrossRef ]
  • Shah, S.M.; Meadows, J.L.; Burg, M.M.; Pfau, S.; Soufer, R. Effects of Psychological Stress on Vascular Physiology: Beyond the Current Imaging Signal. Curr. Cardiol. Rep. 2020 , 22 , 156. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Tawakol, A.; Ishai, A.; Takx, R.A.P.; Figueroa, A.L.; Ali, A.; Kaiser, Y.; Truong, Q.A.; Solomon, C.J.E.; Calcagno, C.; Mani, V.; et al. Relation between resting amygdalar activity and cardiovascular events: A longitudinal and cohort study. Lancet 2017 , 389 , 834–845. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Pondel, N.; Liśkiewicz, D.; Liśkiewicz, A. Dieta ketogeniczna–mechanizm działania i perspektywy zastosowania w terapii: Dane z badań klinicznych. Postępy Biochem. 2020 , 66 , 270–286. [ Google Scholar ] [ CrossRef ]
  • Dyńka, D.; Kowalcze, K.; Paziewska, A. The Role of Ketogenic Diet in the Treatment of Neurological Diseases. Nutrients 2022 , 14 , 5003. [ Google Scholar ] [ CrossRef ]
  • Hwang, C.Y.; Choe, W.; Yoon, K.-S.; Ha, J.; Kim, S.S.; Yeo, E.-J.; Kang, I. Molecular Mechanisms for Ketone Body Metabolism, Signaling Functions, and Therapeutic Potential in Cancer. Nutrients 2022 , 14 , 4932. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Dyńka, D.; Kowalcze, K.; Ambrozkiewicz, F.; Paziewska, A. Effect of the Ketogenic Diet on the Prophylaxis and Treatment of Diabetes Mellitus: A Review of the Meta-Analyses and Clinical Trials. Nutrients 2023 , 15 , 500. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kong, C.; Yan, X.; Liu, Y.; Huang, L.; Zhu, Y.; He, J.; Gao, R.; Kalady, M.F.; Goel, A.; Qin, H.; et al. Ketogenic diet alleviates colitis by reduction of colonic group 3 innate lymphoid cells through altering gut microbiome. Signal Transduct. Target. Ther. 2021 , 6 , 154. [ Google Scholar ] [ CrossRef ]
  • Field, R.; Pourkazemi, F.; Rooney, K. Effects of a Low-Carbohydrate Ketogenic Diet on Reported Pain, Blood Biomarkers and Quality of Life in Patients with Chronic Pain: A Pilot Randomized Clinical Trial. Pain Med. 2022 , 23 , 326–338. [ Google Scholar ] [ CrossRef ]
  • Alkhorayef, N.; Almutery, F.T.; Rasheed, Z.; Althwab, S.A.; Aljohani, A.S.M.; Alhawday, Y.A.N.; Salem, T.; Alharbi, A.M.; Wahaq, A.A.A.B.; Alharbi, F.S.; et al. Regulatory effects of ketogenic diet on the inflammatory response in obese Saudi women. J. Taibah Univ. Med. Sci. 2023 , 18 , 1101–1107. [ Google Scholar ] [ CrossRef ]
  • Mohammadifard, N.; Haghighatdoost, F.; Rahimlou, M.; Rodrigues, A.P.S.; Gaskarei, M.K.; Okhovat, P.; de Oliveira, C.; Silveira, E.A.; Sarrafzadegan, N. The Effect of Ketogenic Diet on Shared Risk Factors of Cardiovascular Disease and Cancer. Nutrients 2022 , 14 , 3499. [ Google Scholar ] [ CrossRef ]
  • Mezzaroma, E.; Toldo, S.; Farkas, D.; Seropian, I.M.; Van Tassell, B.W.; Salloum, F.N.; Kannan, H.R.; Menna, A.C.; Voelkel, N.F.; Abbate, A. The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse. Proc. Natl. Acad. Sci. USA 2011 , 108 , 19725–19730. [ Google Scholar ] [ CrossRef ]
  • Tong, Y.; Wang, Z.; Cai, L.; Lin, L.; Liu, J.; Cheng, J. NLRP3 Inflammasome and Its Central Role in the Cardiovascular Diseases. Oxid. Med. Cell. Longev. 2020 , 2020 , 4293206. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Zheng, Y.; Xu, L.; Dong, N.; Li, F. NLRP3 inflammasome: The rising star in cardiovascular diseases. Front. Cardiovasc. Med. 2022 , 9 , 927061. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Poff, A.; Kesl, S.; Koutnik, A.; Ward, N.; Ari, C.; Deblasi, J.; D’Agostino, D. Characterizing the metabolic effects of exogenous ketone supplementation—An alternative or adjuvant to the ketogenic diet. FASEB J. 2017 , 31 , 970.7. [ Google Scholar ] [ CrossRef ]
  • Youm, Y.H.; Nguyen, K.Y.; Grant, R.W.; Goldberg, E.L.; Bodogai, M.; Kim, D.; D’Agostino, D.; Planavsky, N.; Lupfer, C.; Kanneganti, T.D.; et al. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat. Med. 2015 , 21 , 263–269. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Graff, E.C.; Fang, H.; Wanders, D.; Judd, R.L. Anti-inflammatory effects of the hydroxycarboxylic acid receptor 2. Metabolism 2016 , 65 , 102–113. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Offermanns, S. Free fatty acid (FFA) and hydroxy carboxylic acid (HCA) receptors. Annu. Rev. Pharm. Toxicol. 2014 , 54 , 407–434. [ Google Scholar ] [ CrossRef ]
  • Qi, J.; Gan, L.; Fang, J.; Zhang, J.; Yu, X.; Guo, H.; Cai, D.; Cui, H.; Gou, L.; Deng, J.; et al. Beta-Hydroxybutyrate: A Dual Function Molecular and Immunological Barrier Function Regulator. Front. Immunol. 2022 , 13 , 805881. [ Google Scholar ] [ CrossRef ]
  • Shimazu, T.; Hirschey, M.D.; Newman, J.; He, W.; Shirakawa, K.; Le Moan, N.; Grueter, C.A.; Lim, H.; Saunders, L.R.; Stevens, R.D.; et al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 2013 , 339 , 211–214. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Bae, H.R.; Kim, D.H.; Park, M.H.; Lee, B.; Kim, M.J.; Lee, E.K.; Chung, K.W.; Kim, S.M.; Im, D.S.; Chung, H.Y. β-Hydroxybutyrate suppresses inflammasome formation by ameliorating endoplasmic reticulum stress via AMPK activation. Oncotarget 2016 , 7 , 66444–66454. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Nasser, S.; Vialichka, V.; Biesiekierska, M.; Balcerczyk, A.; Pirola, L. Effects of ketogenic diet and ketone bodies on the cardiovascular system: Concentration matters. World J. Diabetes 2020 , 11 , 584–595. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Yurista, S.R.; Chong, C.R.; Badimon, J.J.; Kelly, D.P.; de Boer, R.A.; Westenbrink, B.D. Therapeutic Potential of Ketone Bodies for Patients with Cardiovascular Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2021 , 77 , 1660–1669. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Westman, E.C.; Feinman, R.D.; Mavropoulos, J.C.; Vernon, M.C.; Volek, J.S.; Wortman, J.A.; Yancy, W.S.; Phinney, S.D. Lowcarbohydrate nutrition and metabolism. Am. J. Clin. Nutr. 2007 , 86 , 276–284. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Ma, X.; Nan, F.; Liang, H.; Shu, P.; Fan, X.; Song, X.; Hou, Y.; Zhang, D. Excessive intake of sugar: An accomplice of inflammation. Front. Immunol. 2022 , 13 , 988481. [ Google Scholar ] [ CrossRef ]
  • Della Corte, K.W.; Perrar, I.; Penczynski, K.J.; Schwingshackl, L.; Herder, C.; Buyken, A.E. Effect of Dietary Sugar Intake on Biomarkers of Subclinical Inflammation: A Systematic Review and Meta-Analysis of Intervention Studies. Nutrients 2018 , 10 , 606. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • O’Connor, L.; Imamura, F.; Brage, S.; Griffin, S.J.; Wareham, N.J.; Forouhi, N.G. Intakes and sources of dietary sugars and their association with metabolic and inflammatory markers. Clin. Nutr. 2018 , 37 , 1313–1322. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • McGandy, R.B.; Hegsted, D.M.; Stare, F.J. Dietary fats, carbohydratesand atherosclerotic vascular disease. N. Engl. J. Med. 1967 , 277 , 186–192. [ Google Scholar ] [ CrossRef ]
  • Carbone, S.; Billingsley, H.E.; Lavie, C.J. The Effects of Dietary Sugars on Cardiovascular Disease and Cardiovascular Disease-Related Mortality: Finding the Sweet Spot. Mayo Clin. Proc. 2019 , 94 , 2375–2377. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Howard, B.V.; Van Horn, L.; Hsia, J.; Manson, J.E.; Stefanick, M.L.; Wassertheil-Smoller, S.; Kuller, L.H.; LaCroix, A.Z.; Langer, L.D.; Lasser, N.L.; et al. Low-fat dietary patternand risk of cardiovascular disease: The Women’s Health Initiative Randomized Controlled Dietary Modification Trial. JAMA 2006 , 295 , 655–666. [ Google Scholar ] [ CrossRef ]
  • Billingsley, H.E.; Carbone, S.; Lavie, C.J. Dietary fats and chronicnoncommunicable diseases. Nutrients 2018 , 10 , 1385. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Huang, C.; Huang, J.; Tian, Y.; Yang, X.; Gu, D. Sugar sweetened beverages consumption and risk of coronary heart disease: A metaanalysis of prospective studies. Atherosclerosis 2014 , 234 , 11–16. [ Google Scholar ] [ CrossRef ]
  • Li, Y.; Hruby, A.; Bernstein, A.M.; Ley, S.H.; Wang, D.D.; Chiuve, S.E.; Sampson, L.; Rexrode, K.M.; Rimm, E.B.; Willett, W.C.; et al. Saturated fats comparedwith unsaturated fats and sources of carbohydrates in relationto risk of coronary heart disease: A prospective cohort study. J. Am. Coll. Cardiol. 2015 , 66 , 1538–1548. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Yang, Q.; Zhang, Z.; Gregg, E.W.; Flanders, W.D.; Merritt, R.; Hu, F.B. Added sugar intake and cardiovascular diseases mortalityamong US adults. JAMA Intern. Med. 2014 , 174 , 516–524. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Available online: https://www.health.harvard.edu/heart-health/the-sweet-danger-of-sugar (accessed on 23 July 2023).
  • Yang, B.; Glenn, A.J.; Liu, Q.; Madsen, T.; Allison, M.A.; Shikany, J.M.; Manson, J.E.; Chan, K.H.K.; Wu, W.C.; Li, J.; et al. Added Sugar, Sugar-Sweetened Beverages, and Artificially Sweetened Beverages and Risk of Cardiovascular Disease: Findings from the Women’s Health Initiative and a Network Meta-Analysis of Prospective Studies. Nutrients 2022 , 14 , 4226. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kelly, R.K.; Tong, T.Y.N.; Watling, C.Z.; Reynolds, A.; Piernas, C.; Schmidt, J.A.; Papier, K.; Carter, J.L.; Key, T.J.; Perez-Cornago, A. Associations between types and sources of dietary carbohydrates and cardiovascular disease risk: A prospective cohort study of UK Biobank participants. BMC Med. 2023 , 21 , 34. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Hosseini, B.; Berthon, B.S.; Saedisomeolia, A.; Starkey, M.R.; Collison, A.; Wark, P.A.B.; Wood, L.G. Effects of fruit and vegetable consumption on inflammatory biomarkers and immune cell populations: A systematic literature review and meta-analysis. Am. J. Clin. Nutr. 2018 , 108 , 136–155. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Cheng, H.; Zhou, J.; Sun, Y.; Zhan, Q.; Zhang, D. High fructose diet: A risk factor for immune system dysregulation. Hum. Immunol. 2022 , 83 , 538–546. [ Google Scholar ] [ CrossRef ]
  • Lubawy, M.; Formanowicz, D. High-Fructose Diet–Induced Hyperuricemia Accompanying Metabolic Syndrome–Mechanisms and Dietary Therapy Proposals. Int. J. Environ. Res. Public Health 2023 , 20 , 3596. [ Google Scholar ] [ CrossRef ]
  • Choi, H.K.; Willett, W.; Curhan, G. Fructose-Rich Beverages and the Risk of Gout in Women. JAMA J. Am. Med. Assoc. 2010 , 304 , 2270–2278. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kanbay, M.; Guler, B.; Ertuglu, L.A.; Dagel, T.; Afsar, B.; Incir, S.; Baygul, A.; Covic, A.; Andres-Hernando, A.; Sánchez-Lozada, L.G.; et al. The Speed of Ingestion of a Sugary Beverage Has an Effect on the Acute Metabolic Response to Fructose. Nutrients 2021 , 13 , 1916. [ Google Scholar ] [ CrossRef ]
  • Public Health England. Why 5%? An Explanation of SACN’s Recommendations about Sugars and Health. PHE Publications Gateway Number 2015193. 2015. Available online: https://www.gov.uk/government/publications/sacns-sugars-and-health-recommendations-why-5 (accessed on 23 July 2023).
  • Rawal, G.; Yadav, S.; Kumar, R.; Singh, A. Glycosylated hemoglobin (HbA1C): A brief overview for clinicians. IP Indian J. Immunol. Respir. Med. 2016 , 1 , 33–36. [ Google Scholar ]
  • Goto, A.; Noda, M.; Matsushita, Y.; Goto, M.; Kato, M.; Isogawa, A.; Takahashi, Y.; Kurotani, K.; Oba, S.; Nanri, A.; et al. JPHC Study Group. Hemoglobin a1c levels and the risk of cardiovascular disease in people without known diabetes: A population-based cohort study in Japan. Medicine 2015 , 94 , e785. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Sinning, C.; Makarova, N.; Völzke, H.; Schnabel, R.B.; Ojeda, F.; Dörr, M.; Felix, S.B.; Koenig, W.; Peters, A.; Rathmann, W.; et al. Association of glycated hemoglobin A1c levels with cardiovascular outcomes in the general population: Results from the BiomarCaRE (Biomarker for Cardiovascular Risk Assessment in Europe) consortium. Cardiovasc. Diabetol. 2021 , 20 , 223. [ Google Scholar ] [ CrossRef ]
  • Prasad, K. Does HbA1cc Play a Role in the Development of Cardiovascular Diseases? Curr. Pharm. Des. 2018 , 24 , 2876–2882. [ Google Scholar ] [ CrossRef ]
  • Zaki, H.A.; Iftikhar, H.; Bashir, K.; Gad, H.; Fahmy, A.S.; Elmoheen, A. A Comparative Study Evaluating the Effectiveness Between Ketogenic and Low-Carbohydrate Diets on Glycemic and Weight Control in Patients with Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Cureus 2022 , 14 , e25528. [ Google Scholar ] [ PubMed ]
  • Zhou, C.; Wang, M.; Liang, J.; He, G.; Chen, N. Ketogenic Diet Benefits to Weight Loss, Glycemic Control, and Lipid Profiles in Overweight Patients with Type 2 Diabetes Mellitus: A Meta-Analysis of Randomized Controlled Trails. Int. J. Environ. Res. Public Health 2022 , 19 , 10429. [ Google Scholar ] [ CrossRef ]
  • Choi, Y.J.; Jeon, S.M.; Shin, S. Impact of a Ketogenic Diet on Metabolic Parameters in Patients with Obesity or Overweight and with or without Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials. Nutrients 2020 , 12 , 2005. [ Google Scholar ] [ CrossRef ]
  • Parry-Strong, A.; Wright-McNaughton, M.; Weatherall, M.; Hall, R.M.; Coppell, K.J.; Barthow, C.; Krebs, J.D. Very low carbohydrate (ketogenic) diets in type 2 diabetes: A systematic review and meta-analysis of randomized controlled trials. Diabetes Obes. Metab. 2022 , 24 , 2431–2442. [ Google Scholar ] [ CrossRef ]
  • Jayedi, A.; Zeraattalab-Motlagh, S.; Jabbarzadeh, B.; Hosseini, Y.; Jibril, A.T.; Shahinfar, H.; Mirrafiei, A.; Hosseini, F.; Bidar, S.S. Dose-dependent effect of carbohydrate restriction for type 2 diabetes management: A systematic review and dose-response meta-analysis of randomized controlled trials. Am. J. Clin. Nutr. 2022 , 116 , 40–56. [ Google Scholar ] [ CrossRef ]
  • Waldman, H.S.; Smith, J.W.; Lamberth, J.; Fountain, B.J.; Bloomer, R.J.; Butawan, M.B.; McAllister, M.J. A 28-Day Carbohydrate-Restricted Diet Improves Markers of Cardiovascular Disease in Professional Firefighters. J. Strength Cond. Res. 2020 , 34 , 2785–2792. [ Google Scholar ] [ CrossRef ]
  • Karimi, E.; Yarizadeh, H.; Setayesh, L.; Sajjadi, S.F.; Ghodoosi, N.; Khorraminezhad, L.; Mirzaei, K. High carbohydrate intakes may predict more inflammatory status than high fat intakes in pre-menopause women with overweight or obesity: A cross-sectional study. BMC Res. Notes 2021 , 14 , 279. [ Google Scholar ] [ CrossRef ]
  • Tavakoli, A.; Mirzababaei, A.; Sajadi, F.; Mirzaei, K. Circulating inflammatory markers may mediate the relationship between low carbohydrate diet and circadian rhythm in overweight and obese women. BMC Women’s Health 2021 , 21 , 87. [ Google Scholar ] [ CrossRef ]
  • Forsythe, C.E.; Phinney, S.D.; Fernandez, M.L.; Quann, E.E.; Wood, R.J.; Bibus, D.M.; Kraemer, W.J.; Feinman, R.D.; Volek, J.S. Comparison of low fat and low carbohydrate diets on circulating fatty acid composition and markers of inflammation. Lipids 2008 , 43 , 65–77. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Wood, R.J.; Volek, J.S.; Davis, S.R.; Dell’Ova, C.; Fernandez, M.L. Effects of a carbohydrate-restricted diet on emerging plasma markers for cardiovascular disease. Nutr. Metab. 2006 , 3 , 19. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Dong, T.; Guo, M.; Zhang, P.; Sun, G.; Chen, B. The effects of low-carbohydrate diets on cardiovascular risk factors: A meta-analysis. PLoS ONE 2020 , 15 , e0225348. [ Google Scholar ] [ CrossRef ]
  • Simonetto, M.; Infante, M.; Sacco, R.L.; Rundek, T.; Della-Morte, D. A Novel Anti-Inflammatory Role of Omega-3 PUFAs in Prevention and Treatment of Atherosclerosis and Vascular Cognitive Impairment and Dementia. Nutrients 2019 , 11 , 2279. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Liu, Y.-X.; Yu, J.-H.; Sun, J.-H.; Ma, W.-Q.; Wang, J.-J.; Sun, G.-J. Effects of Omega-3 Fatty Acids Supplementation on Serum Lipid Profile and Blood Pressure in Patients with Metabolic Syndrome: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Foods 2023 , 12 , 725. [ Google Scholar ] [ CrossRef ]
  • Rodriguez, D.; Lavie, C.J.; Elagizi, A.; Milani, R.V. Update on Omega-3 Polyunsaturated Fatty Acids on Cardiovascular Health. Nutrients 2022 , 14 , 5146. [ Google Scholar ] [ CrossRef ]
  • Fatahi, S.; Sohouli, M.H.; da Silva Magalhães, E.I.; da Cruz Silveira, V.N.; Zanghelini, F.; Rahmani, P.; Kord-Varkaneh, H.; Sharifi-Zahabi, E.; Shidfar, F. Comparing the effects of docosahexaenoic and eicosapentaenoic acids on cardiovascular risk factors: Pairwise and network meta-analyses of randomized controlled trials. Nutr. Metab. Cardiovasc. Dis. 2023 , 33 , 11–21. [ Google Scholar ] [ CrossRef ]
  • Yang, B.; Tseng, P.T.; Hu, X.; Zeng, B.Y.; Chang, J.P.; Liu, Y.; Chu, W.J.; Zhang, S.S.; Zhou, Z.L.; Chu, C.S.; et al. Comparative efficacy of omega-3 polyunsaturated fatty acids on major cardiovascular events: A network meta-analysis of randomized controlled trials. Prog. Lipid Res. 2022 , 88 , 101196, Erratum in Prog. Lipid Res. 2022 , 101206. [ Google Scholar ] [ CrossRef ]
  • Yokoyama, Y.; Kuno, T.; Morita, S.X.; Slipczuk, L.; Takagi, H.; Briasoulis, A.; Latib, A.; Bangalore, S.; Heffron, S.P. Eicosapentaenoic Acid for Cardiovascular Events Reduction- Systematic Review and Network Meta-Analysis of Randomized Controlled Trials. J. Cardiol. 2022 , 80 , 416–422. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Jiang, H.; Wang, L.; Wang, D.; Yan, N.; Li, C.; Wu, M.; Wang, F.; Mi, B.; Chen, F.; Jia, W.; et al. Omega-3 polyunsaturated fatty acid biomarkers and risk of type 2 diabetes, cardiovascular disease, cancer, and mortality. Clin. Nutr. 2022 , 41 , 1798–1807. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • de Luis, D.; Domingo, J.C.; Izaola, O.; Casanueva, F.F.; Bellido, D.; Sajoux, I. Effect of DHA supplementation in a very low-calorie ketogenic diet in the treatment of obesity: A randomized clinical trial. Endocrine 2016 , 54 , 111–122. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Rondanelli, M.; Perna, S.; Ilyas, Z.; Peroni, G.; Bazire, P.; Sajuox, I.; Maugeri, R.; Nichetti, M.; Gasparri, C. Effect of very low-calorie ketogenic diet in combination with omega-3 on inflammation, satiety hormones, body composition, and metabolic markers. A pilot study in class I obese subjects. Endocrine 2022 , 75 , 129–136. [ Google Scholar ] [ CrossRef ]
  • Liu, S.H.; Chen, Y.X.; Tzeng, H.P.; Chiang, M.T. Fish Oil Enriched n-3 Polyunsaturated Fatty Acids Improve Ketogenic Low-Carbohydrate/High-Fat Diet-Caused Dyslipidemia, Excessive Fat Accumulation, and Weight Control in Rats. Nutrients 2022 , 14 , 1796. [ Google Scholar ] [ CrossRef ]
  • Stoll, S.; Leimena, C.; Qiu, H. Mitochondria and Heart Disease ; InTech: London, UK, 2018. [ Google Scholar ] [ CrossRef ]
  • Brown, D.A.; Perry, J.B.; Allen, M.E.; Sabbah, H.N.; Stauffer, B.L.; Shaikh, S.R.; Cleland, J.G.F.; Colucci, W.S.; Butler, J.; Voors, A.A.; et al. Expert consensus document: Mitochondrial function as a therapeutictarget in heart failure. Nat. Rev. Cardiol. 2017 , 14 , 238–250. Available online: https://www.researchgate.net/publication/327299198_Mitochondria_and_Heart_Disease (accessed on 24 April 2023).
