[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (500)

Search Parameters:
Keywords = ketogenic diet

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
8 pages, 956 KiB  
Perspective
Diabetic Kidney Disease: Contribution of Phenyl Sulfate Derived from Dietary Tyrosine upon Gut Microbiota Catabolism
by Haoxin Liu, Tram N. Diep, Ying Wang, Yucheng Wang and Liang-Jun Yan
Biomolecules 2024, 14(9), 1153; https://doi.org/10.3390/biom14091153 - 13 Sep 2024
Viewed by 315
Abstract
Deranged gut microbiota can release increased levels of uremic toxins leading to exacerbated kidney injury. In diabetic kidney disease (DKD), phenyl sulfate (PS) derived from tyrosine catabolism by gut microbiota has been demonstrated to be both an early diagnostic marker and a therapeutic [...] Read more.
Deranged gut microbiota can release increased levels of uremic toxins leading to exacerbated kidney injury. In diabetic kidney disease (DKD), phenyl sulfate (PS) derived from tyrosine catabolism by gut microbiota has been demonstrated to be both an early diagnostic marker and a therapeutic target. In this perspective article, we summarize PS generation pathways and recent findings on PS and kidney injury in DKD. Increasing evidence has shown that the underlying mechanisms of PS-induced kidney injury mainly involve oxidative stress, redox imbalance, and mitochondrial dysfunction, which all may be targeted to attenuate PS-induced kidney injury. For future research directions, we think that a deeper understanding of the pathogenic role of PS in kidney injury using a variety of diabetic animal models should be investigated. Moreover, we also suggest beneficial approaches that could be used to mitigate the deleterious effect of PS on the kidney. These approaches include caloric restriction, tyrosine restriction, and administration of ketogenic drugs, ketogenic diets or natural products; all of which should be conducted under obese and diabetic conditions. Full article
Show Figures

Figure 1

Figure 1
<p>Major pathway of phenyl sulfate formation via gut microbiota catabolism of dietary tyrosine. Tyrosine is converted to phenol by the bacterial enzyme tyrosine phenol lyase followed by further conversion to phenyl sulfate in the liver. Phenyl sulfate is usually eliminated by the kidney but can accumulate in the kidney and cause further kidney damage.</p>
Full article ">Figure 2
<p>Major mechanisms underlying PS-induced kidney injury.</p>
Full article ">
17 pages, 1503 KiB  
Article
Comparative Efficacy of Low-Carbohydrate and Ketogenic Diets on Diabetic Retinopathy and Oxidative Stress in High-Fat Diet-Induced Diabetic Rats
by Monya T. Jawharji, Ghedeir M. Alshammari, Manal Abdulaziz Binobead, Nouf Mohammed Albanyan, Laila Naif Al-Harbi and Mohammed Abdo Yahya
Nutrients 2024, 16(18), 3074; https://doi.org/10.3390/nu16183074 - 12 Sep 2024
Viewed by 373
Abstract
This study examined the effect of a low-carbohydrate diet (LCD) and a low-carbohydrate ketogenic diet (LCKD) on diabetic retinopathy in high-fat diet-induced diabetes mellitus in rats and studied the mechanisms of action. Rats were divided into four groups: the Control group, which was [...] Read more.
This study examined the effect of a low-carbohydrate diet (LCD) and a low-carbohydrate ketogenic diet (LCKD) on diabetic retinopathy in high-fat diet-induced diabetes mellitus in rats and studied the mechanisms of action. Rats were divided into four groups: the Control group, which was fed a normal diet for 16 weeks; the HFD group, which was fed a high-fat diet (HFD) for the first 8 weeks and then switched to a normal diet for 8 weeks; the HFD+LCD group, fed a HFD for 8 weeks followed by an LCD for 8 weeks, and the HFD+LCKD group, which was fed a HFD for 8 weeks followed by an LCKD for 8 more weeks. Both the LCD and the LCKD effectively reduced the final body and total fat weights and decreased fasting serum levels of glucose, insulin, hemoglobin A1 (HbA1C), triglycerides, cholesterol, and LDL-c. They also reduced the levels of malondialdehyde (MDA), tumor necrosis factor-α, vascular endothelial factor, caspapse-3, and bax. In the HFD rats, we found increased serum levels of β-Hydroxybutyrate and upregulated expression of Bcl2, glutathione, superoxide dismutase, and hemeoxygenase-1. Moreover, the LCD and LCKD significantly reduced mRNA levels of Kelch-like ECH-associated protein 1 (Keap1) and enhanced mRNA and nuclear concentrations of nuclear factor erythroid factor 2 (Nrf2). All these effects were associated with improved layers of the retina in the HFD − LCD and HFD + LCKD rats but not in HFD animals. The impact of the LCKD was always more profound on all measured parameters and on improving the structure of the retina compared to the LCD. In conclusion, the LCKD is superior to the LCD in preventing diabetic retinopathy in HFD-fed rats. Mechanistically, our results suggest that the hypoglycemic and hypolipidemic conditions and the Nrf2-dependent antioxidant and anti-inflammatory effects may be involved in the preventative effects of the LCD and LCKD. Full article
Show Figures

Figure 1

Figure 1
<p>The levels of malondialdehyde (MDA) (<b>A</b>), glutathione (GSH) (<b>B</b>), heme oxygenase-1 (HO-1) (<b>C</b>), and superoxide dismutase (<b>D</b>) in retinas of all groups of rats. Data are presented as means ± SD for <span class="html-italic">n</span> = 8 rats/group. ***: significantly different compared with control at <span class="html-italic">p</span> &lt; 0.001; <sup>##</sup> and <sup>###</sup>: significantly different compared with HFD-fed rats at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively; <sup><span>$</span><span>$</span></sup> and <sup><span>$</span><span>$</span><span>$</span></sup>: significantly different compared with HFD + LCD-fed rats at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively. Malondialdehyde (MDA), total glutathione (GSH), heme oxygenase-1 (HO-1), and superoxide dismutase (SOD).</p>
Full article ">Figure 2
<p>Levels of advanced aged glycation products (AGEs) (<b>A</b>), tumor necrosis factor-α (TNF-α) (<b>B</b>), levels of interleukin-6 (IL-6) (<b>C</b>), and nuclear activity of NF-κB (<b>D</b>) in retinas of all groups of rats. Data are presented as means ± SD for <span class="html-italic">n</span> = 8 rats/group. * and ***: significantly different compared with control at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.001, respectively; <sup>##</sup> and <sup>###</sup>: significantly different compared with HFD-fed rats at <span class="html-italic">p</span> &lt; 0.01 and 0.001, respectively; <sup><span>$</span><span>$</span></sup> and <sup><span>$</span><span>$</span><span>$</span></sup>: significantly different compared with HFD + LCD-fed rats at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively. Tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and nuclear factor kappa beta (NF-κB).</p>
Full article ">Figure 3
<p>Levels of caspase-3 (<b>A</b>), Bcl2 (<b>B</b>), BAX (<b>C</b>), and Bax/Bcl2 (<b>D</b>) in the retinas of all groups of rats. Data are presented as means ± SD for <span class="html-italic">n</span> = 8 rats/group. * and ***: significantly different compared with control at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.001, respectively; <sup>###</sup>: significantly different compared with HFD-fed rats at <span class="html-italic">p</span> &lt; 0.001; <sup><span>$</span><span>$</span><span>$</span></sup>: significantly different compared with HFD + LCD-fed rats at <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>mRNA levels of Nrf2 (<b>A</b>) and Keap1 (<b>B</b>), and nuclear levels of Nrf2 (<b>C</b>) in retinas of all groups of rats. Data are presented as means ± SD for <span class="html-italic">n</span> = 8 rats/group. ***: significantly different compared with control at <span class="html-italic">p</span> &lt; 0.001; <sup>##</sup> and <sup>###</sup>: significantly different compared with HFD-fed rats at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively; <sup><span>$</span><span>$</span><span>$</span></sup>: significantly different compared with HFD + LCD-fed rats at <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Histological alterations in retinas of all groups of rats. (<b>A</b>) Control rat: normal retina composed of intact nerve fiber layer (NFL), ganglionic cell layer (GCs), inner nuclear layer (INL), outer plexiform layer (OPL), outer nuclear layer (ONL), and photoreceptor layer (PRs). Note the abundancy of cells in each layer. (<b>B</b>) HFD-fed rat: clear increase in thickness of all layers of retina, damaged NFL (black arrow), reduced number of GCs and increased number of pyknotic GCs (red arrow), reduction in number of pyknotic cells forming the INL (yellow arrow), and vacuolization and loss of PRs (white arrow). However, cells forming the ONL appeared normal but increased in number (green arrow). (<b>C</b>) HFD + LCD rat: intact NFL (black arrow), normal ONL (white arrow), and intact PRs (green arrow). However, pyknotic GCs (red arrow) with partial loss of cells of the INL (yellow arrow) are still seen. (<b>D</b>) HFD + LCKD rat: normal retinal features with intact NLF (black arrow), abundant and intact GCs (red arrow), and intact cells forming the INL (yellow arrow), ONL (white arrow), and PRs (green arrow).</p>
Full article ">
20 pages, 2621 KiB  
Systematic Review
Effects of Different Exercises Combined with Different Dietary Interventions on Body Composition: A Systematic Review and Network Meta-Analysis
by Yongchao Xie, Yu Gu, Zhen Li, Bingchen He and Lei Zhang
Nutrients 2024, 16(17), 3007; https://doi.org/10.3390/nu16173007 - 5 Sep 2024
Viewed by 1964
Abstract
Background: Exercise and dietary interventions are essential for maintaining weight and reducing fat accumulation. With the growing popularity of various dietary strategies, evidence suggests that combining exercise with dietary interventions offers greater benefits than either approach alone. Consequently, this combined strategy has become [...] Read more.
Background: Exercise and dietary interventions are essential for maintaining weight and reducing fat accumulation. With the growing popularity of various dietary strategies, evidence suggests that combining exercise with dietary interventions offers greater benefits than either approach alone. Consequently, this combined strategy has become a preferred method for many individuals aiming to maintain health. Calorie restriction, 5/2 intermittent fasting, time-restricted feeding, and the ketogenic diet are among the most popular dietary interventions today. Aerobic exercise, resistance training, and mixed exercise are the most widely practiced forms of physical activity. Exploring the best combinations of these approaches to determine which yields the most effective results is both meaningful and valuable. Despite this trend, a comparative analysis of the effects of different exercise and diet combinations is lacking. This study uses network meta-analysis to evaluate the impact of various combined interventions on body composition and to compare their efficacy. Methods: We systematically reviewed literature from database inception through May 2024, searching PubMed, Web of Science, Embase, and the Cochrane Library. The study was registered in PROSPERO under the title: “Effects of Exercise Combined with Different Dietary Interventions on Body Composition: A Systematic Review and Network Meta-Analysis” (identifier: CRD42024542184). Studies were meticulously selected based on specific inclusion and exclusion criteria (The included studies must be randomized controlled trials involving healthy adults aged 18 to 65 years. Articles were rigorously screened according to the specified inclusion and exclusion criteria.), and their risk of bias was assessed using the Cochrane risk of bias tool. Data were aggregated and analyzed using network meta-analysis, with intervention efficacy ranked by Surface Under the Cumulative Ranking (SUCRA) curves. Results: The network meta-analysis included 78 randomized controlled trials with 5219 participants, comparing the effects of four combined interventions: exercise with calorie restriction (CR+EX), exercise with time-restricted eating (TRF+EX), exercise with 5/2 intermittent fasting (5/2F+EX), and exercise with a ketogenic diet (KD+EX) on body composition. Intervention efficacy ranking was as follows: (1) Weight Reduction: CR+EX > KD+EX > TRF+EX > 5/2F+EX (Relative to CR+EX, the effect sizes of 5/2F+EX, TRF+EX and KD+EX are 2.94 (−3.64, 9.52); 2.37 (−0.40, 5.15); 1.80 (−1.75, 5.34)). (2) BMI: CR+EX > KD+EX > 5/2F+EX > TRF+EX (Relative to CR+EX, the effect sizes of 5/2F+EX, TRF+EX and KD+EX are 1.95 (−0.49, 4.39); 2.20 (1.08, 3.32); 1.23 (−0.26, 2.71)). (3) Body Fat Percentage: CR+EX > 5/2F+EX > TRF+EX > KD+EX (Relative to CR+EX, the effect sizes of 5/2F+EX, TRF+EX and KD+EX are 2.66 (−1.56, 6.89); 2.84 (0.56, 5.13); 3.14 (0.52, 5.75).). (4) Lean Body Mass in Male: CR+EX > TRF+EX > KD+EX (Relative to CR+EX, the effect sizes of TRF+EX and KD+EX are −1.60 (−6.98, 3.78); −2.76 (−7.93, 2.40)). (5) Lean Body Mass in Female: TRF+EX > CR+EX > 5/2F+EX > KD+EX (Relative to TRF+EX, the effect sizes of CR+EX, 5/2F+EX and KD+EX are −0.52 (−2.58, 1.55); −1.83 (−4.71, 1.04); −2.46 (−5.69,0.76).). Conclusion: Calorie restriction combined with exercise emerged as the most effective strategy for reducing weight and fat percentage while maintaining lean body mass. For women, combining exercise with time-restricted eating proved optimal for preserving muscle mass. While combining exercise with a ketogenic diet effectively reduces weight, it is comparatively less effective at decreasing fat percentage and preserving lean body mass. Hence, the ketogenic diet combined with exercise is considered suboptimal. Full article
(This article belongs to the Section Sports Nutrition)
Show Figures