  • Murashige, D.; Jang, C.; Neinast, M.; Edwards, J.J.; Cowan, A.; Hyman, M.C.; Rabinowitz, J.D.; Frankel, D.S.; Arany, Z. Comprehensive quantification of fuel use by the failing and nonfailing human heart. Science 2020 , 370 , 364–368. [ Google Scholar ] [ CrossRef ]
  • Abdul Kadir, A.; Clarke, K.; Evans, R.D. Cardiac ketone body metabolism. Biochim. Biophys. Acta Mol. Basis Dis. 2020 , 1866 , 165739. [ Google Scholar ] [ CrossRef ]
  • Sack, M.N.; Rader, T.A.; Park, S.; Bastin, J.; McCune, S.A.; Kelly, D.P. Fatty acid oxidation enzyme gene expression is downregulated in the failing heart. Circulation 1996 , 94 , 2837–2842. [ Google Scholar ]
  • Carley, A.N.; Taegtmeyer, H.; Lewandowski, E.D. Matrix revisited: Mechanisms linking energy substrate metabolism to the function of the heart. Circ. Res. 2014 , 114 , 717–729. [ Google Scholar ]
  • Horton, J.L.; Davidson, M.T.; Kurishima, C.; Vega, R.B.; Powers, J.C.; Matsuura, T.R.; Petucci, C.; Lewandowski, E.D.; Crawford, P.A.; Muoio, D.M.; et al. The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight 2019 , 4 , e124079. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Bedi, K.C.; Snyder, N.W.; Brandimarto, J.; Aziz, M.; Mesaros, C.; Worth, A.J.; Wang, L.L.; Javaheri, A.; Blair, I.A.; Margulies, K.B.; et al. Evidence for intramyocardial disruption of lipid metabolism and increased myocardial ketone utilization in advanced human heart failure. Circulation 2016 , 133 , 706–716. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Aubert, G.; Martin, O.J.; Horton, J.L.; Lai, L.; Vega, R.B.; Leone, T.C.; Koves, T.; Gardell, S.J.; Krüger, M.; Hoppel, C.L.; et al. The Failing Heart Relies on Ketone Bodies as a Fuel. Circulation 2016 , 133 , 698–705. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Lommi, M.D.J. Blood ketone bodies in congestive heart failure. J. Am. Coll. Cardiol. 1996 , 28 , 665–672. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Voros, G.; Ector, J.; Garweg, C.; Droogne, W.; Van Cleemput, J.; Peersman, N.; Vermeersch, P.; Janssens, S. Increased cardiac uptake of ketone bodies and free fatty acids in human heart failure and hypertrophic left ventricular remodeling. Circ. Heart Fail. 2018 , 11 , e004953. [ Google Scholar ] [ CrossRef ]
  • Gormsen, L.C.; Svart, M.; Thomsen, H.H.; Sondergaard, E.; Vendelbo, M.H.; Chris-tensen, N.; Tolbod, L.P.; Harms, H.J.; Nielsen, R.; Wiggers, H.; et al. Ketone Body Infusion with 3-Hydroxybutyrate Reduces Myocardial Glucose Uptake and Increases Blood Flow in Humans: A Positron Emission Tomography Study. J. Am. Heart Assoc. 2017 , 6 , e005066. [ Google Scholar ]
  • Svart, M.; Gormsen, L.C.; Hansen, J.; Zeidler, D.; Gejl, M.; Vang, K.; Aanerud, J.; Moeller, N. Regional cerebral effects of ketone body infusion with 3-hydroxybutyrate in humans: Reduced glucose uptake, unchanged oxygen consumption and increased blood flow by positron emission tomography. A randomized, controlled trial. PLoS ONE 2018 , 13 , e0190556. [ Google Scholar ]
  • Lauritsen, K.M.; Søndergaard, E.; Luong, T.V.; Møller, N.; Gormsen, L.C. Acute Hyperketonemia Does Not Affect Glucose or Palmitate Uptake in Abdominal Organs or Skeletal Muscle. J. Clin. Endocrinol. Metab. 2020 , 105 , 1785–1790. [ Google Scholar ] [ CrossRef ]
  • Du, Z.; Shen, A.; Huang, Y.; Su, L.; Lai, W.; Wang, P.; Xie, Z.; Xie, Z.; Zeng, Q.; Ren, H.; et al. 1H-NMR-based metabolic analysis of human serum reveals novel markers of myocardial energy expenditure in heart failure patients. PLoS ONE 2014 , 9 , e88102. [ Google Scholar ] [ CrossRef ]
  • Seki, M.; Powers, J.C.; Maruyama, S.; Zuriaga, M.A.; Wu, C.L.; Kurishima, C.; Kim, L.; Johnson, J.; Poidomani, A.; Wang, T.; et al. Acute and chronic increases of circulating FSTL1 normalize energy substrate metabolism in pacing-induced heart failure. Circulation 2018 , 11 , e004486. [ Google Scholar ]
  • Guo, Y.; Liu, X.; Li, T.; Zhao, J.; Yang, Y.; Yao, Y.; Wang, L.; Yang, B.; Ren, G.; Tan, Y.; et al. Alternate-Day Ketogenic Diet Feeding Protects against Heart Failure through Preservation of Ketogenesis in the Liver. Oxid. Med. Cell. Longev. 2022 , 2022 , 4253651. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Obokata, M.; Negishi, K.; Sunaga, H.; Ishida, H.; Ito, K.; Ogawa, T.; Iso, T.; Ando, Y.; Kurabayashi, M. Association between Circulating Ketone Bodies and Worse Outcomes in Hemodialysis Patients. J. Am. Heart Assoc. 2017 , 6 , e006885. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Flores-Guerrero, J.L.; Westenbrink, B.D.; Connelly, M.A.; Otvos, J.D.; Groothof, D.; Shalaurova, I.; Garcia, E.; Navis, G.; de Boer, R.A.; Bakker, S.J.L.; et al. Association of beta-hydroxybutyrate with development of heart failure: Sex differences in a Dutch population cohort. Eur. J. Clin. Investig. 2021 , 51 , e13468. [ Google Scholar ] [ CrossRef ]
  • Manolis, A.S.; Manolis, T.A.; Manolis, A.A. Ketone Bodies and Cardiovascular Disease: An Alternate Fuel Source to the Rescue. Int. J. Mol. Sci. 2023 , 24 , 3534. [ Google Scholar ] [ CrossRef ]
  • Kashiwagi, Y.; Nagoshi, T.; Inoue, Y.; Tanaka, Y.; Takahashi, H.; Oi, Y.; Kimura, H.; Minai, K.; Yoshimura, M. Close linkage between blood total ketone body levels and B-type natriuretic peptide levels in patients with cardiovascular disorders. Sci. Rep. 2021 , 11 , 6498. [ Google Scholar ] [ CrossRef ]
  • Marcondes-Braga, F.G.; Batista, G.L.; Gutz, I.G.R.; Saldiva, P.H.N.; Mangini, S.; Issa, V.S.; Ayub-Ferreira, S.M.; Bocchi, E.A.; Pereira, A.C.; Bacal, F. Impact of exhaled breath acetone in the prognosis of patients with heart failure with reduced ejection fraction (HFrEF). One year of clinical follow-up. PLoS ONE 2016 , 11 , e0168790. [ Google Scholar ]
  • Voorrips, S.N.; Boorsma, E.M.; Beusekamp, J.C.; DE-Boer, R.A.; Connelly, M.A.; Dullaart, R.P.F.; VAN-DER-Meer, P.; VAN-Veldhuisen, D.J.; Voors, A.A.; Damman, K.; et al. Longitudinal Changes in Circulating Ketone Body Levels in Patients with Acute Heart Failure: A Post Hoc Analysis of the EMPA-Response-AHF Trial. J. Card. Fail. 2023 , 29 , 33–41. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Nielsen, R.; Møller, N.; Gormsen, L.C.; Tolbod, L.P.; Hansson, N.H.; Sorensen, J.; Harms, H.J.; Frøkiær, J.; Eiskjaer, H.; Jespersen, N.R.; et al. Cardiovascular Effects of Treatment With the Ketone Body 3-Hydroxybutyrate in Chronic Heart Failure Patients. Circulation 2019 , 139 , 2129–2141. [ Google Scholar ] [ CrossRef ]
  • Kashiwaya, Y.; Sato, K.; Tsuchiya, N.; Thomas, S.; Fell, D.A.; Veech, R.L.; Passonneau, J.V. Control of glucose utilization in working perfused rat heart. J. Biol. Chem. 1994 , 269 , 25502–25514. [ Google Scholar ] [ CrossRef ]
  • Sato, K.; Kashiwaya, Y.; Keon, C.A.; Tsuchiya, N.; King, M.T.; Radda, G.K.; Chance, B.; Clarke, K.; Veech, R.L. Insulin, ketone bodies, and mitochondrial energy transduction. FASEB J. 1995 , 9 , 651–658. [ Google Scholar ] [ CrossRef ]
  • Schugar, R.C.; Moll, A.R.; André d’Avignon, D.; Weinheimer, C.J.; Kovacs, A.; Crawford, P.A. Cardiomyocyte-specific deficiency of ketone body metabolism promotes accelerated pathological remodeling. Mol. Metab. 2014 , 3 , 754–769. [ Google Scholar ] [ CrossRef ]
  • Luong, T.V.; Abild, C.B.; Bangshaab, M.; Gormsen, L.C.; Søndergaard, E. Ketogenic Diet and Cardiac Substrate Metabolism. Nutrients 2022 , 14 , 1322. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Matsuura, T.R.; Puchalska, P.; Crawford, P.A.; Kelly, D.P. Ketones and the Heart: Meta-bolic Principles and Therapeutic Implications. Circ Res. 2023 , 132 , 882–898. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Likhodii, S.S.; Musa, K.; Mendonca, A.; Dell, C.; Burnham, W.M.; Cunnane, S.C. Dietary fat, ketosis, and seizure resistance in rats on the ketogenic diet. Epilepsia 2000 , 41 , 1400–1410. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Wang, Y.; Liu, Z.; Han, Y.; Xu, J.; Huang, W.; Li, Z. Medium Chain Triglycerides enhances exercise endurance through the increased mitochondrial biogenesis and metabolism. PLoS ONE 2018 , 13 , e0191182. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Cardoso, D.A.; Moreira, A.S.; de Oliveira, G.M.; Raggio Luiz, R.; Rosa, G. A Coconut Extra Virgin Oil-Rich Diet Increases Hdl Cholesterol and Decreases Waist Circumference and Body Mass in Coronary Artery Disease Patients. Nutr. Hosp. 2015 , 32 , 2144–2152. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Galley, H.F.; Webster, N.R. Physiology of the endothelium. Br. J. Anaesth. 2004 , 93 , 105–113. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Nappi, F.; Fiore, A.; Masiglat, J.; Cavuoti, T.; Romandini, M.; Nappi, P.; Avtaar Singh, S.S.; Couetil, J.-P. Endothelium-Derived Relaxing Factors and Endothelial Function: A Systematic Review. Biomedicines 2022 , 10 , 2884. [ Google Scholar ] [ CrossRef ]
  • Weis, E.M.; Puchalska, P.; Nelson, A.B.; Taylor, J.; Moll, I.; Hasan, S.S.; Dewenter, M.; Hagenmüller, M.; Fleming, T.; Poschet, G.; et al. Ketone body oxidation increases cardiac endothelial cell proliferation. EMBO Mol. Med. 2022 , 14 , e14753. [ Google Scholar ] [ CrossRef ]
  • Devaraj, S.; Cheung, A.T.; Jialal, I.; Griffen, S.C.; Nguyen, D.; Glaser, N.; Aoki, T. Evidence of increased inflammation and microcirculatory abnormalities in patients with type 1 diabetes and their role inmicrovascular complications. Diabetes 2007 , 56 , 2790–2796. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • White, N.H. Diabetic ketoacidosis in children. Endocrinol. Metab. Clin. North Am. 2000 , 29 , 657–682. [ Google Scholar ] [ CrossRef ]
  • Bialo, S.R.; Agrawal, S.; Boney, C.M.; Quintos, J.B. Rare complications of pediatric diabetic ketoacidosis. World J. Diabetes 2015 , 6 , 167–174. [ Google Scholar ] [ CrossRef ]
  • Ma, D.; Wang, A.C.; Parikh, I.; Green, S.J.; Hoffman, J.D.; Chlipala, G.; Murphy, M.P.; Sokola, B.S.; Bauer, B.; Hartz, A.M.S.; et al. Ketogenic diet enhances neurovascular function with altered gut microbiome in young healthy mice. Sci Rep. 2018 , 8 , 6670. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Mccarthy, C.G.; Chakraborty, S.; Schreckenberger, Z.; Wenceslau, C.F.; Joe, B. β-hydroxybutyrate (βHOB) increases nitric oxide synthase activity in resistance arteries from dahl salt-sensitive rats. FASEB J. 2019 , 33 , 829. [ Google Scholar ] [ CrossRef ]
  • Newman, J.C.; Covarrubias, A.J.; Zhao, M.; Yu, X.; Gut, P.; Ng, C.P.; Huang, Y.; Haldar, S.; Verdin, E. Ketogenic Diet Reduces Midlife Mortality and Improves Memory in Aging Mice. Cell Metab. 2017 , 26 , 547–557.e8. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Han, Y.M.; Bedarida, T.; Ding, Y.; Somba, B.K.; Lu, Q.; Wang, Q.; Song, P.; Zou, M.H. β-Hydroxybutyrate Prevents Vascular Senescence through hnRNP A1-Mediated Upregulation of Oct4. Mol Cell. 2018 , 71 , 1064–1078.e5. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Meroni, E.; Papini, N.; Criscuoli, F.; Casiraghi, M.C.; Massaccesi, L.; Basilico, N.; Erba, D. Metabolic Responses in Endothelial Cells Following Exposure to Ketone Bodies. Nutrients 2018 , 10 , 250. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Pirola, L.; Balcerczyk, A.; Tothill, R.W.; Haviv, I.; Kaspi, A.; Lunke, S.; Ziemann, M.; Karagiannis, T.; Tonna, S.; Kowalczyk, A.; et al. Genome-wide analysis distinguishes hyperglycemia regulated epigenetic signatures of primary vascular cells. Genome Res. 2011 , 21 , 1601–1615. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Coppola, G.; Natale, F.; Torino, A.; Capasso, R.; D’Aniello, A.; Pironti, E.; Santoro, E.; Calabrò, R.; Verrotti, A. The impact of the ketogenic diet on arterial morphology and en-dothelial function in children and young adults with epilepsy: A case-control study. Seizure 2014 , 23 , 260–265. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Buscemi, S.; Verga, S.; Tranchina, M.R.; Cottone, S.; Cerasola, G. Effects of hypocaloric very-low-carbohydrate diet vs. Mediterranean diet on endothelial function in obese women. Eur. J. Clin. Investig. 2009 , 39 , 339–347. [ Google Scholar ] [ CrossRef ]
  • DeFronzo, R.A. The effect of insulin on renal sodium metabolism. A review with clinical implications. Diabetologia 1981 , 21 , 165–171. [ Google Scholar ] [ CrossRef ]
  • Brands, M.W. Role of Insulin-Mediated Antinatriuresis in Sodium Homeostasis and Hypertension. Hypertension 2018 , 72 , 1255–1262. [ Google Scholar ] [ CrossRef ]
  • Palmer, B.F.; Clegg, D.J. Physiology and pathophysiology of potassium homeostasis. Adv. Physiol. Educ. 2016 , 40 , 480–490. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Harvey, C.J.D.C.; Schofield, G.M.; Williden, M. The use of nutritional supplements to induce ketosis and reduce symptomsassociated with keto-induction: A narrative review. PeerJ 2018 , 6 , e4488. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Zupec-Kania, B.; Zupanc, M.L. Long-term management of the ketogenic diet: Seizure monitoring, nutrition, and supplementation. Epilepsia 2008 , 49 (Suppl. S8), 23–26. [ Google Scholar ] [ CrossRef ]
  • Cordain, L. Nutritional Deficiencies of Ketogenic Diets. 2018. Available online: https://www.researchgate.net/publication/332098774_Nutritional_Deficiencies_of_Ketogenic_Diets?channel=doi&linkId=5c9f99e2a6fdccd46045868c&showFulltext=true (accessed on 23 July 2023). License CC BY-NC-ND 4.0.
  • Iqbal, S.; Klammer, N.; Ekmekcioglu, C. The Effect of Electrolytes on Blood Pressure: A Brief Summary of Meta-Analyses. Nutrients 2019 , 11 , 1362. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Gallo, G.; Volpe, M.; Savoia, C. Endothelial Dysfunction in Hypertension: Current Concepts and Clinical Implications. Front. Med. 2021 , 8 , 798958. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kostov, K. The Causal Relationship between Endothelin-1 and Hypertension: Focusing on Endothelial Dysfunction, Arterial Stiffness, Vascular Remodeling, and Blood Pressure Regulation. Life 2021 , 11 , 986. [ Google Scholar ] [ CrossRef ]
  • Polito, R.; Messina, G.; Valenzano, A.; Scarinci, A.; Villano, I.; Monda, M.; Cibelli, G.; Porro, C.; Pisanelli, D.; Monda, V.; et al. The Role of Very Low Calorie Ketogenic Diet in Sympathetic Activation through Cortisol Secretion in Male Obese Population. J. Clin. Med. 2021 , 10 , 4230. [ Google Scholar ] [ CrossRef ]
  • Polito, R.; Valenzano, A.; Monda, V.; Cibelli, G.; Monda, M.; Messina, G.; Villano, I.; Messina, A. Heart Rate Variability and Sympathetic Activity Is Modulated by Very Low-Calorie Ketogenic Diet. Int. J. Environ. Res. Public Health 2022 , 19 , 2253. [ Google Scholar ] [ CrossRef ]
  • Barrea, L.; Verde, L.; Camajani, E.; Šojat, A.S.; Marina, L.; Savastano, S.; Colao, A.; Caprio, M.; Muscogiuri, G. Effects of very low-calorie ketogenic diet on hypothalamic–pituitary–adrenal axis and renin–angiotensin–aldosterone system. J. Endocrinol. Investig. 2023 , 46 , 1509–1520. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Belany, P.; Kackley, M.L.; Zhao, S.; Kluwe, B.; Buga, A.; Crabtree, C.D.; Nedungadi, D.; Kline, D.; Brock, G.; Simonetti, O.P.; et al. Effects of Hypocaloric Low-Fat, Ketogenic, and Ketogenic and Ketone Supplement Diets on Aldosterone and Renin. J. Clin. Endocrinol. Metab. 2023 , 108 , 1727–1739. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Di Raimondo, D.; Buscemi, S.; Musiari, G.; Rizzo, G.; Pirera, E.; Corleo, D.; Pinto, A.; Tuttolomondo, A. Ketogenic Diet, Physical Activity, and Hypertension—A Narrative Review. Nutrients 2021 , 13 , 2567. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Williams, B.; Mancia, G.; Spiering, W.; Agabiti Rosei, E.; Azizi, M.; Burnier, M.; Clement, D.; Coca, A.; De Simone, G.; Dominiczak, A.; et al. 2018 Practice Guidelines for the management of arterial hypertension of the European Society of Hypertension and the European Society of Cardiology: ESH/ESC Tas k Force for the Management of Arterial Hypertension. J. Hypertens. 2018 , 36 , 2284–2309. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Di Raimondo, D.; Musiari, G.; Miceli, G.; Arnao, V.; Pinto, A. Preventive and Therapeutic Role of Muscle Contraction against Chronic Diseases. Curr. Pharm. Des. 2016 , 22 , 4686–4699. [ Google Scholar ] [ CrossRef ]
  • Barrea, L.; Verde, L.; Santangeli, P.; Lucà, S.; Docimo, A.; Savastano, S.; Colao, A.; Muscogiuri, G. Very low-calorie ketogenic diet (VLCKD): An antihypertensive nutritional approach. J. Transl. Med. 2023 , 21 , 128. [ Google Scholar ] [ CrossRef ]
  • Rinaldi, R.; De Nucci, S.; Castellana, F.; Di Chito, M.; Giannuzzi, V.; Shahini, E.; Zupo, R.; Lampignano, L.; Piazzolla, G.; Triggiani, V.; et al. The Effects of Eight Weeks’ Very Low-Calorie Ketogenic Diet (VLCKD) on Liver Health in Subjects Affected by Overweight and Obesity. Nutrients 2023 , 15 , 825. [ Google Scholar ] [ CrossRef ]
  • Yancy, W.S., Jr.; Westman, E.C.; McDuffie, J.R.; Grambow, S.C.; Jeffreys, A.S.; Bolton, J.; Chalecki, A.; Oddone, E.Z. A randomized trial of a low-carbohydrate diet vs orlistat plus a low-fat diet for weight loss. Arch. Intern. Med. 2010 , 170 , 136–145, Erratum in JAMA Intern. Med. 2015 , 175 , 470. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Foster, G.D.; Wyatt, H.R.; Hill, J.O.; Makris, A.P.; Rosenbaum, D.L.; Brill, C.; Stein, R.I.; Mohammed, B.S.; Miller, B.; Rader, D.J.; et al. Weight and metabolic outcomes after 2 years on a low-carbohydrate versus low-fat diet: A randomized trial. Ann. Intern. Med. 2010 , 153 , 147–157. [ Google Scholar ] [ CrossRef ]
  • Samaha, F.F.; Iqbal, N.; Seshadri, P.; Chicano, K.L.; Daily, D.A.; McGrory, J.; Williams, T.; Williams, M.; Gracely, E.J.; Stern, L. A low-carbohydrate as compared with a low-fat diet in severe obesity. N. Engl. J. Med. 2003 , 348 , 2074–2081. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Tzenios, N.; Lewis, E.D.; Crowley, D.C.; Chahine, M.; Evans, M. Examining the Efficacy of a Very-Low-Carbohydrate Ketogenic Diet on Cardiovascular Health in Adults with Mildly Elevated Low-Density Lipoprotein Cholesterol in an Open-Label Pilot Study. Metab. Syndr. Relat. Disord. 2022 , 20 , 94–103. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Bueno, N.; De Melo, I.; De Oliveira, S.; Da Rocha Ataide, T. Very-low-carbohydrate ketogenic diet v. low-fat diet for long-term weight loss: A meta-analysis of randomised controlled trials. Br. J. Nutr. 2013 , 110 , 1178–1187. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Markovikj, G.; Knights, V.; Kljusurić, J.G. Ketogenic Diet Applied in Weight Reduction of Overweight and Obese Individuals with Progress Prediction by Use of the Modified Wishnofsky Equation. Nutrients 2023 , 15 , 927. [ Google Scholar ] [ CrossRef ]
  • Muscogiuri, G.; Barrea, L.; Laudisio, D.; Pugliese, G.; Salzano, C.; Savastano, S.; Colao, A. The management of very low-calorie ketogenic diet in obesity outpatient clinic: A practical guide. J. Transl. Med. 2019 , 17 , 356. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Powell-Wiley, T.M.; Poirier, P.; Burke, L.E.; Després, J.P.; Gordon-Larsen, P.; Lavie, C.J.; Lear, S.A.; Ndumele, C.E.; Neeland, I.J.; Sanders, P.; et al. American Heart Association Council on Lifestyle and Cardiometabolic Health; Council on Cardiovascular and Stroke Nursing; Council on Clinical Cardiology; Council on Epidemiology and Prevention; and Stroke Council. Obesity and Cardiovascular Disease: A Scientific Statement from the American Heart Association. Circulation 2021 , 143 , e984–e1010. [ Google Scholar ] [ CrossRef ] [ PubMed ]