Figure 1

Figure 1
<p>Flow diagram of study selection.</p>
Full article ">Figure 2
<p>Network Meta-Analysis of Weight: Network Plot, League Table, and SUCRA Plot. (<b>A</b>) Network Plot. The size of the nodes is proportional to the sample size of each dietary intervention, and the thickness of the lines corresponds to the number of available studies. (<b>B</b>) Pairwise comparison League Table, where the estimated effect size differences (SMD with 95% CI) represent the difference between the intervention on the top and the intervention on the right. (<b>C</b>) The SUCRA Plot, where the size of the area under the curve indicates the effectiveness of each intervention.</p>
Full article ">Figure 3
<p>Network Meta-Analysis of BMI: Network Plot, League Table, and SUCRA Plot. (<b>A</b>) Network Plot. The size of the nodes is proportional to the sample size of each dietary intervention, and the thickness of the lines corresponds to the number of available studies. (<b>B</b>) Pairwise comparison League Table, where the estimated effect size differences (SMD with 95% CI) represent the difference between the intervention on the top and the intervention on the right. (<b>C</b>) The SUCRA Plot, where the size of the area under the curve indicates the effectiveness of each intervention.</p>
Full article ">Figure 4
<p>Network Meta-Analysis of Body fat percentage: Network Plot, League Table, and SUCRA Plot. (<b>A</b>) Network Plot. The size of the nodes is proportional to the sample size of each dietary intervention, and the thickness of the lines corresponds to the number of available studies. (<b>B</b>) Pairwise comparison League Table, where the estimated effect size differences (SMD with 95% CI) represent the difference between the intervention on the top and the intervention on the right. (<b>C</b>) The SUCRA Plot, where the size of the area under the curve indicates the effectiveness of each intervention.</p>
Full article ">Figure 5
<p>Network Meta-Analysis of male lean body mass: Network Plot, League Table, and SUCRA Plot. (<b>A</b>) Network Plot. The size of the nodes is proportional to the sample size of each dietary intervention, and the thickness of the lines corresponds to the number of available studies. (<b>B</b>) Pairwise comparison League Table, where the estimated effect size differences (SMD with 95% CI) represent the difference between the intervention on the top and the intervention on the right. (<b>C</b>) The SUCRA Plot, where the size of the area under the curve indicates the effectiveness of each intervention.</p>
Full article ">Figure 6
<p>Network Meta-Analysis of female lean body mass: Network Plot, League Table, and SUCRA Plot. (<b>A</b>) Network Plot. The size of the nodes is proportional to the sample size of each dietary intervention, and the thickness of the lines corresponds to the number of available studies. (<b>B</b>) Pairwise comparison League Table, where the estimated effect size differences (SMD with 95% CI) represent the difference between the intervention on the top and the intervention on the right. (<b>C</b>) The SUCRA Plot, where the size of the area under the curve indicates the effectiveness of each intervention.</p>
Full article ">
25 pages, 655 KiB  
Review
Systematic Review and Clinical Insights: The Role of the Ketogenic Diet in Managing Glioblastoma in Cancer Neuroscience
by Jose Valerio, Matteo Borro, Elisa Proietti, Livia Pisciotta, Immanuel O. Olarinde, Maria Fernandez Gomez and Andres Mauricio Alvarez Pinzon
J. Pers. Med. 2024, 14(9), 929; https://doi.org/10.3390/jpm14090929 - 31 Aug 2024
Viewed by 943
Abstract
Recent scientific research has shown that the ketogenic diet may have potential benefits in a variety of medical fields, which has led to the diet receiving a substantial amount of attention. Clinical and experimental research on brain tumors has shown that the ketogenic [...] Read more.
Recent scientific research has shown that the ketogenic diet may have potential benefits in a variety of medical fields, which has led to the diet receiving a substantial amount of attention. Clinical and experimental research on brain tumors has shown that the ketogenic diet has a satisfactory safety profile. This safety profile has been established in a variety of applications, including the management of obesity and the treatment of drug-resistant epileptic cases. However, in human studies, the impact of ketogenic therapy on the growth of tumors and the life expectancy of patients has not provided results that are well characterized. Consequently, our purpose is to improve the comprehension of these features by succinctly presenting the developments and conclusions that have been gained from the most recent study that pertains to this non-pharmacological technique. According to the findings of our study, patients with brain tumors who stick to a ketogenic diet are more likely to experience improved survival rates. However, it is required to conduct additional research on humans in order to more accurately define the anti-tumor efficiency of this diet as well as the underlying processes that support the therapeutic effects of this dieting regimen. Full article
Show Figures

Figure 1

Figure 1
<p>Paper-selection process for this systematic review.</p>
Full article ">
17 pages, 965 KiB  
Review
Molecular Mechanisms of Healthy Aging: The Role of Caloric Restriction, Intermittent Fasting, Mediterranean Diet, and Ketogenic Diet—A Scoping Review
by Roxana Surugiu, Mihaela Adela Iancu, Ștefănița Bianca Vintilescu, Mioara Desdemona Stepan, Daiana Burdusel, Amelia Valentina Genunche-Dumitrescu, Carmen-Adriana Dogaru and Gheorghe Gindrovel Dumitra
Nutrients 2024, 16(17), 2878; https://doi.org/10.3390/nu16172878 - 28 Aug 2024
Viewed by 2504
Abstract
As the population ages, promoting healthy aging through targeted interventions becomes increasingly crucial. Growing evidence suggests that dietary interventions can significantly impact this process by modulating fundamental molecular pathways. This review focuses on the potential of targeted dietary strategies in promoting healthy aging [...] Read more.
As the population ages, promoting healthy aging through targeted interventions becomes increasingly crucial. Growing evidence suggests that dietary interventions can significantly impact this process by modulating fundamental molecular pathways. This review focuses on the potential of targeted dietary strategies in promoting healthy aging and the mechanisms by which specific nutrients and dietary patterns influence key pathways involved in cellular repair, inflammation, and metabolic regulation. Caloric restriction, intermittent fasting, the Mediterranean diet, as well as the ketogenic diet showed promising effects on promoting healthy aging, possibly by modulating mTORC1 AMPK, an insulin signaling pathway. By understanding the intricate interplay between diet and molecular pathways, we can develop personalized dietary strategies that not only prevent age-related diseases, but also promote overall health and well-being throughout the aging process. Full article
(This article belongs to the Section Geriatric Nutrition)
Show Figures