Click here to enlarge figure

Research Type, YearPurpose of the StudyType of DietChanges in the Lipid ProfileReferences
RCT, 2022Observation of periodic ketogenic diet for effect on overweight or obese patients newly diagnosed as T2DM.Ketogenic diet (KD)
vs.
standard diabetes diet (SDD)
KD vs. SDD:
-decrease in cholesterol from 4.54 ± 0.69 mmol/L to 4.02 ± 0.43 mmol/L (SDD from 4.56 ± 0.67 mmol/L to 4.23 ± 0.47 mmol/L)
-decrease in triglycerides from 1.76 ± 0.59 mmol/L to 1.44 ± 0.26 mmol/L (SDD from 1.81 ± 0.78 mmol/L to 1.66 ± 0.46 mmol/L)
-decrease in LDL from 2.75 ± 0.65 mmol/L to 2.34 ± 0.45 mmol/L (SDD from 2.77 ± 0.69 mmol/L to 2.59 ± 0.58 mmol/L)
-increase in HDL from 1.08 ± 0.11 mmol/L to 1.21 ± 0.23 mmol/L (SDD from 1.09 ± 0.19 mmol/L to 1.12 ± 0.20 mmol/L)
[ ]
RCT, 2022Comparison of 2 low-carbohydrate diets with 3 key similarities and 3 key differences for their effects on glucose control and cardiometabolic risk factors in individuals with prediabetes and T2DM.Well-formulated ketogenic diet (WFKD)
vs.
the Mediterranean-plus diet (Med-Plus)
WFKD vs. Med-Plus:
-reduction in triglycerides from 118.8 mg/dL to 99.5 mg/dL (in Med-Plus from 131.1 mg/dL to 121.7 mg/dL)
-increase in HDL concentration from 49.1 mg/dL to 54.1 mg/dL (in Med-Plus from 48 mg/dL to 47.9 mg/dL)
-increase in LDL concentration from 97.8 mg/dL to 111.3 mg/dL (in Med-Plus from 111.5 mg/dL to 95.3 mg/dL)
[ ]
RCT, 2022Assessment of the clinical advantage of combining two preoperative strategies (continuous positive airway pressure (CPAP) and low-calorie ketogenic diet (LCKD)) compared to CPAP alone, to improve apnea–hypopnea index (AHI) score, hypertension (HTN), dyslipidemia (DLP), insulin resistance (IR) and C-reactive protein (CRP) levels in patients with severe obesity and obstructive sleep apnea syndrome (OSAS) scheduled for bariatric surgery (BS).Low-calorie ketogenic diet (LCKD) + continuous positive airway pressure (CPAP)
vs.
only continuous positive airway pressure (CPAP)
LCKD + CPAP vs. CPAP:
-reduction in total cholesterol from 200.1 ± 30.1 mg/dL to 180.4 ± 35.2 mg/dL (CPAP from 196.1 ± 32.9 mg/dL to 180.8 ± 33.0 mg/dL)
-decrease in LDL from 127.4 ± 26.8 mg/dL to 107.1 ± 37.1 mg/dL (CPAP from 128 ± 30.2 mg/dL to 112.9 ± 34.9 mg/dL)
-decrease in triglycerides from 191 ± 41.7 mg/dL to 130 ± 79 mg/dL (CPAP from 151.6 ± 62.5 mg/dL to 129.7 ± 62.2 mg/dL)
-insignificant increase in HDL from 48.3 ± 9.41 mg/dL to 48.8 ± 10.4 mg/dL (CPAP from 46.4 ± 10.3 mg/dL to 47.3 ± 9.8 mg/dL)
[ ]
RCT, 2021Investigation and comparison of the effects of two iso-energetic hypo-caloric ketogenic hyper-ketonemic and non-ketogenic low-carbohydrate high-fat high-cholesterol diets on body-composition, muscle strength and hormonal profile in experienced resistance-trained middle-aged men.Ketogenic diets (KD)
vs.
non-ketogenic diets (NKD)
(in several variants)
No significant differences in lipid profile.
-In KD—change in TC from 4.44 ± 0.37 mmol/L to 4.43 ± 0.30 mmol/L (NKD from 4.49 ± 0.31 mmol/L to 4.52 ± 0.30 mol/L)
-In KD—change in TG from 0.99 ± 0.25 mmol/L to 0.95 ± 0.26 mmol/L (NKD from 0.90 ± 0.14 mmol/L to 0.85 ± 0.13 mmol/L)
-In KD—change in HDL from 1.28 ± 0.13 mmol/L to 1.36 ± 0.12 mmol/L (NKD from 1.28 ± 0.13 mmol/L to 1.36 ± 0.12 mmol/L)
-In KD—change in LDL from 2.40 ± 0.21 mmol/L to 2.45 ± 0.25 mmol/L (NKD from 2.57 ± 0.41 mmol/L to 2.59 ± 0.39 mmol/L)
[ ]
RCT, 2021Investigation of the effect of a ketogenic LCHF diet on low-density lipoprotein (LDL) cholesterol (primary outcome), LDL cholesterol subfractions and conventional cardiovascular risk factors in the blood of healthy, young, and normal-weight women.Ketogenic low-carbohydrate high-fat (LCHF) diet
vs.
National Food Agency recommended control diet (NFACD)
The LCHF diet:
-increases in LDL cholesterol in every woman with a treatment effect of 1.82 mM (p < 0.001) (primary outcome at baseline = 2.1 ± 0.6 mM)
-increases in apolipoprotein B-100 (ApoB) (treatment effect (95% Cl) = 0.50 [0.35, 0.65], primary outcome at baseline = 0.70 ± 0.15 g/L)
-increases in LDL 1–2 (large, buoyant LDL) (treatment effect (95% Cl) = 31.56 [21.60, 41.51], primary outcome at baseline = 42.1 ± 14.6 mg/dL)
-increases in LDL 3–7 (small, dense LDL) (treatment effect (95% Cl) = 4.51 [1.87, 7.16], primary outcome at baseline = 2.7 ± 2.5 mg/dL)
[ ]
RCT, 2020Comparison of the efficacy, safety and effect of 45-day isocaloric very-low-calorie ketogenic diets (VLCKDs) incorporating whey, vegetable or animal protein on the microbiota in patients with obesity and insulin resistance, to test the hypothesis that protein source may modulate the response to VLCKD interventions.Isocaloric VLCKD regimens (≤800 kcal/day) containing whey (WPG), plant (VPG) or animal protein (APG)Significant reductions in total cholesterol (TC), LDL and triglycerides (TG) in all VLCKD groups:
-TC in WPG from 214.8 ± 31.5 mg/dL to 166.2 ± 43.6 mg/dL, in VPG from 220.9 ± 51.6 mg/dL to 170.7 ± 36.3 mg/dL, in APG from 226.9 ± 32.7 mg/dL to 191.2 ± 34.2 mg/dL
-LDL in WPG from 132.8 ± 30.8 mg/dL to 100.8 ± 38.4 mg/dL, in VPG from 136.1 ± 41.3 mg/dL to 97.5 ± 32.3 mg/dL, in APG from 143.9 ± 25.8 mg/dL to 118.5 ± 23.1 mg/dL
-TG in WPG from 131.0 ± 44.9 mg/dL to 94.6 ± 32.0 mg/dL, in VPG from 170.1 ± 126.9 mg/dL to 117.6 ± 42.7 mg/dL, in APG from 124.25 ± 58 mg/dL to 82.25 ± 33.32 mg/dL
-insignificant changes in HDL: in WPG from 51.7 ± 12.3 mg/dL to 46.1 ± 7.5 mg/dL, in VPG from 51.2 ± 12.8 mg/dL to 49.0 ± 9.5 mg/dL, in APG from 57.9 ± 23.7 mg/dL to 56.2 ± 18.0 mg/dL
[ ]
RCT, 2020Comparison of the influence of a 12-week, well-planned, low-calorie ketogenic diet (LCKD) on hyperglycemic, hyperinsulinemic and lipid profiles in adult, overweight or obese females.Low-calorie ketogenic diet (LCKD)
vs.
control group (CG) (typical diet)
Significant reduction in TG and increase in HDL in LCKD compared to CG:
-TG in LCKD decreased from 213.45 ± 63.60 mg/dL to 129.13 ± 46.23 mg/dL (in CG from 210.57 ± 36.45 mg/dL to 206.44 ± 50.03 mg/dL)
-HDL in LCKD increased from 36.71 ± 4.42 mg/dL to 52.99 ± 7.77 mg/dL (in CG from 44.14 ± 5.07 to 43.01 ± 5.03 mg/dL)
[ ]
RCT, 2017Comparison of the effects of a ketogenic diet vs. a moderate-carbohydrate diet on overweight adults with type 2 diabetes mellitus or pre-diabetes.Very low-carbohydrate ketogenic diet (VLCKD)
vs.
moderate-carbohydrate, calorie-restricted, low-fat diet (MCCRD)
-In VLCKD, there was a significant reduction in TG from 102.6 mg/dL (81.8, 123.4) to 86.2 mg/dL (68.6, 103.7) in the 6th month and 92.7 mg/dL (73.6, 111.7) in the 12th month (in MCCRD from 158.9 mg/dL (128.8, 189.1) to 143.2 mg/dL (115.6, 170.9) in the 6th month and 173.4 mg/dL (138.1, 208.7) in the 12th month)
-In VLCKD, there was an increase in HDL from 48.4 mg/dL (42.6, 54.2) to 51.9 mg/dL (45.7, 58.2) in the 6th month and 53.3 mg/dL (46.8, 59.8) in the 12th month (in MCCRD from 45.8 mg/dL (40.6, 51.0) to 48.1 mg/dL (42.5, 53.6) in the 6th month and 48.9 mg/dL (43.3, 54.5) in the 12th month)
-In VLCKD, there was an increase in LDL from 88.7 mg/dL (76.3, 101.1) to 97.9 mg/dL (85.4, 110.5) in the 6th month and 95.6 mg/dL (82.3, 108.9) in the 12th month (in MCCRD from 98.1 mg/dL (86.4, 109.8) to 88.1 mg/dL (76.0, 100.1) in the 6th month and 96.1 mg/dL (83.7, 108.5) in the 12th month)
[ ]
RCT, 2015Evaluating the effects of ω-3 supplementation during a ketogenic diet in overweight subjects.Ketogenic diet (KD)
vs.
ketogenic diet + ω-3 supplementation (KDO3)
In both dietary versions, there was a reduction in TC, LDL, TG and an increase in HDL.
-TC in KD decreased from 217.25 ± 15.84 mg/dL to 201.28 ± 6.79 mg/dL (in KDO3 from 222.39 ± 6.10 mg/dL to 204.52 ± 9.78 mg/dL)
-LDL in KD decreased from 133.41 ± 15.86 mg/dL to 123.60 ± 7.99 mg/dL (in KDO3 from 136.98 ± 7.06 mg/dL to 127.56 ± 7.19 mg/dL)
-TG in KD decreased from 237.81 ± 20.26 mg/dL to 197.27 ± 6.1 mg/dL (in KDO3 from 230.79 ± 25.66 mg/dL to 185.54 ± 9.64 mg/dL)
-HDL in KD increased slightly from 36.28 ± 2.23 mg/dL to 39.25 ± 1.37 mg/dL (in KDO3 from 39.55 ± 2.99 to 40.25 ± 2.63 mg/dL)
[ ]
RCT, 2012Comparison of the efficacy and metabolic impact of ketogenic and hypocaloric diets in obese children and adolescents.Ketogenic diet (KD)
vs.
hypocaloric diet (HD)
-In KD, there was an increase in TC from 4.4 ± 0.85 mmol/L to 4.63 ± 0.75 mmol/L (in HD from 4.05 ± 0.94 mmol/L to 4.03 ± 0.89 mmol/L)
-In KD, there was an increase in HDL from 1.27 ± 0.26 mmol/L to 1.38 ± 0.25 mmol/L (in HD from 1.13 ± 0.20 mmol/L to 1.23 ± 0.23 mmol/L)
-In KD, there was an increase in LDL from 2.72 ± 0.69 mmol/L to 2.86 ± 0.65 mmol/L (in HD from 2.6 ± 0.83 mmol/L to 2.55 ± 0.77 mmol/L)
-In KD, there was a reduction in TG from 0.83 ± 0.35 mmol/L to 0.81 ± 0.39 mmol/L (in HD from 0.89 ± 0.57 mmol/L to 0.80 ± 0.40 mmol/L)
[ ]
Type of Research, YearPurpose of the StudyDiet TypeBlood Pressure ChangesReferences
Prospective pilot clinical trial,
2023
Evaluate the effect of very low-calorie ketogenic diet (VLCKD) on blood pressure (BP) in women with obesity and hypertension.Very low-calorie ketogenic diet (VLCKD)Relative to baseline values, after 45 days, there was:
-a reduction in systolic blood pressure by an average of −12.89% (from an average of 140.88 ± 8.99 mmHg to 122.56 ± 10.08 mmHg)
-a reduction in diastolic blood pressure by a mean of −10.77% (from a mean of 88.90 ± 6.71 mmHg to 78.94 ± 6.68 mmHg).
[ ]
Prospective study,
2023
Evaluate the efficacy and safety of VLCKD on non-alcoholic fatty liver disease (NAFLD) and parameters commonly associated with this condition in overweight and obese subjects who did not take any drugs.Very low-calorie ketogenic diet (VLCKD)Relative to the initial values, after 8 weeks, there was:
-a reduction in systolic blood pressure from an average of 133.51 ± 12.86 mmHg to 123.27 ± 10.51 mmHg
-a reduction in diastolic blood pressure from a mean of 81.73 ± 8.09 mmHg to 75.27 ± 7.84 mmHg.
[ ]
RCT, 2022Assessment of the clinical advantage of combining two preoperative strategies (continuous positive airway pressure (CPAP) and low-calorie ketogenic diet (LCKD)) compared to CPAP alone, to improve apnea–hypopnea index (AHI) score, hypertension (HTN), dyslipidemia (DLP), insulin resistance (IR) and C-reactive protein (CRP) levels in patients with severe obesity and obstructive sleep apnea syndrome (OSAS) scheduled for bariatric surgery (BS).Low-calorie ketogenic diet (LCKD) + continuous positive airway pressure (CPAP)
vs.
only continuous positive airway pressure (CPAP)
LCKD + CPAP vs. CPAP:
-greater mean reduction in systolic blood pressure from 142.8 ± 13.3 mmHg to 133 ± 11.9 mmHg (in CPAP from 134.2 ± 10.4 mmHg to 130 ± 9.7 mmHg)
-increased mean diastolic blood pressure reduction from 85.4 ± 8.38 mmHg to 78.7 ± 6.43 mmHg (on CPAP from 87 ± 11.6 mmHg to 82 ± 9.5 mmHg).
[ ]
Pilot clinical trial,
2022
Investigate the efficacy of a very-low-carbohydrate ketogenic diet (VLCKD), known as Nic’s Ketogenic Diet, for 140 days on cardiometabolic markers in healthy adults with mildly elevated low-density lipoprotein cholesterol (LDL-C).Very-low-carbohydrate ketogenic diet-Systolic blood pressure decreased by 5.3% from baseline on day 140 of VLCKD.
-There was a significant increase in diastolic blood pressure on day 28; however, there was no significant change on days 56, 70, 84, 112 and 140.
[ ]
RCT, 2020Comparison of the efficacy, safety and effect of 45-day isocaloric very-low-calorie ketogenic diets (VLCKDs) incorporating whey, vegetable or animal protein on the microbiota in patients with obesity and insulin resistance, to test the hypothesis that the protein source may modulate the response to VLCKD interventions.Isocaloric VLCKD regimens (≤800 kcal/day) containing whey (WPG), plant (VPG) or animal protein (APG)Relative to baseline values, after 45 days, there was:
-a reduction in mean systolic pressure values (in WPG from 132 ± 10 mmHg to 124 ± 13 mmHg, in VPG from 131 ± 8 mmHg to 121 ± 10 mmHg, in APG from 129 ± 9 mmHg to 121 ± 16 mmHg)
-a reduction in mean diastolic pressure values (on WPG from 78 ± 11 mmHg to 70 ± 9 mmHg, on VPG from 78 ± 10 mmHg to 72 ± 10 mmHg, on APG from 78 ± 10 mmHg to 71 ± 9 mmHg)
[ ]
Meta-analysis,
2020
Evaluation of the efficacy and safety of VLCKD in overweight and obese patients.Very-low-calorie ketogenic diet (VLCKD)VLCKD was associated with an average reduction in systolic blood pressure of −8 mmHg and diastolic blood pressure of −7 mmHg.[ ]
RCT,
2017
Comparison of the effects of a ketogenic diet vs. a moderate-carbohydrate diet in overweight adults with type 2 diabetes mellitus or pre-diabetes.Very-low-carbohydrate ketogenic diet (VLCKD)
vs.
moderate-carbohydrate, calorie-restricted, low-fat diet (MCCRD)
There was a slight reduction in diastolic blood pressure in both groups:
-in LCK from an average of 77.1 mmHg (74.0, 80.3) to 77.1 mmHg (74.0, 80.1) in the 6th month and to 75.6 mmHg (72.5, 78.8) in the 12th month
-in MCCRD from an average of 81.1 mmHg (78.2, 84.1) to 80.8 mmHg (77.9, 83.7) in the 6th month and 78.4 mmHg (75.5, 81.4) in the 12th month.
There were small changes in systolic blood pressure in both groups:
-in LCK from an average of 127.1 mmHg (121.9, 132.3) to 130.7 mmHg (125.7, 135.7) in the 6th month and 130.3 mmHg (125.2, 135.4) in the 12th month
-in MCCRD from an average of 129.2 mmHg (124.6, 133.7) to 130.4 mmHg (125.6, 135.1) in the 6th month and 127.5 mmHg (122.7, 132.4) in the 12th month.
[ ]
Systematic review with meta-analysis,
2013
Investigate whether individuals assigned to a VLCKD (i.e., a diet with no more than 50 g carbohydrates/d) achieve better long-term body weight and cardiovascular risk factor management when compared with individuals assigned to a conventional low-fat diet (LFD, i.e., a restricted-energy diet with less than 30% of energy from fat).Very-low-carbohydrate ketogenic diet (VLCKD)
vs.
dieta niskotłuszczowa z deficytem kalorycznym (LFD)
-There was a significant difference in favor of the VLCKD in lowering diastolic blood pressure (WMD—1–43 (95% CI—2–49, 0–37) mmHg)
-to a lesser extent, there was a difference in lowering systolic blood pressure (WMD in favor of the VLCKD—1–47 (95% CI—3–44, 0–50) mmHg).
[ ]
RCT, 2012To compare the efficacy and metabolic impact of ketogenic and hypocaloric diets in obese children and adolescents.Ketogenic diet (KD)
vs.
hypocaloric diet (HD)
Mean systolic blood pressure decreased in KD from 110 ± 13 mmHg to 108 ± 13 mmHg, while diastolic blood pressure increased from a mean of 66 ± 10 mmHg to 68 ± 8 mmHg.
In HD, there was a non-significant reduction in systolic blood pressure from 107 ± 9 mmHg to 106 ± 11 mmHg, and diastolic blood pressure from a mean of 65 ± 10 mmHg to 62 ± 11 mmHg.
[ ]
RCT,
2010
Comparison of the effects of a low-carbohydrate ketogenic diet (LCKD) and orlistat therapy in combination with a low-fat diet (O + LFD) as a weight loss therapy on key parameters, i.e., body weight, blood pressure, fasting serum lipids and glycemic parameters.Low-Carb Ketogenic Diet (LCKD) vs. low-fat diet in combination with orlistat (LFD + O)Relative to baseline values after 48 weeks, there was a significantly greater reduction in blood pressure in the LCKD group compared to LFD + O:
-average systolic blood pressure decreased by −5.94 mmHg (−1.5 mmHg in LFD + O)
-average diastolic blood pressure decreased by −4.53 mmHg (in LFD + O by −0.43 mmHg).
[ ]
RCT, 2010To evaluate the effects of 2-year treatment with a low-carbohydrate or low-fat diet, each of which was combined with a comprehensive lifestyle modification program.Low-carbohydrate diet vs. low-fat diet with a caloric deficitThere was a greater reduction in mean diastolic blood pressure in the low-carbohydrate group:
-5.53 mmHg (−6.70 to −4.36) (vs. −3.05 mmHg (−4.29 to −1.81)) in the 3rd month; −5.15 mmHg (−6.49 to −3.82) (vs. −2.50 mmHg (−3.76 to −1.25)) in the 6th month; −3.25 mmHg (−4.74 to −1.76) (vs. −2.19 mmHg (−3.58 to −0.79)) in the 12th month; −3.19 mmHg (−4.66 to −1.73) (vs. −0.50 mmHg (−2.13 to 1.13)) in the 24th month.
There was a slightly greater reduction in mean systolic blood pressure in the low-carbohydrate group:
-7.74 mmHg (−9.59 to −5.89) (vs. −5.20 mmHg (−7.09 to −3.31)) in the 3rd month; −7.36 mmHg (−9.26 to −5.47) (vs. −6.97 mmHg (−8.89 to −5.05)) in the 6th month; −5. 64 mmHg (−7.62 to −3.67) (vs. −4.06 mmHg (−6.07 to −2.05)) in the 12th month; −2.68 mmHg (−5.08 to −0.27) (vs. −2.59 mmHg (−5.07 to −0.12)) in the 24th month.
[ ]
RCT,
2003
Testing the hypothesis that severely obese subjects with a high prevalence of diabetes or metabolic syndrome would achieve greater weight loss, without detrimental effects on risk factors for atherosclerosis, while on a carbohydrate-restricted (low-carbohydrate) diet than on a calorie- and fat-restricted (low-fat) diet.Low-carbohydrate diet (<30 g/d)
vs.
calorie- and fat-restricted diet
Relative to baseline values, after 6 months, there was:
-a non-significant mean reduction in systolic blood pressure of 2 mmHg and diastolic blood pressure of 1 mmHg in the low-carbohydrate group
-a non-significant mean reduction in systolic blood pressure of 2 mmHg and diastolic blood pressure of 2 mmHg in the low-carbohydrate and low-fat groups.
[ ]
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

Dyńka, D.; Kowalcze, K.; Charuta, A.; Paziewska, A. The Ketogenic Diet and Cardiovascular Diseases. Nutrients 2023 , 15 , 3368. https://doi.org/10.3390/nu15153368

Dyńka D, Kowalcze K, Charuta A, Paziewska A. The Ketogenic Diet and Cardiovascular Diseases. Nutrients . 2023; 15(15):3368. https://doi.org/10.3390/nu15153368

Dyńka, Damian, Katarzyna Kowalcze, Anna Charuta, and Agnieszka Paziewska. 2023. "The Ketogenic Diet and Cardiovascular Diseases" Nutrients 15, no. 15: 3368. https://doi.org/10.3390/nu15153368

Article Metrics

Article access statistics, further information, mdpi initiatives, follow mdpi.

MDPI

Subscribe to receive issue release notifications and newsletters from MDPI journals

Log in using your username and password

  • Search More Search for this keyword Advanced search
  • Latest content
  • BMJ Journals

You are here

  • Volume 12, Issue 2
  • The case for a ketogenic diet in the management of kidney disease
  • Article Text
  • Article info
  • Citation Tools
  • Rapid Responses
  • Article metrics

Download PDF

  • http://orcid.org/0000-0001-5427-6748 Shaminie J Athinarayanan 1 ,
  • Caroline G P Roberts 1 ,
  • Chandan Vangala 2 ,
  • Greeshma K Shetty 1 ,
  • Amy L McKenzie 3 ,
  • Thomas Weimbs 4 ,
  • Jeff S Volek 5
  • 1 Virta Health , Denver , Colorado , USA
  • 2 BCM , Houston , Texas , USA
  • 3 Abbott , Wiesbaden , Germany
  • 4 Department of Molecular Cellular & Developmental Biology , University of California Santa Barbara , Santa Barbara , California , USA
  • 5 Department of Human Sciences , The Ohio State University , Columbus , Ohio , USA
  • Correspondence to Dr Shaminie J Athinarayanan; sarassam52{at}gmail.com

Ketogenic diets have been widely used for weight loss and are increasingly used in the management of type 2 diabetes. Despite evidence that ketones have multiple positive effects on kidney function, common misconceptions about ketogenic diets, such as high protein content and acid load, have prevented their widespread use in individuals with impaired kidney function. Clinical trial evidence focusing on major adverse kidney events is sparse. The aim of this review is to explore the effects of a ketogenic diet, with an emphasis on the pleiotropic actions of ketones, on kidney health. Given the minimal concerns in relation to the potential renoprotective effects of a ketogenic diet, future studies should evaluate the safety and efficacy of ketogenic interventions in kidney disease.

  • Kidney Diseases

Data availability statement

No data are available.

This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See:  http://creativecommons.org/licenses/by-nc/4.0/ .

https://doi.org/10.1136/bmjdrc-2024-004101

Statistics from Altmetric.com

Request permissions.

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Introduction

Low carbohydrate eating patterns including a very low carbohydrate or ketogenic diet have been successfully used for weight loss and remitting type 2 diabetes (T2D). Among patients with T2D, the prevalence of chronic kidney disease (CKD), whether characterized as a reduced estimated glomerular filtration rate (eGFR) function or albuminuria is almost 40%. 1 Yet, a ketogenic diet is cautioned against in individuals with impaired kidney function, 2 in part, due to concerns about increased protein intake. The effect of protein intake in CKD is controversial, but high protein intake has been associated with hyperfiltration, increased acid excretion, and potentially, a decline in kidney function. 3 4 However, protein intake on a well-formulated ketogenic diet (WFKD) is moderate to effectively permit nutritional ketosis. Dietary analysis of very low carbohydrate studies usually reports daily protein intake ranging from 0.6 g/kg to 1.4 g/kg, 5–7 which is similar to that in the standard American diet and below the high protein threshold (≥2.0 g/kg) believed to be of concern. 8 The Kidney Disease Outcomes Quality Initiative clinical practice guideline for nutrition in CKD not dependent on dialysis recommends a “low-protein diet providing 0.55–0.6 g of dietary protein/kg body weight/day, or a very low-protein diet providing 0.28–0.43 g of dietary protein/kg of body weight/day with additional keto acid/amino acid analogs to meet protein requirements (0.55–0.60 g/kg/day).” 9 In contrast, Kidney Disease Improving Global Outcomes (KDIGO) 2022 CKD guideline recommended a slightly higher daily protein allowance of 0.8 g/kg/day for individuals with advanced CKD with or without T2D. 10 The Modification of Diet in Renal Disease (MDRD) study, a landmark trial examining the effect of protein restriction among 585 patients with non-diabetic CKD, did not demonstrate a significantly slower progression of disease, 11 and in fact a very low protein diet (0.28 g/kg/day) was associated with increased risk of death at a median follow-up of 3.2 years. 12 The null findings from MDRD are one of numerous inconsistent results studying protein restriction in patients with CKD. Taken altogether, systematic reviews have suggested—at best—a modest benefit for patients on a low protein diet 13 14 and given the aforementioned long-term data noting increased risk of death with very low protein diets, most nephrology experts are more comfortable with moderate protein restriction to the degree of 0.8 g/kg/day as recommended by the KDIGO 2022 guideline.

The impact of carbohydrate restriction interventions on kidney function is poorly understood. Existing studies consistently reported improvements in glycemic control, blood pressure, weight, and insulin resistance, all of which have favorable downstream implications for slowing kidney disease progression ( figure 1 ). In addition, ketone bodies themselves have a myriad of physiologic and signaling effects that could elicit renoprotective effects. For example, the renoprotective effect of sodium-glucose cotransporter 2 inhibitors (SGLT2i) has been postulated to be partially mediated by the modest medication-induced ketosis. 15–17 This low-grade ketosis induced by SGLT2i may directly or indirectly benefit the kidney by serving as an energy source during stress and kidney injury, and through its anti-inflammatory, antifibrotic, and antioxidant effects ( figure 1 ). 15 16 Given that SGLT2i-induced ketosis may be beneficial for the kidney, endogenously produced ketones resulting from a WFKD may prove to be another therapeutic option for diabetic nephropathy or kidney disease. 18 19

  • Download figure
  • Open in new tab
  • Download powerpoint

Summary of pleiotropic renal protective effect of ketones and carbohydrate restriction. BHB, β-hydroxybutyrate; mTORC1, mammalian target of rapamycin complex 1; NLRP3, NOD-, LRR-, and pyrin domain-containing 3; PGC-1ɑ, peroxisome proliferator-activator receptor γ (PPARγ) coactivator-1-alpha; FFAR-3, free fatty acid receptor-3; HCAR, hydroxycarboxylic acid receptor-2; HDAC-3, Histone deacetylase-3; NRF2, nuclear factor erythroid 2-related factor 2; SIRT1, silent information regulator transcript-1.

In this review, we explore the pleiotropic roles and signaling effects of ketones on kidney physiology, address potential concerns of ketogenic therapy, summarize the available literature on the effect of low carbohydrate diets on kidney function, and discuss future studies that could help address the gaps in knowledge and discrepancies in the literature.

Potential roles of ketones on kidney pathophysiology and disease

Ketones as an alternative energy-efficient fuel.

The human body naturally produces ketones, mostly in the liver, at varying rates that result in circulating ketones that span more than four orders of magnitude (<0.01 to >10 mM) 20 21 depending primarily on a person’s carbohydrate intake and insulin level. Increased lipolysis and ketogenesis are upregulated in response to a low insulin-to-glucagon ratio, which occurs during calorie restriction/fasting, prolonged exercise, consumption of a ketogenic diet, or pathologic insulin deficiency. In the liver, when production of fatty acyl coenzyme A (CoA) increases during low insulin secretion and increased lipolysis, fatty acyl CoA is transported to the mitochondrial matrix, where it is then β-oxidized to produce acetyl-CoA. 22 23 Acetyl-CoA is either converted to malonyl CoA or to acetoacetyl CoA ( figure 2 ). Acetoacetyl CoA and acetyl-CoA are further condensed by a rate-limiting enzyme, 3-hydroxy 3-methylglutaryl-CoA synthase 2 (HMGCS2) to generate hydroxymethylglutaryl CoA (HMG-CoA). HMG-CoA is then converted into acetoacetate (AcAc) by hydroxymethylglutaryl coenzyme A lyase (HMGCL). 22 23 Finally, AcAc is reduced to β-hydroxybutyrate (BHB) by BHB dehydrogenase (BDH) ( figure 2 ). Both AcAc and BHB are released from the liver and transported in the blood circulation to extrahepatic tissues where they can have signaling effects and be metabolized and released or oxidized to produce energy. BHB is a vital energy source for the brain with uptake occurring in proportion to circulating levels. As such, during prolonged starvation, ketones can provide over half the brain’s energy requirements. 24 Generally, glucose is considered the most efficient fuel since it produces more ATP per oxygen consumed with a phosphate/oxygen (P/O) ratio of 2.58. 24 25 However, in a state of insulin resistance where glucose uptake and oxidation are impaired, BHB is an effective alternative dense energy molecule with a P/O ratio of 2.50. In contrast to free fatty acids (FFAs), another form of energy-dense fuel, 25 26 BHB gives better ATP yield per oxygen consumed, is water soluble, and generates fewer reactive oxygen species (ROS).

Ketogenesis pathway. AcAc, acetoacetate; BDH, BHB dehydrogenase; CoA, coenzyme A; HMGCL, hydroxymethylglutaryl coenzyme A lyase; HMG-CoA, hydroxymethylglutaryl CoA; HMGCS2, 3-hydroxy 3-methylglutaryl-CoA synthase 2; CPT1/2, carnitine palmitoyltransferase 1/2; MCT, moncarboxylate transporter.