Figure 1

Figure 1
<p>Main effects of dietary interventions on healthy aging.</p>
Full article ">Figure 2
<p>Main metabolic pathways and effect of caloric restriction, intermittent fasting, ketogenic diet, and Mediterranean diet (created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 19 August 2024)).</p>
Full article ">
29 pages, 4245 KiB  
Article
Effects of Ketogenic Diet on Increased Ethanol Consumption Induced by Social Stress in Female Mice
by Laura Torres-Rubio, Marina D. Reguilón, Susana Mellado, María Pascual and Marta Rodríguez-Arias
Nutrients 2024, 16(17), 2814; https://doi.org/10.3390/nu16172814 - 23 Aug 2024
Viewed by 848
Abstract
Stress is a critical factor in the development of mental disorders such as addiction, underscoring the importance of stress resilience strategies. While the ketogenic diet (KD) has shown efficacy in reducing alcohol consumption in male mice without cognitive impairment, its impact on the [...] Read more.
Stress is a critical factor in the development of mental disorders such as addiction, underscoring the importance of stress resilience strategies. While the ketogenic diet (KD) has shown efficacy in reducing alcohol consumption in male mice without cognitive impairment, its impact on the stress response and addiction development, especially in females, remains unclear. This study examined the KD’s effect on increasing ethanol intake due to vicarious social defeat (VSD) in female mice. Sixty-four female OF1 mice were divided into two dietary groups: standard diet (n = 32) and KD (n = 32). These were further split based on exposure to four VSD or exploration sessions, creating four groups: EXP-STD (n = 16), VSD-STD (n = 16), EXP-KD (n = 16), and VSD-KD (n = 16). KD-fed mice maintained ketosis from adolescence until the fourth VSD/EXP session, after which they switched to a standard diet. The Social Interaction Test was performed 24 h after the last VSD session. Three weeks post-VSD, the Drinking in the Dark test and Oral Ethanol Self-Administration assessed ethanol consumption. The results showed that the KD blocked the increase in ethanol consumption induced by VSD in females. Moreover, among other changes, the KD increased the expression of the ADORA1 and CNR1 genes, which are associated with mechanisms modulating neurotransmission. Our results point to the KD as a useful tool to increase resilience to social stress in female mice. Full article
Show Figures

Figure 1

Figure 1
<p>Experimental design.</p>
Full article ">Figure 2
<p>Weekly body weight and kilocalories (kcal) intake: (<b>A</b>) Weekly body weight. Data represented the body weight average per group measured weekly. The shaded area refers to the weeks in which the KD was maintained. Vertical lines show ± SEM. ** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 significant difference in KD-EXP groups with respect to the STD-EXP group. +++ <span class="html-italic">p</span> &lt; 0.001 and + <span class="html-italic">p</span> &lt; 0.05 significant difference in KD-VSD-R group with respect to the STD-VSD-R group. # <span class="html-italic">p</span> &lt; 0.05 significant differences in KD-VSD-S group with respect to the STD-VSD-S group. (<b>B</b>) Weekly kcal intake. Data are represented as the average kcal intake per group measured weekly. Vertical lines show ±SEM. # <span class="html-italic">p</span> &lt; 0.05 and ### <span class="html-italic">p</span> &lt; 0.001 significant difference with respect to the rest of the weeks in each period. *** <span class="html-italic">p</span> &lt; 0.001 and * <span class="html-italic">p</span> &lt; 0.05 significant difference with respect to non-stressed females. +++ <span class="html-italic">p</span> &lt; 0.001, ++ <span class="html-italic">p</span> &lt; 0.01 and + <span class="html-italic">p</span> &lt; 0.05 significant difference with respect to STD females.</p>
Full article ">Figure 3
<p>Weekly and β-hydroxybutyrate blood levels. Data are represented as the average of βHB plasma levels of KD groups measured weekly (black squares). Vertical lines show ±SEM. *** <span class="html-italic">p</span> &lt; 0.001 significant differences with respect to the STD groups. +++ <span class="html-italic">p</span> &lt; 0.001 and + <span class="html-italic">p</span> &lt; 0.05 significant differences in the KD-EXP group with respect to the other weeks. # <span class="html-italic">p</span> &lt; 0.05 significant differences in the KD-VSD-R groups with respect to the 3rd and 4th weeks. &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.001 and &amp; <span class="html-italic">p</span> &lt; 0.05 significant differences in the KD-VSD-S group with respect to the other weeks. The black figures show the βHB plasma levels average of STD groups measured in the 6th week.</p>
Full article ">Figure 4
<p>Corticosterone plasma levels after the 1st and 4th VSD sessions. The bars represent the corticosterone levels and the vertical lines show ±SEM. *** <span class="html-italic">p</span> &lt; 0.001 significant differences with respect to the non-stressed groups. +++ <span class="html-italic">p</span> &lt; 0.001 significant differences with respect to the STD groups. ## <span class="html-italic">p</span> &lt; 0.05 significant differences with respect to the 4th VSD. &amp; <span class="html-italic">p</span> &lt; 0.05 significant differences with respect to the 1st VSD.</p>
Full article ">Figure 5
<p>Social Interaction Test (SIT): (<b>A</b>) VSD and KD effect on the SIT ratio in female mice. The bars represent the ratio of the SIT and the vertical lines show ±SEM. Values &gt;1 indicate preference for social interaction, and &lt;1 indicates social avoidance. *** <span class="html-italic">p</span> &lt; 0.001 significant difference with respect to the non-stressed and VSD-R groups. (<b>B</b>) KD increased the number of susceptible profiles in female mice. Data are represented as the percentage of resilient and susceptible subjects within the KD and STD-fed groups. The light gray portion represents the percentage of Resilient subjects (score &gt; 1 on the SIT ratio), while the dark gray portion represents the percentage of susceptible subjects (score &lt; 1 on the SIT ratio).</p>
Full article ">Figure 6
<p>Drinking in the dark (DID) Test: (<b>A</b>) Female mice increase ethanol consumption with increased exposure time. Data are represented as the average of ethanol consumption per group during each test day. Vertical lines show ±SEM. # <span class="html-italic">p</span> &lt; 0.05 significant differences in ethanol consumption on the fourth day with respect to the other three days. (<b>B</b>) Susceptible profile altered the ethanol consumption during the DID test and KD blocks this effect. The bars show the average total ethanol consumption per group. Vertical lines show ±SEM. * <span class="html-italic">p</span> &lt; 0.05 significant differences with respect to non-stressed and VSD-Resilient females fed on STD. + <span class="html-italic">p</span> &lt; 0.05 significant differences with respect to VSD-Susceptible females fed on KD.</p>
Full article ">Figure 7
<p>Oral ethanol self-administration (SA) procedure: (<b>A</b>) Susceptible profile and KD affected the number of effective responses during the FR1. Data are represented as the number of effective responses per group and vertical lines show ±SEM. * <span class="html-italic">p</span> &lt; 0.05 significant differences in the STD-VSD-S group with respect to the rest of the groups. (<b>B</b>) VSD and KD affected ethanol consumption during FR1. Data are represented as the average of ethanol consumption per group and vertical lines show ±SEM. ** <span class="html-italic">p</span> &lt; 0.01 significant differences in STD-VSD (Resilient and Susceptible) group with respect to the rest of the groups. (<b>C</b>) VSD and KD altered the Breaking Point Value. Data are represented as the average of BP value per group and vertical lines show ±SEM. ** <span class="html-italic">p</span> &lt; 0.01 significant differences with respect to non-stressed females fed on STD. +++ <span class="html-italic">p</span> &lt; 0.001 significant differences with respect to the KD-VSD-S group.</p>
Full article ">Figure 8
<p>Gene expression in the striatum. The bars represent the mRNA expression levels and the vertical lines show ±SEM. *** <span class="html-italic">p</span> &lt; 0.001; ** <span class="html-italic">p</span> &lt; 0.01; * <span class="html-italic">p</span> &lt; 0.05 significant difference with respect to the STD-EXP group. +++ <span class="html-italic">p</span> &lt; 0.001; ++ <span class="html-italic">p</span> &lt; 0.01; + <span class="html-italic">p</span> &lt; 0.05 significant difference with respect to the KD-VSD group. ## <span class="html-italic">p</span> &lt; 0.01; # <span class="html-italic">p</span> &lt; 0.05 significant difference with respect to STD groups.</p>
Full article ">Figure 9
<p>Gene expression in the hippocampus. The bars represent the mRNA expression levels and the vertical lines show ±SEM. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 significant difference with respect to the STD-EXP group. ++ <span class="html-italic">p</span> &lt; 0.01 significant differences with respect to the KD-VSD group. @ <span class="html-italic">p</span> &lt; 0.05 significant difference with respect to the STD-VSD group. &amp; <span class="html-italic">p</span> &lt; 0.05 significant difference with respect to KD-EXP group. # <span class="html-italic">p</span> &lt; 0.05; ### <span class="html-italic">p</span> &lt; 0.001 significant differences with respect to STD groups. % <span class="html-italic">p</span> &lt; 0.05; %% <span class="html-italic">p</span> &lt; 0.01 significant differences with respect to non-stressed groups.</p>
Full article ">Figure 10
<p>Summary of the main results.</p>
Full article ">
27 pages, 1414 KiB  
Review
Beneficial Effects of the Ketogenic Diet on Nonalcoholic Fatty Liver Disease (NAFLD/MAFLD)
by Damian Dyńka, Łukasz Rodzeń, Mateusz Rodzeń, Dorota Łojko, Sebastian Kraszewski, Ali Ibrahim, Maria Hussey, Adam Deptuła, Żaneta Grzywacz, Alexandre Ternianov and David Unwin
J. Clin. Med. 2024, 13(16), 4857; https://doi.org/10.3390/jcm13164857 - 17 Aug 2024
Viewed by 12014
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) is likely to be approaching 38% of the world’s population. It is predicted to become worse and is the main cause of morbidity and mortality due to hepatic pathologies. It is particularly worrying that NAFLD [...] Read more.
The prevalence of nonalcoholic fatty liver disease (NAFLD) is likely to be approaching 38% of the world’s population. It is predicted to become worse and is the main cause of morbidity and mortality due to hepatic pathologies. It is particularly worrying that NAFLD is increasingly diagnosed in children and is closely related, among other conditions, to insulin resistance and metabolic syndrome. Against this background is the concern that the awareness of patients with NAFLD is low; in one study, almost 96% of adult patients with NAFLD in the USA were not aware of their disease. Thus, studies on the therapeutic tools used to treat NAFLD are extremely important. One promising treatment is a well-formulated ketogenic diet (KD). The aim of this paper is to present a review of the available publications and the current state of knowledge of the effect of the KD on NAFLD. This paper includes characteristics of the key factors (from the point of view of NAFLD regression), on which ketogenic diet exerts its effects, i.e., reduction in insulin resistance and body weight, elimination of fructose and monosaccharides, limitation of the total carbohydrate intake, anti-inflammatory ketosis state, or modulation of gut microbiome and metabolome. In the context of the evidence for the effectiveness of the KD in the regression of NAFLD, this paper also suggests the important role of taking responsibility for one’s own health through increasing self-monitoring and self-education. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Potential mechanisms of action of the ketogenic diet in NAFLD (MAFLD). The above figure was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 8 August 2024. Agreement number: CR275R2WWY.</p>
Full article ">Figure 2
<p>Effect of ketogenic diet on NAFLD (MAFLD) in humans. The above figure was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 8 August 2024. Agreement number: KZ275R2EJK.</p>
Full article ">
15 pages, 2684 KiB  
Article
Sex- and Age-Specific Differences in Mice Fed a Ketogenic Diet
by Kenyon W. Sprankle, Mya A. Knappenberger, Erica J. Locke, Jack H. Thompson, Madison F. Vinovrski, Kaylin Knapsack and Stephen C. Kolwicz
Nutrients 2024, 16(16), 2731; https://doi.org/10.3390/nu16162731 - 16 Aug 2024
Viewed by 878
Abstract
The ketogenic diet (KD) is a high-fat, low-carbohydrate diet that results in the elevation of serum ketone bodies, known as ketosis. This metabolic consequence has been suggested as a method for treating neurological conditions, improving exercise performance, and facilitating weight loss for overweight [...] Read more.
The ketogenic diet (KD) is a high-fat, low-carbohydrate diet that results in the elevation of serum ketone bodies, known as ketosis. This metabolic consequence has been suggested as a method for treating neurological conditions, improving exercise performance, and facilitating weight loss for overweight individuals. However, since most research primarily uses male populations, little is known about the potential sex differences during the consumption of the KD. In addition, the effects of the KD on aging are relatively unexplored. Therefore, the purpose of this study was to explore sex- and age-specific differences in mice fed the KD. Male and female C57BL/6N mice at either 12 wks or 24 wks of age were randomly assigned to a KD (90% fat, 1% carbohydrate) or chow (13% fat, 60% carbohydrate) group for 6 wks. KD induced weight gain, increased adiposity, induced hyperlipidemia, caused lipid accumulation in the heart and liver, and led to glycogen depletion in the heart, liver, and muscle with varying degrees of changes depending on age and sex. While younger and older male mice on the KD were prone to glucose intolerance, the KD acutely improved rotarod performance in younger females. Overall, this study highlights potential sex and aging differences in the adaptation to the KD. Full article
(This article belongs to the Special Issue Clinical Impact of Ketogenic Diet)
Show Figures