The kidney is among the most metabolically active organs, with very high oxygen demand and the second-highest mitochondrial density after the myocardium. 27 While oxidative metabolism is the principal source of energy in the kidney, the fuel substrates for metabolism differ across regions of the kidney. In the healthy kidney, both fatty acid oxidation (FAO) in the proximal tubules and glycolysis in the distal tubules support its metabolism. The renal cortex, especially the S1/S2 tubule segments, generates energy primarily from FFAs, lactate, and glutamate versus glucose. 28 The outer medulla uses glucose, lactate, FFAs, and ketones for energy. However, in diseased kidneys, mitochondrial dysfunction has been reported as a key pathologic feature that contributes to disease initiation and progression. 29 30 For example, in diabetic nephropathy (DN), hyperglycemia-induced flux of glycolysis increases oxygen demand with the by-product of amplified ROS. 30 31 Excess glucose use in the kidney shifts the energy reliance from fatty acid metabolism to glycolysis, even in the proximal tubules. 30 31

The renoprotective effect of SGLT2i in diabetic kidney disease is driven by amelioration of the pathologic metabolic shift from FAO to glycolysis. SGLT2i decreases reabsorption of excessive glucose, reduces energy production from glucose in the kidney, and increases fatty acid utilization in the kidney. 32 Furthermore, the glycosuric effect of SGLT2i also augments the BHB level in the kidney mainly through increased production of ketones rather than reduced kidney clearance. 33 The kidney is indeed an avid consumer of ketones. 34 BHB serves as an important alternative source of energy for the kidney during metabolic imbalance. It can be effectively metabolized in all nephron regions, except the S1/S2 proximal tubule segments. 35 During starvation or fasting, the BHB level in the kidney increases 20-fold and is used as a substrate for mitochondrial energy production. 20 In the diabetic kidney disease mouse model, both SGLT2i and exogenous ketone treatment normalized the renal ATP levels by restoring its production and this intervention was also associated with kidney function improvement. 36

Anti-inflammatory effect

Inflammation is critical in both acute kidney disease and CKD, especially through activation of inflammasomes such as NOD-, LRR-, and pyrin domain-containing 3 (NLRP3). 37 Numerous studies highlight the link between DN and NLRP3 inflammasome activation, which negatively impacts podocyte function, escalates the expression of inflammatory markers like IL-1β, and is also linked with albuminuria and tubulointerstitial injury. 38 39 Consequently, targeting NLRP3 inflammasome inhibition emerges as a promising approach for kidney disease treatment, despite concerns over the safety of current experimental drugs. 40 BHB stands out for its wide-ranging anti-inflammatory actions, including its effect on inhibiting NLRP3 inflammasome activation. 41 BHB successfully suppresses NLRP3 inflammasome activation in human monocytes and murine neutrophils in vitro and in animal models of NLRP3-mediated diseases. 42 Likewise, the anti-inflammatory effect of SGLT2i in diabetic rats, characterized by subdued NLRP3 inflammasome activation and lower interleukin (IL)-1β and tumor necrosis factor (TNF)-ɑ levels, correlates with elevated BHB and reduced insulin levels in the bloodstream. 43 BHB’s primary receptor is GPR109A (HCAR2), a G protein coupled receptor (GPCR) that acts by suppressing cyclic adenosine monophosphate (cAMP). 44 Beyond NLRP3 inflammasome suppression, animal studies reveal BHB diminishes other proinflammatory cytokines, including IL-6, chemokine (C–C motif) ligand 2, and monocyte chemoattractant protein-1, through activation of GPR109A, partially influenced by BHB’s effect on nuclear factor kappa B translocation. 45–47 In humans, ketogenic diets consistently reduce inflammation indicators. Individuals with T2D on a ketogenic diet show decreased serum C reactive protein and white cell counts, 48 along with significant reductions in 15 out of 16 inflammatory/immune modulators after 1 and 2 years. 49 This anti-inflammatory benefit aligns with prior findings that observed a greater reduction in 7 out of 14 inflammation/immune modulators with a ketogenic diet compared with a low-fat diet after 12 weeks. 50

Antifibrotic effects

The antifibrotic effect of BHB is mainly mediated through the mammalian target of rapamycin complex 1 (mTORC1) pathway. In diabetic kidney disease, mTORC1 hyperactivation is associated with kidney dysfunction and increased fibrosis. 43 In a mouse model of non-proteinuric diabetic kidney disease, SGLT2i, particularly empagliflozin conferred renal protection by increasing endogenous ketones and suppressing mTORC1 activation in the kidneys. 36 The treatment with empagliflozin mirrored the effect of exogenous ketone supplementation, where both treatments reduced kidney damage as evident through lower plasma cystatin-C levels and decreased interstitial fibrosis. 36 The renoprotective mechanism of SGLT2i hinges on the ketogenesis rate-limiting enzyme HMGCS2 highlighting ketone production’s central role in its antifibrotic effects. 36 51

Antioxidative effects

Ketones, specifically BHB, act as an important signaling molecule influencing gene expression through various regulatory pathways. BHB notably inhibits class I histone deacetylase enzyme activity in kidney tissue, enhancing the expression of genes that respond to oxidative stress, including Foxo3a and Mt2 . 52 This confers protection against oxidative stress in human kidney cells and various animal models. Studies also show that a ketogenic diet or BHB treatment can activate the major detoxification and oxidative stress nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. 53 In a spontaneous mouse model of T2D (db/db mice) treated with dapagliflozin, there was a noticeable reduction in the expression of genes related to oxidative stress compared with those treated with a standard vehicle or glimepiride. 54 This reduction is associated with increased levels of BHB and NRF2 protein expression. 54 Similarly, BHB treatment in human proximal tubular cells (HK-2) led to increased NRF2 expression and induced NRF2 nuclear translocation. 55 Furthermore, a ketogenic diet has been reported to increase the expression of other antioxidants such as NAD(P)H dehydrogenase quinone I (NQO1) and superoxide dismutase (SOD1/2), 56 and ameliorated paraquat (PQ)-induced elevated lipid peroxidation, toxicity, reduced antioxidant activity and decreased Nrf2 expression, 57 highlighting its potential therapeutic role in combating oxidative stress and tissue hypoxia.

Mitochondrial dysfunction

Mitochondrial dysfunction is another key feature of both acute kidney failure and CKD. 58 Ketogenic diet activates the expression of peroxisome proliferator-activator receptor γ (PPARγ) coactivator-1-alpha (PGC-1ɑ) 59 and silent information regulator transcript-1 (SIRT1). 60 61 PGC-1ɑ is the main transcription factor that controls the expression of genes involved in mitochondrial biogenesis and function, while SIRT1 activation protects organelle damage including the mitochondria and reduces oxidative stress. In a recent study on diabetic mice, a ketogenic diet improved mitochondrial function and capacity through its activation of PGC-1ɑ and SIRT1. 62 Further, administration of exogenous BHB was found to increase PGC-1ɑ and mitochondrial copy number in rat kidneys. 63 Human data on mitochondrial function are lacking, but we obtained skeletal muscle biopsies from physically active adults before and after a 12-week ketogenic diet and demonstrated that mitochondrial function and efficiency shifted towards fat oxidation while improving insulin sensitivity. 64

Traditional concerns of a ketogenic diet on kidney function

Common misconceptions about ketogenic diets related to kidney health include potential adverse effects on acid–base and electrolyte balance and risk for kidney stones. The next section briefly discusses the typical renal metabolic response to a ketogenic diet that maintains pH and electrolyte status. Most work in this space has been done in the context of normal kidney function, so we mention how the situation in CKD may differ.

Electrolyte and acid–base imbalance

Ketogenic diets promote a natriuretic and diuretic effect similar to that demonstrated during starvation. 65 66 This in part accounts for the typical rapid weight loss that occurs during the initiation of a ketogenic diet. If sodium intake is not commensurate with the additional loss of sodium, two deleterious outcomes are more likely to manifest: (1) Individuals may develop common signs and symptoms of hypovolemia, colloquially referred to as “Keto-Flu,” which include dizziness when standing, lethargy, and muscle spasms/cramps. (2) Counter-regulatory mechanisms are activated that include sympathetic and aldosterone stimulation that act to preserve plasma volume by increasing sodium reabsorption and a concomitant excretion of potassium and magnesium. These side effects can be eliminated with attention to proper electrolyte intake. For most individuals with normal kidney function consuming a ketogenic diet, it should be emphasized to ingest an additional 1–2 g sodium/day (4–5 g sodium/day total), a maintenance of 3–4 g/day potassium, and sufficient fluid intake.

In CKD, a decrease in viable nephrons and reduction in glomerular filtration rate (GFR) change the kidney’s normal physiology and sodium balance. 67 68 Even though an adaptive fractional increase in sodium excretion per individual nephron unit compensates for the reduced number of working nephrons, the kidney’s inability to excrete sufficient amounts of sodium results in sodium retention, extracellular fluid expansion, and blood pressure increase. 68 69 Likewise, the renin–angiotensin–aldosterone system is activated in CKD, further exacerbating sodium retention and causing vasoconstriction which could significantly raise blood pressure. 69 Sodium retention and its association with blood pressure in CKD are often referred to as “sodium-sensitive hypertension.” 70 Therefore, reducing salt intake is recommended to manage hypertension in patients with CKD. 68 The natriuretic and diuretic effect of the ketogenic diet may help alleviate sodium retention and improve systemic and glomerular blood pressure. Low carbohydrate and ketogenic diet studies often report a reduction in systolic and diastolic blood pressure 71 72 and blood pressure medication requirements. 73 However, the current recommendation of sodium intake in a WFKD is based on individuals with normal kidney function. 74 Recommendations for sodium and electrolyte intake for patients with CKD following a ketogenic diet should be individualized by a healthcare professional based on the patient’s renal function and electrolyte status. Future studies should assess the relationship between ketosis, sodium balance, and blood pressure.

Another misconception associated with ketogenic diets relates to promotion of acidosis owing to specific food items and the weakly acidifying effects of ketones, which could worsen kidney function, bone health, and kidney disease-associated endocrinopathies. 75 76 In healthy subjects provided a carefully prepared ketogenic diet with mean BHB levels >2 mM, serum bicarbonate was modestly reduced but well within normal ranges. 77 A ketogenic diet with mild ketosis (~1 mM) in individuals with normal kidney function has no significant impact on blood pH, serum bicarbonate level, and anion gap over 21 days 78 and 4 months. 79

When faced with an increased acid load, normal kidney function affords compensatory increases in ammonium excretion. In a somewhat mirrored perspective where the acid load is stable and the kidney function is reduced, the fewer working nephrons compensate with increased ammoniagenesis and excretion. This adaptation results in high intrarenal ammonia, which is thought to activate the alternative complement pathway eventually leading to tubulointerstitial fibrosis. Decreases in GFR to levels below 40–50 mL/min diminish the kidney’s ability to excrete more ammonium and overall acid 80 ; hence, metabolic acidosis is more commonly encountered at this level of disease. In patients with kidney disease, clinicians commonly monitor steady-state serum bicarbonate levels to assess overall acid load. However, decreases in serum bicarbonate are often reported at a later stage of the disease and it is considered inadequate to reflect the overall acid load. Eubicarbonatemic hydrogen ion retention among patients with earlier CKD is increasingly an area of focus 81 ; thus, studying ketogenic diet in all stages of CKD requires longer term study of acid excretion and the rate of kidney function decline.

Urinary acid excretion is favored as the gold standard for estimating acid load, and the prevailing wisdom was that an increased dietary acid load would burden the kidneys further and lead to more dysfunction. However, recent observations from the rich data collected in the Chronic Renal Insufficiency Cohort Study have demonstrated pitfalls to that simplistic view. 82 83 Among patients with diabetes, higher levels of net acid excretion were associated with a lower risk of CKD progression. These studies suggested that the changes in acid excretion were diet-independent and may be elicited by changes in energy metabolism and endogenous acid production from insulin resistance. 82 83 Currently, the effect of ketogenic diet on net acid excretion is unknown and this would be worthwhile exploring in patients with T2D and varying stages of CKD.

Kidney stones

Kidney stones, especially genetically driven stones, are associated with an increased risk of CKD. 84 A recent meta-analysis reported a pooled kidney stone incidence of 5.9% among patients on a ketogenic diet followed for a median of 3.7 years, 85 compared with a historical incidence rate of <0.3% per year in the general population. 86 Most studies reporting risk of kidney stones were in children receiving a ketogenic diet therapy for epilepsy 85 87–91 with higher incidence during long-term exposure (ie, 25% over 6 years, 91 which is complicated by concurrent use of antiseizure medications (eg, carbonic anhydrase inhibitors) and other risk factors in this population. In adults with obesity, who are at higher kidney stone risk based on their higher adiposity, 92 consuming a ketogenic diet over 2 years revealed no harmful effects on GFR, albuminuria, or fluid and electrolyte balance compared with a low-fat diet 93 ; and there was one possible, but not confirmed, case of kidney stones out of 153 subjects. 94

Uric acid stones are the most frequently reported by individuals on a ketogenic diet, followed by calcium oxalate stones or mixed stones with calcium and uric acid. 85 A ketogenic diet transiently increases uric acid concentration 25%–50%, which usually peaks at 2–4 weeks, and gradually returns to prediet levels by 8 weeks. 95–97 The initial rise in uric acid is concomitant with the rise in ketones, and it was postulated that the reason for this may be competition between uric acid and ketones for the same organic acid transporters, which are required for renal excretion. 98–101 After several weeks, the kidney conserves ketones, 102 presumably allowing for return of normal renal uric acid excretion and serum levels.

There may be effective strategies to mitigate the kidney stone risk in patients following a ketogenic diet. Increasing fluid intake to maintain dilute urine limits the possibility of mineral crystallization. 103 Urine alkalinization, particularly addressing hypocitraturia, may inhibit supersaturation of calcium salts and aggregation. 104 105 Moreover, studies of kidney stones have largely precluded patients with CKD where their urine parameters change alongside diminishing kidney function. A retrospective study of 811 patients with kidney stones noted that advancing kidney disease afforded reduced calcium stone formation, presumably due to reduced calciuria 106 and increased uric acid stone formation. 107 Metabolic acidosis resulting in acidic urine pH is common among individuals with CKD. 108 Low urine pH is a well-known risk factor for forming uric acid kidney stones due to the low solubility of uric acidic in acidic conditions. 109 110 At the same time, low urine pH leads to hypocitraturia which increases the risk of forming calcium oxalate kidney stones. 111 Hence, future examination of how a ketogenic diet impacts the incidence of kidney stones among patients with T2D and CKD is paramount. Being aware of and addressing the potential kidney stone risk with well-established measures—such as urine alkalization, correcting hypocitraturia, and increasing fluid intake—is prudent. Additionally, understanding that diet-imposed change in risk through modulation of ammonia excretion, uricosuria, calciuria, citraturia, and other urinary parameters will assist with future guidance.

Current evidence on very low or low carbohydrate diet intervention and its effect on kidney function

Evidence from animal studies.

Several rodent studies have specifically investigated the effects of a ketogenic diet on kidney function and disease. Two mouse studies reported benefit of ketogenic diet on DN, even reversing some of the key molecular features of DN. Poplawski et al assessed the effect of ketogenic diet on DN using both type 1 (Akita) and type 2 (db/db) murine diabetes models. In both models, the mice initially developed albuminuria on chow diet, and after transitioning to the ketogenic diet reversed and normalized urinary albumin/creatinine ratio (UACR) within 8 weeks. 112 Furthermore, the expression of several stress-induced genes involved in oxidative stress and toxicity was completely normalized by ketogenic diet in both models, with an observed effect that was more consistently robust in the type 1 mouse model. Likewise, histopathologic features of glomerular sclerosis were also partially reversed by the ketogenic diet in the T2D mouse model. 112 Jung et al examined db/db DN mice fed normal chow diet (dbNCD), high-fat diet (dbHFD), or ketogenic diet (dbKETO). dbKETO animals had lower UACR and blood urea nitrogen to creatinine ratio levels after 5 weeks compared with the dbNCD and dbHFD mice. 55 Histologic analysis of the kidney showed that dbKETO mice had less fibrotic changes than the dbNCD and dbHCD mice suggesting that the dbKETO mice delayed progression of DN histologic phenotypes. Furthermore, in the same report, treatment of the human proximal tubular cell line (HK-2) with BHB led to activated autophagy by increasing the LC3 I to LC3 II ratio, phosphorylation of adenosine 5 monophosphate-activated protein kinase (AMPK), beclin, p62 degradation, NRF2 expression, and decreased glucose-induced ROS levels. 55 Studies in a rat model of a genetic form of CKD, polycystic kidney disease, showed that a ketogenic diet not only slowed disease progression and preserved renal function in young animals but even partially reversed existing renal cystic disease in older animals. 18 The treatment resulted in improvement of renal fibrosis and inhibition of mTORC1 and epithelial proliferation. Remarkably, the effects could be replicated by administering BHB in the drinking water in a dose-dependent manner, without any food changes. 18 63 These results suggest that the actions of BHB may underlie most of the renoprotective mechanisms of nutritional ketosis, and that exogenous BHB can be effectively supplemented.

Evidence from clinical and observational studies

Clinical and observational studies that examined kidney function in response to low-carbohydrate diets ranging from <20 g/day to 30%–40% of energy expenditure are presented in online supplemental table 1 . Three of the six randomized controlled trials (RCTs) reported no significant changes in kidney function in the low carbohydrate arm compared with the comparison diet group. Two of the three studies followed the participants with normal baseline eGFR for 52 weeks 113 114 and the third study followed subjects with slightly lower baseline eGFR (<80) for 12 weeks. 115 Another two RCTs reported renal benefit in the low carbohydrate arm with improvements in serum creatinine, cystatin C, eGFR, and albumin. 93 116 The study by Tirosh et al reported greater eGFR improvement in those following a low-carbohydrate diet versus both a Mediterranean and low-fat diet. 116

Supplemental material

The use of surrogate markers, especially serum creatinine-derived estimates of kidney function, is less accurate at higher eGFRs and may be mischaracterized amidst dietary intervention, highlighting the importance of studying major adverse kidney events and assessing cystatin C-derived kidney function estimates. Thus far, only one RCT has reported hard kidney endpoints including all-cause mortality that compared a carbohydrate-restricted, low-iron, polyphenol enriched diet (CR-LIPE) with a standard protein restriction diet (SPRD). 117 The 191 participants in this study were followed for approximately 4 years. In this study, CR-LIPE significantly decreased doubling of serum creatinine (relative risk, 0.53, 95% CI 0.33 to 0.86, p<0.01), all-cause mortality (relative risk, 0.5, 95% CI 0.2 to 1.12) and also delayed end-stage renal disease and renal replacement therapy when compared with SPRD. 117 However, the CR-LIPE intervention was a multimodal dietary intervention that included carbohydrate restriction (35% of the energy intake) as one of the dietary modifications along with low-iron availability and polyphenol enrichment in the diet. Future study involving major adverse kidney endpoints is warranted to confirm if a ketogenic diet has beneficial impact on kidney disease.

Presumably because eGFR is less accurate at healthier function (eGFR >80 mL/min), some of these studies have shown that the beneficial effect of low carbohydrate diet is greater in those with lower starting baseline eGFR. For example, the study by Tirosh et al reported that the increase in eGFR was greater in those with CKD stage 3 (a 7.1-point; 10% eGFR increase from baseline) than the whole cohort (+5.3% increase from baseline) in the low carbohydrate arm. 116 While other studies included a range of baseline eGFRs, the subset of patients with more significant kidney dysfunction (eGFR <60 mL/min) exhibited a slower decline in function, and no deterioration was evident in participants with normal baseline eGFR. 118 119 Furthermore, caution is warranted when interpreting creatinine-derived eGFR measurements because any change in skeletal muscle mass during a nutritional study may affect the endogenous production of creatinine independent of actual changes in renal function. Hence, corroboration with cystatin-C measurements would strengthen these observations. The single-arm prospective 12 weeks study on individuals with relatively advanced diabetes nephropathy (eGFR <40 mL/min) reported statistically significant improvements in eGFR, serum creatinine, and cystatin C. 118 Three additional retrospective observational studies reported improvements in kidney function in individuals following a low carbohydrate diet 120–122 ( online supplemental table 1 ). One of these studies reported improvement in eGFR and decrease in UACR at an average follow-up of 30 months 119 while the other two studies reported eGFR improvement in individuals with reduced kidney function at baseline (eGFR <90 in one study and eGFR <70 in the other study). 121 122

In contrast, there were only two observational studies frequently cited when suggesting that a low carbohydrate diet is associated with adverse kidney outcomes. These studies did not focus on individuals adhering to a ketogenic diet or on those limiting their carbohydrate intake. For instance, Farhadnejad et al ’s 2018 study, which was a population-based prospective analysis, investigated the association between different tertiles of low carbohydrate high protein (LCHP) scores and the incidence of CKD. 123 Notably, none of the LCHP score tertiles in the study indicated a carbohydrate-restricted diet. Even in the tertile with the lowest LCHP score, carbohydrates contributed to 51.0% of the total energy, resembling the carbohydrate profile of a standard Western diet where 40%–60% of energy typically comes from carbohydrates. The other retrospective observational study by Li et al reported an association between elevated fasting ketone level with abnormal renal function 124 in people with T2D who were admitted to the hospital, and who were not specifically eating a ketogenic diet. The association of ketones and renal function in this study is not relevant to dietary carbohydrate restriction in an ambulatory population.