Figure 1

Figure 1
<p>Changes in anthropometric measures due to ketogenic diet. (<b>A</b>) Weekly percent body mass change from baseline (BL) in male mice. (<b>B</b>) Weekly percent body mass change from BL in female mice. (<b>C</b>) Average daily food intake of chow and ketogenic diets in male and female mice. (<b>D</b>) Average daily caloric intake of chow and ketogenic diets in male and female mice. (<b>E</b>) Adipose tissue mass normalized to tibia length (TL) in mice. (<b>F</b>) Heart weight to TL ratios in mice. (<b>G</b>) Quadriceps to TL ratio in mice. CD-12, mice fed the chow diet starting at 12 weeks of age, <span class="html-italic">n</span> = 9–10. CD-24, mice fed the chow diet starting at 24 weeks of age, <span class="html-italic">n</span> = 13–14. KD-12, mice fed the ketogenic diet starting at 12 weeks of age, <span class="html-italic">n</span> = 11 in each group. KD-24, mice fed the ketogenic diet starting at 24 weeks of age, <span class="html-italic">n</span> = 17–18. * <span class="html-italic">p</span> &lt; 0.05 vs. CD-12 or CD-24, # <span class="html-italic">p</span> &lt; 0.05 vs. KD-12 or KD-24.</p>
Full article ">Figure 2
<p>Ketogenic diet causes ketosis and hypercholesterolemia. β-hydroxybutyrate (β-OHB) was measured in blood in (<b>A</b>) male and (<b>B</b>) female mice at baseline (BL) and at 2-week intervals during the dietary feeding period. (<b>C</b>) β-hydroxybutyrate (β-OHB) measured in blood from male and female mice using a handheld meter prior to tissue harvest. (<b>D</b>) Cholesterol, (<b>E</b>) triglycerides, and (<b>F</b>) non-esterified fatty acids (NEFA) measured in the serum from blood collected at the time of tissue harvest. CD-12, mice fed the chow diet starting at 12 weeks of age, <span class="html-italic">n</span> = 5–10. CD-24, mice fed the chow diet starting at 24 weeks of age, <span class="html-italic">n</span> = 5–14. KD-12, mice fed the ketogenic diet starting at 12 weeks of age, <span class="html-italic">n</span> = 6–11. KD-24, mice fed the ketogenic diet starting at 24 weeks of age, <span class="html-italic">n</span> = 7–17. * <span class="html-italic">p</span> &lt; 0.05 vs. CD-12 or CD-24, # <span class="html-italic">p</span> &lt; 0.05 vs. KD-12.</p>
Full article ">Figure 3
<p>Glucose intolerance in male mice fed a ketogenic diet. (<b>A</b>) The glucose tolerance test performed in male mice. (<b>B</b>) The glucose tolerance test performed in female mice. (<b>C</b>) The area under the curve analysis of the glucose tolerance test in male mice. (<b>D</b>) The area under the curve analysis of the glucose tolerance test in female mice. (<b>E</b>) The blood glucose levels assessed by glucometer in mice prior to tissue harvest. (<b>F</b>) The serum insulin levels in mice from blood collected at the time of tissue harvest. CD-12, mice fed the chow diet starting at 12 weeks of age, <span class="html-italic">n</span> = 5–10. CD-24, mice fed the chow diet starting at 24 weeks of age, <span class="html-italic">n</span> = 6–10. KD-12, mice fed the ketogenic diet starting at 12 weeks of age, <span class="html-italic">n</span> = 7–10. KD-24, mice fed the ketogenic diet starting at 24 weeks of age, <span class="html-italic">n</span> = 7–10. * <span class="html-italic">p</span> &lt; 0.05 vs. CD-12 or CD-24, # <span class="html-italic">p</span> &lt; 0.05 vs. KD-12.</p>
Full article ">Figure 4
<p>Lipid accumulation and glycogen depletion in the heart, liver, and muscle. Triglyceride content measured in extracts from the (<b>A</b>) heart; (<b>B</b>) liver; and (<b>C</b>) quadriceps muscle of male and female mice fed the ketogenic diet for 6 weeks. Glycogen content measured in extracts from the (<b>D</b>) heart; (<b>E</b>) liver; and (<b>F</b>) quadriceps muscle of male and female mice fed the ketogenic diet for 6 weeks. CD-12, mice fed the chow diet starting at 12 weeks of age, <span class="html-italic">n</span> = 9–11. CD-24, mice fed the chow diet starting at 24 weeks of age, <span class="html-italic">n</span> = 7–10. KD-12, mice fed the ketogenic diet starting at 12 weeks of age, <span class="html-italic">n</span> = 8–11. KD-24, mice fed the ketogenic diet starting at 24 weeks of age, <span class="html-italic">n</span> = 5–12. * <span class="html-italic">p</span> &lt; 0.05 vs. CD-12 or CD-24 of same sex, # <span class="html-italic">p</span> &lt; 0.05 vs. KD-12 of same sex. <span>$</span> <span class="html-italic">p</span> &lt; 0.05 vs. male KD-24. ns = <span class="html-italic">p</span> = 0.105.</p>
Full article ">Figure 5
<p>Behavioral analysis of mice on a ketogenic diet. (<b>A</b>) Rotarod performance in cohorts of male mice taken at baseline and every 2 weeks. (<b>B</b>) Rotarod performance in cohorts of female mice taken at baseline and every 2 weeks. (<b>C</b>) Average number of seconds spent in the border zone during open field testing. (<b>D</b>) Average distance traveled by mice measured during open field testing. (<b>E</b>) Average velocity of male and female cohorts during open field testing. CD-12, mice fed the chow diet starting at 12 weeks of age, <span class="html-italic">n</span> = 5 per group. CD-24, mice fed the chow diet starting at 24 weeks of age, <span class="html-italic">n</span> = 5–8. KD-12, mice fed the ketogenic diet starting at 12 weeks of age, <span class="html-italic">n</span> = 6–7. KD-24, mice fed the ketogenic diet starting at 24 weeks of age, <span class="html-italic">n</span> = 5–9. * <span class="html-italic">p</span> &lt; 0.05 vs. CD-12 or CD-24 of same sex, # <span class="html-italic">p</span> &lt; 0.05 vs. KD-12 of same sex. <span>$</span> <span class="html-italic">p</span> &lt; 0.05 vs. male KD-24.</p>
Full article ">Figure 6
<p>Serum testosterone and estradiol concentrations after the ketogenic diet. (<b>A</b>) The serum testosterone concentrations measured in male chow (CD)- or ketogenic diet (KD)-fed mice. (<b>B</b>) The serum estradiol concentrations measured in female chow (CD)- or ketogenic diet (KD)-fed mice. CD-12, mice fed the chow diet starting at 12 weeks of age, <span class="html-italic">n</span> = 7–9. CD-24, mice fed the chow diet starting at 24 weeks of age, <span class="html-italic">n</span> = 6–7. KD-12, mice fed the ketogenic diet starting at 12 weeks of age, <span class="html-italic">n</span> = 7–8. KD-24, mice fed the ketogenic diet starting at 24 weeks of age, <span class="html-italic">n</span> = 6–8. * <span class="html-italic">p</span> &lt; 0.05 vs. CD-12.</p>
Full article ">
17 pages, 1016 KiB  
Review
Ketogenic Interventions in Autosomal Dominant Polycystic Kidney Disease: A Comprehensive Review of Current Evidence
by Carla Pezzuoli, Giuseppe Biagini and Riccardo Magistroni
Nutrients 2024, 16(16), 2676; https://doi.org/10.3390/nu16162676 - 13 Aug 2024
Viewed by 1562
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a genetic disorder characterized by the development and enlargement of multiple kidney cysts, leading to progressive kidney function decline. To date, Tolvaptan, the only approved treatment for this condition, is able to slow down the loss [...] Read more.
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a genetic disorder characterized by the development and enlargement of multiple kidney cysts, leading to progressive kidney function decline. To date, Tolvaptan, the only approved treatment for this condition, is able to slow down the loss of annual kidney function without stopping the progression of the disease. Furthermore, this therapy is approved only for patients with rapid disease progression and its compliance is problematic because of the drug’s impact on quality of life. The recent literature suggests that cystic cells are subject to several metabolic dysregulations, particularly in the glucose pathway, and mitochondrial abnormalities, leading to decreased oxidative phosphorylation and impaired fatty acid oxidation. This finding paved the way for new lines of research targeting potential therapeutic interventions for ADPKD. In particular, this review highlights the latest studies on the use of ketosis, through ketogenic dietary interventions (daily calorie restriction, intermittent fasting, time-restricted feeding, ketogenic diets, and exogenous ketosis), as a potential strategy for patients with ADPKD, and the possible involvement of microbiota in the ketogenic interventions’ effect. Full article
Show Figures