Altogether, these clinical and observational studies show no harm from low or very low carbohydrate diets for people with diabetes in the setting of normal renal function, and a possible beneficial effect in the setting of moderately reduced renal function. The kidney function improvement observed in these studies may be an ancillary outcome associated with other improvements seen in these interventions including weight loss, glycemic control, or blood pressure improvement. Interestingly, Unwin et al reported no association between observed kidney function improvement with the magnitude of weight loss, improvement in blood pressure and HbA1c, 120 while another study reported that the increase in eGFR was significantly associated with a decrease in fasting insulin and systolic blood pressure but not with the level of weight loss and protein intake in the intervention. 116 In our previous study on patients with T2D following a very low carbohydrate intervention, there was a marginally significant increase in eGFR at 1 year. 72 A post hoc analysis of these data revealed that a higher mean BHB at 1 year (β=5.04, p=0.005) was significantly associated with a greater increase in mean eGFR (unpublished data). Furthermore, in a subgroup analysis of 22 trial participants with an eGFR <60 mL/min/1.73 m 2 at baseline who remained in the study for 2 years, 72 the mean eGFR progressively increased from 51 mL/min/1.73 m 2 to 60, 63, and 68 mL/min/1.73 m 2 at 10 weeks, 1 year, and 2 years (unpublished data). Notably, the majority of the 22 participants reverted to stage 2 and no one progressed to stage 4 CKD. Evidently, a dose-dependent association exists between ketosis trajectory classes and the increase in total eGFR slope at 2 years. 125 Participants with higher endogenous ketone concentration and longer duration of ketosis maintenance exhibited the greatest rise in the 2-year eGFR slope compared with those with lower endogenous ketone concentration and unsustainable ketosis maintenance. 125 Hence, available evidence suggests that carbohydrate restriction and ketosis afford benefits to kidney function. It will be important to determine in future trials whether the improvement in kidney function translates to a sustained long-term reduction, or even reversal, in the progression of kidney disease.

Evidence from meta-analyses, systematic and narrative review

A recent review discussing the potential negative effect of purported ketogenic diets on kidney health focused on observational studies that compared low protein versus high protein diets that were not ketogenic or low carbohydrate diets, 126 and raised concern about the association of albuminuria with high animal fat but only referred to observational studies that assessed high animal fat intake in the context of a Western diet 126 negating the relevance of the studies cited for concern.

In contrast, systematic reviews and meta-analyses that assessed pooled effects of RCTs reported beneficial effects of low-carbohydrate diets. The meta-analysis by Oyabu et al evaluated nine RCTs with 861 participants in the low carbohydrate arm and 826 participants in the control group. 127 Despite a large variation in the proportion of carbohydrate intake from 4% to 45% in the low carbohydrate arm of the nine studies with a study duration ranging from 6 to 24 months, the review revealed that there was a significant increase in eGFR in the low carbohydrate group versus control group. 127 Another meta-analysis with 12 RCTs that only included patients with T2D reported no significant difference in the pooled eGFR and creatinine mean estimate between the lower carbohydrate diets (14%–45% of calories from carbohydrate) versus control diets over 5 weeks to 24 months. 128 Similarly, another meta-analysis that included five RCTs with the low carbohydrate arm had carbohydrate intake <45%, and the studies ranging from 5 weeks to 24 months reported no difference in the pooled eGFR estimate between the control and low carbohydrate diets. 129 The current evidence from systematic reviews and meta-analyses with a range of carbohydrate intake suggests that carbohydrate restriction is not associated with adverse effects on kidney function, or in some cases might be beneficial.

Evidence from genetically driven kidney disease

Individuals with autosomal dominant polycystic kidney disease (ADPKD) may benefit from calorie restriction or ketogenic diet. 19 130 131 This chronic progressive condition is characterized by hyperproliferation, inflammation, fibrosis, and cyst growth, leading to deterioration of kidney function over time. 19 132 mTOR is one of the main signaling pathways activated in ADPKD. 132 A study of various polycystic kidney disease animal models showed that time-restricted feeding, administration of a ketogenic diet, or supplementation with exogenous BHB prevented kidney cyst disease progression by inhibiting cell proliferation, fibrosis, and cyst growth. 18 63 Furthermore, the mTOR activity was inhibited in these animal models suggesting that blunting the signaling pathway inhibits cell proliferation, growth, and fibrosis in ADPKD. 18 63 133 In humans, a retrospective observational study of ADPKD patients who self-initiated ketosis either using ketogenic or time-restricted diets reported improvement in eGFR after 6 months. 134 A pilot study on 24 patients with ADPKD demonstrated the feasibility of the ketogenic diet, reporting high adherence rates and improvements in blood pressure, eGFR, and kidney pain. 130 In another exploratory RCT, 66 participants with ADPKD were randomized to ketogenic, water fasting, or control diets. The study confirmed the feasibility of the therapy in the ketogenic arm (KD) and revealed significant improvements in eGFR, including both creatinine and cystatin C-derived eGFR in the KD group but no improvements were observed in the water fasting and control diets. 131 Additionally, there were no significant differences in UACR and blood pressure among the three diets. 131

Perspectives and future direction

There is a considerable body of research suggesting that a very low carbohydrate ketogenic diet is safe in individuals with moderately diminished kidney function, even in studies that had higher protein intake than what is recommended for kidney disease and diets that are not plant-based. The diet can be safely prescribed in patients with T2D for treating and remitting diabetes even if they have underlying stage 2 or 3 CKD or reduced kidney function. Beyond safety, mechanistic plausibility, preclinical data, and even some RCT studies suggest that carbohydrate-restricted diets may be beneficial in improving moderate kidney dysfunction and in reducing progression of CKD. The preliminary proof of concept from small and short duration studies in humans and animals suggests a very low carbohydrate diet could be an effective dietary intervention for patients with CKD. Furthermore, there are predeveloped ketogenic nutritional options to consider when we plan a future trial to assess the impact of ketogenic diet on patients with CKD, such as the recently developed program for treating ADPKD known as Ren.Nu. This program is a plant-focused ketogenic medical nutrition therapy, designed to avoid renal stressors like oxalate, inorganic phosphate, and purines/uric acid. It includes a medical food formulation, KetoCitra, containing BHB with alkaline citrate which helps antagonize kidney stone formation. 129 130 Based on the findings from these different studies and currently available ketogenic medical therapy specific for kidney disease, there is a need for future larger and longer follow-up randomized controlled clinical trials on very low carbohydrate diet, including nutritional ketosis in patients with CKD with or without T2D on kidney hard endpoints including major adverse kidney events (a composite event of death, persistent renal decline >25% decline in eGFR, and a new initiation of dialysis) and other kidney-related outcomes to firmly establish the long-term effectiveness. For example, a head-to-head comparison of the safety and efficacy of ketogenic nutritional therapy versus SGLT2i pharmacologic intervention (that involves the same mechanism of raising ketone levels) could be of high interest. Weight loss from the diet can improve filtration and albuminuria. Thus, including other surrogate endpoints like eGFR slope and microalbuminuria in these studies have the potential to elucidate the degree to which weight loss and blood pressure improvement from the diet affects kidney function markers and also to explore if ketone levels independently have an impact on these markers and endpoints. Furthermore, these studies should also assess the diet’s overall safety in patients with T2D and CKD, specifically exploring its effect on net acid excretion, kidney stone formation, and maybe its beneficial effect on sodium retention hypertension. Finally, another important consideration in the clinical trial design for evaluating the efficacy of a very low carbohydrate diet in patients with CKD is understanding the diet’s additive role, especially how the diet interacts with currently available treatment drugs for patients with CKD including renin–angiotensin system blockade (angiotensin-converting enzyme inhibitor, ACEi and angiotensin receptor blockers, ARBs), SGLT2i, glucagon-like peptide-1 receptor agonists (GLP1-RA), and the non-steroidal mineralocorticoid receptor antagonists (finerenone).

Ethics statements

Patient consent for publication.

Not applicable.

Ethics approval

  • Dean BB , et al
  • ElSayed NA ,
  • Aroda VR , et al
  • Park S , et al
  • Esmeijer K ,
  • Geleijnse JM ,
  • de Fijter JW , et al
  • Guyton JR , et al
  • Seshadri P , et al
  • Westman EC ,
  • Mavropoulos JC , et al
  • Cuenca-Sánchez M ,
  • Navas-Carrillo D ,
  • Orenes-Piñero E
  • Ikizler TA ,
  • Burrowes JD ,
  • Byham-Gray LD , et al
  • Kidney Disease: Improving Global Outcomes (KDIGO) Diabetes Work Group
  • Beck GJ , et al
  • Kopple JD ,
  • Wang X , et al
  • Laville M ,
  • Boissel JP , et al
  • Wen Y , et al
  • Sugahara S , et al
  • Torres JA ,
  • Kruger SL ,
  • Broderick C , et al
  • Saville J ,
  • Kalantar-Zadeh K
  • Balasse EO ,
  • Puchalska P ,
  • Crawford PA
  • Röhling M , et al
  • Mudaliar S ,
  • Bosy-Westphal A , et al
  • Baverel G ,
  • Ferrier B ,
  • Forbes JM ,
  • Thorburn DR
  • Qiao Y , et al
  • Audzeyenka I ,
  • Bierżyńska A ,
  • Fang Y , et al
  • Ferrannini E ,
  • Frascerra S , et al
  • Cuenoud B ,
  • Hartweg M ,
  • Godin J-P , et al
  • McDonough AA ,
  • Shahzad K ,
  • Dong W , et al
  • Qiu J , et al
  • Liu C , et al
  • Nguyen KY ,
  • Grant RW , et al
  • Goldberg EL ,
  • Molony RD , et al
  • Kim SH , et al
  • Spigoni V ,
  • Cinquegrani G ,
  • Iannozzi NT , et al
  • Gambhir D ,
  • Veeranan-Karmegam R , et al
  • Wang J-F , et al
  • Wang JF , et al
  • Bhanpuri NH ,
  • Hallberg SJ ,
  • Williams PT , et al
  • Phinney S ,
  • Athinarayanan S , et al
  • Forsythe CE ,
  • Phinney SD ,
  • Fernandez ML , et al
  • Kogot-Levin A ,
  • Riahi Y , et al
  • Shimazu T ,
  • Hirschey MD ,
  • Newman J , et al
  • Milder JB ,
  • Kim YJ , et al
  • Hovda DA , et al
  • Liu F , et al
  • Pathak SJ ,
  • McCarty MF ,
  • DiNicolantonio JJ ,
  • Scheibye-Knudsen M ,
  • Mitchell SJ ,
  • Fang EF , et al
  • Roberts MN ,
  • Wallace MA ,
  • Tomilov AA , et al
  • Holznecht N ,
  • Asplund DA , et al
  • Miller VJ ,
  • LaFountain RA ,
  • Barnhart E , et al
  • Palmer BF ,
  • Saudek CD ,
  • Boulter PR ,
  • Borrelli S ,
  • Provenzano M ,
  • Gagliardi I , et al
  • Cuevas CA ,
  • Zietse R , et al
  • Murray SW , et al
  • McKenzie AL ,
  • Athinarayanan SJ ,
  • McKenzie AL , et al
  • Dhondup T ,
  • Bistrian BR ,
  • Wolfe RR , et al
  • Sharma AP ,
  • Ross ML , et al
  • Gomez-Arbelaez D ,
  • Crujeiras AB ,
  • Castro AI , et al
  • Pourafshar N ,
  • Pourafshar S ,
  • Soleimani M
  • Scialla JJ ,
  • Dobre M , et al
  • Luciano A ,
  • Pendergast J , et al
  • Bergstralh EJ ,
  • Melton LJ , et al
  • Acharya P ,
  • Acharya C ,
  • Thongprayoon C , et al
  • Pyzik PL , et al
  • Kossoff EH ,
  • Furth SL , et al
  • Herzberg GZ ,
  • Fivush BA ,
  • Kinsman SL , et al
  • Sampath A ,
  • Groesbeck DK ,
  • Taylor EN ,
  • Stampfer MJ ,
  • Friedman AN ,
  • Foster GD , et al
  • Foster GD ,
  • Hill JO , et al
  • Horton ES ,
  • Sims EAH , et al
  • García-Nieto VM ,
  • Claverie-Martín F ,
  • Moraleda-Mesa T , et al
  • Goldfinger S ,
  • Klinenberg E ,
  • Seegmiller JE
  • Lecocq FR ,
  • Agarwal N ,
  • Farooq O , et al
  • McNally MA ,
  • Rubenstein JE , et al
  • Zupec-Kania BA ,
  • Amark PE , et al
  • Cirillo M ,
  • Bilancio G ,
  • Cavallo P , et al
  • Simmons K ,
  • Phadke M , et al
  • Reaven NL , et al
  • Pazos Pérez F
  • Haghighatdoost F ,
  • Sadeghian R ,
  • Clark CCT , et al
  • Poplawski MM ,
  • Mastaitis JW ,
  • Isoda F , et al
  • Thompson CH ,
  • Luscombe-Marsh ND , et al
  • Brinkworth GD ,
  • Buckley JD ,
  • Noakes M , et al
  • Zainordin NA ,
  • Eddy Warman NA ,
  • Mohamad AF , et al
  • Harman-Boehm I , et al
  • Facchini FS ,
  • Chambers M ,
  • Kamendulis LM , et al
  • Tuccinardi D ,
  • Tozzi R , et al
  • Crocombe D , et al
  • Mitchell NS ,
  • Wilmsen N ,
  • Geerlings W , et al
  • Farhadnejad H ,
  • Asghari G ,
  • Emamat H , et al
  • Shen X , et al
  • Roberts CGP ,
  • Adams RN , et al
  • Joshi S , et al
  • Hashimoto Y ,
  • Fukuda T , et al
  • Kingaard JJ ,
  • Munits M , et al
  • Cukoski S ,
  • Lindemann CH ,
  • Arjune S , et al
  • Shillingford JM ,
  • Murcia NS ,
  • Larson CH , et al
  • Torres JA , et al

Supplementary materials

Supplementary data.

This web only file has been produced by the BMJ Publishing Group from an electronic file supplied by the author(s) and has not been edited for content.

  • Data supplement 1

Contributors SJA, JV, and CGPR conceptualized the review topic and formulated the objectives; SJA conducted the comprehensive literature search, synthesized and interpreted the data from the collected literature; SJA drafted the original manuscript; JSV, TW, CGPR, and CV provided critical revisions and edits to the manuscript; ALM and GKS reviewed and edited the manuscript; All authors have read and agreed to the final version of the manuscript.

Funding The authors have not declared a specific grant for this research from any funding agency in the public, commercial or not-for-profit sectors.

Competing interests TW is an inventor on issued and pending patents filed by the University of California, Santa Barbara related to the topic of this article. TW is a founder and shareholder of Santa Barbara Nutrients, Inc., holds a managerial position, and has contributed to the development of the Ren.Nu ketogenic dietary program and the medical food KetoCitra. TW received speaker fees from Otsuka, was a scientific advisor of Chinook Therapeutics, and received research funding from Chinook Therapeutics. JSV is a cofounder and shareholder of Virta Health, serves as a science advisor for Simply Good Foods and Nutrishus Brands, and has authored books on ketogenic diets. SJA, CGPR, and GKS are employees and shareholders of Virta Health. ALM is a shareholder of Virta Health.

Provenance and peer review Not commissioned; externally peer reviewed.

Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.

Read the full text or download the PDF:

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Front Endocrinol (Lausanne)

Ketogenic Diets and Cardio-Metabolic Diseases

Weiyue zhang.

1 Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China

2 Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China

3 Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China

Chaodong Wu

4 Department of Nutrition, Texas A&M University, College Station, TX, United States

While the prevalence of cardio-metabolic diseases (CMDs) has become a worldwide epidemic, much attention is paid to managing CMDs effectively. A ketogenic diet (KD) constitutes a high-fat and low-carbohydrate diet with appropriate protein content and calories. KD has drawn the interests of clinicians and scientists regarding its application in the management of metabolic diseases and related disorders; thus, the current review aimed to examine the evidences surrounding KD and the CMDs to draw the clinical implications. Overall, KD appears to play a significant role in the therapy of various CMDs, which is manifested by the effects of KDs on cardio-metabolic outcomes. KD therapy is generally promising in obesity, heart failure, and hypertension, though different voices still exist. In diabetes and dyslipidemia, the performance of KD remains controversial. As for cardiovascular complications of metabolic diseases, current evidence suggests that KD is generally protective to obese related cardiovascular disease (CVD), while remaining contradictory to diabetes and other metabolic disorder related CVDs. Various factors might account for the controversies, including genetic background, duration of therapy, food composition, quality, and sources of KDs. Therefore, it’s crucial to perform more rigorous researches to focus on clinical safety and appropriate treatment duration and plan of KDs.

Introduction

Cardio-metabolic diseases (CMDs) have become a worldwide epidemic, as demonstrated by an increased prevalence of obesity, diabetes mellitus (DM), metabolic syndrome, cardiovascular disease (CVD), and chronic kidney disease (CKD), and culpable to a significant global financial burden ( 1 ). CVDs comprise a wide range of diseases detrimental to cardiac and vascular function ( 2 , 3 ). To decrease cardiovascular (CV) mortality and related economic burden, it’s important to reduce CV risk factors and employ appropriate therapy in developed countries ( 4 ). Various established risk factors such as age, gender, genetic heritage, smoking, high blood pressure, poor eating habits, type 2 diabetes mellitus, dyslipidemia and obesity had been accounted for the development and progression of cardiovascular diseases (CVD).

Dietary factors that profoundly influence human health are linked to cardiovascular disease and other chronic metabolic conditions such as obesity and type 2 diabetes ( 5 ); thus, dietary interventions have become an essential component in managing cardiovascular risks ( 6 ).

A ketogenic diet (KD) is a high-fat, low-carbohydrate diet with appropriate protein content and calories ( 7 ). A traditional KD consists of a 4:1 ratio of fats to carbohydrates and protein, with 90% of the calories from fat, 8% from protein, and only 2% from carbohydrate ( 8 ). In recent years, to improve compliance and imitate the effects of classic KD, alternative protocols with different formulations of KD have been proposed ( 9 ), including 3:1 KD, 2:1 KD, 1:1 KD, the modified Atkins diet (MAD), the medium-chain triglyceride ketogenic diet (MCTKD), the low glycemic index treatment (LGIT) ( 10 , 11 ) ( Table 1 ). With the implementation of KD therapy, a drastic decrease in dietary carbohydrates reduces glucose utilization. In the human body, KD treatment could imitate the metabolic changes of fasting. In addition, some of the beneficial effects of KDs could be attributable to the production of ketones, e.g., β-hydroxybutyrate (BHB), acetoacetate, and acetone in the liver ( 12 ).