Figure 1

Figure 1
<p>Metabolic pathways in normal and highly proliferative cells under different conditions. The image illustrates the different metabolic pathways of normal differentiated cells and highly proliferative cells under three conditions: oxidative phosphorylation—OXPHOS (in the presence of oxygen), anaerobic glycolysis (in the absence of oxygen), and ketosis. In normal cells in the presence of oxygen, glucose is metabolized through glycolysis, producing pyruvate, which enters the mitochondria to be converted into acetyl-CoA, initiating the tricarboxylic acid (TCA) cycle and ATP production via oxidative phosphorylation, yielding around 30–32 molecules of ATP per molecule of glucose. In the absence of oxygen, pyruvate is converted into lactate, yielding two molecules of ATP per molecule of glucose. During ketosis, the scarcity of glucose and the presence of ketone bodies lead to a suppression of glycolysis, with acetyl-CoA derived from ketone bodies entering the TCA cycle to produce around 20 ATP molecules. In contrast, in highly proliferative cells, even in the presence of oxygen, pyruvate is preferentially converted into lactate (aerobic glycolysis or Warburg Effect), which yields much less energy (approximately four molecules of ATP per molecule of glucose) compared to OXPHOS. Under ketosis conditions, these cells show suppression of both glycolysis and OXPHOS, highlighting the distinctive metabolic phenotype of highly proliferative cells compared to normal cells. OXPHOS: oxidative phosphorylation; ATP: adenosine triphosphate; acetyl-CoA: acetyl coenzyme A; and TCA: tricarboxylic acid cycle.</p>
Full article ">
17 pages, 9976 KiB  
Article
Nifedipine Improves the Ketogenic Diet Effect on Insulin-Resistance-Induced Cognitive Dysfunction in Rats
by Nancy M. Abdel-Kareem, Shimaa M. Elshazly, May A. Abd El Fattah, Afaf A. Aldahish, Sawsan A. Zaitone, Sahar K. Ali and Enas A. Abd El-Haleim
Pharmaceuticals 2024, 17(8), 1054; https://doi.org/10.3390/ph17081054 - 10 Aug 2024
Viewed by 643
Abstract
Insulin resistance, induced by high fructose consumption, affects cognitive function negatively. Nifedipine may be suggested for neurological disorders. This study aimed to assess the effect of nifedipine with either a normal diet (ND) or a ketogenic diet (KD) in cognitive dysfunction. Male Wistar [...] Read more.
Insulin resistance, induced by high fructose consumption, affects cognitive function negatively. Nifedipine may be suggested for neurological disorders. This study aimed to assess the effect of nifedipine with either a normal diet (ND) or a ketogenic diet (KD) in cognitive dysfunction. Male Wistar rats received 10% fructose in drinking water for 8 weeks to induce insulin resistance. Rats received nifedipine (5.2 mg/kg/day; p.o.) later with ND or KD for an additional five weeks. One and two-way ANOVAs were used in analyzing the data. Reversion to the ND improved insulin resistance and lipid profile, besides brain-derived neurotrophic factor (BDNF), glycogen synthase kinase-3 beta (GSK3β), and insulin-degrading enzyme (IDE) levels. Rats fed KD alone and those that received nifedipine with KD did not show similar improvement in the previously mentioned parameters as the ND group. However, nifedipine-ND rats showed improvement in cognitive behavior and insulin resistance. Treatment with nifedipine-KD ameliorated GSK3β, amyloid β (Aβ), and tau protein levels. As the nifedipine-KD combination succeeded in diminishing the accumulated Aβ and tau protein, KD may be used for a while due to its side effects, then nifedipine treatment could be continued with an ND. This conclusion is based on the finding that this combination mitigated insulin resistance with the associated improved behavior. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of the normal diet (ND) and the ketogenic diet (KD) with or without nifedipine (5.2 mg/kg/day, p.o.) on the body weight difference, Final weight of IR at the end of the experiment— their weights after 8 weeks’ treatment with 10% fructose in insulin-resistant (IR) rats. Data are represented as median and interquartile percent (25–75%) and analyzed using one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet; and + <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND + nifedipine group.</p>
Full article ">Figure 2
<p>Effect of 10% fructose treatment after 8 weeks on the oral glucose tolerance test (OGTT). Blood glucose levels were evaluated before glucose administration and after administration of 2.5 mg/kg glucose at 30, 60, and 90 min. Results are represented as the median and interquartile range (min. to max.) (<span class="html-italic">n</span> = 6) and analyzed using two-way ANOVA followed by the Bonferroni test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats.</p>
Full article ">Figure 3
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on the behavioral response in IR rats. Results are represented as median and interquartile range (min. to max.) (<span class="html-italic">n</span> = 6) and analyzed using two-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats.</p>
Full article ">Figure 4
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on serum lipid profile: (<b>a</b>) Cholesterol, (<b>b</b>) triglycerides, (<b>c</b>) high-density lipoprotein (HDL), and (<b>d</b>) low-density lipoprotein (LDL) (<b>d</b>) in (IR) rats. Results are represented as median and interquartile range (min. to max. with all points) (<span class="html-italic">n</span> = 6) and analyzed using one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; and @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet.</p>
Full article ">Figure 5
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on the brain-derived neurotrophic factor (BDNF) in the brain hippocampus of IR rats. Results are represented as median and interquartile range (25–75%) (<span class="html-italic">n</span> = 6) and analyzed using one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats.</p>
Full article ">Figure 6
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on (<b>a</b>) glycogen synthase kinase-3 beta (GSK3β) and (<b>b</b>) insulin-degrading enzyme (IDE) in the brain hippocampus of IR rats. Results are represented as median and interquartile range (25–75%) (<span class="html-italic">n</span> = 6) and analyzed using one-way ANOVA followed by the Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet; and + <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND + nifedipine group.</p>
Full article ">Figure 7
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on histopathological examinations of insulin-resistant rats. Photomicrographs of hematoxylin-and-eosin-stained (<b>a</b>): for cerebral cortex and (<b>b</b>): for hippocampus slices from various groups. (<b>A</b>) Brain tissue that is uniform and without any neuronal damage. Grade 0 for both tissues. (<b>B</b>) The perineuronal edema and neurons with pyknotic nuclei (black arrows), Rosenthal fibers may be seen in gliosis (black arrowheads) regions, and the red arrows represent red neurons for both cerebral cortex and hippocampus, 75% of brain tissue. Grade 3 for both tissues. (<b>C</b>) Red arrows indicate that there are a few scattered red neurons, for both the cerebral cortex and hippocampus. For the cerebral cortex only, tissue edema (red arrowheads) is seen. The red neurons are sparsely distributed (red arrow). Perineuronal edema (black arrows), tissue edema (red arrowheads), and gliosis (black arrowheads) are all seen, 25% of brain tissue. Grade 2 for both tissues. (<b>D</b>) The red neurons are sparsely distributed (red arrow). Perineuronal edema (black arrows), tissue edema (red arrowheads), and gliosis in cerebral cortex only (black arrowheads) are all seen in both tissues, 45% of brain tissue. Grade 3 for cerebral cortex and 15% of brain tissue for hippocampus. Grade 2. (<b>E</b>) Neurons in cerebral cortex and hippocampus with pyknotic nuclei and perineuronal edema are seen (black arrows), 10% of brain tissue. Grade 1 for both tissues. There are gliosis regions with Rosenthal fibers in cerebral cortex only (black arrowheads). There are red neurons (shown by red arrows). Pictures captured at magnification power ×40 for cerebral cortex and ×20 for hippocampus.</p>
Full article ">Figure 8
<p>(<b>a</b>,<b>b</b>) (<b>A</b>) Weak focal levels of Aβ (black arrows). (<b>B</b>) Significant increase in levels of Aβ (black arrows). (<b>C</b>) Significant reduction in levels of Aβ (black arrows). (<b>D</b>) Significant increase in levels of Aβ (black arrows). (<b>E</b>) Weak focal levels of Aβ (black arrows). Pictures captured at magnification power ×40 for cerebral cortex and 20× for hippocampus. (<b>c</b>,<b>d</b>) (<b>A</b>) Weak levels of tau in the cytoplasm of a few neuron cells (black arrows). (<b>B</b>) There is an increase in the levels of tau in many neurons (black arrows). (<b>C</b>) There is a reduction in the levels of tau in many neurons (black arrows). (<b>D</b>) There is an increase in the levels of tau in many neurons (black arrows). (<b>E</b>) There are weak levels of tau in cytoplasm of some neurons (black arrows) in cerebral cortex while a significant increase in the levels of tau in cytoplasm of some neurons (black arrows). Pictures captured at magnification power ×40 for cerebral cortex and 20× for hippocampus. Statistical analysis showed the effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on immunohistochemical staining of cerebral cortex and hippocampal levels of amyloid β (Aβ) (<b>e</b>,<b>f</b>) and tau protein (<b>g</b>,<b>h</b>) of insulin-resistant rats. Results are represented as median and interquartile range (25–75%) (<span class="html-italic">n</span> = 6) and analyzed by one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet; and + <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND + nifedipine group.</p>
Full article ">Figure 8 Cont.
<p>(<b>a</b>,<b>b</b>) (<b>A</b>) Weak focal levels of Aβ (black arrows). (<b>B</b>) Significant increase in levels of Aβ (black arrows). (<b>C</b>) Significant reduction in levels of Aβ (black arrows). (<b>D</b>) Significant increase in levels of Aβ (black arrows). (<b>E</b>) Weak focal levels of Aβ (black arrows). Pictures captured at magnification power ×40 for cerebral cortex and 20× for hippocampus. (<b>c</b>,<b>d</b>) (<b>A</b>) Weak levels of tau in the cytoplasm of a few neuron cells (black arrows). (<b>B</b>) There is an increase in the levels of tau in many neurons (black arrows). (<b>C</b>) There is a reduction in the levels of tau in many neurons (black arrows). (<b>D</b>) There is an increase in the levels of tau in many neurons (black arrows). (<b>E</b>) There are weak levels of tau in cytoplasm of some neurons (black arrows) in cerebral cortex while a significant increase in the levels of tau in cytoplasm of some neurons (black arrows). Pictures captured at magnification power ×40 for cerebral cortex and 20× for hippocampus. Statistical analysis showed the effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on immunohistochemical staining of cerebral cortex and hippocampal levels of amyloid β (Aβ) (<b>e</b>,<b>f</b>) and tau protein (<b>g</b>,<b>h</b>) of insulin-resistant rats. Results are represented as median and interquartile range (25–75%) (<span class="html-italic">n</span> = 6) and analyzed by one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet; and + <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND + nifedipine group.</p>
Full article ">Figure 9
<p>A schematic presentation for the results of the study.</p>
Full article ">
19 pages, 1341 KiB  
Review
The Impacts of Dietary Intervention on Brain Metabolism and Neurological Disorders: A Narrative Review
by Priya Rathor and Ratnasekhar Ch
Dietetics 2024, 3(3), 289-307; https://doi.org/10.3390/dietetics3030023 - 9 Aug 2024
Viewed by 988
Abstract
Neurological disorders are increasing globally due to their complex nature, influenced by genetics and environmental factors. Effective treatments remain limited, and early diagnosis is challenging. Recent evidence indicates that metabolic activities play a crucial role in the onset of neural defects. Molecular changes [...] Read more.
Neurological disorders are increasing globally due to their complex nature, influenced by genetics and environmental factors. Effective treatments remain limited, and early diagnosis is challenging. Recent evidence indicates that metabolic activities play a crucial role in the onset of neural defects. Molecular changes offer new diagnostic markers and dietary targets for disease management. Diets such as MIND, DASH, omega-3 fatty acids, and polyphenols show promise in protecting brain metabolism through their anti-inflammatory properties. Personalized dietary interventions could mitigate neurodegenerative diseases. This review highlights the effects of various dietary interventions, including calorie restriction, fasting, and ketogenic diets, on neurological disorders. Additionally, it emphasizes the nutritional impacts on immunomodulation and the underlying mechanisms, including the influence of gut microbiota on brain function. Dietary interventions could serve as adjunctive therapies in disease management. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of different diets on neurological disease.</p>
Full article ">Figure 2
<p>Examples of macronutrients and micronutrients.</p>
Full article ">Figure 3
<p>Different dietary intervention controls the brain pathways.</p>
Full article ">
22 pages, 704 KiB  
Review
Potential Add-On Benefits of Dietary Intervention in the Treatment of Autosomal Dominant Polycystic Kidney Disease
by Erica Rosati, Giulia Condello, Chiara Tacente, Ilaria Mariani, Valeria Tommolini, Luca Calvaruso, Pierluigi Fulignati, Giuseppe Grandaliano and Francesco Pesce
Nutrients 2024, 16(16), 2582; https://doi.org/10.3390/nu16162582 - 6 Aug 2024
Viewed by 1627
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of renal failure. The pathogenesis of the disease encompasses several pathways and metabolic alterations, including the hyperactivation of mTOR and suppression of AMPK signaling pathways, as well as mitochondrial dysfunction. This [...] Read more.
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of renal failure. The pathogenesis of the disease encompasses several pathways and metabolic alterations, including the hyperactivation of mTOR and suppression of AMPK signaling pathways, as well as mitochondrial dysfunction. This metabolic reprogramming makes epithelial cyst-lining cells highly dependent on glucose for energy and unable to oxidize fatty acids. Evidence suggests that high-carbohydrate diets may worsen the progression of ADPKD, providing the rationale for treating ADPKD patients with calorie restriction and, in particular, with ketogenic dietary interventions, already used for other purposes such as in overweight/obese patients or in the treatment of refractory epilepsy in children. Preclinical studies have demonstrated that calorie restriction may prevent and/or slow disease progression by inducing ketosis, particularly through increased beta-hydroxybutyrate (BHB) levels, which may modulate the metabolic signaling pathways altered in ADKPK. In these patients, although limited, ketogenic intervention studies have shown promising beneficial effects. However, larger and longer randomized controlled trials are needed to confirm their tolerability and safety in long-term maintenance and their additive role in the therapy of polycystic kidney disease. Full article
(This article belongs to the Special Issue Diet Management in Renal Diseases)
Show Figures

Figure 1

Figure 1
<p>Metabolic Reprogramming in ADPKD and the Impact of Ketosis. Metabolic alterations in ADPKD drive cystogenesis and cell proliferation. Dysregulation of the ERK pathway activates the mTOR cascade and increases intracellular levels of cAMP while inhibiting the AMPK pathway. These changes lead to increased glycolysis, higher ATP levels, and extracellular lactate accumulation. Altered mitochondrial function results in elevated production of ROS and reduced fatty acid β-oxidation, further promoting glycolysis. Additionally, systemic inflammation in ADPKD sustains disease progression. Inducing a state of ketosis may help restore metabolic balance and slow cyst formation and growth. During ketosis, reduced glucose availability forces cells to utilize ketone bodies, particularly BHB, and fatty acids as energy sources. This shift restores mitochondrial function and reduces ROS production. Furthermore, reduced glycolysis leads to proper activation of the ERK pathway, resulting in decreased mTOR activation and increased AMPK activity. ADPKD, Autosomal Dominant Polycystic Kidney Disease; mTORC1, mammalian Target of Rapamycin Complex 1; cAMP, cyclic Adenosine Monophosphate; AMPK, Adenosine Monophosphate-Activated Protein Kinase; FAO, fatty acid β-oxidation; BHB, beta-hydroxybutyrate; ROS, Reactive Oxygen Species.</p>
Full article ">
15 pages, 875 KiB  
Review
A Focus on Heart Failure Management through Diet and Nutrition: A Comprehensive Review
by Lee P. Liao, Anushriya Pant, Simone Marschner, Peter Talbot and Sarah Zaman
Hearts 2024, 5(3), 293-307; https://doi.org/10.3390/hearts5030022 - 29 Jul 2024
Viewed by 720
Abstract
There is emerging evidence to suggest that diet and dietary interventions can have an impact on heart failure (HF) outcomes. Currently, the restriction of salt intake is the only dietary advice that is consistently guideline-recommended for the management of HF despite conflicting evidence [...] Read more.
There is emerging evidence to suggest that diet and dietary interventions can have an impact on heart failure (HF) outcomes. Currently, the restriction of salt intake is the only dietary advice that is consistently guideline-recommended for the management of HF despite conflicting evidence for its efficacy. Dietary components that have been investigated in people with HF include middle-chain triglyceride (MCT) oil, beta-hydroxybutyrate (BHB) salts, ketone esters and coenzyme Q10 (CoQ10). Supplementation with these components is thought to be cardioprotective possibly due to an increase in myocardial energy production. There have been research studies on the effectiveness of The Dietary Approaches to Stop Hypertension (DASH) diet and the Mediterranean Diet (MedDiet) in the treatment of HF, but with conflicting results. The ketogenic diet (KD) has come to the forefront of interest due to evidence indicating its effectiveness in addressing the metabolic shift that occurs in HF. However, there is a lack of randomised controlled trials (RCT) centred around the KD. In any dietary intervention, factors such as adherence and compliance affect the validity of the results. Malnutrition, sarcopenia and/or cardiac cachexia can be present in the more advanced stages of heart failure. Nutritional screening, assessment and support/intervention are important aspects of treatment in the advanced stages of heart failure. Furthermore, HF management through dietary intervention is further complicated by the presence of comorbidities, such as diabetes mellitus (DM) and coronary artery disease (CAD). Long-term studies on the use of dietary modifications in people with HF are warranted to ascertain their efficacy, safety and side effects. Full article
Show Figures