Table 1

Formulations of common ketogenic diets (KDs).

DietPercent Total Daily Energy Intake
Fat %Carbohydrate %Proteins % (g)
Classic KD (4:1 KD)9028
3:1 KD8749
2:1 KD82810
1:1 KD701020
MAD60-655-1030
MCTKD70-7515-1910
LGIT601030

MAD, the modified Atkins diet; MCTKD, the medium chain triglyceride ketogenic diet; LGIT, the low glycemic index treatment.

KD was firstly used as a dietary treatment for epilepsy in the 1920s ( 8 ). However, with the progress in antiepileptic drugs (AEDs) development and application, the clinical use of KDs in epilepsy has dramatically decreased. Interestingly, about one-third of patients receiving epilepsy treatment couldn’t gain significant relief from the disease, and the KD regained scientists’ attention and became a choice for application in drug-resistant or difficult-to-treat epilepsies ( 13 , 14 ). Apart from neurological diseases, KD has recently shown promising efficacy in a wide variety of diseases, including various cancers and metabolic diseases. Ovarian cancer, for instance, may reveal significantly better clinical outcomes under KD intervention, as revealed by a systematic review of randomized controlled trials ( 15 ). Furthermore, KD intervention has been found to inhibit tumor progression or mitigate cachexia symptoms ( 16 ).

KD has drawn more interest and gradually become an elective dietary intervention choice for CMDs ( 17 ). Moreover, it is significantly effective in mitigating various metabolic diseases, including obesity ( 18 , 19 ), glucose transporter type 1 deficiency syndrome (GLUT1DS) ( 20 ), and pyruvate dehydrogenase (PDH) deficiency ( 21 ). Meanwhile, due to the uncertainty of dietary interventions, different voices have also occurred regarding the safety issues and drawbacks of employing KD. The clarity on how KD influences cardiovascular and metabolic diseases remains unclear. Therefore, the current review highlighted pertinent information concerning KD and CMDs ( Figure 1 ). Qualified studies reflecting the advantages or disadvantages of KD in CMDs were all equally considered and incorporated without bias.

An external file that holds a picture, illustration, etc.
Object name is fendo-12-753039-g001.jpg

Graphic abstract.

Effects of Ketogenic Diets on Metabolic Diseases

Various metabolic diseases have been recognized as cardiovascular risk factors, including diabetes mellitus, obesity and other metabolic diseases ( 22 ). The disrupted glucose and lipid metabolism lead to abnormal oxidative stress, inflammatory, vasoactive factors, cardiac and vascular function, and finally elevate the risk of CVD ( 23 ). KD can regulate metabolic profiles and may consequently regulate the risk of CVD.

As for the management of obesity, most studies indicated that KDs were efficient at weight loss ( 24 , 25 ), especially in reducing food intake in humans and elevating energy consumption in animals ( 18 , 19 ). Studies concerning the effects of KDs on body composition changes reported that KD-fed mice had an increased fat mass percentage than regular-chow fed mice ( 26 , 27 ), and reduced ( 28 ) or no differences ( 26 ) in the percentage of lean body mass between diets; whereas in humans, weight loss affects both fat and lean mass ( 29 ). Obesity is commonly connected with insulin resistance and type 2 diabetes mellitus (T2DM), in which systemic ketone body metabolism is perturbed ( 30 , 31 ). As such, the weight-loss effect of KDs is expected to be beneficial for diabetes; although it remains disputable that KD induces insulin resistance. There are studies indicating that KD led to ameliorated glucose homeostasis and reduced antidiabetic medications in T2DM subjects ( 32 , 33 ), even with reduced baseline insulin levels and elevated insulin sensitivity in diabetic rats ( 34 , 35 ). Moreover, the study by Farrés et al. ( 33 ) appeared to offer a plausible explanation of how KDs bring about the anti-diabetic effect, which might be attributable to the anti-inflammatory effect of KD itself and beneficial effects of the altered lipid metabolism on diabetes effector proteins. However, certain studies suggested that KD reduced glucose and insulin levels while inducing insulin resistance and glucose intolerance in rats ( 27 , 36 ). Besides, utilizing KD in adults with type 1 diabetes mellitus (T1DM) is associated with dyslipidaemia and a high number of hypoglycaemic episodes apart from excellent HbA1c levels and little glycaemic variability ( 37 ). Thus, in T1DM, the safety issues are considerable; while in T2DM, more studies are needed to address how KD might impact on insulin resistance and other aspects.

In Polycystic ovarian syndrome (PCOS), KD appears to be a valuable non-pharmacological treatment. A 24-week low-carbohydrate KD ( 38 ) and a 12-week ketogenic Mediterranean diet with phyoextracts ( 39 ) were both reported to lead to remarkable improvement in body weight, percentages of free testosterone, LH/FSH ratios, and insulin levels in women with PCOS and obesity/overweight. Besides, KD has also been proven effective in other metabolic diseases including glucose transporter type 1 deficiency syndrome (GLUT1DS) ( 20 ), pyruvate dehydrogenase (PDH) deficiency ( 40 ), phosphofructokinase (PFK) deficiency and glycogenosis type V (McArdle disease) ( 41 ).

In summary, KDs are recommended in some inherited metabolic diseases and PCOS, while the effects of KDs on diabetes and some other metabolic diseases remain controversial. Rigorously-designed long-term studies are warranted to evaluate the effects and the safety problems of KDs and further evaluate whether the impact of KDs can be maintained.

Cardiometabolic Effects of Ketogenic Diets

The occurrence and development of CMDs are closely related to systemic chronic low-grade inflammation characterized by the continuous increase of circulatory inflammatory factors ( 42 ). Dietary pattern is one of the important factors that affect chronic inflammatory states ( 43 ). Thus, the effects of dietary pattern on CMDs arouse scientists’ interest and a large number of studies have focused on the cardiometabolic effects of KD. Apart from the above-mentioned metabolic diseases, the effects of KD on cardiac function, hypertension, vascular function and lipid profile have also been studied.

Ketogenic Diets Regulate Cardiac Function

The effects of KD on cardiac health have been widely investigated, but researches concerning the effects of KD on cardiac functions provided a few relatively controversial data. Studies generally suggest that KD intake benefited cardiac metabolic efficiency and acted as a cardioprotective antioxidant. Selvaraj et al. ( 44 ) reviewed current evidence surrounding the use of therapeutic ketosis including KD in heart failure (HF) and pointed out its potential benefit in HF, particularly in HF with reduced ejection fraction. Further, Balietti et al. ( 45 ) found that an 8-week supplementation of medium-chain triglycerides KD (MCT-KD) to late-adult rats partly restored age-related decrease of succinic dehydrogenase (SDH) activity and metabolically active mitochondria, which might offset senescent alterations leading to apoptosis-induced myocardial atrophy and failure. Another study with a similar conclusion indicated that a 19-week low carbohydrate KD following global ischemic injury significantly increased the numbers of mitochondria in cardiac muscles and the reperfusion recovery of coronary flow ( 46 ). As such, the two studies demonstrated that KD was cardio-protective in terms of regulating cardiac energy metabolism including mitochondrial capability. However, some studies suggested that KD might be just not harmful to cardiac functions. A study utilizing KD for at least 12 months on cardiac functions in intractable epilepsy patients suggested that the KD used appeared to have no negative impact on ventricular functions in epileptic children in the midterm ( 47 ). Similarly, a 6-month KD therapy didn’t affect electrocardiogram outcomes in the drug-resistant children with epilepsy ( 48 ). The subjects in these two studies are both epileptic children, which cannot represent all the patients who might use KD therapy. Thus, we can still stay optimistic about the effects of KD on cardiac functions.

Studies have also been conducted concerning the mechanism of how KD might affect cardiac health. Abnormal substrate metabolism is one of the major changes of insulin resistance and diabetic myocardium ( 49 ). Given this, changes in the regulation of myocardial ketone body metabolism appear to be a novel diagnostic biomarker of altered ketolytic capacity. Wentz et al. ( 50 ) utilized ketogenic nutritional mouse models (24 h of fasting and a very low carbohydrate ketogenic diet) to demonstrate that cardiac muscle engages a metabolic response that limits ketone body utilization. Specifically, the results revealed that unmetabolized substrate concentrations were higher within the hearts of ketogenic diet-fed mice. Furthermore, a recent study suggested that a KD or a high-fat diet could reverse the structural, metabolic and functional remodeling of non-stressed cMPC1-/- (cardiomyocyte-restricted deletion of subunit 1 of mitochondrial pyruvate carrier) mouse hearts ( 51 ). A KD of 3 weeks before transverse aortic constriction was already enough to rescue cMPC1-/- hearts from rapid decompensation and early mortality after pressure overload. Another study also indicated that a high-fat, low-carbohydrate KD could completely reverse progressively developed cardiac dilation and contractile dysfunction in mice with cardiac-specific deletion of Mpc2 (CS-MPC2-/-) ( 52 ). Accordingly, KD therapy might be promising in improving cardiac fat metabolism to prevent or reverse cardiac dysfunction and remodeling in MPC deficiency.

As mentioned above, KDs are generally cardioprotective, which might be attributable to the effects of KDs on cardiac metabolism, such as ketone body metabolism and energy metabolism including mitochondrial capability ( Figure 2 ). Despite the evidence supporting the cardioprotective effect of KDs, another study utilizing KD on cardiac remodeling in spontaneously hypertensive rats (SHRs) suggested that KD might deteriorate cardiac remodeling in the hypertensive heart and warranted fully evaluation of its reliability before clinical use ( 53 ). The different pathogenesis backgrounds of hypertension might account for the different results. More studies with larger samples, longer follow-up duration, and standardized basic health status can be conducted to further clarify the role of KDs in cardiac functions and other potential mechanisms.

An external file that holds a picture, illustration, etc.
Object name is fendo-12-753039-g002.jpg

The role and mechanism of ketogenic diets in cardiac function and vascular function. Various pathways might underly the effects of ketogenic diets in cardiomyocytes and endothelial cells in different models. In cardiomyocytes, βHB regulates PI3K/Akt pathway and SDH in mitochondria to finally ameliorate cell apoptosis. However, elevated βHB might also acts through inhibiting HDAC2 and influencing mitochondrial biogenesis, leading to myocardial fibrosis. In endothelial cells, ketone bodies can functions through inhibiting mTOR pathway to regulate the level of eNOS, subsequently dilating blood vessels and enhancing vascular function. Besides, elevated ketone bodies can give rise to mild oxidative/electrophilic stress, activate Nrf2 in cytoplasm and enhance antioxidant gene expression, which lead to lowered ROS level and improved vascular functions. Created with BioRender.com .

Ketogenic Diets and Hypertension

Attention has also been paid to the effects of KD in hypertension. Most studies showed positive effects of KD in hypertension. Castellana et al. ( 54 ) suggested that very-low-calorie ketogenic diet (VLCKD) manifested improvements in hypertension, type 2 diabetes and dyslipidemia, apart from being a promising lifestyle intervention for overweight and obesity. Another study incorporating 377 patients across Italy drew a similar conclusion that VLCKD could significantly lower SBP in three months ( 55 ). Even a short-term 4-week KD with micronutrient supplementation could result in improved hypertension control and in a reduction for the usage of hypertension medications in patients with preoperative T2DM and hypertension ( 56 ). Increasing ketone bodies by nutritional interventions of ketone bodies or their precursors, such as 1,3-Butanediol, was also reported to attenuate hypertension ( 57 ). However, Guo et al. ( 58 ) revealed that subjecting spontaneously hypertensive rats (SHRs) to KD for 4 weeks aggravated hypertension, increased the expression of IL1-β and TNF-α, impaired endothelium-dependent relaxation and decreased CD31 and eNOS expression in mesenteric arteries. This finding is opposite to the previous results; thus, it remind us to be cautious in treating hypertension with KD, and perform more studies to explore the effects of KD on hypertension in human studies and animal models.

The Dietary Approaches to Stop Hypertension (DASH) diet is a classic dietary approach that has been endorsed for patients with elevated blood pressure (BP). Besides, the addition of exercise and weight loss to the DASH diet resulted in even larger BP reductions, greater improvements in vascular and reduced left ventricular mass for obese people with elevated BP ( 59 ). The main drawback of DASH might be the difficulty in long-term adherence to this diet. Considering the different components of DASH and KD, KD might become another choice for those people who love a high-fat diet, although we suggest cautious application since the antihypertensive efficacy and side effects of KD under the background of hypertension remain unclear. Interestingly, a review suggested that intermittent fasting could lower both systolic and diastolic blood pressure in human studies and animal studies, possibly through reducing oxidative stress, syncing with circadian rhythm, and inducing a ketogenic state ( 60 ). The less consumption of fats is assumed as the reason why intermittent fasting appears to be more beneficial than KD in treating hypertension. Thus, the type of fats consumed in KD therapy is crucial to be considered both in treating hypertension and evaluating its effects.

Ketogenic Diets Regulate Vascular Functions and Vascular Blood Flow

A study by Keogh et al. ( 61 ) has indicated that a very-low-carbohydrate, high-saturated-fat weight-loss diet did not impair FMD. How would the actual KD impact on vascular function and vascular blood flow?

In some studies, KD appears to play a protective role in vascular functions. Ischemic tolerance can reduce brain injury and neurological dysfunction after brain ischemia. Additional to the cardiovascular effects such as higher reperfusion recovery of coronary flow, KD also can enhance brain vascular function. As supported by the results from a study upon feeding a KD to young healthy mice, KD intervention enhanced neurovascular function through reducing mTOR protein expression and increasing eNOS levels ( 62 ). Yang et al. ( 63 ) discovered that feeding mice with KD-fed mice could remarkably decrease infarct volume and elevate regional cerebral blood flow in both ischemic and reperfusion phases. Besides, while investigating the effects of KB level on HMEC-1 endothelial cells, one study indicated that KB activated transcription factor Nrf2 and elevated the expression of cell antioxidant defending genes via inducing moderate oxidative stress ( 64 ). Thus, the increased KB level by KD might also lead to these protective effects.

However, as for big vessels such as carotid and aortic artery, the effects of KD remain controversial. For instance, after observing the effect of KD on the vascular structure and functions for at least one year, it was found that KD notably elevated the serum levels of lipids but didn’t significantly affect carotid intima-media thickness, aortic and carotid strain, the stiffness index, distensibility, and elastic modulus ( 65 ). Another study by Doksoz et al. ( 66 ) also demonstrated that a 6-month KD didn’t affect carotid intima-media thickness and elastic properties of the carotid artery and the aorta. In contrast, in the research of Coppola et al. ( 67 ), participants prescribed with KD had higher arterial stiffness parameters, including AIx and beta-index and higher serum levels of cholesterol or triglycerides. Another study revealed that a high-fat KD notably elevated atherogenic apolipoprotein B (apoB)-containing lipoproteins and decreased antiatherogenic HDL cholesterol and urged further researches to investigate whether this diet deteriorates endothelial function and facilitates inflammation and formation of atherosclerotic lesions ( 68 ). However, a clinical study involving 26 children after one year and 13 children after two years of KD suggested that the initial influences on arterial function observed within the first year of KD-treatment were reversible and were no longer significant after 2 years of the therapy ( 69 ). Therefore, the effects of KD on big vessels such as carotid and aortic artery were reversible and were no longer significant after 1-2 years, which might explain the above results.

Ketogenic Diets Regulate Serum Lipids

Apart from the cardiometabolic effects mentioned above, the impact of KD on serum CVD biomarkers has also been investigated. Research on 20 normal-weight, normolipidemic men indicated that a 6-week KD notably decreased fasting serum triglyceride, postprandial lipemia, and fasting serum insulin concentrations, tended to increase HDL cholesterol, while not affecting fasting serum total and LDL cholesterol and oxidized LDL ( 70 ). These results revealed that short-term KD would not deteriorate CVD risk profile and, indeed, appeared to ameliorate lipid disorders that are characteristics of atherogenic dyslipidemia. Another research also indicated that changes in the ratio of protein to carbohydrate toward higher protein proportion could provide beneficial effects on serum lipids apart from lowering body weight ( 71 ).

However, Özdemir et al. ( 65 ) pointed out that prescribing patients with at least 12 months KD could significantly elevate serum total and LDL cholesterol and triglyceride at a median of 12.6 months while not affecting HDL level. Moreover, another research ( 72 ) found that a 6-month KD could notably increase median triglyceride, total cholesterol, LDL, and HDL. They suggested that classic KD was indeed efficient in treating refractory seizures in children but might give rise to hypercholesterolemia and hypertriglyceridemia.

As such, disputable voices concerning the impact of KDs on serum lipids remain to be settled by future work in this field.

Ketogenic Diets and Cardiovascular Complications of Metabolic Diseases

The potential effects of KDs on the prevention or treatment of cardiovascular risk factors or diseases have been significantly studied over the past decades. Moreover, various animal and human studies have investigated the role of KDs in regulating cardiovascular complications of obesity, insulin resistance and type 2 diabetes, dyslipidemia, NAFLD, and/or GLUT1DS. However, whether and how KDs influence the cardiovascular risk factors or complications in metabolic diseases remains undetermined.

Obesity-Associated Cardiovascular Disease

Obesity is closely related to CVD, and complications of CVD are often witnessed in obese patients. Cicero et al. ( 55 ) evaluated the effect of a very low carbohydrate ketogenic diet (VLCKD) on overweight-related risk factors of CVD such as blood pressure, lipid levels, and glucose metabolism, and the study found that VLCKD intervention for 3 months was generally safe and found effective in inducing weight loss and improved CV risk factors levels.

Another study recruited a hundred obese patients and prescribed them a ketogenic diet for over six months showed significant improvement in patients’ cardiovascular status in addition to weight reduction ( 73 ). Moreover, a meta-analysis study by Bueno et al. ( 74 ) assessed the long-term effects of VLCKD on body weight and cardiovascular risk factors. The results indicated that under VLCKD, the participants had a significant reduction in body weight, TAG, and diastolic blood pressure, while increased HDL-C and LDL-C levels were observed. Apart from long-term studies, a study by Ministrini et al. ( 75 ) that treated obese patients with VLCKD for 25 days also concluded that VLCKD had positive effects on cardiovascular risk factors, and such a beneficial outcome in the short term is remarkable.

Other studies investigated the effects of modified KDs on cardiovascular risks in obese participants. Perez-Guisado et al. ( 76 ) carried out a prospective evaluation in 31 obese participants with “Spanish Ketogenic Mediterranean Diet” (incorporating virgin olive oil as a principal source of fat, moderate red wine intake, green vegetables and salads as the primary source of carbohydrates and fish as the main source of protein, SKMD). The SKMD was found safe and effective interventional approach for improving non-atherogenic lipid profiles and lowering blood pressure while lowering body weight. Similarly, Paoli et al. ( 77 ) applied another modified KD that incorporated phytoextracts and ingredients imitating the taste of carbohydrates (ketogenic Mediterranean with phytoextracts, KEMEPHY). The study recruited 106 overweight Rome council employees and revealed a remarkable reduction in body weight, BMI, percentage of fat mass, total cholesterol, LDL-C, TAG and blood glucose while displaying a significant increase in HDL-C after the intervention with KEMEPHY. In addition to good compliance, extra beneficial effects on cardiovascular risk markers and waist circumference were also achieved by the KEMEPHY diet.

Since the beneficial effects of KDs on metabolism and cardiovascular risk factors are similar to those seen after n-3 polyunsaturated fatty acids (omega-3) supplementation, Paoli’s team ( 78 ) modified the ketogenic Mediterranean diet with phytoextracts after their previous research through combining with omega-3 supplementation. The results suggested that this newly modified diet can further enhance the beneficial effects on cardiovascular risk factors and inflammation in overweight participants.