Figure 1

Figure 1
<p>Current pharmacological, dietary patterns and dietary supplements that have been investigated in the management of HF.</p>
Full article ">
27 pages, 2977 KiB  
Systematic Review
Influence of Carbohydrate Intake on Caprylic Acid (C8:0)-Induced Ketogenesis—A Systematic Review and Meta-Analysis
by Marius Frenser, Tobias Fischer, Isabel Albrecht and Thorsten Marquardt
Nutrients 2024, 16(15), 2456; https://doi.org/10.3390/nu16152456 - 29 Jul 2024
Viewed by 1199
Abstract
The ketogenic diet is used worldwide to treat various diseases, especially drug-resistant epilepsies. Medium-chain triglycerides or medium-chain fatty acids, primarily the major ketogenic compound caprylic acid (C8; C8:0), can significantly support ketogenesis. This review examines the effects of concurrent carbohydrate intake on C8-induced [...] Read more.
The ketogenic diet is used worldwide to treat various diseases, especially drug-resistant epilepsies. Medium-chain triglycerides or medium-chain fatty acids, primarily the major ketogenic compound caprylic acid (C8; C8:0), can significantly support ketogenesis. This review examines the effects of concurrent carbohydrate intake on C8-induced ketogenesis. A systematic literature search (PubMed and Web of Science) with subsequent data extraction was performed according to PRISMA guidelines and the Cochrane Handbook. Studies investigating the metabolic response to C8-containing MCT interventions with carbohydrate intake were included. The studies did not include a ketogenic diet. Three intervention groups were created. The quality of the studies was assessed using the RoB II tool, and the meta-analysis was performed using the Cochrane RevMan software. A total of 7 trials, including 4 RCTs, met the inclusion criteria. Ketone production was lower when C8 was combined with carbohydrates compared to MCT intake alone. The lower C8 dose group (11 g) did not show a significantly lower ketogenic effect than the higher dose group (19 g). Forest plot analysis showed heterogeneous data. The data suggest a non-linear relationship between C8, carbohydrate intake and ketone production. Further studies are needed to investigate the influence of different carbohydrates on C8-induced ketogenesis. Limitations include heterogeneous intervention conditions, such as different types of dispersions, caffeine intake, limited number of studies and variability in study design. Full article
(This article belongs to the Section Nutrition and Metabolism)
Show Figures

Figure 1

Figure 1
<p>Modified flow chart of the literature search and selection based on the PRISMA flow diagram for systematic reviews [<a href="#B36-nutrients-16-02456" class="html-bibr">36</a>].</p>
Full article ">Figure 2
<p>Temporal comparison of MCT and meal interventions in the included trials [<a href="#B20-nutrients-16-02456" class="html-bibr">20</a>,<a href="#B21-nutrients-16-02456" class="html-bibr">21</a>,<a href="#B22-nutrients-16-02456" class="html-bibr">22</a>,<a href="#B24-nutrients-16-02456" class="html-bibr">24</a>,<a href="#B25-nutrients-16-02456" class="html-bibr">25</a>,<a href="#B42-nutrients-16-02456" class="html-bibr">42</a>,<a href="#B43-nutrients-16-02456" class="html-bibr">43</a>].</p>
Full article ">Figure 3
<p>Modified visualisation of the results of the risk of bias assessment using RoB II tools of the available studies [<a href="#B20-nutrients-16-02456" class="html-bibr">20</a>,<a href="#B21-nutrients-16-02456" class="html-bibr">21</a>,<a href="#B22-nutrients-16-02456" class="html-bibr">22</a>,<a href="#B24-nutrients-16-02456" class="html-bibr">24</a>,<a href="#B25-nutrients-16-02456" class="html-bibr">25</a>,<a href="#B42-nutrients-16-02456" class="html-bibr">42</a>,<a href="#B43-nutrients-16-02456" class="html-bibr">43</a>,<a href="#B45-nutrients-16-02456" class="html-bibr">45</a>].</p>
Full article ">Figure 4
<p>The course of total ketones (ßHB + AcAc) (mean values + standard deviation, only positive values shown here) (<b>A</b>) (groups 2 and 3) and of ßHB (group 1) of the three intervention groups. The number of studies (n) of the individual measurement points: group 1: t<sub>0</sub> = 3, t<sub>0,5</sub> = 3, t<sub>1</sub> = 3, t<sub>2</sub> = 3, t<sub>3</sub> = 3, t<sub>4</sub> = 3, t<sub>5</sub> = 2, t<sub>6</sub> = 1; group 2: t<sub>0</sub> = 4, t<sub>0,5</sub> = 4, t<sub>1</sub> = 4, t<sub>2</sub> = 4, t<sub>3</sub> = 4, t<sub>4</sub> = 4, t<sub>5</sub> = 3, t<sub>6</sub> = 3, t<sub>7</sub> = 3, T<sub>8</sub> = 3; group 3: t<sub>0</sub> = 6, t<sub>0,5</sub> = 6, t<sub>1</sub> = 6, t<sub>2</sub> = 6, t<sub>3</sub> = 6, t<sub>4</sub> = 6, t<sub>5</sub> = 3, t<sub>6</sub> = 2, t<sub>7</sub> = 2, T<sub>8</sub> = 2 (<b>B</b>) of group 2 (values multiplied by a factor of 1.87 to achieve a uniform kcal/MCT density of 5.36 between groups 2 and 3) and of group 3 (<b>C</b>) of the individual studies from group 2 (<b>D</b>) of the individual studies from group 3 <sup>1</sup> [<a href="#B20-nutrients-16-02456" class="html-bibr">20</a>,<a href="#B21-nutrients-16-02456" class="html-bibr">21</a>,<a href="#B22-nutrients-16-02456" class="html-bibr">22</a>,<a href="#B24-nutrients-16-02456" class="html-bibr">24</a>,<a href="#B25-nutrients-16-02456" class="html-bibr">25</a>,<a href="#B42-nutrients-16-02456" class="html-bibr">42</a>,<a href="#B43-nutrients-16-02456" class="html-bibr">43</a>]. <sup>1</sup> Norgren et al. (2020) [<a href="#B20-nutrients-16-02456" class="html-bibr">20</a>]: ßHB instead of total ketones.</p>
Full article ">Figure 5
<p>Forest plots for the change in total ketones (ßHB and AcAc) compared to t<sub>0</sub> after (<b>A</b>) 30 min (t<sub>0.5</sub>; group 2), (<b>B</b>) 120 min (t<sub>2</sub>; group 2), (<b>C</b>) 240 min (t<sub>4</sub>; group 2), (<b>D</b>) 30 min (t<sub>0.5</sub>; group 3), (<b>E</b>) 120 min (t<sub>2</sub>; group 3) and (<b>F</b>) 240 min (t<sub>4</sub>; group 3) [<a href="#B24-nutrients-16-02456" class="html-bibr">24</a>,<a href="#B25-nutrients-16-02456" class="html-bibr">25</a>,<a href="#B42-nutrients-16-02456" class="html-bibr">42</a>,<a href="#B43-nutrients-16-02456" class="html-bibr">43</a>]. Figure created with Cochrane RevMan software (<a href="https://revman.cochrane.org/info" target="_blank">https://revman.cochrane.org/info</a> accessed on 21 February 2024).</p>
Full article ">Figure 5 Cont.
<p>Forest plots for the change in total ketones (ßHB and AcAc) compared to t<sub>0</sub> after (<b>A</b>) 30 min (t<sub>0.5</sub>; group 2), (<b>B</b>) 120 min (t<sub>2</sub>; group 2), (<b>C</b>) 240 min (t<sub>4</sub>; group 2), (<b>D</b>) 30 min (t<sub>0.5</sub>; group 3), (<b>E</b>) 120 min (t<sub>2</sub>; group 3) and (<b>F</b>) 240 min (t<sub>4</sub>; group 3) [<a href="#B24-nutrients-16-02456" class="html-bibr">24</a>,<a href="#B25-nutrients-16-02456" class="html-bibr">25</a>,<a href="#B42-nutrients-16-02456" class="html-bibr">42</a>,<a href="#B43-nutrients-16-02456" class="html-bibr">43</a>]. Figure created with Cochrane RevMan software (<a href="https://revman.cochrane.org/info" target="_blank">https://revman.cochrane.org/info</a> accessed on 21 February 2024).</p>
Full article ">Figure 6
<p>The course of the C8 and C10 concentrations in plasma based on the analysis of the total plasma lipids of groups 2 and 3 (mean values ± standard deviation). The number of studies (n) of the individual measurement time points: group 2 (C8 and C10): t<sub>0</sub> = 3, t<sub>0,5</sub> = 3, t<sub>1</sub> = 3, t<sub>2</sub> = 3, t<sub>3</sub> = 3, t<sub>4</sub> = 3, t<sub>5</sub> = 1, t<sub>6</sub> = 1, t<sub>7</sub> = 1, t<sub>8</sub> = 1; group 3 (C8): t<sub>0</sub> = 3, t<sub>0,5</sub> = 3, t<sub>1</sub> = 3, t<sub>2</sub> = 3, t<sub>3</sub> = 3, t<sub>4</sub> = 3, t<sub>5</sub> = 1, t<sub>6</sub> = 1, t<sub>7</sub> = 1, T<sub>8</sub> = 1; group 3 (C10): t<sub>0</sub> = 2, t<sub>0,5</sub> = 2, t<sub>1</sub> = 2, t<sub>2</sub> = 2, t<sub>3</sub> = 2, t<sub>4</sub> = 2.</p>
Full article ">
18 pages, 6366 KiB  
Article
In Silico Screening Identification of Fatty Acids and Fatty Acid Derivatives with Antiseizure Activity: In Vitro and In Vivo Validation
by Emilia Mercedes Barrionuevo, Estefanía Peralta, Agustín Manzur De Nardi, Juliana Monat, Maximiliano José Fallico, Manuel Augusto Llanos, Luciana Gavernet, Emilio Román Mustafá, Pedro Martin and Alan Talevi
Pharmaceutics 2024, 16(8), 996; https://doi.org/10.3390/pharmaceutics16080996 - 27 Jul 2024
Viewed by 708
Abstract
High fat diets have been used as complementary treatments for seizure disorders for more than a century. Moreover, many fatty acids and derivatives, including the broad-spectrum antiseizure medication valproic acid, have been explored and used as pharmacological agents to treat epilepsy. In this [...] Read more.
High fat diets have been used as complementary treatments for seizure disorders for more than a century. Moreover, many fatty acids and derivatives, including the broad-spectrum antiseizure medication valproic acid, have been explored and used as pharmacological agents to treat epilepsy. In this work, we have explored the anticonvulsant potential of a large library of fatty acids and fatty acid derivatives, the LIPID MAPS Structure Database, using structure-based virtual screening to assess their ability to block the voltage-gated sodium channel 1.2 (NaV1.2), a validated target for antiseizure medications. Four of the resulting in silico hits were submitted for experimental confirmation using in vitro patch clamp experiments, and their protective role was evaluated in an acute mice seizure model, the Maximal Electroshock seizure model. These four compounds were found to protect mice against seizures. Two of them exhibited blocking effects on NaV1.2, CaV2.2, and CaV3.1. Full article
(This article belongs to the Special Issue Computer-Aided Development: Recent Advances and Expectations)
Show Figures

Figure 1

Figure 1
<p>Docking poses vs. hNaV1.2 of the selected in silico hits submitted to in vivo and in vitro assays. The top scoring pose for each ligand is shown. Residues constituting the DEKA ring are represented in pink, while residues F1118 (FS6) and Y1125 from the Ab site are depicted in yellow. Compounds were selected based on the ligand’s efficiency, visual inspection of the poses, and favorable drug-like properties: 5-hexenoic acid (orange), 2-aminooctanoic acid (green), 9-phenylnonanoic acid (blue), and 9-hydroxydecanoic acid (violet). (<b>A</b>) Front view of hNaV1.2 along with the top scoring docking poses for the selected compounds. (<b>B</b>) 5-hexenoic acid’s highest score docking pose showing a 2.7 Å hydrogen bond with SER815 and a 3.7 Å minimal distance to FS6. (<b>C</b>) 2-aminooctanoic acid’s highest score docking pose showing a 1.9 Å hydrogen bond with SER815 and a 3.6 Å minimal distance to FS6. (<b>D</b>) 9-phenylnonanoic acid’s highest score docking pose showing a 3.6 Å pi-interaction with PHE535, a 3.0 Å hydrogen bond with TYR1125, and a 4.9 Å minimal distance to FS6. (<b>E</b>) 9-hydroxydecanoic acid’s highest score docking pose showing a 2.7 Å hydrogen bond with SER815, a 2.2 Å hydrogen bond to SER1117, and a 3.6 Å minimal distance to FS6.</p>
Full article ">Figure 2
<p>NaV1.1 current inhibition. Representative traces (<b>top</b>) and time courses (<b>middle</b>) of normalized NaV1.1 current (INaV1.1) from HEK293 cells with stable expression of NaV1.1 channels in control condition and 5-hexanoic acid (100 µM) application (n = 5, panel (<b>A</b>)); 2-aminooctanoic acid (100 µM) application (n = 7, panel (<b>B</b>)); 9-hydroxydecanoic acid (100 µM) application (n = 8, panel (<b>C</b>)); or 9-phenylnonanoic acid (100 µM) application (n = 7, panel (<b>D</b>)). Bars (<b>bottom</b>) represent normalized INaV1.1 in control condition and with each compound. Statistical significance was evaluated by one-sample <span class="html-italic">t</span>-test against 1.</p>
Full article ">Figure 3
<p>NaV1.2 current inhibition. Representative traces (<b>top</b>) and time courses (<b>middle</b>) of normalized NaV1.2 current (INaV1.2) from HEK293 cells with stable expression of NaV1.2 channels in control condition and 5-hexanoic acid (100 µM) application (n = 7, panel (<b>A</b>)); 2-aminooctanoic acid (100 µM) application (n = 7, panel (<b>B</b>)); 9-hydroxydecanoic acid (100 µM) application (n = 7, panel (<b>C</b>)); or 9-phenylnonanoicacid (100 µM) application (n = 10, panel (<b>D</b>)). Bars (<b>bottom</b>) represent normalized INaV1.2 in control condition and with each compound. Statistical significance was evaluated by one-sample <span class="html-italic">t</span>-test against 1.</p>
Full article ">Figure 4
<p>Mechanism of NaV channel inhibition by fatty acids. (<b>A</b>). Typical traces of human NaV1.1 (<b>left</b>) and NaV1.2 (<b>right</b>) currents evoked by the steady-state inactivation protocol under control conditions and after the application of 100 μM 9-hydroxydecanoic acid. (<b>B</b>). Mean h-curves obtained for control conditions and after stable effect of 100 µM 5-hexanoic acid (5-HEX, orange, NaV1.1: n = 5, NaV1.2: n = 5), 2-aminooctanoic acid (2-AO, green, NaV1.1: n = 7, NaV1.2: n = 5), 9-hydroxydecanoic acid (9-HiD, violet, NaV1.1: n = 8, NaV1.2: n = 7), and 9-phenylnonanoic acid (9-PhN, blue, NaV1.1: n = 6, NaV1.2: n = 10). (<b>C</b>). Mean change in half-maximal voltage inactivation (ΔV1/2) values obtained from the h-curves calculated for each fatty acid. # indicates statistically significant differences between ΔV1/2 values and 0 mV (one-sample <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05); * indicates statistically significant differences between isoforms in the obtained ΔV1/2 values (unpaired <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05). The slope parameter remained unchanged in all the treatments. (<b>D</b>). Mean voltage-independent inhibition, observed as a reduction in the Na+ current elicited after a pre-conditioning pulse of −130 mV (Imax) obtained after fatty acid treatment and normalized to control current value. # indicates statistically significant differences between Imax values and 1 (one-sample <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05); * indicates statistically significant differences between isoforms in the obtained Imax values (unpaired <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>CaV3.1 current inhibition. Representative traces (<b>top</b>) and time courses (<b>middle</b>) of normalized CaV3.1 current (ICaV3.1) from HEK293T cells transfected with CaV3.1 in control condition and 5-hexanoic acid (100 µM) application (n = 4, panel (<b>A</b>)); 2-aminooctanoic acid (100 µM) application (n = 5, panel (<b>B</b>)); 9-hydroxydecanoic acid (100 µM) application (n = 5, panel (<b>C</b>)); or 9-phenylnonanoic acid (100 µM) application (n = 5, panel (<b>D</b>)). Bars (<b>bottom</b>) represent normalized ICaV3.1 in control condition and with each compound. Statistical significance was evaluated by one-sample <span class="html-italic">t</span>-test against 1.</p>
Full article ">Figure 6
<p>CaV2.2 current inhibition. Representative traces (<b>top</b>) and time courses (<b>middle</b>) of normalized CaV2.2 current (ICaV2.2) from HEK293T cells transfected with CaV2.2 in control condition and 5-hexanoic acid (100 µM) application (n = 3, panel (<b>A</b>)); 2-aminooctanoic acid (100 µM) application (n = 4, panel (<b>B</b>)); 9-hydroxydecanoic acid (100 µM) application (n = 3, panel (<b>C</b>)); or 9-phenylnonanoic acid (100 µM) application (n = 4, panel (<b>D</b>)). Bars (<b>bottom</b>) represent normalized ICaV2.2 in control condition and with each compound. Statistical significance was evaluated by one-sample <span class="html-italic">t</span>-test against 1.</p>
Full article ">Figure 7
<p>Molecular dynamics simulations. The distance between the carbonyl carbon of carboxylic acid and the oxygen of the protein side chain was monitored along 25 ns of simulation (n = 3). (<b>A</b>) 9-phenylnonanoic acid interaction with Tyr1125; (<b>B</b>) 9-hydroxydecanoic acid interaction with Ser815.</p>
Full article ">
Back to TopTop