The influence of a multi-step dietary program including different dietary patterns has also been evaluated. In an open-label study by Castaldo et al. ( 79 ), 73 obese patients entered a rehabilitative multi-step dietary program: a 3-week protein-sparing, very low-calorie KD (<500 kcal/day; Oloproteic Diet) and a 6-week hypocaloric (25–30 kcal/kg of ideal body weight/day), low glycemic index, Mediterranean-like diet (hypo-MD). In both phases, improved glucose and lipid metabolism and blood pressure were observed. Based on this, it was concluded that a dietary program consisting of a KD and a subsequent MD could decrease cardiovascular risks efficaciously in obese patients.

CVD During Diabetes Mellitus

Diabetes mellitus is often associated with obesity ( 80 ), and recent estimates showed that 87.5% of T2DM patients are overweight or obese ( 24 ). Moreover, obese subjects are prone to developing hypertension, CVD, and strokes, and the risk is even higher if it co-exists with T2DM ( 81 ).

In patients with both T2DM and obesity, LC diets not only cause weight loss but also improve postprandial plasma glucose levels, glucose variability, serum triglycerides, and HDL-C levels ( 82 ). Similar results were observed by a 2-year randomized clinical trial study ( 39 ) that investigated the effect of an LC diet with high unsaturated fat and low saturated fat on glycemic control and CVD risk factors in overweight or obese patients with T2DM. KD is a low-carbohydrate (LC) and high-fat (HF) diet, which sort of belongs to one type of LC diets. However, one of the potential concerns of KDs is postprandial hyperlipidemia, which leads to significant cardiovascular risks ( 83 ).

Studies have found that individuals with pre-diabetes or diabetes who received an earlier LCHF diet revealed several beneficial outcomes, including weight loss, improved insulin sensitivity, glucose homeostasis, and lower fasting blood glucose levels. These outcome improvements also decreased the risks of cardiovascular diseases development ( 38 , 84 ). Mobbs et al. ( 28 ) analyzed the evidence concerning the treatment of diabetes and diabetic complications with a KD. They revealed that a classic KD significantly reduced blood glucose in animal models of type 1 and 2 diabetes and reversed diabetic nephropathy without producing significant cardiovascular risks. Moreover, a study on db/db mice revealed that KD ameliorates cardiac dysfunction by inhibiting apoptosis via activating the PI3K-Akt pathway in type 2 diabetic mice and suggested KD as a promising lifestyle intervention against diabetic cardiomyopathy ( 85 ).

However, there also are studies with conflicting or controversial findings and opinions. Westman et al. ( 86 ) stated that LCHF diets gave rise to decreased appetite, thus the improved surrogate markers of cardiovascular disease resulted from weight loss but not from low carbohydrate intake itself. In an animal study, Abdurrachim et al. ( 87 ) investigated the effects of long-term KD on cardiac metabolism and diabetic cardiomyopathy status in lean diabetic Goto-Kakizaki (GK) rats. Upon KDs for 62 weeks, diabetic GK rats displayed decreased blood glucose, triglyceride, and insulin levels, revealing increased blood ketone body levels. Additionally, KDs decreased myocardial ketone body and glucose oxidation and induced cardiac hypertrophy. These results suggested that KDs might lead to maladaptive cardiac metabolic modulation and lipotoxicity and deteriorate diabetic cardiomyopathy in GK rats. Given this, the possible role of KDs in cardiovascular risks of DM remains controversial in rodent models and humans, which warrants more studies for elucidation.

Dyslipidemia-Associated CVD

As for patients with dyslipidemia, Westman et al. ( 88 ) investigated the effect of KDs on serum lipoprotein subclasses to address the concern of KDs on cardiovascular risks. The study was a randomized, two-arm clinical trial involving overweight and hyperlipidemic participants motivated to lose weight. After 6 months, the KD group displayed more significant decreases in medium VLDL, small VLDL, and medium LDL, and more significant increases in VLDL particle size, large LDL, and HDL particle size than the control group. Although the KDs did not decrease total LDL cholesterol, they shifted from small, dense LDL to large, buoyant LDL, thus decreasing CVD risks in these participants.

NAFLD-Related CVD

NAFLD contributes to CVD through various mechanisms. Weight loss has been commonly recommended for treating obesity-associated NAFLD; meanwhile, LCKD benefits weight loss. Recent studies have revealed an association between LC diets and NAFLD in both rodents and humans. In the study by Garbow et al. ( 28 ), mice fed a KD for 12 weeks were lean, euglycemic, ketotic, and hypo-insulinemic but were glucose intolerant and with NAFLD. Also, obese subjects on LC diets displayed enhanced weight loss, improved metabolic parameters and decreased intrahepatic triglyceride content. Nevertheless, long-term KDs led to NAFLD and systemic glucose intolerance in mice ( 89 ), negatively impacting CVD. As such, current evidence is insufficient to conclude, and more related studies are warranted to explore how KDs might influence NAFLD-related CVD in the long run.

CVD During GLUT1DS

GLUT1DS is an inherited but treatable disease concerning cerebral energy metabolism ( 90 ). KDs are currently a treatment option for GLUT1DS from infancy into adulthood, raising concerns about long-term cardiovascular risks ( 43 , 60 , 69 ). To address this problem, Heussinger et al. ( 91 ) performed a 10-year follow-up study on cardiovascular risk of KDs in GLUT1DS and revealed that dyslipidemia caused by KDs might be transient; and carotid intimal wall thickness (CIMT), BMI and blood pressure parameters remained normal after 10 years. Because of this, Heussinger et al. suggested that cardiovascular risks of KDs in some previous studies appeared to be attributable to inadequate follow-up. Also, a period of at least five years appears to be necessary for evaluating the effect of KDs on lipid parameters. Moreover, the authors recommended KDs as a treatment of choice for GLUT1DS. Another study by Alter et al. ( 90 ) also characterized the long-term course of GLUT1DS and followed up for an average of 14.2 years (range = 8.9-23.6). The results indicated that earlier introduction of KDs correlated with better long-term outcomes and KDs seemed to be protective of vital organs. However, GLUT1DS is a rare disease, and therefore, the study cohort’s size and external validity are limited. Long-term follow-up studies are warranted to confirm the above findings further.

Potential Safety Concerns on KD

The majority of the studies had found KD to be beneficial, but some studies had shown concerns regarding heart functions, liver inflammation and so on.

The Effects of KD on Heart Functions in Rodents

One study found KD treatment ameliorates cardiac dysfunction by inhibiting apoptosis via activating the PI3K-Akt pathway in type 2 diabetic mice, suggesting that the KD is a promising lifestyle intervention offering protection against diabetic cardiomyopathy ( 85 ). In contrast, a ketogenic diet may lead to adverse effects on the remodeling in the hypertensive heart via mechanisms involving increased mTOR signaling, and they underscore the necessity to evaluate its reliability before clinical use ( 53 ). Preclinical studies results indicate that KD also has a potential safety concern, although much evidence suggests that KD is a promising approach for managing CVD.

The Effects of KD on Hepatic Inflammation in Rodents

As a key factor that triggers or exacerbates CVD risk, liver inflammation is a potential safety concern related to KD. In support of this, a study observed that mice fed with KD sustained unimpaired insulin-induced hepatic Akt phosphorylation and whole-body insulin responsiveness but ultimately developed hepatic endoplasmic reticulum stress, steatosis, cellular injury, and macrophage accumulation ( 28 ).

The Effects of KD on Lipid Profile

KD is enriched in lipid contents, and it’s natural to speculate the potential risk of elevated levels of lipids. Apart from studies indicating the beneficial effects of KD, concerns regarding the elevated level of lipids, including serum total and LDL cholesterol and triglyceride, are subjective while prescribing KD ( 65 , 72 ). It is reported that KDs are likely to deteriorate levels of total, high-density lipoprotein (HDL) and low-density lipoprotein (LDL) cholesterol, and triglycerides ( 27 , 34 , 92 ) in rodents while doing the opposite in humans ( 39 ). These contradictory results might be attributable to the different composition of diets since animal researches generally employ diets higher not only in total fat but also in saturated fat ( 93 ). Considering this, it is necessary to compare the fat composition, e.g., content of saturated fat versus unsaturated fat in KDs in long-term studies involving both rodents and humans.

As described above, while considering KD as an exciting approach for managing CMDs, it also is important to be cautious about the potential safety concern associated with KD. While future studies are warranted to confirm and elucidate whether and how KD causes potential safety concerns, it would also be important to consider to modifying KD or combining KD with other healthy diets for managing CMDs.

Despite particular safety concerns, the beneficial and advantageous aspects of KD cannot be denied. Because multiple factors are affecting the results, including using different mouse strains, providing KD with different food compositions, short study duration, etc. In the future, studies essentially need to explore the possible factors influencing the responses to KD and improve KD dietary plan for utilizing KD as a dietary therapy to minimize safety concerns.

Factors Affecting Ketogenic Diets Responses

Genetic control of the responses to a ketogenic diet.

Nutrigenetic research suggested that genetic markers critically regulate nutritional interactions that impact body weight and composition, which lays the foundation for personalized nutrition therapy ( 94 , 95 ).

Barrington et al. ( 96 ) observed that mouse genetic backgrounds determined dietary outcomes on CVD risk. Specifically, the study included mice from four inbred strains (A, B6, FVB, and NOD), which accounted for genetic and phenotypic diversity and examined mice’s metabolic responses to four human-comparable mice diets (American, Mediterranean, Japanese and ketogenic diets). The authors revealed that the effects of these diets on metabolic health were indeed dependent on genetic backgrounds. The outcomes of KD on body composition, glucose metabolism and liver health varied markedly among different strains.

The different diet responses could be partly attributable to the genetic background related to varying dietary therapy compliance. Parnell et al. ( 97 ) analyzed the interactions between single nucleotide polymorphisms (SNP) in various cardiometabolic pathways and the intake of different nutrients. Their results indicated that gene-environment (GxE) genes had better responses to plasma cholesterol-lowering or regression of atherosclerotic plaques, primarily through high-energy diets and fat intake.

As mentioned above, genetic background plays a vital role in individual responses to KDs and may consequently influence the effects of KDs. It is of great importance to take genetic background into account when initiating KD therapy.

Food Composition, Quality and Sources of KDs Influence the Outcome

As KDs are a kind of macronutrient-focused diet, we should fully consider the food composition, quality and sources to avoid potential drawbacks when starting a KD. As indicated in the research by Seidelmann et al. ( 98 ), there was a U-shaped association between the percentage of energy consumed from carbohydrates and mortality: 50–55% carbohydrate intake was associated with minimal mortality risk. In comparison, a percentage of <40% or >70% led to greater mortality risk. Besides, different types of dietary fatty acids have different effects on CVD risk and replacing saturated fatty acid (SFA) with unsaturated fats especially polyunsaturated fatty acids can lead to a significant reduction in CVD risk ( 99 ). Moreover, diets that favored plant-derived protein and fat intake were associated with lower mortality than animal-derived protein and fat sources. Thus, the nutrient composition, types and sources should be taken into consideration when prescribing a KD therapy; the diversity in these nutrient details could affect the effects of KD and should be relatively standardized to compare the results.

Duration of KD Therapy Affects the Responses

Interestingly, increasing the therapeutic duration of KDs appears to reduce some safety-related problems ( 34 , 35 ). For instance, long-term follow-up research has demonstrated that dyslipidemia caused by KDs is transient. Moreover, over 10 years, KD therapy has ended with normal vascular function as indicated by carotid artery ultrasound ( 91 ).

Modified KD Dietary Plan

Because of the irreconcilable options on the therapeutic use of KD, several studies concerning modified KD and cardiovascular risks have been performed. The “Spanish Ketogenic Mediterranean Diet” carried out by Perez-Guisado et al. ( 76 ) and two modified KDs (KEMEPHY ( 77 ) and KEMEPHY with omega-3 supplementation ( 78 )) employed by Paoli et al. have all displayed beneficial effects on cardiovascular risk factors. A combined diet consisting of KD and a subsequent Mediterranean-like diet has been proven to decrease cardiovascular risks in patients ( 79 ). Therefore, a modified KD or multi-step dietary program including different diet patterns is promising in resolving the safety concerns associated with KDs.

Based on the currently available evidence, KD appears to play a significant role in treating various cardio-metabolic diseases and reveals remarkable effects on cardiovascular function. KD therapy is generally promising in obesity, heart failure, and hypertension, though different voices still exist. In diabetes and dyslipidemia, the performance of KD remains controversial. As for cardiovascular complications of metabolic diseases, current evidence suggests that KD is generally protective to obese related cardiovascular disease (CVD), while remaining contradictory to diabetes and other metabolic disorder related CVDs. Various factors might account for the controversies, including genetic background, duration of therapy, food composition, quality and sources of KDs. Therefore, further studies are warranted to provide concrete and more conclusive opinions. Also, it is vital to monitor safety-related signs and biomarkers during the KD intervention, although most are reversible or transient. In addition, modified KD could be adequately designed and utilized to enhance compliance as a therapeutic approach. Overall, there is a critical need to conduct more rigorous research focusing on the clinical implication and safety issues of KD.

Author Contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work, and approved it for publication. WYZ: designing, conceptualization, writing, figure plotting, revising; JZ and CDW: designing, funding acquisition, review & editing; XG: funding acquisition, review & editing; LLC: supervision, editing, revising; TC and JYY: figure plotting.

The development of this review was supported in whole or in part by grants from the National Natural Science Foundation of China (81770772 to JZ), the Hubei Province Natural Science Foundation (2019CFB701 to JZ), National Natural Science Foundation of China (81803224 to XG) and Young Scholars Program of Shandong University (2018WLJH33 to XG).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

AcknowleDgments

We especially express our appreciation to Mohammad Ishraq Zafar for his assistance with the English language of our review.

COMMENTS

  1. The Potential Health Benefits of the Ketogenic Diet: A Narrative Review

    1. Introduction Ketogenic diets have started to increase in popularity as doctors and researchers investigate the potential benefits. Nutritional ketosis, the aspirational endpoint of ketogenic diets, is achieved by restricting carbohydrate intake, moderating protein consumption, and increasing the number of calories obtained from fat [ 1 ].

  2. Ketogenic Diet

    The ketogenic or keto diet is a dietary approach characterized by high-fat and low-carbohydrate intake, aiming to facilitate weight loss, enhance mental clarity, and boost energy levels. [4] By significantly reducing carbohydrate consumption and increasing fat and protein intake, this diet induces a metabolic state called ketosis, where the body utilizes fat as its primary fuel source instead ...

  3. Advantages and Disadvantages of the Ketogenic Diet: A Review Article

    The ketogenic diet (KD) has gained immense popularity during the last decade, primarily because of its successful short-term effect on weight loss. In the United States, KD is utilized in a variety of patient populations for weight management, despite ...

  4. Ketogenic Diet: Risks and Downfalls

    Review of current evidence and clinical recommendations on the effects of low-carbohydrate and very-low-carbohydrate (including ketogenic) diets for the management of body weight and other cardiometabolic risk factors: a scientific statement from the National Lipid Association Nutrition and Lifestyle Task Force..

  5. Effect of the ketogenic diet on glycemic control, insulin resistance

    Background At present, the beneficial effect of the ketogenic diet (KD) on weight loss in obese patients is generally recognized. However, a systematic research on the role of KD in the ...

  6. Effects of ketogenic diet on health outcomes: an umbrella review of

    Table 2 Summary of significant associations of ketogenic diet with health outcomes supported by moderate to high quality of evidence Full size table Types of KD showed different effects on health outcomes with changes more than the MCID thresholds in different populations (Fig. 2 ).

  7. Impact of the ketogenic diet as a dietary approach on cardiovascular

    Conclusions Although the ketogenic diet demonstrates benefits in terms of triglyceride, blood pressure, weight, and glycemic control, its impact on CVD risk factors, especially the elevated total cholesterol and low-density lipoprotein cholesterol concentrations, warrants a cautious approach.

  8. Ketogenic Diet for Weight Loss and Its Implication on Health: a

    This literature review aimed to discuss the mechanism of ketogenic diet in weight lost as well as its long and short term eff ects on health. Ketogenic diet is a very low-carbohydrate and high-fat ...

  9. The Effect of Ketogenic-Diet on Health

    The ketogenic diet (KD) is a high-fat, adequate-protein, and very-lowcarbohydrate diet regiment that mimics the metabolism of the fasting state (1), for the purpose of harnessing its high fat ...

  10. Impact of ketogenic diet on cardiovascular disease

    Impact of ketogenic diet on serum circulating lipids. The link between serum levels of circulating lipids and CVD is well established. 37,60-62 Dyslipidemia is a hallmark of atheroprogression, and dietary interventions are classically associated with the overall reduction of lipid and fat intake to reduce hyperlipidemia.

  11. A Review of Ketogenic Diet and Lifestyle

    The ketogenic diet has become increasing popular in recent years. With 25.4 million unique searches, the keto diet was the most Googled diet in the United States in 2020. With increased consumer interest, the "keto" food industry has grown ...

  12. Ketogenic Diet: Overview

    In order to prevent unnecessary health issues and achieve the desired effect, nutritionists offer patients to use the rules of the popular ketogenic diet.

  13. PDF Review of current evidence and clinical recommendations on the ...

    This scientific statement was developed after the NLA Scientific Statements Committee and the NLA Board of Directors approved a proposal for its development to address the recent popularity of using low-carbohydrate and very-low-carbohydrate diets, including ketogenic diets, for the management of cardiometabolic risk factors and type 2 diabetes.

  14. The Effects of Ketogenic Diet on the Immune System: A Meta-Analysis

    In this regard, they are focusing on promoting the intake of foods comprising various diets with health benefits, such as the ketogenic diet.

  15. Very-low-calorie ketogenic diet (VLCKD) in the management of metabolic

    Very-low calorie ketogenic diets (VLCKDs) are increasingly used in clinical practice for weight loss and management of obesity-related comorbidities. Despite evidence on the clinical benefits of VLCKDs is rapidly emerging, some concern still exists about their potential risks and their use in the long-term, due to paucity of clinical studies.

  16. The Ketogenic Diet (Argument Essay)

    An argument essay arguing in support of the ketogenic diet, a low-carb, high-fat diet. Formatted according to MLA style. 1165 words with a works cited page that has five sources.

  17. Ketogenic Diets: Side Effects, Attitude, and Quality of Life

    The outcomes of the participants approved the efficacy of the ketogenic diet in weight reduction. Different symptoms and side effects occurred with varying intensities, especially in the first few days of adopting this diet. Overall, the ketogenic diet did not affect the quality of life and yielded a very positive overall experience.

  18. The Ketogenic Diet and Cardiovascular Diseases

    The ketogenic diet has a beneficial effect on blood pressure and other CVD risk factors through, among other aspects, weight loss. The evidence cited is often superior to that for standard diets, making it likely that the ketogenic diet shows advantages over other dietary models in the prevention and treatment of cardiovascular diseases.

  19. The Ketogenic Diet and Cardiovascular Diseases

    The most common and increasing causes of death worldwide are cardiovascular diseases (CVD). Taking into account the fact that diet is a key factor, it is worth exploring this aspect of CVD prevention and therapy. The aim of this article is to assess the potential of the ketogenic diet in the prevention and treatment of CVD. The article is a comprehensive, meticulous analysis of the literature ...

  20. The case for a ketogenic diet in the management of kidney disease

    Ketogenic diets have been widely used for weight loss and are increasingly used in the management of type 2 diabetes. Despite evidence that ketones have multiple positive effects on kidney function, common misconceptions about ketogenic diets, such as high protein content and acid load, have prevented their widespread use in individuals with impaired kidney function. Clinical trial evidence ...

  21. Ketogenic Diets and Cardio-Metabolic Diseases

    A ketogenic diet (KD) constitutes a high-fat and low-carbohydrate diet with appropriate protein content and calories. KD has drawn the interests of clinicians and scientists regarding its application in the management of metabolic diseases and related disorders; thus, the current review aimed to examine the evidences surrounding KD and the CMDs ...